1
|
Zhang L, Huang R, Zhou H, Lin X, Guo F, Jing X, Zhang Y, Li F, Li F, Yu Q, Wang D, Chen G, Fu F, Pan M, Han J, Li D, Li R. Prenatal diagnosis in fetal right aortic arch using chromosomal microarray analysis and whole exome sequencing: a Chinese single-center retrospective study. Mol Cytogenet 2024; 17:22. [PMID: 39334424 PMCID: PMC11438370 DOI: 10.1186/s13039-024-00691-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 04/23/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Right aortic arch (RAA) is a common congenital aortic arch abnormality. Fetuses with RAA frequently have good outcomes after birth. However, chromosomal abnormalities and genetic syndromes suggest poor prognosis for these patients. So far the underlying genetic etiology is still not identified in most RAA patients based on traditional genetic techniques and a problem is still debated whether fetuses with isolated RAA should be referred for CMA. Our study aims to investigate the genetic etiology of fetuses with right aortic arch (RAA) by chromosomal microarray analysis (CMA) and whole exome sequencing (WES) and evaluate the efficacy of CMA in fetal isolated RAA. RESULTS Among these 153 fetuses, 99 (64.7%) with isolated RAA and 54 (35.3%) with non-isolated RAA; 25.5% (39/153) with additional intracardiac anomalies (ICA), and 19.0% (29/153) with extracardiac anomalies (ECA). Tetralogy of Fallot (n = 10) and persistent left superior vena cava (n = 11) are the most common ICA and ECA, respectively. CMA detected 15 clinically significant copy number variations (CNVs) in 14 cases (9.2%); microdeletion of 22q11.21 was the most common pathogenic CNVs (7.8%). The chromosomal abnormalities rate was higher in non-isolated RAA and RAA with ICA groups than in isolated RAA group (16.7% vs. 5.1%; 20% vs. 5.1%, both p < 0.05). From five cases further undergoing WES, a diagnostic variant in MTOR gene (c.7255G > A, de novo) was first reported in prenatal, extending the prenatal manifestation of Smith-Kingsmore syndrome (OMIM: 616638); a clinically relevant variant c.3407A > T in STAG2 was identified, being inherited from the healthy mother. Moreover, the premature birth and termination rates were higher in non-isolated RAA group than in isolated RAA group (11.1% vs. 1.0%; 37.0% vs. 2.0%, both p < 0.01). CONCLUSIONS We demonstrate that CMA and WES are useful diagnostic tools for fetal RAA, particularly non-isolated RAA, and all fetuses with RAA should be referred for CMA. The data probably aids in prenatal diagnosis and prenatal counseling of fetal RAA.
Collapse
Affiliation(s)
- Lu Zhang
- Prenatal Diagnostic Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510620, Guangdong, China
| | - Ruibin Huang
- Prenatal Diagnostic Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510620, Guangdong, China
| | - Hang Zhou
- Prenatal Diagnostic Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510620, Guangdong, China
| | - Xiaomei Lin
- Prenatal Diagnostic Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510620, Guangdong, China
| | - Fei Guo
- Prenatal Diagnostic Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510620, Guangdong, China
| | - Xiangyi Jing
- Prenatal Diagnostic Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510620, Guangdong, China
| | - Yongling Zhang
- Prenatal Diagnostic Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510620, Guangdong, China
| | - Fucheng Li
- Prenatal Diagnostic Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510620, Guangdong, China
| | - Fatao Li
- Prenatal Diagnostic Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510620, Guangdong, China
| | - Qiuxia Yu
- Prenatal Diagnostic Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510620, Guangdong, China
| | - Dan Wang
- Prenatal Diagnostic Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510620, Guangdong, China
| | - Guilan Chen
- Prenatal Diagnostic Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510620, Guangdong, China
| | - Fang Fu
- Prenatal Diagnostic Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510620, Guangdong, China
| | - Min Pan
- Prenatal Diagnostic Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510620, Guangdong, China
| | - Jin Han
- Prenatal Diagnostic Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510620, Guangdong, China
| | - Dongzhi Li
- Prenatal Diagnostic Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510620, Guangdong, China
| | - Ru Li
- Prenatal Diagnostic Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510620, Guangdong, China.
| |
Collapse
|
2
|
Oscorbin IP, Gordukova MA, Davydova NV, Zinovieva NV, Kovzel EF, Andries L, Kudlay DA, Filipenko ML. Multiplex droplet digital PCR for 22q11.2 microdeletions screening and DiGeorge syndrome diagnostics. Clin Chim Acta 2024; 563:119903. [PMID: 39127298 DOI: 10.1016/j.cca.2024.119903] [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: 05/31/2024] [Revised: 08/01/2024] [Accepted: 08/04/2024] [Indexed: 08/12/2024]
Abstract
BACKGROUND AND AIMS DiGeorge syndrome (DGS) is a genetic disorder manifesting in polymorphic symptoms related to developmental abnormalities of various organs including thymus. DGS is caused by microdeletions in the 22q11.2 region between several low copy repeats (LCR) occurring in approximately 1 in 4000 live births. Diagnosis of DGS relies on phenotypic examination, qPCR, ultrasound, FISH, MLPA and NGS which can be relatively inaccurate, time-consuming, and costly. MATERIALS AND METHODS A novel multiplex droplet digital PCR (ddPCR) assay was designed, optimized and validated for detection and mapping 22q11.2 microdeletions by simultaneous amplification of three targets - TUPLE1, ZNF74, D22S936 - within the deletion areas and one reference target - RPP30 - as an internal control. RESULTS The assay reliable identified microdeletions when the template concentration was >32 copies per reaction and successfully detected LCR22A-B, LCR22A-C, LCR22A-D, and LCR22B-C deletions in clinical samples from 153 patients with signs of immunodeficiency. In patients with the microdeletions, flow cytometry detected a significant increase in B-cell and natural killer cell counts and percentages, while T-cell percentages and T-cell receptor excision circle (TREC) numbers decreased. CONCLUSION The designed ddPCR assay is suitable for diagnosing DGS using whole blood and blood spots.
Collapse
Affiliation(s)
- Igor Petrovich Oscorbin
- Laboratory of Pharmacogenomics, The Institute of Chemical Biology and Fundamental Medicine of the Siberian Branch of the Russian Academy of Sciences (ICBFM SB RAS), Novosibirsk 630090, Russia.
| | | | | | | | - Elena Fedorovna Kovzel
- Clinical Immunology, Allergology, Pulmonology Program, Corporate Fund "University Medical Center" of Nazarbayev University, Astana, Kazakhstan
| | - Lucia Andries
- Laboratory of Clinical Immunology and Allergology, Nicolae Testemitanu State University of Medicine and Pharmacy of the Republic of Moldova, Chișinău, Moldova
| | - Dmitry Anatolyevich Kudlay
- The Department of Pharmacology, Faculty of Medicine, I.M. Sechenov First Moscow State Medical University, Pogodinskaya St. 1, Moscow 119991, Russia
| | - Maxim Leonidovich Filipenko
- Laboratory of Pharmacogenomics, The Institute of Chemical Biology and Fundamental Medicine of the Siberian Branch of the Russian Academy of Sciences (ICBFM SB RAS), Novosibirsk 630090, Russia
| |
Collapse
|
3
|
Kuenstner W, Rapisuwon S, Shobab L. DiGeorge Syndrome Diagnosed at Age 38: Challenges in Low-resource Settings and Implications of a Missed Diagnosis. JCEM CASE REPORTS 2024; 2:luae136. [PMID: 39049863 PMCID: PMC11267221 DOI: 10.1210/jcemcr/luae136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Indexed: 07/27/2024]
Abstract
22q11.2 deletion syndrome (22.q11.2 DS) is a genetic syndrome resulting from a microdeletion on chromosome 22. It has a diverse array of manifestations, and most cases are diagnosed early in childhood. We present the case of a 38-year-old female born in a developing country who presented to our clinic to establish care for a history of primary hypothyroidism. She was clinically and biochemically euthyroid on thyroid supplementation. She was also noted to have hypocalcemia in the setting of low PTH, for which the patient was previously prescribed calcitriol. Given a history of cleft palate, abnormal facial features, mild recurrent sinopulmonary infections, and her endocrine history (including short stature with height in the 6th percentile), genetic testing was obtained. She was diagnosed with a heterozygous whole gene deletion of the TBX1 gene. Additional genetic evaluation demonstrated a 2.6-Mb microdeleted segment of the 22a11.2 region encompassing 62 genes. The patient was referred to cardiology for evaluation of cardiac involvement given a history of tachyarrhythmia. This case highlights challenges in diagnosis and the implications of a delayed diagnosis of 22.q11.2 DS.
Collapse
Affiliation(s)
- William Kuenstner
- Division of Endocrinology, Department of Medicine, MedStar Georgetown University Hospital, Washington, DC 20007, USA
| | - Suthee Rapisuwon
- Division of Hematology and Oncology, Department of Medicine, MedStar Washington Hospital Center, Washington, DC 20010, USA
| | - Leila Shobab
- Division of Endocrinology, Department of Medicine, MedStar Washington Hospital Center, Washington, DC 20010, USA
| |
Collapse
|
4
|
Ramirez A, Vyzas CA, Zhao H, Eng K, Degenhardt K, Astrof S. Buffering Mechanism in Aortic Arch Artery Formation and Congenital Heart Disease. Circ Res 2024; 134:e112-e132. [PMID: 38618720 PMCID: PMC11081845 DOI: 10.1161/circresaha.123.322767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 03/27/2024] [Indexed: 04/16/2024]
Abstract
BACKGROUND The resiliency of embryonic development to genetic and environmental perturbations has been long appreciated; however, little is known about the mechanisms underlying the robustness of developmental processes. Aberrations resulting in neonatal lethality are exemplified by congenital heart disease arising from defective morphogenesis of pharyngeal arch arteries (PAAs) and their derivatives. METHODS Mouse genetics, lineage tracing, confocal microscopy, and quantitative image analyses were used to investigate mechanisms of PAA formation and repair. RESULTS The second heart field (SHF) gives rise to the PAA endothelium. Here, we show that the number of SHF-derived endothelial cells (ECs) is regulated by VEGFR2 (vascular endothelial growth factor receptor 2) and Tbx1. Remarkably, when the SHF-derived EC number is decreased, PAA development can be rescued by the compensatory endothelium. Blocking such compensatory response leads to embryonic demise. To determine the source of compensating ECs and mechanisms regulating their recruitment, we investigated 3-dimensional EC connectivity, EC fate, and gene expression. Our studies demonstrate that the expression of VEGFR2 by the SHF is required for the differentiation of SHF-derived cells into PAA ECs. The deletion of 1 VEGFR2 allele (VEGFR2SHF-HET) reduces SHF contribution to the PAA endothelium, while the deletion of both alleles (VEGFR2SHF-KO) abolishes it. The decrease in SHF-derived ECs in VEGFR2SHF-HET and VEGFR2SHF-KO embryos is complemented by the recruitment of ECs from the nearby veins. Compensatory ECs contribute to PAA derivatives, giving rise to the endothelium of the aortic arch and the ductus in VEGFR2SHF-KO mutants. Blocking the compensatory response in VEGFR2SHF-KO mutants results in embryonic lethality shortly after mid-gestation. The compensatory ECs are absent in Tbx1+/- embryos, a model for 22q11 deletion syndrome, leading to unpredictable arch artery morphogenesis and congenital heart disease. Tbx1 regulates the recruitment of the compensatory endothelium in an SHF-non-cell-autonomous manner. CONCLUSIONS Our studies uncover a novel buffering mechanism underlying the resiliency of PAA development and remodeling.
Collapse
Affiliation(s)
- AnnJosette Ramirez
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, 07103
- Multidisciplinary Ph.D. Program in Biomedical Sciences: Cell Biology, Neuroscience and Physiology Track, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, 07103
| | - Christina A. Vyzas
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, 07103
- Multidisciplinary Ph.D. Program in Biomedical Sciences: Cell Biology, Neuroscience and Physiology Track, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, 07103
| | - Huaning Zhao
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, 07103
| | - Kevin Eng
- Department of Statistics, Rutgers University, School of Arts and Sciences, Piscataway, NJ 08854
| | - Karl Degenhardt
- Children's Hospital of Pennsylvania, University of Pennsylvania, Philadelphia, PA 19107
| | - Sophie Astrof
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, 07103
- Multidisciplinary Ph.D. Program in Biomedical Sciences: Cell Biology, Neuroscience and Physiology Track, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, 07103
| |
Collapse
|
5
|
Stanton E, Sheridan S, Urata M, Chai Y. From Bedside to Bench and Back: Advancing Our Understanding of the Pathophysiology of Cleft Palate and Implications for the Future. Cleft Palate Craniofac J 2024; 61:759-773. [PMID: 36457208 DOI: 10.1177/10556656221142098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2024] Open
Abstract
OBJECTIVE To provide a comprehensive understanding of the pathophysiology of cleft palate (CP) and future perspectives. DESIGN Literature review. SETTING Setting varied across studies by level of care and geographical locations. INTERVENTIONS No interventions were performed. MAIN OUTCOME MEASURE(S) Primary outcome measures were to summarize our current understanding of palatogenesis in humans and animal models, the pathophysiology of CP, and potential future treatment modalities. RESULTS Animal research has provided considerable insight into the pathophysiology, molecular and cellular mechanisms of CP that have allowed for the development of novel treatment strategies. However, much work has yet to be done to connect our mouse model investigations and discoveries to CP in humans. The success of innovative strategies for tissue regeneration in mice provides promise for an exciting new avenue for improved and more targeted management of cleft care with precision medicine in patients. However, significant barriers to clinical translation remain. Among the most notable challenges include the differences in some aspects of palatogenesis and tissue repair between mice and humans, suggesting that potential therapies that have worked in animal models may not provide similar benefits to humans. CONCLUSIONS Increased translation of pathophysiological and tissue regeneration studies to clinical trials will bridge a wide gap in knowledge between animal models and human disease. By enhancing interaction between basic scientists and clinicians, and employing our animal model findings of disease mechanisms in concert with what we glean in the clinic, we can generate a more targeted and improved treatment algorithm for patients with CP.
Collapse
Affiliation(s)
- Eloise Stanton
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA, USA
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Samuel Sheridan
- Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA
| | - Mark Urata
- Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA
- Division of Plastic and Maxillofacial Surgery, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Yang Chai
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA, USA
- Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
6
|
Bremer SJ, Boxnick A, Glau L, Biermann D, Joosse SA, Thiele F, Billeb E, May J, Kolster M, Hackbusch R, Fortmann MI, Kozlik-Feldmann R, Hübler M, Tolosa E, Sachweh JS, Gieras A. Thymic Atrophy and Immune Dysregulation in Infants with Complex Congenital Heart Disease. J Clin Immunol 2024; 44:69. [PMID: 38393459 PMCID: PMC10891212 DOI: 10.1007/s10875-024-01662-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 01/24/2024] [Indexed: 02/25/2024]
Abstract
Congenital heart disease (CHD) is the most common birth defect, and up to 50% of infants with CHD require cardiovascular surgery early in life. Current clinical practice often involves thymus resection during cardiac surgery, detrimentally affecting T-cell immunity. However, epidemiological data indicate that CHD patients face an elevated risk for infections and immune-mediated diseases, independent of thymectomy. Hence, we examined whether the cardiac defect impacts thymus function in individuals with CHD. We investigated thymocyte development in 58 infants categorized by CHD complexity. To assess the relationship between CHD complexity and thymic function, we analyzed T-cell development, thymic output, and biomarkers linked to cardiac defects, stress, or inflammation. Patients with highly complex CHD exhibit thymic atrophy, resulting in low frequencies of recent thymic emigrants in peripheral blood, even prior to thymectomy. Elevated plasma cortisol levels were detected in all CHD patients, while high NT-proBNP and IL-6 levels were associated with thymic atrophy. Our findings reveal an association between complex CHD and thymic atrophy, resulting in reduced thymic output. Consequently, thymus preservation during cardiovascular surgery could significantly enhance immune function and the long-term health of CHD patients.
Collapse
Affiliation(s)
- Sarah-Jolan Bremer
- Department of Immunology, University Medical Center Hamburg-Eppendorf, N27, Martinistraße 52, 20246, Hamburg, Germany
- University Children's Research, UCR@Kinder-UKE, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Annika Boxnick
- Department of Immunology, University Medical Center Hamburg-Eppendorf, N27, Martinistraße 52, 20246, Hamburg, Germany
| | - Laura Glau
- Department of Immunology, University Medical Center Hamburg-Eppendorf, N27, Martinistraße 52, 20246, Hamburg, Germany
| | - Daniel Biermann
- Congenital and Pediatric Heart Surgery, Children's Heart Clinic, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Simon A Joosse
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Mildred Scheel Cancer Career Center HaTriCS4, University, Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Friederike Thiele
- Department of Immunology, University Medical Center Hamburg-Eppendorf, N27, Martinistraße 52, 20246, Hamburg, Germany
| | - Elena Billeb
- Department of Immunology, University Medical Center Hamburg-Eppendorf, N27, Martinistraße 52, 20246, Hamburg, Germany
- University Children's Research, UCR@Kinder-UKE, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jonathan May
- Department of Immunology, University Medical Center Hamburg-Eppendorf, N27, Martinistraße 52, 20246, Hamburg, Germany
| | - Manuela Kolster
- Department of Immunology, University Medical Center Hamburg-Eppendorf, N27, Martinistraße 52, 20246, Hamburg, Germany
| | - Romy Hackbusch
- Department of Immunology, University Medical Center Hamburg-Eppendorf, N27, Martinistraße 52, 20246, Hamburg, Germany
| | | | - Rainer Kozlik-Feldmann
- Department of Pediatric Cardiology, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Michael Hübler
- Congenital and Pediatric Heart Surgery, Children's Heart Clinic, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Eva Tolosa
- Department of Immunology, University Medical Center Hamburg-Eppendorf, N27, Martinistraße 52, 20246, Hamburg, Germany
| | - Jörg Siegmar Sachweh
- Congenital and Pediatric Heart Surgery, Children's Heart Clinic, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Anna Gieras
- Department of Immunology, University Medical Center Hamburg-Eppendorf, N27, Martinistraße 52, 20246, Hamburg, Germany.
| |
Collapse
|
7
|
Ramirez A, Vyzas CA, Zhao H, Eng K, Degenhardt K, Astrof S. Identification of novel buffering mechanisms in aortic arch artery development and congenital heart disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.03.02.530833. [PMID: 38370627 PMCID: PMC10871175 DOI: 10.1101/2023.03.02.530833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Rationale The resiliency of embryonic development to genetic and environmental perturbations has been long appreciated; however, little is known about the mechanisms underlying the robustness of developmental processes. Aberrations resulting in neonatal lethality are exemplified by congenital heart disease (CHD) arising from defective morphogenesis of pharyngeal arch arteries (PAA) and their derivatives. Objective To uncover mechanisms underlying the robustness of PAA morphogenesis. Methods and Results The second heart field (SHF) gives rise to the PAA endothelium. Here, we show that the number of SHF-derived ECs is regulated by VEGFR2 and Tbx1 . Remarkably, when SHF-derived EC number is decreased, PAA development can be rescued by the compensatory endothelium. Blocking such compensatory response leads to embryonic demise. To determine the source of compensating ECs and mechanisms regulating their recruitment, we investigated three-dimensional EC connectivity, EC fate, and gene expression. Our studies demonstrate that the expression of VEGFR2 by the SHF is required for the differentiation of SHF-derived cells into PAA ECs. The deletion of one VEGFR2 allele (VEGFR2 SHF-HET ) reduces SHF contribution to the PAA endothelium, while the deletion of both alleles (VEGFR2 SHF-KO ) abolishes it. The decrease in SHF-derived ECs in VEGFR2 SHF-HET and VEGFR2 SHF-KO embryos is complemented by the recruitment of ECs from the nearby veins. Compensatory ECs contribute to PAA derivatives, giving rise to the endothelium of the aortic arch and the ductus in VEGFR2 SHF-KO mutants. Blocking the compensatory response in VEGFR2 SHF-KO mutants results in embryonic lethality shortly after mid-gestation. The compensatory ECs are absent in Tbx1 +/- embryos, a model for 22q11 deletion syndrome, leading to unpredictable arch artery morphogenesis and CHD. Tbx1 regulates the recruitment of the compensatory endothelium in an SHF-non-cell-autonomous manner. Conclusions Our studies uncover a novel buffering mechanism underlying the resiliency of PAA development and remodeling. Nonstandard Abbreviations and Acronyms in Alphabetical Order CHD - congenital heart disease; ECs - endothelial cells; IAA-B - interrupted aortic arch type B; PAA - pharyngeal arch arteries; RERSA - retro-esophageal right subclavian artery; SHF - second heart field; VEGFR2 - Vascular endothelial growth factor receptor 2.
Collapse
|
8
|
Quilez S, Dumontier E, Baim C, Kam J, Cloutier JF. Loss of Neogenin alters branchial arch development and leads to craniofacial skeletal defects. Front Cell Dev Biol 2024; 12:1256465. [PMID: 38404688 PMCID: PMC10884240 DOI: 10.3389/fcell.2024.1256465] [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: 07/10/2023] [Accepted: 01/22/2024] [Indexed: 02/27/2024] Open
Abstract
The formation of complex structures, such as the craniofacial skeleton, requires precise and intricate two-way signalling between populations of cells of different embryonic origins. For example, the lower jaw, or mandible, arises from cranial neural crest cells (CNCCs) in the mandibular portion of the first branchial arch (mdBA1) of the embryo, and its development is regulated by signals from the ectoderm and cranial mesoderm (CM) within this structure. The molecular mechanisms underlying CM cell influence on CNCC development in the mdBA1 remain poorly defined. Herein we identified the receptor Neogenin as a key regulator of craniofacial development. We found that ablation of Neogenin expression via gene-targeting resulted in several craniofacial skeletal defects, including reduced size of the CNCC-derived mandible. Loss of Neogenin did not affect the formation of the mdBA1 CM core but resulted in altered Bmp4 and Fgf8 expression, increased apoptosis, and reduced osteoblast differentiation in the mdBA1 mesenchyme. Reduced BMP signalling in the mdBA1 of Neogenin mutant embryos was associated with alterations in the gene regulatory network, including decreased expression of transcription factors of the Hand, Msx, and Alx families, which play key roles in the patterning and outgrowth of the mdBA1. Tissue-specific Neogenin loss-of-function studies revealed that Neogenin expression in mesodermal cells contributes to mandible formation. Thus, our results identify Neogenin as a novel regulator of craniofacial skeletal formation and demonstrates it impinges on CNCC development via a non-cell autonomous mechanism.
Collapse
Affiliation(s)
- Sabrina Quilez
- The Neuro—Montreal Neurological Institute and Hospital, 3801 University, Montréal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada
| | - Emilie Dumontier
- The Neuro—Montreal Neurological Institute and Hospital, 3801 University, Montréal, QC, Canada
| | - Christopher Baim
- The Neuro—Montreal Neurological Institute and Hospital, 3801 University, Montréal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada
| | - Joseph Kam
- The Neuro—Montreal Neurological Institute and Hospital, 3801 University, Montréal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada
| | - Jean-François Cloutier
- The Neuro—Montreal Neurological Institute and Hospital, 3801 University, Montréal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada
- Department of Anatomy and Cell Biology, McGill University, Montréal, QC, Canada
| |
Collapse
|
9
|
Ghazanfar S, Guibentif C, Marioni JC. Stabilized mosaic single-cell data integration using unshared features. Nat Biotechnol 2024; 42:284-292. [PMID: 37231260 PMCID: PMC10869270 DOI: 10.1038/s41587-023-01766-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 03/28/2023] [Indexed: 05/27/2023]
Abstract
Currently available single-cell omics technologies capture many unique features with different biological information content. Data integration aims to place cells, captured with different technologies, onto a common embedding to facilitate downstream analytical tasks. Current horizontal data integration techniques use a set of common features, thereby ignoring non-overlapping features and losing information. Here we introduce StabMap, a mosaic data integration technique that stabilizes mapping of single-cell data by exploiting the non-overlapping features. StabMap first infers a mosaic data topology based on shared features, then projects all cells onto supervised or unsupervised reference coordinates by traversing shortest paths along the topology. We show that StabMap performs well in various simulation contexts, facilitates 'multi-hop' mosaic data integration where some datasets do not share any features and enables the use of spatial gene expression features for mapping dissociated single-cell data onto a spatial transcriptomic reference.
Collapse
Affiliation(s)
- Shila Ghazanfar
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK.
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, UK.
- School of Mathematics and Statistics, The University of Sydney, Camperdown, New South Wales, Australia.
- Charles Perkins Centre, The University of Sydney, Camperdown, New South Wales, Australia.
| | - Carolina Guibentif
- Sahlgrenska Center for Cancer Research, Inst. Biomedicine, Dept. Microbiology and Immunology, University of Gothenburg, Gothenburg, Sweden
| | - John C Marioni
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK.
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, UK.
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK.
| |
Collapse
|
10
|
Gill E, Bamforth SD. Molecular Pathways and Animal Models of Truncus Arteriosus. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1441:853-865. [PMID: 38884754 DOI: 10.1007/978-3-031-44087-8_52] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
In normal cardiovascular development in birds and mammals, the outflow tract of the heart is divided into two distinct channels to separate the oxygenated systemic blood flow from the deoxygenated pulmonary circulation. When the process of outflow tract septation fails, a single common outflow vessel persists resulting in a serious clinical condition known as persistent truncus arteriosus or common arterial trunk. In this chapter, we will review molecular pathways and the cells that are known to play a role in the formation and development of the outflow tract and how genetic manipulation of these pathways in animal models can result in common arterial trunk.
Collapse
Affiliation(s)
- Eleanor Gill
- Newcastle University Biosciences Institute, Newcastle, UK
| | | |
Collapse
|
11
|
Watanabe Y, Wang Y, Tanaka Y, Iwase A, Kawamura T, Saga Y, Yashiro K, Kurihara H, Nakagawa O. Hey2 enhancer activity defines unipotent progenitors for left ventricular cardiomyocytes in juxta-cardiac field of early mouse embryo. Proc Natl Acad Sci U S A 2023; 120:e2307658120. [PMID: 37669370 PMCID: PMC10500178 DOI: 10.1073/pnas.2307658120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 07/31/2023] [Indexed: 09/07/2023] Open
Abstract
The cardiac crescent is the first structure of the heart and contains progenitor cells of the first heart field, which primarily differentiate into left ventricular cardiomyocytes. The interface between the forming cardiac crescent and extraembryonic tissue is known as the juxta-cardiac field (JCF), and progenitor cells in this heart field contribute to the myocardium of the left ventricle and atrioventricular canal as well as the epicardium. However, it is unclear whether there are progenitor cells that differentiate specifically into left ventricular cardiomyocytes. We have previously demonstrated that an enhancer of the gene encoding the Hey2 bHLH transcriptional repressor is activated in the ventricular myocardium during mouse embryonic development. In this study, we aimed to investigate the characteristics of cardiomyocyte progenitor cells and their cell lineages by analyzing Hey2 enhancer activity at the earliest stages of heart formation. We found that the Hey2 enhancer initiated its activity prior to cardiomyocyte differentiation within the JCF. Hey2 enhancer-active cells were present rostrally to the Tbx5-expressing region at the early phase of cardiac crescent formation and differentiated exclusively into left ventricular cardiomyocytes in a lineage distinct from the Tbx5-positive lineage. By the late phase of cardiac crescent formation, Hey2 enhancer activity became significantly overlapped with Tbx5 expression in cells that contribute to the left ventricular myocardium. Our study reveals that a population of unipotent progenitor cells for left ventricular cardiomyocytes emerge in the JCF, providing further insight into the mode of cell type diversification during early cardiac development.
Collapse
Affiliation(s)
- Yusuke Watanabe
- Department of Molecular Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka564-8565, Japan
| | - Yunce Wang
- Department of Molecular Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka564-8565, Japan
- Laboratory of Stem Cell & Regenerative Medicine, Department of Biomedical Sciences, College of Life Sciences, Ritsumeikan University, Kusatsu, Shiga525-8577, Japan
| | - Yuki Tanaka
- Department of Molecular Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka564-8565, Japan
- Laboratory of Stem Cell & Regenerative Medicine, Department of Biomedical Sciences, College of Life Sciences, Ritsumeikan University, Kusatsu, Shiga525-8577, Japan
| | - Akiyasu Iwase
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo113-0033, Japan
| | - Teruhisa Kawamura
- Laboratory of Stem Cell & Regenerative Medicine, Department of Biomedical Sciences, College of Life Sciences, Ritsumeikan University, Kusatsu, Shiga525-8577, Japan
| | - Yumiko Saga
- Mammalian Development Laboratory, Department of Gene Function and Phenomics, National Institute of Genetics, Mishima, Shizuoka411-8582, Japan
| | - Kenta Yashiro
- Division of Anatomy and Developmental Biology, Department of Anatomy, Kyoto Prefectural University of Medicine, Kamigyo, Kyoto602-8566, Japan
| | - Hiroki Kurihara
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo113-0033, Japan
| | - Osamu Nakagawa
- Department of Molecular Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka564-8565, Japan
| |
Collapse
|
12
|
Doering L, Cornean A, Thumberger T, Benjaminsen J, Wittbrodt B, Kellner T, Hammouda OT, Gorenflo M, Wittbrodt J, Gierten J. CRISPR-based knockout and base editing confirm the role of MYRF in heart development and congenital heart disease. Dis Model Mech 2023; 16:dmm049811. [PMID: 37584388 PMCID: PMC10445736 DOI: 10.1242/dmm.049811] [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: 08/01/2022] [Accepted: 07/21/2023] [Indexed: 08/17/2023] Open
Abstract
High-throughput DNA sequencing studies increasingly associate DNA variants with congenital heart disease (CHD). However, functional modeling is a crucial prerequisite for translating genomic data into clinical care. We used CRISPR-Cas9-mediated targeting of 12 candidate genes in the vertebrate model medaka (Oryzias latipes), five of which displayed a novel cardiovascular phenotype spectrum in F0 (crispants): mapre2, smg7, cdc42bpab, ankrd11 and myrf, encoding a transcription factor recently linked to cardiac-urogenital syndrome. Our myrf mutant line showed particularly prominent embryonic cardiac defects recapitulating phenotypes of pediatric patients, including hypoplastic ventricle. Mimicking human mutations, we edited three sites to generate specific myrf single-nucleotide variants via cytosine and adenine base editors. The Glu749Lys missense mutation in the conserved intramolecular chaperon autocleavage domain fully recapitulated the characteristic myrf mutant phenotype with high penetrance, underlining the crucial function of this protein domain. The efficiency and scalability of base editing to model specific point mutations accelerate gene validation studies and the generation of human-relevant disease models.
Collapse
Affiliation(s)
- Lino Doering
- Centre for Organismal Studies, Heidelberg University, 69120 Heidelberg, Germany
- Department of Pediatric Cardiology, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Alex Cornean
- Centre for Organismal Studies, Heidelberg University, 69120 Heidelberg, Germany
- Heidelberg Biosciences International Graduate School, Heidelberg University, 69120 Heidelberg, Germany
| | - Thomas Thumberger
- Centre for Organismal Studies, Heidelberg University, 69120 Heidelberg, Germany
| | - Joergen Benjaminsen
- Centre for Organismal Studies, Heidelberg University, 69120 Heidelberg, Germany
| | - Beate Wittbrodt
- Centre for Organismal Studies, Heidelberg University, 69120 Heidelberg, Germany
| | - Tanja Kellner
- Centre for Organismal Studies, Heidelberg University, 69120 Heidelberg, Germany
| | - Omar T. Hammouda
- Centre for Organismal Studies, Heidelberg University, 69120 Heidelberg, Germany
| | - Matthias Gorenflo
- Department of Pediatric Cardiology, University Hospital Heidelberg, 69120 Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, 69120 Heidelberg, Germany
| | - Joachim Wittbrodt
- Centre for Organismal Studies, Heidelberg University, 69120 Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, 69120 Heidelberg, Germany
| | - Jakob Gierten
- Centre for Organismal Studies, Heidelberg University, 69120 Heidelberg, Germany
- Department of Pediatric Cardiology, University Hospital Heidelberg, 69120 Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, 69120 Heidelberg, Germany
| |
Collapse
|
13
|
Zbasnik N, Fish JL. Fgf8 regulates first pharyngeal arch segmentation through pouch-cleft interactions. Front Cell Dev Biol 2023; 11:1186526. [PMID: 37287454 PMCID: PMC10242020 DOI: 10.3389/fcell.2023.1186526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 05/09/2023] [Indexed: 06/09/2023] Open
Abstract
Introduction: The pharyngeal arches are transient developmental structures that, in vertebrates, give rise to tissues of the head and neck. A critical process underlying the specification of distinct arch derivatives is segmentation of the arches along the anterior-posterior axis. Formation of ectodermal-endodermal interfaces is a key mediator of this process, and although it is essential, mechanisms regulating the establishment of these interfaces vary between pouches and between taxa. Methods: Here, we focus on the patterning and morphogenesis of epithelia associated with the first pharyngeal arch, the first pharyngeal pouch (pp1) and the first pharyngeal cleft (pc1), and the role of Fgf8 dosage in these processes in the mouse model system. Results: We find that severe reductions of Fgf8 levels disrupt both pp1 and pc1 development. Notably, out-pocketing of pp1 is largely robust to Fgf8 reductions, however, pp1 extension along the proximal-distal axis fails when Fgf8 is low. Our data indicate that Fgf8 is required for specification of regional identity in both pp1 and pc1, for localized changes in cell polarity, and for elongation and extension of both pp1 and pc1. Discussion: Based on Fgf8-mediated changes in tissue relationships between pp1 and pc1, we hypothesize that extension of pp1 requires physical interaction with pc1. Overall, our data indicate a critical role for the lateral surface ectoderm in segmentation of the first pharyngeal arch that has previously been under-appreciated.
Collapse
|
14
|
De Bono C, Liu Y, Ferrena A, Valentine A, Zheng D, Morrow BE. Single-cell transcriptomics uncovers a non-autonomous Tbx1-dependent genetic program controlling cardiac neural crest cell development. Nat Commun 2023; 14:1551. [PMID: 36941249 PMCID: PMC10027855 DOI: 10.1038/s41467-023-37015-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 02/28/2023] [Indexed: 03/23/2023] Open
Abstract
Disruption of cardiac neural crest cells (CNCCs) results in congenital heart disease, yet we do not understand the cell fate dynamics as these cells differentiate to vascular smooth muscle cells. Here we performed single-cell RNA-sequencing of NCCs from the pharyngeal apparatus with the heart in control mouse embryos and when Tbx1, the gene for 22q11.2 deletion syndrome, is inactivated. We uncover three dynamic transitions of pharyngeal NCCs expressing Tbx2 and Tbx3 through differentiated CNCCs expressing cardiac transcription factors with smooth muscle genes. These transitions are altered non-autonomously by loss of Tbx1. Further, inactivation of Tbx2 and Tbx3 in early CNCCs results in aortic arch branching defects due to failed smooth muscle differentiation. Loss of Tbx1 interrupts mesoderm to CNCC cell-cell communication with upregulation and premature activation of BMP signaling and reduced MAPK signaling, as well as alteration of other signaling, and failed dynamic transitions of CNCCs leading to disruption of aortic arch artery formation and cardiac outflow tract septation.
Collapse
Affiliation(s)
- Christopher De Bono
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA.
| | - Yang Liu
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Alexander Ferrena
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
- Institute for Clinical and Translational Research, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Aneesa Valentine
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Deyou Zheng
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Neurology, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Bernice E Morrow
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA.
- Departments of Obstetrics and Gynecology; and Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
15
|
Zbasnik N, Fish JL. Fgf8 regulates first pharyngeal arch segmentation through pouch-cleft interactions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.15.532781. [PMID: 36993764 PMCID: PMC10055162 DOI: 10.1101/2023.03.15.532781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
The pharyngeal arches are transient developmental structures that, in vertebrates, give rise to tissues of the head and neck. A critical process underlying the specification of distinct arch derivatives is segmentation of the arches along the anterior-posterior axis. Out-pocketing of the pharyngeal endoderm between the arches is a key mediator of this process, and although it is essential, mechanisms regulating out-pocketing vary between pouches and between taxa. Here, we focus on the patterning and morphogenesis of epithelia associated with the first pharyngeal arch, the first pharyngeal pouch (pp1) and the first pharyngeal cleft (pc1), and the role of Fgf8 dosage in these processes. We find that severe reductions of Fgf8 levels disrupt both pp1 and pc1 development. Notably, out-pocketing of pp1 is largely robust to Fgf8 reductions, however, pp1 extension along the proximal-distal axis fails when Fgf8 is low. Our data indicate that extension of pp1 requires physical interaction with pc1, and that multiple aspects of pc1 morphogenesis require Fgf8 . In particular, Fgf8 is required for specification of regional identity in both pp1 and pc1, for localized changes in cell polarity, and for elongation and extension of both pp1 and pc1. Overall, our data indicate a critical role for the lateral surface ectoderm in segmentation of the first pharyngeal arch that has previously been under-appreciated.
Collapse
|
16
|
Cen H, Luo H, Luo B, Fan P, Zhang Y, Zhang Y. TBX1 regulates myogenic differentiation by activating the TGFβ-Smad2/3 pathway in myoblasts. Exp Biol Med (Maywood) 2023; 248:61-69. [PMID: 36036218 PMCID: PMC9989151 DOI: 10.1177/15353702221112087] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
TBX1 is systematically conserved in the T-box transcription factor family and regulates craniofacial muscle development during various stages of myogenesis, including commitment, proliferation, terminal differentiation, and survival. However, the role and mechanism by which TBX1 regulates the myogenic development of myoblasts remains unclear. In our study, we overexpressed TBX1 in mouse C2C12 myoblasts using a lentivirus method. We found that TBX1 inhibited cell proliferation and muscle differentiation, which had no effect on apoptosis. During myogenic differentiation, we also found that TBX1 overexpressing cells regulate myogenic differentiation by upregulating the expression levels of Smad2 and Smad3 and downregulating the expression level of MEF2C. After treatment with a specific inhibitor of Smad3 (SIS3), the myogenic differentiation of wild-type and TBX1 overexpressing cells increased. Thus, TBX1 may regulate myoblast muscle differentiation by enhancing the expression of Smad2 and Smad3. TBX1 may be a therapeutic target for muscular dystrophy.
Collapse
Affiliation(s)
- Haimei Cen
- Department of Neurology, The First Affiliated Hospital, Jinan University, Guangzhou 510630, China
| | - Hong Luo
- Department of Neurology, The First Affiliated Hospital, Jinan University, Guangzhou 510630, China
| | - Bin Luo
- Department of Neurology, The First Affiliated Hospital, Jinan University, Guangzhou 510630, China
| | - Pin Fan
- Department of Neurology, The First Affiliated Hospital, Jinan University, Guangzhou 510630, China
| | - Yusheng Zhang
- Department of Neurology, The First Affiliated Hospital, Jinan University, Guangzhou 510630, China
| | - Yu Zhang
- Department of Neurology, The First Affiliated Hospital, Jinan University, Guangzhou 510630, China
| |
Collapse
|
17
|
Hankey PB, Ghulmiyyah J, Yeh HW, Tracy M, Arganbright J. Airway anomalies in patients with 22q11.2 deletion syndrome: A scoping review. Int J Pediatr Otorhinolaryngol 2022; 163:111373. [PMID: 36335759 DOI: 10.1016/j.ijporl.2022.111373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 10/22/2022] [Accepted: 10/30/2022] [Indexed: 11/06/2022]
Abstract
OBJECTIVE People with 22q11.2 deletion syndrome (22q11DS) can present with a wide variety of findings. Various airway anomalies have been described intermittently within this syndrome, but this feature has not been extensively investigated. Increased provider awareness of these findings may help guide clinical decision-making and improve overall patient outcomes. The objectives of this review are to identify the types of airway anomalies in 22q11DS and the prevalence of airway anomalies within symptomatic individuals. METHODS PubMed/MEDLINE, Cochrane Library, and EMBASE databases were searched in February 2022 for all available articles. Search terms included those that described 22q11DS or one of its synonymous conditions AND those that described airway anatomy and anomalies. The term airway anomaly was defined as any structural aberration in the conductive airway from the oral or nasal vestibule region to the mainstem bronchus. Studies were screened by two authors. A review of references was conducted. Eligible manuscripts underwent full-text review for quality appraisal and data extraction. RESULTS From a total of 909 unique manuscripts, 58 studies were selected, describing 328 people. The prevalence of airway anomaly diagnosis within symptomatic individuals ranged from 14% to 74%. Twenty-two unique airway anomalies were described. Laryngeal web was the most frequently described anomaly, followed by airway malacia and subglottic stenosis. Laryngeal web was 40% sensitive for suggesting a diagnosis of 22q11DS. Among affected individuals, as many as 46% had multiple concomitant airway anomalies. Aside from respiratory symptoms, other features that prompted airway evaluation included difficult intubation or failed extubation. CONCLUSION The findings within this review support the notion that a wide variety of airway anomalies may be seen in people with 22q11DS and that these findings have been discovered frequently in those with airway symptoms. Providers should maintain a low threshold to perform an airway examination in those with 22q11DS, especially when airway symptoms are present.
Collapse
Affiliation(s)
- Paul Bryan Hankey
- Kansas City University College of Osteopathic Medicine, 1750 Independence Ave, Kansas City, MO, 64106, USA; Children's Mercy Hospital, Division of Otolaryngology, 2401 Gillham Road, Kansas City, MO, 64108, USA.
| | - Jana Ghulmiyyah
- Children's Mercy Hospital, Division of Otolaryngology, 2401 Gillham Road, Kansas City, MO, 64108, USA.
| | - Hung-Wen Yeh
- Children's Mercy Hospital, Division of Otolaryngology, 2401 Gillham Road, Kansas City, MO, 64108, USA.
| | - Meghan Tracy
- Children's Mercy Hospital, Division of Otolaryngology, 2401 Gillham Road, Kansas City, MO, 64108, USA.
| | - Jill Arganbright
- Children's Mercy Hospital, Division of Otolaryngology, 2401 Gillham Road, Kansas City, MO, 64108, USA.
| |
Collapse
|
18
|
Mannstadt M, Cianferotti L, Gafni RI, Giusti F, Kemp EH, Koch CA, Roszko KL, Yao L, Guyatt GH, Thakker RV, Xia W, Brandi ML. Hypoparathyroidism: Genetics and Diagnosis. J Bone Miner Res 2022; 37:2615-2629. [PMID: 36375809 DOI: 10.1002/jbmr.4667] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 07/22/2022] [Accepted: 07/31/2022] [Indexed: 01/05/2023]
Abstract
This narrative report summarizes diagnostic criteria for hypoparathyroidism and describes the clinical presentation and underlying genetic causes of the nonsurgical forms. We conducted a comprehensive literature search from January 2000 to January 2021 and included landmark articles before 2000, presenting a comprehensive update of these topics and suggesting a research agenda to improve diagnosis and, eventually, the prognosis of the disease. Hypoparathyroidism, which is characterized by insufficient secretion of parathyroid hormone (PTH) leading to hypocalcemia, is diagnosed on biochemical grounds. Low albumin-adjusted calcium or ionized calcium with concurrent inappropriately low serum PTH concentration are the hallmarks of the disease. In this review, we discuss the characteristics and pitfalls in measuring calcium and PTH. We also undertook a systematic review addressing the utility of measuring calcium and PTH within 24 hours after total thyroidectomy to predict long-term hypoparathyroidism. A summary of the findings is presented here; results of the detailed systematic review are published separately in this issue of JBMR. Several genetic disorders can present with hypoparathyroidism, either as an isolated disease or as part of a syndrome. A positive family history and, in the case of complex diseases, characteristic comorbidities raise the clinical suspicion of a genetic disorder. In addition to these disorders' phenotypic characteristics, which include autoimmune diseases, we discuss approaches for the genetic diagnosis. © 2022 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Michael Mannstadt
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Luisella Cianferotti
- Bone Metabolic Diseases Unit, Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Rachel I Gafni
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | | | | | - Christian A Koch
- Department of Medicine/Endocrinology, Fox Chase Cancer Center, Philadelphia, PA, USA.,Department of Medicine/Endocrinology, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Kelly L Roszko
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Liam Yao
- Department of Health Research Methods, Evidence, and Impact, and Department of Medicine, McMaster University, Hamilton, Canada
| | - Gordon H Guyatt
- Department of Health Research Methods, Evidence, and Impact, and Department of Medicine, McMaster University, Hamilton, Canada
| | - Rajesh V Thakker
- Academic Endocrine Unit, Radcliffe Department of Medicine, University of Oxford, Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), Churchill Hospital, Headington, Oxford, UK.,Oxford National Institute for Health Research (NIHR) Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, UK
| | - Weibo Xia
- Department of Endocrinology, Peking Union Medical Collage Hospital, Beijing, China
| | - Maria-Luisa Brandi
- Fondazione Italiana sulla Ricerca sulle Malattie dell'Osso (F.I.R.M.O. Foundation), Florence, Italy
| |
Collapse
|
19
|
Bhalla P, Du Q, Kumar A, Xing C, Moses A, Dozmorov I, Wysocki CA, Cleaver OB, Pirolli TJ, Markert ML, de la Morena MT, Baldini A, van Oers NS. Mesenchymal cell replacement corrects thymic hypoplasia in murine models of 22q11.2 deletion syndrome. J Clin Invest 2022; 132:e160101. [PMID: 36136514 PMCID: PMC9663160 DOI: 10.1172/jci160101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 09/21/2022] [Indexed: 11/17/2022] Open
Abstract
22q11.2 deletion syndrome (22q11.2DS) is the most common human chromosomal microdeletion, causing developmentally linked congenital malformations, thymic hypoplasia, hypoparathyroidism, and/or cardiac defects. Thymic hypoplasia leads to T cell lymphopenia, which most often results in mild SCID. Despite decades of research, the molecular underpinnings leading to thymic hypoplasia in 22q11.2DS remain unknown. Comparison of embryonic thymuses from mouse models of 22q11.2DS (Tbx1neo2/neo2) revealed proportions of mesenchymal, epithelial, and hematopoietic cell types similar to those of control thymuses. Yet, the small thymuses were growth restricted in fetal organ cultures. Replacement of Tbx1neo2/neo2 thymic mesenchymal cells with normal ones restored tissue growth. Comparative single-cell RNA-Seq of embryonic thymuses uncovered 17 distinct cell subsets, with transcriptome differences predominant in the 5 mesenchymal subsets from the Tbx1neo2/neo2 cell line. The transcripts affected included those for extracellular matrix proteins, consistent with the increased collagen deposition we observed in the small thymuses. Attenuating collagen cross-links with minoxidil restored thymic tissue expansion for hypoplastic lobes. In colony-forming assays, the Tbx1neo2/neo2-derived mesenchymal cells had reduced expansion potential, in contrast to the normal growth of thymic epithelial cells. These findings suggest that mesenchymal cells were causal to the small embryonic thymuses in the 22q11.2DS mouse models, which was correctable by substitution with normal mesenchyme.
Collapse
Affiliation(s)
| | | | - Ashwani Kumar
- Eugene McDermott Center for Human Growth and Development
| | - Chao Xing
- Eugene McDermott Center for Human Growth and Development
- Departments of Bioinformatics and
- Population and Data Sciences, Departments of
| | | | | | | | | | - Timothy J. Pirolli
- Division of Pediatric Cardiothoracic Surgery, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Mary Louise Markert
- Departments of Pediatrics and Immunology, Duke University Medical Center, Durham, North Carolina, USA
| | - Maria Teresa de la Morena
- Division of Immunology, Department of Pediatrics, University of Washington, and Seattle Children’s Hospital, Seattle, Washington, USA
| | - Antonio Baldini
- Department Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Nicolai S.C. van Oers
- Department of Immunology
- Pediatrics
- Department of Microbiology, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
20
|
Lania G, Franzese M, Noritaka A, Bilio M, Flore G, Russo A, D'Agostino E, Angelini C, Kelly RG, Baldini A. A phenotypic rescue approach identifies lineage regionalization defects in a mouse model of DiGeorge syndrome. Dis Model Mech 2022; 15:276264. [PMID: 35946435 PMCID: PMC9555768 DOI: 10.1242/dmm.049415] [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: 12/04/2021] [Accepted: 07/26/2022] [Indexed: 11/24/2022] Open
Abstract
TBX1 is a key regulator of pharyngeal apparatus (PhAp) development. Vitamin B12 (vB12) treatment partially rescues aortic arch patterning defects of Tbx1+/− embryos. Here, we show that it also improves cardiac outflow tract septation and branchiomeric muscle anomalies of Tbx1 hypomorphic mutants. At the molecular level, in vivo vB12 treatment enabled us to identify genes that were dysregulated by Tbx1 haploinsufficiency and rescued by treatment. We found that SNAI2, also known as SLUG, encoded by the rescued gene Snai2, identified a population of mesodermal cells that was partially overlapping with, but distinct from, ISL1+ and TBX1+ populations. In addition, SNAI2+ cells were mislocalized and had a greater tendency to aggregate in Tbx1+/− and Tbx1−/− embryos, and vB12 treatment restored cellular distribution. Adjacent neural crest-derived mesenchymal cells, which do not express TBX1, were also affected, showing enhanced segregation from cardiopharyngeal mesodermal cells. We propose that TBX1 regulates cell distribution in the core mesoderm and the arrangement of multiple lineages within the PhAp.
Collapse
Affiliation(s)
- Gabriella Lania
- Institute of Genetics and Biophysics, National Research Council (CNR), Naples, Italy
| | - Monica Franzese
- Istituto per le Applicazione del Calcolo, National Research Council (CNR), Naples, Italy.,IRCCS SDN, Naples, Italy
| | - Adachi Noritaka
- Aix-Marseille Université, CNRS UMR 7288, IBDM, Marseille, France
| | - Marchesa Bilio
- Institute of Genetics and Biophysics, National Research Council (CNR), Naples, Italy
| | - Gemma Flore
- Institute of Genetics and Biophysics, National Research Council (CNR), Naples, Italy
| | - Annalaura Russo
- Institute of Genetics and Biophysics, National Research Council (CNR), Naples, Italy
| | - Erika D'Agostino
- Institute of Genetics and Biophysics, National Research Council (CNR), Naples, Italy
| | - Claudia Angelini
- Istituto per le Applicazione del Calcolo, National Research Council (CNR), Naples, Italy
| | - Robert G Kelly
- Aix-Marseille Université, CNRS UMR 7288, IBDM, Marseille, France
| | - Antonio Baldini
- Institute of Genetics and Biophysics, National Research Council (CNR), Naples, Italy.,Department of Molecular Medicine and Medical Biotechnology, University Federico II, Naples, Italy
| |
Collapse
|
21
|
Jeon H, Jin S, Choe CP. inka1b expression in the head mesoderm is dispensable for facial cartilage development. Gene Expr Patterns 2022; 45:119262. [PMID: 35811016 DOI: 10.1016/j.gep.2022.119262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 07/04/2022] [Accepted: 07/05/2022] [Indexed: 11/16/2022]
Abstract
Inka box actin regulator 1 (Inka1) is a novel protein identified in Xenopus and is found in vertebrates. While Inka1 is required for facial skeletal development in Xenopus and zebrafish, it is dispensable in mice despite its conserved expression in the cranial neural crest, indicating that Inka1 function in facial skeletal development is not conserved among vertebrates. Zebrafish bears two paralogs of inka1 (inka1a and inka1b) in the genome, with the biological roles of inka1b barely known. Here, we analyzed the expression and function of inka1b during facial skeletal development in zebrafish. inka1b was expressed sequentially in the head mesoderm adjacent to the pharyngeal pouches essential for facial skeletal development at the stage of arch segmentation. However, a loss-of-function mutation in inka1b displayed normal head development, including the pouches and facial cartilages. The normal head of inka1b mutant fish was unlikely a result of the genetic redundancy of inka1b with inka1a, given the distinct expression of inka1a and inka1b in the cranial neural crest and head mesoderm, respectively, during craniofacial development. Our findings suggest that the inka1b expression in the head mesoderm might not be essential for head development in zebrafish.
Collapse
Affiliation(s)
- Haewon Jeon
- Division of Applied Life Science, Plant Molecular Biology and Biotechnology Research Center, Gyeongsang National University, Jinju, 52828, South Korea
| | - Sil Jin
- Division of Applied Life Science, Plant Molecular Biology and Biotechnology Research Center, Gyeongsang National University, Jinju, 52828, South Korea
| | - Chong Pyo Choe
- Division of Applied Life Science, Plant Molecular Biology and Biotechnology Research Center, Gyeongsang National University, Jinju, 52828, South Korea; Division of Life Science, Gyeongsang National University, Jinju, 52828, South Korea.
| |
Collapse
|
22
|
Funato N. Craniofacial Phenotypes and Genetics of DiGeorge Syndrome. J Dev Biol 2022; 10:jdb10020018. [PMID: 35645294 PMCID: PMC9149807 DOI: 10.3390/jdb10020018] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/11/2022] [Accepted: 05/11/2022] [Indexed: 02/06/2023] Open
Abstract
The 22q11.2 deletion is one of the most common genetic microdeletions, affecting approximately 1 in 4000 live births in humans. A 1.5 to 2.5 Mb hemizygous deletion of chromosome 22q11.2 causes DiGeorge syndrome (DGS) and velocardiofacial syndrome (VCFS). DGS/VCFS are associated with prevalent cardiac malformations, thymic and parathyroid hypoplasia, and craniofacial defects. Patients with DGS/VCFS manifest craniofacial anomalies involving the cranium, cranial base, jaws, pharyngeal muscles, ear-nose-throat, palate, teeth, and cervical spine. Most craniofacial phenotypes of DGS/VCFS are caused by proximal 1.5 Mb microdeletions, resulting in a hemizygosity of coding genes, microRNAs, and long noncoding RNAs. TBX1, located on chromosome 22q11.21, encodes a T-box transcription factor and is a candidate gene for DGS/VCFS. TBX1 regulates the fate of progenitor cells in the cranial and pharyngeal apparatus during embryogenesis. Tbx1-null mice exhibit the most clinical features of DGS/VCFS, including craniofacial phenotypes. Despite the frequency of DGS/VCFS, there has been a limited review of the craniofacial phenotypes of DGC/VCFS. This review focuses on these phenotypes and summarizes the current understanding of the genetic factors that impact DGS/VCFS-related phenotypes. We also review DGS/VCFS mouse models that have been designed to better understand the pathogenic processes of DGS/VCFS.
Collapse
Affiliation(s)
- Noriko Funato
- Department of Signal Gene Regulation, Advanced Therapeutic Sciences, Medical and Dental Sciences, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo 113-8510, Japan
| |
Collapse
|
23
|
Lozano-Velasco E, Garcia-Padilla C, del Mar Muñoz-Gallardo M, Martinez-Amaro FJ, Caño-Carrillo S, Castillo-Casas JM, Sanchez-Fernandez C, Aranega AE, Franco D. Post-Transcriptional Regulation of Molecular Determinants during Cardiogenesis. Int J Mol Sci 2022; 23:ijms23052839. [PMID: 35269981 PMCID: PMC8911333 DOI: 10.3390/ijms23052839] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/19/2022] [Accepted: 02/26/2022] [Indexed: 12/15/2022] Open
Abstract
Cardiovascular development is initiated soon after gastrulation as bilateral precardiac mesoderm is progressively symmetrically determined at both sides of the developing embryo. The precardiac mesoderm subsequently fused at the embryonic midline constituting an embryonic linear heart tube. As development progress, the embryonic heart displays the first sign of left-right asymmetric morphology by the invariably rightward looping of the initial heart tube and prospective embryonic ventricular and atrial chambers emerged. As cardiac development progresses, the atrial and ventricular chambers enlarged and distinct left and right compartments emerge as consequence of the formation of the interatrial and interventricular septa, respectively. The last steps of cardiac morphogenesis are represented by the completion of atrial and ventricular septation, resulting in the configuration of a double circuitry with distinct systemic and pulmonary chambers, each of them with distinct inlets and outlets connections. Over the last decade, our understanding of the contribution of multiple growth factor signaling cascades such as Tgf-beta, Bmp and Wnt signaling as well as of transcriptional regulators to cardiac morphogenesis have greatly enlarged. Recently, a novel layer of complexity has emerged with the discovery of non-coding RNAs, particularly microRNAs and lncRNAs. Herein, we provide a state-of-the-art review of the contribution of non-coding RNAs during cardiac development. microRNAs and lncRNAs have been reported to functional modulate all stages of cardiac morphogenesis, spanning from lateral plate mesoderm formation to outflow tract septation, by modulating major growth factor signaling pathways as well as those transcriptional regulators involved in cardiac development.
Collapse
Affiliation(s)
- Estefania Lozano-Velasco
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain; (E.L.-V.); (C.G.-P.); (M.d.M.M.-G.); (F.J.M.-A.); (S.C.-C.); (J.M.C.-C.); (C.S.-F.); (A.E.A.)
- Fundación Medina, 18007 Granada, Spain
| | - Carlos Garcia-Padilla
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain; (E.L.-V.); (C.G.-P.); (M.d.M.M.-G.); (F.J.M.-A.); (S.C.-C.); (J.M.C.-C.); (C.S.-F.); (A.E.A.)
- Department of Anatomy, Embryology and Zoology, School of Medicine, University of Extremadura, 06006 Badajoz, Spain
| | - Maria del Mar Muñoz-Gallardo
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain; (E.L.-V.); (C.G.-P.); (M.d.M.M.-G.); (F.J.M.-A.); (S.C.-C.); (J.M.C.-C.); (C.S.-F.); (A.E.A.)
| | - Francisco Jose Martinez-Amaro
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain; (E.L.-V.); (C.G.-P.); (M.d.M.M.-G.); (F.J.M.-A.); (S.C.-C.); (J.M.C.-C.); (C.S.-F.); (A.E.A.)
| | - Sheila Caño-Carrillo
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain; (E.L.-V.); (C.G.-P.); (M.d.M.M.-G.); (F.J.M.-A.); (S.C.-C.); (J.M.C.-C.); (C.S.-F.); (A.E.A.)
| | - Juan Manuel Castillo-Casas
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain; (E.L.-V.); (C.G.-P.); (M.d.M.M.-G.); (F.J.M.-A.); (S.C.-C.); (J.M.C.-C.); (C.S.-F.); (A.E.A.)
| | - Cristina Sanchez-Fernandez
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain; (E.L.-V.); (C.G.-P.); (M.d.M.M.-G.); (F.J.M.-A.); (S.C.-C.); (J.M.C.-C.); (C.S.-F.); (A.E.A.)
- Fundación Medina, 18007 Granada, Spain
| | - Amelia E. Aranega
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain; (E.L.-V.); (C.G.-P.); (M.d.M.M.-G.); (F.J.M.-A.); (S.C.-C.); (J.M.C.-C.); (C.S.-F.); (A.E.A.)
- Fundación Medina, 18007 Granada, Spain
| | - Diego Franco
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain; (E.L.-V.); (C.G.-P.); (M.d.M.M.-G.); (F.J.M.-A.); (S.C.-C.); (J.M.C.-C.); (C.S.-F.); (A.E.A.)
- Fundación Medina, 18007 Granada, Spain
- Correspondence:
| |
Collapse
|
24
|
Palmquist-Gomes P, Meilhac SM. Shaping the mouse heart tube from the second heart field epithelium. Curr Opin Genet Dev 2022; 73:101896. [PMID: 35026527 DOI: 10.1016/j.gde.2021.101896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 12/12/2021] [Accepted: 12/15/2021] [Indexed: 11/03/2022]
Abstract
As other tubular organs, the embryonic heart develops from an epithelial sheet of cells, referred to as the heart field. The second heart field, which lies in the dorsal pericardial wall, constitutes a transient cell reservoir, integrating patterning and polarity cues. Conditional mutants have shown that impairment of the epithelial architecture of the second heart field is associated with congenital heart defects. Here, taking the mouse as a model, we review the epithelial properties of the second heart field and how they are modulated upon cardiomyocyte differentiation. Compared to other cases of tubulogenesis, the cellular dynamics in the second heart field are only beginning to be revealed. A challenge for the future will be to unravel key physical forces driving heart tube morphogenesis.
Collapse
Affiliation(s)
- Paul Palmquist-Gomes
- Université de Paris, Imagine- Institut Pasteur, Unit of Heart Morphogenesis, INSERM UMR1163, Paris, F-75015, France
| | - Sigolène M Meilhac
- Université de Paris, Imagine- Institut Pasteur, Unit of Heart Morphogenesis, INSERM UMR1163, Paris, F-75015, France.
| |
Collapse
|
25
|
Single cell multi-omic analysis identifies a Tbx1-dependent multilineage primed population in murine cardiopharyngeal mesoderm. Nat Commun 2021; 12:6645. [PMID: 34789765 PMCID: PMC8599455 DOI: 10.1038/s41467-021-26966-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 10/18/2021] [Indexed: 12/11/2022] Open
Abstract
The poles of the heart and branchiomeric muscles of the face and neck are formed from the cardiopharyngeal mesoderm within the pharyngeal apparatus. They are disrupted in patients with 22q11.2 deletion syndrome, due to haploinsufficiency of TBX1, encoding a T-box transcription factor. Here, using single cell RNA-sequencing, we now identify a multilineage primed population within the cardiopharyngeal mesoderm, marked by Tbx1, which has bipotent properties to form cardiac and branchiomeric muscle cells. The multilineage primed cells are localized within the nascent mesoderm of the caudal lateral pharyngeal apparatus and provide a continuous source of cardiopharyngeal mesoderm progenitors. Tbx1 regulates the maturation of multilineage primed progenitor cells to cardiopharyngeal mesoderm derivatives while restricting ectopic non-mesodermal gene expression. We further show that TBX1 confers this balance of gene expression by direct and indirect regulation of enriched genes in multilineage primed progenitors and downstream pathways, partly through altering chromatin accessibility, the perturbation of which can lead to congenital defects in individuals with 22q11.2 deletion syndrome.
Collapse
|
26
|
Khasawneh RR, Kist R, Queen R, Hussain R, Coxhead J, Schneider JE, Mohun TJ, Zaffran S, Peters H, Phillips HM, Bamforth SD. Msx1 haploinsufficiency modifies the Pax9-deficient cardiovascular phenotype. BMC DEVELOPMENTAL BIOLOGY 2021; 21:14. [PMID: 34615475 PMCID: PMC8493722 DOI: 10.1186/s12861-021-00245-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 09/23/2021] [Indexed: 01/16/2023]
Abstract
BACKGROUND Successful embryogenesis relies on the coordinated interaction between genes and tissues. The transcription factors Pax9 and Msx1 genetically interact during mouse craniofacial morphogenesis, and mice deficient for either gene display abnormal tooth and palate development. Pax9 is expressed specifically in the pharyngeal endoderm at mid-embryogenesis, and mice deficient for Pax9 on a C57Bl/6 genetic background also have cardiovascular defects affecting the outflow tract and aortic arch arteries giving double-outlet right ventricle, absent common carotid arteries and interruption of the aortic arch. RESULTS In this study we have investigated both the effect of a different genetic background and Msx1 haploinsufficiency on the presentation of the Pax9-deficient cardiovascular phenotype. Compared to mice on a C57Bl/6 background, congenic CD1-Pax9-/- mice displayed a significantly reduced incidence of outflow tract defects but aortic arch defects were unchanged. Pax9-/- mice with Msx1 haploinsufficiency, however, have a reduced incidence of interrupted aortic arch, but more cases with cervical origins of the right subclavian artery and aortic arch, than seen in Pax9-/- mice. This alteration in arch artery defects was accompanied by a rescue in third pharyngeal arch neural crest cell migration and smooth muscle cell coverage of the third pharyngeal arch arteries. Although this change in phenotype could theoretically be compatible with post-natal survival, using tissue-specific inactivation of Pax9 to maintain correct palate development whilst inducing the cardiovascular defects was unable to prevent postnatal death in the mutant mice. Hyoid bone and thyroid cartilage formation were abnormal in Pax9-/- mice. CONCLUSIONS Msx1 haploinsufficiency mitigates the arch artery defects in Pax9-/- mice, potentially by maintaining the survival of the 3rd arch artery through unimpaired migration of neural crest cells to the third pharyngeal arches. With the neural crest cell derived hyoid bone and thyroid cartilage also being defective in Pax9-/- mice, we speculate that the pharyngeal endoderm is a key signalling centre that impacts on neural crest cell behaviour highlighting the ability of cells in different tissues to act synergistically or antagonistically during embryo development.
Collapse
Affiliation(s)
- Ramada R. Khasawneh
- grid.419328.50000 0000 9225 6820Newcastle University Biosciences Institute, Centre for Life, Newcastle, NE1 3BZ UK ,grid.14440.350000 0004 0622 5497Present Address: Department of Basic Medical Sciences, Faculty of Medicine, Yarmouk University, Irbid, Jordan
| | - Ralf Kist
- grid.419328.50000 0000 9225 6820Newcastle University Biosciences Institute, Centre for Life, Newcastle, NE1 3BZ UK ,grid.1006.70000 0001 0462 7212School of Dental Sciences, Newcastle University, Newcastle, NE2 4BW UK
| | - Rachel Queen
- grid.1006.70000 0001 0462 7212Bioinformatics Support Unit, Newcastle University, Newcastle, NE1 3BZ UK
| | - Rafiqul Hussain
- grid.1006.70000 0001 0462 7212Genomics Core Facility, Newcastle University, Newcastle, NE1 3BZ UK
| | - Jonathan Coxhead
- grid.1006.70000 0001 0462 7212Genomics Core Facility, Newcastle University, Newcastle, NE1 3BZ UK
| | - Jürgen E. Schneider
- grid.9909.90000 0004 1936 8403Biomedical Imaging, University of Leeds, Leeds, LS2 9JT UK
| | - Timothy J. Mohun
- grid.451388.30000 0004 1795 1830The Francis Crick Institute, London, NW1 1AT UK
| | - Stéphane Zaffran
- grid.5399.60000 0001 2176 4817INSERM, Marseille Medical Genetics, U1251, Aix Marseille University, Marseille, France
| | - Heiko Peters
- grid.419328.50000 0000 9225 6820Newcastle University Biosciences Institute, Centre for Life, Newcastle, NE1 3BZ UK
| | - Helen M. Phillips
- grid.419328.50000 0000 9225 6820Newcastle University Biosciences Institute, Centre for Life, Newcastle, NE1 3BZ UK
| | - Simon D. Bamforth
- grid.419328.50000 0000 9225 6820Newcastle University Biosciences Institute, Centre for Life, Newcastle, NE1 3BZ UK
| |
Collapse
|
27
|
Jiang X, Li T, Liu S, Fu Q, Li F, Chen S, Sun K, Xu R, Xu Y. Variants in a cis-regulatory element of TBX1 in conotruncal heart defect patients impair GATA6-mediated transactivation. Orphanet J Rare Dis 2021; 16:334. [PMID: 34332615 PMCID: PMC8325851 DOI: 10.1186/s13023-021-01981-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 07/25/2021] [Indexed: 12/14/2022] Open
Abstract
Background TBX1 (T-box transcription factor 1) is a major candidate gene that likely contributes to the etiology of velo-cardio-facial syndrome/DiGeorge syndrome (VCFS/DGS). Although the haploinsufficiency of TBX1 in both mice and humans results in congenital cardiac malformations, little has been elucidated about its upstream regulation. We aimed to explore the transcriptional regulation and dysregulation of TBX1. Methods Different TBX1 promoter reporters were constructed. Luciferase assays and electrophoretic mobility shift assays (EMSAs) were used to identify a cis-regulatory element within the TBX1 promoter region and its trans-acting factor. The expression of proteins was identified by immunohistochemistry and immunofluorescence. Variants in the cis-regulatory element were screened in conotruncal defect (CTD) patients. In vitro functional assays were performed to show the effects of the variants found in CTD patients on the transactivation of TBX1. Results We identified a cis-regulatory element within intron 1 of TBX1 that was found to be responsive to GATA6 (GATA binding protein 6), a transcription factor crucial for cardiogenesis. The expression patterns of GATA6 and TBX1 overlapped in the pharyngeal arches of human embryos. Transfection experiments and EMSA indicated that GATA6 could activate the transcription of TBX1 by directly binding with its GATA cis-regulatory element in vitro. Furthermore, sequencing analyses of 195 sporadic CTD patients without the 22q11.2 deletion or duplication identified 3 variants (NC_000022.11:g.19756832C > G, NC_000022.11:g.19756845C > T, and NC_000022.11:g. 19756902G > T) in the non-coding cis-regulatory element of TBX1. Luciferase assays showed that all 3 variants led to reduced transcription of TBX1 when incubated with GATA6. Conclusions Our findings showed that TBX1 might be a direct transcriptional target of GATA6, and variants in the non-coding cis-regulatory element of TBX1 disrupted GATA6-mediated transactivation. Supplementary Information The online version contains supplementary material available at 10.1186/s13023-021-01981-4.
Collapse
Affiliation(s)
- Xuechao Jiang
- Scientific Research Center, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Tingting Li
- Department of Pediatric Cardiology, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Sijie Liu
- Department of Pediatric Cardiology, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Qihua Fu
- Medical Laboratory, Shanghai Children's Medical Center, Affiliated to Shanghai Jiao Tong University School of Medicine , Shanghai, 200127, China
| | - Fen Li
- Department of Pediatric Cardiology, Shanghai Children's Medical Center, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Sun Chen
- Department of Pediatric Cardiology, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Kun Sun
- Department of Pediatric Cardiology, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Rang Xu
- Scientific Research Center, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| | - Yuejuan Xu
- Department of Pediatric Cardiology, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| |
Collapse
|
28
|
Sarac Sivrikoz T, Basaran S, Has R, Karaman B, Kalelioglu IH, Kirgiz M, Altunoglu U, Yuksel A. Prenatal sonographic and cytogenetic/molecular findings of 22q11.2 microdeletion syndrome in 48 confirmed cases in a single tertiary center. Arch Gynecol Obstet 2021; 305:323-342. [PMID: 34145474 DOI: 10.1007/s00404-021-06125-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 06/12/2021] [Indexed: 11/26/2022]
Abstract
PURPOSE We aimed to present the fetal ultrasound, cytogenetic/molecular testing and postmortem or postnatal clinical findings of cases with 22q11.2DS diagnosed prenatally. MATERIALS AND METHODS A retrospective medical record review of 48 prenatal cases diagnosed with 22q11.2DS were evaluated in our institution. Detailed ultrasound examination was performed on all fetuses. Postmortem and postnatal examinations were evaluated. The microdeletions were detected by karyotyping or microarray, then confirmed by FISH. Descriptive statistical analysis was performed. RESULTS Demographic data of 48 prenatal cases including 46 singletons and 1 dichorionic diamniotic twin pregnancy were evaluated. The most common extracardiac anomaly was skeletal system anomalies (25%), in which PEV was the most frequent one (20.8%). Polyhydramnios rate was detected as 31%, in 6.6% as an isolated finding. Microdeletion has been detected by karyotyping in 13 cases (13/47, 27.7%) (including 2 unbalanced translocations), by FISH in 28 cases (28/48, 58.3%), by microarray/a-CGH testing in 7 cases. Microarray analysis showed that in one case with unbalanced translocation had two consecutive deletions; one was proximal and other one distal to critical region and not encompassing TBX1 gene but CRKL and LZTR1 genes. CONCLUSION The current study demonstrates the whole spectrum of atypical phenotypic and genotypic variations of 22q11.2DS in the largest prenatal case series reported to date. Therefore, differential diagnosis should be considered not solely in CHD, but also in the presence of isolated clubfeet and polyhydramnios. Establishing the diagnosis in the prenatal period may allow a postnatal multidisciplinary approach, as well as affect the actual prevalence of the disease.
Collapse
Affiliation(s)
- Tugba Sarac Sivrikoz
- Division of Perinatology, Department of Obstetrics and Gynecology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey.
| | - Seher Basaran
- Department of Medical Genetics, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
- PREMED, Center for Genetic Diagnosis and Research, Mecidiyekoy, Istanbul, Turkey
| | - Recep Has
- Division of Perinatology, Department of Obstetrics and Gynecology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Birsen Karaman
- Department of Medical Genetics, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
- Department of Pediatric Basic Science, Child Health Institute, Istanbul University, Istanbul, Turkey
| | - Ibrahim Halil Kalelioglu
- Division of Perinatology, Department of Obstetrics and Gynecology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Melike Kirgiz
- PREMED, Center for Genetic Diagnosis and Research, Mecidiyekoy, Istanbul, Turkey
| | - Umut Altunoglu
- Department of Medical Genetics, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Atil Yuksel
- Division of Perinatology, Department of Obstetrics and Gynecology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| |
Collapse
|
29
|
Abstract
Congenital heart disease is the most frequent birth defect and the leading cause of death for the fetus and in the first year of life. The wide phenotypic diversity of congenital heart defects requires expert diagnosis and sophisticated repair surgery. Although these defects have been described since the seventeenth century, it was only in 2005 that a consensus international nomenclature was adopted, followed by an international classification in 2017 to help provide better management of patients. Advances in genetic engineering, imaging, and omics analyses have uncovered mechanisms of heart formation and malformation in animal models, but approximately 80% of congenital heart defects have an unknown genetic origin. Here, we summarize current knowledge of congenital structural heart defects, intertwining clinical and fundamental research perspectives, with the aim to foster interdisciplinary collaborations at the cutting edge of each field. We also discuss remaining challenges in better understanding congenital heart defects and providing benefits to patients.
Collapse
Affiliation(s)
- Lucile Houyel
- Unité de Cardiologie Pédiatrique et Congénitale and Centre de Référence des Malformations Cardiaques Congénitales Complexes (M3C), Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), 75015 Paris, France.,Université de Paris, 75015 Paris, France
| | - Sigolène M Meilhac
- Université de Paris, 75015 Paris, France.,Imagine-Institut Pasteur Unit of Heart Morphogenesis, INSERM UMR 1163, 75015 Paris, France;
| |
Collapse
|
30
|
Fan X, Loebel DAF, Bildsoe H, Wilkie EE, Qin J, Wang J, Tam PPL. Tissue interactions, cell signaling and transcriptional control in the cranial mesoderm during craniofacial development. AIMS GENETICS 2021. [DOI: 10.3934/genet.2016.1.74] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
AbstractThe cranial neural crest and the cranial mesoderm are the source of tissues from which the bone and cartilage of the skull, face and jaws are constructed. The development of the cranial mesoderm is not well studied, which is inconsistent with its importance in craniofacial morphogenesis as a source of precursor tissue of the chondrocranium, muscles, vasculature and connective tissues, mechanical support for tissue morphogenesis, and the signaling activity that mediate interactions with the cranial neural crest. Phenotypic analysis of conditional knockout mouse mutants, complemented by the transcriptome analysis of differentially enriched genes in the cranial mesoderm and cranial neural crest, have identified signaling pathways that may mediate cross-talk between the two tissues. In the cranial mesenchyme, Bmp4 is expressed in the mesoderm cells while its signaling activity could impact on both the mesoderm and the neural crest cells. In contrast, Fgf8 is predominantly expressed in the cranial neural crest cells and it influences skeletal development and myogenesis in the cranial mesoderm. WNT signaling, which emanates from the cranial neural crest cells, interacts with BMP and FGF signaling in monitoring the switch between tissue progenitor expansion and differentiation. The transcription factor Twist1, a critical molecular regulator of many aspects of craniofacial development, coordinates the activity of the above pathways in cranial mesoderm and cranial neural crest tissue compartments.
Collapse
Affiliation(s)
- Xiaochen Fan
- Embryology Unit, Children's Medical Research Institute, Westmead NSW 2145, Australia
| | - David A F Loebel
- Embryology Unit, Children's Medical Research Institute, Westmead NSW 2145, Australia
| | - Heidi Bildsoe
- Embryology Unit, Children's Medical Research Institute, Westmead NSW 2145, Australia
| | - Emilie E Wilkie
- Embryology Unit, Children's Medical Research Institute, Westmead NSW 2145, Australia
- Bioinformatics Group, Children's Medical Research Institute, Westmead NSW 2145, Australia
| | - Jing Qin
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, NT, Hong Kong
| | - Junwen Wang
- Department of Health Sciences Research, Center for Individualized Medicine, Mayo Clinic, and Department of Biomedical Informatics, Arizona State University, Scottsdale AZ 85259, USA
| | - Patrick P L Tam
- Embryology Unit, Children's Medical Research Institute, Westmead NSW 2145, Australia
- School of Medical Sciences, Sydney Medical School, University of Sydney, NSW 2006, Australia
| |
Collapse
|
31
|
Goliusova DV, Klementieva NV, Panova AV, Mokrysheva NG, Kiselev SL. The Role of Genetic Factors in Endocrine Tissues Development and Its Regulation In Vivo and In Vitro. RUSS J GENET+ 2021. [DOI: 10.1134/s102279542103008x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
32
|
Fischer C, Koblmüller S, Börger C, Michelitsch G, Trajanoski S, Schlötterer C, Guelly C, Thallinger GG, Sturmbauer C. Genome sequences of Tropheus moorii and Petrochromis trewavasae, two eco-morphologically divergent cichlid fishes endemic to Lake Tanganyika. Sci Rep 2021; 11:4309. [PMID: 33619328 PMCID: PMC7900123 DOI: 10.1038/s41598-021-81030-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 12/28/2020] [Indexed: 01/01/2023] Open
Abstract
With more than 1000 species, East African cichlid fishes represent the fastest and most species-rich vertebrate radiation known, providing an ideal model to tackle molecular mechanisms underlying recurrent adaptive diversification. We add high-quality genome reconstructions for two phylogenetic key species of a lineage that diverged about ~ 3-9 million years ago (mya), representing the earliest split of the so-called modern haplochromines that seeded additional radiations such as those in Lake Malawi and Victoria. Along with the annotated genomes we analysed discriminating genomic features of the study species, each representing an extreme trophic morphology, one being an algae browser and the other an algae grazer. The genomes of Tropheus moorii (TM) and Petrochromis trewavasae (PT) comprise 911 and 918 Mbp with 40,300 and 39,600 predicted genes, respectively. Our DNA sequence data are based on 5 and 6 individuals of TM and PT, and the transcriptomic sequences of one individual per species and sex, respectively. Concerning variation, on average we observed 1 variant per 220 bp (interspecific), and 1 variant per 2540 bp (PT vs PT)/1561 bp (TM vs TM) (intraspecific). GO enrichment analysis of gene regions affected by variants revealed several candidates which may influence phenotype modifications related to facial and jaw morphology, such as genes belonging to the Hedgehog pathway (SHH, SMO, WNT9A) and the BMP and GLI families.
Collapse
Affiliation(s)
- C Fischer
- Institute of Biology, University of Graz, Graz, Austria
- Institute of Biomedical Informatics, Graz University of Technology, Graz, Austria
| | - S Koblmüller
- Institute of Biology, University of Graz, Graz, Austria
| | - C Börger
- Institute of Biology, University of Graz, Graz, Austria
| | - G Michelitsch
- Center for Medical Research, Medical University of Graz, Graz, Austria
| | - S Trajanoski
- Center for Medical Research, Medical University of Graz, Graz, Austria
| | - C Schlötterer
- Institut für Populationsgenetik, Vetmeduni Vienna, Vienna, Austria
| | - C Guelly
- Center for Medical Research, Medical University of Graz, Graz, Austria
| | - G G Thallinger
- Institute of Biomedical Informatics, Graz University of Technology, Graz, Austria.
- BioTechMed-Graz, Graz, Austria.
| | - C Sturmbauer
- Institute of Biology, University of Graz, Graz, Austria.
- BioTechMed-Graz, Graz, Austria.
| |
Collapse
|
33
|
Caprio C, Lania G, Bilio M, Ferrentino R, Chen L, Baldini A. EZH2 is required for parathyroid and thymic development through differentiation of the third pharyngeal pouch endoderm. Dis Model Mech 2021; 14:dmm.046789. [PMID: 33608392 PMCID: PMC7969367 DOI: 10.1242/dmm.046789] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 02/10/2021] [Indexed: 12/18/2022] Open
Abstract
The Ezh2 gene encodes a histone methyltransferase of the polycomb repressive complex 2 that methylates histone H3 lysine 27. In this study, we investigated whether EZH2 has a role in the development of the pharyngeal apparatus and whether it regulates the expression of the Tbx1 gene, which encodes a key transcription factor required in pharyngeal development. To these ends, we performed genetic in vivo experiments with mouse embryos and used mouse embryonic stem cell (ESC)-based protocols to probe endoderm and cardiogenic mesoderm differentiation. Results showed that EZH2 occupies the Tbx1 gene locus in mouse embryos, and that suppression of EZH2 was associated with reduced expression of Tbx1 in differentiated mouse ESCs. Conditional deletion of Ezh2 in the Tbx1 expression domain, which includes the pharyngeal endoderm, did not cause cardiac defects but revealed that the gene has an important role in the morphogenesis of the third pharyngeal pouch (PP). We found that in conditionally deleted embryos the third PP was hypoplastic, had reduced expression of Tbx1, lacked the expression of Gcm2, a gene that marks the parathyroid domain, but expressed FoxN1, a gene marking the thymic domain. Consistently, the parathyroids did not develop, and the thymus was hypoplastic. Thus, Ezh2 is required for parathyroid and thymic development, probably through a function in the pouch endoderm. This discovery also provides a novel interpretational key for the finding of Ezh2 activating mutations in hyperparathyroidism and parathyroid cancer. Summary: EZH2 in the pharyngeal apparatus is required in the endoderm for the development of the parathyroids and thymus, and positively regulates the expression of the Tbx1 gene.
Collapse
Affiliation(s)
- Cinzia Caprio
- Institute of Genetics and Biophysics, CNR, Naples, Italy
| | | | - Marchesa Bilio
- Institute of Genetics and Biophysics, CNR, Naples, Italy
| | | | - Li Chen
- Department of Biology and Biochemistry, University of Houston, TX
| | - Antonio Baldini
- Institute of Genetics and Biophysics, CNR, Naples, Italy .,Department of Molecular Medicine and Medical Biotechnologies, University Federico II, Naples, Italy
| |
Collapse
|
34
|
Watanabe Y, Seya D, Ihara D, Ishii S, Uemoto T, Kubo A, Arai Y, Isomoto Y, Nakano A, Abe T, Shigeta M, Kawamura T, Saito Y, Ogura T, Nakagawa O. Importance of endothelial Hey1 expression for thoracic great vessel development and its distal enhancer for Notch-dependent endothelial transcription. J Biol Chem 2020; 295:17632-17645. [PMID: 33454003 PMCID: PMC7762959 DOI: 10.1074/jbc.ra120.015003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 10/12/2020] [Indexed: 12/19/2022] Open
Abstract
Thoracic great vessels such as the aorta and subclavian arteries are formed through dynamic remodeling of embryonic pharyngeal arch arteries (PAAs). Previous work has shown that loss of a basic helix-loop-helix transcription factor Hey1 in mice causes abnormal fourth PAA development and lethal great vessel anomalies resembling congenital malformations in humans. However, how Hey1 mediates vascular formation remains unclear. In this study, we revealed that Hey1 in vascular endothelial cells, but not in smooth muscle cells, played essential roles for PAA development and great vessel morphogenesis in mouse embryos. Tek-Cre-mediated Hey1 deletion in endothelial cells affected endothelial tube formation and smooth muscle differentiation in embryonic fourth PAAs and resulted in interruption of the aortic arch and other great vessel malformations. Cell specificity and signal responsiveness of Hey1 expression were controlled through multiple cis-regulatory regions. We found two distal genomic regions that had enhancer activity in endothelial cells and in the pharyngeal epithelium and somites, respectively. The novel endothelial enhancer was conserved across species and was specific to large-caliber arteries. Its transcriptional activity was regulated by Notch signaling in vitro and in vivo, but not by ALK1 signaling and other transcription factors implicated in endothelial cell specificity. The distal endothelial enhancer was not essential for basal Hey1 expression in mouse embryos but may likely serve for Notch-dependent transcriptional control in endothelial cells together with the proximal regulatory region. These findings help in understanding the significance and regulation of endothelial Hey1 as a mediator of multiple signaling pathways in embryonic vascular formation.
Collapse
Affiliation(s)
- Yusuke Watanabe
- Department of Molecular Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan; Graduate School of Medical Sciences, Nara Medical University, Kashihara, Nara, Japan.
| | - Daiki Seya
- Department of Molecular Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
| | - Dai Ihara
- Department of Molecular Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan; Laboratory of Stem Cell and Regenerative Medicine, Department of Biomedical Sciences, College of Life Sciences, Ritsumeikan University, Kusatsu, Shiga, Japan
| | - Shuhei Ishii
- Department of Molecular Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan; Graduate School of Medical Sciences, Nara Medical University, Kashihara, Nara, Japan
| | - Taiki Uemoto
- Department of Molecular Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan; Graduate School of Medical Sciences, Nara Medical University, Kashihara, Nara, Japan
| | - Atsushi Kubo
- Department of Developmental Neurobiology, Institute of Development, Aging, and Cancer, Tohoku University, Sendai, Miyagi, Japan
| | - Yuji Arai
- Department of Molecular Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan; Laboratory of Animal Experiment and Medical Management, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
| | - Yoshie Isomoto
- Laboratory of Animal Experiment and Medical Management, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
| | - Atsushi Nakano
- Laboratory of Animal Experiment and Medical Management, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
| | - Takaya Abe
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Mayo Shigeta
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Teruhisa Kawamura
- Laboratory of Stem Cell and Regenerative Medicine, Department of Biomedical Sciences, College of Life Sciences, Ritsumeikan University, Kusatsu, Shiga, Japan
| | - Yoshihiko Saito
- Graduate School of Medical Sciences, Nara Medical University, Kashihara, Nara, Japan; Department of Cardiovascular Medicine, Nara Medical University, Kashihara, Nara, Japan
| | - Toshihiko Ogura
- Department of Developmental Neurobiology, Institute of Development, Aging, and Cancer, Tohoku University, Sendai, Miyagi, Japan
| | - Osamu Nakagawa
- Department of Molecular Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan; Graduate School of Medical Sciences, Nara Medical University, Kashihara, Nara, Japan.
| |
Collapse
|
35
|
Wei L, Wang W, Yang J, Huang X, Baldini A, Zhang Z. Pharyngeal epithelial deletion of Tbx1 causes caudal pharyngeal arch defect but not cardiac conotruncal anomaly. Biochem Biophys Res Commun 2020; 533:1315-1322. [PMID: 33066956 DOI: 10.1016/j.bbrc.2020.10.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 10/06/2020] [Indexed: 11/29/2022]
Abstract
TBX1 is a major disease gene of 22q11.2 deletion syndrome (22q11.2DS). It is expressed in all three germ layers of pharyngeal apparatus to control the complicated morphogenesis. The haploinsufficiency of pharyngeal endodermal or ectodermal, but not mesodermal Tbx1 causes aortic arch patterning defect. However, the mesodermal deletion of Tbx1 causes much severer pharyngeal and cardiovascular defect than either pharyngeal endodermal or ectodermal Tbx1 deletion does. It is inconsistent with the conventional thought that the invagination of pharyngeal epithelia drives pharyngeal segmentation. Therefore, we asked whether pharyngeal ectodermal and ectodermal Tbx1 can compensate the loss of each other. Here we carefully characterized pharyngeal epithelia-specific Fgf15Cre and Fgf15HspCre lines and used them to perform pharyngeal epithelia-specific deletion. Our data showed that the percentage of E18.5 Fgf15Cre;Tbx1flox/+ embryos with aortic arch patterning defects was similar to that of E10.5 Fgf15Cre;Tbx1flox/+ embryos with the 4th pharyngeal arch artery (PAA) defect, indicating that there is no significant recovery from the initial PAA defect, in contrast to germ line haploinsufficiency. Fgf15Cre;Tbx1flox/flox embryos had hypoplastic caudal pharyngeal arch and defective derivatives, but cardiac OFT development was not affected. The phenotypic spectrum of simultaneous Tbx1 deletion in both pharyngeal ectoderm and endoderm is strikingly similar to what presents with single pharyngeal endoderm or ectoderm-specific deletion of Tbx1. The absence of synergistic effect indicates intimate topographic interactions among pharyngeal endoderm and ectoderm, through which deletion of a gene in one tissue may disrupt the development of adjacent tissues and thereby lead to similar morphological phenotypes in either tissue-specific deletion.
Collapse
Affiliation(s)
- Lu Wei
- Pediatric Translational Medicine Institute and Shanghai Pediatric Congenital Heart Disease Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenfeng Wang
- Pediatric Translational Medicine Institute and Shanghai Pediatric Congenital Heart Disease Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junjie Yang
- Pediatric Translational Medicine Institute and Shanghai Pediatric Congenital Heart Disease Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xu Huang
- School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Antonio Baldini
- Department of Molecular Medicine and Medical Biotechnologies, University Federico II, Naples, Italy; Institute of Genetics and Biophysics, CNR Naples, Italy
| | - Zhen Zhang
- Pediatric Translational Medicine Institute and Shanghai Pediatric Congenital Heart Disease Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
36
|
Lungova V, Thibeault SL. Mechanisms of larynx and vocal fold development and pathogenesis. Cell Mol Life Sci 2020; 77:3781-3795. [PMID: 32253462 PMCID: PMC7511430 DOI: 10.1007/s00018-020-03506-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 03/12/2020] [Accepted: 03/16/2020] [Indexed: 12/12/2022]
Abstract
The larynx and vocal folds sit at the crossroad between digestive and respiratory tracts and fulfill multiple functions related to breathing, protection and phonation. They develop at the head and trunk interface through a sequence of morphogenetic events that require precise temporo-spatial coordination. We are beginning to understand some of the molecular and cellular mechanisms that underlie critical processes such as specification of the laryngeal field, epithelial lamina formation and recanalization as well as the development and differentiation of mesenchymal cell populations. Nevertheless, many gaps remain in our knowledge, the filling of which is essential for understanding congenital laryngeal disorders and the evaluation and treatment approaches in human patients. This review highlights recent advances in our understanding of the laryngeal embryogenesis. Proposed genes and signaling pathways that are critical for the laryngeal development have a potential to be harnessed in the field of regenerative medicine.
Collapse
Affiliation(s)
- Vlasta Lungova
- Department of Surgery, University of Wisconsin Madison, 5103 WIMR, 1111 Highland Ave, Madison, WI, 53705, USA
| | - Susan L Thibeault
- Department of Surgery, University of Wisconsin Madison, 5103 WIMR, 1111 Highland Ave, Madison, WI, 53705, USA.
| |
Collapse
|
37
|
Thymus Inception: Molecular Network in the Early Stages of Thymus Organogenesis. Int J Mol Sci 2020; 21:ijms21165765. [PMID: 32796710 PMCID: PMC7460828 DOI: 10.3390/ijms21165765] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 08/07/2020] [Indexed: 11/17/2022] Open
Abstract
The thymus generates central immune tolerance by producing self-restricted and self-tolerant T-cells as a result of interactions between the developing thymocytes and the stromal microenvironment, mainly formed by the thymic epithelial cells. The thymic epithelium derives from the endoderm of the pharyngeal pouches, embryonic structures that rely on environmental cues from the surrounding mesenchyme for its development. Here, we review the most recent advances in our understanding of the molecular mechanisms involved in early thymic organogenesis at stages preceding the expression of the transcription factor Foxn1, the early marker of thymic epithelial cells identity. Foxn1-independent developmental stages, such as the specification of the pharyngeal endoderm, patterning of the pouches, and thymus fate commitment are discussed, with a special focus on epithelial–mesenchymal interactions.
Collapse
|
38
|
Imamoto A, Ki S, Li L, Iwamoto K, Maruthamuthu V, Devany J, Lu O, Kanazawa T, Zhang S, Yamada T, Hirayama A, Fukuda S, Suzuki Y, Okada M. Essential role of the Crk family-dosage in DiGeorge-like anomaly and metabolic homeostasis. Life Sci Alliance 2020; 3:3/2/e201900635. [PMID: 32041892 PMCID: PMC7010317 DOI: 10.26508/lsa.201900635] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 01/20/2020] [Accepted: 01/21/2020] [Indexed: 12/12/2022] Open
Abstract
CRK and CRKL (CRK-like) encode adapter proteins with similar biochemical properties. Here, we show that a 50% reduction of the family-combined dosage generates developmental defects, including aspects of DiGeorge/del22q11 syndrome in mice. Like the mouse homologs of two 22q11.21 genes CRKL and TBX1, Crk and Tbx1 also genetically interact, thus suggesting that pathways shared by the three genes participate in organogenesis affected in the syndrome. We also show that Crk and Crkl are required during mesoderm development, and Crk/Crkl deficiency results in small cell size and abnormal mesenchyme behavior in primary embryonic fibroblasts. Our systems-wide analyses reveal impaired glycolysis, associated with low Hif1a protein levels as well as reduced histone H3K27 acetylation in several key glycolysis genes. Furthermore, Crk/Crkl deficiency sensitizes MEFs to 2-deoxy-D-glucose, a competitive inhibitor of glycolysis, to induce cell blebbing. Activated Rapgef1, a Crk/Crkl-downstream effector, rescues several aspects of the cell phenotype, including proliferation, cell size, focal adhesions, and phosphorylation of p70 S6k1 and ribosomal protein S6. Our investigations demonstrate that Crk/Crkl-shared pathways orchestrate metabolic homeostasis and cell behavior through widespread epigenetic controls.
Collapse
Affiliation(s)
- Akira Imamoto
- The Ben May Department for Cancer Research, The University of Chicago, Chicago, IL, USA
| | - Sewon Ki
- RIKEN Integrative Medical Sciences, Tsurumi, Yokohama, Kanagawa, Japan
| | - Leiming Li
- The Ben May Department for Cancer Research, The University of Chicago, Chicago, IL, USA
| | - Kazunari Iwamoto
- Institute for Protein Research, Osaka University, Suita, Osaka, Japan
| | - Venkat Maruthamuthu
- Department of Mechanical and Aerospace Engineering, Old Dominion University, Norfolk, VA, USA
| | - John Devany
- Department of Physics, The University of Chicago, Chicago, IL, USA
| | - Ocean Lu
- The Ben May Department for Cancer Research, The University of Chicago, Chicago, IL, USA
| | - Tomomi Kanazawa
- Institute for Protein Research, Osaka University, Suita, Osaka, Japan
| | - Suxiang Zhang
- Institute for Protein Research, Osaka University, Suita, Osaka, Japan
| | - Takuji Yamada
- Department of Life Science and Technology, Tokyo Institute of Technology, Meguro, Tokyo, Japan
| | - Akiyoshi Hirayama
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata, Japan
| | - Shinji Fukuda
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata, Japan.,Intestinal Microbiota Project, Kanagawa Institute of Industrial Science and Technology, Kawasaki, Kanagawa, Japan.,Transborder Medical Research Center, University of Tsukuba, Tsukuba, Ibaraki, Japan.,PRESTO, Japan Science and Technology Agency, Kawaguchi, Saitama, Japan
| | - Yutaka Suzuki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, Japan
| | - Mariko Okada
- RIKEN Integrative Medical Sciences, Tsurumi, Yokohama, Kanagawa, Japan .,Institute for Protein Research, Osaka University, Suita, Osaka, Japan.,Center for Drug Design and Research, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka, Japan
| |
Collapse
|
39
|
Failed Progenitor Specification Underlies the Cardiopharyngeal Phenotypes in a Zebrafish Model of 22q11.2 Deletion Syndrome. Cell Rep 2019; 24:1342-1354.e5. [PMID: 30067987 PMCID: PMC6261257 DOI: 10.1016/j.celrep.2018.06.117] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 05/08/2018] [Accepted: 06/28/2018] [Indexed: 12/13/2022] Open
Abstract
Microdeletions involving TBX1 result in variable congenital malformations known collectively as 22q11.2 deletion syndrome (22q11.2DS). Tbx1-deficient mice and zebrafish recapitulate several disease phenotypes, including pharyngeal arch artery (PAA), head muscle (HM), and cardiac outflow tract (OFT) deficiencies. In zebrafish, these structures arise from nkx2.5+ progenitors in pharyngeal arches 2-6. Because pharyngeal arch morphogenesis is compromised in Tbx1-deficient animals, the malformations were considered secondary. Here, we report that the PAA, HM, and OFT phenotypes in tbx1 mutant zebrafish are primary and arise prior to pharyngeal arch morphogenesis from failed specification of the nkx2.5+ pharyngeal lineage. Through in situ analysis and lineage tracing, we reveal that nkx2.5 and tbx1 are co-expressed in this progenitor population. Furthermore, we present evidence suggesting that gdf3-ALK4 signaling is a downstream mediator of nkx2.5+ pharyngeal lineage specification. Collectively, these studies support a cellular mechanism potentially underlying the cardiovascular and craniofacial defects observed in the 22q11.2DS population.
Collapse
|
40
|
Sullivan KE. Chromosome 22q11.2 deletion syndrome and DiGeorge syndrome. Immunol Rev 2019; 287:186-201. [PMID: 30565249 DOI: 10.1111/imr.12701] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 07/30/2018] [Indexed: 12/19/2022]
Abstract
Chromosome 22q11.2 deletion syndrome is the most common microdeletion syndrome in humans. The effects are protean and highly variable, making a unified approach difficult. Nevertheless, commonalities have been identified and white papers with recommended evaluations and anticipatory guidance have been published. This review will cover the immune system in detail and discuss both the primary features and the secondary features related to thymic hypoplasia. A brief discussion of the other organ system involvement will be provided for context. The immune system, percolating throughout the body can impact the function of other organs through allergy or autoimmune disease affecting organs in deleterious manners. Our work has shown that the primary effect of thymic hypoplasia is to restrict T cell production. Subsequent homeostatic proliferation and perhaps other factors drive a Th2 polarization, most obvious in adulthood. This contributes to atopic risk in this population. Thymic hypoplasia also contributes to low regulatory T cells and this may be part of the overall increased risk of autoimmunity. Collectively, the effects are complex and often age-dependent. Future goals of improving thymic function or augmenting thymic volume may offer a direct intervention to ameliorate infections, atopy, and autoimmunity.
Collapse
Affiliation(s)
- Kathleen E Sullivan
- The Children's Hospital of Philadelphia, The University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| |
Collapse
|
41
|
Morrow BE, McDonald-McGinn DM, Emanuel BS, Vermeesch JR, Scambler PJ. Molecular genetics of 22q11.2 deletion syndrome. Am J Med Genet A 2019; 176:2070-2081. [PMID: 30380194 DOI: 10.1002/ajmg.a.40504] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 06/11/2018] [Accepted: 07/17/2018] [Indexed: 02/02/2023]
Abstract
The 22q11.2 deletion syndrome (22q11.2DS) is a congenital malformation and neuropsychiatric disorder caused by meiotic chromosome rearrangements. One of the goals of this review is to summarize the current state of basic research studies of 22q11.2DS. It highlights efforts to understand the mechanisms responsible for the 22q11.2 deletion that occurs in meiosis. This mechanism involves the four sets of low copy repeats (LCR22) that are dispersed in the 22q11.2 region and the deletion is mediated by nonallelic homologous recombination events. This review also highlights selected genes mapping to the 22q11.2 region that may contribute to the typical clinical findings associated with the disorder and explain that mutations in genes on the remaining allele can uncover rare recessive conditions. Another important aspect of 22q11.2DS is the existence of phenotypic heterogeneity. While some patients are mildly affected, others have severe medical, cognitive, and/or psychiatric challenges. Variability may be due in part to the presence of genetic modifiers. This review discusses current genome-wide efforts to identify such modifiers that could shed light on molecular pathways required for normal human development, cognition or behavior.
Collapse
Affiliation(s)
- Bernice E Morrow
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York
| | - Donna M McDonald-McGinn
- Division of Human Genetics, Children's Hospital of Philadelphia and Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Beverly S Emanuel
- Division of Human Genetics, Children's Hospital of Philadelphia and Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Joris R Vermeesch
- Center for Human Genetics, Katholieke Universiteit Leuven (KU Leuven), Leuven, Belgium
| | - Peter J Scambler
- Institute of Child Health, University College London, London, UK
| |
Collapse
|
42
|
Comai G, Heude E, Mella S, Paisant S, Pala F, Gallardo M, Langa F, Kardon G, Gopalakrishnan S, Tajbakhsh S. A distinct cardiopharyngeal mesoderm genetic hierarchy establishes antero-posterior patterning of esophagus striated muscle. eLife 2019; 8:e47460. [PMID: 31535973 PMCID: PMC6752947 DOI: 10.7554/elife.47460] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 08/27/2019] [Indexed: 02/06/2023] Open
Abstract
In most vertebrates, the upper digestive tract is composed of muscularized jaws linked to the esophagus that permits food ingestion and swallowing. Masticatory and esophagus striated muscles (ESM) share a common cardiopharyngeal mesoderm (CPM) origin, however ESM are unusual among striated muscles as they are established in the absence of a primary skeletal muscle scaffold. Using mouse chimeras, we show that the transcription factors Tbx1 and Isl1 are required cell-autonomously for myogenic specification of ESM progenitors. Further, genetic loss-of-function and pharmacological studies point to MET/HGF signaling for antero-posterior migration of esophagus muscle progenitors, where Hgf ligand is expressed in adjacent smooth muscle cells. These observations highlight the functional relevance of a smooth and striated muscle progenitor dialogue for ESM patterning. Our findings establish a Tbx1-Isl1-Met genetic hierarchy that uniquely regulates esophagus myogenesis and identify distinct genetic signatures that can be used as framework to interpret pathologies arising within CPM derivatives.
Collapse
Affiliation(s)
- Glenda Comai
- Department of Developmental and Stem Cell BiologyInstitut PasteurParisFrance
- CNRS UMR 3738ParisFrance
| | - Eglantine Heude
- Department of Developmental and Stem Cell BiologyInstitut PasteurParisFrance
- CNRS UMR 3738ParisFrance
- Department Adaptation du VivantCNRS/MNHN UMR 7221, Muséum national d’Histoire naturelleParisFrance
| | - Sebastian Mella
- Department of Developmental and Stem Cell BiologyInstitut PasteurParisFrance
- CNRS UMR 3738ParisFrance
| | - Sylvain Paisant
- Department of Developmental and Stem Cell BiologyInstitut PasteurParisFrance
- CNRS UMR 3738ParisFrance
| | - Francesca Pala
- Department of Developmental and Stem Cell BiologyInstitut PasteurParisFrance
- CNRS UMR 3738ParisFrance
- Laboratory of Clinical Immunology and Microbiology (LCIM)National Institutes of HealthBethesdaUnited States
| | - Mirialys Gallardo
- Department of Human GeneticsUniversity of UtahSalt Lake CityUnited States
| | - Francina Langa
- Mouse Genetics Engineering CenterInstitut PasteurParisFrance
| | - Gabrielle Kardon
- Department of Human GeneticsUniversity of UtahSalt Lake CityUnited States
| | - Swetha Gopalakrishnan
- Department of Developmental and Stem Cell BiologyInstitut PasteurParisFrance
- CNRS UMR 3738ParisFrance
- Institute of Biotechnology, HiLIFEUniversity of HelsinkiHelsinkiFinland
| | - Shahragim Tajbakhsh
- Department of Developmental and Stem Cell BiologyInstitut PasteurParisFrance
- CNRS UMR 3738ParisFrance
| |
Collapse
|
43
|
Hasten E, Morrow BE. Tbx1 and Foxi3 genetically interact in the pharyngeal pouch endoderm in a mouse model for 22q11.2 deletion syndrome. PLoS Genet 2019; 15:e1008301. [PMID: 31412026 PMCID: PMC6709926 DOI: 10.1371/journal.pgen.1008301] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Revised: 08/26/2019] [Accepted: 07/10/2019] [Indexed: 11/18/2022] Open
Abstract
We investigated whether Tbx1, the gene for 22q11.2 deletion syndrome (22q11.2DS) and Foxi3, both required for segmentation of the pharyngeal apparatus (PA) to individual arches, genetically interact. We found that all Tbx1+/-;Foxi3+/- double heterozygous mouse embryos had thymus and parathyroid gland defects, similar to those in 22q11.2DS patients. We then examined Tbx1 and Foxi3 heterozygous, null as well as conditional Tbx1Cre and Sox172A-iCre/+ null mutant embryos. While Tbx1Cre/+;Foxi3f/f embryos had absent thymus and parathyroid glands, Foxi3-/- and Sox172A-iCre/+;Foxi3f/f endoderm conditional mutant embryos had in addition, interrupted aortic arch type B and retroesophageal origin of the right subclavian artery, which are all features of 22q11.2DS. Tbx1Cre/+;Foxi3f/f embryos had failed invagination of the third pharyngeal pouch with greatly reduced Gcm2 and Foxn1 expression, thereby explaining the absence of thymus and parathyroid glands. Immunofluorescence on tissue sections with E-cadherin and ZO-1 antibodies in wildtype mouse embryos at E8.5-E10.5, revealed that multilayers of epithelial cells form where cells are invaginating as a normal process. We noted that excessive multilayers formed in Foxi3-/-, Sox172A-iCre/+;Foxi3f/f as well as Tbx1 null mutant embryos where invagination should have occurred. Several genes expressed in the PA epithelia were downregulated in both Tbx1 and Foxi3 null mutant embryos including Notch pathway genes Jag1, Hes1, and Hey1, suggesting that they may, along with other genes, act downstream to explain the observed genetic interaction. We found Alcam and Fibronectin extracellular matrix proteins were reduced in expression in Foxi3 null but not Tbx1 null embryos, suggesting that some, but not all of the downstream mechanisms are shared.
Collapse
Affiliation(s)
- Erica Hasten
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Bernice E. Morrow
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, United States of America
| |
Collapse
|
44
|
A single-cell transcriptional roadmap for cardiopharyngeal fate diversification. Nat Cell Biol 2019; 21:674-686. [PMID: 31160712 PMCID: PMC7491489 DOI: 10.1038/s41556-019-0336-z] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 04/29/2019] [Indexed: 01/06/2023]
Abstract
In vertebrates, multipotent progenitors located in the pharyngeal mesoderm form cardiomyocytes and branchiomeric head muscles, but the dynamic gene expression programmes and mechanisms underlying cardiopharyngeal multipotency and heart versus head muscle fate choices remain elusive. Here, we used single-cell genomics in the simple chordate model Ciona to reconstruct developmental trajectories forming first and second heart lineages and pharyngeal muscle precursors and characterize the molecular underpinnings of cardiopharyngeal fate choices. We show that FGF-MAPK signalling maintains multipotency and promotes the pharyngeal muscle fate, whereas signal termination permits the deployment of a pan-cardiac programme, shared by the first and second heart lineages, to define heart identity. In the second heart lineage, a Tbx1/10-Dach pathway actively suppresses the first heart lineage programme, conditioning later cell diversity in the beating heart. Finally, cross-species comparisons between Ciona and the mouse evoke the deep evolutionary origins of cardiopharyngeal networks in chordates.
Collapse
|
45
|
Involvement of CXCR4 in Normal and Abnormal Development. Cells 2019; 8:cells8020185. [PMID: 30791675 PMCID: PMC6406665 DOI: 10.3390/cells8020185] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 01/30/2019] [Accepted: 02/13/2019] [Indexed: 02/06/2023] Open
Abstract
CXC motif chemokine receptor type 4 (CXCR4) is associated with normal and abnormal development, including oncogenesis. The ligand of CXCR4 is stromal cell-derived factor (SDF), also known as CXC motif ligand (CXCL) 12. Through the SDF-1/CXCR4 axis, both homing and migration of hematopoietic (stem) cells are regulated through niches in the bone marrow. Outside of the bone marrow, however, SDF-1 can recruit CXCR4-positive cells from the bone marrow. SDF/CXCR4 has been implicated in the maintenance and/or differentiation of stemness, and tissue-derived stem cells can be associated with SDF-1 and CXCR4 activity. CXCR4 plays a role in multiple pathways involved in carcinogenesis and other pathologies. Here, we summarize reports detailing the functions of CXCR4. We address the molecular signature of CXCR4 and how this molecule and cells expressing it are involved in either normal (maintaining stemness or inducing differentiation) or abnormal (developing cancer and other pathologies) events. As a constituent of stem cells, the SDF-1/CXCR4 axis influences downstream signal transduction and the cell microenvironment.
Collapse
|
46
|
Hasten E, McDonald-McGinn DM, Crowley TB, Zackai E, Emanuel BS, Morrow BE, Racedo SE. Dysregulation of TBX1 dosage in the anterior heart field results in congenital heart disease resembling the 22q11.2 duplication syndrome. Hum Mol Genet 2019; 27:1847-1857. [PMID: 29509905 DOI: 10.1093/hmg/ddy078] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 02/27/2018] [Indexed: 12/27/2022] Open
Abstract
Non-allelic homologous recombination events on chromosome 22q11.2 during meiosis can result in either the deletion (22q11.2DS) or duplication (22q11.2DupS) syndrome. Although the spectrum and frequency of congenital heart disease (CHD) are known for 22q11.2DS, there is less known for 22q11.2DupS. We now evaluated cardiac phenotypes in 235 subjects with 22q11.2DupS including 102 subjects we collected and 133 subjects that were previously reported as a confirmation and found 25% have CHD, mostly affecting the cardiac outflow tract (OFT). Previous studies have shown that global loss or gain of function (LOF; GOF) of mouse Tbx1, encoding a T-box transcription factor mapping to the region of synteny to 22q11.2, results in similar OFT defects. To further evaluate Tbx1 function in the progenitor cells forming the cardiac OFT, termed the anterior heart field, Tbx1 was overexpressed using the Mef2c-AHF-Cre driver (Tbx1 GOF). Here we found that all resulting conditional GOF embryos had a persistent truncus arteriosus (PTA), similar to what was previously reported for conditional Tbx1 LOF mutant embryos. To understand the basis for the PTA in the conditional GOF embryos, we found that proliferation in the Mef2c-AHF-Cre lineage cells before migrating to the heart, was reduced and critical genes were oppositely changed in this tissue in Tbx1 GOF embryos versus conditional LOF embryos. These results suggest that a major function of TBX1 in the AHF is to maintain the normal balance of expression of key cardiac developmental genes required to form the aorta and pulmonary trunk, which is disrupted in 22q11.2DS and 22q11.2DupS.
Collapse
Affiliation(s)
- Erica Hasten
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Donna M McDonald-McGinn
- Division of Human Genetics, Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Terrence B Crowley
- Division of Human Genetics, Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Elaine Zackai
- Division of Human Genetics, Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Beverly S Emanuel
- Division of Human Genetics, Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Bernice E Morrow
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Silvia E Racedo
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
47
|
Heude E, Tesarova M, Sefton EM, Jullian E, Adachi N, Grimaldi A, Zikmund T, Kaiser J, Kardon G, Kelly RG, Tajbakhsh S. Unique morphogenetic signatures define mammalian neck muscles and associated connective tissues. eLife 2018; 7:40179. [PMID: 30451684 PMCID: PMC6310459 DOI: 10.7554/elife.40179] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 11/17/2018] [Indexed: 12/16/2022] Open
Abstract
In vertebrates, head and trunk muscles develop from different mesodermal populations and are regulated by distinct genetic networks. Neck muscles at the head-trunk interface remain poorly defined due to their complex morphogenesis and dual mesodermal origins. Here, we use genetically modified mice to establish a 3D model that integrates regulatory genes, cell populations and morphogenetic events that define this transition zone. We show that the evolutionary conserved cucullaris-derived muscles originate from posterior cardiopharyngeal mesoderm, not lateral plate mesoderm, and we define new boundaries for neural crest and mesodermal contributions to neck connective tissue. Furthermore, lineage studies and functional analysis of Tbx1- and Pax3-null mice reveal a unique developmental program for somitic neck muscles that is distinct from that of somitic trunk muscles. Our findings unveil the embryological and developmental requirements underlying tetrapod neck myogenesis and provide a blueprint to investigate how muscle subsets are selectively affected in some human myopathies.
Collapse
Affiliation(s)
- Eglantine Heude
- Department of Developmental and Stem Cell Biology, Institut Pasteur, Paris, France.,CNRS UMR 3738, Paris, France
| | - Marketa Tesarova
- Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic
| | - Elizabeth M Sefton
- Department of Human Genetics, University of Utah, Salt Lake City, United States
| | - Estelle Jullian
- Aix-Marseille Université, CNRS UMR 7288, IBDM, Marseille, France
| | - Noritaka Adachi
- Aix-Marseille Université, CNRS UMR 7288, IBDM, Marseille, France
| | - Alexandre Grimaldi
- Department of Developmental and Stem Cell Biology, Institut Pasteur, Paris, France.,CNRS UMR 3738, Paris, France
| | - 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
| | - Gabrielle Kardon
- Department of Human Genetics, University of Utah, Salt Lake City, United States
| | - Robert G Kelly
- Aix-Marseille Université, CNRS UMR 7288, IBDM, Marseille, France
| | - Shahragim Tajbakhsh
- Department of Developmental and Stem Cell Biology, Institut Pasteur, Paris, France.,CNRS UMR 3738, Paris, France
| |
Collapse
|
48
|
Calmont A, Anderson N, Suntharalingham JP, Ang R, Tinker A, Scambler PJ. Defective Vagal Innervation in Murine Tbx1 Mutant Hearts. J Cardiovasc Dev Dis 2018; 5:jcdd5040049. [PMID: 30249045 PMCID: PMC6306933 DOI: 10.3390/jcdd5040049] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 09/17/2018] [Accepted: 09/20/2018] [Indexed: 02/07/2023] Open
Abstract
Haploinsufficiency of the T-box transcription factor TBX1 is responsible for many features of 22q11.2 deletion syndrome. Tbx1 is expressed dynamically in the pharyngeal apparatus during mouse development and Tbx1 homozygous mutants display numerous severe defects including abnormal cranial ganglion formation and neural crest cell defects. These abnormalities prompted us to investigate whether parasympathetic (vagal) innervation of the heart was affected in Tbx1 mutant embryos. In this report, we used an allelic series of Tbx1 mouse mutants, embryo tissue explants and cardiac electrophysiology to characterise, in detail, the function of Tbx1 in vagal innervation of the heart. We found that total nerve branch length was significantly reduced in Tbx1+/- and Tbx1neo2/- mutant hearts expressing 50% and 15% levels of Tbx1. We also found that neural crest cells migrated normally to the heart of Tbx1+/-, but not in Tbx1neo2 mutant embryos. In addition, we showed that cranial ganglia IXth and Xth were fused in Tbx1neo2/- but neuronal differentiation appeared intact. Finally, we used telemetry to monitor heart response to carbachol, a cholinergic receptor agonist, and found that heart rate recovered more quickly in Tbx1+/- animals versus controls. We speculate that this condition of decreased parasympathetic drive could result in a pro-arrhythmic substrate in some 22q11.2DS patients.
Collapse
Affiliation(s)
- Amélie Calmont
- INSERM UMRS 1155, Centre for Kidney Research, 4 Rue de la Chine, 75020 Paris, France.
- UCL Great Ormond Street-Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK.
| | - Naomi Anderson
- UCL Great Ormond Street-Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK.
- William Harvey Heart Centre, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London EC1M 6BQ, UK.
| | | | - Richard Ang
- William Harvey Heart Centre, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London EC1M 6BQ, UK.
- Department of Medicine, Rayne Institute, University College London, London WC1E 6JJ, UK.
| | - Andrew Tinker
- William Harvey Heart Centre, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London EC1M 6BQ, UK.
- Department of Medicine, Rayne Institute, University College London, London WC1E 6JJ, UK.
| | - Peter J Scambler
- UCL Great Ormond Street-Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK.
| |
Collapse
|
49
|
Zoupa M, Xavier GM, Bryan S, Theologidis I, Arno M, Cobourne MT. Gene expression profiling in the developing secondary palate in the absence of Tbx1 function. BMC Genomics 2018; 19:429. [PMID: 29866044 PMCID: PMC5987606 DOI: 10.1186/s12864-018-4782-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 05/11/2018] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Microdeletion of chromosome 22q11 is associated with significant developmental anomalies, including disruption of the cardiac outflow tract, thymic/parathyroid aplasia and cleft palate. Amongst the genes within this region, TBX1 is a major candidate for many of these developmental defects. Targeted deletion of Tbx1 in the mouse has provided significant insight into the function of this transcription factor during early development of the cardiac and pharyngeal systems. However, less is known about its role during palatogenesis. To assess the influence of Tbx1 function on gene expression profile within the developing palate we performed a microarray screen using total RNA isolated from the secondary palate of E13.5 mouse embryos wild type, heterozygous and mutant for Tbx1. RESULTS Expression-level filtering and statistical analysis revealed a total of 577 genes differentially expressed across genotypes. Data were clustered into 3 groups based on comparison between genotypes. Group A was composed of differentially expressed genes in mutant compared to wild type (n = 89); Group B included differentially expressed genes in heterozygous compared to wild type (n = 400) and Group C included differentially expressed genes in mutant compared to heterozygous (n = 88). High-throughput quantitative real-time PCR (RT-PCR) confirmed a total of 27 genes significantly changed between wild type and mutant; and 27 genes between heterozygote and mutant. Amongst these, the majority were present in both groups A and C (26 genes). Associations existed with hypertrophic cardiomyopathy, cardiac muscle contraction, dilated cardiomyopathy, focal adhesion, tight junction and calcium signalling pathways. No significant differences in gene expression were found between wild type and heterozygous palatal shelves. CONCLUSIONS Significant differences in gene expression profile within the secondary palate of wild type and mutant embryos is consistent with a primary role for Tbx1 during palatogenesis.
Collapse
Affiliation(s)
- Maria Zoupa
- Centre for Craniofacial Development and Regeneration, King's College London Dental Institute, Floor 27, Guy's Tower, London, SE1 9RT, UK
| | - Guilherme Machado Xavier
- Centre for Craniofacial Development and Regeneration, King's College London Dental Institute, Floor 27, Guy's Tower, London, SE1 9RT, UK.,Department of Orthodontics, King's College London Dental Institute, London, UK
| | - Stephanie Bryan
- Department of Orthodontics, King's College London Dental Institute, London, UK
| | - Ioannis Theologidis
- Division of Development and Gene Expression, Institute of Molecular Biology and BiotechnologyFoundation for Research & Technology, Crete, Greece
| | - Matthew Arno
- Genomics Centre, King's College London, London, UK
| | - Martyn T Cobourne
- Centre for Craniofacial Development and Regeneration, King's College London Dental Institute, Floor 27, Guy's Tower, London, SE1 9RT, UK. .,Department of Orthodontics, King's College London Dental Institute, London, UK.
| |
Collapse
|
50
|
Razy-Krajka F, Gravez B, Kaplan N, Racioppi C, Wang W, Christiaen L. An FGF-driven feed-forward circuit patterns the cardiopharyngeal mesoderm in space and time. eLife 2018; 7:e29656. [PMID: 29431097 PMCID: PMC5809146 DOI: 10.7554/elife.29656] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 01/26/2018] [Indexed: 12/16/2022] Open
Abstract
In embryos, multipotent progenitors divide to produce distinct progeny and express their full potential. In vertebrates, multipotent cardiopharyngeal progenitors produce second-heart-field-derived cardiomyocytes, and branchiomeric skeletal head muscles. However, the mechanisms underlying these early fate choices remain largely elusive. The tunicate Ciona emerged as an attractive model to study early cardiopharyngeal development at high resolution: through two asymmetric and oriented divisions, defined cardiopharyngeal progenitors produce distinct first and second heart precursors, and pharyngeal muscle (aka atrial siphon muscle, ASM) precursors. Here, we demonstrate that differential FGF-MAPK signaling distinguishes between heart and ASM precursors. We characterize a feed-forward circuit that promotes the successive activations of essential ASM determinants, Hand-related, Tbx1/10 and Ebf. Finally, we show that coupling FGF-MAPK restriction and cardiopharyngeal network deployment with cell divisions defines the timing of gene expression and permits the emergence of diverse cell types from multipotent progenitors.
Collapse
Affiliation(s)
- Florian Razy-Krajka
- Center for Developmental Genetics, Department of BiologyCollege of Arts and Science, New York UniversityNew YorkUnited States
| | - Basile Gravez
- Center for Developmental Genetics, Department of BiologyCollege of Arts and Science, New York UniversityNew YorkUnited States
| | - Nicole Kaplan
- Center for Developmental Genetics, Department of BiologyCollege of Arts and Science, New York UniversityNew YorkUnited States
| | - Claudia Racioppi
- Center for Developmental Genetics, Department of BiologyCollege of Arts and Science, New York UniversityNew YorkUnited States
| | - Wei Wang
- Center for Developmental Genetics, Department of BiologyCollege of Arts and Science, New York UniversityNew YorkUnited States
| | - Lionel Christiaen
- Center for Developmental Genetics, Department of BiologyCollege of Arts and Science, New York UniversityNew YorkUnited States
| |
Collapse
|