1
|
Griffin AHC, Small AM, Johnson RD, Medina AM, Kollar KT, Nazir RA, McGuire AM, Schumacher JA. Retinoic acid promotes second heart field addition and regulates ventral aorta patterning in zebrafish. Dev Biol 2025; 522:143-155. [PMID: 40147741 DOI: 10.1016/j.ydbio.2025.03.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 03/09/2025] [Accepted: 03/23/2025] [Indexed: 03/29/2025]
Abstract
Retinoic acid (RA) signaling is used reiteratively during vertebrate heart development. Its earliest known role is to restrict formation of the earlier-differentiating first heart field (FHF) progenitors, while promoting the differentiation of second heart field (SHF) progenitors that give rise to the arterial pole of the ventricle and outflow tract (OFT). However, requirements for RA signaling at later stages of cardiogenesis remain poorly understood. Here, we investigated the role of RA signaling after the later differentiating SHF cells have begun to add to the OFT. We found that inhibiting RA production in zebrafish beginning at 26 hours post fertilization (hpf) produced embryos that have smaller ventricles with fewer ventricular cardiomyocytes, and reduced number of smooth muscle cells in the bulbus arteriosus (BA) of the OFT. Our results suggest that the deficiency of the ventricular cardiomyocytes is due to reduced SHF addition to the arterial pole. In contrast to smaller ventricles and BA, later RA deficiency also results in a dramatically elongated posterior branch of the adjacent ventral aorta, which is surrounded by an increased number of smooth muscle cells. Altogether, our results reveal that RA signaling is required during the period of SHF addition to promote addition of ventricular cardiomyocytes, partition smooth muscle cells onto the BA and posterior ventral aorta, and to establish proper ventral aorta anterior-posterior patterning.
Collapse
Affiliation(s)
| | - Allison M Small
- Department of Biology, Miami University, Oxford, OH, 45056, USA
| | - Riley D Johnson
- Department of Biology, Miami University, Oxford, OH, 45056, USA
| | - Anna M Medina
- Department of Biology, Miami University, Oxford, OH, 45056, USA
| | - Kiki T Kollar
- Department of Biology, Miami University, Oxford, OH, 45056, USA
| | - Ridha A Nazir
- Department of Biology, Miami University, Oxford, OH, 45056, USA
| | | | - Jennifer A Schumacher
- Department of Biology, Miami University, Oxford, OH, 45056, USA; Department of Biological Sciences, Miami University, Hamilton, OH, 45011, USA.
| |
Collapse
|
2
|
Bisson JA, Gordillo M, Kumar R, de Silva N, Yang E, Banks KM, Shi ZD, Lee K, Yang D, Chung WK, Huangfu D, Evans T. GATA6 regulates WNT and BMP programs to pattern precardiac mesoderm during the earliest stages of human cardiogenesis. eLife 2025; 13:RP100797. [PMID: 40080060 PMCID: PMC11906159 DOI: 10.7554/elife.100797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/15/2025] Open
Abstract
Haploinsufficiency for GATA6 is associated with congenital heart disease (CHD) with variable comorbidity of pancreatic or diaphragm defects, although the etiology of disease is not well understood. Here, we used cardiac directed differentiation from human embryonic stem cells (hESCs) as a platform to study GATA6 function during early cardiogenesis. GATA6 loss-of-function hESCs had a profound impairment in cardiac progenitor cell (CPC) specification and cardiomyocyte (CM) generation due to early defects during the mesendoderm and lateral mesoderm patterning stages. Profiling by RNA-seq and CUT&RUN identified genes of the WNT and BMP programs regulated by GATA6 during early mesoderm patterning. Furthermore, interactome analysis detected GATA6 binding with developmental transcription factors and chromatin remodelers, suggesting cooperative regulation of cardiac lineage gene accessibility. We show that modulating WNT and BMP inputs during the first 48 hr of cardiac differentiation is sufficient to partially rescue CPC and CM defects in GATA6 heterozygous and homozygous mutant hESCs. This study provides evidence of the regulatory functions for GATA6 directing human precardiac mesoderm patterning during the earliest stages of cardiogenesis to further our understanding of haploinsufficiency causing CHD and the co-occurrence of cardiac and other organ defects caused by human GATA6 mutations.
Collapse
Affiliation(s)
- Joseph A Bisson
- Department of Surgery, Weill Cornell Medicine, New York, United States
| | - Miriam Gordillo
- Department of Surgery, Weill Cornell Medicine, New York, United States
| | - Ritu Kumar
- Department of Surgery, Weill Cornell Medicine, New York, United States
| | - Neranjan de Silva
- Department of Surgery, Weill Cornell Medicine, New York, United States
| | - Ellen Yang
- Department of Surgery, Weill Cornell Medicine, New York, United States
| | - Kelly M Banks
- Department of Surgery, Weill Cornell Medicine, New York, United States
| | - Zhong-Dong Shi
- Developmental Biology Program, Sloan Kettering Institute, New York, United States
| | - Kihyun Lee
- Developmental Biology Program, Sloan Kettering Institute, New York, United States
| | - Dapeng Yang
- Developmental Biology Program, Sloan Kettering Institute, New York, United States
| | - Wendy K Chung
- Childrens Hospital, Harvard Medical School, Boston, United States
| | - Danwei Huangfu
- Developmental Biology Program, Sloan Kettering Institute, New York, United States
| | - Todd Evans
- Department of Surgery, Weill Cornell Medicine, New York, United States
- Hartman Institute for Therapeutic Organ Regeneration, Weill Cornell Medicine, New York, United States
- Center for Genomic Health, Weill Cornell Medicine, New York, United States
| |
Collapse
|
3
|
Gans IM, Chepurko E, Chepurko V, Mangoba GA, Asson-Batres MA, Vary CP, Sawyer DB. Vitamin A deficiency and male-specific effects on heart function in mice. Biochem Biophys Res Commun 2025; 748:151300. [PMID: 39818189 PMCID: PMC11801225 DOI: 10.1016/j.bbrc.2025.151300] [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: 12/19/2024] [Revised: 12/30/2024] [Accepted: 01/08/2025] [Indexed: 01/18/2025]
Abstract
Hepatic stores of Vitamin A (retinol) are mobilized and metabolized in the heart following myocardial infarction. The physiological consequences of this mobilization are poorly understood. Here we used dietary depletion in a lecithin retinol acyltransferase mutant mouse line to induce Vitamin A deficiency and investigate the effects on cardiac function and recovery from myocardial infarction. We found that uninjured Vitamin A-depleted hearts had decreased contractile function but, paradoxically, improved recovery after injury. These effects on cardiac function were specific to male mice, which experienced more rapid and severe depletion of circulating Vitamin A. Following injury, Vitamin A deficiency also caused hepatic hypolipidemia and gene expression changes in heart and liver suggesting altered metabolism contributed to cardiac phenotypes.
Collapse
Affiliation(s)
- Ian M Gans
- Center for Molecular Medicine, Maine Health Institute for Research, 81 Research Drive, Scarborough, ME, USA.
| | - Elena Chepurko
- Center for Molecular Medicine, Maine Health Institute for Research, 81 Research Drive, Scarborough, ME, USA
| | - Vadim Chepurko
- Center for Molecular Medicine, Maine Health Institute for Research, 81 Research Drive, Scarborough, ME, USA
| | - Grazina A Mangoba
- Center for Molecular Medicine, Maine Health Institute for Research, 81 Research Drive, Scarborough, ME, USA
| | - Mary Ann Asson-Batres
- Center for Molecular Medicine, Maine Health Institute for Research, 81 Research Drive, Scarborough, ME, USA; Tennessee State University, Nashville, TN, USA
| | - Calvin P Vary
- Center for Molecular Medicine, Maine Health Institute for Research, 81 Research Drive, Scarborough, ME, USA; Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME, USA
| | - Douglas B Sawyer
- Center for Molecular Medicine, Maine Health Institute for Research, 81 Research Drive, Scarborough, ME, USA
| |
Collapse
|
4
|
Bisson JA, Gordillo M, Kumar R, de Silva N, Yang E, Banks KM, Shi ZD, Lee K, Yang D, Chung WK, Huangfu D, Evans T. GATA6 regulates WNT and BMP programs to pattern precardiac mesoderm during the earliest stages of human cardiogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.07.09.602666. [PMID: 39026742 PMCID: PMC11257636 DOI: 10.1101/2024.07.09.602666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Haploinsufficiency for GATA6 is associated with congenital heart disease (CHD) with variable comorbidity of pancreatic or diaphragm defects, although the etiology of disease is not well understood. Here, we used cardiac directed differentiation from human embryonic stem cells (hESCs) as a platform to study GATA6 function during early cardiogenesis. GATA6 loss-of-function hESCs had a profound impairment in cardiac progenitor cell (CPC) specification and cardiomyocyte (CM) generation due to early defects during the mesendoderm and lateral mesoderm patterning stages. Profiling by RNA-seq and CUT&RUN identified genes of the WNT and BMP programs regulated by GATA6 during early mesoderm patterning. Furthermore, interactome analysis detected GATA6 binding with developmental transcription factors and chromatin remodelers suggesting cooperative regulation of cardiac lineage gene accessibility. We show that modulating WNT and BMP inputs during the first 48 hours of cardiac differentiation is sufficient to partially rescue CPC and CM defects in GATA6 heterozygous and homozygous mutant hESCs. This study provides evidence of the regulatory functions for GATA6 directing human precardiac mesoderm patterning during the earliest stages of cardiogenesis to further our understanding of haploinsufficiency causing CHD and the co-occurrence of cardiac and other organ defects caused by human GATA6 mutations.
Collapse
Affiliation(s)
- Joseph A. Bisson
- Department of Surgery, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Miriam Gordillo
- Department of Surgery, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Ritu Kumar
- Department of Surgery, Weill Cornell Medicine, New York, NY, 10065, USA
- current address: Gladstone Institutes, San Francisco, CA, 94158, USA
| | - Neranjan de Silva
- Department of Surgery, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Ellen Yang
- Department of Surgery, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Kelly M. Banks
- Department of Surgery, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Zhong-Dong Shi
- Developmental Biology Program, Sloan Kettering Institute, New York, NY, 10065, USA
| | - Kihyun Lee
- Developmental Biology Program, Sloan Kettering Institute, New York, NY, 10065, USA
- current address: College of Pharmacy, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Dapeng Yang
- Developmental Biology Program, Sloan Kettering Institute, New York, NY, 10065, USA
| | - Wendy K. Chung
- Childrens Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Danwei Huangfu
- Developmental Biology Program, Sloan Kettering Institute, New York, NY, 10065, USA
| | - Todd Evans
- Department of Surgery, Weill Cornell Medicine, New York, NY, 10065, USA
- Hartman Institute for Therapeutic Organ Regeneration, Weill Cornell Medicine, New York, NY 10065, USA
- Center for Genomic Health, Weill Cornell Medicine, New York, NY 10065, USA
| |
Collapse
|
5
|
Fainsod A, Vadigepalli R. Rethinking retinoic acid self-regulation: A signaling robustness network approach. Curr Top Dev Biol 2024; 161:113-141. [PMID: 39870431 DOI: 10.1016/bs.ctdb.2024.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2025]
Abstract
All-trans retinoic acid (ATRA) signaling is a major pathway regulating numerous differentiation, proliferation, and patterning processes throughout life. ATRA biosynthesis depends on the nutritional availability of vitamin A and other retinoids and carotenoids, while it is sensitive to dietary and environmental toxicants. This nutritional and environmental influence requires a robustness response that constantly fine-tunes the ATRA metabolism to maintain a context-specific, physiological range of signaling levels. The ATRA metabolic and signaling network is characterized by the existence of multiple enzymes, transcription factors, and binding proteins capable of performing the same activity. The partial spatiotemporal expression overlap of these enzymes and proteins yields different network compositions in the cells and tissues where this pathway is active. Genetic polymorphisms affecting the activity of individual network components further impact the network composition variability and the self-regulatory feedback response to ATRA fluctuations. Experiments directly challenging the robustness response uncovered a Pareto optimality in the ATRA network, such that some genetic backgrounds efficiently deal with excess ATRA but are very limited in their robustness response to reduced ATRA and vice versa. We discuss a network-focused framework to describe the robustness response and the Pareto optimality of the ATRA metabolic and signaling network.
Collapse
Affiliation(s)
- Abraham Fainsod
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
| | - Rajanikanth Vadigepalli
- Daniel Baugh Institute for Functional Genomics and Computational Biology, Department of Pathology and Genomic Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States.
| |
Collapse
|
6
|
Bolesani E, Bornhorst D, Iyer LM, Zawada D, Friese N, Morgan M, Lange L, Gonzalez DM, Schrode N, Leffler A, Wunder J, Franke A, Drakhlis L, Sebra R, Schambach A, Goedel A, Dubois NC, Dobreva G, Moretti A, Zelaráyan LC, Abdelilah-Seyfried S, Zweigerdt R. Transient stabilization of human cardiovascular progenitor cells from human pluripotent stem cells in vitro reflects stage-specific heart development in vivo. Cardiovasc Res 2024; 120:1295-1311. [PMID: 38836637 DOI: 10.1093/cvr/cvae118] [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] [Received: 08/09/2023] [Revised: 01/11/2024] [Accepted: 04/06/2024] [Indexed: 06/06/2024] Open
Abstract
AIMS Understanding the molecular identity of human pluripotent stem cell (hPSC)-derived cardiac progenitors and mechanisms controlling their proliferation and differentiation is valuable for developmental biology and regenerative medicine. METHODS AND RESULTS Here, we show that chemical modulation of histone acetyl transferases (by IQ-1) and WNT (by CHIR99021) synergistically enables the transient and reversible block of directed cardiac differentiation progression on hPSCs. The resulting stabilized cardiovascular progenitors (SCPs) are characterized by ISL1pos/KI-67pos/NKX2-5neg expression. In the presence of the chemical inhibitors, SCPs maintain a proliferation quiescent state. Upon small molecules, removal SCPs resume proliferation and concomitant NKX2-5 up-regulation triggers cell-autonomous differentiation into cardiomyocytes. Directed differentiation of SCPs into the endothelial and smooth muscle lineages confirms their full developmental potential typical of bona fide cardiovascular progenitors. Single-cell RNA-sequencing-based transcriptional profiling of our in vitro generated human SCPs notably reflects the dynamic cellular composition of E8.25-E9.25 posterior second heart field of mouse hearts, hallmarked by nuclear receptor sub-family 2 group F member 2 expression. Investigating molecular mechanisms of SCP stabilization, we found that the cell-autonomously regulated retinoic acid and BMP signalling is governing SCP transition from quiescence towards proliferation and cell-autonomous differentiation, reminiscent of a niche-like behaviour. CONCLUSION The chemically defined and reversible nature of our stabilization approach provides an unprecedented opportunity to dissect mechanisms of cardiovascular progenitors' specification and reveal their cellular and molecular properties.
Collapse
Affiliation(s)
- Emiliano Bolesani
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiac, Thoracic, Transplantation and Vascular Surgery (HTTG), Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Dorothee Bornhorst
- Institute of Molecular Biology, Hannover Medical School, Hannover, Germany
- Institute of Biochemistry and Biology, Potsdam University, Potsdam, Germany
| | - Lavanya M Iyer
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany
- Epigenetic Regulation and Chromatin Architecture Group, Berlin Institute for Medical Systems Biology, Max-Delbrück Centre for Molecular Medicine, Berlin, Germany
| | - Dorota Zawada
- First Department of Medicine, Cardiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
| | - Nina Friese
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiac, Thoracic, Transplantation and Vascular Surgery (HTTG), Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Michael Morgan
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Lucas Lange
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - David M Gonzalez
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Nadine Schrode
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Andreas Leffler
- Department of Anesthesiology and Intensive Care Medicine, Hannover Medical School, Hannover, Germany
| | - Julian Wunder
- Department of Anesthesiology and Intensive Care Medicine, Hannover Medical School, Hannover, Germany
| | - Annika Franke
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiac, Thoracic, Transplantation and Vascular Surgery (HTTG), Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Lika Drakhlis
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiac, Thoracic, Transplantation and Vascular Surgery (HTTG), Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Robert Sebra
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Axel Schambach
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Alexander Goedel
- First Department of Medicine, Cardiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
| | - Nicole C Dubois
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Gergana Dobreva
- Department of Anatomy and Developmental Biology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Alessandra Moretti
- First Department of Medicine, Cardiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
| | - Laura C Zelaráyan
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany
| | - Salim Abdelilah-Seyfried
- Institute of Molecular Biology, Hannover Medical School, Hannover, Germany
- Institute of Biochemistry and Biology, Potsdam University, Potsdam, Germany
| | - Robert Zweigerdt
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiac, Thoracic, Transplantation and Vascular Surgery (HTTG), Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| |
Collapse
|
7
|
Edwards W, Bussey OK, Conlon FL. The Tbx20-TLE interaction is essential for the maintenance of the second heart field. Development 2023; 150:dev201677. [PMID: 37756602 PMCID: PMC10629681 DOI: 10.1242/dev.201677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023]
Abstract
T-box transcription factor 20 (Tbx20) plays a multifaceted role in cardiac morphogenesis and controls a broad gene regulatory network. However, the mechanism by which Tbx20 activates and represses target genes in a tissue-specific and temporal manner remains unclear. Studies show that Tbx20 directly interacts with the Transducin-like Enhancer of Split (TLE) family of proteins to mediate transcriptional repression. However, a function for the Tbx20-TLE transcriptional repression complex during heart development has yet to be established. We created a mouse model with a two amino acid substitution in the Tbx20 EH1 domain, thereby disrupting the Tbx20-TLE interaction. Disruption of this interaction impaired crucial morphogenic events, including cardiac looping and chamber formation. Transcriptional profiling of Tbx20EH1Mut hearts and analysis of putative direct targets revealed misexpression of the retinoic acid pathway and cardiac progenitor genes. Further, we show that altered cardiac progenitor development and function contribute to the severe cardiac defects in our model. Our studies indicate that TLE-mediated repression is a primary mechanism by which Tbx20 controls gene expression.
Collapse
Affiliation(s)
- Whitney Edwards
- Department of Biology and Genetics, McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Integrative Program for Biological & Genome Sciences, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Olivia K. Bussey
- Department of Biology and Genetics, McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Integrative Program for Biological & Genome Sciences, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Frank L. Conlon
- Department of Biology and Genetics, McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Integrative Program for Biological & Genome Sciences, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
8
|
Hu P, Wang B, Jin D, Gu Y, He H, Meng X, Zhu W, Chiang DY, Li W, MacRae CA, Zu Y. Modeling of large-scale hoxbb cluster deletions in zebrafish uncovers a role for segmentation pathways in atrioventricular boundary specification. Cell Mol Life Sci 2023; 80:317. [PMID: 37801106 PMCID: PMC11072906 DOI: 10.1007/s00018-023-04933-2] [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: 03/20/2023] [Accepted: 08/19/2023] [Indexed: 10/07/2023]
Abstract
Hox genes orchestrate the segmental specification of the muscular circulatory system in invertebrates but it has not proven straightforward to decipher segmental parallels in the vertebrate heart. Recently, patients with HOXB gene cluster deletion were found to exhibit abnormalities including atrioventricular canal defects. Using CRISPR, we established a mutant with the orthologous hoxbb cluster deletion in zebrafish. The mutant exhibited heart failure and atrioventricular regurgitation at 5 days. Analyzing the four genes in the hoxbb cluster, isolated deletion of hoxb1b-/- recapitulated the cardiac abnormalities, supporting hoxb1b as the causal gene. Both in situ and in vitro data indicated that hoxb1b regulates gata5 to inhibit hand2 expression and ultimately is required to pattern the vertebrate atrioventricular boundary. Together, these data reveal a role for segmental specification in vertebrate cardiac development and highlight the utility of CRISPR techniques for efficiently exploring the function of large structural genomic lesions.
Collapse
Affiliation(s)
- Peinan Hu
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, China
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China
| | - Bingqi Wang
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, China
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China
| | - Dongxu Jin
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, China
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China
| | - Yedan Gu
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, China
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China
| | - Hongyang He
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, China
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China
| | - Xiangli Meng
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, China
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China
| | - Wandi Zhu
- Cardiovascular Medicine Division, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - David Y Chiang
- Cardiovascular Medicine Division, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Weiming Li
- Department of Fisheries and Wildlife, Michigan State University, East Lansing, MI, 48824, USA
| | - Calum A MacRae
- Cardiovascular Medicine Division, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, 02115, USA.
| | - Yao Zu
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, China.
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China.
- Cardiovascular Medicine Division, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, 02115, USA.
| |
Collapse
|
9
|
Cherianidou A, Kappenberg F, Seidel F, Acharya A, Papazoglou P, Srinivasan SP, Hescheler J, Peng L, Leist M, Hengstler JG, Rahnenführer J, Sachinidis A. Transcriptome-based prediction of drugs, inhibiting cardiomyogenesis in human induced pluripotent stem cells. Cell Death Discov 2023; 9:321. [PMID: 37644023 PMCID: PMC10465524 DOI: 10.1038/s41420-023-01616-6] [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: 06/06/2023] [Revised: 07/31/2023] [Accepted: 08/16/2023] [Indexed: 08/31/2023] Open
Abstract
Animal studies for embryotoxicity evaluation of potential therapeutics and environmental factors are complex, costly, and time-consuming. Often, studies are not of human relevance because of species differences. In the present study, we recapitulated the process of cardiomyogenesis in human induced pluripotent stem cells (hiPSCs) by modulation of the Wnt signaling pathway to identify a key cardiomyogenesis gene signature that can be applied to identify compounds and/or stress factors compromising the cardiomyogenesis process. Among the 23 tested teratogens and 16 non-teratogens, we identified three retinoids including 13-cis-retinoic acid that completely block the process of cardiomyogenesis in hiPSCs. Moreover, we have identified an early gene signature consisting of 31 genes and associated biological processes that are severely affected by the retinoids. To predict the inhibitory potential of teratogens and non-teratogens in the process of cardiomyogenesis we established the "Developmental Cardiotoxicity Index" (CDI31g) that accurately differentiates teratogens and non-teratogens to do or do not affect the differentiation of hiPSCs to functional cardiomyocytes.
Collapse
Affiliation(s)
- Anna Cherianidou
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Physiology, Working Group Sachinidis, 50931 Cologne, Germany Robert-Koch-Str. 39, 50931, Cologne, Germany
| | - Franziska Kappenberg
- Department of Statistics, TU Dortmund University, Vogelpothsweg 87, 44227, Dortmund, Germany
| | - Florian Seidel
- Leibniz Research Centre for Working Environment and Human Factors at the Technical University of Dortmund (IfADo), Ardeystrasse 67, 44139, Dortmund, Germany
| | - Aviseka Acharya
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Physiology, Working Group Sachinidis, 50931 Cologne, Germany Robert-Koch-Str. 39, 50931, Cologne, Germany
| | - Panagiota Papazoglou
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Physiology, Working Group Sachinidis, 50931 Cologne, Germany Robert-Koch-Str. 39, 50931, Cologne, Germany
| | - Sureshkumar Perumal Srinivasan
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Physiology, Working Group Sachinidis, 50931 Cologne, Germany Robert-Koch-Str. 39, 50931, Cologne, Germany
| | - Jürgen Hescheler
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Physiology, Working Group Sachinidis, 50931 Cologne, Germany Robert-Koch-Str. 39, 50931, Cologne, Germany
| | - Luying Peng
- Heart Health Center, Shanghai East Hospital, School of Medicine, Tongji University, 200120, Shanghai and Research Units of Origin and Regulation of Heart Rhythm, Chinese Academy of Medical Sciences, 100730, Beijing, China
| | - Marcel Leist
- In Vitro Toxicology and Biomedicine, Department of Biology, University of Konstanz, Universitätsstr. 10, PO, Box M657, 78457, Constance, Germany
| | - Jan G Hengstler
- Leibniz Research Centre for Working Environment and Human Factors at the Technical University of Dortmund (IfADo), Ardeystrasse 67, 44139, Dortmund, Germany
| | - Jörg Rahnenführer
- Department of Statistics, TU Dortmund University, Vogelpothsweg 87, 44227, Dortmund, Germany
| | - Agapios Sachinidis
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Physiology, Working Group Sachinidis, 50931 Cologne, Germany Robert-Koch-Str. 39, 50931, Cologne, Germany.
| |
Collapse
|
10
|
Prodan N, Ershad F, Reyes-Alcaraz A, Li L, Mistretta B, Gonzalez L, Rao Z, Yu C, Gunaratne PH, Li N, Schwartz RJ, McConnell BK. Direct reprogramming of cardiomyocytes into cardiac Purkinje-like cells. iScience 2022; 25:105402. [PMID: 36388958 PMCID: PMC9646947 DOI: 10.1016/j.isci.2022.105402] [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: 06/06/2022] [Revised: 09/30/2022] [Accepted: 10/14/2022] [Indexed: 11/06/2022] Open
Abstract
Currently, there are no treatments that ameliorate cardiac cell death, the underlying basis of cardiovascular disease. An unexplored cell type in cardiac regeneration is cardiac Purkinje cells; specialized cells from the cardiac conduction system (CCS) responsible for propagating electrical signals. Purkinje cells have tremendous potential as a regenerative treatment because they may intrinsically integrate with the CCS of a recipient myocardium, resulting in more efficient electrical conduction in diseased hearts. This study is the first to demonstrate an effective protocol for the direct reprogramming of human cardiomyocytes into cardiac Purkinje-like cells using small molecules. The cells generated were genetically and functionally similar to native cardiac Purkinje cells, where expression of key cardiac Purkinje genes such as CNTN2, ETV1, PCP4, IRX3, SCN5a, HCN2 and the conduction of electrical signals with increased velocity was observed. This study may help to advance the quest to finding an optimized cell therapy for heart regeneration.
Collapse
Affiliation(s)
- Nicole Prodan
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, 4349 Martin Luther King Blvd, Health-2 (H2) Building, Room 5024, Houston, TX 77204-5037, USA
| | - Faheem Ershad
- Department of Biomedical Engineering, Cullen College of Engineering, University of Houston, Houston, TX 77204, USA
| | - Arfaxad Reyes-Alcaraz
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, 4349 Martin Luther King Blvd, Health-2 (H2) Building, Room 5024, Houston, TX 77204-5037, USA
| | - Luge Li
- Department of Medicine (Section of Cardiovascular Research), Baylor College of Medicine, Houston, TX 77030, USA
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA
| | - Brandon Mistretta
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
- Department of Biology and Biochemistry, UH-Sequencing & Gene Editing Core, University of Houston, Houston, TX 77204, USA
| | - Lei Gonzalez
- Department of Biomedical Engineering, Cullen College of Engineering, University of Houston, Houston, TX 77204, USA
| | - Zhoulyu Rao
- Department of Mechanical Engineering, Cullen College of Engineering, University of Houston, Houston, TX 77204, USA
| | - Cunjiang Yu
- Department of Biomedical Engineering, Cullen College of Engineering, University of Houston, Houston, TX 77204, USA
- Department of Mechanical Engineering, Cullen College of Engineering, University of Houston, Houston, TX 77204, USA
| | - Preethi H. Gunaratne
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
- Department of Biology and Biochemistry, UH-Sequencing & Gene Editing Core, University of Houston, Houston, TX 77204, USA
| | - Na Li
- Department of Medicine (Section of Cardiovascular Research), Baylor College of Medicine, Houston, TX 77030, USA
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA
| | - Robert J. Schwartz
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| | - Bradley K. McConnell
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, 4349 Martin Luther King Blvd, Health-2 (H2) Building, Room 5024, Houston, TX 77204-5037, USA
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| |
Collapse
|
11
|
Gonzalez DM, Schrode N, Ebrahim TAM, Broguiere N, Rossi G, Drakhlis L, Zweigerdt R, Lutolf MP, Beaumont KG, Sebra R, Dubois NC. Dissecting mechanisms of chamber-specific cardiac differentiation and its perturbation following retinoic acid exposure. Development 2022; 149:275658. [DOI: 10.1242/dev.200557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 05/26/2022] [Indexed: 11/20/2022]
Abstract
ABSTRACT
The specification of distinct cardiac lineages occurs before chamber formation and acquisition of bona fide atrial or ventricular identity. However, the mechanisms underlying these early specification events remain poorly understood. Here, we performed single cell analysis at the murine cardiac crescent, primitive heart tube and heart tube stages to uncover the transcriptional mechanisms underlying formation of atrial and ventricular cells. We find that progression towards differentiated cardiomyocytes occurs primarily based on heart field progenitor identity, and that progenitors contribute to ventricular or atrial identity through distinct differentiation mechanisms. We identify new candidate markers that define such differentiation processes and examine their expression dynamics using computational lineage trajectory methods. We further show that exposure to exogenous retinoic acid causes defects in ventricular chamber size, dysregulation in FGF signaling and a shunt in differentiation towards orthogonal lineages. Retinoic acid also causes defects in cell-cycle exit resulting in formation of hypomorphic ventricles. Collectively, our data identify, at a single cell level, distinct lineage trajectories during cardiac specification and differentiation, and the precise effects of manipulating cardiac progenitor patterning via retinoic acid signaling.
Collapse
Affiliation(s)
- David M. Gonzalez
- Icahn School of Medicine at Mount Sinai 1 Department of Cell, Developmental, and Regenerative Biology , , New York, NY 10029 , USA
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai 2 , New York, NY 10029 , USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai 3 , New York, NY 10029 , USA
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai 4 , New York, NY 10029 , USA
| | - Nadine Schrode
- Icahn School of Medicine at Mount Sinai 5 Department of Genetics and Genomic Sciences , , New York, NY 10029 , USA
| | - Tasneem A. M. Ebrahim
- Icahn School of Medicine at Mount Sinai 1 Department of Cell, Developmental, and Regenerative Biology , , New York, NY 10029 , USA
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai 2 , New York, NY 10029 , USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai 3 , New York, NY 10029 , USA
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai 4 , New York, NY 10029 , USA
| | - Nicolas Broguiere
- School of Life Sciences, EPFL 6 Laboratory of Stem Cell Bioengineering , , Lausanne CH-1015 , Switzerland
| | - Giuliana Rossi
- School of Life Sciences, EPFL 6 Laboratory of Stem Cell Bioengineering , , Lausanne CH-1015 , Switzerland
| | - Lika Drakhlis
- Roche Institute for Translational Bioengineering 7 , Roche Pharma Research and Early Development , Basel 4052 , Switzerland
| | - Robert Zweigerdt
- Roche Institute for Translational Bioengineering 7 , Roche Pharma Research and Early Development , Basel 4052 , Switzerland
| | - Matthias P. Lutolf
- School of Life Sciences, EPFL 6 Laboratory of Stem Cell Bioengineering , , Lausanne CH-1015 , Switzerland
- Roche Institute for Translational Bioengineering 7 , Roche Pharma Research and Early Development , Basel 4052 , Switzerland
| | - Kristin G. Beaumont
- Icahn School of Medicine at Mount Sinai 5 Department of Genetics and Genomic Sciences , , New York, NY 10029 , USA
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO) 8 , Department of Cardiothoracic, Transplantation and Vascular Surgery (HTTG) , , Hannover , Germany
- REBIRTH–Research Center for Translational Regenerative Medicine, Hannover Medical School 8 , Department of Cardiothoracic, Transplantation and Vascular Surgery (HTTG) , , Hannover , Germany
| | - Robert Sebra
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai 3 , New York, NY 10029 , USA
- Icahn School of Medicine at Mount Sinai 5 Department of Genetics and Genomic Sciences , , New York, NY 10029 , USA
- Sema4, a Mount Sinai venture 9 , Stamford, CT 06902 , USA
| | - Nicole C. Dubois
- Icahn School of Medicine at Mount Sinai 1 Department of Cell, Developmental, and Regenerative Biology , , New York, NY 10029 , USA
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai 2 , New York, NY 10029 , USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai 3 , New York, NY 10029 , USA
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai 4 , New York, NY 10029 , USA
| |
Collapse
|
12
|
Inhibition of RhoA and Cdc42 by miR-133a Modulates Retinoic Acid Signalling during Early Development of Posterior Cardiac Tube Segment. Int J Mol Sci 2022; 23:ijms23084179. [PMID: 35456995 PMCID: PMC9025022 DOI: 10.3390/ijms23084179] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 04/06/2022] [Accepted: 04/07/2022] [Indexed: 12/15/2022] Open
Abstract
It is well known that multiple microRNAs play crucial roles in cardiovascular development, including miR-133a. Additionally, retinoic acid regulates atrial marker expression. In order to analyse the role of miR-133a as a modulator of retinoic acid signalling during the posterior segment of heart tube formation, we performed functional experiments with miR-133a and retinoic acid by means of microinjections into the posterior cardiac precursors of both primitive endocardial tubes in chick embryos. Subsequently, we subjected embryos to whole mount in situ hybridisation, immunohistochemistry and qPCR analysis. Our results demonstrate that miR-133a represses RhoA and Cdc42, as well as Raldh2/Aldh1a2, and the specific atrial markers Tbx5 and AMHC1, which play a key role during differentiation. Furthermore, we observed that miR-133a upregulates p21 and downregulates cyclin A by repressing RhoA and Cdc42, respectively, thus functioning as a cell proliferation inhibitor. Additionally, retinoic acid represses miR-133a, while it increases Raldh2, Tbx5 and AMHC1. Given that RhoA and Cdc42 are involved in Raldh2 expression and that they are modulated by miR-133a, which is influenced by retinoic acid signalling, our results suggest the presence of a negative feedback mechanism between miR-133a and retinoic acid during early development of the posterior cardiac tube segment. Despite additional unexplored factors being possible contributors to this negative feedback mechanism, miR-133a might also be considered as a potential therapeutic tool for the diagnosis, therapy and prognosis of cardiac diseases.
Collapse
|
13
|
Coppola U, Waxman JS. Origin and evolutionary landscape of Nr2f transcription factors across Metazoa. PLoS One 2021; 16:e0254282. [PMID: 34807940 PMCID: PMC8608329 DOI: 10.1371/journal.pone.0254282] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 11/07/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Nuclear Receptor Subfamily 2 Group F (Nr2f) orphan nuclear hormone transcription factors (TFs) are fundamental regulators of many developmental processes in invertebrates and vertebrates. Despite the importance of these TFs throughout metazoan development, previous work has not clearly outlined their evolutionary history. RESULTS We integrated molecular phylogeny with comparisons of intron/exon structure, domain architecture, and syntenic conservation to define critical evolutionary events that distinguish the Nr2f gene family in Metazoa. Our data indicate that a single ancestral eumetazoan Nr2f gene predated six main Bilateria subfamilies, which include single Nr2f homologs, here referred to as Nr2f1/2/5/6, that are present in invertebrate protostomes and deuterostomes, Nr2f1/2 homologs in agnathans, and Nr2f1, Nr2f2, Nr2f5, and Nr2f6 orthologs that are found in gnathostomes. Four cnidarian Nr2f1/2/5/6 and three agnathan Nr2f1/2 members are each due to independent expansions, while the vertebrate Nr2f1/Nr2f2 and Nr2f5/Nr2f6 members each form paralogous groups that arose from the established series of whole-genome duplications (WGDs). Nr2f6 members are the most divergent Nr2f subfamily in gnathostomes. Interestingly, in contrast to the other gnathostome Nr2f subfamilies, Nr2f5 has been independently lost in numerous vertebrate lineages. Furthermore, our analysis shows there are differential expansions and losses of Nr2f genes in teleosts following their additional rounds of WGDs. CONCLUSION Overall, our analysis of Nr2f gene evolution helps to reveal the origins and previously unrecognized relationships of this ancient TF family, which may allow for greater insights into the conservation of Nr2f functions that shape Metazoan body plans.
Collapse
Affiliation(s)
- Ugo Coppola
- Molecular Cardiovascular Biology Division and Heart Institute, Cincinnati Children’s Research Foundation, Cincinnati, Ohio, United States of America
| | - Joshua S. Waxman
- Molecular Cardiovascular Biology Division and Heart Institute, Cincinnati Children’s Research Foundation, Cincinnati, Ohio, United States of America
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| |
Collapse
|
14
|
Duong TB, Waxman JS. Patterning of vertebrate cardiac progenitor fields by retinoic acid signaling. Genesis 2021; 59:e23458. [PMID: 34665508 DOI: 10.1002/dvg.23458] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 09/13/2021] [Accepted: 09/17/2021] [Indexed: 01/10/2023]
Abstract
The influence of retinoic acid (RA) signaling on vertebrate development has a well-studied history. Cumulatively, we now understand that RA signaling has a conserved requirement early in development restricting cardiac progenitors within the anterior lateral plate mesoderm of vertebrate embryos. Moreover, genetic and pharmacological manipulations of RA signaling in vertebrate models have shown that proper heart development is achieved through the deployment of positive and negative feedback mechanisms, which maintain appropriate RA levels. In this brief review, we present a chronological overview of key work that has led to a current model of the critical role for early RA signaling in limiting the generation of cardiac progenitors within vertebrate embryos. Furthermore, we integrate the previous work in mice and our recent findings using zebrafish, which together show that RA signaling has remarkably conserved influences on the later-differentiating progenitor populations at the arterial and venous poles. We discuss how recognizing the significant conservation of RA signaling on the differentiation of these progenitor populations offers new perspectives and may impact future work dedicated to examining vertebrate heart development.
Collapse
Affiliation(s)
- Tiffany B Duong
- Molecular Genetics Graduate Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Molecular Cardiovascular Biology Division and Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Joshua S Waxman
- Molecular Cardiovascular Biology Division and Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
15
|
Rankin SA, Steimle JD, Yang XH, Rydeen AB, Agarwal K, Chaturvedi P, Ikegami K, Herriges MJ, Moskowitz IP, Zorn AM. Tbx5 drives Aldh1a2 expression to regulate a RA-Hedgehog-Wnt gene regulatory network coordinating cardiopulmonary development. eLife 2021; 10:69288. [PMID: 34643182 PMCID: PMC8555986 DOI: 10.7554/elife.69288] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Accepted: 09/23/2021] [Indexed: 12/14/2022] Open
Abstract
The gene regulatory networks that coordinate the development of the cardiac and pulmonary systems are essential for terrestrial life but poorly understood. The T-box transcription factor Tbx5 is critical for both pulmonary specification and heart development, but how these activities are mechanistically integrated remains unclear. Here using Xenopus and mouse embryos, we establish molecular links between Tbx5 and retinoic acid (RA) signaling in the mesoderm and between RA signaling and sonic hedgehog expression in the endoderm to unveil a conserved RA-Hedgehog-Wnt signaling cascade coordinating cardiopulmonary (CP) development. We demonstrate that Tbx5 directly maintains expression of aldh1a2, the RA-synthesizing enzyme, in the foregut lateral plate mesoderm via an evolutionarily conserved intronic enhancer. Tbx5 promotes posterior second heart field identity in a positive feedback loop with RA, antagonizing a Fgf8-Cyp regulatory module to restrict FGF activity to the anterior. We find that Tbx5/Aldh1a2-dependent RA signaling directly activates shh transcription in the adjacent foregut endoderm through a conserved MACS1 enhancer. Hedgehog signaling coordinates with Tbx5 in the mesoderm to activate expression of wnt2/2b, which induces pulmonary fate in the foregut endoderm. These results provide mechanistic insight into the interrelationship between heart and lung development informing CP evolution and birth defects.
Collapse
Affiliation(s)
- Scott A Rankin
- Center for Stem Cell and Organoid Medicine (CuSTOM), Division of Developmental Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, United States
| | - Jeffrey D Steimle
- Department of Pediatrics, University of Chicago, Chicago, United States.,Department of Pathology, University of Chicago, Chicago, United States.,Department of Human Genetics, University of Chicago, Chicago, United States
| | - Xinan H Yang
- Department of Pediatrics, University of Chicago, Chicago, United States.,Department of Pathology, University of Chicago, Chicago, United States.,Department of Human Genetics, University of Chicago, Chicago, United States
| | - Ariel B Rydeen
- Department of Pediatrics, University of Chicago, Chicago, United States.,Department of Pathology, University of Chicago, Chicago, United States.,Department of Human Genetics, University of Chicago, Chicago, United States
| | - Kunal Agarwal
- Center for Stem Cell and Organoid Medicine (CuSTOM), Division of Developmental Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, United States
| | - Praneet Chaturvedi
- Center for Stem Cell and Organoid Medicine (CuSTOM), Division of Developmental Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, United States
| | - Kohta Ikegami
- Department of Pediatrics, University of Chicago, Chicago, United States
| | | | - Ivan P Moskowitz
- Department of Pediatrics, University of Chicago, Chicago, United States.,Department of Pathology, University of Chicago, Chicago, United States.,Department of Human Genetics, University of Chicago, Chicago, United States
| | - Aaron M Zorn
- Center for Stem Cell and Organoid Medicine (CuSTOM), Division of Developmental Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, United States.,University of Cincinnati, College of Medicine, Department of Pediatrics, Chicago, United States
| |
Collapse
|
16
|
Duong TB, Holowiecki A, Waxman JS. Retinoic acid signaling restricts the size of the first heart field within the anterior lateral plate mesoderm. Dev Biol 2021; 473:119-129. [PMID: 33607112 DOI: 10.1016/j.ydbio.2021.02.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 02/09/2021] [Accepted: 02/10/2021] [Indexed: 01/27/2023]
Abstract
Retinoic acid (RA) signaling is required to restrict heart size through limiting the posterior boundary of the vertebrate cardiac progenitor field within the anterior lateral plate mesoderm (ALPM). However, we still do not fully understand how different cardiac progenitor populations that contribute to the developing heart, including earlier-differentiating first heart field (FHF), later-differentiating second heart field (SHF), and neural crest-derived progenitors, are each affected in RA-deficient embryos. Here, we quantified the number of cardiac progenitors and differentiating cardiomyocytes (CMs) in RA-deficient zebrafish embryos. While Nkx2.5+ cells were increased overall in the nascent hearts of RA-deficient embryos, unexpectedly, we found that the major effect within this population was a significant expansion in the number of differentiating FHF CMs. In contrast to the expansion of the FHF, there was a progressive decrease in SHF progenitors at the arterial pole as the heart tube elongated. Temporal differentiation assays and immunostaining in RA-deficient embryos showed that the outflow tracts (OFTs) of the hearts were significantly smaller, containing fewer differentiated SHF-derived ventricular CMs and a complete absence of SHF-derived smooth muscle at later stages. At the venous pole of the heart, pacemaker cells of the sinoatrial node also failed to differentiate in RA-deficient embryos. Interestingly, genetic lineage tracing showed that the number of neural-crest derived CMs was not altered within the enlarged hearts of RA-deficient zebrafish embryos. Altogether, our data show that the enlarged hearts in RA-deficient zebrafish embryos are comprised of an expansion in earlier differentiating FHF-derived CMs coupled with a progressive depletion of the SHF, suggesting RA signaling determines the relative ratios of earlier- and later-differentiation cardiac progenitors within an expanded cardiac progenitor pool.
Collapse
Affiliation(s)
- Tiffany B Duong
- Molecular Genetics Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Molecular Cardiovascular Biology Division and Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Andrew Holowiecki
- Molecular Cardiovascular Biology Division and Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Joshua S Waxman
- Molecular Cardiovascular Biology Division and Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
17
|
Kemmler CL, Riemslagh FW, Moran HR, Mosimann C. From Stripes to a Beating Heart: Early Cardiac Development in Zebrafish. J Cardiovasc Dev Dis 2021; 8:17. [PMID: 33578943 PMCID: PMC7916704 DOI: 10.3390/jcdd8020017] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 02/05/2021] [Accepted: 02/07/2021] [Indexed: 12/18/2022] Open
Abstract
The heart is the first functional organ to form during vertebrate development. Congenital heart defects are the most common type of human birth defect, many originating as anomalies in early heart development. The zebrafish model provides an accessible vertebrate system to study early heart morphogenesis and to gain new insights into the mechanisms of congenital disease. Although composed of only two chambers compared with the four-chambered mammalian heart, the zebrafish heart integrates the core processes and cellular lineages central to cardiac development across vertebrates. The rapid, translucent development of zebrafish is amenable to in vivo imaging and genetic lineage tracing techniques, providing versatile tools to study heart field migration and myocardial progenitor addition and differentiation. Combining transgenic reporters with rapid genome engineering via CRISPR-Cas9 allows for functional testing of candidate genes associated with congenital heart defects and the discovery of molecular causes leading to observed phenotypes. Here, we summarize key insights gained through zebrafish studies into the early patterning of uncommitted lateral plate mesoderm into cardiac progenitors and their regulation. We review the central genetic mechanisms, available tools, and approaches for modeling congenital heart anomalies in the zebrafish as a representative vertebrate model.
Collapse
Affiliation(s)
| | | | | | - Christian Mosimann
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine and Children’s Hospital Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA; (C.L.K.); (F.W.R.); (H.R.M.)
| |
Collapse
|
18
|
Sharma A, Wasson LK, Willcox JAL, Morton SU, Gorham JM, DeLaughter DM, Neyazi M, Schmid M, Agarwal R, Jang MY, Toepfer CN, Ward T, Kim Y, Pereira AC, DePalma SR, Tai A, Kim S, Conner D, Bernstein D, Gelb BD, Chung WK, Goldmuntz E, Porter G, Tristani-Firouzi M, Srivastava D, Seidman JG, Seidman CE. GATA6 mutations in hiPSCs inform mechanisms for maldevelopment of the heart, pancreas, and diaphragm. eLife 2020; 9:e53278. [PMID: 33054971 PMCID: PMC7593088 DOI: 10.7554/elife.53278] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 10/14/2020] [Indexed: 12/18/2022] Open
Abstract
Damaging GATA6 variants cause cardiac outflow tract defects, sometimes with pancreatic and diaphragmic malformations. To define molecular mechanisms for these diverse developmental defects, we studied transcriptional and epigenetic responses to GATA6 loss of function (LoF) and missense variants during cardiomyocyte differentiation of isogenic human induced pluripotent stem cells. We show that GATA6 is a pioneer factor in cardiac development, regulating SMYD1 that activates HAND2, and KDR that with HAND2 orchestrates outflow tract formation. LoF variants perturbed cardiac genes and also endoderm lineage genes that direct PDX1 expression and pancreatic development. Remarkably, an exon 4 GATA6 missense variant, highly associated with extra-cardiac malformations, caused ectopic pioneer activities, profoundly diminishing GATA4, FOXA1/2, and PDX1 expression and increasing normal retinoic acid signaling that promotes diaphragm development. These aberrant epigenetic and transcriptional signatures illuminate the molecular mechanisms for cardiovascular malformations, pancreas and diaphragm dysgenesis that arise in patients with distinct GATA6 variants.
Collapse
Affiliation(s)
- Arun Sharma
- Department of Genetics, Harvard Medical SchoolBostonUnited States
- Smidt Heart Institute, Cedars-Sinai Medical CenterLos AngelesUnited States
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical CenterLos AngelesUnited States
| | - Lauren K Wasson
- Department of Genetics, Harvard Medical SchoolBostonUnited States
- Howard Hughes Medical Institute, Harvard Medical SchoolBostonUnited States
| | - Jon AL Willcox
- Department of Genetics, Harvard Medical SchoolBostonUnited States
| | - Sarah U Morton
- Department of Genetics, Harvard Medical SchoolBostonUnited States
- Division of Newborn Medicine, Boston Children's HospitalBostonUnited States
| | - Joshua M Gorham
- Department of Genetics, Harvard Medical SchoolBostonUnited States
| | | | - Meraj Neyazi
- Department of Genetics, Harvard Medical SchoolBostonUnited States
- Hannover Medical SchoolHannoverGermany
| | - Manuel Schmid
- Department of Genetics, Harvard Medical SchoolBostonUnited States
- Deutsches Herzzentrum München, Technische Universität MünchenMunichGermany
| | - Radhika Agarwal
- Department of Genetics, Harvard Medical SchoolBostonUnited States
| | - Min Young Jang
- Department of Genetics, Harvard Medical SchoolBostonUnited States
| | - Christopher N Toepfer
- Department of Genetics, Harvard Medical SchoolBostonUnited States
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of OxfordOxfordUnited Kingdom
- Wellcome Centre for Human Genetics, University of OxfordOxfordUnited Kingdom
| | - Tarsha Ward
- Department of Genetics, Harvard Medical SchoolBostonUnited States
| | - Yuri Kim
- Department of Genetics, Harvard Medical SchoolBostonUnited States
| | - Alexandre C Pereira
- Department of Genetics, Harvard Medical SchoolBostonUnited States
- Laboratory of Genetics and Molecular Cardiology, Heart Institute, Medical School of University of Sao PauloSao PauloBrazil
| | - Steven R DePalma
- Department of Genetics, Harvard Medical SchoolBostonUnited States
| | - Angela Tai
- Department of Genetics, Harvard Medical SchoolBostonUnited States
| | - Seongwon Kim
- Department of Genetics, Harvard Medical SchoolBostonUnited States
| | - David Conner
- Department of Genetics, Harvard Medical SchoolBostonUnited States
| | - Daniel Bernstein
- Department of Pediatrics, Stanford University School of MedicineStanfordUnited States
| | - Bruce D Gelb
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Wendy K Chung
- Department of Medicine, Columbia University Medical CenterNew YorkUnited States
| | - Elizabeth Goldmuntz
- Department of Pediatrics, The Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - George Porter
- Department of Pediatrics, University of Rochester Medical CenterRochesterUnited States
| | - Martin Tristani-Firouzi
- Division of Pediatric Cardiology, University of Utah School of MedicineSalt Lake CityUnited States
| | | | | | - Christine E Seidman
- Department of Genetics, Harvard Medical SchoolBostonUnited States
- Howard Hughes Medical Institute, Harvard Medical SchoolBostonUnited States
- Cardiovascular Division, Department of Medicine, Brigham and Women's HospitalBostonUnited States
| | | |
Collapse
|
19
|
Caregiver-reported physical health status of children and young people with fetal alcohol spectrum disorder. J Dev Orig Health Dis 2020; 12:420-427. [PMID: 32513328 DOI: 10.1017/s2040174420000537] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
While fetal alcohol spectrum disorder (FASD) has primarily been thought of as a neurodevelopmental condition, research is beginning to highlight its 'whole-body' implications. Accordingly, the current study sought to provide a snapshot of potential health issues. Caregivers of children (median age of 12 years) with an FASD diagnosis were invited to participate in an online survey. Information relating to sample demographics, FASD status of the child and health outcomes were collected. The prevalence of health conditions reported in the FASD sample was compared against national prevalence data. Multiple linear regression utilising a stepwise approach was used to investigate potential predictors of the number of diagnosed health conditions. Survey data were from an international cohort (n = 197), with the majority of respondents based in Australia (40.2%) or the United States (27.7%). The most commonly reported diagnosed health conditions were eye conditions (44.7%), asthma (34.5%), heart conditions (34.0%) and skin conditions (27.4%). Binomial testing indicated the proportion of children diagnosed with these disorders was generally higher in the current FASD population, compared to national prevalence data. Indicators of metabolic dysfunction including diabetes and obesity were not significantly different compared to national prevalence data. Age of FASD diagnosis, existence of comorbid mental health conditions and the primary caregiver being in paid work were identified as being associated with the prevalence of diagnosed health conditions. Overall, the study has provided an up-to-date snapshot of health problems reported in a sample of children with FASD, confirming their increased risk of adverse health outcomes.
Collapse
|
20
|
Roberts C. Regulating Retinoic Acid Availability during Development and Regeneration: The Role of the CYP26 Enzymes. J Dev Biol 2020; 8:jdb8010006. [PMID: 32151018 PMCID: PMC7151129 DOI: 10.3390/jdb8010006] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 02/17/2020] [Accepted: 02/17/2020] [Indexed: 12/16/2022] Open
Abstract
This review focuses on the role of the Cytochrome p450 subfamily 26 (CYP26) retinoic acid (RA) degrading enzymes during development and regeneration. Cyp26 enzymes, along with retinoic acid synthesising enzymes, are absolutely required for RA homeostasis in these processes by regulating availability of RA for receptor binding and signalling. Cyp26 enzymes are necessary to generate RA gradients and to protect specific tissues from RA signalling. Disruption of RA homeostasis leads to a wide variety of embryonic defects affecting many tissues. Here, the function of CYP26 enzymes is discussed in the context of the RA signalling pathway, enzymatic structure and biochemistry, human genetic disease, and function in development and regeneration as elucidated from animal model studies.
Collapse
Affiliation(s)
- Catherine Roberts
- Developmental Biology of Birth Defects, UCL-GOS Institute of Child Health, 30 Guilford St, London WC1N 1EH, UK;
- Institute of Medical and Biomedical Education St George’s, University of London, Cranmer Terrace, Tooting, London SW17 0RE, UK
| |
Collapse
|
21
|
Xu C, Dai Y, Mohsin A, Hang H, Zhuang Y, Guo M. Mapping molecular pathways for embryonic Sertoli cells derivation based on differentiation model of mouse embryonic stem cells. Stem Cell Res Ther 2020; 11:85. [PMID: 32102677 PMCID: PMC7045406 DOI: 10.1186/s13287-020-01600-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 02/07/2020] [Accepted: 02/11/2020] [Indexed: 12/27/2022] Open
Abstract
Background Embryonic Sertoli cells (eSCs) have been known for playing important roles in male reproductive development system. In current studies, eSCs were mainly generated from induced intermediate mesoderm. The deriving mechanism of eSCs has been unclear so far. Therefore, this work was aimed to reveal the molecular pathways during derivation of eSCs. Methods In this scenario, a differentiation model from mouse embryonic stem cells (mESCs) to eSCs was established through spatiotemporal control of 5 key factors, Wilms tumor 1 homolog (Wt1), GATA binding protein 4 (Gata4), nuclear receptor subfamily 5, group A, member 1 (Nr5a1, i.e., Sf1), SRY (sex determining region Y)-box 9 (Sox9), doublesex, and mab-3 related transcription factor 1 (Dmrt1). To investigate the molecular mechanism, these key factors were respectively manipulated through a light-switchable (light-on) system, tetracycline-switchable (Tet-on) system, and CRISPR/Cas9 knock out (KO) system. Results Via the established approach, some embryonic Sertoli-like cells (eSLCs) were induced from mESCs and formed ring-like or tubular-like structures. The key factors were respectively manipulated and revealed their roles in the derivation of these eSLCs. Based on these results, some molecular pathways were mapped during the development of coelomic epithelial somatic cells to eSCs. Conclusions This differentiation model provided a high controllability of some key factors and brought a novel insight into the deriving mechanism of Sertoli cells. Supplementary information accompanies this paper at 10.1186/s13287-020-01600-2.
Collapse
Affiliation(s)
- Chenze Xu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, People's Republic of China
| | - Yichen Dai
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, People's Republic of China
| | - Ali Mohsin
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, People's Republic of China
| | - Haifeng Hang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, People's Republic of China.,Engineering Research Centre of Processes System, Ministry of Education, East China University of Science and Technology, 130 Meilong Rd., Shanghai, 200237, China
| | - Yingping Zhuang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, People's Republic of China.,Engineering Research Centre of Processes System, Ministry of Education, East China University of Science and Technology, 130 Meilong Rd., Shanghai, 200237, China
| | - Meijin Guo
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, People's Republic of China. .,Engineering Research Centre of Processes System, Ministry of Education, East China University of Science and Technology, 130 Meilong Rd., Shanghai, 200237, China. .,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|
22
|
Sirbu IO, Chiş AR, Moise AR. Role of carotenoids and retinoids during heart development. Biochim Biophys Acta Mol Cell Biol Lipids 2020; 1865:158636. [PMID: 31978553 DOI: 10.1016/j.bbalip.2020.158636] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 01/15/2020] [Accepted: 01/16/2020] [Indexed: 02/08/2023]
Abstract
The nutritional requirements of the developing embryo are complex. In the case of dietary vitamin A (retinol, retinyl esters and provitamin A carotenoids), maternal derived nutrients serve as precursors to signaling molecules such as retinoic acid, which is required for embryonic patterning and organogenesis. Despite variations in the composition and levels of maternal vitamin A, embryonic tissues need to generate a precise amount of retinoic acid to avoid congenital malformations. Here, we summarize recent findings regarding the role and metabolism of vitamin A during heart development and we survey the association of genes known to affect retinoid metabolism or signaling with various inherited disorders. A better understanding of the roles of vitamin A in the heart and of the factors that affect retinoid metabolism and signaling can help design strategies to meet nutritional needs and to prevent birth defects and disorders associated with altered retinoid metabolism. This article is part of a Special Issue entitled Carotenoids recent advances in cell and molecular biology edited by Johannes von Lintig and Loredana Quadro.
Collapse
Affiliation(s)
- Ioan Ovidiu Sirbu
- Biochemistry Department, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Nr. 2, 300041 Timisoara, Romania; Timisoara Institute of Complex Systems, V. Lucaciu 18, 300044 Timisoara, Romania.
| | - Aimée Rodica Chiş
- Biochemistry Department, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Nr. 2, 300041 Timisoara, Romania
| | - Alexander Radu Moise
- Medical Sciences Division, Northern Ontario School of Medicine, Sudbury, ON P3E 2C6, Canada; Department of Chemistry and Biochemistry, Biology and Biomolecular Sciences Program, Laurentian University, Sudbury, ON P3E 2C6, Canada.
| |
Collapse
|
23
|
Witman N, Zhou C, Grote Beverborg N, Sahara M, Chien KR. Cardiac progenitors and paracrine mediators in cardiogenesis and heart regeneration. Semin Cell Dev Biol 2019; 100:29-51. [PMID: 31862220 DOI: 10.1016/j.semcdb.2019.10.011] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 10/13/2019] [Accepted: 10/21/2019] [Indexed: 12/17/2022]
Abstract
The mammalian hearts have the least regenerative capabilities among tissues and organs. As such, heart regeneration has been and continues to be the ultimate goal in the treatment against acquired and congenital heart diseases. Uncovering such a long-awaited therapy is still extremely challenging in the current settings. On the other hand, this desperate need for effective heart regeneration has developed various forms of modern biotechnologies in recent years. These involve the transplantation of pluripotent stem cell-derived cardiac progenitors or cardiomyocytes generated in vitro and novel biochemical molecules along with tissue engineering platforms. Such newly generated technologies and approaches have been shown to effectively proliferate cardiomyocytes and promote heart repair in the diseased settings, albeit mainly preclinically. These novel tools and medicines give somehow credence to breaking down the barriers associated with re-building heart muscle. However, in order to maximize efficacy and achieve better clinical outcomes through these cell-based and/or cell-free therapies, it is crucial to understand more deeply the developmental cellular hierarchies/paths and molecular mechanisms in normal or pathological cardiogenesis. Indeed, the morphogenetic process of mammalian cardiac development is highly complex and spatiotemporally regulated by various types of cardiac progenitors and their paracrine mediators. Here we discuss the most recent knowledge and findings in cardiac progenitor cell biology and the major cardiogenic paracrine mediators in the settings of cardiogenesis, congenital heart disease, and heart regeneration.
Collapse
Affiliation(s)
- Nevin Witman
- Department of Cell and Molecular Biology, Karolinska Institutet, SE-171 77 Stockholm, Sweden; Department of Medicine, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Chikai Zhou
- Department of Cell and Molecular Biology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Niels Grote Beverborg
- Department of Cell and Molecular Biology, Karolinska Institutet, SE-171 77 Stockholm, Sweden; Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Makoto Sahara
- Department of Cell and Molecular Biology, Karolinska Institutet, SE-171 77 Stockholm, Sweden; Department of Medicine, Karolinska Institutet, SE-171 77 Stockholm, Sweden; Department of Surgery, Yale University School of Medicine, CT, USA.
| | - Kenneth R Chien
- Department of Cell and Molecular Biology, Karolinska Institutet, SE-171 77 Stockholm, Sweden; Department of Medicine, Karolinska Institutet, SE-171 77 Stockholm, Sweden.
| |
Collapse
|
24
|
Maldonado E, Rangel-Huerta E, González-Gómez R, Fajardo-Alvarado G, Morillo-Velarde PS. Octopus insularis as a new marine model for evolutionary developmental biology. Biol Open 2019; 8:bio046086. [PMID: 31666222 PMCID: PMC6899024 DOI: 10.1242/bio.046086] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 10/15/2019] [Indexed: 11/20/2022] Open
Abstract
Octopuses are intriguing organisms that, together with squids and cuttlefishes, form the extant coleoid cephalopods. This group includes many species that can potentially be used as models in the fields of biomedicine, developmental biology, evolution, neuroscience and even for robotics research. The purpose of this work is to first present a simple method for maintaining Octopus insularis embryos under a laboratory setup. Second, we show that these embryos are suitable for detailed analyses of specific traits that appear during developmental stages, including the eyes, hearts, arms, suckers, chromatophores and Kölliker's organs. Similar complex traits between cephalopods and vertebrates such as the visual, cardiovascular, neural and pigmentation systems are generally considered to be a result of parallel evolution. We propose that O. insularis embryos could be used as a model for evolutionary developmental biology (or EvoDevo) research, where comparisons of the morphogenetic steps in the building of equivalent organs between cephalopods and known vertebrate model systems could shed light on evolutionary convergences and deep homologies.
Collapse
Affiliation(s)
- Ernesto Maldonado
- EvoDevo Research Group, Unidad de Sistemas Arrecifales, Instituto de Ciencias del Mar y Limnología, Universidad Nacional Autónoma de México, Puerto Morelos, Quintana Roo, México 77580
| | - Emma Rangel-Huerta
- EvoDevo Research Group, Unidad de Sistemas Arrecifales, Instituto de Ciencias del Mar y Limnología, Universidad Nacional Autónoma de México, Puerto Morelos, Quintana Roo, México 77580
- Doctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de México, UNAM, México 77580
| | - Roberto González-Gómez
- Posgrado en Ecología y Pesquerías, Universidad Veracruzana, Boca del Río, Veracruz, México 94290
- Instituto de Ciencias Marinas y Pesquerías, Universidad Veracruzana, Boca del Río, Veracruz, México 94290
| | - Gabriel Fajardo-Alvarado
- Posgrado en Ecología y Pesquerías, Universidad Veracruzana, Boca del Río, Veracruz, México 94290
- Instituto de Ciencias Marinas y Pesquerías, Universidad Veracruzana, Boca del Río, Veracruz, México 94290
| | - Piedad S Morillo-Velarde
- Instituto de Ciencias Marinas y Pesquerías, Universidad Veracruzana, Boca del Río, Veracruz, México 94290
- CONACyT-Instituto de Ciencias Marinas y Pesquerías, Universidad Veracruzana, Boca del Río, Veracruz, México 94290
| |
Collapse
|