1
|
Lu J, Peng B, Wang W, Zou Y. Epithelial-mesenchymal crosstalk: the scriptwriter of craniofacial morphogenesis. Front Cell Dev Biol 2024; 12:1497002. [PMID: 39583201 PMCID: PMC11582012 DOI: 10.3389/fcell.2024.1497002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 10/25/2024] [Indexed: 11/26/2024] Open
Abstract
Epithelial-mesenchymal interactions (EMI) are fundamental mechanisms in regulating development and organogenesis. Here we summarized the signaling mechanisms involved in EMI in the major developmental events during craniofacial morphogenesis, including neural crest cell induction, facial primordial growth as well as fusion processes. Regional specificity/polarity are demonstrated in the expression of most signaling molecules that usually act in a mutually synergistic/antagonistic manner. The underlying mechanisms of pathogenesis due to disrupted EMI was also discussed in this review.
Collapse
Affiliation(s)
- Junjie Lu
- School of Life Science and Technology, Jinan University, Guangzhou, China
| | - Bo Peng
- Institute for Environmental and Climate Research, Jinan University, Guangzhou, China
| | - Wenyi Wang
- School of Life Science and Technology, Jinan University, Guangzhou, China
| | - Yi Zou
- School of Life Science and Technology, Jinan University, Guangzhou, China
| |
Collapse
|
2
|
Kim S, Koppitch K, Parvez RK, Guo J, Achieng M, Schnell J, Lindström NO, McMahon AP. Comparative single-cell analyses identify shared and divergent features of human and mouse kidney development. Dev Cell 2024; 59:2912-2930.e7. [PMID: 39121855 DOI: 10.1016/j.devcel.2024.07.013] [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: 05/20/2023] [Revised: 04/02/2024] [Accepted: 07/12/2024] [Indexed: 08/12/2024]
Abstract
The mammalian kidney maintains fluid homeostasis through diverse epithelial cell types generated from nephron and ureteric progenitor cells. To extend a developmental understanding of the kidney's epithelial networks, we compared chromatin organization (single-nuclear assay for transposase-accessible chromatin sequencing [ATAC-seq]; 112,864 nuclei) and gene expression (single-cell/nuclear RNA sequencing [RNA-seq]; 109,477 cells/nuclei) in the developing human (10.6-17.6 weeks; n = 10) and mouse (post-natal day [P]0; n = 10) kidney, supplementing analysis with published mouse datasets from earlier stages. Single-cell/nuclear datasets were analyzed at a species level, and then nephron and ureteric cellular lineages were extracted and integrated into a common, cross-species, multimodal dataset. Comparative computational analyses identified conserved and divergent features of chromatin organization and linked gene activity, identifying species-specific and cell-type-specific regulatory programs. In situ validation of human-enriched gene activity points to human-specific signaling interactions in kidney development. Further, human-specific enhancer regions were linked to kidney diseases through genome-wide association studies (GWASs), highlighting the potential for clinical insight from developmental modeling.
Collapse
Affiliation(s)
- Sunghyun Kim
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Kari Koppitch
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Riana K Parvez
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Jinjin Guo
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - MaryAnne Achieng
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Jack Schnell
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Nils O Lindström
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Andrew P McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA.
| |
Collapse
|
3
|
Jang J, Jung H, Jeong J, Jeon J, Lee K, Jang HR, Han JW, Lee J. Modeling doxorubicin-induced-cardiotoxicity through breast cancer patient specific iPSC-derived heart organoid. Heliyon 2024; 10:e38714. [PMID: 39640743 PMCID: PMC11620051 DOI: 10.1016/j.heliyon.2024.e38714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 09/26/2024] [Accepted: 09/27/2024] [Indexed: 12/07/2024] Open
Abstract
Heart organoid (HO) technology has successfully overcome the limitations of two-dimensional (2D) disease modeling and drug testing, thereby emerging as a valuable tool in drug discovery for assessing toxicity and efficacy. However, its ability to distinguish drug responses among individuals remain unclear, which is crucial for developing predictive models. We addressed this gap by comparing human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) with human induced pluripotent stem cell-derived heart organoids (hiPSC-HOs) in the context of doxorubicin-induced cardiotoxicity (DIC). For this study, we utilized hiPSCs generated from breast cancer patients who had previously been treated with doxorubicin. By comparing groups with and without DIC, we examined various parameters, including cell viability, mRNA expression, protein expression and electrophysiological variations. The results of our analysis revealed significant differences between these groups, providing insights into hiPSC-HOs as a potential platform for testing differences in drug responses among patients.
Collapse
Affiliation(s)
- Jiye Jang
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Epigenome Dynamics Control Research Center (EDCRC), School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Hyewon Jung
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Epigenome Dynamics Control Research Center (EDCRC), School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Jaekyun Jeong
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Epigenome Dynamics Control Research Center (EDCRC), School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Junseok Jeon
- Division of Nephrology, Department of Medicine, Cell and Gene Therapy Institute, Samsung Medical Center, Sungkyunkwan University, Seoul, Republic of Korea
| | - Kyungho Lee
- Division of Nephrology, Department of Medicine, Cell and Gene Therapy Institute, Samsung Medical Center, Sungkyunkwan University, Seoul, Republic of Korea
| | - Hye Ryoun Jang
- Division of Nephrology, Department of Medicine, Cell and Gene Therapy Institute, Samsung Medical Center, Sungkyunkwan University, Seoul, Republic of Korea
| | - Jeung-Whan Han
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Epigenome Dynamics Control Research Center (EDCRC), School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Jaecheol Lee
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Epigenome Dynamics Control Research Center (EDCRC), School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Department of Biohealth Regulatory Science, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| |
Collapse
|
4
|
Plummer NW, Smith KG, Jensen P. A knock-in allele of Hand2 expressing Dre recombinase. Genesis 2024; 62:e23601. [PMID: 38703044 PMCID: PMC11088872 DOI: 10.1002/dvg.23601] [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: 02/26/2024] [Revised: 04/05/2024] [Accepted: 04/23/2024] [Indexed: 05/06/2024]
Abstract
HAND2 is a basic helix-loop-helix transcription factor with diverse functions during development. To facilitate the investigation of genetic and functional diversity among Hand2-expressing cells in the mouse, we have generated Hand2Dre, a knock-in allele expressing Dre recombinase. To avoid disrupting Hand2 function, the Dre cDNA is inserted at the 3' end of the Hand2 coding sequence following a viral 2A peptide. Hand2Dre homozygotes can therefore be used in complex crosses to increase the proportion of useful genotypes among offspring. Dre expression in mid-gestation Hand2Dre embryos is indistinguishable from wild-type Hand2 expression, and HandDre efficiently recombines rox target sites in vivo. In combination with existing Cre and Flp mouse lines, Hand2Dre will therefore extend the ability to perform genetic intersectional labeling, fate mapping, and functional manipulation of subpopulations of cells characterized by developmental expression of Hand2.
Collapse
Affiliation(s)
- Nicholas W. Plummer
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina 27709, USA
| | - Kathleen G. Smith
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina 27709, USA
| | - Patricia Jensen
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina 27709, USA
| |
Collapse
|
5
|
Soukup J, Manethova M, Stejskal V, Novakova M, Duskova J, Hornychova H, Hacova M, Staniczkova-Zambo I, Zelinka T, Kosak M, Cesak T, Netuka D, Ryska A, Gabalec F. Hand2 Immunohistochemistry in the Diagnosis of Paragangliomas and Other Neuroendocrine Neoplasms. Endocr Pathol 2024; 35:14-24. [PMID: 38416360 DOI: 10.1007/s12022-024-09803-6] [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: 02/08/2024] [Indexed: 02/29/2024]
Abstract
Hand2 is a core transcription factor responsible for chromaffin cell differentiation. However, its potential utility in surgical pathology has not been studied. Thus, we aimed to investigate its expression in paragangliomas, other neuroendocrine neoplasms (NENs), and additional non-neuroendocrine tumors. We calibrated Hand2 immunohistochemistry on adrenal medulla cells and analyzed H-scores in 46 paragangliomas (PGs), 9 metastatic PGs, 21 cauda equina neuroendocrine tumors (CENETs), 48 neuroendocrine carcinomas (NECs), 8 olfactory neuroblastomas (ONBs), 110 well-differentiated NETs (WDNETs), 10 adrenal cortical carcinomas, 29 adrenal cortical adenomas, 8 melanomas, 41 different carcinomas, and 10 gastrointestinal stromal tumors (GISTs). Both tissue microarrays (TMAs) and whole sections (WSs) were studied. In 171 NENs, previously published data on Phox2B and GATA3 were correlated with Hand2. Hand2 was positive in 98.1% (54/55) PGs, but only rarely in WDNETs (9.6%, 10/104), CENETs (9.5%, 2/21), NECs (4.2%, 2/48), or ONBs (12.5%, 1/8). Any Hand2 positivity was 98.1% sensitive and 91.7% specific for the diagnosis of PG. The Hand2 H-score was significantly higher in primary PGs compared to Hand2-positive WDNETs (median 166.3 vs. 7.5; p < 0.0001). Metastatic PGs were positive in 88.9% (8/9). No Hand2 positivity was observed in any adrenal cortical neoplasm or other non-neuroendocrine tumors, with exception of 8/10 GISTs. Parasympathetic PGs showed a higher Hand2 H-score compared to sympathetic PGs (median H-scores 280 vs. 104, p < 0.0001). Hand2 positivity in NENs serves as a reliable marker of primary and metastatic PG, since other NENs only rarely exhibit limited Hand2 positivity.
Collapse
Affiliation(s)
- Jiri Soukup
- Department of Pathology, Military University Hospital Prague, U Vojenske Nemocnice 1200, Praha 6, 169 02, Prague, Czech Republic.
- The Fingerland Department of Pathology, Charles University, Faculty of Medicine in Hradec Králové and University Hospital Hradec Králové, Sokolska 582, 500 05, Hradec Kralove, Czech Republic.
- Department of Pathology, Charles University, First Faculty of Medicine and General University Hospital in Prague, Studnickova, 2039, 128 00, Nové Mesto, Prague, Czech Republic.
| | - Monika Manethova
- The Fingerland Department of Pathology, Charles University, Faculty of Medicine in Hradec Králové and University Hospital Hradec Králové, Sokolska 582, 500 05, Hradec Kralove, Czech Republic
| | - Vaclav Stejskal
- The Fingerland Department of Pathology, Charles University, Faculty of Medicine in Hradec Králové and University Hospital Hradec Králové, Sokolska 582, 500 05, Hradec Kralove, Czech Republic
| | - Marie Novakova
- Department of Pathology, Military University Hospital Prague, U Vojenske Nemocnice 1200, Praha 6, 169 02, Prague, Czech Republic
| | - Jaroslava Duskova
- Department of Pathology, Charles University, First Faculty of Medicine and General University Hospital in Prague, Studnickova, 2039, 128 00, Nové Mesto, Prague, Czech Republic
| | - Helena Hornychova
- The Fingerland Department of Pathology, Charles University, Faculty of Medicine in Hradec Králové and University Hospital Hradec Králové, Sokolska 582, 500 05, Hradec Kralove, Czech Republic
| | - Maria Hacova
- Department of Pathology, The Regional Hospital Pardubice, Pardubice, Czech Republic
| | - Iva Staniczkova-Zambo
- 1st Department of Pathology, St. Anne's University Hospital and Faculty of Medicine , Masaryk University, Pekarská 664/53, 602 00, Brno-stred, Brno, Czech Republic
| | - Tomas Zelinka
- Centre for Hypertension, 3rd Department of Medicine, Charles University, First Faculty of Medicine and General University Hospital in Prague, Prague, Czech Republic
| | - Mikulas Kosak
- Department of Internal Medicine, First Faculty of Medicine, Charles University and Military University Hospital, Prague, Czech Republic
| | - Tomas Cesak
- Department of Neurosurgery, Charles University, Faculty of Medicine in Hradec Králové and University Hospital Hradec Králové, Sokolska 582, 500 05, Hradec Kralove, Czech Republic
| | - David Netuka
- Department of Neurosurgery and Neurooncology, 1st Medical Faculty, Charles University, Military University Hospital Prague, U Vojenske Nemocnice 1200, Praha 6, 169 02, Prague, Czech Republic
| | - Ales Ryska
- The Fingerland Department of Pathology, Charles University, Faculty of Medicine in Hradec Králové and University Hospital Hradec Králové, Sokolska 582, 500 05, Hradec Kralove, Czech Republic
| | - Filip Gabalec
- 4th Department of Internal Medicine, Charles University, Faculty of Medicine in Hradec Králové and University Hospital Hradec Králové, Sokolska 582, 500 05, Hradec Kralove, Czech Republic
| |
Collapse
|
6
|
Kim S, Koppitch K, Parvez RK, Guo J, Achieng M, Schnell J, Lindström NO, McMahon AP. Comparative single-cell analyses identify shared and divergent features of human and mouse kidney development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.16.540880. [PMID: 37293066 PMCID: PMC10245679 DOI: 10.1101/2023.05.16.540880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Mammalian kidneys maintain fluid homeostasis through the cellular activity of nephrons and the conjoined collecting system. Each epithelial network originates from distinct progenitor cell populations that reciprocally interact during development. To extend our understanding of human and mouse kidney development, we profiled chromatin organization (ATAC-seq) and gene expression (RNA-seq) in developing human and mouse kidneys. Data were analyzed at a species level and then integrated into a common, cross-species multimodal data set. Comparative analysis of cell types and developmental trajectories identified conserved and divergent features of chromatin organization and linked gene activity, revealing species- and cell-type specific regulatory programs. Identification of human-specific enhancer regions linked through GWAS studies to kidney disease highlights the potential of developmental modeling to provide clinical insight.
Collapse
|
7
|
George RM, Firulli BA, Podicheti R, Rusch DB, Mannion BJ, Pennacchio LA, Osterwalder M, Firulli AB. Single cell evaluation of endocardial Hand2 gene regulatory networks reveals HAND2-dependent pathways that impact cardiac morphogenesis. Development 2023; 150:dev201341. [PMID: 36620995 PMCID: PMC10110492 DOI: 10.1242/dev.201341] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 12/26/2022] [Indexed: 01/10/2023]
Abstract
The transcription factor HAND2 plays essential roles during cardiogenesis. Hand2 endocardial deletion (H2CKO) results in tricuspid atresia or double inlet left ventricle with accompanying intraventricular septum defects, hypo-trabeculated ventricles and an increased density of coronary lumens. To understand the regulatory mechanisms of these phenotypes, single cell transcriptome analysis of mouse E11.5 H2CKO hearts was performed revealing a number of disrupted endocardial regulatory pathways. Using HAND2 DNA occupancy data, we identify several HAND2-dependent enhancers, including two endothelial enhancers for the shear-stress master regulator KLF2. A 1.8 kb enhancer located 50 kb upstream of the Klf2 TSS imparts specific endothelial/endocardial expression within the vasculature and endocardium. This enhancer is HAND2-dependent for ventricular endocardium expression but HAND2-independent for Klf2 vascular and valve expression. Deletion of this Klf2 enhancer results in reduced Klf2 expression within ventricular endocardium. These data reveal that HAND2 functions within endocardial gene regulatory networks including shear-stress response.
Collapse
Affiliation(s)
- Rajani M. George
- Herman B Wells Center for Pediatric Research, Departments of Pediatrics, Anatomy and Medical and Molecular Genetics, Indiana Medical School, Indianapolis, IN 46202, USA
| | - Beth A. Firulli
- Herman B Wells Center for Pediatric Research, Departments of Pediatrics, Anatomy and Medical and Molecular Genetics, Indiana Medical School, Indianapolis, IN 46202, USA
| | - Ram Podicheti
- Center for Genomics and Bioinformatics, Indiana University, Bloomington, IN 47405, USA
| | - Douglas B. Rusch
- Center for Genomics and Bioinformatics, Indiana University, Bloomington, IN 47405, USA
| | - Brandon J. Mannion
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
- Comparative Biochemistry Program, University of California, Berkeley, CA 94720, USA
| | - Len A. Pennacchio
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
- Comparative Biochemistry Program, University of California, Berkeley, CA 94720, USA
- US Department of Energy Joint Genome Institute, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Marco Osterwalder
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
- Department for BioMedical Research (DBMR), University of Bern, Bern 3008, Switzerland
- Department of Cardiology, Bern University Hospital, Bern 3010, Switzerland
| | - Anthony B. Firulli
- Herman B Wells Center for Pediatric Research, Departments of Pediatrics, Anatomy and Medical and Molecular Genetics, Indiana Medical School, Indianapolis, IN 46202, USA
| |
Collapse
|
8
|
Parkinson LM, Gillen SL, Woods LM, Chaytor L, Marcos D, Ali FR, Carroll JS, Philpott A. The proneural transcription factor ASCL1 regulates cell proliferation and primes for differentiation in neuroblastoma. Front Cell Dev Biol 2022; 10:942579. [PMID: 36263020 PMCID: PMC9574099 DOI: 10.3389/fcell.2022.942579] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 08/26/2022] [Indexed: 11/27/2022] Open
Abstract
Neuroblastoma is believed to arise from sympathetic neuroblast precursors that fail to engage the neuronal differentiation programme, but instead become locked in a pro-proliferative developmental state. Achaete-scute homolog 1 (ASCL1) is a proneural master regulator of transcription which modulates both proliferation and differentiation of sympathetic neuroblast precursor cells during development, while its expression has been implicated in the maintenance of an oncogenic programme in MYCN-amplified neuroblastoma. However, the role of ASCL1 expression in neuroblastoma is not clear, especially as its levels vary considerably in different neuroblastoma cell lines. Here, we have investigated the role of ASCL1 in maintaining proliferation and controlling differentiation in both MYCN amplified and Anaplastic Lymphoma Kinase (ALK)-driven neuroblastoma cells. Using CRISPR deletion, we generated neuroblastoma cell lines lacking ASCL1 expression, and these grew more slowly than parental cells, indicating that ASCL1 contributes to rapid proliferation of MYCN amplified and non-amplified neuroblastoma cells. Genome-wide analysis after ASCL1 deletion revealed reduced expression of genes associated with neuronal differentiation, while chromatin accessibility at regulatory regions associated with differentiation genes was also attenuated by ASCL1 knock-out. In neuroblastoma, ASCL1 has been described as part of a core regulatory circuit of developmental regulators whose high expression is maintained by mutual cross-activation of a network of super enhancers and is further augmented by the activity of MYC/MYCN. Surprisingly, ASCL1 deletion had little effect on the transcription of CRC gene transcripts in these neuroblastoma cell lines, but the ability of MYC/MYCN and CRC component proteins, PHOX2B and GATA3, to bind to chromatin was compromised. Taken together, our results demonstrate several roles for endogenous ASCL1 in neuroblastoma cells: maintaining a highly proliferative phenotype, regulating DNA binding of the core regulatory circuit genes to chromatin, while also controlling accessibility and transcription of differentiation targets. Thus, we propose a model where ASCL1, a key developmental regulator of sympathetic neurogenesis, plays a pivotal role in maintaining proliferation while simultaneously priming cells for differentiation in neuroblastoma.
Collapse
Affiliation(s)
- Lydia M. Parkinson
- Department of Oncology, University of Cambridge, Cambridge, United Kingdom
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Sarah L. Gillen
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Laura M. Woods
- Department of Oncology, University of Cambridge, Cambridge, United Kingdom
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Lewis Chaytor
- Department of Oncology, University of Cambridge, Cambridge, United Kingdom
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Daniel Marcos
- Department of Oncology, University of Cambridge, Cambridge, United Kingdom
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Fahad R. Ali
- Department of Oncology, University of Cambridge, Cambridge, United Kingdom
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, United Kingdom
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
| | - Jason S. Carroll
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| | - Anna Philpott
- Department of Oncology, University of Cambridge, Cambridge, United Kingdom
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, United Kingdom
| |
Collapse
|
9
|
Kameneva P, Melnikova VI, Kastriti ME, Kurtova A, Kryukov E, Murtazina A, Faure L, Poverennaya I, Artemov AV, Kalinina TS, Kudryashov NV, Bader M, Skoda J, Chlapek P, Curylova L, Sourada L, Neradil J, Tesarova M, Pasqualetti M, Gaspar P, Yakushov VD, Sheftel BI, Zikmund T, Kaiser J, Fried K, Alenina N, Voronezhskaya EE, Adameyko I. Serotonin limits generation of chromaffin cells during adrenal organ development. Nat Commun 2022; 13:2901. [PMID: 35614045 PMCID: PMC9133002 DOI: 10.1038/s41467-022-30438-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 04/23/2022] [Indexed: 11/12/2022] Open
Abstract
Adrenal glands are the major organs releasing catecholamines and regulating our stress response. The mechanisms balancing generation of adrenergic chromaffin cells and protecting against neuroblastoma tumors are still enigmatic. Here we revealed that serotonin (5HT) controls the numbers of chromaffin cells by acting upon their immediate progenitor "bridge" cells via 5-hydroxytryptamine receptor 3A (HTR3A), and the aggressive HTR3Ahigh human neuroblastoma cell lines reduce proliferation in response to HTR3A-specific agonists. In embryos (in vivo), the physiological increase of 5HT caused a prolongation of the cell cycle in "bridge" progenitors leading to a smaller chromaffin population and changing the balance of hormones and behavioral patterns in adulthood. These behavioral effects and smaller adrenals were mirrored in the progeny of pregnant female mice subjected to experimental stress, suggesting a maternal-fetal link that controls developmental adaptations. Finally, these results corresponded to a size-distribution of adrenals found in wild rodents with different coping strategies.
Collapse
Affiliation(s)
- Polina Kameneva
- Department of Neuroimmunology, Center for Brain Research, Medical University Vienna, Vienna, Austria
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - Victoria I Melnikova
- Koltsov Institute of Developmental Biology, Russian Academy of Sciences, Moscow, Russia
| | - Maria Eleni Kastriti
- Department of Neuroimmunology, Center for Brain Research, Medical University Vienna, Vienna, Austria
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - Anastasia Kurtova
- Koltsov Institute of Developmental Biology, Russian Academy of Sciences, Moscow, Russia
| | - Emil Kryukov
- Department of Neuroimmunology, Center for Brain Research, Medical University Vienna, Vienna, Austria
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - Aliia Murtazina
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - Louis Faure
- Department of Neuroimmunology, Center for Brain Research, Medical University Vienna, Vienna, Austria
| | - Irina Poverennaya
- Department of Neuroimmunology, Center for Brain Research, Medical University Vienna, Vienna, Austria
| | - Artem V Artemov
- Department of Neuroimmunology, Center for Brain Research, Medical University Vienna, Vienna, Austria
- National Medical Research Center for Endocrinology, Moscow, Russia
| | - Tatiana S Kalinina
- Federal state budgetary institution "Research Zakusov Institute of Pharmacology" (FSBI "Zakusov Institute of Pharmacology"), Russian Academy of Sciences, Moscow, Russia
| | - Nikita V Kudryashov
- Federal state budgetary institution "Research Zakusov Institute of Pharmacology" (FSBI "Zakusov Institute of Pharmacology"), Russian Academy of Sciences, Moscow, Russia
- Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Michael Bader
- Max-Delbrück Center for Molecular Medicine (MDC), 13125, Berlin-Buch, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Germany
- Charité-Universitätsmedizin Berlin, 10117, Berlin, Germany
- Institute for Biology, University of Lübeck, 23562, Lübeck, Germany
| | - Jan Skoda
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
| | - Petr Chlapek
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
| | - Lucie Curylova
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
| | - Lukas Sourada
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
| | - Jakub Neradil
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
| | - Marketa Tesarova
- Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic
| | - Massimo Pasqualetti
- Unit of Cell and Developmental Biology, Department of Biology, University of Pisa, Pisa, Italy
- Center for Neuroscience and Cognitive Systems @UniTn, Istituto Italiano di Tecnologia, Rovereto, Italy
| | | | - Vasily D Yakushov
- Severtsov Institute of Ecology and Evolution, Russian Academy of Sciences, Moscow, Russia
| | - Boris I Sheftel
- Severtsov Institute of Ecology and Evolution, Russian Academy of Sciences, Moscow, Russia
| | - Tomas Zikmund
- Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic
| | - Jozef Kaiser
- Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic
| | - Kaj Fried
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Natalia Alenina
- Max-Delbrück Center for Molecular Medicine (MDC), 13125, Berlin-Buch, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Germany
| | - Elena E Voronezhskaya
- Koltsov Institute of Developmental Biology, Russian Academy of Sciences, Moscow, Russia.
| | - Igor Adameyko
- Department of Neuroimmunology, Center for Brain Research, Medical University Vienna, Vienna, Austria.
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden.
| |
Collapse
|
10
|
Videira RF, Koop AMC, Ottaviani L, Poels EM, Kocken JMM, Dos Remedios C, Mendes-Ferreira P, Van De Kolk KW, Du Marchie Sarvaas GJ, Lourenço A, Llucià-Valldeperas A, Nascimento DS, de Windt LJ, De Man FS, Falcão-Pires I, Berger RMF, da Costa Martins P. The adult heart requires baseline expression of the transcription factor Hand2 to withstand RV pressure overload. Cardiovasc Res 2021; 118:2688-2702. [PMID: 34550326 PMCID: PMC9491876 DOI: 10.1093/cvr/cvab299] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Indexed: 11/14/2022] Open
Abstract
AIMS Research on the pathophysiology of right ventricular (RV) failure has, in spite of the associated high mortality and morbidity, lagged behind compared to the left ventricle (LV).Previous work from our lab revealed that the embryonic basic helix-loop-helix transcription factor heart and neural crest derivatives expressed-2 (Hand2) is re-expressed in the adult heart and activates a 'fetal gene program' contributing to pathological cardiac remodeling under conditions of LV pressure overload. As such, ablation of cardiac expression of Hand2 conferred protection to cardiac stress and abrogated the maladaptive effects that were observed upon increased expression levels. In this study, we aimed to understand the contribution of Hand2 to RV remodeling in response to pressure overload induced by pulmonary artery banding (PAB). METHODS AND RESULTS In the present study, Hand2F/F and MCM- Hand2F/F mice were treated with tamoxifen (control and knockout, respectively) and subjected to six weeks of RV pressure overload induced by PAB. Echocardiographic- and MRI-derived hemodynamic parameters as well as molecular remodeling were assessed for all experimental groups and compared to sham-operated controls. Six weeks after PAB, levels of Hand2 expression increased in the control banded animals but, as expected, remained absent in the knockout hearts. Despite the dramatic differences in Hand2 expression, pressure overload resulted in impaired cardiac function independently of the genotype. In fact, Hand2 depletion seems to sensitize the RV to pressure overload as these mice develop more hypertrophy and more severe cardiac dysfunction. Higher expression levels of HAND2 were also observed in RV samples of human hearts from patients with pulmonary hypertension. In turn, the LV of RV-pressure overloaded hearts was also dramatically affected as reflected by changes in shape, decreased LV mass and impaired cardiac function. RNA sequencing revealed a distinct set of genes that are dysregulated in the pressure-overloaded RV, compared to the previously described pressure-overloaded LV. CONCLUSIONS Cardiac-specific depletion of Hand2 is associated with severe cardiac dysfunction in conditions of RV pressure overload. While inhibiting Hand2 expression can prevent cardiac dysfunction in conditions of LV pressure overload, the same does not hold true for conditions of RV pressure overload. This study highlights the need to better understand the molecular mechanisms driving pathological remodeling of the RV in contrast to the LV, in order to better diagnose and treat patients with RV or LV failure. TRANSLATIONAL PERSPECTIVE RV failure associated with pulmonary hypertension reduces long-term survival rate to 55% within 3 years, suggesting that 3 years after diagnosis almost half of the patients will die. To revert these numbers an adequate RV-specific and, therefore, more efficient treatment is needed. Our work suggests that current therapies and potential mechanisms underlying LV failure may not be suitable for RV failure. While Hand2 deletion is favorable in LV response to stress, it is particularly detrimental in the RV under similar conditions, and thus, highlighting potential severe consequences of not differentiating therapeutic targets or treatment for RV or LV failure.
Collapse
Affiliation(s)
- R F Videira
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, Netherlands.,Department of Molecular Genetics, Faculty of Sciences and Engineering, Maastricht University, Maastricht, The Netherlands.,Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, University of Porto, Porto, Portuga
| | - A M C Koop
- University of Groningen, University Medical Center Groningen, Department of Pediatrics, Center for Congenital Heart Diseases, Groningen, Netherlands
| | - L Ottaviani
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, Netherlands.,Department of Molecular Genetics, Faculty of Sciences and Engineering, Maastricht University, Maastricht, The Netherlands
| | - E M Poels
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, Netherlands
| | - J M M Kocken
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, Netherlands.,Department of Molecular Genetics, Faculty of Sciences and Engineering, Maastricht University, Maastricht, The Netherlands
| | - C Dos Remedios
- University of Sidney, Sidney, and Victor Chang Cardiac Research Institute, Darlinghurst, Australia
| | - P Mendes-Ferreira
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, University of Porto, Porto, Portuga
| | - K W Van De Kolk
- University Medical Center Groningen, The Central Animal Facility, Groningen, Netherlands.,University Medical Center Groningen, Gronsai (Groningen Small Animal Imaging Facility), Groningen, Netherlands
| | - G J Du Marchie Sarvaas
- University of Groningen, University Medical Center Groningen, Department of Pediatrics, Center for Congenital Heart Diseases, Groningen, Netherlands
| | - A Lourenço
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, University of Porto, Porto, Portuga
| | - A Llucià-Valldeperas
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Pulmonary Medicine, PHEniX laboratory, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - D S Nascimento
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal.,ICBAS-Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| | - L J de Windt
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, Netherlands.,Department of Molecular Genetics, Faculty of Sciences and Engineering, Maastricht University, Maastricht, The Netherlands
| | - F S De Man
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Pulmonary Medicine, PHEniX laboratory, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - I Falcão-Pires
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, University of Porto, Porto, Portuga
| | - R M F Berger
- University of Groningen, University Medical Center Groningen, Department of Pediatrics, Center for Congenital Heart Diseases, Groningen, Netherlands
| | - Paula da Costa Martins
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, Netherlands.,Department of Molecular Genetics, Faculty of Sciences and Engineering, Maastricht University, Maastricht, The Netherlands.,Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, University of Porto, Porto, Portuga
| |
Collapse
|
11
|
Kameda Y. Comparative morphological and molecular studies on the oxygen-chemoreceptive cells in the carotid body and fish gills. Cell Tissue Res 2021; 384:255-273. [PMID: 33852077 DOI: 10.1007/s00441-021-03421-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 01/20/2021] [Indexed: 11/30/2022]
Abstract
Oxygen-chemoreceptive cells play critical roles for the respiration control. This review summarizes the chemoreceptive cells in the carotid body and fish gills from a morphological and molecular perspective. The cells synthesize and secrete biogenic amines, neuropeptides, and neuroproteins and also express many signaling molecules and transcription factors. In mammals, birds, reptiles, and amphibians, the carotid body primordium is consistently formed in the wall of the third arch artery which gives rise to the common carotid artery and the basal portion of the internal carotid artery. Consequently, the carotid body is located in the carotid bifurcation region, except birds in which the organ is situated at the lateral side of the common carotid artery. The carotid body receives branches of the cranial nerves IX and/or X dependent on the location of the organ. The glomus cell progenitors in mammals and birds are derived from the neighboring ganglion, i.e., the superior cervical sympathetic ganglion and the nodose ganglion, respectively, and immigrate into the carotid body primordium, constituting a solid cell cluster. In other animal species, the glomus cells are dispersed singly or forming small cell groups in intervascular stroma of the carotid body. In fishes, the neuroepithelial cells, corresponding to the glomus cells, are distributed in the gill filaments and lamellae. All oxygen-chemoreceptive cells sensitively respond to acute or chronic hypoxia, exhibiting degranulation, hypertrophy, hyperplasia, and upregulated expression of many genes.
Collapse
Affiliation(s)
- Yoko Kameda
- Department of Anatomy, Kitasato University School of Medicine, Sagamihara, Kanagawa, 252-0374, Japan.
| |
Collapse
|
12
|
Vincentz JW, Firulli BA, Toolan KP, Osterwalder M, Pennacchio LA, Firulli AB. HAND transcription factors cooperatively specify the aorta and pulmonary trunk. Dev Biol 2021; 476:1-10. [PMID: 33757801 DOI: 10.1016/j.ydbio.2021.03.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 03/11/2021] [Accepted: 03/15/2021] [Indexed: 01/11/2023]
Abstract
Congenital heart defects (CHDs) affecting the cardiac outflow tract (OFT) constitute a significant cause of morbidity and mortality. The OFT develops from migratory cell populations which include the cardiac neural crest cells (cNCCs) and secondary heart field (SHF) derived myocardium and endocardium. The related transcription factors HAND1 and HAND2 have been implicated in human CHDs involving the OFT. Although Hand1 is expressed within the OFT, Hand1 NCC-specific conditional knockout mice (H1CKOs) are viable. Here we show that these H1CKOs present a low penetrance of OFT phenotypes, whereas SHF-specific Hand1 ablation does not reveal any cardiac phenotypes. Further, HAND1 and HAND2 appear functionally redundant within the cNCCs, as a reduction/ablation of Hand2 on an NCC-specific H1CKO background causes pronounced OFT defects. Double conditional Hand1 and Hand2 NCC knockouts exhibit persistent truncus arteriosus (PTA) with 100% penetrance. NCC lineage-tracing and Sema3c in situ mRNA expression reveal that Sema3c-expressing cells are mis-localized, resulting in a malformed septal bridge within the OFTs of H1CKO;H2CKO embryos. Interestingly, Hand1 and Hand2 also genetically interact within the SHF, as SHF H1CKOs on a heterozygous Hand2 background exhibit Ventricular Septal Defects (VSDs) with incomplete penetrance. Previously, we identified a BMP, HAND2, and GATA-dependent Hand1 OFT enhancer sufficient to drive reporter gene expression within the nascent OFT and aorta. Using these transcription inputs as a probe, we identify a novel Hand2 OFT enhancer, suggesting that a conserved BMP-GATA dependent mechanism transcriptionally regulates both HAND factors. These findings support the hypothesis that HAND factors interpret BMP signaling within the cNCCs to cooperatively coordinate OFT morphogenesis.
Collapse
Affiliation(s)
- Joshua W Vincentz
- Herman B Wells Center for Pediatric Research Department of Pediatrics, Anatomy, Biochemistry, and Medical and Molecular Genetics, Indiana University School of Medicine, 1044 W. Walnut St., Indianapolis, IN, 46202-5225, USA.
| | - Beth A Firulli
- Herman B Wells Center for Pediatric Research Department of Pediatrics, Anatomy, Biochemistry, and Medical and Molecular Genetics, Indiana University School of Medicine, 1044 W. Walnut St., Indianapolis, IN, 46202-5225, USA
| | - Kevin P Toolan
- Herman B Wells Center for Pediatric Research Department of Pediatrics, Anatomy, Biochemistry, and Medical and Molecular Genetics, Indiana University School of Medicine, 1044 W. Walnut St., Indianapolis, IN, 46202-5225, USA
| | - Marco Osterwalder
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA; Department for BioMedical Research (DBMR), University of Bern, Murtenstrasse 35, 3008, Bern, Switzerland
| | - Len A Pennacchio
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA; U.S. Department of Energy Joint Genome Institute, Berkeley, CA, 94720, USA; Comparative Biochemistry Program, University of California, Berkeley, CA, 94720, USA
| | - Anthony B Firulli
- Herman B Wells Center for Pediatric Research Department of Pediatrics, Anatomy, Biochemistry, and Medical and Molecular Genetics, Indiana University School of Medicine, 1044 W. Walnut St., Indianapolis, IN, 46202-5225, USA.
| |
Collapse
|
13
|
Fedele L, Brand T. The Intrinsic Cardiac Nervous System and Its Role in Cardiac Pacemaking and Conduction. J Cardiovasc Dev Dis 2020; 7:jcdd7040054. [PMID: 33255284 PMCID: PMC7712215 DOI: 10.3390/jcdd7040054] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 11/19/2020] [Accepted: 11/20/2020] [Indexed: 12/11/2022] Open
Abstract
The cardiac autonomic nervous system (CANS) plays a key role for the regulation of cardiac activity with its dysregulation being involved in various heart diseases, such as cardiac arrhythmias. The CANS comprises the extrinsic and intrinsic innervation of the heart. The intrinsic cardiac nervous system (ICNS) includes the network of the intracardiac ganglia and interconnecting neurons. The cardiac ganglia contribute to the tight modulation of cardiac electrophysiology, working as a local hub integrating the inputs of the extrinsic innervation and the ICNS. A better understanding of the role of the ICNS for the modulation of the cardiac conduction system will be crucial for targeted therapies of various arrhythmias. We describe the embryonic development, anatomy, and physiology of the ICNS. By correlating the topography of the intracardiac neurons with what is known regarding their biophysical and neurochemical properties, we outline their physiological role in the control of pacemaker activity of the sinoatrial and atrioventricular nodes. We conclude by highlighting cardiac disorders with a putative involvement of the ICNS and outline open questions that need to be addressed in order to better understand the physiology and pathophysiology of the ICNS.
Collapse
Affiliation(s)
- Laura Fedele
- Correspondence: (L.F.); (T.B.); Tel.: +44-(0)-207-594-6531 (L.F.); +44-(0)-207-594-8744 (T.B.)
| | - Thomas Brand
- Correspondence: (L.F.); (T.B.); Tel.: +44-(0)-207-594-6531 (L.F.); +44-(0)-207-594-8744 (T.B.)
| |
Collapse
|
14
|
Elliott KH, Chen X, Salomone J, Chaturvedi P, Schultz PA, Balchand SK, Servetas JD, Zuniga A, Zeller R, Gebelein B, Weirauch MT, Peterson KA, Brugmann SA. Gli3 utilizes Hand2 to synergistically regulate tissue-specific transcriptional networks. eLife 2020; 9:e56450. [PMID: 33006313 PMCID: PMC7556880 DOI: 10.7554/elife.56450] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 10/01/2020] [Indexed: 12/17/2022] Open
Abstract
Despite a common understanding that Gli TFs are utilized to convey a Hh morphogen gradient, genetic analyses suggest craniofacial development does not completely fit this paradigm. Using the mouse model (Mus musculus), we demonstrated that rather than being driven by a Hh threshold, robust Gli3 transcriptional activity during skeletal and glossal development required interaction with the basic helix-loop-helix TF Hand2. Not only did genetic and expression data support a co-factorial relationship, but genomic analysis revealed that Gli3 and Hand2 were enriched at regulatory elements for genes essential for mandibular patterning and development. Interestingly, motif analysis at sites co-occupied by Gli3 and Hand2 uncovered mandibular-specific, low-affinity, 'divergent' Gli-binding motifs (dGBMs). Functional validation revealed these dGBMs conveyed synergistic activation of Gli targets essential for mandibular patterning and development. In summary, this work elucidates a novel, sequence-dependent mechanism for Gli transcriptional activity within the craniofacial complex that is independent of a graded Hh signal.
Collapse
Affiliation(s)
- Kelsey H Elliott
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical CenterCincinnatiUnited States
- Division of Plastic Surgery, Department of Surgery, Cincinnati Children’s Hospital Medical CenterCincinnatiUnited States
- Graduate Program in Molecular and Developmental Biology, Cincinnati Children's Hospital Research FoundationCincinnatiUnited States
| | - Xiaoting Chen
- Center for Autoimmune Genomics and Etiology, Department of Pediatrics, Cincinnati Children’s Hospital Medical CenterCincinnatiUnited States
| | - Joseph Salomone
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical CenterCincinnatiUnited States
- Graduate Program in Molecular and Developmental Biology, Cincinnati Children's Hospital Research FoundationCincinnatiUnited States
- Medical-Scientist Training Program, University of Cincinnati College of MedicineCincinnatiUnited States
| | - Praneet Chaturvedi
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical CenterCincinnatiUnited States
| | - Preston A Schultz
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical CenterCincinnatiUnited States
- Division of Plastic Surgery, Department of Surgery, Cincinnati Children’s Hospital Medical CenterCincinnatiUnited States
| | - Sai K Balchand
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical CenterCincinnatiUnited States
- Division of Plastic Surgery, Department of Surgery, Cincinnati Children’s Hospital Medical CenterCincinnatiUnited States
| | | | - Aimée Zuniga
- Developmental Genetics, Department of Biomedicine, University of BaselBaselSwitzerland
| | - Rolf Zeller
- Developmental Genetics, Department of Biomedicine, University of BaselBaselSwitzerland
| | - Brian Gebelein
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical CenterCincinnatiUnited States
| | - Matthew T Weirauch
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical CenterCincinnatiUnited States
- Center for Autoimmune Genomics and Etiology, Department of Pediatrics, Cincinnati Children’s Hospital Medical CenterCincinnatiUnited States
| | | | - Samantha A Brugmann
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical CenterCincinnatiUnited States
- Division of Plastic Surgery, Department of Surgery, Cincinnati Children’s Hospital Medical CenterCincinnatiUnited States
- Shriners Children’s HospitalCincinnatiUnited States
| |
Collapse
|
15
|
The diversity of neuronal phenotypes in rodent and human autonomic ganglia. Cell Tissue Res 2020; 382:201-231. [PMID: 32930881 PMCID: PMC7584561 DOI: 10.1007/s00441-020-03279-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 08/10/2020] [Indexed: 12/29/2022]
Abstract
Selective sympathetic and parasympathetic pathways that act on target organs represent the terminal actors in the neurobiology of homeostasis and often become compromised during a range of neurodegenerative and traumatic disorders. Here, we delineate several neurotransmitter and neuromodulator phenotypes found in diverse parasympathetic and sympathetic ganglia in humans and rodent species. The comparative approach reveals evolutionarily conserved and non-conserved phenotypic marker constellations. A developmental analysis examining the acquisition of selected neurotransmitter properties has provided a detailed, but still incomplete, understanding of the origins of a set of noradrenergic and cholinergic sympathetic neuron populations, found in the cervical and trunk region. A corresponding analysis examining cholinergic and nitrergic parasympathetic neurons in the head, and a range of pelvic neuron populations, with noradrenergic, cholinergic, nitrergic, and mixed transmitter phenotypes, remains open. Of particular interest are the molecular mechanisms and nuclear processes that are responsible for the correlated expression of the various genes required to achieve the noradrenergic phenotype, the segregation of cholinergic locus gene expression, and the regulation of genes that are necessary to generate a nitrergic phenotype. Unraveling the neuron population-specific expression of adhesion molecules, which are involved in axonal outgrowth, pathway selection, and synaptic organization, will advance the study of target-selective autonomic pathway generation.
Collapse
|
16
|
Kameda Y. Molecular and cellular mechanisms of the organogenesis and development of the mammalian carotid body. Dev Dyn 2019; 249:592-609. [DOI: 10.1002/dvdy.144] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 12/08/2019] [Accepted: 12/08/2019] [Indexed: 12/16/2022] Open
Affiliation(s)
- Yoko Kameda
- Department of AnatomyKitasato University School of Medicine Sagamihara Japan
| |
Collapse
|
17
|
MicroRNA-92b-3p suppresses angiotensin II-induced cardiomyocyte hypertrophy via targeting HAND2. Life Sci 2019; 232:116635. [PMID: 31283925 DOI: 10.1016/j.lfs.2019.116635] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 06/29/2019] [Accepted: 07/04/2019] [Indexed: 11/22/2022]
Abstract
AIMS The pathological cardiac hypertrophy will develop into heart failure, which has no effective treatment currently. Previous studies have proved that microRNAs (miRNAs) participate in the development of cardiac hypertrophy and regulate the pathological progress. In this study, we want to investigate the role of microRNA-92b-3p (miR-92b-3p) in cardiomyocyte hypertrophy and the mechanisms involved. MATERIALS AND METHODS Neonatal mouse ventricular cells (NMVCs) were isolated from the hearts of 1-3-d-old newborn C57BL6 mice. The isolated NMVCs were induced hypertrophic phenotype by Angiotensin-II (Ang-II) and the cell size was examined by FITC-phalloidin staining assay. The expression of miR-92b-3p was determined by quantitative real-time PCR (qRT-qPCR). MRNA and protein level of β-MHC, ACTA1 and HAND2 in NMVCs transfected with miR-92b-3p mimic and inhibition were assessed by RT-qPCR assay and western blot assay, respectively. Dual luciferase assay was used to verify the interaction between miR-92b-3p and the 3'-untranslated region (UTR) of HAND2 gene. KEY FINDINGS MiR-92b-3p and HAND2 were significantly increased in Ang-II-induced NMVCs. Overexpression of miR-92b-3p can ameliorate Ang-II-induced cardiomyocyte hypertrophy. MiR-92b-3p negatively regulated HAND2 expression at the transcriptional level. Both miR-92b-3p mimic and HAND2 siRNA could efficiently inhibit Ang-II-induced hypertrophy in mouse cardiomyocytes. SIGNIFICANCE MiR-92b-3p inhibits Ang-II-induced cardiomyocyte hypertrophy via targeting HAND2.
Collapse
|
18
|
Han X, Zhang J, Liu Y, Fan X, Ai S, Luo Y, Li X, Jin H, Luo S, Zheng H, Yue Y, Chang Z, Yang Z, Tang F, He A, Shen X. The lncRNA Hand2os1/ Uph locus orchestrates heart development through regulation of precise expression of Hand2. Development 2019; 146:146/13/dev176198. [PMID: 31273086 DOI: 10.1242/dev.176198] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 05/09/2019] [Indexed: 12/30/2022]
Abstract
Exploration and dissection of potential actions and effects of long noncoding RNA (lncRNA) in animals remain challenging. Here, using multiple knockout mouse models and single cell RNA sequencing, we demonstrate that the divergent lncRNA Hand2os1/Uph has a key complex modulatory effect on the expression of its neighboring gene HAND2 and subsequently on heart development and function. Short deletion of the Hand2os1 promoter in mouse diminishes Hand2os1 transcription to ∼8-32%, but fails to affect HAND2 expression and yields no discernable heart phenotypes. Interestingly, full-length deletion of Hand2os1 in mouse causes moderate yet prevalent upregulation of HAND2 in hundreds of cardiac cells, leading to profound biological consequences, including dysregulated cardiac gene programs, congenital heart defects and perinatal lethality. We propose that the Hand2os1 locus dampens HAND2 expression to restrain cardiomyocyte proliferation, thereby orchestrating a balanced development of cardiac cell lineages. This study highlights the regulatory complexity of the lncRNA Hand2os1 on HAND2 expression, emphasizing the need for complementary genetic and single cell approaches to delineate the function and primary molecular effects of an lncRNA in animals.
Collapse
Affiliation(s)
- Xue Han
- Tsinghua Center for Life Sciences, School of Medicine, and School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Jiejie Zhang
- Peking Center for Life Sciences, Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing 100871, China
| | - Yaxi Liu
- Peking Center for Life Sciences, Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing 100871, China
| | - Xiaoying Fan
- Beijing Advanced Innovation Center for Genomics, College of Life Sciences, Peking University, Beijing 100871, China
| | - Shanshan Ai
- Peking Center for Life Sciences, Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing 100871, China
| | - Yingjie Luo
- Peking Center for Life Sciences, Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing 100871, China
| | - Xin Li
- Peking Center for Life Sciences, Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing 100871, China
| | - Hengwei Jin
- Model Animal Research Center, Nanjing University, Nanjing 210061, China
| | - Sai Luo
- Tsinghua Center for Life Sciences, School of Medicine, and School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Hui Zheng
- Tsinghua Center for Life Sciences, School of Medicine, and School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yanzhu Yue
- Peking Center for Life Sciences, Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing 100871, China
| | - Zai Chang
- Tsinghua Center for Life Sciences, School of Medicine, and School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Zhongzhou Yang
- Model Animal Research Center, Nanjing University, Nanjing 210061, China
| | - Fuchou Tang
- Beijing Advanced Innovation Center for Genomics, College of Life Sciences, Peking University, Beijing 100871, China
| | - Aibin He
- Peking Center for Life Sciences, Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing 100871, China
| | - Xiaohua Shen
- Tsinghua Center for Life Sciences, School of Medicine, and School of Life Sciences, Tsinghua University, Beijing 100084, China
| |
Collapse
|
19
|
Reynolds K, Kumari P, Sepulveda Rincon L, Gu R, Ji Y, Kumar S, Zhou CJ. Wnt signaling in orofacial clefts: crosstalk, pathogenesis and models. Dis Model Mech 2019; 12:12/2/dmm037051. [PMID: 30760477 PMCID: PMC6398499 DOI: 10.1242/dmm.037051] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Diverse signaling cues and attendant proteins work together during organogenesis, including craniofacial development. Lip and palate formation starts as early as the fourth week of gestation in humans or embryonic day 9.5 in mice. Disruptions in these early events may cause serious consequences, such as orofacial clefts, mainly cleft lip and/or cleft palate. Morphogenetic Wnt signaling, along with other signaling pathways and transcription regulation mechanisms, plays crucial roles during embryonic development, yet the signaling mechanisms and interactions in lip and palate formation and fusion remain poorly understood. Various Wnt signaling and related genes have been associated with orofacial clefts. This Review discusses the role of Wnt signaling and its crosstalk with cell adhesion molecules, transcription factors, epigenetic regulators and other morphogenetic signaling pathways, including the Bmp, Fgf, Tgfβ, Shh and retinoic acid pathways, in orofacial clefts in humans and animal models, which may provide a better understanding of these disorders and could be applied towards prevention and treatments.
Collapse
Affiliation(s)
- Kurt Reynolds
- Department of Biochemistry and Molecular Medicine, University of California at Davis, School of Medicine, Sacramento, CA 95817, USA.,Institute for Pediatric Regenerative Medicine of Shriners Hospitals for Children, University of California at Davis, School of Medicine, Sacramento, CA 95817, USA.,Biochemistry, Molecular, Cellular, and Developmental Biology (BMCDB) Graduate Group, University of California, Davis, CA 95616, USA
| | - Priyanka Kumari
- Department of Biochemistry and Molecular Medicine, University of California at Davis, School of Medicine, Sacramento, CA 95817, USA.,Institute for Pediatric Regenerative Medicine of Shriners Hospitals for Children, University of California at Davis, School of Medicine, Sacramento, CA 95817, USA
| | - Lessly Sepulveda Rincon
- Department of Biochemistry and Molecular Medicine, University of California at Davis, School of Medicine, Sacramento, CA 95817, USA.,Institute for Pediatric Regenerative Medicine of Shriners Hospitals for Children, University of California at Davis, School of Medicine, Sacramento, CA 95817, USA
| | - Ran Gu
- Department of Biochemistry and Molecular Medicine, University of California at Davis, School of Medicine, Sacramento, CA 95817, USA.,Institute for Pediatric Regenerative Medicine of Shriners Hospitals for Children, University of California at Davis, School of Medicine, Sacramento, CA 95817, USA
| | - Yu Ji
- Department of Biochemistry and Molecular Medicine, University of California at Davis, School of Medicine, Sacramento, CA 95817, USA.,Institute for Pediatric Regenerative Medicine of Shriners Hospitals for Children, University of California at Davis, School of Medicine, Sacramento, CA 95817, USA.,Biochemistry, Molecular, Cellular, and Developmental Biology (BMCDB) Graduate Group, University of California, Davis, CA 95616, USA
| | - Santosh Kumar
- Department of Biochemistry and Molecular Medicine, University of California at Davis, School of Medicine, Sacramento, CA 95817, USA.,Institute for Pediatric Regenerative Medicine of Shriners Hospitals for Children, University of California at Davis, School of Medicine, Sacramento, CA 95817, USA
| | - Chengji J Zhou
- Department of Biochemistry and Molecular Medicine, University of California at Davis, School of Medicine, Sacramento, CA 95817, USA .,Institute for Pediatric Regenerative Medicine of Shriners Hospitals for Children, University of California at Davis, School of Medicine, Sacramento, CA 95817, USA.,Biochemistry, Molecular, Cellular, and Developmental Biology (BMCDB) Graduate Group, University of California, Davis, CA 95616, USA
| |
Collapse
|
20
|
Ernsberger U, Rohrer H. Sympathetic tales: subdivisons of the autonomic nervous system and the impact of developmental studies. Neural Dev 2018; 13:20. [PMID: 30213267 PMCID: PMC6137933 DOI: 10.1186/s13064-018-0117-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 08/12/2018] [Indexed: 02/06/2023] Open
Abstract
Remarkable progress in a range of biomedical disciplines has promoted the understanding of the cellular components of the autonomic nervous system and their differentiation during development to a critical level. Characterization of the gene expression fingerprints of individual neurons and identification of the key regulators of autonomic neuron differentiation enables us to comprehend the development of different sets of autonomic neurons. Their individual functional properties emerge as a consequence of differential gene expression initiated by the action of specific developmental regulators. In this review, we delineate the anatomical and physiological observations that led to the subdivision into sympathetic and parasympathetic domains and analyze how the recent molecular insights melt into and challenge the classical description of the autonomic nervous system.
Collapse
Affiliation(s)
- Uwe Ernsberger
- Institute for Clinical Neuroanatomy, Goethe University, Theodor-Stern-Kai 7, 60590 Frankfurt/Main, Germany
| | - Hermann Rohrer
- Institute for Clinical Neuroanatomy, Goethe University, Theodor-Stern-Kai 7, 60590 Frankfurt/Main, Germany
| |
Collapse
|
21
|
Barald KF, Shen YC, Bianchi LM. Chemokines and cytokines on the neuroimmunoaxis: Inner ear neurotrophic cytokines in development and disease. Prospects for repair? Exp Neurol 2018; 301:92-99. [DOI: 10.1016/j.expneurol.2017.10.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 09/18/2017] [Accepted: 10/12/2017] [Indexed: 01/22/2023]
|
22
|
Urraca N, Hope K, Victor AK, Belgard TG, Memon R, Goorha S, Valdez C, Tran QT, Sanchez S, Ramirez J, Donaldson M, Bridges D, Reiter LT. Significant transcriptional changes in 15q duplication but not Angelman syndrome deletion stem cell-derived neurons. Mol Autism 2018; 9:6. [PMID: 29423132 PMCID: PMC5787244 DOI: 10.1186/s13229-018-0191-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 01/15/2018] [Indexed: 01/09/2023] Open
Abstract
Background The inability to analyze gene expression in living neurons from Angelman (AS) and Duplication 15q (Dup15q) syndrome subjects has limited our understanding of these disorders at the molecular level. Method Here, we use dental pulp stem cells (DPSC) from AS deletion, 15q Duplication, and neurotypical control subjects for whole transcriptome analysis. We identified 20 genes unique to AS neurons, 120 genes unique to 15q duplication, and 3 shared transcripts that were differentially expressed in DPSC neurons vs controls. Results Copy number correlated with gene expression for most genes across the 15q11.2-q13.1 critical region. Two thirds of the genes differentially expressed in 15q duplication neurons were downregulated compared to controls including several transcription factors, while in AS differential expression was restricted primarily to the 15q region. Here, we show significant downregulation of the transcription factors FOXO1 and HAND2 in neurons from 15q duplication, but not AS deletion subjects suggesting that disruptions in transcriptional regulation may be a driving factor in the autism phenotype in Dup15q syndrome. Downstream analysis revealed downregulation of the ASD associated genes EHPB2 and RORA, both genes with FOXO1 binding sites. Genes upregulated in either Dup15q cortex or idiopathic ASD cortex both overlapped significantly with the most upregulated genes in Dup15q DPSC-derived neurons. Conclusions Finding a significant increase in both HERC2 and UBE3A in Dup15q neurons and significant decrease in these two genes in AS deletion neurons may explain differences between AS deletion class and UBE3A specific classes of AS mutation where HERC2 is expressed at normal levels. Also, we identified an enrichment for FOXO1-regulated transcripts in Dup15q neurons including ASD-associated genes EHPB2 and RORA indicating a possible connection between this syndromic form of ASD and idiopathic cases.
Collapse
Affiliation(s)
- Nora Urraca
- Department of Neurology, The University of Tennessee Health Science Center, 855 Monroe Ave., Link 415, Memphis, TN 38163 USA
| | - Kevin Hope
- Department of Neurology, The University of Tennessee Health Science Center, 855 Monroe Ave., Link 415, Memphis, TN 38163 USA
- IPBS Program, The University of Tennessee Health Science Center, Memphis, TN 38163 USA
| | - A. Kaitlyn Victor
- Department of Neurology, The University of Tennessee Health Science Center, 855 Monroe Ave., Link 415, Memphis, TN 38163 USA
- IPBS Program, The University of Tennessee Health Science Center, Memphis, TN 38163 USA
| | - T. Grant Belgard
- MRC Functional Genomics Unit, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, OX1 3QX UK
| | - Rawaha Memon
- Department of Pediatric Dentistry, The University of Tennessee Health Science Center, Memphis, TN 38163 USA
| | - Sarita Goorha
- Department of Neurology, The University of Tennessee Health Science Center, 855 Monroe Ave., Link 415, Memphis, TN 38163 USA
| | - Colleen Valdez
- Department of Neurology, The University of Tennessee Health Science Center, 855 Monroe Ave., Link 415, Memphis, TN 38163 USA
| | - Quynh T. Tran
- Department of Preventive Medicine, The University of Tennessee Health Science Center, Memphis, TN 38163 USA
| | - Silvia Sanchez
- Instituto Nacional de Pediatria, 04530 Mexico City, Mexico
| | - Juanma Ramirez
- Department of Biochemistry and Molecular Biology, University of Basque Country, Bilbao, Spain
| | - Martin Donaldson
- Department of Pediatric Dentistry, The University of Tennessee Health Science Center, Memphis, TN 38163 USA
| | - Dave Bridges
- Department of Nutritional Sciences, University of Michigan School of Public Health, 1415 Washington Heights, Ann Arbor, MI 48109 USA
| | - Lawrence T. Reiter
- Department of Neurology, The University of Tennessee Health Science Center, 855 Monroe Ave., Link 415, Memphis, TN 38163 USA
- Department of Pediatrics, The University of Tennessee Health Science Center, Memphis, TN 38163 USA
| |
Collapse
|
23
|
Abstract
Fell et al. deleted KIF1Bβ in the mouse sympathetic nervous system and observed impaired sympathetic nervous function and misexpression of genes required for sympathoadrenal lineage differentiation. They discovered that KIF1Bβ is required for NGF-dependent neuronal differentiation through anterograde transport of the NGF receptor TRKA. We recently identified pathogenic KIF1Bβ mutations in sympathetic nervous system malignancies that are defective in developmental apoptosis. Here we deleted KIF1Bβ in the mouse sympathetic nervous system and observed impaired sympathetic nervous function and misexpression of genes required for sympathoadrenal lineage differentiation. We discovered that KIF1Bβ is required for nerve growth factor (NGF)-dependent neuronal differentiation through anterograde transport of the NGF receptor TRKA. Moreover, pathogenic KIF1Bβ mutations identified in neuroblastoma impair TRKA transport. Expression of neuronal differentiation markers is ablated in both KIF1Bβ-deficient mouse neuroblasts and human neuroblastomas that lack KIF1Bβ. Transcriptomic analyses show that unfavorable neuroblastomas resemble mouse sympathetic neuroblasts lacking KIF1Bβ independent of MYCN amplification and the loss of genes neighboring KIF1B on chromosome 1p36. Thus, defective precursor cell differentiation, a common trait of aggressive childhood malignancies, is a pathogenic effect of KIF1Bβ loss in neuroblastomas. Furthermore, neuropathy-associated KIF1Bβ mutations impede cargo transport, providing a direct link between neuroblastomas and neurodegeneration.
Collapse
|
24
|
Chan WH, Anderson CR, Gonsalvez DG. From proliferation to target innervation: signaling molecules that direct sympathetic nervous system development. Cell Tissue Res 2017; 372:171-193. [PMID: 28971249 DOI: 10.1007/s00441-017-2693-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 08/30/2017] [Indexed: 02/07/2023]
Abstract
The sympathetic division of the autonomic nervous system includes a variety of cells including neurons, endocrine cells and glial cells. A recent study (Furlan et al. 2017) has revised thinking about the developmental origin of these cells. It now appears that sympathetic neurons and chromaffin cells of the adrenal medulla do not have an immediate common ancestor in the form a "sympathoadrenal cell", as has been long believed. Instead, chromaffin cells arise from Schwann cell precursors. This review integrates the new findings with the expanding body of knowledge on the signalling pathways and transcription factors that regulate the origin of cells of the sympathetic division of the autonomic nervous system.
Collapse
Affiliation(s)
- W H Chan
- Department of Anatomy and Neuroscience, School of Biomedical Sciences, The University of Melbourne, Parkville, 3010, Australia
| | - C R Anderson
- Department of Anatomy and Neuroscience, School of Biomedical Sciences, The University of Melbourne, Parkville, 3010, Australia
| | - David G Gonsalvez
- Department of Anatomy and Neuroscience, School of Biomedical Sciences, The University of Melbourne, Parkville, 3010, Australia.
| |
Collapse
|
25
|
Moore SW. Advances in understanding functional variations in the Hirschsprung disease spectrum (variant Hirschsprung disease). Pediatr Surg Int 2017; 33:285-298. [PMID: 27988850 DOI: 10.1007/s00383-016-4038-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/05/2016] [Indexed: 12/11/2022]
Abstract
Hirschsprung disease (HSCR) is a fairly well understood congenital, genetically based functional obstruction due to the congenital absence of ganglion cells in the distal bowel. However, although over 90% of Hirschsprung cases conform to the normally accepted histological diagnostic criteria, it has become increasingly clear that in addition to HSCR, there is a group of functional disturbances relating to a number of other congenital neurodysplastic conditions causing some degree of gastrointestinal tract malfunction. Although these represent a variety of possibly separate conditions of the enteric nervous system, this spectrum it would appear to be also influenced by similar developmental processes. The term "variant Hirschsprung" is commonly used to describe these conditions, but ganglion cells are mostly present if abnormal in number and distribution. These conditions are a problem group being amongst the most difficult to diagnose and treat with possible practical and legal consequences. The problem appears to be possibly one of definition which has proven difficult in the relative paucity of normal values, especially when correlated to age and gestation. It is the purpose of this paper to review the current position on these conditions and to explore possible shared common pathogenetic and genetic mechanisms. This article explores those conditions where a similar pathogenetic mechanisms to HSCR can be demonstrated (e.g. hypoganglionosis) as well as other neural features, which appear to represent separate conditions possibly linked to certain syndromes.
Collapse
Affiliation(s)
- S W Moore
- Division of Paediatric Surgery, Faculty of Medicine, University of Stellenbosch, P.O. Box 19063, Tygerberg, 7505, South Africa.
| |
Collapse
|
26
|
Végh AMD, Duim SN, Smits AM, Poelmann RE, Ten Harkel ADJ, DeRuiter MC, Goumans MJ, Jongbloed MRM. Part and Parcel of the Cardiac Autonomic Nerve System: Unravelling Its Cellular Building Blocks during Development. J Cardiovasc Dev Dis 2016; 3:jcdd3030028. [PMID: 29367572 PMCID: PMC5715672 DOI: 10.3390/jcdd3030028] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Revised: 09/05/2016] [Accepted: 09/07/2016] [Indexed: 02/06/2023] Open
Abstract
The autonomic nervous system (cANS) is essential for proper heart function, and complications such as heart failure, arrhythmias and even sudden cardiac death are associated with an altered cANS function. A changed innervation state may underlie (part of) the atrial and ventricular arrhythmias observed after myocardial infarction. In other cardiac diseases, such as congenital heart disease, autonomic dysfunction may be related to disease outcome. This is also the case after heart transplantation, when the heart is denervated. Interest in the origin of the autonomic nerve system has renewed since the role of autonomic function in disease progression was recognized, and some plasticity in autonomic regeneration is evident. As with many pathological processes, autonomic dysfunction based on pathological innervation may be a partial recapitulation of the early development of innervation. As such, insight into the development of cardiac innervation and an understanding of the cellular background contributing to cardiac innervation during different phases of development is required. This review describes the development of the cANS and focuses on the cellular contributions, either directly by delivering cells or indirectly by secretion of necessary factors or cell-derivatives.
Collapse
Affiliation(s)
- Anna M D Végh
- Department of Molecular Cell Biology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands.
| | - Sjoerd N Duim
- Department of Molecular Cell Biology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands.
| | - Anke M Smits
- Department of Molecular Cell Biology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands.
| | - Robert E Poelmann
- Department of Cardiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZC Leiden, The Netherlands.
- Institute of Biology Leiden, Leiden University, Sylviusweg 20, 2311 EZ Leiden, The Netherlands.
| | - Arend D J Ten Harkel
- Department of Pediatric Cardiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZC Leiden, The Netherlands.
| | - Marco C DeRuiter
- Department of Anatomy & Embryology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands.
| | - Marie José Goumans
- Department of Molecular Cell Biology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands.
| | - Monique R M Jongbloed
- Department of Cardiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZC Leiden, The Netherlands.
- Department of Pediatric Cardiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZC Leiden, The Netherlands.
| |
Collapse
|
27
|
Funato N, Kokubo H, Saga Y. Transcriptomic analyses of Hand2 transgenic embryos. GENOMICS DATA 2016; 9:60-2. [PMID: 27408813 PMCID: PMC4932614 DOI: 10.1016/j.gdata.2016.06.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 06/23/2016] [Indexed: 11/17/2022]
Abstract
In this article, we further provide the data generated for the previously published research article “Specification of jaw identity by the Hand2 transcription factor.” To better understand the downstream genes of the basic helix-loop-helix transcription factor Hand2, we generated double-transgenic mice (Hand2NC) by intercrossing CAG-floxed CAT-Hand2 mice with Wnt1-Cre mice for conditional activation of Hand2 expression in the neural crest. Altered expression of Hand2 induces transformation of the upper jaw to the lower jaw in Hand2NC mutant mice. This data article provides Tables detailing the differentially expressed genes between wild-type and Hand2NC mutant embryos. The raw array data of our transcriptomes as generated using Affymetrix microarrays are available on the NCBI Gene Expression Omnibus (GEO) browser (Reference number GSE75805).
Collapse
Affiliation(s)
- Noriko Funato
- Department of Signal Gene Regulation, Research Center for Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Hiroki Kokubo
- Division of Mammalian Development, National Institute of Genetics, Yata 1111, Mishima, Shizuoka 411-8540, Japan; Department of Genetics, The Graduate University for Advanced Studies, Yata 1111, Mishima, Shizuoka 411-8540, Japan; Department of Cardiovascular Physiology and Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minamiku, Hiroshima 734-8551, Japan
| | - Yumiko Saga
- Division of Mammalian Development, National Institute of Genetics, Yata 1111, Mishima, Shizuoka 411-8540, Japan; Department of Genetics, The Graduate University for Advanced Studies, Yata 1111, Mishima, Shizuoka 411-8540, Japan
| |
Collapse
|
28
|
Bondurand N, Southard-Smith EM. Mouse models of Hirschsprung disease and other developmental disorders of the enteric nervous system: Old and new players. Dev Biol 2016; 417:139-57. [PMID: 27370713 DOI: 10.1016/j.ydbio.2016.06.042] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 06/27/2016] [Accepted: 06/27/2016] [Indexed: 12/18/2022]
Abstract
Hirschsprung disease (HSCR, intestinal aganglionosis) is a multigenic disorder with variable penetrance and severity that has a general population incidence of 1/5000 live births. Studies using animal models have contributed to our understanding of the developmental origins of HSCR and the genetic complexity of this disease. This review summarizes recent progress in understanding control of enteric nervous system (ENS) development through analyses in mouse models. An overview of signaling pathways that have long been known to control the migration, proliferation and differentiation of enteric neural progenitors into and along the developing gut is provided as a framework for the latest information on factors that influence enteric ganglia formation and maintenance. Newly identified genes and additional factors beyond discrete genes that contribute to ENS pathology including regulatory sequences, miRNAs and environmental factors are also introduced. Finally, because HSCR has become a paradigm for complex oligogenic diseases with non-Mendelian inheritance, the importance of gene interactions, modifier genes, and initial studies on genetic background effects are outlined.
Collapse
Affiliation(s)
- Nadege Bondurand
- INSERM, U955, Equipe 6, F-94000 Creteil, France; Universite Paris-Est, UPEC, F-94000 Creteil, France.
| | - E Michelle Southard-Smith
- Vanderbilt University Medical Center, Department of Medicine, 2215 Garland Ave, Nashville, TN 37232, USA.
| |
Collapse
|
29
|
Morrison MA, Zimmerman MW, Look AT, Stewart RA. Studying the peripheral sympathetic nervous system and neuroblastoma in zebrafish. Methods Cell Biol 2016; 134:97-138. [PMID: 27312492 DOI: 10.1016/bs.mcb.2015.12.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The zebrafish serves as an excellent model to study vertebrate development and disease. Optically clear embryos, combined with tissue-specific fluorescent reporters, permit direct visualization and measurement of peripheral nervous system formation in real time. Additionally, the model is amenable to rapid cellular, molecular, and genetic approaches to determine how developmental mechanisms contribute to disease states, such as cancer. In this chapter, we describe the development of the peripheral sympathetic nervous system (PSNS) in general, and our current understanding of genetic pathways important in zebrafish PSNS development specifically. We also illustrate how zebrafish genetics is used to identify new mechanisms controlling PSNS development and methods for interrogating the potential role of PSNS developmental pathways in neuroblastoma pathogenesis in vivo using the zebrafish MYCN-driven neuroblastoma model.
Collapse
Affiliation(s)
- M A Morrison
- University of Utah, Salt Lake City, UT, United States
| | | | - A T Look
- Harvard Medical School, Boston, MA, United States
| | - R A Stewart
- University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
30
|
Stanzel S, Stubbusch J, Pataskar A, Howard MJ, Deller T, Ernsberger U, Tiwari VK, Rohrer H, Tsarovina K. Distinct roles of hand2 in developing and adult autonomic neurons. Dev Neurobiol 2016; 76:1111-24. [PMID: 26818017 DOI: 10.1002/dneu.22378] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 01/05/2016] [Accepted: 01/07/2016] [Indexed: 11/08/2022]
Abstract
The bHLH transcription factor Hand2 is essential for the acquisition and maintenance of noradrenergic properties of embryonic sympathetic neurons and controls neuroblast proliferation. Hand2 is also expressed in embryonic and postnatal parasympathetic ganglia and remains expressed in sympathetic neurons up to the adult stage. Here, we address its function in developing parasympathetic and adult sympathetic neurons. We conditionally deleted Hand2 in the parasympathetic sphenopalatine ganglion by crossing a line of floxed Hand2 mice with DbhiCre transgenic mice, taking advantage of the transient Dbh expression in parasympathetic ganglia. Hand2 elimination does not affect Dbh expression and sphenopalatine ganglion size at E12.5 and E16.5, in contrast to sympathetic ganglia. These findings demonstrate different functions for Hand2 in the parasympathetic and sympathetic lineage. Our previous Hand2 knockdown in postmitotic, differentiated chick sympathetic neurons resulted in decreased expression of noradrenergic marker genes but it was unclear whether Hand2 is required for maintaining noradrenergic neuron identity in adult animals. We now show that Hand2 elimination in adult Dbh-expressing sympathetic neurons does not decrease the expression of Th and Dbh, in contrast to the situation during development. However, gene expression profiling of adult sympathetic neurons identified 75 Hand2-dependent target genes. Interestingly, a notable proportion of down-regulated genes (15%) encode for proteins with synaptic and neurotransmission functions. These results demonstrate a change in Hand2 target genes during maturation of sympathetic neurons. Whereas Hand2 controls genes regulating noradrenergic differentiation during development, Hand2 seems to be involved in the regulation of genes controlling neurotransmission in adult sympathetic neurons. © 2016 Wiley Periodicals, Inc. Develop Neurobiol 76: 1111-1124, 2016.
Collapse
Affiliation(s)
- Sabine Stanzel
- Developmental Neurobiology, Max-Planck-Institute for Brain Research, Max-von-Laue-Str. 4, Frankfurt/M, 60438, Germany
| | - Jutta Stubbusch
- Developmental Neurobiology, Max-Planck-Institute for Brain Research, Max-von-Laue-Str. 4, Frankfurt/M, 60438, Germany
| | - Abhijeet Pataskar
- Institute of Molecular Biology (IMB) Boehringer Ingelheim Foundation, Ackermannweg 4, Mainz, 55128, Germany
| | - Marthe J Howard
- Department of Neurosciences and Program in Neurosciences and Neurological Disorders, University of Toledo Health Sciences Campus, Toledo, Ohio, 43614
| | - Thomas Deller
- Institute of Clinical Neuroanatomy, Goethe University Frankfurt/M, Theodor-Stern-Kai 7, Frankfurt/M, 60590, Germany
| | - Uwe Ernsberger
- Developmental Neurobiology, Max-Planck-Institute for Brain Research, Max-von-Laue-Str. 4, Frankfurt/M, 60438, Germany.,Institute of Clinical Neuroanatomy, Goethe University Frankfurt/M, Theodor-Stern-Kai 7, Frankfurt/M, 60590, Germany.,Ernst-Strüngmann-Institute, Deutschordenstr. 46, Frankfurt/M, 60528, Germany
| | - Vijay K Tiwari
- Institute of Molecular Biology (IMB) Boehringer Ingelheim Foundation, Ackermannweg 4, Mainz, 55128, Germany
| | - Hermann Rohrer
- Developmental Neurobiology, Max-Planck-Institute for Brain Research, Max-von-Laue-Str. 4, Frankfurt/M, 60438, Germany.,Institute of Clinical Neuroanatomy, Goethe University Frankfurt/M, Theodor-Stern-Kai 7, Frankfurt/M, 60590, Germany.,Ernst-Strüngmann-Institute, Deutschordenstr. 46, Frankfurt/M, 60528, Germany
| | - Konstantina Tsarovina
- Developmental Neurobiology, Max-Planck-Institute for Brain Research, Max-von-Laue-Str. 4, Frankfurt/M, 60438, Germany
| |
Collapse
|
31
|
Chan WH, Gonsalvez DG, Young HM, Southard-Smith EM, Cane KN, Anderson CR. Differences in CART expression and cell cycle behavior discriminate sympathetic neuroblast from chromaffin cell lineages in mouse sympathoadrenal cells. Dev Neurobiol 2015; 76:137-49. [PMID: 25989220 DOI: 10.1002/dneu.22304] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Revised: 04/22/2015] [Accepted: 05/13/2015] [Indexed: 12/31/2022]
Abstract
Adrenal medullary chromaffin cells and peripheral sympathetic neurons originate from a common sympathoadrenal (SA) progenitor cell. The timing and phenotypic changes that mark this lineage diversification are not fully understood. The present study investigated the expression patterns of phenotypic markers, and cell cycle dynamics, in the adrenal medulla and the neighboring suprarenal ganglion of embryonic mice. The noradrenergic marker, tyrosine hydroxylase (TH), was detected in both presumptive adrenal medulla and sympathetic ganglion cells, but with significantly stronger immunostaining in the former. There was intense cocaine and amphetamine-regulated transcript (CART) peptide immunostaining in most neuroblasts, whereas very few adrenal chromaffin cells showed detectable CART immunostaining. This phenotypic segregation appeared as early as E12.5, before anatomical segregation of the two cell types. Cell cycle dynamics were also examined. Initially, 88% of Sox10 positive (+) neural crest progenitors were proliferating at E10.5. Many SA progenitor cells withdrew from the cell cycle at E11.5 as they started to express TH. Whereas 70% of neuroblasts (TH+/CART+ cells) were back in the cell cycle at E12.5, only around 20% of chromaffin (CART negative) cells were in the cell cycle at E12.5 and subsequent days. Thus, chromaffin cell and neuroblast lineages showed differences in proliferative behavior from their earliest appearance. We conclude that the intensity of TH immunostaining and the expression of CART permit early discrimination of chromaffin cells and sympathetic neuroblasts, and that developing chromaffin cells exhibit significantly lower proliferative activity relative to sympathetic neuroblasts.
Collapse
Affiliation(s)
- Wing Hei Chan
- Department of Anatomy and Neuroscience, University of Melbourne, Victoria, 3010, Australia
| | - David G Gonsalvez
- Department of Anatomy and Neuroscience, University of Melbourne, Victoria, 3010, Australia
| | - Heather M Young
- Department of Anatomy and Neuroscience, University of Melbourne, Victoria, 3010, Australia
| | - E Michelle Southard-Smith
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University School of Medicine, 529 Light Hall, 2215 Garland Avenue, Nashville, Tennessee
| | - Kylie N Cane
- Department of Anatomy and Neuroscience, University of Melbourne, Victoria, 3010, Australia
| | - Colin R Anderson
- Department of Anatomy and Neuroscience, University of Melbourne, Victoria, 3010, Australia
| |
Collapse
|
32
|
Nishimura Y, Murakami S, Ashikawa Y, Sasagawa S, Umemoto N, Shimada Y, Tanaka T. Zebrafish as a systems toxicology model for developmental neurotoxicity testing. Congenit Anom (Kyoto) 2015; 55:1-16. [PMID: 25109898 DOI: 10.1111/cga.12079] [Citation(s) in RCA: 137] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 07/29/2014] [Indexed: 12/18/2022]
Abstract
The developing brain is extremely sensitive to many chemicals. Exposure to neurotoxicants during development has been implicated in various neuropsychiatric and neurological disorders, including autism spectrum disorder, attention deficit hyperactive disorder, schizophrenia, Parkinson's disease, and Alzheimer's disease. Although rodents have been widely used for developmental neurotoxicity testing, experiments using large numbers of rodents are time-consuming, expensive, and raise ethical concerns. Using alternative non-mammalian animal models may relieve some of these pressures by allowing testing of large numbers of subjects while reducing expenses and minimizing the use of mammalian subjects. In this review, we discuss some of the advantages of using zebrafish in developmental neurotoxicity testing, focusing on central nervous system development, neurobehavior, toxicokinetics, and toxicodynamics in this species. We also describe some important examples of developmental neurotoxicity testing using zebrafish combined with gene expression profiling, neuroimaging, or neurobehavioral assessment. Zebrafish may be a systems toxicology model that has the potential to reveal the pathways of developmental neurotoxicity and to provide a sound basis for human risk assessments.
Collapse
Affiliation(s)
- Yuhei Nishimura
- Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Mie University Graduate School of Medicine, Tsu, Japan; Mie University Medical Zebrafish Research Center, Tsu, Japan; Depertment of Systems Pharmacology, Mie University Graduate School of Medicine, Tsu, Japan; Department of Omics Medicine, Mie University Industrial Technology Innovation Institute, Tsu, Japan; Department of Bioinformatics, Mie University Life Science Research Center, Tsu, Japan
| | | | | | | | | | | | | |
Collapse
|
33
|
Segregation of neuronal and neuroendocrine differentiation in the sympathoadrenal lineage. Cell Tissue Res 2014; 359:333-41. [PMID: 25038743 DOI: 10.1007/s00441-014-1947-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 06/06/2014] [Indexed: 10/25/2022]
Abstract
Neuronal and neuroendocrine cells possess the capacity for Ca(2+)-regulated discharge of messenger molecules, which they release into synapses or the blood stream, respectively. The neural-crest-derived sympathoadrenal lineage gives rise to the sympathetic neurons of the autonomic nervous system and the neuroendocrine chromaffin cells of the adrenal medulla. These cells provide an excellent model system for studying common and distinct developmental mechanisms underlying the acquisition of neuroendocrine and neuronal properties. As catecholaminergic cells, they possess common markers related to noradrenaline synthesis, storage and release, but they also display diverging gene expression patterns and are morphologically and functionally different. The precise mechanisms that underlie the diversification of sympathoadrenal cells into neurons and neuroendocrine cells are not fully understood. However, in the past we could show that the establishment of a chromaffin phenotype does not depend on signals from the adrenal cortex and that chromaffin cells and sympathetic neurons apparently differ from the onset of their catecholaminergic differentiation. Nevertheless, the cues that specifically induce neuroendocrine features remain elusive. The early development of the progenitors of chromaffin cells and sympathetic neurons depends on a common set of transcription factors with overlapping but distinct influences on their development. In addition to the well-defined role of transcription factors as developmental regulators, our understanding of post-transcriptional gene regulation by microRNAs has substantially increased within the last few decades. This review highlights the major similarities and differences between chromaffin cells and sympathetic neurons, summarizes our current knowledge of the roles of selected transcription factors, microRNAs and environmental signals for the neuroendocrine differentiation of sympathoadrenal cells, and draws comparisons with the development of other endocrine and neuronal cells.
Collapse
|
34
|
Kameda Y. Signaling molecules and transcription factors involved in the development of the sympathetic nervous system, with special emphasis on the superior cervical ganglion. Cell Tissue Res 2014; 357:527-48. [PMID: 24770894 DOI: 10.1007/s00441-014-1847-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Accepted: 02/12/2014] [Indexed: 12/16/2022]
Abstract
The cells that constitute the sympathetic nervous system originate from the neural crest. This review addresses the current understanding of sympathetic ganglion development viewed from molecular and morphological perspectives. Development of the sympathetic nervous system is categorized into three main steps, as follows: (1) differentiation and migration of cells in the neural crest lineage for formation of the primary sympathetic chain, (2) differentiation of sympathetic progenitors, and (3) growth and survival of sympathetic ganglia. The signaling molecules and transcription factors involved in each of these developmental stages are elaborated mostly on the basis of the results of targeted mutation of respective genes. Analyses in mutant mice revealed differences between the superior cervical ganglion (SCG) and the other posterior sympathetic ganglia. This review provides a summary of the similarities and differences in the development of the SCG and other posterior sympathetic ganglia. Relevant to the development of sympathetic ganglia is the demonstration that neuroendocrine cells, such as adrenal chromaffin cells and carotid body glomus cells, share a common origin with the sympathetic ganglia. Neural crest cells at the trunk level give rise to common sympathoadrenal progenitors of sympathetic neurons and chromaffin cells, while progenitors segregated from the SCG give rise to glomus cells. After separation from the sympathetic primordium, the progenitors of both chromaffin cells and glomus cells colonize the anlage of the adrenal gland and carotid body, respectively. This review highlights the biological properties of chromaffin cells and glomus cells, because, although both cell types are derivatives of sympathetic primordium, they are distinct in many respects.
Collapse
Affiliation(s)
- Yoko Kameda
- Department of Anatomy, Kitasato University School of Medicine, Sagamihara, Kanagawa, 252-0374, Japan,
| |
Collapse
|
35
|
Ascl1 as a novel player in the Ptf1a transcriptional network for GABAergic cell specification in the retina. PLoS One 2014; 9:e92113. [PMID: 24643195 PMCID: PMC3958475 DOI: 10.1371/journal.pone.0092113] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Accepted: 02/17/2014] [Indexed: 11/19/2022] Open
Abstract
In contrast with the wealth of data involving bHLH and homeodomain transcription factors in retinal cell type determination, the molecular bases underlying neurotransmitter subtype specification is far less understood. Using both gain and loss of function analyses in Xenopus, we investigated the putative implication of the bHLH factor Ascl1 in this process. We found that in addition to its previously characterized proneural function, Ascl1 also contributes to the specification of the GABAergic phenotype. We showed that it is necessary for retinal GABAergic cell genesis and sufficient in overexpression experiments to bias a subset of retinal precursor cells towards a GABAergic fate. We also analysed the relationships between Ascl1 and a set of other bHLH factors using an in vivo ectopic neurogenic assay. We demonstrated that Ascl1 has unique features as a GABAergic inducer and is epistatic over factors endowed with glutamatergic potentialities such as Neurog2, NeuroD1 or Atoh7. This functional specificity is conferred by the basic DNA binding domain of Ascl1 and involves a specific genetic network, distinct from that underlying its previously demonstrated effects on catecholaminergic differentiation. Our data show that GABAergic inducing activity of Ascl1 requires the direct transcriptional regulation of Ptf1a, providing therefore a new piece of the network governing neurotransmitter subtype specification during retinogenesis.
Collapse
|
36
|
Loss of Hand2 in a population of Periostin lineage cells results in pronounced bradycardia and neonatal death. Dev Biol 2014; 388:149-58. [PMID: 24565998 DOI: 10.1016/j.ydbio.2014.02.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Revised: 01/26/2014] [Accepted: 02/08/2014] [Indexed: 11/21/2022]
Abstract
The Periostin Cre (Postn-Cre) lineage includes endocardial and neural crest derived mesenchymal cells of the cardiac cushions, neural crest-derived components of the sympathetic and enteric nervous systems, and cardiac fibroblasts. In this study, we use the Postn-Cre transgenic allele to conditionally ablate Hand2 (H2CKO). We find that Postn-Cre H2CKOs die shortly after birth despite a lack of obvious cardiac structural defects. To ascertain the cause of death, we performed a detailed comparison of the Postn-Cre lineage and Hand2 expression at mid and late stages of embryonic development. Gene expression analyses demonstrate that Postn-Cre ablates Hand2 from the adrenal medulla as well as the sphenopalatine ganglia of the head. In both cases, Hand2 loss-of-function dramatically reduces expression of Dopamine Beta Hydroxylase (Dbh), a gene encoding a crucial catecholaminergic biosynthetic enzyme. Expression of the genes Tyrosine Hydroxylase (Th) and Phenylethanolamine N-methyltransferase (Pnmt), which also encode essential catecholaminergic enzymes, were severely reduced in postnatal adrenal glands. Electrocardiograms demonstrate that 3-day postnatal Postn-Cre H2CKO pups exhibit sinus bradycardia. In conjunction with the aforementioned gene expression analyses, these results strongly suggest that the observed postnatal lethality occurs due to a catecholamine deficiency and subsequent heart failure.
Collapse
|
37
|
Tamura M, Amano T, Shiroishi T. The Hand2 Gene Dosage Effect in Developmental Defects and Human Congenital Disorders. Curr Top Dev Biol 2014; 110:129-52. [DOI: 10.1016/b978-0-12-405943-6.00003-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
38
|
Dirkx E, Gladka MM, Philippen LE, Armand AS, Kinet V, Leptidis S, el Azzouzi H, Salic K, Bourajjaj M, da Silva GJJ, Olieslagers S, van der Nagel R, de Weger R, Bitsch N, Kisters N, Seyen S, Morikawa Y, Chanoine C, Heymans S, Volders PGA, Thum T, Dimmeler S, Cserjesi P, Eschenhagen T, da Costa Martins PA, De Windt LJ. Nfat and miR-25 cooperate to reactivate the transcription factor Hand2 in heart failure. Nat Cell Biol 2013; 15:1282-93. [DOI: 10.1038/ncb2866] [Citation(s) in RCA: 112] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Accepted: 09/24/2013] [Indexed: 01/05/2023]
|
39
|
Abstract
Autonomic cardiac neurons have a common origin in the neural crest but undergo distinct developmental differentiation as they mature toward their adult phenotype. Progenitor cells respond to repulsive cues during migration, followed by differentiation cues from paracrine sources that promote neurochemistry and differentiation. When autonomic axons start to innervate cardiac tissue, neurotrophic factors from vascular tissue are essential for maintenance of neurons before they reach their targets, upon which target-derived trophic factors take over final maturation, synaptic strength and postnatal survival. Although target-derived neurotrophins have a central role to play in development, alternative sources of neurotrophins may also modulate innervation. Both developing and adult sympathetic neurons express proNGF, and adult parasympathetic cardiac ganglion neurons also synthesize and release NGF. The physiological function of these “non-classical” cardiac sources of neurotrophins remains to be determined, especially in relation to autocrine/paracrine sustenance during development.
Cardiac autonomic nerves are closely spatially associated in cardiac plexuses, ganglia and pacemaker regions and so are sensitive to release of neurotransmitter, neuropeptides and trophic factors from adjacent nerves. As such, in many cardiac pathologies, it is an imbalance within the two arms of the autonomic system that is critical for disease progression. Although this crosstalk between sympathetic and parasympathetic nerves has been well established for adult nerves, it is unclear whether a degree of paracrine regulation occurs across the autonomic limbs during development. Aberrant nerve remodeling is a common occurrence in many adult cardiovascular pathologies, and the mechanisms regulating outgrowth or denervation are disparate. However, autonomic neurons display considerable plasticity in this regard with neurotrophins and inflammatory cytokines having a central regulatory function, including in possible neurotransmitter changes. Certainly, neurotrophins and cytokines regulate transcriptional factors in adult autonomic neurons that have vital differentiation roles in development. Particularly for parasympathetic cardiac ganglion neurons, additional examinations of developmental regulatory mechanisms will potentially aid in understanding attenuated parasympathetic function in a number of conditions, including heart failure.
Collapse
Affiliation(s)
- Wohaib Hasan
- Knight Cardiovascular Institute; Oregon Health & Science University; Portland, OR USA
| |
Collapse
|
40
|
Huber K, Narasimhan P, Shtukmaster S, Pfeifer D, Evans SM, Sun Y. The LIM-Homeodomain transcription factor Islet-1 is required for the development of sympathetic neurons and adrenal chromaffin cells. Dev Biol 2013; 380:286-98. [PMID: 23648511 PMCID: PMC5544970 DOI: 10.1016/j.ydbio.2013.04.027] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Revised: 04/04/2013] [Accepted: 04/17/2013] [Indexed: 12/25/2022]
Abstract
Islet-1 is a LIM-Homeodomain transcription factor with important functions for the development of distinct neuronal and non-neuronal cell populations. We show here that Islet-1 acts genetically downstream of Phox2B in cells of the sympathoadrenal cell lineage and that the development of sympathetic neurons and chromaffin cells is impaired in mouse embryos with a conditional deletion of Islet-1 controlled by the wnt1 promotor. Islet-1 is not essential for the initial differentiation of sympathoadrenal cells, as indicated by the correct expression of pan-neuronal and catecholaminergic subtype specific genes in primary sympathetic ganglia of Islet-1 deficient mouse embryos. However, our data indicate that the subsequent survival of sympathetic neuron precursors and their differentiation towards TrkA expressing neurons depends on Islet-1 function. In contrast to spinal sensory neurons, sympathetic neurons of Islet-1 deficient mice did not display ectopic expression of genes normally present in the CNS. In Islet-1 deficient mouse embryos the numbers of chromaffin cells were only mildly reduced, in contrast to that of sympathetic neurons, but the initiation of the adrenaline synthesizing enzyme PNMT was abrogated and the expression level of chromogranin A was diminished. Microarray analysis revealed that developing chromaffin cells of Islet-1 deficient mice displayed normal expression levels of TH, DBH and the transcription factors Phox2B, Mash-1, Hand2, Gata3 and Insm1, but the expression levels of the transcription factors Gata2 and Hand1, and AP-2β were significantly reduced. Together our data indicate that Islet-1 is not essentially required for the initial differentiation of sympathoadrenal cells, but has an important function for the correct subsequent development of sympathetic neurons and chromaffin cells.
Collapse
Affiliation(s)
- Katrin Huber
- Institute for Anatomy and Cell Biology, Department of Molecular Embryology, Albert-Ludwigs-University, Freiburg, Germany.
| | | | | | | | | | | |
Collapse
|
41
|
The transcription factor Hmx1 and growth factor receptor activities control sympathetic neurons diversification. EMBO J 2013; 32:1613-25. [PMID: 23591430 DOI: 10.1038/emboj.2013.85] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Accepted: 03/15/2013] [Indexed: 01/17/2023] Open
Abstract
The sympathetic nervous system relies on distinct populations of neurons that use noradrenaline or acetylcholine as neurotransmitter. We show that fating of the sympathetic lineage at early stages results in hybrid precursors from which, genetic cell-lineage tracing reveals, all types progressively emerge by principal mechanisms of maintenance, repression and induction of phenotypes. The homeobox transcription factor HMX1 represses Tlx3 and Ret, induces TrkA and maintains tyrosine hydroxylase (Th) expression in precursors, thus driving segregation of the noradrenergic sympathetic fate. Cholinergic sympathetic neurons develop through cross-regulatory interactions between TRKC and RET in precursors, which lead to Hmx1 repression and sustained Tlx3 expression, thereby resulting in failure of TrkA induction and loss of maintenance of Th expression. Our results provide direct evidence for a model in which diversification of noradrenergic and cholinergic sympathetic neurons is based on a principle of cross-repressive functions in which the specific cell fates are directed by an active suppression of the expression of transcription factors and receptors that direct the alternative fate.
Collapse
|
42
|
Vincentz JW, Firulli BA, Lin A, Spicer DB, Howard MJ, Firulli AB. Twist1 controls a cell-specification switch governing cell fate decisions within the cardiac neural crest. PLoS Genet 2013; 9:e1003405. [PMID: 23555309 PMCID: PMC3605159 DOI: 10.1371/journal.pgen.1003405] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Accepted: 02/07/2013] [Indexed: 01/31/2023] Open
Abstract
Neural crest cells are multipotent progenitor cells that can generate both ectodermal cell types, such as neurons, and mesodermal cell types, such as smooth muscle. The mechanisms controlling this cell fate choice are not known. The basic Helix-loop-Helix (bHLH) transcription factor Twist1 is expressed throughout the migratory and post-migratory cardiac neural crest. Twist1 ablation or mutation of the Twist-box causes differentiation of ectopic neuronal cells, which molecularly resemble sympathetic ganglia, in the cardiac outflow tract. Twist1 interacts with the pro-neural factor Sox10 via its Twist-box domain and binds to the Phox2b promoter to repress transcriptional activity. Mesodermal cardiac neural crest trans-differentiation into ectodermal sympathetic ganglia-like neurons is dependent upon Phox2b function. Ectopic Twist1 expression in neural crest precursors disrupts sympathetic neurogenesis. These data demonstrate that Twist1 functions in post-migratory neural crest cells to repress pro-neural factors and thereby regulate cell fate determination between ectodermal and mesodermal lineages. During vertebrate development, a unique population of cells, termed neural crest cells, migrates throughout the developing embryo, generating various cell types, for example, the smooth muscle that divides the aorta and pulmonary artery where they connect to the heart, and the autonomic neurons, which coordinate organ function. The distinctions between neural crest cells that will form smooth muscle and those that will become neurons are thought to occur prior to migration. Here, we show that, in mice with mutations of the transcription factor Twist1, a subpopulation of presumptive smooth muscle cells, following migration to the heart, instead mis-specify to resemble autonomic neurons. Twist1 represses transcription of the pro-neural factor Phox2b both through antagonism of its upstream effector, Sox10, and through direct binding to its promoter. Phox2b is absolutely required for autonomic neuron development, and indeed, the aberrant neurons in Twist1 mutants disappear when Phox2b is also mutated. Ectopic Twist1 expression within all neural crest cells disrupts the specification of normal autonomic neurons. Collectively, these data reveal that neural crest cells can alter their cell fate from mesoderm to ectoderm after they have migrated and that Twist1 functions to maintain neural crest cell potency during embryonic development.
Collapse
Affiliation(s)
- Joshua W Vincentz
- Riley Heart Research Center, Herman B Wells Center for Pediatric Research, Division of Pediatrics Cardiology, Departments of Anatomy, Indiana University Medical School, Indianapolis, Indiana, United States of America
| | | | | | | | | | | |
Collapse
|
43
|
Vincentz JW, Rubart M, Firulli AB. Ontogeny of cardiac sympathetic innervation and its implications for cardiac disease. Pediatr Cardiol 2012; 33:923-8. [PMID: 22395650 PMCID: PMC3391355 DOI: 10.1007/s00246-012-0248-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2012] [Accepted: 02/15/2012] [Indexed: 12/16/2022]
Abstract
The vertebrate heart is innervated by the sympathetic and parasympathetic components of the peripheral autonomic nervous system, which regulates its contractile rate and force. Understanding the mechanisms that control sympathetic neuronal growth, differentiation, and innervation of the heart may provide insight into the etiology of cardiac arrhythmogenesis. This review provides an overview of the cell signaling pathways and transcriptional effectors that regulate both the noradrenergic gene program during sympathetic neurogenesis and regional nerve density during cardiac innervation. Recent studies exploring transcriptional regulation of the bHLH transcription factor Hand1 in developing sympathetic neurons are explored, and how the Hand1 sympathetic neuron-specific cis-regulatory element may be used further to assess the contribution of altered sympathetic innervation to human cardiac disease is discussed.
Collapse
|
44
|
A Phox2- and Hand2-dependent Hand1 cis-regulatory element reveals a unique gene dosage requirement for Hand2 during sympathetic neurogenesis. J Neurosci 2012; 32:2110-20. [PMID: 22323723 DOI: 10.1523/jneurosci.3584-11.2012] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Neural crest cell specification and differentiation to a sympathetic neuronal fate serves as an important model for neurogenesis and depends upon the function of both bHLH transcription factors, notably Hand2, and homeodomain transcription factors, including Phox2b. Here, we define a 1007 bp cis-regulatory element 5' of the Hand1 gene sufficient to drive reporter expression within the sympathetic chain of transgenic mice. Comparative genomic analyses uncovered evolutionarily conserved consensus-binding sites within this element, which chromatin immunoprecipitation and electrophoretic mobility shift assays confirm are bound by Hand2 and Phox2b. Mutational analyses revealed that the conserved Phox2 and E-box binding sites are necessary for proper cis-regulatory element activity, and expression analyses on both Hand2 conditionally null and hypomorphic backgrounds demonstrate that Hand2 is required for reporter activation in a gene dosage-dependent manner. We demonstrate that Hand2 and Hand1 differentially bind the E-boxes in this cis-regulatory element, establishing molecular differences between these two factors. Finally, we demonstrate that Hand1 is dispensable for normal tyrosine hydroxylase (TH) and dopamine β-hydroxylase (DBH) expression in sympathetic neurons, even when Hand2 gene dosage is concurrently reduced by half. Together, these data define a tissue-specific Hand1 cis-regulatory element controlled by two factors essential for the development of the sympathetic nervous system and provide in vivo regulatory evidence to support previous findings that Hand2, rather than Hand1, is predominantly responsible for regulating TH, DBH, and Hand1 expression in developing sympathetic neurons.
Collapse
|
45
|
Abstract
Autonomic neuron development is controlled by a network of transcription factors, which is induced by bone morphogenetic protein signalling in neural crest progenitor cells. This network intersects with a transcriptional program in migratory neural crest cells that pre-specifies autonomic neuron precursor cells. Recent findings demonstrate that the transcription factors acting in the initial specification and differentiation of sympathetic neurons are also important for the proliferation of progenitors and immature neurons during neurogenesis. Elimination of Phox2b, Hand2 and Gata3 in differentiated neurons affects the expression of subtype-specific and/or generic neuronal properties or neuron survival. Taken together, transcription factors previously shown to act in initial neuron specification and differentiation display a much broader spectrum of functions, including control of neurogenesis and the maintenance of subtype characteristics and survival of mature neurons.
Collapse
Affiliation(s)
- Hermann Rohrer
- Research Group Developmental Neurobiology, Max-Planck-Institute for Brain Research, 60528 Frankfurt/Main, Germany.
| |
Collapse
|
46
|
Lei J, Howard MJ. Targeted deletion of Hand2 in enteric neural precursor cells affects its functions in neurogenesis, neurotransmitter specification and gangliogenesis, causing functional aganglionosis. Development 2011; 138:4789-800. [PMID: 21989918 DOI: 10.1242/dev.060053] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Targeted deletion of the bHLH DNA-binding protein Hand2 in the neural crest, impacts development of the enteric nervous system (ENS), possibly by regulating the transition from neural precursor cell to neuron. We tested this hypothesis by targeting Hand2 deletion in nestin-expressing neural precursor (NEP) cells. The mutant mice showed abnormal ENS development, resulting in lethal neurogenic pseudo-obstruction. Neurogenesis of neurons derived from NEP cells identified a second nestin non-expressing neural precursor (NNEP) cell in the ENS. There was substantial compensation for the loss of neurons derived from the NEP pool by the NNEP pool but this was insufficient to abrogate the negative impact of Hand2 deletion. Hand2-mediated regulation of proliferation affected both neural precursor and neuron numbers. Differentiation of glial cells derived from the NEP cells was significantly decreased with no compensation from the NNEP pool of cells. Our data indicate differential developmental potential of NEPs and NNEPs; NNEPs preferentially differentiate as neurons, whereas NEPs give rise to both neurons and glial cells. Deletion of Hand2 also resulted in complete loss of NOS and VIP and a significant decrease in expression of choline acetyltransferase and calretinin, demonstrating a role for Hand2 in neurotransmitter specification and/or expression. Loss of Hand2 resulted in a marked disruption of the developing neural network, exemplified by lack of a myenteric plexus and extensive overgrowth of fibers. Thus, Hand2 is essential for neurogenesis, neurotransmitter specification and neural network patterning in the developing ENS.
Collapse
Affiliation(s)
- Jun Lei
- Department of Neurosciences and Program in Neurosciences and Neurodegenerative Diseases, University of Toledo Health Sciences Campus, Toledo, OH 43614, USA
| | | |
Collapse
|
47
|
D’Autréaux F, Margolis KG, Roberts J, Stevanovic K, Mawe G, Li Z, Karamooz N, Ahuja A, Morikawa Y, Cserjesi P, Setlick W, Gershon MD. Expression level of Hand2 affects specification of enteric neurons and gastrointestinal function in mice. Gastroenterology 2011; 141:576-87, 587.e1-6. [PMID: 21669203 PMCID: PMC3152642 DOI: 10.1053/j.gastro.2011.04.059] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2010] [Revised: 03/20/2011] [Accepted: 04/12/2011] [Indexed: 01/08/2023]
Abstract
BACKGROUND & AIMS Hand2 is a basic helix-loop-helix transcription factor required for terminal differentiation of enteric neurons. We studied Hand2 haploinsufficient mice, to determine whether reduced expression of Hand2 allows sufficient enteric neurogenesis for survival, but not for development of a normal enteric nervous system (ENS). METHODS Enteric transcripts that encode Hand2 and the neuron-specific embryonic lethal abnormal vision proteins HuB, HuC, and HuD were quantified. Immunocytochemistry was used to identify and quantify neurons. Apoptosis was analyzed with the terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labeling procedure. Intracellular microelectrodes were used to record inhibitory junction potentials. Gastrointestinal transit and colonic motility were measured in vivo. RESULTS Levels of of enteric Hand2 transcripts were associated with genotypes of mice, in the following order: Hand2(+/+) > Hand2(LoxP/+) > Hand2(+/-) > Hand2(LoxP/-). Parallel reductions were found in expression of HuD and in regional and phenotypic manners. Numbers of neurons, numbers of neuronal nitric oxide synthase(+) and calretinin(+), but not substance P(+) or vasoactive intestinal peptide(+) neurons, decreased. No effects were observed in stomach or cecum. Apoptosis was not detected, consistent with the concept that Hand2 inhibits neuronal differentiation, rather than regulates survival. The amplitude of inhibitory junction potentials in colonic circular muscle was similar in Hand2 wild-type and haploinsufficient mice, although in haploinsufficient mice, the purinergic component was reduced and a nitrergic component appeared. The abnormal ENS of haploinsufficient mice slowed gastrointestinal motility but protected mice against colitis. CONCLUSIONS Reduced expression of factors required for development of the ENS can cause defects in the ENS that are subtle enough to escape detection yet cause significant abnormalities in bowel function.
Collapse
Affiliation(s)
- Fabien D’Autréaux
- Département de Biologie - Dévelopement et évolution du système nerveux, CNRS - Ecole Normale Supérieure, Paris, France
| | - Kara G. Margolis
- Department of Pediatrics, Columbia University, College of P&S, New York, NY, U.S.A
| | - Jane Roberts
- Department of Anatomy and Neurobiology, University of Vermont, Burlington, VT, U.S.A
| | - Korey Stevanovic
- Department of Pediatrics, Columbia University, College of P&S, New York, NY, U.S.A
| | - Gary Mawe
- Department of Anatomy and Neurobiology, University of Vermont, Burlington, VT, U.S.A
| | - Zhishan Li
- Pathology and Cell Biology, Columbia University, College of P&S, New York, NY, U.S.A
| | - Nima Karamooz
- Pathology and Cell Biology, Columbia University, College of P&S, New York, NY, U.S.A
| | - Ankur Ahuja
- Pathology and Cell Biology, Columbia University, College of P&S, New York, NY, U.S.A
| | - Yuka Morikawa
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, U.S.A
| | - Peter Cserjesi
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, U.S.A
| | - Wanda Setlick
- Pathology and Cell Biology, Columbia University, College of P&S, New York, NY, U.S.A
| | - Michael D. Gershon
- Pathology and Cell Biology, Columbia University, College of P&S, New York, NY, U.S.A
| |
Collapse
|
48
|
Flames N, Hobert O. Transcriptional Control of the Terminal Fate of Monoaminergic Neurons. Annu Rev Neurosci 2011; 34:153-84. [DOI: 10.1146/annurev-neuro-061010-113824] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Nuria Flames
- Department of Biochemistry and Molecular Biophysics, Howard Hughes Medical Institute, Columbia University Medical Center, New York, New York 10032;
- Genes & Disease Program, Center for Genomic Regulation (CRG), Barcelona, Spain E-08003;
- Present address: Instituto de Biomedicina de Valencia IBV-CSIC, E-46010 Valencia, Spain
| | - Oliver Hobert
- Department of Biochemistry and Molecular Biophysics, Howard Hughes Medical Institute, Columbia University Medical Center, New York, New York 10032;
| |
Collapse
|
49
|
Barron F, Woods C, Kuhn K, Bishop J, Howard MJ, Clouthier DE. Downregulation of Dlx5 and Dlx6 expression by Hand2 is essential for initiation of tongue morphogenesis. Development 2011; 138:2249-59. [PMID: 21558373 DOI: 10.1242/dev.056929] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Lower jaw development is a complex process in which multiple signaling cascades establish a proximal-distal organization. These cascades are regulated both spatially and temporally and are constantly refined through both induction of normal signals and inhibition of inappropriate signals. The connective tissue of the tongue arises from cranial neural crest cell-derived ectomesenchyme within the mandibular portion of the first pharyngeal arch and is likely to be impacted by this signaling. Although the developmental mechanisms behind later aspects of tongue development, including innervation and taste acquisition, have been elucidated, the early patterning signals driving ectomesenchyme into a tongue lineage are largely unknown. We show here that the basic helix-loop-helix transcription factor Hand2 plays key roles in establishing the proximal-distal patterning of the mouse lower jaw, in part through establishing a negative-feedback loop in which Hand2 represses Dlx5 and Dlx6 expression in the distal arch ectomesenchyme following Dlx5- and Dlx6-mediated induction of Hand2 expression in the same region. Failure to repress distal Dlx5 and Dlx6 expression results in upregulation of Runx2 expression in the mandibular arch and the subsequent formation of aberrant bone in the lower jaw along with proximal-distal duplications. In addition, there is an absence of lateral lingual swelling expansion, from which the tongue arises, resulting in aglossia. Hand2 thus appears to establish a distal mandibular arch domain that is conducive for lower jaw development, including the initiation of tongue mesenchyme morphogenesis.
Collapse
Affiliation(s)
- Francie Barron
- Department of Craniofacial Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | | | | | | | | | | |
Collapse
|
50
|
Vincentz JW, Barnes RM, Firulli AB. Hand factors as regulators of cardiac morphogenesis and implications for congenital heart defects. BIRTH DEFECTS RESEARCH. PART A, CLINICAL AND MOLECULAR TERATOLOGY 2011; 91:485-94. [PMID: 21462297 PMCID: PMC3119928 DOI: 10.1002/bdra.20796] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2010] [Revised: 01/06/2011] [Accepted: 02/02/2011] [Indexed: 11/08/2022]
Abstract
Almost 15 years of careful study have established the related basic Helix-Loop-Helix (bHLH) transcription factors Hand1 and Hand2 as critical for heart development across evolution. Hand factors make broad contributions, revealed through animal models, to the development of multiple cellular lineages that ultimately contribute to the heart. They perform critical roles in ventricular cardiomyocyte growth, differentiation, morphogenesis, and conduction. They are also important for the proper development of the cardiac outflow tract, epicardium, and endocardium. Molecularly, they function both through DNA binding and through protein-protein interactions, which are regulated transcriptionally, posttranscriptionally by microRNAs, and posttranslationally through phosphoregulation. Although direct Hand factor transcriptional targets are progressively being identified, confirmed direct targets of Hand factor transcriptional activity in the heart are limited. Identification of these targets will be critical to model the mechanisms by which Hand factor bHLH interactions affect developmental pathways. Improved understanding of Hand factor-mediated transcriptional cascades will be necessary to determine how Hand factor dysregulation translates to human disease phenotypes. This review summarizes the insight that animal models have provided into the regulation and function of these factors during heart development, in addition to the recent findings that suggest roles for HAND1 and HAND2 in human congenital heart disease.
Collapse
Affiliation(s)
- Joshua W. Vincentz
- Riley Heart Research Center, Wells Center for Pediatric Research, Division of Pediatric Cardiology, Departments of Anatomy, Biochemistry and Medical and Molecular Genetics, Indiana Medical School, 1044 W. Walnut St., Indianapolis, IN 46202-5225, USA
| | - Ralston M. Barnes
- Riley Heart Research Center, Wells Center for Pediatric Research, Division of Pediatric Cardiology, Departments of Anatomy, Biochemistry and Medical and Molecular Genetics, Indiana Medical School, 1044 W. Walnut St., Indianapolis, IN 46202-5225, USA
| | - Anthony B. Firulli
- Riley Heart Research Center, Wells Center for Pediatric Research, Division of Pediatric Cardiology, Departments of Anatomy, Biochemistry and Medical and Molecular Genetics, Indiana Medical School, 1044 W. Walnut St., Indianapolis, IN 46202-5225, USA
| |
Collapse
|