Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 15(1): 1982, 2024 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-38438357

RESUMO

De novo synthesis of the pyrimidine, cytidine triphosphate (CTP), is crucial for DNA/RNA metabolism and depends on the CTP synthetases, CTPS1 and -2. Partial CTPS1 deficiency in humans has previously been shown to lead to immunodeficiency, with impaired expansion of T and B cells. Here, we examine the effects of conditional and inducible inactivation of Ctps1 and/or Ctps2 on mouse embryonic development and immunity. We report that deletion of Ctps1, but not Ctps2, is embryonic-lethal. Tissue and cells with high proliferation and renewal rates, such as intestinal epithelium, erythroid and thymic lineages, activated B and T lymphocytes, and memory T cells strongly rely on CTPS1 for their maintenance and growth. However, both CTPS1 and CTPS2 are required for T cell proliferation following TCR stimulation. Deletion of Ctps1 in T cells or treatment with a CTPS1 inhibitor rescued Foxp3-deficient mice from fatal systemic autoimmunity and reduced the severity of experimental autoimmune encephalomyelitis. These findings support that CTPS1 may represent a target for immune suppression.


Assuntos
Autoimunidade , Desenvolvimento Embrionário , Feminino , Gravidez , Humanos , Animais , Camundongos , Citidina Trifosfato , Autoimunidade/genética , Linfócitos B , Proliferação de Células
2.
Dev Cell ; 58(21): 2217-2234.e8, 2023 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-37852253

RESUMO

Despite their burden, most congenital defects remain poorly understood, due to lack of knowledge of embryological mechanisms. Here, we identify Greb1l mutants as a mouse model of crisscross heart. Based on 3D quantifications of shape changes, we demonstrate that torsion of the atrioventricular canal occurs together with supero-inferior ventricles at E10.5, after heart looping. Mutants phenocopy partial deficiency in retinoic acid signaling, which reflect overlapping pathways in cardiac precursors. Spatiotemporal gene mapping and cross-correlated transcriptomic analyses further reveal the role of Greb1l in maintaining a pool of dorsal pericardial wall precursor cells during heart tube elongation, likely by controlling ribosome biogenesis and cell differentiation. Consequently, we observe growth arrest and malposition of the outflow tract, which are predictive of abnormal tube remodeling in mutants. Our work on a rare cardiac malformation opens novel perspectives on the origin of a broader spectrum of congenital defects associated with GREB1L in humans.


Assuntos
Coração Entrecruzado , Humanos , Animais , Camundongos , Morfogênese/genética , Coração , Ventrículos do Coração , Células-Tronco
3.
Curr Opin Genet Dev ; 73: 101896, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35026527

RESUMO

As other tubular organs, the embryonic heart develops from an epithelial sheet of cells, referred to as the heart field. The second heart field, which lies in the dorsal pericardial wall, constitutes a transient cell reservoir, integrating patterning and polarity cues. Conditional mutants have shown that impairment of the epithelial architecture of the second heart field is associated with congenital heart defects. Here, taking the mouse as a model, we review the epithelial properties of the second heart field and how they are modulated upon cardiomyocyte differentiation. Compared to other cases of tubulogenesis, the cellular dynamics in the second heart field are only beginning to be revealed. A challenge for the future will be to unravel key physical forces driving heart tube morphogenesis.


Assuntos
Coração , Organogênese , Animais , Diferenciação Celular/genética , Epitélio , Camundongos , Morfogênese , Organogênese/genética
4.
Annu Rev Genomics Hum Genet ; 22: 257-284, 2021 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-34061573

RESUMO

Congenital heart disease is the most frequent birth defect and the leading cause of death for the fetus and in the first year of life. The wide phenotypic diversity of congenital heart defects requires expert diagnosis and sophisticated repair surgery. Although these defects have been described since the seventeenth century, it was only in 2005 that a consensus international nomenclature was adopted, followed by an international classification in 2017 to help provide better management of patients. Advances in genetic engineering, imaging, and omics analyses have uncovered mechanisms of heart formation and malformation in animal models, but approximately 80% of congenital heart defects have an unknown genetic origin. Here, we summarize current knowledge of congenital structural heart defects, intertwining clinical and fundamental research perspectives, with the aim to foster interdisciplinary collaborations at the cutting edge of each field. We also discuss remaining challenges in better understanding congenital heart defects and providing benefits to patients.


Assuntos
Cardiopatias Congênitas , Animais , Cardiopatias Congênitas/genética , Humanos , Modelos Animais
5.
Dev Cell ; 55(4): 413-431.e6, 2020 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-33171097

RESUMO

The secreted factor Nodal, known as a major left determinant, is associated with severe heart defects. Yet, it has been unclear how it regulates asymmetric morphogenesis such as heart looping, which align cardiac chambers to establish the double blood circulation. Here, we report that Nodal is transiently active in precursors of the mouse heart tube poles, before looping. In conditional mutants, we show that Nodal is not required to initiate asymmetric morphogenesis. We provide evidence of a heart-specific random generator of asymmetry that is independent of Nodal. Using 3D quantifications and simulations, we demonstrate that Nodal functions as a bias of this mechanism: it is required to amplify and coordinate opposed left-right asymmetries at the heart tube poles, thus generating a robust helical shape. We identify downstream effectors of Nodal signaling, regulating asymmetries in cell proliferation, differentiation, and extracellular matrix composition. Our study uncovers how Nodal regulates asymmetric organogenesis.


Assuntos
Padronização Corporal , Coração/embriologia , Proteína Nodal/metabolismo , Transdução de Sinais , Animais , Diferenciação Celular , Proliferação de Células , Simulação por Computador , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/patologia , Matriz Extracelular/metabolismo , Cardiopatias Congênitas/metabolismo , Mesoderma/metabolismo , Camundongos , Miocárdio/metabolismo , Miocárdio/patologia , Transgenes
6.
Philos Trans R Soc Lond B Biol Sci ; 375(1809): 20190556, 2020 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-32829679

RESUMO

Retinoic acid (RA), derived from vitamin A, is a major teratogen, clinically recognized in 1983. Identification of its natural presence in the embryo and dissection of its molecular mechanism of action became possible in the animal model with the advent of molecular biology, starting with the cloning of its nuclear receptor. In normal development, the dose of RA is tightly controlled to regulate organ formation. Its production depends on enzymes, which have a dynamic expression profile during embryonic development. As a small molecule, it diffuses rapidly and acts as a morphogen. Here, we review advances in deciphering how endogenously produced RA provides positional information to cells. We compare three mesodermal tissues, the limb, the somites and the heart, and discuss how RA signalling regulates antero-posterior and left-right patterning. A common principle is the establishment of its spatio-temporal dynamics by positive and negative feedback mechanisms and by antagonistic signalling by FGF. However, the response is cell-specific, pointing to the existence of cofactors and effectors, which are as yet incompletely characterized. This article is part of a discussion meeting issue 'Contemporary morphogenesis'.


Assuntos
Padronização Corporal , Embrião de Mamíferos/embriologia , Embrião não Mamífero/embriologia , Mesoderma/embriologia , Transdução de Sinais , Tretinoína/metabolismo , Animais , Camundongos , Peixe-Zebra
7.
Cell Rep ; 32(3): 107932, 2020 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-32698004

RESUMO

Cilia and the intraflagellar transport (IFT) proteins involved in ciliogenesis are associated with congenital heart diseases (CHDs). However, the molecular links between cilia, IFT proteins, and cardiogenesis are yet to be established. Using a combination of biochemistry, genetics, and live-imaging methods, we show that IFT complex B proteins (Ift88, Ift54, and Ift20) modulate the Hippo pathway effector YAP1 in zebrafish and mouse. We demonstrate that this interaction is key to restrict the formation of the proepicardium and the myocardium. In cellulo experiments suggest that IFT88 and IFT20 interact with YAP1 in the cytoplasm and functionally modulate its activity, identifying a molecular link between cilia-related proteins and the Hippo pathway. Taken together, our results highlight a noncanonical role for IFT complex B proteins during cardiogenesis and shed light on a mechanism of action for ciliary proteins in YAP1 regulation.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas de Transporte/metabolismo , Proteínas de Ciclo Celular/metabolismo , Flagelos/metabolismo , Coração/embriologia , Organogênese , Proteínas Serina-Treonina Quinases/metabolismo , Transativadores/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Animais , Transporte Biológico , Proteínas Morfogenéticas Ósseas/metabolismo , Cílios/metabolismo , Células HEK293 , Células HeLa , Humanos , Camundongos Endogâmicos C57BL , Pericárdio/metabolismo , Ligação Proteica , Transdução de Sinais , Proteínas de Sinalização YAP
8.
Dis Model Mech ; 12(7)2019 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-31208960

RESUMO

Laterality defects are developmental disorders resulting from aberrant left/right patterning. In the most severe cases, such as in heterotaxy, they are associated with complex malformations of the heart. Advances in understanding the underlying physiopathological mechanisms have been hindered by the lack of a standardised and exhaustive procedure in mouse models for phenotyping left/right asymmetries of all visceral organs. Here, we have developed a multimodality imaging pipeline, which combines non-invasive micro-ultrasound imaging, micro-computed tomography (micro-CT) and high-resolution episcopic microscopy (HREM) to acquire 3D images at multiple stages of development and at multiple scales. On the basis of the position in the uterine horns, we track in a single individual, the progression of organ asymmetry, the situs of all visceral organs in the thoracic or abdominal environment, and the fine anatomical left/right asymmetries of cardiac segments. We provide reference anatomical images and organ reconstructions in the mouse, and discuss differences with humans. This standardised pipeline, which we validated in a mouse model of heterotaxy, offers a fast and easy-to-implement framework. The extensive 3D phenotyping of organ asymmetry in the mouse uses the clinical nomenclature for direct comparison with patient phenotypes. It is compatible with automated and quantitative image analyses, which is essential to compare mutant phenotypes with incomplete penetrance and to gain mechanistic insight into laterality defects.


Assuntos
Padronização Corporal , Cardiopatias Congênitas/diagnóstico por imagem , Fenótipo , Animais , Modelos Animais de Doenças , Feto/diagnóstico por imagem , Coração/diagnóstico por imagem , Coração/embriologia , Imageamento Tridimensional , Camundongos , Ultrassonografia , Microtomografia por Raio-X
9.
Development ; 145(22)2018 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-30467108

RESUMO

Extensive studies have shown how bilateral symmetry of the vertebrate embryo is broken during early development, resulting in a molecular left-right bias in the mesoderm. However, how this early asymmetry drives the asymmetric morphogenesis of visceral organs remains poorly understood. The heart provides a striking model of left-right asymmetric morphogenesis, undergoing rightward looping to shape an initially linear heart tube and align cardiac chambers. Importantly, abnormal left-right patterning is associated with severe congenital heart defects, as exemplified in heterotaxy syndrome. Here, we compare the mechanisms underlying the rightward looping of the heart tube in fish, chick and mouse embryos. We propose that heart looping is not only a question of direction, but also one of fine-tuning shape. This is discussed in the context of evolutionary and clinical perspectives.


Assuntos
Padronização Corporal , Cardiopatias/embriologia , Coração/embriologia , Animais , Humanos , Modelos Biológicos , Morfogênese , Vertebrados/embriologia
10.
Nat Rev Cardiol ; 15(11): 705-724, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30266935

RESUMO

The function of the mammalian heart depends on the interplay between different cardiac cell types. The deployment of these cells, with precise spatiotemporal regulation, is also important during development to establish the heart structure. In this Review, we discuss the diverse origins of cardiac cell types and the lineage relationships between cells of a given type that contribute to different parts of the heart. The emerging lineage tree shows the progression of cell fate diversification, with patterning cues preceding cell type segregation, as well as points of convergence, with overlapping lineages contributing to a given tissue. Several cell lineage markers have been identified. However, caution is required with genetic-tracing experiments in comparison with clonal analyses. Genetic studies on cell populations provided insights into the mechanisms for lineage decisions. In the past 3 years, results of single-cell transcriptomics are beginning to reveal cell heterogeneity and early developmental trajectories. Equating this information with the in vivo location of cells and their lineage history is a current challenge. Characterization of the progenitor cells that form the heart and of the gene regulatory networks that control their deployment is of major importance for understanding the origin of congenital heart malformations and for producing cardiac tissue for use in regenerative medicine.


Assuntos
Diferenciação Celular , Linhagem da Célula , Proliferação de Células , Coração Fetal/anormalidades , Cardiopatias Congênitas/patologia , Miócitos Cardíacos/patologia , Animais , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Coração Fetal/metabolismo , Coração Fetal/fisiopatologia , Fibroblastos/metabolismo , Fibroblastos/patologia , Regulação da Expressão Gênica no Desenvolvimento , Redes Reguladoras de Genes , Cardiopatias Congênitas/metabolismo , Cardiopatias Congênitas/fisiopatologia , Humanos , Miócitos Cardíacos/metabolismo , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Organogênese , Fenótipo , Regeneração , Medicina Regenerativa/métodos , Transdução de Sinais
11.
Elife ; 62017 11 28.
Artigo em Inglês | MEDLINE | ID: mdl-29179813

RESUMO

How left-right patterning drives asymmetric morphogenesis is unclear. Here, we have quantified shape changes during mouse heart looping, from 3D reconstructions by HREM. In combination with cell labelling and computer simulations, we propose a novel model of heart looping. Buckling, when the cardiac tube grows between fixed poles, is modulated by the progressive breakdown of the dorsal mesocardium. We have identified sequential left-right asymmetries at the poles, which bias the buckling in opposite directions, thus leading to a helical shape. Our predictive model is useful to explore the parameter space generating shape variations. The role of the dorsal mesocardium was validated in Shh-/- mutants, which recapitulate heart shape changes expected from a persistent dorsal mesocardium. Our computer and quantitative tools provide novel insight into the mechanism of heart looping and the contribution of different factors, beyond the simple description of looping direction. This is relevant to congenital heart defects.


Assuntos
Coração/embriologia , Morfogênese , Animais , Simulação por Computador , Imageamento Tridimensional , Camundongos , Microscopia
12.
Nat Commun ; 8: 14582, 2017 02 27.
Artigo em Inglês | MEDLINE | ID: mdl-28239148

RESUMO

Although in flies the atypical cadherin Fat is an upstream regulator of Hippo signalling, the closest mammalian homologue, Fat4, has been shown to regulate tissue polarity rather than growth. Here we show in the mouse heart that Fat4 modulates Hippo signalling to restrict growth. Fat4 mutant myocardium is thicker, with increased cardiomyocyte size and proliferation, and this is mediated by an upregulation of the transcriptional activity of Yap1, an effector of the Hippo pathway. Fat4 is not required for the canonical activation of Hippo kinases but it sequesters a partner of Yap1, Amotl1, out of the nucleus. The nuclear translocation of Amotl1 is accompanied by Yap1 to promote cardiomyocyte proliferation. We, therefore, identify Amotl1, which is not present in flies, as a mammalian intermediate for non-canonical Hippo signalling, downstream of Fat4. This work uncovers a mechanism for the restriction of heart growth at birth, a process which impedes the regenerative potential of the mammalian heart.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Caderinas/metabolismo , Coração/crescimento & desenvolvimento , Proteínas de Membrana/metabolismo , Fosfoproteínas/metabolismo , Proteína 1 Semelhante a Angiopoietina , Animais , Animais Recém-Nascidos , Cardiomegalia/genética , Cardiomegalia/patologia , Proteínas de Ciclo Celular , Proliferação de Células , Desmossomos/metabolismo , Desmossomos/ultraestrutura , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Modelos Biológicos , Ligação Proteica , Ratos , Transdução de Sinais , Proteínas de Sinalização YAP
13.
Cilia ; 4: 5, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25974046

RESUMO

The Cilia 2014 conference was organised by four European networks: the Ciliopathy Alliance, the Groupement de Recherche CIL, the Nordic Cilia and Centrosome Network and the EU FP7 programme SYSCILIA. More than 400 delegates from 27 countries gathered at the Institut Pasteur conference centre in Paris, including 30 patients and patient representatives. The meeting offered a unique opportunity for exchange between different scientific and medical communities. Major highlights included new discoveries about the roles of motile and immotile cilia during development and homeostasis, the mechanism of cilium construction, as well as progress in diagnosis and possible treatment of ciliopathies. The contributions to the cilia field of flagellated infectious eukaryotes and of systems biology were also presented.

14.
Methods Cell Biol ; 127: 55-73, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25837386

RESUMO

The primary cilium is a small sensory organelle that is required for different aspects of embryonic development, including the formation of the heart. The structure and composition of cilia have been extensively studied, so that several markers of primary cilia have now been identified. However, the role of cilia in specific cell types remains poorly understood. We describe here a series of approaches to image primary cilia in the rodent heart or in primary cultures of cells dissociated from the heart. As the cilium is a marker of cell polarity, we also provide, for quantitative image analysis of cilium orientation, tools which are generally applicable to other types of tissues.


Assuntos
Cílios/fisiologia , Coração/fisiologia , Miocárdio/citologia , Miócitos Cardíacos/fisiologia , Animais , Diferenciação Celular , Polaridade Celular/fisiologia , Células Cultivadas , Dissecação , Embrião de Mamíferos/fisiologia , Desenvolvimento Embrionário , Processamento de Imagem Assistida por Computador , Camundongos , Miócitos Cardíacos/citologia , Cultura Primária de Células , Transdução de Sinais , Coloração e Rotulagem/métodos , Fixação de Tecidos/métodos
16.
Med Sci (Paris) ; 30(11): 996-1003, 2014 Nov.
Artigo em Francês | MEDLINE | ID: mdl-25388582

RESUMO

After the seminal discovery in 2000 that primary cilia are functional organelles which are essential for embryonic development, several mouse models of ciliopathies have been generated. The heart is frequently affected, with a large spectrum of malformations. The cilia of the node are required early in development in the determination of the left/right laterality of the embryo, which has secondary consequences on the formation of the heart. Thus, abnormal looping of the heart is a recurrent phenotype in models of ciliopathies. However, the function of primary cilia in cardiac cells remains poorly understood. Receptors such as polycystins or hedgehog receptors are usually localized in the primary cilium, raising the possibility that these signalling pathways, which are important for the septation and the growth of the heart, are transduced in primary cilia of cardiac cells. Knowledge of the roles of primary cilia at different steps of heart development and in different cardiac cell types will be essential to better understand the origin of human cardiopathies associated with ciliopathies.


Assuntos
Cílios/fisiologia , Coração/embriologia , Animais , Transtornos da Motilidade Ciliar/genética , Transtornos da Motilidade Ciliar/patologia , Vasos Coronários/embriologia , Desenvolvimento Embrionário , Coração Fetal/crescimento & desenvolvimento , Coração Fetal/ultraestrutura , Cardiopatias Congênitas/genética , Cardiopatias Congênitas/patologia , Proteínas Hedgehog/genética , Proteínas Hedgehog/fisiologia , Humanos , Camundongos , Morfogênese/fisiologia , Receptores Patched , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/fisiologia , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/fisiologia , Transdução de Sinais/fisiologia , Receptor Smoothened , Síndrome , Canais de Cátion TRPP/genética , Canais de Cátion TRPP/fisiologia , Vertebrados/embriologia
17.
Artigo em Inglês | MEDLINE | ID: mdl-25286050

RESUMO

Insights into the mechanisms of development of the mammalian four-chambered heart are based on biological observations at organ, tissue, cell, and molecular levels, but the full integration of these experimental data awaits a systems biology approach. Such an approach can be employed to formulate and test conceptual models in a computational simulation. To illustrate how this can be applied to heart development, we used the process of trabeculation, which is the formation of muscular strands during chamber development. We selected this process because it is localized, involves a restricted number of cell types, and a range of experimental data is available. Trabeculation of the ventricles is based on the interplay between endocardial and myocardial cells and involves molecular pathways underlying cell-cell interactions and tissue-specific cell behavior. A cellular Potts model was used for the simulation of these multi-scale processes. With fairly simple inputs, of which the relative contributions are unknown, an iterative exploration achieved an outcome that resembles the trabeculation process and allows further investigation of contributing factors. The systems biology pipeline from biological observations and conceptual modeling to a mathematical model and computational algorithms is described and discussed. The multi-level biological observations provide the components and their connections of the conceptual model. However, the true strength of systems biology must be found in the biological test of the predictions that result from an experimental change in the computational model. These validated predictions will ultimately elucidate the functional role of a component or interaction in the process of heart development.


Assuntos
Ventrículos do Coração/embriologia , Modelos Cardiovasculares , Animais , Simulação por Computador , Humanos , Imageamento Tridimensional , Camundongos , Reprodutibilidade dos Testes , Biologia de Sistemas
18.
Cold Spring Harb Perspect Med ; 4(9): a013888, 2014 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-25183852

RESUMO

Myocardial cells ensure the contractility of the heart, which also depends on other mesodermal cell types for its function. Embryological experiments had identified the sources of cardiac precursor cells. With the advent of genetic engineering, novel tools have been used to reconstruct the lineage tree of cardiac cells that contribute to different parts of the heart, map the development of cardiac regions, and characterize their genetic signature. Such knowledge is of fundamental importance for our understanding of cardiogenesis and also for the diagnosis and treatment of heart malformations.


Assuntos
Linhagem da Célula , Cardiopatias Congênitas/embriologia , Coração/embriologia , Miócitos Cardíacos/citologia , Animais , Diferenciação Celular , Humanos
19.
Dev Dyn ; 242(6): 665-77, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23526457

RESUMO

BACKGROUND: The ventricular conduction system (VCS) coordinates the heartbeat and is composed of central components (the atrioventricular node, bundle, and right and left bundle branches) and a peripheral Purkinje fiber network. Conductive myocytes develop from common progenitor cells with working myocytes in a bimodal process of lineage restriction followed by limited outgrowth. The lineage relationship between progenitor cells giving rise to different components of the VCS is unclear. RESULTS: Cell lineage contributions to different components of the VCS were analysed by a combination of retrospective clonal analysis, regionalized transgene expression studies, and genetic tracing experiments using Connexin40-GFP mice that precisely delineate the VCS. Analysis of a library of hearts containing rare large clusters of clonally related myocytes identifies two VCS lineages encompassing either the right Purkinje fiber network or left bundle branch. Both lineages contribute to the atrioventricular bundle and right bundle branch that segregate early from working myocytes. Right and left VCS lineages share the transcriptional program of the respective ventricular working myocytes and genetic tracing experiments discount alternate progenitor cell contributions to the VCS. CONCLUSIONS: The mammalian VCS is comprised of cells derived from two lineages, supporting a dual contribution of first and second heart field progenitor cells.


Assuntos
Linhagem da Célula , Conexinas/genética , Sistema de Condução Cardíaco/embriologia , Ventrículos do Coração/embriologia , Células-Tronco/citologia , Alelos , Animais , Fascículo Atrioventricular/metabolismo , Conexinas/fisiologia , Feminino , Proteínas de Fluorescência Verde/metabolismo , Masculino , Camundongos , Microscopia de Fluorescência , Miocárdio/citologia , Miócitos Cardíacos/citologia , Fatores de Tempo , Transcrição Gênica , Transgenes
20.
Bioinformatics ; 29(6): 772-9, 2013 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-23337749

RESUMO

MOTIVATION: In developmental biology, quantitative tools to extract features from fluorescence microscopy images are becoming essential to characterize organ morphogenesis at the cellular level. However, automated image analysis in this context is a challenging task, owing to perturbations induced by the acquisition process, especially in organisms where the tissue is dense and opaque. RESULTS: We propose an automated framework for the segmentation of 3D microscopy images of highly cluttered environments such as developing tissues. The approach is based on a partial differential equation framework that jointly takes advantage of the nuclear and cellular membrane information to enable accurate extraction of nuclei and cells in dense tissues. This framework has been used to study the developing mouse heart, allowing the extraction of quantitative information such as the cell cycle duration; the method also provides qualitative information on cell division and cell polarity through the creation of 3D orientation maps that provide novel insight into tissue organization during organogenesis.


Assuntos
Imageamento Tridimensional/métodos , Microscopia de Fluorescência/métodos , Miocárdio/citologia , Animais , Ciclo Celular , Divisão Celular , Núcleo Celular/ultraestrutura , Centrossomo/ultraestrutura , Coração/embriologia , Camundongos , Miocárdio/ultraestrutura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...