Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 145
Filtrar
1.
Cell ; 185(5): 794-814.e30, 2022 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-35182466

RESUMEN

Congenital heart disease (CHD) is present in 1% of live births, yet identification of causal mutations remains challenging. We hypothesized that genetic determinants for CHDs may lie in the protein interactomes of transcription factors whose mutations cause CHDs. Defining the interactomes of two transcription factors haplo-insufficient in CHD, GATA4 and TBX5, within human cardiac progenitors, and integrating the results with nearly 9,000 exomes from proband-parent trios revealed an enrichment of de novo missense variants associated with CHD within the interactomes. Scoring variants of interactome members based on residue, gene, and proband features identified likely CHD-causing genes, including the epigenetic reader GLYR1. GLYR1 and GATA4 widely co-occupied and co-activated cardiac developmental genes, and the identified GLYR1 missense variant disrupted interaction with GATA4, impairing in vitro and in vivo function in mice. This integrative proteomic and genetic approach provides a framework for prioritizing and interrogating genetic variants in heart disease.


Asunto(s)
Factor de Transcripción GATA4/metabolismo , Cardiopatías Congénitas , Proteínas Nucleares/metabolismo , Oxidorreductasas/metabolismo , Factores de Transcripción , Animales , Cardiopatías Congénitas/genética , Ratones , Mutación , Proteómica , Proteínas de Dominio T Box/genética , Factores de Transcripción/genética
2.
Cell ; 167(7): 1734-1749.e22, 2016 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-27984724

RESUMEN

Mutation of highly conserved residues in transcription factors may affect protein-protein or protein-DNA interactions, leading to gene network dysregulation and human disease. Human mutations in GATA4, a cardiogenic transcription factor, cause cardiac septal defects and cardiomyopathy. Here, iPS-derived cardiomyocytes from subjects with a heterozygous GATA4-G296S missense mutation showed impaired contractility, calcium handling, and metabolic activity. In human cardiomyocytes, GATA4 broadly co-occupied cardiac enhancers with TBX5, another transcription factor that causes septal defects when mutated. The GATA4-G296S mutation disrupted TBX5 recruitment, particularly to cardiac super-enhancers, concomitant with dysregulation of genes related to the phenotypic abnormalities, including cardiac septation. Conversely, the GATA4-G296S mutation led to failure of GATA4 and TBX5-mediated repression at non-cardiac genes and enhanced open chromatin states at endothelial/endocardial promoters. These results reveal how disease-causing missense mutations can disrupt transcriptional cooperativity, leading to aberrant chromatin states and cellular dysfunction, including those related to morphogenetic defects.


Asunto(s)
Factor de Transcripción GATA4/genética , Cardiopatías Congénitas/genética , Cardiopatías Congénitas/patología , Cromatina , Elementos de Facilitación Genéticos , Femenino , Corazón/crecimiento & desarrollo , Humanos , Células Madre Pluripotentes Inducidas , Masculino , Mutación Missense , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal , Proteínas de Dominio T Box/genética
3.
J Neurosci ; 44(39)2024 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-39174351

RESUMEN

Nerve injury can induce aberrant changes in the spine; these changes are due to, or at least partly governed by, transcription factors that contribute to the genesis of neuropathic allodynia. Here, we showed that spinal nerve ligation (SNL, a clinical neuropathic allodynia model) increased the expression of the transcription factor Tbx5 in the injured dorsal horn in male Sprague Dawley rats. In contrast, blocking this upregulation alleviated SNL-induced mechanical allodynia, and there was no apparent effect on locomotor function. Moreover, SNL-induced Tbx5 upregulation promoted the recruitment and interaction of GATA4 and Brd4 by enhancing its binding activity to H3K9Ac, which was enriched at the Trpv1 promotor, leading to an increase in TRPV1 transcription and the development of neuropathic allodynia. In addition, nerve injury-induced expression of Fbxo3, which abates Fbxl2-dependent Tbx5 ubiquitination, promoted the subsequent Tbx5-dependent epigenetic modification of TRPV1 expression during SNL-induced neuropathic allodynia. Collectively, our findings indicated that spinal Tbx5-dependent TRPV1 transcription signaling contributes to the development of neuropathic allodynia via Fbxo3-dependent Fbxl2 ubiquitination and degradation. Thus, we propose a potential medical treatment strategy for neuropathic allodynia by targeting Tbx5.


Asunto(s)
Epigénesis Genética , Hiperalgesia , Neuralgia , Ratas Sprague-Dawley , Asta Dorsal de la Médula Espinal , Proteínas de Dominio T Box , Canales Catiónicos TRPV , Animales , Proteínas de Dominio T Box/metabolismo , Proteínas de Dominio T Box/genética , Masculino , Ratas , Canales Catiónicos TRPV/metabolismo , Canales Catiónicos TRPV/genética , Hiperalgesia/metabolismo , Hiperalgesia/genética , Hiperalgesia/fisiopatología , Neuralgia/metabolismo , Neuralgia/genética , Asta Dorsal de la Médula Espinal/metabolismo
4.
Dev Dyn ; 253(1): 157-172, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37083132

RESUMEN

BACKGROUND: Essential patterning processes transform the heart tube into a compartmentalized organ with distinct chambers separated by an atrioventricular canal (AVC). This transition involves the refinement of expression of genes that are first found broadly throughout the heart tube and then become restricted to the AVC. Despite the importance of cardiac patterning, we do not fully understand the mechanisms that limit gene expression to the AVC. RESULTS: We show that the zebrafish gene smarcc1a, encoding a BAF chromatin remodeling complex subunit homologous to mammalian BAF155, is critical for cardiac patterning. In smarcc1a mutants, myocardial differentiation and heart tube assembly appear to proceed normally. Subsequently, the smarcc1a mutant heart fails to exhibit refinement of gene expression patterns to the AVC, and the persistence of broad gene expression is accompanied by failure of chamber expansion. In addition to their cardiac defects, smarcc1a mutants lack pectoral fins, indicating similarity to tbx5a mutants. However, comparison of smarcc1a and tbx5a mutants suggests that perturbation of tbx5a function is not sufficient to cause the smarcc1a mutant phenotype. CONCLUSIONS: Our data indicate an important role for Smarcc1a-containing chromatin remodeling complexes in regulating the changes in gene expression and morphology that distinguish the AVC from the cardiac chambers.


Asunto(s)
Cojinetes Endocárdicos , Pez Cebra , Animales , Pez Cebra/genética , Pez Cebra/metabolismo , Ensamble y Desensamble de Cromatina , Proteínas de Pez Cebra/metabolismo , Corazón , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Regulación del Desarrollo de la Expresión Génica , Mamíferos/metabolismo
5.
Biochem Biophys Res Commun ; 718: 150037, 2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-38735135

RESUMEN

BACKGROUND: Non-small cell lung cancer (NSCLC) accounts for more than 80 % of lung cancer (LC) cases, making it the primary cause of cancer-related mortality worldwide. T-box transcription factor 5 (TBX5) is an important regulator of embryonic and organ development and plays a key role in cancer development. Here, our objective was to investigate the involvement of TBX5 in ferroptosis within LC cells and the underlying mechanisms. METHODS: First, TBX5 expression was examined in human LC cells. Next, overexpression of TBX5 and Yes1-associated transcriptional regulator (YAP1) and knockdown of TEA domain 1 (TEAD1) were performed in A549 and NCI-H1703 cells. The proliferation ability of A549 and NCI-H1703 cells, GSH, MDA, ROS, and Fe2+ levels were measured. Co-immunoprecipitation (Co-IP) was performed to verify whether TBX5 protein could bind YAP1. Then TBX5, YAP1, TEAD1, GPX4, p53, FTH1, SLC7A11 and PTGS2 protein levels were assessed. Finally, we verified the effect of TBX5 on ferroptosis in LC cells in vivo. RESULTS: TBX5 expression was down-regulated in LC cells, especially in A549 and NCI-H1703 cells. Overexpression of TBX5 significantly decreased proliferation ability of A549 and NCI-H1703 cells, downregulated GPX4 and GSH levels, and upregulated MDA, ROS, and Fe2+ levels. Co-IP verified that TBX5 protein could bind YAP1. Moreover, oe-YAP1 promoted proliferation ability of A549 and NCI-H1703 cells transfected with Lv-TBX5, upregulated GPX4 and GSH levels and downregulated MDA, ROS, and Fe2+ levels. Additionally, oe-YAP1 promoted FTH1 and SLC7A11 levels and inhibited p53 and PTGS2 levels in A549 and NCI-H1703 cells transfected with Lv-TBX5. However, transfection with si-TEAD1 further reversed these effects. In vivo experiments further validated that TBX5 promoted ferroptosis in LC cells. CONCLUSIONS: TBX5 inhibited the activation of YAP1-TEAD1 pathway to promote ferroptosis in LC cells.


Asunto(s)
Ferroptosis , Neoplasias Pulmonares , Proteínas de Dominio T Box , Factores de Transcripción de Dominio TEA , Factores de Transcripción , Proteínas Señalizadoras YAP , Ferroptosis/genética , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/genética , Proteínas Señalizadoras YAP/metabolismo , Proteínas Señalizadoras YAP/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Factores de Transcripción de Dominio TEA/metabolismo , Proteínas de Dominio T Box/metabolismo , Proteínas de Dominio T Box/genética , Animales , Línea Celular Tumoral , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Pulmón de Células no Pequeñas/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Ratones Desnudos , Proliferación Celular , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/genética , Ratones , Regulación Neoplásica de la Expresión Génica , Células A549 , Transducción de Señal , Especies Reactivas de Oxígeno/metabolismo
6.
Genet Med ; : 101267, 2024 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-39268717

RESUMEN

PURPOSE: Predicting effects of genomic variants has become a real challenge in the diagnosis of rare human diseases. Holt-Oram syndrome (HOS) is an autosomal condition characterized by the association of radial and heart defects, due to variants in TBX5. Most variants are predicted to be truncating and result in haploinsufficiency. The pathogenicity of missense or splice variants is harder to demonstrate. METHODS: Fourteen TBX5 variants of uncertain significance (VUS) (5 missense, 9 splice) and 6 likely pathogenic missense variants were selected for functional testing, depending on the variant-type (immunolocalization, western blot, reporter assays, minigene splice assays and RT-PCR). Results were compared with in silico predictions. RESULTS: Functional tests allowed to reclassify 9/14 VUS in TBX5 as likely pathogenic, confirming their role in HOS. We demonstrated loss-of-function (n=8) or gain-of-function (n=1) for 9 of the 11 missense variants, whereas no functional impact was shown for the 2 variants: p.(Gly195Ala) and p.(Ser261Cys), as suggested by contradictory predictions of in silico approaches. Of 9 splice variants predicted to affect splicing by SpliceAI, we observed partial or complete exon skipping (n=6), intron retention (n=2) or exon shortening (n=1), inducing frame-shifting with premature stop codons. CONCLUSION: Bioinformatic and biological approaches are complementary, together with a good knowledge of clinical conditions, for accurate ACMG classification in human rare diseases.

7.
Acta Pharmacol Sin ; 45(8): 1644-1659, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38589686

RESUMEN

Cardiopulmonary progenitor cells (CPPs) constitute a minor subpopulation of cells that are commonly associated with heart and lung morphogenesis during embryonic development but completely subside after birth. This fact offers the possibility for the treatment of pulmonary heart disease (PHD), in which the lung and heart are both damaged. A reliable source of CPPs is urgently needed. In this study, we reprogrammed human cardiac fibroblasts (HCFs) into CPP-like cells (or induced CPPs, iCPPs) and evaluated the therapeutic potential of iCPP-derived exosomes for acute lung injury (ALI). iCPPs were created in passage 3 primary HCFs by overexpressing GLI1, WNT2, ISL1 and TBX5 (GWIT). Exosomes were isolated from the culture medium of passage 6-8 GWIT-iCPPs. A mouse ALI model was established by intratracheal instillation of LPS. Four hours after LPS instillation, ALI mice were treated with GWIT-iCPP-derived exosomes (5 × 109, 5 × 1010 particles/mL) via intratracheal instillation. We showed that GWIT-iCPPs could differentiate into cell lineages, such as cardiomyocyte-like cells, endothelial cells, smooth muscle cells and alveolar epithelial cells, in vitro. Transcription analysis revealed that GWIT-iCPPs have potential for heart and lung development. Intratracheal instillation of iCPP-derived exosomes dose-dependently alleviated LPS-induced ALI in mice by attenuating lung inflammation, promoting endothelial function and restoring capillary endothelial cells and the epithelial cells barrier. This study provides a potential new method for the prevention and treatment of cardiopulmonary injury, especially lung injury, and provides a new cell model for drug screening.


Asunto(s)
Lesión Pulmonar Aguda , Exosomas , Células Madre , Animales , Exosomas/metabolismo , Exosomas/trasplante , Lesión Pulmonar Aguda/terapia , Humanos , Ratones , Células Madre/citología , Células Madre/metabolismo , Fibroblastos/metabolismo , Masculino , Ratones Endogámicos C57BL , Diferenciación Celular , Células Cultivadas , Lipopolisacáridos/farmacología , Pulmón/metabolismo , Pulmón/patología , Modelos Animales de Enfermedad
8.
Adv Exp Med Biol ; 1441: 535-549, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38884730

RESUMEN

Ventricular septation is a complex process which involves the major genes of cardiac development, acting on myocardial cells from first and second heart fields, and on mesenchymal cells from endocardial cushions. These genes, coding for transcription factors, interact with each other, and their differential expression conditions the severity of the phenotype. In this chapter, we will describe the formation of the ventricular septum in the normal heart, as well as the molecular mechanisms leading to the four main anatomic types of ventricular septal defects: outlet, inlet, muscular, and central perimembranous, resulting from failure of development of the different parts of the ventricular septum. Experiments on animal models, particularly transgenic mouse lines, have helped us to decipher the molecular determinants of ventricular septation. However, a precise description of the anatomic phenotypes found in these models is mandatory to achieve a better comprehension of the complex mechanisms responsible for the various types of VSDs.


Asunto(s)
Modelos Animales de Enfermedad , Defectos del Tabique Interventricular , Animales , Humanos , Ratones , Regulación del Desarrollo de la Expresión Génica , Defectos del Tabique Interventricular/genética , Defectos del Tabique Interventricular/patología , Defectos del Tabique Interventricular/metabolismo , Ratones Transgénicos , Transducción de Señal/genética , Tabique Interventricular/patología , Tabique Interventricular/metabolismo , Tabique Interventricular/embriología
9.
Adv Exp Med Biol ; 1441: 937-945, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38884762

RESUMEN

Hypoplastic left heart syndrome (HLHS) is a severe congenital cardiovascular malformation characterized by hypoplasia of the left ventricle, aorta, and other structures on the left side of the heart. The pathologic definition includes atresia or stenosis of both the aortic and mitral valves. Despite considerable progress in clinical and surgical management of HLHS, mortality and morbidity remain concerns. One barrier to progress in HLHS management is poor understanding of its cause. Several lines of evidence point to genetic origins of HLHS. First, some HLHS cases have been associated with cytogenetic abnormalities (e.g., Turner syndrome). Second, studies of family clustering of HLHS and related cardiovascular malformations have determined HLHS is heritable. Third, genomic regions that encode genes influencing the inheritance of HLHS have been identified. Taken together, these diverse studies provide strong evidence for genetic origins of HLHS and related cardiac phenotypes. However, using simple Mendelian inheritance models, identification of single genetic variants that "cause" HLHS has remained elusive, and in most cases, the genetic cause remains unknown. These results suggest that HLHS inheritance is complex rather than simple. The implication of this conclusion is that researchers must move beyond the expectation that a single disease-causing variant can be found. Utilization of complex models to analyze high-throughput genetic data requires careful consideration of study design.


Asunto(s)
Síndrome del Corazón Izquierdo Hipoplásico , Humanos , Predisposición Genética a la Enfermedad/genética , Síndrome del Corazón Izquierdo Hipoplásico/genética , Fenotipo
10.
Adv Exp Med Biol ; 1441: 481-493, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38884727

RESUMEN

The relative simplicity of the clinical presentation and management of an atrial septal defect belies the complexity of the developmental pathogenesis. Here, we describe the anatomic development of the atrial septum and the venous return to the atrial chambers. Experimental models suggest how mutations and naturally occurring genetic variation could affect developmental steps to cause a defect within the oval fossa, the so-called secundum defect, or other interatrial communications, such as the sinus venosus defect or ostium primum defect.


Asunto(s)
Modelos Animales de Enfermedad , Defectos del Tabique Interatrial , Defectos del Tabique Interatrial/genética , Defectos del Tabique Interatrial/patología , Defectos del Tabique Interatrial/fisiopatología , Animales , Humanos , Mutación , Tabique Interatrial/patología , Transducción de Señal/genética
11.
Adv Exp Med Biol ; 1441: 915-928, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38884760

RESUMEN

Ebstein's anomaly is a congenital malformation of the tricuspid valve characterized by abnormal attachment of the valve leaflets, resulting in varying degrees of valve dysfunction. The anatomic hallmarks of this entity are the downward displacement of the attachment of the septal and posterior leaflets of the tricuspid valve. Additional intracardiac malformations are common. From an embryological point of view, the cavity of the future right atrium does not have a direct orifice connected to the developing right ventricle. This chapter provides an overview of current insight into how this connection is formed and how malformations of the tricuspid valve arise from dysregulation of molecular and morphological events involved in this process. Furthermore, mouse models that show features of Ebstein's anomaly and the naturally occurring model of canine tricuspid valve malformation are described and compared to the human model. Although Ebstein's anomaly remains one of the least understood cardiac malformations to date, the studies summarized here provide, in aggregate, evidence for monogenic and oligogenic factors driving pathogenesis.


Asunto(s)
Modelos Animales de Enfermedad , Anomalía de Ebstein , Válvula Tricúspide , Anomalía de Ebstein/genética , Anomalía de Ebstein/patología , Anomalía de Ebstein/fisiopatología , Animales , Humanos , Perros , Ratones , Válvula Tricúspide/anomalías , Válvula Tricúspide/patología
12.
Adv Exp Med Biol ; 1441: 885-900, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38884757

RESUMEN

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.


Asunto(s)
Modelos Animales de Enfermedad , Transducción de Señal , Atresia Tricúspide , Animales , Atresia Tricúspide/genética , Atresia Tricúspide/metabolismo , Atresia Tricúspide/patología , Humanos , Ratones , Corazón Univentricular/genética , Corazón Univentricular/metabolismo , Corazón Univentricular/fisiopatología , Corazón Univentricular/patología , Factores de Transcripción NFATC/metabolismo , Factores de Transcripción NFATC/genética , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta/genética , Receptores Notch/metabolismo , Receptores Notch/genética
13.
Adv Exp Med Biol ; 1441: 629-644, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38884738

RESUMEN

Tetralogy of Fallot (TOF) and double-outlet right ventricle (DORV) are conotruncal defects resulting from disturbances of the second heart field and the neural crest, which can occur as isolated malformations or as part of multiorgan syndromes. Their etiology is multifactorial and characterized by overlapping genetic causes. In this chapter, we present the different genetic alterations underlying the two diseases, which range from chromosomal abnormalities like aneuploidies and structural mutations to rare single nucleotide variations affecting distinct genes. For example, mutations in the cardiac transcription factors NKX2-5, GATA4, and HAND2 have been identified in isolated TOF cases, while mutations of TBX5 and 22q11 deletion, leading to haploinsufficiency of TBX1, cause Holt-Oram and DiGeorge syndrome, respectively. Moreover, genes involved in signaling pathways, laterality determination, and epigenetic mechanisms have also been found mutated in TOF and/or DORV patients. Finally, genome-wide association studies identified common single nucleotide polymorphisms associated with the risk for TOF.


Asunto(s)
Ventrículo Derecho con Doble Salida , Tetralogía de Fallot , Humanos , Tetralogía de Fallot/genética , Ventrículo Derecho con Doble Salida/genética , Mutación , Estudio de Asociación del Genoma Completo , Polimorfismo de Nucleótido Simple/genética , Predisposición Genética a la Enfermedad/genética , Factores de Transcripción/genética
14.
Adv Exp Med Biol ; 1441: 761-775, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38884747

RESUMEN

Lesions of the semilunar valve and the aortic arch can occur either in isolation or as part of well-described clinical syndromes. The polygenic cause of calcific aortic valve disease will be discussed including the key role of NOTCH1 mutations. In addition, the complex trait of bicuspid aortic valve disease will be outlined, both in sporadic/familial cases and in the context of associated syndromes, such as Alagille, Williams, and Kabuki syndromes. Aortic arch abnormalities particularly coarctation of the aorta and interrupted aortic arch, including their association with syndromes such as Turner and 22q11 deletion, respectively, are also discussed. Finally, the genetic basis of congenital pulmonary valve stenosis is summarized, with particular note to Ras-/mitogen-activated protein kinase (Ras/MAPK) pathway syndromes and other less common associations, such as Holt-Oram syndrome.


Asunto(s)
Aorta Torácica , Válvula Aórtica , Humanos , Aorta Torácica/anomalías , Aorta Torácica/patología , Válvula Aórtica/anomalías , Válvula Aórtica/patología , Anomalías Múltiples/genética , Anomalías Múltiples/patología , Cardiopatías Congénitas/genética , Cardiopatías Congénitas/patología , Enfermedad de la Válvula Aórtica Bicúspide/genética , Estenosis de la Válvula Pulmonar/genética , Mutación , Receptor Notch1/genética , Enfermedad de la Válvula Aórtica/genética , Enfermedades de las Válvulas Cardíacas/genética , Enfermedades de las Válvulas Cardíacas/patología , Calcinosis/genética , Calcinosis/patología , Enfermedades Hematológicas/genética , Enfermedades Hematológicas/patología , Enfermedades Vestibulares/genética , Enfermedades Vestibulares/patología
15.
Adv Exp Med Biol ; 1441: 467-480, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38884726

RESUMEN

Although atrial septal defects (ASD) can be subdivided based on their anatomical location, an essential aspect of human genetics and genetic counseling is distinguishing between isolated and familiar cases without extracardiac features and syndromic cases with the co-occurrence of extracardiac abnormalities, such as developmental delay. Isolated or familial cases tend to show genetic alterations in genes related to important cardiac transcription factors and genes encoding for sarcomeric proteins. By contrast, the spectrum of genes with genetic alterations observed in syndromic cases is diverse. Currently, it points to different pathways and gene networks relevant to the dysregulation of cardiomyogenesis and ASD pathogenesis. Therefore, this chapter reflects the current knowledge and highlights stable associations observed in human genetics studies. It gives an overview of the different types of genetic alterations in these subtypes, including common associations based on genome-wide association studies (GWAS), and it highlights the most frequently observed syndromes associated with ASD pathogenesis.


Asunto(s)
Estudio de Asociación del Genoma Completo , Defectos del Tabique Interatrial , Humanos , Defectos del Tabique Interatrial/genética , Predisposición Genética a la Enfermedad/genética , Mutación
16.
Adv Exp Med Biol ; 1441: 295-311, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38884718

RESUMEN

Cardiac development is a fine-tuned process governed by complex transcriptional networks, in which transcription factors (TFs) interact with other regulatory layers. In this chapter, we introduce the core cardiac TFs including Gata, Hand, Nkx2, Mef2, Srf, and Tbx. These factors regulate each other's expression and can also act in a combinatorial manner on their downstream targets. Their disruption leads to various cardiac phenotypes in mice, and mutations in humans have been associated with congenital heart defects. In the second part of the chapter, we discuss different levels of regulation including cis-regulatory elements, chromatin structure, and microRNAs, which can interact with transcription factors, modulate their function, or are downstream targets. Finally, examples of disturbances of the cardiac regulatory network leading to congenital heart diseases in human are provided.


Asunto(s)
Redes Reguladoras de Genes , Cardiopatías Congénitas , Factores de Transcripción , Animales , Humanos , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Cardiopatías Congénitas/genética , Cardiopatías Congénitas/metabolismo , Regulación del Desarrollo de la Expresión Génica , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Corazón/fisiología , Miocardio/metabolismo
17.
Adv Exp Med Biol ; 1441: 505-534, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38884729

RESUMEN

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.


Asunto(s)
Defectos del Tabique Interventricular , Humanos , Aberraciones Cromosómicas , Variaciones en el Número de Copia de ADN/genética , Predisposición Genética a la Enfermedad/genética , Defectos del Tabique Interventricular/genética , Mutación , Factores de Transcripción/genética
18.
Dev Biol ; 481: 201-214, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34756968

RESUMEN

The T-box containing family member, TBX5, has been shown to play important functional roles in the pectoral appendages of a variety of vertebrate species. While a single TBX5 gene exists in all tetrapods studied to date, the zebrafish genome retains two paralogues, designated as tbx5a and tbx5b, resulting from a whole genome duplication in the teleost lineage. Zebrafish deficient in tbx5a lack pectoral fin buds, whereas zebrafish deficient in tbx5b exhibit misshapen pectoral fins, showing that both paralogues function in fin development. The mesenchymal cells of the limb/fin bud are derived from the Lateral Plate Mesoderm (LPM). Previous fate mapping work in zebrafish has shown that wildtype (wt) fin field cells are initially located adjacent to somites (s)1-4. The wt fin field cells migrate in opposing diagonal directions placing the limb bud between s2-3 and lateral to the main body. To better characterize tbx5 paralogue functions in zebrafish, time-lapse analyses of the migrations of fin bud precursors under conditions of tbx5a knock-down, tbx5b knock-down and double-knock-down were performed. Our data suggest that zebrafish tbx5a and tbx5b have functionally separated migration direction vectors, that when combined recapitulate the migration of the wt fin field. We and others have shown that loss of Tbx5a function abolishes an fgf24 signaling cue resulting in fin field cells failing to converge in an Antero-Posterior (AP) direction and migrating only in a mediolateral (ML) direction. We show here that loss of Tbx5b function affects initial ML directed movements so that fin field cells fail to migrate laterally but continue to converge along the AP axis. Furthermore, fin field cells in the double Tbx5a/Tbx5b knock-down zebrafish do not engage in directed migrations along either the ML or AP axis. Therefore, these two paralogues may be acting to instruct separate vectors of fin field migration in order to direct proper fin bud formation.


Asunto(s)
Aletas de Animales/embriología , Movimiento Celular , Factores de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/embriología , Animales , Factores de Crecimiento de Fibroblastos/genética , Técnicas de Silenciamiento del Gen , Factores de Transcripción/genética , Pez Cebra/genética , Proteínas de Pez Cebra/genética
19.
Biochem Biophys Res Commun ; 638: 184-191, 2023 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-36462492

RESUMEN

Ubiquitin specific proteinase 28 (USP28) is a member of the deubiquitylating enzymes, which are mainly involved in cell cycle, apoptosis and DNA damage repair. Although USP28 has been found to be upregulated in some tumors, its role in ovarian cancer (OV) remains unclear. Here we show that USP28 was highly expressed in OV samples compared with normal ovarian tissue, and OV patients with higher USP28 levels had a worse prognosis. We found that the abnormal expression of USP28 mRNA in OV was related to the activation of ß-catenin signaling pathway, and USP28 was a transcriptional target gene of the ß-catenin/YAP1/TBX5 complex. In addition, genetic ablation or pharmacological inhibition of USP28 impaired the proliferation ability of OV cells in vitro and in vivo. In conclusion, our findings show that ß-catenin/YAP1/TBX5-mediated aberrant expression of USP28 promotes the malignant phenotype of OV, suggesting that USP28 may be a therapeutic target for OV.


Asunto(s)
Neoplasias Ováricas , beta Catenina , Humanos , Femenino , beta Catenina/genética , Ubiquitina , Péptido Hidrolasas , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Enzimas Desubicuitinizantes , Línea Celular Tumoral , Ubiquitina Tiolesterasa/genética , Ubiquitina Tiolesterasa/metabolismo , Proliferación Celular/genética
20.
Dev Biol ; 472: 52-66, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33482174

RESUMEN

In this study, we elucidate a single cell resolution fate map in the zebrafish in a sub-section of the anterior Lateral Plate Mesoderm (aLPM) at 18 hpf. Our results show that this tissue is not organized into segregated regions but gives rise to intermingled pericardial sac, peritoneum, pharyngeal arch and cardiac precursors. We further report upon asymmetrical contributions of lateral aLPM-derived heart precursors-specifically that twice as many heart precursors arise from the right side versus the left side of the embryo. Cell tracking analyses and large-scale cell labeling of the lateral aLPM corroborate these differences and show that the observed asymmetries are dependent upon Tbx5a expression. Previously, it was shown that cardiac looping was affected in Tbx5a knock-down and knock-out zebrafish (Garrity et al., 2002; Parrie et al., 2013); our present data also implicate tbx5a function in cell specification, establishment and maintenance of cardiac left-right asymmetry.


Asunto(s)
Tipificación del Cuerpo/genética , Diferenciación Celular/genética , Movimiento Celular/genética , Mesodermo/metabolismo , Miocitos Cardíacos/metabolismo , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Proteínas de Pez Cebra/deficiencia , Proteínas de Pez Cebra/genética , Pez Cebra/genética , Pez Cebra/metabolismo , Animales , Animales Modificados Genéticamente , Región Branquial/metabolismo , Embrión no Mamífero/metabolismo , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Organogénesis/genética , Transducción de Señal/genética , Pez Cebra/embriología
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda