Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Dev Dyn ; 242(5): 485-502, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23441045

RESUMEN

BACKGROUND: T-box genes constitute a large family of transcriptional regulators involved in developmental patterning. Homozygous mutation of tbx5 leads to embryonic lethal cardiac phenotypes and forelimb malformations in vertebrate models. Haploinsufficiency of tbx5 results in Holt-Oram syndrome, a human congenital disease characterized by cardiac and forelimb defects. Homozygous mutation of zebrafish tbx5a leads to lethal defects in cardiac looping morphogenesis, blocks pectoral fin initiation, and impairs outgrowth. Recently, a second zebrafish tbx5 gene was described, termed tbx5b. RESULTS: Our phylogenetic analyses confirm tbx5b as a paralog that likely arose in the teleost-specific whole genome duplication ∼270 MYA. Using morpholino depletion studies, we find that tbx5b is required in the heart for embryonic survival, and influences the timing and morphogenesis of pectoral fin development. Because tbx5a hypomorphic mutations are embryonic lethal, tbx5a and tbx5b functions in the heart must not be completely redundant. Consistent with this hypothesis, simultaneous depletion of both tbx5 paralogs did not lead to more severe phenotypes, and injection of wild-type mRNA from one tbx5 paralog was not sufficient to cross-rescue phenotypes of the paralogous gene. CONCLUSIONS: Collectively, these data indicate that, despite similar spatio-temporal expression patterns, tbx5a and tbx5b have independent functions in heart and fin development.


Asunto(s)
Aletas de Animales/embriología , Corazón/embriología , Proteínas de Dominio T Box/fisiología , Pez Cebra , Secuencia de Aminoácidos , Aletas de Animales/metabolismo , Animales , Tipificación del Cuerpo/genética , Tipificación del Cuerpo/fisiología , Embrión no Mamífero , Duplicación de Gen , Regulación del Desarrollo de la Expresión Génica , Especiación Genética , Genoma/genética , Datos de Secuencia Molecular , Morfogénesis/genética , Filogenia , Isoformas de Proteínas/genética , Homología de Secuencia , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo , Pez Cebra/embriología , Pez Cebra/genética , Pez Cebra/metabolismo
2.
J Biomech Eng ; 135(4): 041006, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24231901

RESUMEN

Congenital heart defects arise during the early stages of development, and studies have linked abnormal blood flow and irregular cardiac function to improper cardiac morphogenesis. The embryonic zebrafish offers superb optical access for live imaging of heart development. Here, we build upon previously used techniques to develop a methodology for quantifying cardiac function in the embryonic zebrafish model. Imaging was performed using bright field microscopy at 1500 frames/s at 0.76 µm/pixel. Heart function was manipulated in a wild-type zebrafish at ∼55 h post fertilization (hpf). Blood velocity and luminal diameter were measured at the atrial inlet and atrioventricular junction (AVJ) by analyzing spatiotemporal plots. Control volume analysis was used to estimate the flow rate waveform, retrograde fractions, stroke volume, and cardiac output. The diameter and flow waveforms at the inlet and AVJ are highly repeatable between heart beats. We have developed a methodology for quantifying overall heart function, which can be applied to early stages of zebrafish development.


Asunto(s)
Corazón/embriología , Corazón/fisiología , Animales , Circulación Coronaria , Frecuencia Cardíaca , Hemodinámica , Procesamiento de Imagen Asistido por Computador , Pez Cebra/embriología
3.
Dev Dyn ; 241(4): 648-62, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22274990

RESUMEN

BACKGROUND: L-type calcium channels (LTCC) regulate calcium entry into cardiomyocytes. CACNB2 (ß2) LTCC auxiliary subunits traffic the pore-forming CACNA subunit to the membrane and modulate channel kinetics. ß2 is a membrane associated guanylate kinase (MAGUK) protein. A major role of MAGUK proteins is to scaffold cellular junctions and multiprotein complexes. RESULTS: To investigate developmental functions for ß2.1, we depleted it in zebrafish using morpholinos. ß2.1-depleted embryos developed compromised cardiac function by 48 hr postfertilization, which was ultimately lethal. ß2.1 contractility defects were mimicked by pharmacological depression of LTCC, and rescued by LTCC stimulation, suggesting ß2.1 phenotypes are at least in part LTCC-dependent. Morphological studies indicated that ß2.1 contributes to heart size by regulating the rate of ventricle cell proliferation, and by modulating the transition of outer curvature cells to an elongated cell shape during chamber ballooning. In addition, ß2.1-depleted cardiomyocytes failed to accumulate N-cadherin at the membrane, and dissociated easily from neighboring myocytes under stress. CONCLUSIONS: Hence, we propose that ß2.1 may also function in the heart as a MAGUK scaffolding unit to maintain N-cadherin-based adherens junctions and heart tube integrity.


Asunto(s)
Canales de Calcio Tipo L/fisiología , Corazón/embriología , Proteínas de Pez Cebra/fisiología , Pez Cebra/embriología , Animales , Calcio/metabolismo , Canales de Calcio Tipo L/genética , Proliferación Celular , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , Corazón/fisiología , Organogénesis/fisiología , Pez Cebra/fisiología , Proteínas de Pez Cebra/genética
4.
Front Physiol ; 14: 1086050, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37007999

RESUMEN

In the adult heart, acute adaptation of electrical and mechanical activity to changes in mechanical load occurs via feedback processes known as "mechano-electric coupling" and "mechano-mechanical coupling." Whether this occurs during cardiac development is ill-defined, as acutely altering the heart's mechanical load while measuring functional responses in traditional experimental models is difficult, as embryogenesis occurs in utero, making the heart inaccessible. These limitations can be overcome with zebrafish, as larvae develop in a dish and are nearly transparent, allowing for in vivo manipulation and measurement of cardiac structure and function. Here we present a novel approach for the in vivo study of mechano-electric and mechano-mechanical coupling in the developing zebrafish heart. This innovative methodology involves acute in vivo atrial dilation (i.e., increased atrial preload) in larval zebrafish by injection of a controlled volume into the venous circulation immediately upstream of the heart, combined with optical measurement of the acute electrical (change in heart rate) and mechanical (change in stroke area) response. In proof-of-concept experiments, we applied our new method to 48 h post-fertilisation zebrafish, which revealed differences between the electrical and mechanical response to atrial dilation. In response to an acute increase in atrial preload there is a large increase in atrial stroke area but no change in heart rate, demonstrating that in contrast to the fully developed heart, during early cardiac development mechano-mechanical coupling alone drives the adaptive increase in atrial output. Overall, in this methodological paper we present our new experimental approach for the study of mechano-electric and mechano-mechanical coupling during cardiac development and demonstrate its potential for understanding the essential adaptation of heart function to acute changes in mechanical load.

5.
J Cardiovasc Dev Dis ; 9(1)2022 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-35050232

RESUMEN

Heart valve development is governed by both genetic and biomechanical inputs. Prior work has demonstrated that oscillating shear stress associated with blood flow is required for normal atrioventricular (AV) valve development. Cardiac afterload is defined as the pressure the ventricle must overcome in order to pump blood throughout the circulatory system. In human patients, conditions of high afterload can cause valve pathology. Whether high afterload adversely affects embryonic valve development remains poorly understood. Here we describe a zebrafish model exhibiting increased myocardial afterload, caused by vasopressin, a vasoconstrictive drug. We show that the application of vasopressin reliably produces an increase in afterload without directly acting on cardiac tissue in zebrafish embryos. We have found that increased afterload alters the rate of growth of the cardiac chambers and causes remodeling of cardiomyocytes. Consistent with pathology seen in patients with clinically high afterload, we see defects in both the form and the function of the valve leaflets. Our results suggest that valve defects are due to changes in atrioventricular myocyte signaling, rather than pressure directly acting on the endothelial valve leaflet cells. Cardiac afterload should therefore be considered a biomechanical factor that particularly impacts embryonic valve development.

6.
Dev Biol ; 330(1): 175-84, 2009 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-19345202

RESUMEN

Mutations in the T-box transcription factor, TBX5, result in Holt-Oram syndrome (HOS), a human condition in which cardiac development is defective and forelimbs are stunted. Similarly, zebrafish tbx5 morphants and mutants (heartstrings; hst) lack pectoral fins and exhibit a persistently elongated heart that does not undergo chamber looping. Tbx5 is expressed in the developing atrium, ventricle and in pectoral fin fields, but its genetic targets are still being uncovered. In this study, evidence is provided that Tbx5 induces the expression of a specific member of the CaMK-II (the type II multifunctional Ca(2+)/calmodulin-dependent protein kinase) family; this CaMK-II is necessary for proper heart and fin development. Morphants of beta2 CaMK-II (camk2b2), but not the beta1 CaMK-II (camk2b1) paralog, exhibit bradycardia, elongated hearts and diminished pectoral fin development. Normal cardiac phenotypes can be restored by ectopic cytosolic CaMK-II expression in tbx5 morphants. Like tbx5, camk2b2 is expressed in the pectoral fin and looping heart, but this expression is diminished in both tbx5 morphant and hst embryos. Conversely, the introduction of excess Tbx5 into zebrafish embryos and mouse fibroblasts doubles CaMK-II expression. We conclude that beta CaMK-II expression and activity are necessary for proper cardiac and limb morphogenesis. These findings not only identify a morphogenic target for Ca(2+) during heart development, but support implied roles for CaMK-II in adult heart remodeling.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Corazón/embriología , Morfogénesis , Proteínas de Dominio T Box/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/embriología , Secuencia de Aminoácidos , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Embrión no Mamífero/metabolismo , Regulación del Desarrollo de la Expresión Génica , Corazón/crecimiento & desarrollo , Ratones , Datos de Secuencia Molecular , Morfogénesis/genética , Células 3T3 NIH , Transfección , Proteínas de Pez Cebra/genética
7.
Dev Biol ; 317(2): 486-96, 2008 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-18407257

RESUMEN

NDRG4 is a novel member of the NDRG family (N-myc downstream-regulated gene). The roles of NDRG4 in development have not previously been evaluated. We show that, during zebrafish embryonic development, ndrg4 is expressed exclusively in the embryonic heart, the central nervous system (CNS) and the sensory system. Ndrg4 knockdown in zebrafish embryos causes a marked reduction in proliferative myocytes and results in hypoplastic hearts. This growth defect is associated with cardiac phenotypes in morphogenesis and function, including abnormal heart looping, inefficient circulation and weak contractility. We reveal that ndrg4 is required for restricting the expression of versican and bmp4 to the developing atrioventricular canal. This constellation of ndrg4 cardiac defects phenocopies those seen in mutant hearts of heartstrings (hst), the tbx5 loss-of-function mutants in zebrafish. We further show that ndrg4 expression is significantly decreased in hearts with reduced tbx5 activities. Conversely, increased expression of tbx5 that is due to tbx20 knockdown leads to an increase in ndrg4 expression. Together, our studies reveal an essential role of ndrg4 in regulating proliferation and growth of cardiomyocytes, suggesting that ndrg4 may function downstream of tbx5 during heart development and growth.


Asunto(s)
Proteínas Musculares/metabolismo , Miocitos Cardíacos/fisiología , Proteínas del Tejido Nervioso/metabolismo , Fenotipo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/embriología , Animales , Proteína Morfogenética Ósea 4 , Proteínas Morfogenéticas Óseas/metabolismo , Proliferación Celular , Clonación Molecular , Corazón/embriología , Hibridación in Situ , Proteínas Musculares/genética , Mutación/genética , Miocitos Cardíacos/metabolismo , Proteínas del Tejido Nervioso/genética , Oligonucleótidos/genética , Proteínas de Dominio T Box/metabolismo , Versicanos/metabolismo , Proteínas de Pez Cebra/genética
8.
Physiol Genomics ; 35(2): 133-44, 2008 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-18682574

RESUMEN

The Ca(2+) channel beta-subunits, encoded by CACNB genes 1-4, are membrane-associated guanylate kinase (MAGUK) proteins. As auxiliary subunits of voltage-gated Ca(2+) channels, the beta-subunits facilitate membrane trafficking of the pore-forming alpha1 subunits and regulate voltage-dependent channel gating. In this report, we investigate whether two zebrafish beta4 genes, beta4.1 and beta4.2, have diverged in structure and function over time. Comparative expression analyses indicated that beta4.1 and beta4.2 were expressed in separable domains within the developing brain and other tissues. Alternative splicing in both genes was subject to differential temporal and spatial regulation, with some organs expressing different subsets of beta4.1 and beta4.2 transcript variants. We used several genomic tools to identify and compare predicted cDNAs for eight teleost and five tetrapod beta4 genes. Teleost species had either one or two beta4 paralogs, whereas each tetrapod species contained only one. Teleost beta4.1 and beta4.2 genes had regions of sequence divergence, but compared with tetrapod beta4s, they exhibited similar exon/intron structure, strong conservation of residues involved in alpha1 subunit binding, and similar 5' alternative splicing. Phylogenetic results are consistent with the duplicate teleost beta4 genes resulting from the teleost whole genome duplication. Following duplication, the beta4.1 genes have evolved faster than beta4.2 genes. We identified disproportionately large second and third introns in several beta4 genes, which we propose may provide regulatory elements contributing to their differential tissue expression. In sum, both mRNA expression data and phylogenetic analysis support the evolutionary divergence of beta4.1 and beta4.2 subunit function.


Asunto(s)
Canales de Calcio/clasificación , Canales de Calcio/genética , Genoma , Empalme Alternativo , Secuencia de Aminoácidos , Animales , Canales de Calcio/metabolismo , Evolución Molecular , Expresión Génica , Genómica , Humanos , Hibridación in Situ , Intrones , Datos de Secuencia Molecular , Filogenia , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , ARN Mensajero/metabolismo , Alineación de Secuencia , Vertebrados , Pez Cebra/genética , Pez Cebra/metabolismo
9.
BMC Mol Biol ; 9: 38, 2008 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-18419826

RESUMEN

BACKGROUND: Cardiomyocyte contraction is initiated by influx of extracellular calcium through voltage-gated calcium channels. These oligomeric channels utilize auxiliary beta subunits to chaperone the pore-forming alpha subunit to the plasma membrane, and to modulate channel electrophysiology 1. Several beta subunit family members are detected by RT-PCR in the embryonic heart. Null mutations in mouse beta2, but not in the other three beta family members, are embryonic lethal at E10.5 due to defects in cardiac contractility 2. However, a drawback of the mouse model is that embryonic heart rhythm is difficult to study in live embryos due to their intra-uterine development. Moreover, phenotypes may be obscured by secondary effects of hypoxia. As a first step towards developing a model for contributions of beta subunits to the onset of embryonic heart rhythm, we characterized the structure and expression of beta2 subunits in zebrafish and other teleosts. RESULTS: Cloning of two zebrafish beta2 subunit genes (beta2.1 and beta2.2) indicated they are membrane-associated guanylate kinase (MAGUK)-family genes. Zebrafish beta2 genes show high conservation with mammals within the SH3 and guanylate kinase domains that comprise the "core" of MAGUK proteins, but beta2.2 is much more divergent in sequence than beta2.1. Alternative splicing occurs at the N-terminus and within the internal HOOK domain. In both beta2 genes, alternative short ATG-containing first exons are separated by some of the largest introns in the genome, suggesting that individual transcript variants could be subject to independent cis-regulatory control. In the Tetraodon nigrovidis and Fugu rubripes genomes, we identified single beta2 subunit gene loci. Comparative analysis of the teleost and human beta2 loci indicates that the short 5' exon sequences are highly conserved. A subset of 5' exons appear to be unique to teleost genomes, while others are shared with mammals. Alternative splicing is temporally and spatially regulated in embryo and adult. Moreover, a different subset of spliced beta2 transcript variants is detected in the embryonic heart compared to the adult. CONCLUSION: These studies refine our understanding of beta2 subunit diversity arising from alternative splicing, and provide the groundwork for functional analysis of beta2 subunit diversity in the embryonic heart.


Asunto(s)
Canales de Calcio Tipo L/química , Canales de Calcio Tipo L/genética , Subunidades de Proteína/genética , Tetraodontiformes/genética , Proteínas de Pez Cebra/genética , Pez Cebra/genética , Empalme Alternativo/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Embrión no Mamífero/metabolismo , Regulación del Desarrollo de la Expresión Génica , Variación Genética , Genoma , Intrones/genética , Datos de Secuencia Molecular , Filogenia , Estructura Terciaria de Proteína , Subunidades de Proteína/química , ARN Mensajero/genética , ARN Mensajero/metabolismo , Alineación de Secuencia , Tetraodontiformes/embriología , Pez Cebra/embriología , Proteínas de Pez Cebra/química
10.
JACC Clin Electrophysiol ; 4(4): 504-514, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-30067491

RESUMEN

OBJECTIVES: The purpose of this study was to assess the phenotype of Filamin C (FLNC) truncating variants in dilated cardiomyopathy (DCM) and understand the mechanism leading to an arrhythmogenic phenotype. BACKGROUND: Mutations in FLNC are known to lead to skeletal myopathies, which may have an associated cardiac component. Recently, the clinical spectrum of FLNC mutations has been recognized to include a cardiac-restricted presentation in the absence of skeletal muscle involvement. METHODS: A population of 319 U.S. and European DCM cardiomyopathy families was evaluated using whole-exome and targeted next-generation sequencing. FLNC truncation probands were identified and evaluated by clinical examination, histology, transmission electron microscopy, and immunohistochemistry. RESULTS: A total of 13 individuals in 7 families (2.2%) were found to harbor 6 different FLNC truncation variants (2 stopgain, 1 frameshift, and 3 splicing). Of the 13 FLNC truncation carriers, 11 (85%) had either ventricular arrhythmias or sudden cardiac death, and 5 (38%) presented with evidence of right ventricular dilation. Pathology analysis of 2 explanted hearts from affected FLNC truncation carriers showed interstitial fibrosis in the right ventricle and epicardial fibrofatty infiltration in the left ventricle. Ultrastructural findings included occasional disarray of Z-discs within the sarcomere. Immunohistochemistry showed normal plakoglobin signal at cell-cell junctions, but decreased signals for desmoplakin and synapse-associated protein 97 in the myocardium and buccal mucosa. CONCLUSIONS: We found FLNC truncating variants, present in 2.2% of DCM families, to be associated with a cardiac-restricted arrhythmogenic DCM phenotype characterized by a high risk of life-threatening ventricular arrhythmias and a pathological cellular phenotype partially overlapping with arrhythmogenic right ventricular cardiomyopathy.


Asunto(s)
Cardiomiopatía Dilatada , Filaminas/genética , Mutación/genética , Miocardio , Arritmias Cardíacas , Cardiomiopatía Dilatada/epidemiología , Cardiomiopatía Dilatada/genética , Cardiomiopatía Dilatada/patología , Adhesión Celular/genética , Análisis Mutacional de ADN , Europa (Continente) , Humanos , Inmunohistoquímica , Miocardio/citología , Miocardio/patología , Polimorfismo de Nucleótido Simple/genética , Estudios Prospectivos , Estados Unidos
11.
Plant Methods ; 13: 113, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29270209

RESUMEN

BACKGROUND: Switchgrass (Panicum virgatum), a robust perennial C4-type grass, has been evaluated and designated as a model bioenergy crop by the U.S. DOE and USDA. Conventional breeding of switchgrass biomass is difficult because it displays self-incompatible hindrance. Therefore, direct genetic modifications of switchgrass have been considered the more effective approach to tailor switchgrass with traits of interest. Successful transformations have demonstrated increased biomass yields, reduction in the recalcitrance of cell walls and enhanced saccharification efficiency. Several tissue culture protocols have been previously described to produce transgenic switchgrass lines using different nutrient-based media, co-cultivation approaches, and antibiotic strengths for selection. RESULTS: After evaluating the published protocols, we consolidated these approaches and optimized the process to develop a more efficient protocol for producing transgenic switchgrass. First, seed sterilization was optimized, which led to a 20% increase in yield of induced calluses. Second, we have selected a N6 macronutrient/B5 micronutrient (NB)-based medium for callus induction from mature seeds of the Alamo cultivar, and chose a Murashige and Skoog-based medium to regenerate both Type I and Type II calluses. Third, Agrobacterium-mediated transformation was adopted that resulted in 50-100% positive regenerated transformants after three rounds (2 weeks/round) of selection with antibiotic. Genomic DNA PCR, RT-PCR, Southern blot, visualization of the red fluorescent protein and histochemical ß-glucuronidase (GUS) staining were conducted to confirm the positive switchgrass transformants. The optimized methods developed here provide an improved strategy to promote the production and selection of callus and generation of transgenic switchgrass lines. CONCLUSION: The process for switchgrass transformation has been evaluated and consolidated to devise an improved approach for transgenic switchgrass production. With the optimization of seed sterilization, callus induction, and regeneration steps, a reliable and effective protocol is established to facilitate switchgrass engineering.

12.
JACC Basic Transl Sci ; 1(5): 344-359, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28008423

RESUMEN

OBJECTIVE: To identify novel dilated cardiomyopathy (DCM) causing genes, and to elucidate the pathological mechanism leading to DCM by utilizing zebrafish as a model organism. BACKGROUND: DCM, a major cause of heart failure, is frequently familial and caused by a genetic defect. However, only 50% of DCM cases can be attributed to a known DCM gene variant, motivating the ongoing search for novel disease genes. METHODS: We performed whole exome sequencing (WES) in two multigenerational Italian families and one US family with arrhythmogenic DCM without skeletal muscle defects, in whom prior genetic testing had been unrevealing. Pathogenic variants were sought by a combination of bioinformatic filtering and cosegregation testing among affected individuals within the families. We performed function assays and generated a zebrafish morpholino knockdown model. RESULTS: A novel filamin C gene splicing variant (FLNC c.7251+1 G>A) was identified by WES in all affected family members in the two Italian families. A separate novel splicing mutation (FLNC c.5669-1delG) was identified in the US family. Western blot analysis of cardiac heart tissue from an affected individual showed decreased FLNC protein, supporting a haploinsufficiency model of pathogenesis. To further analyze this model, a morpholino knockdown of the ortholog filamin Cb in zebrafish was created which resulted in abnormal cardiac function and ultrastructure. CONCLUSIONS: Using WES, we identified two novel FLNC splicing variants as the likely cause of DCM in three families. We provided protein expression and in vivo zebrafish data supporting haploinsufficiency as the pathogenic mechanism leading to DCM.

13.
Cardiovasc Eng Technol ; 4(3): 246-255, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29637499

RESUMEN

Several studies have linked abnormal blood flow dynamics to the formation of congenital heart defects during the early stages of development. The objective of this study is to document the transition of pumping mechanics from the early tube stage to the late looping stage of the embryonic heart. The optically transparent zebrafish embryonic heart was utilized as the in vivo model and was studied using standard bright field microscopy at three relevant stages within the transitional period: (1) tube stage at 30 hours post-fertilization (hpf); (2) early cardiac looping stage at 36 hpf; and (3) late cardiac looping stage at 48 hpf. High-speed videos were collected at 1000 fps at a spatial resolution of 1.1 µm/pixel at each of these stages and were post-processed to yield blood velocity patterns as well as wall kinematics. Results show that several relevant trends exist. Morphological trends from tube through late looping include: (a) ballooning of the chambers, (b) increasing constriction at the atrioventricular junction (AVJ), and (c) repositioning of the ventricle toward the side of the atrium. Blood flow trends include: (a) higher blood velocities, (b) increased AVJ regurgitation, and (c) larger percentages of blood from the upper atrium expelled backward toward the atrial inlet. Pumping mechanics trends include: (a) increasing contraction wave delay at the AVJ, (b) the AVJ begins acting as a rudimentary valve, (c) decreasing chamber constriction during maximum contraction, and (d) a transition in ventricular kinematics from a pronounced propagating wave to an independent, full-chamber contraction. The above results provide new insight into the transitional pumping mechanics from peristalsis-like pumping to a displacement pumping mechanism.

15.
Development ; 129(19): 4635-45, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12223419

RESUMEN

Holt-Oram syndrome is one of the autosomal dominant human "heart-hand" disorders, with a combination of upper limb malformations and cardiac defects. Holt-Oram syndrome is caused by mutations in the TBX5 gene, a member of a large family of T-box transcription factors that play important roles in cell-type specification and morphogenesis. In a screen for mutations affecting zebrafish cardiac function, we isolated the recessive lethal mutant heartstrings, which lacks pectoral fins and exhibits severe cardiac dysfunction, beginning with a slow heart rate and progressing to a stretched, non-functional heart. We mapped and cloned the heartstrings mutation and find it to encode the zebrafish ortholog of the TBX5 gene. The heartstrings mutation causes premature termination at amino acid 316. Homozygous mutant embryos never develop pectoral fin buds and do not express several markers of early fin differentiation. The total absence of any fin bud differentiation distinguishes heartstrings from most other mutations that affect zebrafish fin development, suggesting that Tbx5 functions very early in the pectoral fin induction pathway. Moderate reduction of Tbx5 by morpholino causes fin malformations, revealing an additional early requirement for Tbx5 in coordinating the axes of fin outgrowth. The heart of heartstrings mutant embryos appears to form and function normally through the early heart tube stage, manifesting only a slight bradycardia compared with wild-type siblings. However, the heart fails to loop and then progressively deteriorates, a process affecting the ventricle as well as the atrium. Relative to mammals, fish require lower levels of Tbx5 to produce malformed appendages and display whole-heart rather than atrial-predominant cardiac defects. However, the syndromic deficiencies of tbx5 mutation are remarkably well retained between fish and mammals.


Asunto(s)
Cardiopatías Congénitas/metabolismo , Corazón/embriología , Factores de Transcripción/fisiología , Proteínas de Pez Cebra/fisiología , Pez Cebra/embriología , Animales , Mapeo Cromosómico , Perfilación de la Expresión Génica , Cardiopatías Congénitas/genética , Biosíntesis de Proteínas , Síndrome , Factores de Transcripción/genética , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética
16.
Development ; 129(4): 973-82, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11861480

RESUMEN

Little is known about how vascular patterns are generated in the embryo. The vasculature of the zebrafish trunk has an extremely regular pattern. One intersegmental vessel (ISV) sprouts from the aorta, runs between each pair of somites, and connects to the dorsal longitudinal anastomotic vessel (DLAV). We now define the cellular origins, migratory paths and cell fates that generate these metameric vessels of the trunk. Additionally, by a genetic screen we define one gene, out of bounds (obd), that constrains this angiogenic growth to a specific path. We have performed lineage analysis, using laser activation of a caged dye and mosaic construction to determine the origin of cells that constitute the ISV. Individual angioblasts destined for the ISVs arise from the lateral posterior mesoderm (LPM), and migrate to the dorsal aorta, from where they migrate between somites to their final position in the ISVs and dorsal longitudinal anastomotic vessel (DLAV). Cells of each ISV leave the aorta only between the ventral regions of two adjacent somites, and migrate dorsally to assume one of three ISV cell fates. Most dorsal is a T-shaped cell, based in the DLAV and branching ventrally; the second constitutes a connecting cell; and the third an inverted T-shaped cell, based in the aorta and branching dorsally. The ISV remains between somites during its ventral course, but changes to run mid-somite dorsally. This suggests that the pattern of ISV growth ventrally and dorsally is guided by different cues. We have also performed an ENU mutagenesis screen of 750 mutagenized genomes and identified one mutation, obd that disrupts this pattern. In obd mutant embryos, ISVs sprout precociously at abnormal sites and migrate anomalously in the vicinity of ventral somite. The dorsal extent of the ISV is less perturbed. Precocious sprouting can be inhibited in a VEGF morphant, but the anomalous site of origin of obd ISVs remains. In mosaic embryos, obd somite causes adjacent wild-type endothelial cells to assume the anomalous ISV pattern of obd embryos. Thus, the launching position of the new sprout and its initial trajectory are directed by inhibitory signals from ventral somites. Zebrafish ISVs are a tractable system for defining the origins and fates of vessels, and for dissecting elements that govern patterns of vessel growth.


Asunto(s)
Tipificación del Cuerpo/fisiología , Neovascularización Fisiológica/fisiología , Células Madre/fisiología , Pez Cebra/embriología , Animales , Diferenciación Celular , Movimiento Celular/fisiología , Mesodermo , Fenotipo
17.
Development ; 129(12): 3009-19, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12050147

RESUMEN

The zebrafish mutant violet beauregarde (vbg) can be identified at two days post-fertilization by an abnormal circulation pattern in which most blood cells flow through a limited number of dilated cranial vessels and fail to perfuse the trunk and tail. This phenotype cannot be explained by caudal vessel abnormalities or by a defect in cranial vessel patterning, but instead stems from an increase in endothelial cell number in specific cranial vessels. We show that vbg encodes activin receptor-like kinase 1 (Acvrl1; also known as Alk1), a TGFbeta type I receptor that is expressed predominantly in the endothelium of the vessels that become dilated in vbg mutants. Thus, vbg provides a model for the human autosomal dominant disorder, hereditary hemorrhagic telangiectasia type 2, in which disruption of ACVRL1 causes vessel malformations that may result in hemorrhage or stroke. Movies available on-line


Asunto(s)
Receptores de Activinas/genética , Receptores de Activinas/metabolismo , Vasos Sanguíneos/embriología , Cabeza/irrigación sanguínea , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , Pez Cebra/genética , Secuencia de Aminoácidos , Animales , Animales Modificados Genéticamente , Vasos Sanguíneos/anomalías , Circulación Cerebrovascular , Mapeo Cromosómico , Proteínas de Unión al ADN/metabolismo , Embrión no Mamífero , Regulación del Desarrollo de la Expresión Génica , Cabeza/embriología , Humanos , Datos de Secuencia Molecular , Mutación , Fosfoproteínas/metabolismo , Homología de Secuencia de Aminoácido , Transducción de Señal , Proteínas Smad , Proteína Smad5 , Proteína Smad8 , Telangiectasia Hemorrágica Hereditaria/genética , Telangiectasia Hemorrágica Hereditaria/fisiopatología , Transactivadores/metabolismo , Pez Cebra/embriología
18.
Development ; 129(22): 5161-70, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12399308

RESUMEN

A major gap in our knowledge of development is how the growth and identity of tissues and organs are linked during embryogenesis. The vertebrate limb is one of the best models to study these processes. Combining mutant analyses with gain- and loss-of-function approaches in zebrafish and chick embryos, we show that Tbx5, in addition to its role governing forelimb identity, is both necessary and sufficient for limb outgrowth. We find that Tbx5 functions downstream of WNT signaling to regulate Fgf10, which, in turn, maintains Tbx5 expression during limb outgrowth. Furthermore, our results indicate that Tbx5 and Wnt2b function together to initiate and specify forelimb outgrowth and identity. The molecular interactions governed by members of the T-box, Wnt and Fgf gene families uncovered in this study provide a framework for understanding not only limb development, but how outgrowth and identity of other tissues and organs of the embryo may be regulated.


Asunto(s)
Extremidades/embriología , Factores de Crecimiento de Fibroblastos/genética , Glicoproteínas/genética , Péptidos y Proteínas de Señalización Intercelular , Proteínas de Dominio T Box/genética , Proteínas de Pez Cebra/genética , Pez Cebra/embriología , Secuencia de Aminoácidos , Animales , Embrión de Pollo , Embrión no Mamífero , Factor 10 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Glicoproteínas/metabolismo , Esbozos de los Miembros/fisiología , Datos de Secuencia Molecular , Mutación , Transducción de Señal , Proteínas de Dominio T Box/metabolismo , Proteínas Wnt , Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA