Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 81
Filtrar
1.
Cell ; 186(12): 2610-2627.e18, 2023 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-37209682

RESUMO

The hourglass model describes the convergence of species within the same phylum to a similar body plan during development; however, the molecular mechanisms underlying this phenomenon in mammals remain poorly described. Here, we compare rabbit and mouse time-resolved differentiation trajectories to revisit this model at single-cell resolution. We modeled gastrulation dynamics using hundreds of embryos sampled between gestation days 6.0 and 8.5 and compared the species using a framework for time-resolved single-cell differentiation-flows analysis. We find convergence toward similar cell-state compositions at E7.5, supported by the quantitatively conserved expression of 76 transcription factors, despite divergence in surrounding trophoblast and hypoblast signaling. However, we observed noticeable changes in specification timing of some lineages and divergence of primordial germ cell programs, which in the rabbit do not activate mesoderm genes. Comparative analysis of temporal differentiation models provides a basis for studying the evolution of gastrulation dynamics across mammals.


Assuntos
Gastrulação , Mesoderma , Animais , Coelhos , Camundongos , Gastrulação/genética , Mesoderma/fisiologia , Diferenciação Celular/fisiologia , Mamíferos/genética , Trofoblastos , Regulação da Expressão Gênica no Desenvolvimento
2.
Cell ; 176(4): 844-855.e15, 2019 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-30712870

RESUMO

In developing organisms, spatially prescribed cell identities are thought to be determined by the expression levels of multiple genes. Quantitative tests of this idea, however, require a theoretical framework capable of exposing the rules and precision of cell specification over developmental time. We use the gap gene network in the early fly embryo as an example to show how expression levels of the four gap genes can be jointly decoded into an optimal specification of position with 1% accuracy. The decoder correctly predicts, with no free parameters, the dynamics of pair-rule expression patterns at different developmental time points and in various mutant backgrounds. Precise cellular identities are thus available at the earliest stages of development, contrasting the prevailing view of positional information being slowly refined across successive layers of the patterning network. Our results suggest that developmental enhancers closely approximate a mathematically optimal decoding strategy.


Assuntos
Proteínas Ativadoras de GTPase/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Redes Reguladoras de Genes/genética , Animais , Padronização Corporal/genética , Diferenciação Celular/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Embrião não Mamífero/metabolismo , Desenvolvimento Embrionário/genética , Proteínas Ativadoras de GTPase/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Modelos Genéticos , Fatores de Transcrição/metabolismo
3.
Annu Rev Cell Dev Biol ; 32: 47-75, 2016 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-27576120

RESUMO

Land plants can grow to tremendous body sizes, yet even the most complex architectures are the result of iterations of the same developmental processes: organ initiation, growth, and pattern formation. A central question in plant biology is how these processes are regulated and coordinated to allow for the formation of ordered, 3D structures. All these elementary processes first occur in early embryogenesis, during which, from a fertilized egg cell, precursors for all major tissues and stem cells are initiated, followed by tissue growth and patterning. Here we discuss recent progress in our understanding of this phase of plant life. We consider the cellular basis for multicellular development in 3D and focus on the genetic regulatory mechanisms that direct specific steps during early embryogenesis.


Assuntos
Morfogênese , Sementes/embriologia , Padronização Corporal , Nicho de Células-Tronco
4.
Dev Biol ; 511: 26-38, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38580174

RESUMO

In a developing embryo, formation of tissues and organs is remarkably precise in both time and space. Through cell-cell interactions, neighboring progenitors coordinate their activities, sequentially generating distinct types of cells. At present, we only have limited knowledge, rather than a systematic understanding, of the underlying logic and mechanisms responsible for cell fate transitions. The formation of the dorsal aspect of the spinal cord is an outstanding model to tackle these dynamics, as it first generates the peripheral nervous system and is later responsible for transmitting sensory information from the periphery to the brain and for coordinating local reflexes. This is reflected first by the ontogeny of neural crest cells, progenitors of the peripheral nervous system, followed by formation of the definitive roof plate of the central nervous system and specification of adjacent interneurons, then a transformation of roof plate into dorsal radial glia and ependyma lining the forming central canal. How do these peripheral and central neural branches segregate from common progenitors? How are dorsal radial glia established concomitant with transformation of the neural tube lumen into a central canal? How do the dorsal radial glia influence neighboring cells? This is only a partial list of questions whose clarification requires the implementation of experimental paradigms in which precise control of timing is crucial. Here, we outline some available answers and still open issues, while highlighting the contributions of avian models and their potential to address mechanisms of neural patterning and function.


Assuntos
Tubo Neural , Medula Espinal , Animais , Medula Espinal/embriologia , Tubo Neural/embriologia , Crista Neural/embriologia , Crista Neural/citologia , Crista Neural/fisiologia , Diferenciação Celular/fisiologia , Neuroglia/fisiologia , Células Neuroepiteliais/citologia , Células Neuroepiteliais/fisiologia , Humanos
5.
Development ; 149(12)2022 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-35758255

RESUMO

The architecture of gene regulatory networks determines the specificity and fidelity of developmental outcomes. We report that the core regulatory circuitry for endoderm development in Caenorhabditis elegans operates through a transcriptional cascade consisting of six sequentially expressed GATA-type factors that act in a recursive series of interlocked feedforward modules. This structure results in sequential redundancy, in which removal of a single factor or multiple alternate factors in the cascade leads to a mild or no effect on gut development, whereas elimination of any two sequential factors invariably causes a strong phenotype. The phenotypic strength is successfully predicted with a computational model based on the timing and levels of transcriptional states. We found that one factor in the middle of the cascade, END-1, which straddles the distinct events of specification and differentiation, functions in both processes. Finally, we reveal roles for key GATA factors in establishing spatial regulatory state domains by repressing other fates, thereby defining boundaries in the digestive tract. Our findings provide a paradigm that could account for the genetic redundancy observed in many developmental regulatory systems.


Assuntos
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Animais , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Endoderma/metabolismo , Fatores de Transcrição GATA/genética , Fatores de Transcrição GATA/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Lógica , Fatores de Transcrição/metabolismo
6.
Development ; 149(5)2022 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-35245348

RESUMO

The hypothalamus displays staggering cellular diversity, chiefly established during embryogenesis by the interplay of several signalling pathways and a battery of transcription factors. However, the contribution of epigenetic cues to hypothalamus development remains unclear. We mutated the polycomb repressor complex 2 gene Eed in the developing mouse hypothalamus, which resulted in the loss of H3K27me3, a fundamental epigenetic repressor mark. This triggered ectopic expression of posteriorly expressed regulators (e.g. Hox homeotic genes), upregulation of cell cycle inhibitors and reduced proliferation. Surprisingly, despite these effects, single cell transcriptomic analysis revealed that most neuronal subtypes were still generated in Eed mutants. However, we observed an increase in glutamatergic/GABAergic double-positive cells, as well as loss/reduction of dopamine, hypocretin and Tac2-Pax6 neurons. These findings indicate that many aspects of the hypothalamic gene regulatory flow can proceed without the key H3K27me3 epigenetic repressor mark, but points to a unique sensitivity of particular neuronal subtypes to a disrupted epigenomic landscape.


Assuntos
Desenvolvimento Embrionário/fisiologia , Hipotálamo/fisiologia , Neurônios/fisiologia , Complexo Repressor Polycomb 2/genética , Proteínas do Grupo Polycomb/genética , Animais , Proliferação de Células/genética , Repressão Epigenética/genética , Feminino , Masculino , Camundongos , Mutação/genética , Transcriptoma/genética
7.
Annu Rev Physiol ; 83: 17-37, 2021 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-33035429

RESUMO

Embryonic definitive hematopoiesis generates hematopoietic stem and progenitor cells (HSPCs) essential for establishment and maintenance of the adult blood system. This process requires the specification of a subset of vascular endothelial cells to become blood-forming, or hemogenic, and the subsequent endothelial-to-hematopoietic transition to generate HSPCs therefrom. The mechanisms that regulate these processes are under intensive investigation, as their recapitulation in vitro from human pluripotent stem cells has the potential to generate autologous HSPCs for clinical applications. In this review, we provide an overview of hemogenic endothelial cell development and highlight the molecular events that govern hemogenic specification of vascular endothelial cells and the generation of multilineage HSPCs from hemogenic endothelium. We also discuss the impact of hemogenic endothelial cell development on adult hematopoiesis.


Assuntos
Células Endoteliais/fisiologia , Endotélio Vascular/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Animais , Diferenciação Celular/fisiologia , Hemangioblastos/fisiologia , Hematopoese/fisiologia , Humanos
8.
J Biol Chem ; 299(8): 104964, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37364687

RESUMO

The epithelium of the pulmonary airway is composed of several distinct cell types that differentiate from common progenitor cells to provide defense against environmental insults. Epigenetic mechanisms regulating lineage differentiation of airway epithelial progenitors remain poorly understood. Protein arginine methyltransferase 5 (Prmt5) is a predominant type II arginine methyltransferase that methylates >85% of symmetric arginine residues. Here, we provide evidence for the function of Prmt5 in promoting ciliated cell fate specification of airway epithelial progenitors. We show that lung epithelial-specific deletion of Prmt5 resulted in a complete loss of ciliated cells, an increased number of basal cells, and ecotopic-expressed Tp63-Krt5+ putative cells in the proximal airway. We further identified that transcription factor Tp63 is a direct target of Prmt5, and Prmt5 inhibited Tp63 transcription expression through H4R3 symmetric dimethylation (H4R3sme2). Moreover, inhibition of Tp63 expression in Prmt5-deficient tracheal progenitors could partially restore the ciliated cell deficient phenotype. Together, our data support a model where Prmt5-mediated H4R3sme2 represses Tp63 expression to promote ciliated cell fate specification of airway progenitors.


Assuntos
Regulação da Expressão Gênica , Fatores de Transcrição , Animais , Humanos , Camundongos , Diferenciação Celular , Linhagem Celular Tumoral , Pulmão/metabolismo , Proteína-Arginina N-Metiltransferases/genética , Proteína-Arginina N-Metiltransferases/metabolismo , Fatores de Transcrição/metabolismo , Camundongos Endogâmicos C57BL
9.
Development ; 148(14)2021 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-34137447

RESUMO

Arabidopsis VASCULATURE COMPLEXITY AND CONNECTIVITY (VCC) is a plant-specific transmembrane protein that controls the development of veins in cotyledons. Here, we show that the expression and localization of the auxin efflux carrier PIN-FORMED1 (PIN1) is altered in vcc developing cotyledons and that overexpression of PIN1-GFP partially rescues vascular defects of vcc in a dosage-dependent manner. Genetic analyses suggest that VCC and PINOID (PID), a kinase that regulates PIN1 polarity, are both required for PIN1-mediated control of vasculature development. VCC expression is upregulated by auxin, likely as part of a positive feedback loop for the progression of vascular development. VCC and PIN1 localized to the plasma membrane in pre-procambial cells but were actively redirected to vacuoles in procambial cells for degradation. In the vcc mutant, PIN1 failed to properly polarize in pre-procambial cells during the formation of basal strands, and instead, it was prematurely degraded in vacuoles. VCC plays a role in the localization and stability of PIN1, which is crucial for the transition of pre-procambial cells into procambial cells that are involved in the formation of basal lateral strands in embryonic cotyledons.


Assuntos
Proteínas de Arabidopsis/metabolismo , Arabidopsis/embriologia , Arabidopsis/genética , Desenvolvimento Embrionário , Regulação da Expressão Gênica de Plantas , Proteínas de Arabidopsis/genética , Transporte Biológico , Cotilédone/citologia , Cotilédone/genética , Cotilédone/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/metabolismo , Mutação , Folhas de Planta/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo
10.
J Exp Bot ; 2024 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-38642102

RESUMO

The development of the embryo sac is an important factor affecting seed setting in rice. Numerous genes associated with embryo sac (ES) development have been identified in plants. However, the function of the DEAD-box RNA helicase family genes on ES is poorly known in rice. Here, we characterized a rice DEAD-box protein, OsRH52A, which was localized in the nucleus and cytoplasm and highly expressed in the floral organs in rice. The knockout mutant, rh52a, displayed partial ES sterility, including degenerated ES (21.0%) and the presence of double-female-gametophyte (DFG) structure (11.8%). The DFG developed from two functional megaspores (FM) near the chalazal end in one ovule, and 3.4% of DFG could fertilize via the sac near the micropylar pole in rh52a. OsRH52A was found to interact with OsMFS1 and ZIP4, both of which play a role in homologous recombination in rice meiosis. RNA-seq identified 234 down-regulated differentially expressed genes (DEGs) associated with reproductive development, including the two genes, OsMSP1 and HSA1b, required for female germline cell specification. Taken together, our study demonstrated that OsRH52A is essential for the development of the embryo sac and provided cytological evidence regarding the formation of DFG.

11.
Development ; 147(12)2020 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-32467243

RESUMO

Retinoic acid (RA) signaling is essential for multiple developmental processes, including appropriate pancreas formation from the foregut endoderm. RA is also required to generate pancreatic progenitors from human pluripotent stem cells. However, the role of RA signaling during endocrine specification has not been fully explored. In this study, we demonstrate that the disruption of RA signaling within the NEUROG3-expressing endocrine progenitor population impairs mouse ß cell differentiation and induces ectopic expression of crucial δ cell genes, including somatostatin. In addition, the inhibition of the RA pathway in hESC-derived pancreatic progenitors downstream of NEUROG3 induction impairs insulin expression. We further determine that RA-mediated regulation of endocrine cell differentiation occurs through Wnt pathway components. Together, these data demonstrate the importance of RA signaling in endocrine specification and identify conserved mechanisms by which RA signaling directs pancreatic endocrine cell fate.


Assuntos
Células Secretoras de Insulina/metabolismo , Pâncreas/metabolismo , Transdução de Sinais , Tretinoína/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/deficiência , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Diferenciação Celular , Embrião de Mamíferos/metabolismo , Proteínas de Homeodomínio/genética , Células-Tronco Embrionárias Humanas/citologia , Células-Tronco Embrionárias Humanas/metabolismo , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas do Tecido Nervoso/deficiência , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Pâncreas/citologia , Receptores do Ácido Retinoico/deficiência , Receptores do Ácido Retinoico/genética , Somatostatina/genética , Somatostatina/metabolismo , Células Secretoras de Somatostatina/citologia , Células Secretoras de Somatostatina/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Transativadores/deficiência , Transativadores/genética , Proteínas Wnt/metabolismo
12.
Cell Tissue Res ; 391(2): 269-286, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36512054

RESUMO

The central nervous system contains a daunting number of different cell types. Understanding how each cell acquires its fate remains a major challenge for neurobiology. The developing embryonic ventral nerve cord (VNC) of Drosophila melanogaster has been a powerful model system for unraveling the basic principles of cell fate specification. This pertains specifically to neuropeptide neurons, which typically are stereotypically generated in discrete subsets, allowing for unambiguous single-cell resolution in different genetic contexts. Here, we study the specification of the OrcoA-LA neurons, characterized by the expression of the neuropeptide Orcokinin A and located laterally in the A1-A5 abdominal segments of the VNC. We identified the progenitor neuroblast (NB; NB5-3) and the temporal window (castor/grainyhead) that generate the OrcoA-LA neurons. We also describe the role of the Ubx, abd-A, and Abd-B Hox genes in the segment-specific generation of these neurons. Additionally, our results indicate that the OrcoA-LA neurons are "Notch Off" cells, and neither programmed cell death nor the BMP pathway appears to be involved in their specification. Finally, we performed a targeted genetic screen of 485 genes known to be expressed in the CNS and identified nab, vg, and tsh as crucial determinists for OrcoA-LA neurons. This work provides a new neuropeptidergic model that will allow for addressing new questions related to neuronal specification mechanisms in the future.


Assuntos
Proteínas de Drosophila , Neuropeptídeos , Animais , Drosophila , Drosophila melanogaster/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Neurônios/metabolismo , Neuropeptídeos/genética , Neuropeptídeos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Fatores de Transcrição/metabolismo , Proteínas de Homeodomínio/metabolismo , Proteínas Repressoras
13.
Cell Mol Life Sci ; 79(10): 522, 2022 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-36114905

RESUMO

The transcription factor SOX9 is essential for the development of multiple organs including bone, testis, heart, lung, pancreas, intestine and nervous system. Mutations in the human SOX9 gene led to campomelic dysplasia, a haploinsufficiency disorder with several skeletal malformations frequently accompanied by 46, XY sex reversal. The mechanisms underlying the diverse SOX9 functions during organ development including its post-translational modifications, the availability of binding partners, and tissue-specific accessibility to target gene chromatin. Here we summarize the expression, activities, and downstream target genes of SOX9 in molecular genetic pathways essential for organ development, maintenance, and function. We also provide an insight into understanding the mechanisms that regulate the versatile roles of SOX9 in different organs.


Assuntos
Organogênese , Fatores de Transcrição SOX9/metabolismo , Displasia Campomélica , Cromatina , Transtornos do Desenvolvimento Sexual/genética , Humanos , Masculino , Mutação , Fatores de Transcrição SOX9/genética
14.
Proc Natl Acad Sci U S A ; 117(11): 6231-6236, 2020 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-32132210

RESUMO

Double fertilization is a key innovation for the evolutionary success of angiosperms by which the two fertilized female gametes, the egg cell and central cell, generate the embryo and endosperm, respectively. The female gametophyte (embryo sac) enclosed in the sporophyte is derived from a one-celled haploid cell lineage. It undergoes successive events of mitotic divisions, cellularization, and cell specification to give rise to the mature embryo sac, which contains the two female gametes accompanied by two types of accessory cells, namely synergids and antipodals. How the cell fate of the central cell is specified has long been equivocal and is further complicated by the structural diversity of female gametophyte across plant taxa. Here, MADS-box protein AGL80 was verified as a transcriptional repressor that directly suppresses the expression of accessory cell-specific genes to specify the central cell. Further genetic rescue and phylogenetic assay of the AGL80 orthologs revealed a possible conserved mechanism in the Brassicaceae family. Results from this study provide insight into the molecular determination of the second female gamete cell in Brassicaceae.


Assuntos
Proteína AGAMOUS de Arabidopsis/metabolismo , Arabidopsis/fisiologia , Regulação da Expressão Gênica de Plantas/fisiologia , Óvulo Vegetal/genética , Transcrição Gênica , Proteína AGAMOUS de Arabidopsis/genética , Proteínas de Arabidopsis/genética , Endosperma/metabolismo , Fertilização/genética , Mutação , Filogenia , Plantas Geneticamente Modificadas , Fatores de Transcrição/genética
15.
Plant J ; 103(5): 1869-1884, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32530511

RESUMO

Cereal endosperm represents the most important source of the world's food; nevertheless, the molecular mechanisms underlying cell and tissue differentiation in cereal grains remain poorly understood. Endosperm cellularization commences at the maternal-filial intersection of grains and generates endosperm transfer cells (ETCs), a cell type with a prominent anatomy optimized for efficient nutrient transport. Barley HISTIDINE KINASE1 (HvHK1) was identified as a receptor component with spatially restricted expression in the syncytial endosperm where ETCs emerge. Here, we demonstrate its function in ETC fate acquisition using RNA interference-mediated downregulation of HvHK1. Repression of HvHK1 impairs cell specification in the central ETC region and the development of transfer cell morphology, and consecutively defects differentiation of adjacent endosperm tissues. Coinciding with reduced expression of HvHK1, disturbed cell plate formation and fusion were observed at the initiation of endosperm cellularization, revealing that HvHK1 triggers initial cytokinesis of ETCs. Cell-type-specific RNA sequencing confirmed loss of transfer cell identity, compromised cell wall biogenesis and reduced transport capacities in aberrant cells and elucidated two-component signaling and hormone pathways that are mediated by HvHK1. Gene regulatory network modeling was used to specify the direct targets of HvHK1; this predicted non-canonical auxin signaling elements as the main regulatory links governing cellularization of ETCs, potentially through interaction with type-B response regulators. This work provides clues to previously unknown molecular mechanisms directing ETC specification, a process with fundamental impact on grain yield in cereals.


Assuntos
Diferenciação Celular , Endosperma/crescimento & desenvolvimento , Histidina Quinase/metabolismo , Hordeum/metabolismo , Proteínas de Plantas/metabolismo , Divisão Celular , Polaridade Celular , Grão Comestível/crescimento & desenvolvimento , Grão Comestível/metabolismo , Endosperma/metabolismo , Regulação da Expressão Gênica de Plantas , Histidina Quinase/fisiologia , Hordeum/enzimologia , Hordeum/crescimento & desenvolvimento , Proteínas de Plantas/fisiologia
16.
Angiogenesis ; 24(2): 213-236, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33844116

RESUMO

There are two vascular networks in mammals that coordinately function as the main supply and drainage systems of the body. The blood vasculature carries oxygen, nutrients, circulating cells, and soluble factors to and from every tissue. The lymphatic vasculature maintains interstitial fluid homeostasis, transports hematopoietic cells for immune surveillance, and absorbs fat from the gastrointestinal tract. These vascular systems consist of highly organized networks of specialized vessels including arteries, veins, capillaries, and lymphatic vessels that exhibit different structures and cellular composition enabling distinct functions. All vessels are composed of an inner layer of endothelial cells that are in direct contact with the circulating fluid; therefore, they are the first responders to circulating factors. However, endothelial cells are not homogenous; rather, they are a heterogenous population of specialized cells perfectly designed for the physiological demands of the vessel they constitute. This review provides an overview of the current knowledge of the specification of arterial, venous, capillary, and lymphatic endothelial cell identities during vascular development. We also discuss how the dysregulation of these processes can lead to vascular malformations, and therapeutic approaches that have been developed for their treatment.


Assuntos
Vasos Sanguíneos/metabolismo , Células Endoteliais/metabolismo , Vasos Linfáticos/metabolismo , Malformações Vasculares/metabolismo , Animais , Vasos Sanguíneos/patologia , Células Endoteliais/patologia , Humanos , Vasos Linfáticos/patologia , Malformações Vasculares/patologia
17.
Biochem Biophys Res Commun ; 551: 121-126, 2021 04 30.
Artigo em Inglês | MEDLINE | ID: mdl-33725573

RESUMO

We have investigated the mechanism of action of SWITCH1/DYAD (SWI1), an important regulator of plant meiosis in Arabidopsis that is required for meiotic chromosome organization including maintenance of sister chromatid cohesion. The central portion of SWI1 contains a domain of unknown function that shows strong conservation between SWI1 and its orthologs in maize and rice and is also found in paralogs including MALE MEIOCYTE DEATH 1 (MMD1). In order to examine the role of this domain we performed domain swap experiments into SWI1 in a swi1 mutant background. Domain swap analysis revealed functional conservation of the central domain between SWI1 and its orthologs but not with the domain from MMD1 suggesting that the domain plays an important role in SWI1 function that has been conserved in orthologs and diverged in paralogs in plant evolution. Analysis of expression of the non-complementing MMD1 domain swap SWI1(DSMMD1)::GFP transgenic lines revealed an altered pattern of expression that suggests a role for SWI1 in commitment to female meiocyte differentiation and meiosis. The results suggest that SWI1 may also play a developmental role as an identity determinant in the female germ cell lineage in addition to its known role in meiotic chromosome organization.


Assuntos
Proteínas de Arabidopsis/metabolismo , Arabidopsis/citologia , Proteínas de Ciclo Celular/metabolismo , Diferenciação Celular , Expressão Ectópica do Gene , Meiose , Proteínas Nucleares/metabolismo , Óvulo Vegetal/citologia , Domínios Proteicos , Arabidopsis/genética , Arabidopsis/metabolismo , Proteínas de Arabidopsis/química , Proteínas de Arabidopsis/genética , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Linhagem da Célula , Regulação da Expressão Gênica de Plantas , Proteínas Nucleares/química , Proteínas Nucleares/genética , Oryza/genética , Domínios Proteicos/genética , Alinhamento de Sequência , Zea mays/genética
18.
Development ; 145(21)2018 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-30337377

RESUMO

Patterning of a continuously growing naive field in the context of a life-long growing organ such as the teleost eye is of high functional relevance. Intrinsic and extrinsic signals have been proposed to regulate lineage specification in progenitors that exit the stem cell niche in the ciliary marginal zone (CMZ). The proper cell-type composition arising from those progenitors is a prerequisite for retinal function. Our findings in the teleost medaka (Oryzias latipes) uncover that the Notch-Atoh7 axis continuously patterns the CMZ. The complement of cell types originating from the two juxtaposed progenitors marked by Notch or Atoh7 activity contains all constituents of a retinal column. Modulation of Notch signalling specifically in Atoh7-expressing cells demonstrates the crucial role of this axis in generating the correct cell-type proportions. After transiently blocking Notch signalling, retinal patterning and differentiation is re-initiated de novo Taken together, our data show that Notch activity in the CMZ continuously structures the growing retina by juxtaposing Notch and Atoh7 progenitors that give rise to distinct complementary lineages, revealing coupling of de novo patterning and cell-type specification in the respective lineages.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Oryzias/crescimento & desenvolvimento , Oryzias/metabolismo , Receptores Notch/metabolismo , Retina/crescimento & desenvolvimento , Retina/metabolismo , Transdução de Sinais , Células Amácrinas/citologia , Células Amácrinas/metabolismo , Animais , Contagem de Células , Linhagem da Célula , Embrião não Mamífero/metabolismo , Células Ependimogliais/citologia , Células Ependimogliais/metabolismo , Modelos Biológicos , Retina/citologia , Células Bipolares da Retina/citologia , Células Bipolares da Retina/metabolismo , Células-Tronco/metabolismo
19.
Int J Mol Sci ; 22(6)2021 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-33802099

RESUMO

Blood vessels are essential for the formation and maintenance of almost all functional tissues. They play fundamental roles in the supply of oxygen and nutrition, as well as development and morphogenesis. Vascular endothelial cells are the main factor in blood vessel formation. Recently, research findings showed heterogeneity in vascular endothelial cells in different tissue/organs. Endothelial cells alter their gene expressions depending on their cell fate or angiogenic states of vascular development in normal and pathological processes. Studies on gene regulation in endothelial cells demonstrated that the activator protein 1 (AP-1) transcription factors are implicated in angiogenesis and vascular development. In particular, it has been revealed that JunB (a member of the AP-1 transcription factor family) is transiently induced in endothelial cells at the angiogenic frontier and controls them on tip cells specification during vascular development. Moreover, JunB plays a role in tissue-specific vascular maturation processes during neurovascular interaction in mouse embryonic skin and retina vasculatures. Thus, JunB appears to be a new angiogenic factor that induces endothelial cell migration and sprouting particularly in neurovascular interaction during vascular development. In this review, we discuss the recently identified role of JunB in endothelial cells and blood vessel formation.


Assuntos
Indutores da Angiogênese/metabolismo , Movimento Celular , Células Endoteliais/metabolismo , Neovascularização Fisiológica , Fator de Transcrição AP-1/metabolismo , Fatores de Transcrição/metabolismo , Animais , Humanos
20.
Am J Physiol Gastrointest Liver Physiol ; 318(4): G803-G815, 2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-32116021

RESUMO

Liver sinusoidal endothelial cells (LSECs) are the first liver cells to encounter waste macromolecules, pathogens, and toxins in blood. LSECs are highly specialized to mediate the clearance of these substances via endocytic scavenger receptors and are equipped with fenestrae that mediate the passage of macromolecules toward hepatocytes. Although some transcription factors (TFs) are known to play a role in LSEC specialization, information about the specialized LSEC signature and its transcriptional determinants remains incomplete.Based on a comparison of liver, heart, and brain endothelial cells (ECs), we established a 30-gene LSEC signature comprising both established and newly identified markers, including 7 genes encoding TFs. To evaluate the LSEC TF regulatory network, we artificially increased the expression of the 7 LSEC-specific TFs in human umbilical vein ECs. Although Zinc finger E-box-binding protein 2, homeobox B5, Cut-like homolog 2, and transcription factor EC (TCFEC) had limited contributions, musculoaponeurotic fibrosarcoma (C-MAF), GATA binding protein 4 (GATA4), and MEIS homeobox 2 (MEIS2) emerged as stronger inducers of LSEC marker expression. Furthermore, a combination of C-MAF, GATA4, and MEIS2 showed a synergistic effect on the increase of LSEC signature genes, including liver/lymph node-specific ICAM-3 grabbing non-integrin (L-SIGN) (or C-type lectin domain family member M (CLEC4M)), mannose receptor C-Type 1 (MRC1), legumain (LGMN), G protein-coupled receptor 182 (GPR182), Plexin C1 (PLXNC1), and solute carrier organic anion transporter family member 2A1 (SLCO2A1). Accordingly, L-SIGN, MRC1, pro-LGMN, GPR182, PLXNC1, and SLCO2A1 protein levels were elevated by this combined overexpression. Although receptor-mediated endocytosis was not significantly induced by the triple TF combination, it enhanced binding to E2, the hepatitis C virus host-binding protein. We conclude that C-MAF, GATA4, and MEIS2 are important transcriptional regulators of the unique LSEC fingerprint and LSEC interaction with viruses. Additional factors are however required to fully recapitulate the molecular, morphological, and functional LSEC fingerprint.NEW & NOTEWORTHY Liver sinusoidal endothelial cells (LSECs) are the first liver cells to encounter waste macromolecules, pathogens, and toxins in the blood and are highly specialized. Although some transcription factors are known to play a role in LSEC specialization, information about the specialized LSEC signature and its transcriptional determinants remains incomplete. Here, we show that Musculoaponeurotic Fibrosarcoma (C-MAF), GATA binding protein 4 (GATA4), and Meis homeobox 2 (MEIS2) are important transcriptional regulators of the unique LSEC signature and that they affect the interaction of LSECs with viruses.


Assuntos
Células Endoteliais/fisiologia , Regulação da Expressão Gênica/fisiologia , Fígado/citologia , Animais , Marcadores Genéticos , Humanos , Fígado/metabolismo , Masculino , Especificidade de Órgãos , Ratos , Transcriptoma
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA