Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 16.544
Filtrar
1.
Curr Top Dev Biol ; 159: 232-271, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38729677

RESUMO

The anterior-to-posterior (head-to-tail) body axis is extraordinarily diverse among vertebrates but conserved within species. Body axis development requires a population of axial progenitors that resides at the posterior of the embryo to sustain elongation and is then eliminated once axis extension is complete. These progenitors occupy distinct domains in the posterior (tail-end) of the embryo and contribute to various lineages along the body axis. The subset of axial progenitors with neuromesodermal competency will generate both the neural tube (the precursor of the spinal cord), and the trunk and tail somites (producing the musculoskeleton) during embryo development. These axial progenitors are called Neuromesodermal Competent cells (NMCs) and Neuromesodermal Progenitors (NMPs). NMCs/NMPs have recently attracted interest beyond the field of developmental biology due to their clinical potential. In the mouse, the maintenance of neuromesodermal competency relies on a fine balance between a trio of known signals: Wnt/ß-catenin, FGF signalling activity and suppression of retinoic acid signalling. These signals regulate the relative expression levels of the mesodermal transcription factor Brachyury and the neural transcription factor Sox2, permitting the maintenance of progenitor identity when co-expressed, and either mesoderm or neural lineage commitment when the balance is tilted towards either Brachyury or Sox2, respectively. Despite important advances in understanding key genes and cellular behaviours involved in these fate decisions, how the balance between mesodermal and neural fates is achieved remains largely unknown. In this chapter, we provide an overview of signalling and gene regulatory networks in NMCs/NMPs. We discuss mutant phenotypes associated with axial defects, hinting at the potential significant role of lesser studied proteins in the maintenance and differentiation of the progenitors that fuel axial elongation.


Assuntos
Padronização Corporal , Mesoderma , Animais , Padronização Corporal/genética , Mesoderma/metabolismo , Mesoderma/citologia , Mesoderma/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Transdução de Sinais , Proteínas com Domínio T/metabolismo , Proteínas com Domínio T/genética , Diferenciação Celular , Cabeça/embriologia
2.
Curr Top Dev Biol ; 159: 372-405, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38729682

RESUMO

The Segmentation Clock is a tissue-level patterning system that enables the segmentation of the vertebral column precursors into transient multicellular blocks called somites. This patterning system comprises a set of elements that are essential for correct segmentation. Under the so-called "Clock and Wavefront" model, the system consists of two elements, a genetic oscillator that manifests itself as traveling waves of gene expression, and a regressing wavefront that transforms the temporally periodic signal encoded in the oscillations into a permanent spatially periodic pattern of somite boundaries. Over the last twenty years, every new discovery about the Segmentation Clock has been tightly linked to the nomenclature of the "Clock and Wavefront" model. This constrained allocation of discoveries into these two elements has generated long-standing debates in the field as what defines molecularly the wavefront and how and where the interaction between the two elements establishes the future somite boundaries. In this review, we propose an expansion of the "Clock and Wavefront" model into three elements, "Clock", "Wavefront" and signaling gradients. We first provide a detailed description of the components and regulatory mechanisms of each element, and we then examine how the spatiotemporal integration of the three elements leads to the establishment of the presumptive somite boundaries. To be as exhaustive as possible, we focus on the Segmentation Clock in zebrafish. Furthermore, we show how this three-element expansion of the model provides a better understanding of the somite formation process and we emphasize where our current understanding of this patterning system remains obscure.


Assuntos
Padronização Corporal , Regulação da Expressão Gênica no Desenvolvimento , Mesoderma , Somitos , Animais , Padronização Corporal/genética , Somitos/embriologia , Somitos/metabolismo , Mesoderma/embriologia , Mesoderma/metabolismo , Mesoderma/citologia , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Transdução de Sinais , Relógios Biológicos/genética
3.
Development ; 151(10)2024 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-38757779

RESUMO

Collective migration of caudal visceral mesoderm (CVM) cells in Drosophila embryos helps form the longitudinal muscles of the larval gut. In their study, Angelike Stathopoulos and colleagues reveal that cell division coordinates two gene expression programmes in migrating CVM cells. To know more about their work, we spoke to the first author, Jingjing Sun, and the corresponding author, Angelike Stathopoulos, Professor in the Division of Biology at the California Institute of Technology, USA.


Assuntos
Biologia do Desenvolvimento , Animais , Biologia do Desenvolvimento/história , História do Século XX , História do Século XXI , Mesoderma/metabolismo , Drosophila/embriologia , Movimento Celular , Humanos
4.
Cell Syst ; 15(5): 445-461.e4, 2024 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-38692274

RESUMO

BMP signaling is essential for mammalian gastrulation, as it initiates a cascade of signals that control self-organized patterning. As development is highly dynamic, it is crucial to understand how time-dependent combinatorial signaling affects cellular differentiation. Here, we show that BMP signaling duration is a crucial control parameter that determines cell fates upon the exit from pluripotency through its interplay with the induced secondary signal WNT. BMP signaling directly converts cells from pluripotent to extraembryonic fates while simultaneously upregulating Wnt signaling, which promotes primitive streak and mesodermal specification. Using live-cell imaging of signaling and cell fate reporters together with a simple mathematical model, we show that this circuit produces a temporal morphogen effect where, once BMP signal duration is above a threshold for differentiation, intermediate and long pulses of BMP signaling produce specification of mesoderm and extraembryonic fates, respectively. Our results provide a systems-level picture of how these signaling pathways control the landscape of early human development.


Assuntos
Proteínas Morfogenéticas Ósseas , Diferenciação Celular , Linha Primitiva , Transdução de Sinais , Linha Primitiva/metabolismo , Linha Primitiva/embriologia , Proteínas Morfogenéticas Ósseas/metabolismo , Humanos , Transdução de Sinais/fisiologia , Animais , Mesoderma/metabolismo , Mesoderma/embriologia , Via de Sinalização Wnt/fisiologia , Proteínas Wnt/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Gastrulação/fisiologia
5.
Development ; 151(10)2024 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-38742434

RESUMO

During mouse development, presomitic mesoderm cells synchronize Wnt and Notch oscillations, creating sequential phase waves that pattern somites. Traditional somitogenesis models attribute phase waves to a global modulation of the oscillation frequency. However, increasing evidence suggests that they could arise in a self-organizing manner. Here, we introduce the Sevilletor, a novel reaction-diffusion system that serves as a framework to compare different somitogenesis patterning hypotheses. Using this framework, we propose the Clock and Wavefront Self-Organizing model that considers an excitable self-organizing region where phase waves form independent of global frequency gradients. The model recapitulates the change in relative phase of Wnt and Notch observed during mouse somitogenesis and provides a theoretical basis for understanding the excitability of mouse presomitic mesoderm cells in vitro.


Assuntos
Receptores Notch , Somitos , Animais , Camundongos , Somitos/embriologia , Somitos/metabolismo , Receptores Notch/metabolismo , Receptores Notch/genética , Mesoderma/embriologia , Mesoderma/metabolismo , Modelos Biológicos , Padronização Corporal/genética , Proteínas Wnt/metabolismo , Proteínas Wnt/genética , Desenvolvimento Embrionário/genética , Desenvolvimento Embrionário/fisiologia , Relógios Biológicos/fisiologia
6.
Artigo em Chinês | MEDLINE | ID: mdl-38563180

RESUMO

Cartilage mesenchyme hamartoma originates from the mesoderm and contains a blend of interstitium and cartilage, which is mostly benign tumor and is a non-neoplastic cartilage lesion with self-limiting hyperplasia. This article reports a infant with cervical chondromesenchymal hamartoma in the neck, the main clinical manifestations of which are asphyxia and acute respiratory distress, and the imaging features are often similar to those of malignant tumors.Radical resection operation under general anesthesia is the main treatment method, and the postoperative pathological diagnosis was cartilage mesenchyme, and immunohistochemistry showed Catenin(-),MDM2(+),CDK4(-),H3K36M(+),Myogenin (-),SMA (-).The clinical characteristics and diagnosis and treatment process of this case are reported and related literature is reviewed.


Assuntos
Cartilagem , Hamartoma , Humanos , Recém-Nascido , Imuno-Histoquímica , Mesoderma/patologia
7.
FASEB J ; 38(9): e23632, 2024 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-38686936

RESUMO

The upper Müllerian duct (MD) is patterned and specified into two morphologically and functionally distinct organs, the oviduct and uterus. It is known that this regionalization process is instructed by inductive signals from the adjacent mesenchyme. However, the interaction landscape between epithelium and mesenchyme during upper MD development remains largely unknown. Here, we performed single-cell transcriptomic profiling of mouse neonatal oviducts and uteri at the initiation of MD epithelial differentiation (postnatal day 3). We identified major cell types including epithelium, mesenchyme, pericytes, mesothelium, endothelium, and immune cells in both organs with established markers. Moreover, we uncovered region-specific epithelial and mesenchymal subpopulations and then deduced region-specific ligand-receptor pairs mediating mesenchymal-epithelial interactions along the craniocaudal axis. Unexpectedly, we discovered a mesenchymal subpopulation marked by neurofilaments with specific localizations at the mesometrial pole of both the neonatal oviduct and uterus. Lastly, we analyzed and revealed organ-specific signature genes of pericytes and mesothelial cells. Taken together, our study enriches our knowledge of upper MD development, and provides a manageable list of potential genes, pathways, and region-specific cell subtypes for future functional studies.


Assuntos
Ductos Paramesonéfricos , Oviductos , Análise de Célula Única , Transcriptoma , Útero , Animais , Feminino , Camundongos , Útero/metabolismo , Útero/citologia , Ductos Paramesonéfricos/metabolismo , Oviductos/metabolismo , Oviductos/citologia , Perfilação da Expressão Gênica , Animais Recém-Nascidos , Diferenciação Celular , Mesoderma/metabolismo , Mesoderma/citologia , Células Epiteliais/metabolismo , Camundongos Endogâmicos C57BL , Regulação da Expressão Gênica no Desenvolvimento
8.
PLoS Biol ; 22(4): e3002590, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38683849

RESUMO

Brain pericytes are one of the critical cell types that regulate endothelial barrier function and activity, thus ensuring adequate blood flow to the brain. The genetic pathways guiding undifferentiated cells into mature pericytes are not well understood. We show here that pericyte precursor populations from both neural crest and head mesoderm of zebrafish express the transcription factor nkx3.1 develop into brain pericytes. We identify the gene signature of these precursors and show that an nkx3.1-, foxf2a-, and cxcl12b-expressing pericyte precursor population is present around the basilar artery prior to artery formation and pericyte recruitment. The precursors later spread throughout the brain and differentiate to express canonical pericyte markers. Cxcl12b-Cxcr4 signaling is required for pericyte attachment and differentiation. Further, both nkx3.1 and cxcl12b are necessary and sufficient in regulating pericyte number as loss inhibits and gain increases pericyte number. Through genetic experiments, we have defined a precursor population for brain pericytes and identified genes critical for their differentiation.


Assuntos
Encéfalo , Diferenciação Celular , Pericitos , Fatores de Transcrição , Proteínas de Peixe-Zebra , Peixe-Zebra , Pericitos/metabolismo , Pericitos/citologia , Animais , Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Encéfalo/metabolismo , Encéfalo/embriologia , Proteínas de Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética , Fatores de Transcrição/metabolismo , Fatores de Transcrição/genética , Diferenciação Celular/genética , Proteínas de Homeodomínio/metabolismo , Proteínas de Homeodomínio/genética , Regulação da Expressão Gênica no Desenvolvimento , Crista Neural/metabolismo , Crista Neural/citologia , Mesoderma/metabolismo , Mesoderma/citologia , Transdução de Sinais , Receptores CXCR4/metabolismo , Receptores CXCR4/genética , Quimiocina CXCL12/metabolismo , Quimiocina CXCL12/genética
9.
PLoS Biol ; 22(4): e3002611, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38683880

RESUMO

As tissues grow and change shape during animal development, they physically pull and push on each other, and these mechanical interactions can be important for morphogenesis. During Drosophila gastrulation, mesoderm invagination temporally overlaps with the convergence and extension of the ectodermal germband; the latter is caused primarily by Myosin II-driven polarised cell intercalation. Here, we investigate the impact of mesoderm invagination on ectoderm extension, examining possible mechanical and mechanotransductive effects on Myosin II recruitment and polarised cell intercalation. We find that the germband ectoderm is deformed by the mesoderm pulling in the orthogonal direction to germband extension (GBE), showing mechanical coupling between these tissues. However, we do not find a significant change in Myosin II planar polarisation in response to mesoderm invagination, nor in the rate of junction shrinkage leading to neighbour exchange events. We conclude that the main cellular mechanism of axis extension, polarised cell intercalation, is robust to the mesoderm invagination pull. We find, however, that mesoderm invagination slows down the rate of anterior-posterior cell elongation that contributes to axis extension, counteracting the tension from the endoderm invagination, which pulls along the direction of GBE.


Assuntos
Drosophila melanogaster , Ectoderma , Gastrulação , Mesoderma , Miosina Tipo II , Animais , Mesoderma/embriologia , Mesoderma/citologia , Gastrulação/fisiologia , Ectoderma/citologia , Ectoderma/embriologia , Ectoderma/metabolismo , Miosina Tipo II/metabolismo , Drosophila melanogaster/embriologia , Polaridade Celular , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Embrião não Mamífero , Morfogênese , Padronização Corporal/fisiologia , Drosophila/embriologia
10.
Dev Cell ; 59(10): 1302-1316.e5, 2024 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-38569553

RESUMO

The planar cell polarity (PCP) complex is speculated to function in murine lung development, where branching morphogenesis generates an epithelial tree whose distal tips expand dramatically during sacculation. Here, we show that PCP is dispensable in the airway epithelium for sacculation. Rather, we find a Celsr1-independent role for the PCP component Vangl in the pulmonary mesenchyme: loss of Vangl1/2 inhibits mesenchymal thinning and expansion of the saccular epithelium. Further, loss of mesenchymal Wnt5a mimics sacculation defects observed in Vangl2-mutant lungs, implicating mesenchymal Wnt5a/Vangl signaling as a key regulator of late lung morphogenesis. A computational model predicts that sacculation requires a fluid mesenchymal compartment. Lineage-tracing and cell-shape analyses are consistent with the mesenchyme acting as a fluid tissue, suggesting that loss of Vangl1/2 impacts the ability of mesenchymal cells to exchange neighbors. Our data thus identify an explicit function for Vangl and the pulmonary mesenchyme in actively shaping the saccular epithelium.


Assuntos
Polaridade Celular , Pulmão , Mesoderma , Morfogênese , Proteínas do Tecido Nervoso , Animais , Mesoderma/metabolismo , Camundongos , Pulmão/metabolismo , Pulmão/patologia , Pulmão/embriologia , Proteínas do Tecido Nervoso/metabolismo , Proteínas do Tecido Nervoso/genética , Proteína Wnt-5a/metabolismo , Proteína Wnt-5a/genética , Proteínas de Membrana/metabolismo , Proteínas de Membrana/genética , Transdução de Sinais , Organogênese/genética , Receptores Acoplados a Proteínas G
11.
Stem Cell Reports ; 19(5): 618-628, 2024 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-38579708

RESUMO

SOX2 is a transcription factor involved in the regulatory network maintaining the pluripotency of embryonic stem cells in culture as well as in early embryos. In addition, SOX2 plays a pivotal role in neural stem cell formation and neurogenesis. How SOX2 can serve both processes has remained elusive. Here, we identified a set of SOX2-dependent neural-associated enhancers required for neural lineage priming. They form a distinct subgroup (1,898) among 8,531 OCT4/SOX2/NANOG-bound enhancers characterized by enhanced SOX2 binding and chromatin accessibility. Activation of these enhancers is triggered by neural induction of wild-type cells or by default in Smad4-ablated cells resistant to mesoderm induction and is antagonized by mesodermal transcription factors via Sox2 repression. Our data provide mechanistic insight into the transition from the pluripotency state to the early neural fate and into the regulation of early neural versus mesodermal specification in embryonic stem cells and embryos.


Assuntos
Elementos Facilitadores Genéticos , Mesoderma , Células-Tronco Neurais , Fatores de Transcrição SOXB1 , Fatores de Transcrição SOXB1/metabolismo , Fatores de Transcrição SOXB1/genética , Animais , Camundongos , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/citologia , Mesoderma/citologia , Mesoderma/metabolismo , Neurogênese , Regulação da Expressão Gênica no Desenvolvimento , Fator 3 de Transcrição de Octâmero/metabolismo , Fator 3 de Transcrição de Octâmero/genética , Diferenciação Celular/genética , Proteína Homeobox Nanog/metabolismo , Proteína Homeobox Nanog/genética , Linhagem da Célula/genética , Proteína Smad4/metabolismo , Proteína Smad4/genética , Células-Tronco Embrionárias/metabolismo , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias Murinas/metabolismo , Células-Tronco Embrionárias Murinas/citologia , Cromatina/metabolismo , Ligação Proteica
12.
Development ; 151(10)2024 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-38646822

RESUMO

The precise assembly of tissues and organs relies on spatiotemporal regulation of gene expression to coordinate the collective behavior of cells. In Drosophila embryos, the midgut musculature is formed through collective migration of caudal visceral mesoderm (CVM) cells, but how gene expression changes as cells migrate is not well understood. Here, we have focused on ten genes expressed in the CVM and the cis-regulatory sequences controlling their expression. Although some genes are continuously expressed, others are expressed only early or late during migration. Late expression relates to cell cycle progression, as driving string/Cdc25 causes earlier division of CVM cells and accelerates the transition to late gene expression. In particular, we found that the cell cycle effector transcription factor E2F1 is a required input for the late gene CG5080. Furthermore, whereas late genes are broadly expressed in all CVM cells, early gene transcripts are polarized to the anterior or posterior ends of the migrating collective. We show this polarization requires transcription factors Snail, Zfh1 and Dorsocross. Collectively, these results identify two sequential gene expression programs bridged by cell division that support long-distance directional migration of CVM cells.


Assuntos
Divisão Celular , Movimento Celular , Proteínas de Drosophila , Regulação da Expressão Gênica no Desenvolvimento , Animais , Movimento Celular/genética , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Divisão Celular/genética , Mesoderma/metabolismo , Mesoderma/citologia , Fatores de Transcrição/metabolismo , Fatores de Transcrição/genética , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Drosophila melanogaster/embriologia , Fator de Transcrição E2F1/metabolismo , Fator de Transcrição E2F1/genética , Embrião não Mamífero/metabolismo , Embrião não Mamífero/citologia , Drosophila/genética , Drosophila/metabolismo , Drosophila/embriologia , Fatores de Transcrição da Família Snail/metabolismo , Fatores de Transcrição da Família Snail/genética
13.
PLoS One ; 19(4): e0297853, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38635504

RESUMO

During vertebrate embryo development, the body is progressively segmented along the anterior-posterior (A-P) axis early in development. The rate of somite formation is controlled by the somitogenesis embryo clock (EC), which was first described as gene expression oscillations of hairy1 (hes4) in the presomitic mesoderm of chick embryos with 15-20 somites. Here, the EC displays the same periodicity as somite formation, 90 min, whereas the posterior-most somites (44-52) only arise every 150 minutes, matched by a corresponding slower pace of the EC. Evidence suggests that the rostral-most somites are formed faster, however, their periodicity and the EC expression dynamics in these early stages are unknown. In this study, we used time-lapse imaging of chicken embryos from primitive streak to somitogenesis stages with high temporal resolution (3-minute intervals). We measured the length between the anterior-most and the last formed somitic clefts in each captured frame and developed a simple algorithm to automatically infer both the length and time of formation of each somite. We found that the occipital somites (up to somite 5) form at an average rate of 75 minutes, while somites 6 onwards are formed approximately every 90 minutes. We also assessed the expression dynamics of hairy1 using half-embryo explants cultured for different periods of time. This showed that EC hairy1 expression is highly dynamic prior to somitogenesis and assumes a clear oscillatory behaviour as the first somites are formed. Importantly, using ex ovo culture and live-imaging techniques, we showed that the hairy1 expression pattern recapitulates with the formation of each new pair of somites, indicating that somite segmentation is coupled with EC oscillations since the onset of somitogenesis.


Assuntos
Proteínas Aviárias , Somitos , Animais , Embrião de Galinha , Galinhas , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Proteínas Aviárias/genética , Mesoderma/metabolismo
14.
Int J Oral Sci ; 16(1): 33, 2024 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-38654018

RESUMO

Precise orchestration of cell fate determination underlies the success of scaffold-based skeletal regeneration. Despite extensive studies on mineralized parenchymal tissue rebuilding, regenerating and maintaining undifferentiated mesenchyme within calvarial bone remain very challenging with limited advances yet. Current knowledge has evidenced the indispensability of rebuilding suture mesenchymal stem cell niches to avoid severe brain or even systematic damage. But to date, the absence of promising therapeutic biomaterials/scaffolds remains. The reason lies in the shortage of fundamental knowledge and methodological evidence to understand the cellular fate regulations of scaffolds. To address these issues, in this study, we systematically investigated the cellular fate determinations and transcriptomic mechanisms by distinct types of commonly used calvarial scaffolds. Our data elucidated the natural processes without scaffold transplantation and demonstrated how different scaffolds altered in vivo cellular responses. A feasible scaffold, polylactic acid electrospinning membrane (PLA), was next identified to precisely control mesenchymal ingrowth and self-renewal to rebuild non-osteogenic suture-like tissue at the defect center, meanwhile supporting proper osteointegration with defect bony edges. Especially, transcriptome analysis and cellular mechanisms underlying the well-orchestrated cell fate determination of PLA were deciphered. This study for the first time cellularly decoded the fate regulations of scaffolds in suture-bony composite defect healing, offering clinicians potential choices for regenerating such complicated injuries.


Assuntos
Regeneração Óssea , Alicerces Teciduais , Transcriptoma , Animais , Regeneração Óssea/fisiologia , Poliésteres , Crânio/cirurgia , Células-Tronco Mesenquimais , Mesoderma/citologia , Diferenciação Celular , Engenharia Tecidual/métodos , Suturas Cranianas , Materiais Biocompatíveis
15.
Development ; 151(8)2024 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-38602485

RESUMO

Alveologenesis, the final stage in lung development, substantially remodels the distal lung, expanding the alveolar surface area for efficient gas exchange. Secondary crest myofibroblasts (SCMF) exist transiently in the neonatal distal lung and are crucial for alveologenesis. However, the pathways that regulate SCMF function, proliferation and temporal identity remain poorly understood. To address this, we purified SCMFs from reporter mice, performed bulk RNA-seq and found dynamic changes in Hippo-signaling components during alveologenesis. We deleted the Hippo effectors Yap/Taz from Acta2-expressing cells at the onset of alveologenesis, causing a significant arrest in alveolar development. Using single cell RNA-seq, we identified a distinct cluster of cells in mutant lungs with altered expression of marker genes associated with proximal mesenchymal cell types, airway smooth muscle and alveolar duct myofibroblasts. In vitro studies confirmed that Yap/Taz regulates myofibroblast-associated gene signature and contractility. Together, our findings show that Yap/Taz is essential for maintaining functional myofibroblast identity during postnatal alveologenesis.


Assuntos
Diferenciação Celular , Via de Sinalização Hippo , Morfogênese , Miofibroblastos , Proteínas Serina-Treonina Quinases , Alvéolos Pulmonares , Transdução de Sinais , Proteínas de Sinalização YAP , Animais , Camundongos , Miofibroblastos/metabolismo , Miofibroblastos/citologia , Proteínas de Sinalização YAP/metabolismo , Proteínas de Sinalização YAP/genética , Alvéolos Pulmonares/metabolismo , Alvéolos Pulmonares/citologia , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Serina-Treonina Quinases/genética , Morfogênese/genética , Mesoderma/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Pulmão/metabolismo , Organogênese/genética , Regulação da Expressão Gênica no Desenvolvimento
16.
Biol Open ; 13(3)2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38451093

RESUMO

Loss of Cdx2 in vivo leads to stunted development of the allantois, an extraembryonic mesoderm-derived structure critical for nutrient delivery and waste removal in the early embryo. Here, we investigate how CDX2 dose-dependently influences the gene regulatory network underlying extraembryonic mesoderm development. By engineering human induced pluripotent stem cells (hiPSCs) consisting of wild-type (WT), heterozygous (CDX2-Het), and homozygous null CDX2 (CDX2-KO) genotypes, differentiating these cells in a 2D gastruloid model, and subjecting these cells to single-nucleus RNA and ATAC sequencing, we identify several pathways that are dose-dependently regulated by CDX2 including VEGF and non-canonical WNT. snATAC-seq reveals that CDX2-Het cells retain a WT-like chromatin accessibility profile, suggesting accessibility alone is not sufficient to drive this variability in gene expression. Because the loss of CDX2 or TBXT phenocopy one another in vivo, we compared differentially expressed genes in our CDX2-KO to those from TBXT-KO hiPSCs differentiated in an analogous experiment. This comparison identifies several communally misregulated genes that are critical for cytoskeletal integrity and tissue permeability. Together, these results clarify how CDX2 dose-dependently regulates gene expression in the extraembryonic mesoderm and reveal pathways that may underlie the defects in vascular development and allantoic elongation seen in vivo.


Assuntos
Fator de Transcrição CDX2 , Dosagem de Genes , Redes Reguladoras de Genes , Células-Tronco Pluripotentes Induzidas , Humanos , Fator de Transcrição CDX2/genética , Diferenciação Celular/genética , Embrião de Mamíferos , Mesoderma
17.
Cells ; 13(6)2024 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-38534378

RESUMO

Pluripotent stem cells can be differentiated into all three germ-layers including ecto-, endo-, and mesoderm in vitro. However, the early identification and rapid characterization of each germ-layer in response to chemical and physical induction of differentiation is limited. This is a long-standing issue for rapid and high-throughput screening to determine lineage specification efficiency. Here, we present deep learning (DL) methodologies for predicting and classifying early mesoderm cells differentiated from embryoid bodies (EBs) based on cellular and nuclear morphologies. Using a transgenic murine embryonic stem cell (mESC) line, namely OGTR1, we validated the upregulation of mesodermal genes (Brachyury (T): DsRed) in cells derived from EBs for the deep learning model training. Cells were classified into mesodermal and non-mesodermal (representing endo- and ectoderm) classes using a convolutional neural network (CNN) model called InceptionV3 which achieved a very high classification accuracy of 97% for phase images and 90% for nuclei images. In addition, we also performed image segmentation using an Attention U-Net CNN and obtained a mean intersection over union of 61% and 69% for phase-contrast and nuclear images, respectively. This work highlights the potential of integrating cell culture, imaging technologies, and deep learning methodologies in identifying lineage specification, thus contributing to the advancements in regenerative medicine. Collectively, our trained deep learning models can predict the mesoderm cells with high accuracy based on cellular and nuclear morphologies.


Assuntos
Aprendizado Profundo , Células-Tronco Pluripotentes , Animais , Camundongos , Diferenciação Celular/fisiologia , Camadas Germinativas/metabolismo , Mesoderma/metabolismo
18.
Curr Top Dev Biol ; 157: 1-42, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38556456

RESUMO

This article is about how the famous organizer experiment has been perceived since it was first published in 1924. The experiment involves the production of a secondary embryo under the influence of a graft of a dorsal lip from an amphibian gastrula to a host embryo. The early experiments of Spemann and his school gave rise to a view that the whole early amphibian embryo was "indifferent" in terms of determination, except for a special region called "the organizer". This was viewed mainly as an agent of neural induction, also having the ability to generate an anteroposterior body pattern. Early biochemical efforts to isolate a factor emitted by the organizer were not successful but culminated in the definition of "neuralizing (N)" and "mesodermalizing (M)" factors present in a wide variety of animal tissues. By the 1950s this view became crystallized as a "two gradient" model involving the N and M factors, which explained the anteroposterior patterning effect. In the 1970s, the phenomenon of mesoderm induction was characterized as a process occurring before the commencement of gastrulation. Reinvestigation of the organizer effect using lineage labels gave rise to a more precise definition of the sequence of events. Since the 1980s, modern research using the tools of molecular biology, combined with microsurgery, has explained most of the processes involved. The organizer graft should now be seen as an experiment which involves multiple interactions: dorsoventral polarization following fertilization, mesoderm induction, the dorsalizing signal responsible for neuralization and dorsoventral patterning of the mesoderm, and additional factors responsible for anteroposterior patterning.


Assuntos
Desenvolvimento Embrionário , Mesoderma , Animais , Anfíbios , Biologia do Desenvolvimento , Padronização Corporal , Indução Embrionária , Organizadores Embrionários , Regulação da Expressão Gênica no Desenvolvimento
19.
Curr Top Dev Biol ; 157: 83-123, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38556460

RESUMO

For almost a century, developmental biologists have appreciated that the ability of the embryonic organizer to induce and pattern the body plan is intertwined with its differentiation into axial mesoderm. Despite this, we still have a relatively poor understanding of the contribution of axial mesoderm to induction and patterning of different body regions, and the manner in which axial mesoderm-derived information is interpreted in tissues of changing competence. Here, with a particular focus on the nervous system, we review the evidence that axial mesoderm notochord and prechordal mesoderm/mesendoderm act as organizers, discuss how their influence extends through the different axes of the developing organism, and describe how the ability of axial mesoderm to direct morphogenesis impacts on its role as a local organizer.


Assuntos
Encéfalo/embriologia , Face/embriologia , Camadas Germinativas , Mesoderma , Sistema Nervoso , Mesoderma/fisiologia , Morfogênese , Padronização Corporal
20.
Curr Top Dev Biol ; 157: 43-65, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38556458

RESUMO

In avian and mammalian embryos the "organizer" property associated with neural induction of competent ectoderm into a neural plate and its subsequent patterning into rostro-caudal domains resides at the tip of the primitive streak before neurulation begins, and before a morphological Hensen's node is discernible. The same region and its later derivatives (like the notochord) also have the ability to "dorsalize" the adjacent mesoderm, for example by converting lateral plate mesoderm into paraxial (pre-somitic) mesoderm. Both neural induction and dorsalization of the mesoderm involve inhibition of BMP, and the former also requires other signals. This review surveys the key experiments done to elucidate the functions of the organizer and the mechanisms of neural induction in amniotes. We conclude that the mechanisms of neural induction in amniotes and anamniotes are likely to be largely the same; apparent differences are likely to be due to differences in experimental approaches dictated by embryo topology and other practical constraints. We also discuss the relationships between "neural induction" assessed by grafts of the organizer and normal neural plate development, as well as how neural induction relates to the generation of neuronal cells from embryonic and other stem cells in vitro.


Assuntos
Mesoderma , Somitos , Animais , Indução Embrionária/fisiologia , Aves , Mamíferos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...