Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 88
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Proc Natl Acad Sci U S A ; 120(37): e2307658120, 2023 09 12.
Article in English | MEDLINE | ID: mdl-37669370

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.


Subject(s)
Heart Ventricles , Myocytes, Cardiac , Female , Pregnancy , Animals , Mice , Embryonic Development , Myocardium , Regulatory Sequences, Nucleic Acid , Transcription Factors , Repressor Proteins , Basic Helix-Loop-Helix Transcription Factors
2.
Dev Dyn ; 253(1): 107-118, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37042466

ABSTRACT

BACKGROUND: Endocardial cushion tissue is primordia of the valves and septa of the adult heart, and its malformation causes various congenital heart diseases (CHDs). Tricuspid atresia (TA) is defined as congenital absence or agenesis of the tricuspid valve caused by endocardial cushion defects. However, little is known about what type of endocardial cushion defect causes TA. RESULTS: Using three-dimensional volume rendering image analysis, we demonstrated morphological changes of endocardial cushion tissue in developing Hey2/Hrt2 KO mouse embryos that showed malformation of the tricuspid valve, which resembled human TA at neonatal period. In control embryos, atrioventricular (AV) endocardial cushion tissues showed rightward shift to form a tricuspid valve. However, the rightward shift of endocardial cushion tissue was disrupted in Hey2/Hrt2 KO embryos, leading to the misalignment of AV cushions. We also found that muscular tissue filled up the space between the right atrium and ventricle, resulting in the absence of the tricuspid valve. Moreover, analysis using tissue-specific conditional KO mice showed that HEY2/HRT2-expressing myocardium may physically regulate the AV shift. CONCLUSION: Disruption of rightward cushion movement is an initial cue of TA phenotype, and myocardial HEY2/HRT2 is necessary for the regulation of proper alignment of AV endocardial cushion tissue.


Subject(s)
Endocardial Cushion Defects , Tricuspid Atresia , Animals , Mice , Humans , Heart , Myocardium , Transcription Factors , Basic Helix-Loop-Helix Transcription Factors/genetics , Repressor Proteins
3.
Adv Exp Med Biol ; 1441: 885-900, 2024.
Article in English | MEDLINE | ID: mdl-38884757

ABSTRACT

The process of valve formation is a complex process that involves intricate interplay between various pathways at precise times. Although we have not completely elucidated the molecular pathways that lead to normal valve formation, we have identified a few major players in this process. We are now able to implicate TGF-ß, BMP, and NOTCH as suspects in tricuspid atresia (TA), as well as their downstream targets: NKX2-5, TBX5, NFATC1, GATA4, and SOX9. We know that the TGF-ß and the BMP pathways converge on the SMAD4 molecule, and we believe that this molecule plays a very important role to tie both pathways to TA. Similarly, we look at the NOTCH pathway and identify the HEY2 as a potential link between this pathway and TA. Another transcription factor that has been implicated in TA is NFATC1. While several mouse models exist that include part of the TA abnormality as their phenotype, no true mouse model can be said to represent TA. Bridging this gap will surely shed light on this complex molecular pathway and allow for better understanding of the disease process.


Subject(s)
Disease Models, Animal , Signal Transduction , Tricuspid Atresia , Animals , Tricuspid Atresia/genetics , Tricuspid Atresia/metabolism , Tricuspid Atresia/pathology , Humans , Mice , Univentricular Heart/genetics , Univentricular Heart/metabolism , Univentricular Heart/physiopathology , Univentricular Heart/pathology , NFATC Transcription Factors/metabolism , NFATC Transcription Factors/genetics , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/genetics , Receptors, Notch/metabolism , Receptors, Notch/genetics
4.
Adv Exp Med Biol ; 1441: 875-884, 2024.
Article in English | MEDLINE | ID: mdl-38884756

ABSTRACT

Tricuspid atresia (TA) is a rare congenital heart condition that presents with a complete absence of the right atrioventricular valve. Because of the rarity of familial and/or isolated cases of TA, little is known about the potential genetic abnormalities contributing to this condition. Potential responsible chromosomal abnormalities were identified in exploratory studies and include deletions in 22q11, 4q31, 8p23, and 3p as well as trisomies 13 and 18. In parallel, potential culprit genes include the ZFPM2, HEY2, NFATC1, NKX2-5, MYH6, and KLF13 genes. The aim of this chapter is to expose the genetic components that are potentially involved in the pathogenesis of TA in humans. The large variability in phenotypes and genotypes among cases of TA suggests a genetic network that involves many components yet to be unraveled.


Subject(s)
Tricuspid Atresia , Humans , Chromosome Aberrations , Phenotype , Tricuspid Atresia/genetics , Univentricular Heart/genetics
5.
Adv Exp Med Biol ; 1441: 505-534, 2024.
Article in English | MEDLINE | ID: mdl-38884729

ABSTRACT

Ventricular septal defects (VSDs) are recognized as one of the commonest congenital heart diseases (CHD), accounting for up to 40% of all cardiac malformations, and occur as isolated CHDs as well as together with other cardiac and extracardiac congenital malformations in individual patients and families. The genetic etiology of VSD is complex and extraordinarily heterogeneous. Chromosomal abnormalities such as aneuploidy and structural variations as well as rare point mutations in various genes have been reported to be associated with this cardiac defect. This includes both well-defined syndromes with known genetic cause (e.g., DiGeorge syndrome and Holt-Oram syndrome) and so far undefined syndromic forms characterized by unspecific symptoms. Mutations in genes encoding cardiac transcription factors (e.g., NKX2-5 and GATA4) and signaling molecules (e.g., CFC1) have been most frequently found in VSD cases. Moreover, new high-resolution methods such as comparative genomic hybridization enabled the discovery of a high number of different copy number variations, leading to gain or loss of chromosomal regions often containing multiple genes, in patients with VSD. In this chapter, we will describe the broad genetic heterogeneity observed in VSD patients considering recent advances in this field.


Subject(s)
Heart Septal Defects, Ventricular , Humans , Chromosome Aberrations , DNA Copy Number Variations/genetics , Genetic Predisposition to Disease/genetics , Heart Septal Defects, Ventricular/genetics , Mutation , Transcription Factors/genetics
6.
Clin Anat ; 37(1): 102-113, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37732460

ABSTRACT

Dick van Velzen practiced as a pediatric pathologist at Alder Hey Children's Hospital in Liverpool, England from September 1988 until December 1995; he then relocated to the IWK-Grace Health Centre, a children's and maternity hospital in Halifax, Nova Scotia, Canada, where he practiced until he was fired for cause in January 1998. About a year and a half later, his practice in Liverpool came under increasing scrutiny, with the initial focus on the massive collection of post-mortem pediatric organs he had accumulated for planned future research on sudden infant death syndrome. Soon, a Parliamentary Inquiry began investigating the full scope of his Liverpool practice. During the Inquiry, another organ-hoarding scandal erupted; van Velzen, when leaving Halifax after his dismissal, had put his family's personal belongings into a storage facility at Burnside Industrial Park and then did not pay bills. As his belongings were being prepared for auction, formalin-fixed organs were found, and a Canada-wide arrest warrant for disrespect for human remains was issued by the Halifax Police. While the Alder Hey scandal resulted in a 535-page-long Parliamentary Report and the Human Tissue Act, van Velzen was never charged criminally in the UK. The largely unknown story of his second organ scandal in Halifax, is related here. Although he had obtained the body parts with the consent of the parents of the child to which they had belonged, his failure to properly identify and store them traumatized parents already impacted by his organ-hoarding in the UK, traumatized additional parents in Halifax, and resulted in significant waste of public resources in investigating the case. He pled guilty to "indignity to a human body" in Canada and was fined and placed on 12 months' probation.


Subject(s)
Human Body , Female , Pregnancy , Humans , Child , Nova Scotia , Autopsy , England
7.
Int J Mol Sci ; 25(10)2024 May 08.
Article in English | MEDLINE | ID: mdl-38791169

ABSTRACT

The Notch pathway is a key cancer driver and is important in tumor progression. Early research suggested that Notch activity was highly dependent on the expression of the intracellular cleaved domain of Notch-1 (NICD). However, recent insights into Notch signaling reveal the presence of Notch pathway signatures, which may vary depending on different cancer types and tumor microenvironments. Herein, we perform a comprehensive investigation of the Notch signaling pathway in adult T-cell leukemia (ATL) primary patient samples. Using gene arrays, we demonstrate that the Notch pathway is constitutively activated in ATL patient samples. Furthermore, the activation of Notch in ATL cells remains elevated irrespective of the presence of activating mutations in Notch itself or its repressor, FBXW7, and that ATL cells are dependent upon Notch-1 expression for proliferation and survival. We demonstrate that ATL cells exhibit the expression of pivotal Notch-related genes, including notch-1, hes1, c-myc, H19, and hes4, thereby defining a critical Notch signature associated with ATL disease. Finally, we demonstrate that lncRNA H19 is highly expressed in ATL patient samples and ATL cells and contributes to Notch signaling activation. Collectively, our results shed further light on the Notch pathway in ATL leukemia and reveal new therapeutic approaches to inhibit Notch activation in ATL cells.


Subject(s)
Leukemia-Lymphoma, Adult T-Cell , MicroRNAs , RNA, Long Noncoding , Signal Transduction , Humans , Leukemia-Lymphoma, Adult T-Cell/genetics , Leukemia-Lymphoma, Adult T-Cell/metabolism , Leukemia-Lymphoma, Adult T-Cell/pathology , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Cell Line, Tumor , Receptor, Notch1/metabolism , Receptor, Notch1/genetics , Gene Expression Regulation, Leukemic , Receptors, Notch/metabolism , Receptors, Notch/genetics , Cell Proliferation/genetics , F-Box-WD Repeat-Containing Protein 7/metabolism , F-Box-WD Repeat-Containing Protein 7/genetics , Gene Expression Regulation, Neoplastic , Adult
8.
Genes Chromosomes Cancer ; 62(3): 171-175, 2023 03.
Article in English | MEDLINE | ID: mdl-36416671

ABSTRACT

HEY1-NCOA2 fusion is most described in mesenchymal chondrosarcoma. This is the first case report of a primary renal spindle cell neoplasm of uncertain malignant potential with a HEY1::NCOA2 fusion identified by Fusionplex RNA-sequencing that is histologically distinct from mesenchymal chondrosarcoma. The neoplasm was identified in a 33-year-old woman without significant past medical history who underwent partial nephrectomy for an incidentally discovered renal mass. The histologic features of the mass included spindle cells with variable cellularity and monotonous bland cytology forming vague fascicles and storiform architecture within a myxoedematous and collagenous stroma with areas of calcification. The morphologic and immunophenotypic features were not specific for any entity but were most similar to low-grade fibromyxoid sarcoma. To date, the patient has not had recurrence, and the malignant potential of the neoplasm is uncertain.


Subject(s)
Bone Neoplasms , Chondrosarcoma, Mesenchymal , Female , Humans , Adult , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Chondrosarcoma, Mesenchymal/genetics , Chondrosarcoma, Mesenchymal/surgery , Chondrosarcoma, Mesenchymal/pathology , Nephrectomy , Nuclear Receptor Coactivator 2/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics
9.
Development ; 147(18)2020 09 28.
Article in English | MEDLINE | ID: mdl-32988975

ABSTRACT

Teleost zebrafish and neonatal mammalian hearts exhibit the remarkable capacity to regenerate through dedifferentiation and proliferation of pre-existing cardiomyocytes (CMs). Although many mitogenic signals that stimulate zebrafish heart regeneration have been identified, transcriptional programs that restrain injury-induced CM renewal are incompletely understood. Here, we report that mutations in gridlock (grl; also known as hey2), encoding a Hairy-related basic helix-loop-helix transcriptional repressor, enhance CM proliferation and reduce fibrosis following damage. In contrast, myocardial grl induction blunts CM dedifferentiation and regenerative responses to heart injury. RNA sequencing analyses uncover Smyd2 lysine methyltransferase (KMT) as a key transcriptional target repressed by Grl. Reduction in Grl protein levels triggered by injury induces smyd2 expression at the wound myocardium, enhancing CM proliferation. We show that Smyd2 functions as a methyltransferase and modulates the Stat3 methylation and phosphorylation activity. Inhibition of the KMT activity of Smyd2 reduces phosphorylated Stat3 at cardiac wounds, suppressing the elevated CM proliferation in injured grl mutant hearts. Our findings establish an injury-specific transcriptional repression program in governing CM renewal during heart regeneration, providing a potential strategy whereby silencing Grl repression at local regions might empower regeneration capacity to the injured mammalian heart.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Heart/physiology , Lysine/genetics , Methyltransferases/genetics , Regeneration/genetics , Transcription, Genetic/genetics , Vertebrates/genetics , Zebrafish Proteins/genetics , Animals , Animals, Newborn , Cell Differentiation/genetics , Cell Proliferation/genetics , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Phosphorylation/genetics , STAT3 Transcription Factor/genetics , Signal Transduction/genetics , Zebrafish/genetics
10.
Exp Dermatol ; 32(12): 2176-2179, 2023 12.
Article in English | MEDLINE | ID: mdl-37649203

ABSTRACT

Dermal papilla (DP) cells are specialized mesenchymal cells that play a crucial role in regulating hair morphology, colour and growth through the secretion of specific factors. It is still unclear what the source of progenitor cells is for dermal cell regeneration during wound healing, and whether DP cells are involved in this process. We analyzed the gene expression profile of various skin cell populations using existing datasets and found that the Hey2 gene was predominantly expressed in DP cells. We introduced Hey2-CreERT2 knockin mice and crossed them with Rosa26-ZsGreen reporter mice. After induction in the double transgenic mice by administration of tamoxifen, the reporter ZsGreen was found to be predominantly expressed in DP cells both at anagen and telogen phases, and broadly expressed in some other dermal cells at anagen. We also created a wound after tamoxifen induction, and found there were abundant ZsGreen+ cells in the regenerated dermis. We conclude that the HEY2+ DP cells and dermal cells exhibit some stemness properties and can contribute to the dermal cell regeneration during wound healing.


Subject(s)
Hair Follicle , Wound Healing , Mice , Animals , Hair Follicle/metabolism , Regeneration , Mice, Transgenic , Cells, Cultured , Tamoxifen/pharmacology , Tamoxifen/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism
11.
J Pathol ; 257(5): 579-592, 2022 08.
Article in English | MEDLINE | ID: mdl-35342947

ABSTRACT

Mesenchymal chondrosarcoma is a rare, high-grade, primitive mesenchymal tumor. It accounts for around 2-10% of all chondrosarcomas and mainly affects adolescents and young adults. We previously described the HEY1-NCOA2 as a recurrent gene fusion in mesenchymal chondrosarcoma, an important breakthrough for characterizing this disease; however, little study had been done to characterize the fusion protein functionally, in large part due to a lack of suitable models for evaluating the impact of HEY1-NCOA2 expression in the appropriate cellular context. We used iPSC-derived mesenchymal stem cells (iPSC-MSCs), which can differentiate into chondrocytes, and generated stable transduced iPSC-MSCs with inducible expression of HEY1-NCOA2 fusion protein, wildtype HEY1 or wildtype NCOA2. We next comprehensively analyzed both the DNA binding properties and transcriptional impact of HEY1-NCOA2 expression by integrating genome-wide chromatin immunoprecipitation sequencing (ChIP-seq) and expression profiling (RNA-seq). We demonstrated that HEY1-NCOA2 fusion protein preferentially binds to promoter regions of canonical HEY1 targets, resulting in transactivation of HEY1 targets, and significantly enhances cell proliferation. Intriguingly, we identified that both PDGFB and PDGFRA were directly targeted and upregulated by HEY1-NCOA2; and the fusion protein, but not wildtype HEY1 or NCOA2, dramatically increased the level of phospho-AKT (Ser473). Our findings provide a rationale for exploring PDGF/PI3K/AKT inhibition in treating mesenchymal chondrosarcoma. © 2022 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Subject(s)
Bone Neoplasms , Chondrosarcoma, Mesenchymal , Adolescent , Basic Helix-Loop-Helix Transcription Factors/genetics , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Carcinogenesis , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Transformation, Neoplastic , Chondrosarcoma, Mesenchymal/genetics , Chondrosarcoma, Mesenchymal/metabolism , Chondrosarcoma, Mesenchymal/pathology , Gene Fusion , Genomics , Humans , Nuclear Receptor Coactivator 2/genetics , Nuclear Receptor Coactivator 2/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Young Adult
12.
Genes Chromosomes Cancer ; 61(11): 670-677, 2022 11.
Article in English | MEDLINE | ID: mdl-35672279

ABSTRACT

BACKGROUND: Mesenchymal chondrosarcoma (MCS) is a rare translocation-associated sarcoma, driven by a canonical HEY1::NCOA2 fusion. The tumors typically have a biphasic phenotype of primitive small blue round cells intermixed with hyaline cartilage. The head and neck (HN) region is a common site for MCS, accounting for 12-45% of all cases reported. AIMS: We assembled a relatively large cohort of 13 molecularly confirmed HN MCS for a detailed clinicopathologic analysis. The underlying fusion events were determined using fluorescence in situ hybridization and/or targeted RNA sequencing. RESULTS: The median age of presentation was 19 years. Five MCSs (39%) had an intraosseous presentation (skull, maxilla, palate, and mandible), while the remaining eight cases occurred in the brain/meninges, orbit, and nasal cavity. Microscopically, HN MCSs were characterized by primitive round cells arranged in a distinctive nested architecture and a rich staghorn vasculature. A cartilaginous component of hyaline cartilage islands and/or single chondrocytes were present in 69% cases. A combined immunoprofile of CD99(+)/SATB2(+)/CD34(-)/STAT6(-) was typically noted. As this immunoprofile is non-specific, the referral diagnoses in cases lacking a cartilaginous component included Ewing sarcoma family and osteosarcoma. Among the seven patients with follow-up data, three developed distant metastasis and one died of disease. CONCLUSION: HN MCS may arise at intra- or extra-osseous sites. The HN MCS appears to have a more prolonged survival compared other MCS sites. Testing for HEY1::NCOA2 fusion is recommended in HN tumors with nested round cell morphology and staghorn vasculature that lack a distinctive cartilaginous component.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors , Chondrosarcoma, Mesenchymal , Gene Fusion , Head and Neck Neoplasms , Nuclear Receptor Coactivator 2 , Adult , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Cycle Proteins/genetics , Child , Chondrosarcoma, Mesenchymal/genetics , Chondrosarcoma, Mesenchymal/pathology , Female , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/pathology , Humans , In Situ Hybridization, Fluorescence , Male , Nuclear Receptor Coactivator 2/genetics , Young Adult
13.
Neurochem Res ; 47(5): 1442-1458, 2022 May.
Article in English | MEDLINE | ID: mdl-35316462

ABSTRACT

To investigate the function of hairy/enhancer-of-split related with YRPW motif protein 1 (HEY1) and Notch receptor 3 (NOTCH3) in ischemic stroke. Stroke models were established by middle cerebral artery occlusion (MCAO) and oxygen glucose deprivation (OGD) in rats and rat brain microvascular endothelial cells (BMVECs), respectively. Neurological deficit evaluation and 2,3,5-triphenyltetrazolium chloride staining were used to assess cerebral injury. The expression of HEY1 and NOTCH3 was manipulated using gain and loss of function approaches. Terminal deoxynucleotidyl transferase dUTP nick end labeling and Western blotting analysis of cleaved caspase-3 and B-cell lymphoma-2 (Bcl2) were used to evaluate apoptosis. Enzyme-linked immunosorbent assay was performed to measure the expression levels of interleukin (IL)-1ß, IL-6 and IL-18. The proliferation and migration of BMVECs were analyzed by Ki-67 immunofluorescence and scratch assay, respectively. Tube formation assay was conducted to measure the length of capillary-like tubes formed by BMVECs. Co-immunoprecipitation was used to testify the relationship between HEY1 and NOTCH3. HEY1 and NOTCH3 were upregulated in MCAO and OGD models. HEY1 ameliorated ischemic injuries in MCAO rats. Knockdown of HEY1 or NOTCH3 promoted OGD-induced apoptosis and inflammation and inhibited proliferation and migration in BMVECs. NOTCH3 was a binding protein of HEY1. Overexpression of HEY1 offset the disease-promoting effect of NOTCH3 silencing. HEY1 suppresses apoptosis and inflammation and promotes proliferation and migration in BMVECs by upregulating NOTCH3, thereby ameliorating ischemic stroke.


Subject(s)
Ischemic Stroke , Stroke , Animals , Brain/metabolism , Endothelial Cells/metabolism , Rats , Receptor, Notch3/metabolism , Stroke/metabolism , Transcription Factors/metabolism
14.
Dev Biol ; 461(2): 124-131, 2020 05 15.
Article in English | MEDLINE | ID: mdl-32035085

ABSTRACT

Development of multi-chambered heart is associated with spatio-temporal regulation of gene expression. A basic helix-loop-helix transcription factor Hey2 is specifically expressed in the embryonic mouse ventricles and is indispensable for ventricular myocyte differentiation, compartment identity and morphogenesis of the heart. However, how Hey2 transcription is precisely regulated in the heart remains unclear. In this study, we identified a distal Hey2 enhancer conserved in the mouse and human to possess specific transcriptional activity in ventricular free wall myocytes at the looping stage of cardiac development. Deletion of the enhancer significantly decreased endogenous Hey2 expression in the ventricular myocardium but not in other tissues of mouse embryos. Mutation/deletion of the conserved binding sites for T-box and Gata proteins, but not NK-2 proteins, abolished the enhancer activity, and Tbx20 null mice completely lost the enhancer activity in the embryonic ventricles. Luciferase reporter analysis suggested that the ventricular enhancer activity was controlled by Tbx20 through its DNA binding and cooperative function with cardiac Gata proteins. These results delineate a regulatory mechanism of ventricular Hey2 expression and help fully understand molecular cascades in myocardial cell differentiation and cardiac morphogenesis during embryonic development.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Enhancer Elements, Genetic , GATA4 Transcription Factor/physiology , Gene Expression Regulation, Developmental , Heart Ventricles/embryology , Repressor Proteins/biosynthesis , T-Box Domain Proteins/physiology , Animals , Base Sequence , Basic Helix-Loop-Helix Transcription Factors/genetics , Conserved Sequence , Genes, Reporter , Heart Ventricles/metabolism , Humans , Mammals/genetics , Mice , Mice, Transgenic , Repressor Proteins/genetics , Sequence Alignment , Sequence Deletion , Sequence Homology, Nucleic Acid , Species Specificity
15.
J Biol Chem ; 295(51): 17632-17645, 2020 12 18.
Article in English | MEDLINE | ID: mdl-33454003

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.


Subject(s)
Cell Cycle Proteins/metabolism , Endothelium/metabolism , Receptors, Notch/metabolism , Animals , Arteries/growth & development , Arteries/metabolism , Branchial Region/blood supply , Branchial Region/growth & development , Cell Cycle Proteins/deficiency , Cell Cycle Proteins/genetics , Cell Differentiation , Embryo, Mammalian/metabolism , Endothelium/cytology , Female , Humans , Mice , Mice, Knockout , Morphogenesis , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , RNA, Guide, Kinetoplastida/metabolism , Regulatory Sequences, Nucleic Acid , Signal Transduction , Transcriptional Activation
16.
Am J Physiol Gastrointest Liver Physiol ; 320(5): G816-G828, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33236952

ABSTRACT

Upregulating the expression of long noncoding RNA LINC00982 controlled cell proliferation in gastric cancer, but the regulatory molecular mechanisms are yet to be expounded. We here aimed to elaborate how LINC00982 regulated the malignancy of gastric cancer cells. RT-qPCR and Western blot analysis were used to detect the expression of LINC00982 and cathepsin F (CTSF) in gastric cancer tissues and cells. Modulatory effect of LINC00982 on gastric cancer cells was assessed by CCK-8, colony formation, Transwell migration, and invasion assays. The relationship between LINC00982, YRPW motif 1 (HEY1), and CTSF was examined by RNA-binding protein immunoprecipitation, luciferase assay, and chromatin immunoprecipitation, and their interaction in the regulation of gastric cancer cellular functions was analyzed by performing gain-of-function and rescue assays. The nude mouse model of tumor formation was developed to examine the effects of LINC00982 on tumorigenesis. LINC00982 was lowly expressed in gastric cancer tissues, whereas its overexpression impaired the proliferative, migratory, and invasive properties of gastric cancer cells. Furthermore, LINC00982 could bind to transcription factor HEY1 and inhibited its expression. Through blocking the binding of HEY1 to CTSF promoter, LINC00982 promoted the expression of CTSF. Overexpression of HEY1 or inhibition of CTSF could reverse the antitumor effects of LINC00982 on gastric cancer, which were further demonstrated in vivo. All these taken together, LINC00982 acted as a tumor suppressor in gastric cancer, which is therefore suggested to be a potential antitumor target for gastric cancer.NEW & NOTEWORTHY We identified LINC00982 as a promising antitumor target for the treatment of patients with gastric cancer. We also determined a regulatory network involved in the pathophysiology of gastric cancer wherein LINC00982 could bind to HEY1 to impair its binding to cathepsin F (CTSF) promoter and hence promote CTSF expression, which aids in better understanding of molecular mechanisms related to gastric tumorigenesis.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cathepsin F/metabolism , Cell Cycle Proteins/metabolism , RNA, Long Noncoding/metabolism , Stomach Neoplasms/metabolism , Up-Regulation , Aged , Basic Helix-Loop-Helix Transcription Factors/genetics , Biomarkers, Tumor , Carcinogenesis/genetics , Carcinogenesis/pathology , Cathepsin F/genetics , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Databases, Factual , Disease Progression , Female , Humans , Male , Middle Aged , RNA, Long Noncoding/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology
17.
Development ; 145(22)2018 11 21.
Article in English | MEDLINE | ID: mdl-30355727

ABSTRACT

A key event in heart development is the timely addition of cardiac progenitor cells, defects in which can lead to congenital heart defects. However, how the balance and proportion of progenitor proliferation versus addition to the heart is regulated remains poorly understood. Here, we demonstrate that Hey2 functions to regulate the dynamics of cardiac progenitor addition to the zebrafish heart. We found that the previously noted increase in myocardial cell number found in the absence of Hey2 function was due to a pronounced expansion in the size of the cardiac progenitor pool. Expression analysis and lineage tracing of hey2-expressing cells showed that hey2 is active in cardiac progenitors. Hey2 acted to limit proliferation of cardiac progenitors, prior to heart tube formation. Use of a transplantation approach demonstrated a likely cell-autonomous (in cardiac progenitors) function for Hey2. Taken together, our data suggest a previously unappreciated role for Hey2 in controlling the proliferative capacity of cardiac progenitors, affecting the subsequent contribution of late-differentiating cardiac progenitors to the developing vertebrate heart.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Heart/embryology , Stem Cells/cytology , Stem Cells/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Zebrafish/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cardiovascular Diseases/pathology , Cell Count , Cell Lineage , Cell Proliferation , Cell Size , Fibroblast Growth Factors/metabolism , Gene Expression Regulation, Developmental , Mutation/genetics , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Signal Transduction , Zebrafish/genetics , Zebrafish Proteins/genetics
18.
Development ; 145(10)2018 05 15.
Article in English | MEDLINE | ID: mdl-29695612

ABSTRACT

Neural stem cells (NSCs) in the adult vertebrate brain are found in a quiescent state and can preserve long-lasting progenitor potential (stemness). Whether and how these two properties are linked, and to what extent they can be independently controlled by NSC maintenance pathways, is unresolved. We have previously identified Notch3 signalling as a major quiescence-promoting pathway in adult NSCs of the zebrafish pallium. We now show that Notch3 also controls NSC stemness. Using parallel transcriptomic characterizations of notch3 mutant NSCs and adult NSC physiological states, we demonstrate that a set of potentially direct Notch3 target genes distinguishes quiescence and stemness control. As a proof of principle, we focus on one 'stemness' target, encoding the bHLH transcription factor Hey1, that has not yet been analysed in adult NSCs. We show that abrogation of Hey1 function in adult pallial NSCs in vivo, including quiescent NSCs, leads to their differentiation without affecting their proliferation state. These results demonstrate that quiescence and stemness are molecularly distinct outputs of Notch3 signalling, and identify Hey1 as a major Notch3 effector controlling NSC stemness in the vertebrate adult brain.


Subject(s)
Brain/metabolism , Neural Stem Cells/cytology , Neurogenesis/physiology , Receptor, Notch3/metabolism , Zebrafish Proteins/metabolism , Animals , Animals, Genetically Modified , Basic Helix-Loop-Helix Transcription Factors , Cell Differentiation/genetics , Cell Proliferation/physiology , Gene Knockout Techniques , Neurogenesis/genetics , Receptor, Notch3/genetics , Signal Transduction/physiology , Zebrafish , Zebrafish Proteins/genetics
19.
Genet Med ; 23(1): 103-110, 2021 01.
Article in English | MEDLINE | ID: mdl-32820247

ABSTRACT

PURPOSE: In this study we aimed to establish the genetic cause of a myriad of cardiovascular defects prevalent in individuals from a genetically isolated population, who were found to share a common ancestor in 1728. METHODS: Trio genome sequencing was carried out in an index patient with critical congenital heart disease (CHD); family members had either exome or Sanger sequencing. To confirm enrichment, we performed a gene-based association test and meta-analysis in two independent validation cohorts: one with 2685 CHD cases versus 4370 . These controls were also ancestry-matched (same as FTAA controls), and the other with 326 cases with familial thoracic aortic aneurysms (FTAA) and dissections versus 570 ancestry-matched controls. Functional consequences of identified variants were evaluated using expression studies. RESULTS: We identified a loss-of-function variant in the Notch target transcription factor-encoding gene HEY2. The homozygous state (n = 3) causes life-threatening congenital heart defects, while 80% of heterozygous carriers (n = 20) had cardiovascular defects, mainly CHD and FTAA of the ascending aorta. We confirm enrichment of rare risk variants in HEY2 functional domains after meta-analysis (MetaSKAT p = 0.018). Furthermore, we show that several identified variants lead to dysregulation of repression by HEY2. CONCLUSION: A homozygous germline loss-of-function variant in HEY2 leads to critical CHD. The majority of heterozygotes show a myriad of cardiovascular defects.


Subject(s)
Aortic Aneurysm, Thoracic , Heart Defects, Congenital , Aortic Aneurysm, Thoracic/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Genetic Predisposition to Disease , Germ Cells , Heart Defects, Congenital/genetics , Humans , Pedigree , Repressor Proteins
20.
Dev Growth Differ ; 63(1): 82-92, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33410138

ABSTRACT

A basic helix-loop-helix transcription factor Hey2 is expressed in the ventricular myocardium and endocardium of mouse embryos, and Hey2 null mice die perinatally showing ventricular septal defect, dysplastic tricuspid valve and hypoplastic right ventricle. In order to understand region-specific roles of Hey2 during cardiac morphogenesis, we generated Hey2 conditional knockout (cKO) mice using Mef2c-AHF-Cre, which was active in the anterior part of the second heart field and the right ventricle and outflow tract of the heart. Hey2 cKO neonates reproduced three anomalies commonly observed in Hey2 null mice. An earliest morphological defect was the lack of right ventricular extension along the apico-basal axis at midgestational stages. Underdevelopment of the right ventricle was present in all cKO neonates including those without apparent atresia of right-sided atrioventricular connection. RNA sequencing analysis of cKO embryos identified that the gene expression of a non-chamber T-box factor Tbx2 was ectopically induced in the chamber myocardium of the right ventricle. Consistently, mRNA expression of the Mycn transcription factor, which was a cell cycle regulator transcriptionally repressed by Tbx2, was down regulated, and the number of S-phase cells was significantly decreased in the right ventricle of cKO heart. These results suggest that Hey2 plays an important role in right ventricle development during cardiac morphogenesis, at least in part, through mitigating Tbx2-dependent inhibition of Mycn expression.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Heart Ventricles/growth & development , Heart/growth & development , N-Myc Proto-Oncogene Protein/metabolism , Repressor Proteins/metabolism , T-Box Domain Proteins/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/deficiency , Female , Heart Ventricles/metabolism , Male , Mice , Mice, Knockout , Mice, Transgenic , Morphogenesis , N-Myc Proto-Oncogene Protein/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Repressor Proteins/deficiency , T-Box Domain Proteins/genetics , Ventricular Function, Right
SELECTION OF CITATIONS
SEARCH DETAIL