Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros












Base de dados
Intervalo de ano de publicação
1.
Science ; 380(6649): eabn9257, 2023 06 09.
Artigo em Inglês | MEDLINE | ID: mdl-37289866

RESUMO

Aging is associated with changes in circulating levels of various molecules, some of which remain undefined. We find that concentrations of circulating taurine decline with aging in mice, monkeys, and humans. A reversal of this decline through taurine supplementation increased the health span (the period of healthy living) and life span in mice and health span in monkeys. Mechanistically, taurine reduced cellular senescence, protected against telomerase deficiency, suppressed mitochondrial dysfunction, decreased DNA damage, and attenuated inflammaging. In humans, lower taurine concentrations correlated with several age-related diseases and taurine concentrations increased after acute endurance exercise. Thus, taurine deficiency may be a driver of aging because its reversal increases health span in worms, rodents, and primates and life span in worms and rodents. Clinical trials in humans seem warranted to test whether taurine deficiency might drive aging in humans.


Assuntos
Envelhecimento , Taurina , Animais , Humanos , Camundongos , Envelhecimento/sangue , Envelhecimento/efeitos dos fármacos , Envelhecimento/metabolismo , Senescência Celular , Haplorrinos , Longevidade/efeitos dos fármacos , Longevidade/fisiologia , Taurina/sangue , Taurina/deficiência , Taurina/farmacologia , Suplementos Nutricionais , Dano ao DNA/efeitos dos fármacos , Telomerase/metabolismo
2.
Sci Adv ; 9(7): eade4814, 2023 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-36800428

RESUMO

Alternative polyadenylation (APA) creates distinct transcripts from the same gene by cleaving the pre-mRNA at poly(A) sites that can lie within the 3' untranslated region (3'UTR), introns, or exons. Most studies focus on APA within the 3'UTR; however, here, we show that CPSF6 insufficiency alters protein levels and causes a developmental syndrome by deregulating APA throughout the transcript. In neonatal humans and zebrafish larvae, CPSF6 insufficiency shifts poly(A) site usage between the 3'UTR and internal sites in a pathway-specific manner. Genes associated with neuronal function undergo mostly intronic APA, reducing their expression, while genes associated with heart and skeletal function mostly undergo 3'UTR APA and are up-regulated. This suggests that, under healthy conditions, cells toggle between internal and 3'UTR APA to modulate protein expression.


Assuntos
Poliadenilação , Peixe-Zebra , Animais , Humanos , Recém-Nascido , Regiões 3' não Traduzidas , Éxons , Íntrons/genética , Peixe-Zebra/genética , Embrião não Mamífero
3.
Nat Commun ; 13(1): 2970, 2022 05 27.
Artigo em Inglês | MEDLINE | ID: mdl-35624100

RESUMO

The cardiac developmental network has been associated with myocardial regenerative potential. However, the embryonic signals triggered following injury have yet to be fully elucidated. Nkx2.5 is a key causative transcription factor associated with human congenital heart disease and one of the earliest markers of cardiac progenitors, thus it serves as a promising candidate. Here, we show that cardiac-specific RNA-sequencing studies reveal a disrupted embryonic transcriptional profile in the adult Nkx2.5 loss-of-function myocardium. nkx2.5-/- fish exhibit an impaired ability to recover following ventricular apex amputation with diminished dedifferentiation and proliferation. Complex network analyses illuminate that Nkx2.5 is required to provoke proteolytic pathways necessary for sarcomere disassembly and to mount a proliferative response for cardiomyocyte renewal. Moreover, Nkx2.5 targets embedded in these distinct gene regulatory modules coordinate appropriate, multi-faceted injury responses. Altogether, our findings support a previously unrecognized, Nkx2.5-dependent regenerative circuit that invokes myocardial cell cycle re-entry, proteolysis, and mitochondrial metabolism to ensure effective regeneration in the teleost heart.


Assuntos
Miocárdio , Miócitos Cardíacos , Animais , Ventrículos do Coração/metabolismo , Proteína Homeobox Nkx-2.5/genética , Proteína Homeobox Nkx-2.5/metabolismo , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Fatores de Transcrição/metabolismo
4.
Nat Commun ; 11(1): 2141, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-32358538

RESUMO

Optogenetic genome engineering tools enable spatiotemporal control of gene expression and provide new insight into biological function. Here, we report the new version of genetically encoded photoactivatable (PA) Cre recombinase, PA-Cre 3.0. To improve PA-Cre technology, we compare light-dimerization tools and optimize for mammalian expression using a CAG promoter, Magnets, and 2A self-cleaving peptide. To prevent background recombination caused by the high sequence similarity in the dimerization domains, we modify the codons for mouse gene targeting and viral production. Overall, these modifications significantly reduce dark leak activity and improve blue-light induction developing our new version, PA-Cre 3.0. As a resource, we have generated and validated AAV-PA-Cre 3.0 as well as two mouse lines that can conditionally express PA-Cre 3.0. Together these new tools will facilitate further biological and biomedical research.


Assuntos
Integrases/metabolismo , Recombinação Genética/genética , Animais , Códon/genética , Engenharia Genética/métodos , Integrases/genética , Luz , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Optogenética , Regiões Promotoras Genéticas/genética , Regiões Promotoras Genéticas/efeitos da radiação , Recombinação Genética/efeitos da radiação
5.
Dev Biol ; 462(2): 223-234, 2020 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-32272116

RESUMO

Vertebrate heart development requires spatiotemporal regulation of gene expression to specify cardiomyocytes, increase the cardiomyocyte population through proliferation, and to establish and maintain atrial and ventricular cardiac chamber identities. The evolutionarily conserved chromatin factor Gon4-like (Gon4l), encoded by the zebrafish ugly duckling (udu) locus, has previously been implicated in cell proliferation, cell survival, and specification of mesoderm-derived tissues including blood and somites, but its role in heart formation has not been studied. Here we report two distinct roles of Gon4l/Udu in heart development: regulation of cell proliferation and maintenance of ventricular identity. We show that zygotic loss of udu expression causes a significant reduction in cardiomyocyte number at one day post fertilization that becomes exacerbated during later development. We present evidence that the cardiomyocyte deficiency in udu mutants results from reduced cell proliferation, unlike hematopoietic deficiencies attributed to TP53-dependent apoptosis. We also demonstrate that expression of the G1/S-phase cell cycle regulator, cyclin E2 (ccne2), is reduced in udu mutant hearts, and that the Gon4l protein associates with regulatory regions of the ccne2 gene during early embryogenesis. Furthermore, udu mutant hearts exhibit a decrease in the proportion of ventricular cardiomyocytes compared to atrial cardiomyocytes, concomitant with progressive reduction of nkx2.5 expression. We further demonstrate that udu and nkx2.5 interact to maintain the proportion of ventricular cardiomyocytes during development. However, we find that ectopic expression of nkx2.5 is not sufficient to restore ventricular chamber identity suggesting that Gon4l regulates cardiac chamber patterning via multiple pathways. Together, our findings define a novel role for zygotically-expressed Gon4l in coordinating cardiomyocyte proliferation and chamber identity maintenance during cardiac development.


Assuntos
Fatores de Ligação de DNA Eritroide Específicos/metabolismo , Coração/embriologia , Miócitos Cardíacos/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Animais , Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Cromatina/metabolismo , Fatores de Ligação de DNA Eritroide Específicos/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/genética , Átrios do Coração/embriologia , Átrios do Coração/metabolismo , Miocárdio/metabolismo , Miócitos Cardíacos/fisiologia , Fase S/genética , Fatores de Transcrição/metabolismo , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/fisiologia
6.
Nat Methods ; 16(10): 1054-1062, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31562489

RESUMO

The limited per-pixel bandwidth of most microscopy methods requires compromises between field of view, sampling density and imaging speed. This limitation constrains studies involving complex motion or fast cellular signaling, and presents a major bottleneck for high-throughput structural imaging. Here, we combine high-speed intensified camera technology with a versatile, reconfigurable and dramatically improved Swept, Confocally Aligned Planar Excitation (SCAPE) microscope design that can achieve high-resolution volumetric imaging at over 300 volumes per second and over 1.2 GHz pixel rates. We demonstrate near-isotropic sampling in freely moving Caenorhabditis elegans, and analyze real-time blood flow and calcium dynamics in the beating zebrafish heart. The same system also permits high-throughput structural imaging of mounted, intact, cleared and expanded samples. SCAPE 2.0's significantly lower photodamage compared to point-scanning techniques is also confirmed. Our results demonstrate that SCAPE 2.0 is a powerful, yet accessible imaging platform for myriad emerging high-speed dynamic and high-throughput volumetric microscopy applications.


Assuntos
Microscopia/métodos , Animais , Caenorhabditis elegans/metabolismo , Cálcio/metabolismo , Coração/embriologia , Coração/fisiologia , Fótons , Peixe-Zebra/embriologia , Peixe-Zebra/metabolismo , Peixe-Zebra/fisiologia
7.
Development ; 145(3)2018 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-29361575

RESUMO

NKX2-5 is the most commonly mutated gene associated with human congenital heart defects (CHDs), with a predilection for cardiac pole abnormalities. This homeodomain transcription factor is a central regulator of cardiac development and is expressed in both the first and second heart fields (FHF and SHF). We have previously revealed essential functions of nkx2.5 and nkx2.7, two Nkx2-5 homologs expressed in zebrafish cardiomyocytes, in maintaining ventricular identity. However, the differential roles of these genes in the specific subpopulations of the anterior (aSHF) and posterior (pSHF) SHFs have yet to be fully defined. Here, we show that Nkx genes regulate aSHF and pSHF progenitors through independent mechanisms. We demonstrate that Nkx genes restrict proliferation of aSHF progenitors in the outflow tract, delimit the number of pSHF progenitors at the venous pole and pattern the sinoatrial node acting through Isl1 repression. Moreover, optical mapping highlights the requirement for Nkx gene dose in establishing electrophysiological chamber identity and in integrating the physiological connectivity of FHF and SHF cardiomyocytes. Ultimately, our results may shed light on the discrete errors responsible for NKX2-5-dependent human CHDs of the cardiac outflow and inflow tracts.


Assuntos
Coração/embriologia , Proteína Homeobox Nkx-2.5/genética , Proteínas de Homeodomínio/genética , Proteínas com Homeodomínio LIM/genética , Mioblastos Cardíacos/citologia , Mioblastos Cardíacos/metabolismo , Fatores de Transcrição/genética , Proteínas de Peixe-Zebra/genética , Animais , Animais Geneticamente Modificados , Padronização Corporal/genética , Diferenciação Celular , Proliferação de Células , Regulação da Expressão Gênica no Desenvolvimento , Cardiopatias Congênitas/embriologia , Cardiopatias Congênitas/genética , Humanos , Mutação
8.
Am J Physiol Regul Integr Comp Physiol ; 313(3): R265-R271, 2017 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-28615160

RESUMO

Heart rate variability (HRV) has become an important clinical marker of cardiovascular health and a research measure for the study of the cardiac conduction system and its autonomic controls. While the zebrafish (Danio rerio) is an ideal vertebrate model for understanding heart development, HRV has only recently been investigated in this system. We have previously demonstrated that nkx2.5 and nkx2.7, two homologues of Nkx2-5 expressed in zebrafish cardiomyocytes, play vital roles in maintaining cardiac chamber-specific characteristics. Given observed defects in ventricular and atrial chamber identities in nkx2.5-/- embryos coupled with conduction system abnormalities in murine models of Nkx2.5 insufficiency, we postulated that reduced HRV would serve as a marker of poor cardiac health in nkx2.5 mutants and in other zebrafish models of human congenital heart disease. Using live video image acquisition, we derived beat-to-beat intervals to compare HRV in wild-type and nkx2.5-/- embryos. Our data illustrate that the nkx2.5 loss-of-function model exhibits increased heart rate and decreased HRV when compared with wild type during embryogenesis. These findings validate HRV analysis as a useful quantitative tool for assessment of cardiac health in zebrafish and underscore the importance of nkx2.5 in maintaining normal heart rate and HRV during early conduction system development.


Assuntos
Desenvolvimento Embrionário/fisiologia , Frequência Cardíaca/fisiologia , Proteína Homeobox Nkx-2.5/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Peixe-Zebra/fisiologia , Animais
9.
Development ; 144(7): 1328-1338, 2017 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-28232600

RESUMO

Atrial and ventricular cardiac chambers behave as distinct subunits with unique morphological, electrophysiological and contractile properties. Despite the importance of chamber-specific features, chamber fate assignments remain relatively plastic, even after differentiation is underway. In zebrafish, Nkx transcription factors are essential for the maintenance of ventricular characteristics, but the signaling pathways that operate upstream of Nkx factors in this context are not well understood. Here, we show that FGF signaling plays an essential part in enforcing ventricular identity. Loss of FGF signaling results in a gradual accumulation of atrial cells, a corresponding loss of ventricular cells, and the appearance of ectopic atrial gene expression within the ventricle. These phenotypes reflect important roles for FGF signaling in promoting ventricular traits, both in early-differentiating cells that form the initial ventricle and in late-differentiating cells that append to its arterial pole. Moreover, we find that FGF signaling functions upstream of Nkx genes to inhibit ectopic atrial gene expression. Together, our data suggest a model in which sustained FGF signaling acts to suppress cardiomyocyte plasticity and to preserve the integrity of the ventricular chamber.


Assuntos
Fatores de Crescimento de Fibroblastos/metabolismo , Ventrículos do Coração/embriologia , Ventrículos do Coração/metabolismo , Organogênese , Transdução de Sinais , Peixe-Zebra/embriologia , Peixe-Zebra/metabolismo , Animais , Diferenciação Celular , Embrião não Mamífero/citologia , Embrião não Mamífero/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Átrios do Coração/citologia , Ventrículos do Coração/citologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Organogênese/genética , Transdução de Sinais/genética , Fatores de Tempo , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
10.
Genesis ; 55(3)2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28109039

RESUMO

In gnathostomes, dorsoventral (D-V) patterning of neural crest cells (NCC) within the pharyngeal arches is crucial for the development of hinged jaws. One of the key signals that mediate this process is Endothelin-1 (EDN1). Loss of EDN1 binding to the Endothelin-A receptor (EDNRA) results in loss of EDNRA signaling and subsequent facial birth defects in humans, mice and zebrafish. A rate-limiting step in this crucial signaling pathway is the conversion of immature EDN1 into a mature active form by Endothelin converting enzyme-1 (ECE1). However, surprisingly little is known about how Ece1 transcription is induced or regulated. We show here that Nkx2.5 is required for proper craniofacial development in zebrafish and acts in part by upregulating ece1 expression. Disruption of nkx2.5 in zebrafish embryos results in defects in both ventral and dorsal pharyngeal arch-derived elements, with changes in ventral arch gene expression consistent with a disruption in Ednra signaling. ece1 mRNA rescues the nkx2.5 morphant phenotype, indicating that Nkx2.5 functions through modulating Ece1 expression or function. These studies illustrate a new function for Nkx2.5 in embryonic development and provide new avenues with which to pursue potential mechanisms underlying human facial disorders.


Assuntos
Enzimas Conversoras de Endotelina/genética , Regulação da Expressão Gênica no Desenvolvimento , Proteína Homeobox Nkx-2.5/genética , Crista Neural/metabolismo , Proteínas de Peixe-Zebra/genética , Animais , Enzimas Conversoras de Endotelina/metabolismo , Proteína Homeobox Nkx-2.5/metabolismo , Camundongos , Crista Neural/embriologia , Faringe/embriologia , Faringe/metabolismo , Regulação para Cima , Peixe-Zebra , Proteínas de Peixe-Zebra/metabolismo
11.
Curr Biol ; 25(16): 2099-110, 2015 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-26255850

RESUMO

The pharyngeal arch arteries (PAAs) are a series of paired embryonic blood vessels that give rise to several major arteries that connect directly to the heart. During development, the PAAs emerge from nkx2.5-expressing mesodermal cells and connect the dorsal head vasculature to the outflow tract of the heart. Despite their central role in establishing the circulatory system, the embryonic origins of the PAA progenitors are only coarsely defined, and the factors that specify them and their regenerative potential are unclear. Using fate mapping and mutant analysis, we find that PAA progenitors are derived from the tcf21 and nkx2.5 double-positive head mesoderm and require these two transcription factors for their specification and survival. Unexpectedly, cell ablation shows that the tcf21+; nkx2.5+ PAA progenitors are not required for PAA formation. We find that this compensation is due to the replacement of ablated tcf21+; nkx2.5+ PAA cells by endothelial cells from the dorsal head vasculature. Together, these studies assign the embryonic origin of the great vessel progenitors to the interface between the pharyngeal and cardiac mesoderm, identify the transcription factor code required for their specification, and reveal an unexpected plasticity in the formation of the great vessels.


Assuntos
Coração/embriologia , Peixe-Zebra/embriologia , Animais , Região Branquial/embriologia , Vasos Coronários/embriologia , Proteínas de Peixes/genética , Proteínas de Peixes/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Peixe-Zebra/genética
12.
ACS Chem Biol ; 10(3): 901-8, 2015 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-25560305

RESUMO

Protein metabolism, consisting of both synthesis and degradation, is highly complex, playing an indispensable regulatory role throughout physiological and pathological processes. Over recent decades, extensive efforts, using approaches such as autoradiography, mass spectrometry, and fluorescence microscopy, have been devoted to the study of protein metabolism. However, noninvasive and global visualization of protein metabolism has proven to be highly challenging, especially in live systems. Recently, stimulated Raman scattering (SRS) microscopy coupled with metabolic labeling of deuterated amino acids (D-AAs) was demonstrated for use in imaging newly synthesized proteins in cultured cell lines. Herein, we significantly generalize this notion to develop a comprehensive labeling and imaging platform for live visualization of complex protein metabolism, including synthesis, degradation, and pulse-chase analysis of two temporally defined populations. First, the deuterium labeling efficiency was optimized, allowing time-lapse imaging of protein synthesis dynamics within individual live cells with high spatial-temporal resolution. Second, by tracking the methyl group (CH3) distribution attributed to pre-existing proteins, this platform also enables us to map protein degradation inside live cells. Third, using two subsets of structurally and spectroscopically distinct D-AAs, we achieved two-color pulse-chase imaging, as demonstrated by observing aggregate formation of mutant hungtingtin proteins. Finally, going beyond simple cell lines, we demonstrated the imaging ability of protein synthesis in brain tissues, zebrafish, and mice in vivo. Hence, the presented labeling and imaging platform would be a valuable tool to study complex protein metabolism with high sensitivity, resolution, and biocompatibility for a broad spectrum of systems ranging from cells to model animals and possibly to humans.


Assuntos
Aminoácidos/metabolismo , Microscopia/métodos , Biossíntese de Proteínas , Proteínas/metabolismo , Proteólise , Análise Espectral Raman/métodos , Animais , Encéfalo/metabolismo , Encéfalo/ultraestrutura , Células Cultivadas , Deutério/metabolismo , Embrião não Mamífero , Células HeLa , Humanos , Marcação por Isótopo/métodos , Camundongos , Microscopia/instrumentação , Imagem Molecular/instrumentação , Imagem Molecular/métodos , Neurônios/metabolismo , Neurônios/ultraestrutura , Proteínas/química , Análise Espectral Raman/instrumentação , Peixe-Zebra
13.
Dev Biol ; 400(1): 10-22, 2015 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-25536398

RESUMO

Temporally controlled mechanisms that define the unique features of ventricular and atrial cardiomyocyte identities are essential for the construction of a coordinated, morphologically intact heart. We have previously demonstrated an important role for nkx genes in maintaining ventricular identity, however, the specific timing of nkx2.5 function in distinct cardiomyocyte populations has yet to be elucidated. Here, we show that heat-shock induction of a novel transgenic line, Tg(hsp70l:nkx2.5-EGFP), during the initial stages of cardiomyocyte differentiation leads to rescue of chamber shape and identity in nkx2.5(-/-) embryos as chambers emerge. Intriguingly, our findings link an early role of this essential cardiac transcription factor with a later function. Moreover, these data reveal that nkx2.5 is also required in the second heart field as the heart tube forms, reflecting the temporal delay in differentiation of this population. Thus, our results support a model in which nkx genes induce downstream targets that are necessary to maintain chamber-specific identity in both early- and late-differentiating cardiomyocytes at discrete stages in cardiac morphogenesis. Furthermore, we show that overexpression of nkx2.5 during the first and second heart field development not only rescues the mutant phenotype, but also is sufficient for proper function of the adult heart. Taken together, these results shed new light on the stage-dependent mechanisms that sculpt chamber-specific cardiomyocytes and, therefore, have the potential to improve in vitro generation of ventricular cells to treat myocardial infarction and congenital heart disease.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Ventrículos do Coração/embriologia , Morfogênese/fisiologia , Miócitos Cardíacos/fisiologia , Fatores de Transcrição/fisiologia , Proteínas de Peixe-Zebra/fisiologia , Peixe-Zebra/embriologia , Animais , Western Blotting , Contagem de Células , Diferenciação Celular/fisiologia , Primers do DNA/genética , Técnicas de Inativação de Genes , Genótipo , Proteínas de Fluorescência Verde/metabolismo , Proteína Homeobox Nkx-2.5 , Processamento de Imagem Assistida por Computador , Hibridização In Situ , Microscopia de Fluorescência , Reação em Cadeia da Polimerase , Fatores de Transcrição/genética , Proteínas de Peixe-Zebra/genética
14.
Development ; 140(20): 4203-13, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24026123

RESUMO

Establishment of specific characteristics of each embryonic cardiac chamber is crucial for development of a fully functional adult heart. Despite the importance of defining and maintaining unique features in ventricular and atrial cardiomyocytes, the regulatory mechanisms guiding these processes are poorly understood. Here, we show that the homeodomain transcription factors Nkx2.5 and Nkx2.7 are necessary to sustain ventricular chamber attributes through repression of atrial chamber identity. Mutation of nkx2.5 in zebrafish yields embryos with diminutive ventricular and bulbous atrial chambers. These chamber deformities emerge gradually during development, with a severe collapse in the number of ventricular cardiomyocytes and an accumulation of excess atrial cardiomyocytes as the heart matures. Removal of nkx2.7 function from nkx2.5 mutants exacerbates the loss of ventricular cells and the gain of atrial cells. Moreover, in these Nkx-deficient embryos, expression of vmhc, a ventricular gene, fades, whereas expression of amhc, an atrial gene, expands. Cell-labeling experiments suggest that ventricular cardiomyocytes can transform into atrial cardiomyocytes in the absence of Nkx gene function. Through suggestion of transdifferentiation from ventricular to atrial fate, our data reveal a pivotal role for Nkx genes in maintaining ventricular identity and highlight remarkable plasticity in differentiated myocardium. Thus, our results are relevant to the etiologies of fetal and neonatal cardiac pathology and could direct future innovations in cardiac regenerative medicine.


Assuntos
Átrios do Coração/embriologia , Ventrículos do Coração/embriologia , Proteínas de Homeodomínio/metabolismo , Fatores de Transcrição/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Animais , Miosinas Atriais/biossíntese , Diferenciação Celular , Proliferação de Células , Regulação da Expressão Gênica no Desenvolvimento , Genótipo , Átrios do Coração/metabolismo , Ventrículos do Coração/metabolismo , Proteína Homeobox Nkx-2.5 , Proteínas de Homeodomínio/genética , Mutação , Miócitos Cardíacos/metabolismo , Fatores de Transcrição/genética , Transcrição Gênica , Miosinas Ventriculares/biossíntese , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética
15.
Radiat Environ Biophys ; 52(3): 411-7, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23708525

RESUMO

The Radiological Research Accelerator Facility at Columbia University has recently added a UV microspot irradiator to a microbeam irradiation platform. This UV microspot irradiator applies multiphoton excitation at the focal point of an incident laser as the source for cell damage, and with this approach, a single cell within a 3D sample can be targeted and exposed to damaging UV. The UV microspot's ability to impart cellular damage within 3D is an advantage over all other microbeam techniques, which instead impart damage to numerous cells along microbeam tracks. This short communication is an overview, and a description of the UV microspot including the following applications and demonstrations of selective damage to live single cell targets: DNA damage foci formation, patterned irradiation, photoactivation, targeting of mitochondria, and targeting of individual cardiomyocytes in a live zebrafish embryo.


Assuntos
Microscopia de Fluorescência por Excitação Multifotônica/instrumentação , Radiobiologia/instrumentação , Raios Ultravioleta , Animais , Linhagem Celular Tumoral , Núcleo Celular/efeitos da radiação , Dano ao DNA , Embrião não Mamífero/efeitos da radiação , Fibroblastos/efeitos da radiação , Proteínas de Fluorescência Verde , Células HeLa , Coração/embriologia , Coração/efeitos da radiação , Humanos , Mitocôndrias/efeitos da radiação , Miócitos Cardíacos/efeitos da radiação , New York , Universidades , Peixe-Zebra
16.
Dev Biol ; 322(2): 314-21, 2008 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-18718462

RESUMO

Heart formation is a complex morphogenetic process, and perturbations in cardiac morphogenesis lead to congenital heart disease. NKX2-5 is a key causative gene associated with cardiac birth defects, presumably because of its essential roles during the early steps of cardiogenesis. Previous studies in model organisms implicate NKX2-5 homologs in numerous processes, including cardiac progenitor specification, progenitor proliferation, and chamber morphogenesis. By inhibiting function of the zebrafish NKX2-5 homologs, nkx2.5 and nkx2.7, we show that nkx genes are essential to establish the original dimensions of the linear heart tube. The nkx-deficient heart tube fails to elongate normally: its ventricular portion is atypically short and wide, and its atrial portion is disorganized and sprawling. This atrial phenotype is associated with a surplus of atrial cardiomyocytes, whereas ventricular cell number is normal at this stage. However, ventricular cell number is decreased in nkx-deficient embryos later in development, when cardiac chambers are emerging. Thus, we conclude that nkx genes regulate heart tube extension and exert differential effects on ventricular and atrial cell number. Our data suggest that morphogenetic errors could originate during early stages of heart tube assembly in patients with NKX2-5 mutations.


Assuntos
Coração/embriologia , Proteínas de Homeodomínio/fisiologia , Fatores de Transcrição/fisiologia , Proteínas de Peixe-Zebra/fisiologia , Peixe-Zebra/embriologia , Animais , Átrios do Coração/citologia , Átrios do Coração/embriologia , Átrios do Coração/metabolismo , Ventrículos do Coração/citologia , Ventrículos do Coração/embriologia , Ventrículos do Coração/metabolismo , Proteína Homeobox Nkx-2.5 , Morfogênese , Miocárdio/citologia , Miocárdio/metabolismo , Especificidade de Órgãos , Peixe-Zebra/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...