Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 337
Filtrar
1.
Nature ; 618(7965): 543-549, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37225983

RESUMEN

The development of paired appendages was a key innovation during evolution and facilitated the aquatic to terrestrial transition of vertebrates. Largely derived from the lateral plate mesoderm (LPM), one hypothesis for the evolution of paired fins invokes derivation from unpaired median fins via a pair of lateral fin folds located between pectoral and pelvic fin territories1. Whilst unpaired and paired fins exhibit similar structural and molecular characteristics, no definitive evidence exists for paired lateral fin folds in larvae or adults of any extant or extinct species. As unpaired fin core components are regarded as exclusively derived from paraxial mesoderm, any transition presumes both co-option of a fin developmental programme to the LPM and bilateral duplication2. Here, we identify that the larval zebrafish unpaired pre-anal fin fold (PAFF) is derived from the LPM and thus may represent a developmental intermediate between median and paired fins. We trace the contribution of LPM to the PAFF in both cyclostomes and gnathostomes, supporting the notion that this is an ancient trait of vertebrates. Finally, we observe that the PAFF can be bifurcated by increasing bone morphogenetic protein signalling, generating LPM-derived paired fin folds. Our work provides evidence that lateral fin folds may have existed as embryonic anlage for elaboration to paired fins.


Asunto(s)
Aletas de Animales , Evolución Biológica , Mesodermo , Pez Cebra , Animales , Aletas de Animales/anatomía & histología , Aletas de Animales/embriología , Aletas de Animales/crecimiento & desarrollo , Larva/anatomía & histología , Larva/crecimiento & desarrollo , Mesodermo/anatomía & histología , Mesodermo/embriología , Mesodermo/crecimiento & desarrollo , Pez Cebra/anatomía & histología , Pez Cebra/embriología , Pez Cebra/crecimiento & desarrollo , Proteínas Morfogenéticas Óseas/metabolismo
2.
Physiol Rev ; 103(3): 1899-1964, 2023 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-36656056

RESUMEN

The teeth are vertebrate-specific, highly specialized organs performing fundamental functions of mastication and speech, the maintenance of which is crucial for orofacial homeostasis and is further linked to systemic health and human psychosocial well-being. However, with limited ability for self-repair, the teeth can often be impaired by traumatic, inflammatory, and progressive insults, leading to high prevalence of tooth loss and defects worldwide. Regenerative medicine holds the promise to achieve physiological restoration of lost or damaged organs, and in particular an evolving framework of developmental engineering has pioneered functional tooth regeneration by harnessing the odontogenic program. As a key event of tooth morphogenesis, mesenchymal condensation dictates dental tissue formation and patterning through cellular self-organization and signaling interaction with the epithelium, which provides a representative to decipher organogenetic mechanisms and can be leveraged for regenerative purposes. In this review, we summarize how mesenchymal condensation spatiotemporally assembles from dental stem cells (DSCs) and sequentially mediates tooth development. We highlight condensation-mimetic engineering efforts and mechanisms based on ex vivo aggregation of DSCs, which have achieved functionally robust and physiologically relevant tooth regeneration after implantation in animals and in humans. The discussion of this aspect will add to the knowledge of development-inspired tissue engineering strategies and will offer benefits to propel clinical organ regeneration.


Asunto(s)
Regeneración Ósea , Mesodermo , Odontogénesis , Ingeniería de Tejidos , Pérdida de Diente , Diente , Diente/crecimiento & desarrollo , Ingeniería de Tejidos/métodos , Humanos , Animales , Mesodermo/crecimiento & desarrollo , Pérdida de Diente/terapia
3.
Exp Cell Res ; 410(1): 112931, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34798131

RESUMEN

Branchiomeric muscles of the head and neck originate in a population of cranial mesoderm termed cardiopharyngeal mesoderm that also contains progenitor cells contributing to growth of the embryonic heart. Retrospective lineage analysis has shown that branchiomeric muscles share a clonal origin with parts of the heart, indicating the presence of common heart and head muscle progenitor cells in the early embryo. Genetic lineage tracing and functional studies in the mouse, as well as in Ciona and zebrafish, together with recent experiments using single cell transcriptomics and multipotent stem cells, have provided further support for the existence of bipotent head and heart muscle progenitor cells. Current challenges concern defining where and when such common progenitor cells exist in mammalian embryos and how alternative myogenic derivatives emerge in cardiopharyngeal mesoderm. Addressing these questions will provide insights into mechanisms of cell fate acquisition and the evolution of vertebrate musculature, as well as clinical insights into the origins of muscle restricted myopathies and congenital defects affecting craniofacial and cardiac development.


Asunto(s)
Desarrollo Embrionario/genética , Corazón/crecimiento & desarrollo , Mesodermo/crecimiento & desarrollo , Desarrollo de Músculos/genética , Animales , Diferenciación Celular/genética , Embrión de Mamíferos , Regulación del Desarrollo de la Expresión Génica/genética , Cabeza/crecimiento & desarrollo , Ratones , Músculo Esquelético/crecimiento & desarrollo , Células Madre/citología , Pez Cebra/genética
4.
Exp Cell Res ; 410(1): 112950, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34838813

RESUMEN

Drosophila embryonic somatic muscles represent a simple and tractable model system to study the gene regulatory networks that control diversification of cell types. Somatic myogenesis in Drosophila is initiated by intrinsic action of the mesodermal master gene twist, which activates a cascade of transcriptional outputs including myogenic differentiation factor Mef2, which triggers all aspects of the myogenic differentiation program. In parallel, the expression of a combinatorial code of identity transcription factors (iTFs) defines discrete particular features of each muscle fiber, such as number of fusion events, and specific attachment to tendon cells or innervation, thus ensuring diversification of muscle types. Here, we take the example of a subset of lateral transverse (LT) muscles and discuss how the iTF code and downstream effector genes progressively define individual LT properties such as fusion program, attachment and innervation. We discuss new challenges in the field including the contribution of posttranscriptional and epitranscriptomic regulation of gene expression in the diversification of cell types.


Asunto(s)
Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Desarrollo Embrionario/genética , Desarrollo de Músculos/genética , Factores Reguladores Miogénicos/genética , Animales , Diferenciación Celular/genética , Drosophila melanogaster/crecimiento & desarrollo , Embrión no Mamífero , Regulación del Desarrollo de la Expresión Génica/genética , Mesodermo/crecimiento & desarrollo , Mesodermo/metabolismo , Músculos/metabolismo
5.
Development ; 148(23)2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34905617

RESUMEN

Development of the Drosophila visceral muscle depends on Anaplastic Lymphoma Kinase (Alk) receptor tyrosine kinase (RTK) signaling, which specifies founder cells (FCs) in the circular visceral mesoderm (VM). Although Alk activation by its ligand Jelly Belly (Jeb) is well characterized, few target molecules have been identified. Here, we used targeted DamID (TaDa) to identify Alk targets in embryos overexpressing Jeb versus embryos with abrogated Alk activity, revealing differentially expressed genes, including the Snail/Scratch family transcription factor Kahuli (Kah). We confirmed Kah mRNA and protein expression in the VM, and identified midgut constriction defects in Kah mutants similar to those of pointed (pnt). ChIP and RNA-Seq data analysis defined a Kah target-binding site similar to that of Snail, and identified a set of common target genes putatively regulated by Kah and Pnt during midgut constriction. Taken together, we report a rich dataset of Alk-responsive loci in the embryonic VM and functionally characterize the role of Kah in the regulation of embryonic midgut morphogenesis.


Asunto(s)
Quinasa de Linfoma Anaplásico , Proteínas de Unión al ADN , Proteínas de Drosophila , Desarrollo Embrionario , Proteínas del Tejido Nervioso , Proteínas Proto-Oncogénicas , Factores de Transcripción , Animales , Quinasa de Linfoma Anaplásico/genética , Diferenciación Celular/genética , Proteínas de Unión al ADN/genética , Drosophila melanogaster/genética , Drosophila melanogaster/crecimiento & desarrollo , Proteínas de Drosophila/genética , Desarrollo Embrionario/genética , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica/genética , Mesodermo/crecimiento & desarrollo , Mesodermo/metabolismo , Desarrollo de Músculos/genética , Músculos/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas Proto-Oncogénicas/genética , ARN Mensajero/genética , RNA-Seq , Transducción de Señal/genética , Análisis de la Célula Individual , Factores de Transcripción/genética
6.
PLoS Genet ; 17(12): e1009982, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34928956

RESUMEN

Sonic Hedgehog/GLI3 signaling is critical in regulating digit number, such that Gli3-deficiency results in polydactyly and Shh-deficiency leads to digit number reductions. SHH/GLI3 signaling regulates cell cycle factors controlling mesenchymal cell proliferation, while simultaneously regulating Grem1 to coordinate BMP-induced chondrogenesis. SHH/GLI3 signaling also coordinates the expression of additional genes, however their importance in digit formation remain unknown. Utilizing genetic and molecular approaches, we identified HES1 as a downstream modifier of the SHH/GLI signaling axis capable of inducing preaxial polydactyly (PPD), required for Gli3-deficient PPD, and capable of overcoming digit number constraints of Shh-deficiency. Our data indicate that HES1, a direct SHH/GLI signaling target, induces mesenchymal cell proliferation via suppression of Cdkn1b, while inhibiting chondrogenic genes and the anterior autopod boundary regulator, Pax9. These findings establish HES1 as a critical downstream effector of SHH/GLI3 signaling in the development of PPD.


Asunto(s)
Proteínas Hedgehog/genética , Proteínas del Tejido Nervioso/genética , Factor de Transcripción PAX9/genética , Polidactilia/genética , Pulgar/anomalías , Factor de Transcripción HES-1/genética , Proteína Gli3 con Dedos de Zinc/genética , Animales , División Celular/genética , Proliferación Celular/genética , Condrogénesis/genética , Cromatina/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Modelos Animales de Enfermedad , Humanos , Esbozos de los Miembros/crecimiento & desarrollo , Esbozos de los Miembros/metabolismo , Mesodermo/crecimiento & desarrollo , Ratones , Polidactilia/patología , Pulgar/patología
7.
Development ; 148(23)2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34822716

RESUMEN

The node-streak border region comprising notochord progenitor cells (NPCs) at the posterior node and neuro-mesodermal progenitor cells (NMPs) in the adjacent epiblast is the prime organizing center for axial elongation in mouse embryos. The T-box transcription factor brachyury (T) is essential for both formation of the notochord and maintenance of NMPs, and thus is a key regulator of trunk and tail development. The T promoter controlling T expression in NMPs and nascent mesoderm has been characterized in detail; however, control elements for T expression in the notochord have not been identified yet. We have generated a series of deletion alleles by CRISPR/Cas9 genome editing in mESCs, and analyzed their effects in mutant mouse embryos. We identified a 37 kb region upstream of T that is essential for notochord function and tailbud outgrowth. Within that region, we discovered a T-binding enhancer required for notochord cell specification and differentiation. Our data reveal a complex regulatory landscape controlling cell type-specific expression and function of T in NMP/nascent mesoderm and node/notochord, allowing proper trunk and tail development.


Asunto(s)
Desarrollo Embrionario/genética , Elementos de Facilitación Genéticos/genética , Proteínas Fetales/genética , Proteínas de Dominio T Box/genética , Cola (estructura animal)/crecimiento & desarrollo , Secuencia de Aminoácidos/genética , Animales , Sistemas CRISPR-Cas/genética , Edición Génica/métodos , Regulación del Desarrollo de la Expresión Génica/genética , Mesodermo/crecimiento & desarrollo , Mesodermo/metabolismo , Ratones , Células Madre Embrionarias de Ratones/citología , Células Madre Embrionarias de Ratones/metabolismo , Notocorda/crecimiento & desarrollo , Notocorda/metabolismo , Regiones Promotoras Genéticas/genética , Secuencias Reguladoras de Ácidos Nucleicos/genética , Cola (estructura animal)/metabolismo
8.
Sci Rep ; 11(1): 18030, 2021 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-34504115

RESUMEN

The mesoderm is considered the youngest of the three germ layers. Although its morphogenesis has been studied in some metazoans, the molecular components underlying this process remain obscure for numerous phyla including the highly diverse Mollusca. Here, expression of Hairy and enhancer of split (HES), Mox, and myosin heavy chain (MHC) was investigated in Acanthochitona fascicularis, a representative of Polyplacophora with putative ancestral molluscan features. While AfaMHC is expressed throughout myogenesis, AfaMox1 is only expressed during early stages of mesodermal band formation and in the ventrolateral muscle, an autapomorphy of the polyplacophoran trochophore. Comparing our findings to previously published data across Metazoa reveals Mox expression in the mesoderm in numerous bilaterians including gastropods, polychaetes, and brachiopods. It is also involved in myogenesis in molluscs, annelids, tunicates, and craniates, suggesting a dual role of Mox in mesoderm and muscle formation in the last common bilaterian ancestor. AfaHESC2 is expressed in the ectoderm of the polyplacophoran gastrula and later in the mesodermal bands and in putative neural tissue, whereas AfaHESC7 is expressed in the trochoblasts of the gastrula and during foregut formation. This confirms the high developmental variability of HES gene expression and demonstrates that Mox and HES genes are pleiotropic.


Asunto(s)
Pleiotropía Genética , Proteínas de Homeodominio/genética , Mesodermo/metabolismo , Cadenas Pesadas de Miosina/genética , Poliplacóforos/genética , Factor de Transcripción HES-1/genética , Animales , Anélidos/clasificación , Anélidos/genética , Evolución Biológica , Gastrulación/genética , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Mesodermo/citología , Mesodermo/crecimiento & desarrollo , Morfogénesis/genética , Cadenas Pesadas de Miosina/metabolismo , Filogenia , Poliplacóforos/clasificación , Poliplacóforos/crecimiento & desarrollo , Poliplacóforos/metabolismo , Factor de Transcripción HES-1/metabolismo , Urocordados/clasificación , Urocordados/genética
9.
Genes (Basel) ; 12(8)2021 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-34440466

RESUMEN

Mesodermal cells of holothurian Eupentacta fraudatrix can transdifferentiate into enterocytes during the regeneration of the digestive system. In this study, we investigated the expression of several genes involved in gut regeneration in E. fraudatrix. Moreover, the localization of progenitor cells of coelomocytes, juvenile cells, and their participation in the formation of the luminal epithelium of the digestive tube were studied. It was shown that Piwi-positive cells were not involved in the formation of the luminal epithelium of the digestive tube. Ef-72 kDa type IV collagenase and Ef-MMP16 had an individual expression profile and possibly different functions. The Ef-tensilin3 gene exhibited the highest expression and indicates its potential role in regeneration. Ef-Sox9/10 and Ef-Sox17 in E. fraudatrix may participate in the mechanism of transdifferentiation of coelomic epithelial cells. Their transcripts mark the cells that plunge into the connective tissue of the gut anlage and give rise to enterocytes. Ef-Sox9/10 probably controls the switching of mesodermal cells to the enterocyte phenotype, while Ef-Sox17 may be involved in the regulation of the initial stages of transdifferentiation.


Asunto(s)
Sistema Digestivo/crecimiento & desarrollo , Tracto Gastrointestinal/crecimiento & desarrollo , Regeneración/genética , Pepinos de Mar/genética , Animales , Transdiferenciación Celular/genética , Sistema Digestivo/metabolismo , Células Epiteliales/metabolismo , Tracto Gastrointestinal/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Metaloproteinasas de la Matriz/genética , Mesodermo/crecimiento & desarrollo , Mesodermo/metabolismo , ARN Interferente Pequeño/genética , Factores de Transcripción SOX/genética , Pepinos de Mar/crecimiento & desarrollo , Inhibidores Tisulares de Metaloproteinasas/genética
10.
Genes (Basel) ; 12(7)2021 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-34202311

RESUMEN

Transforming growth factor ß (TGFß) signaling plays an important role in skeletal development. We previously demonstrated that the loss of TGFß receptor II (Tgfbr2) in Osterix-Cre-expressing mesenchyme results in defects in bones and teeth due to reduced proliferation and differentiation in pre-osteoblasts and pre-odontoblasts. These Osterix-Cre;Tgfbr2f/f mice typically die within approximately four weeks for unknown reasons. To investigate the cause of death, we performed extensive pathological analysis on Osterix-Cre- (Cre-), Osterix-Cre+;Tgfbr2f/wt (HET), and Osterix-Cre+;Tgfbr2f/f (CKO) mice. We also crossed Osterix-Cre mice with the ROSA26mTmG reporter line to identify potential off-target Cre expression. The findings recapitulated published skeletal and tooth abnormalities and revealed previously unreported osteochondral dysplasia throughout both the appendicular and axial skeletons in the CKO mice, including the calvaria. Alterations to the nasal area and teeth suggest a potentially reduced capacity to sense and process food, while off-target Cre expression in the gastrointestinal tract may indicate an inability to absorb nutrients. Additionally, altered nasal passages and unexplained changes in diaphragmatic muscle support the possibility of hypoxia. We conclude that these mice likely died due to a combination of breathing difficulties, malnutrition, and starvation resulting primarily from skeletal deformities that decreased their ability to sense, gather, and process food.


Asunto(s)
Osteogénesis/genética , Receptor Tipo II de Factor de Crecimiento Transformador beta/genética , Esqueleto/anomalías , Factor de Transcripción Sp7/genética , Animales , Huesos/anomalías , Huesos/fisiopatología , Diferenciación Celular/genética , Modelos Animales de Enfermedad , Regulación del Desarrollo de la Expresión Génica/genética , Humanos , Integrasas/genética , Mesodermo/crecimiento & desarrollo , Mesodermo/metabolismo , Ratones , Osteoblastos/metabolismo , Osteoblastos/patología , Transducción de Señal/genética , Esqueleto/diagnóstico por imagen , Esqueleto/metabolismo , Esqueleto/fisiopatología
11.
Development ; 148(13)2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-34228796

RESUMEN

The trachea delivers inhaled air into the lungs for gas exchange. Anomalies in tracheal development can result in life-threatening malformations, such as tracheoesophageal fistula and tracheomalacia. Given the limitations of current therapeutic approaches, development of technologies for the reconstitution of a three-dimensional trachea from stem cells is urgently required. Recently, single-cell sequencing technologies and quantitative analyses from cell to tissue scale have been employed to decipher the cellular basis of tracheal morphogenesis. In this Review, recent advances in mammalian tracheal development and the generation of tracheal tissues from pluripotent stem cells are summarized.


Asunto(s)
Pulmón/crecimiento & desarrollo , Morfogénesis/fisiología , Tráquea/crecimiento & desarrollo , Fístula Traqueoesofágica/patología , Animales , Cartílago/crecimiento & desarrollo , Diferenciación Celular , Epitelio , Humanos , Mesodermo/crecimiento & desarrollo , Ratones , Morfogénesis/genética , Sistema Respiratorio , Tráquea/anomalías , Traqueomalacia , Transcriptoma
12.
Genes Genomics ; 43(9): 1087-1094, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34302633

RESUMEN

BACKGROUND: In tooth bioengineering for replacement therapy of missing teeth, the utilized cells must possess an inductive signal-forming ability to initiate odontogenesis. This ability is called odontogenic potential. In mice, the odontogenic potential signal is known to be translocated from the epithelium to the mesenchyme at the early bud stage in the developing molar tooth germ. However, the identity of the molecular constituents of this process remains unclear. OBJECTIVE: The purpose of this study is to determine the molecular identity of odontogenic potential and to provide a new perspective in the field of tooth development research. METHODS: In this study, whole transcriptome profiles of the mouse molar tooth germ epithelium and mesenchyme were investigated using the RNA sequencing (RNA-seq) technique. The analyzed transcriptomes corresponded to two developmental stages, embryonic day 11.5 (E11.5) and 14.5 (E14.5), which represent the odontogenic potential shifts. RESULTS: We identified differentially expressed genes (DEGs), which were specifically overexpressed in both the E11.5 epithelium and E14.5 mesenchyme, but not expressed in their respective counterparts. Of the 55 DEGs identified, the top three most expressed transcription factor genes (transcription factor AP-2 beta isoform 3 [TFAP2B], developing brain homeobox protein 2 [DBX2], and insulin gene enhancer protein ISL-1 [ISL1]) and three tooth development-related genes (transcription factor HES-5 [HES5], platelet-derived growth factor D precursor [PDGFD], semaphrin-3 A precursor [SEMA3A]) were selected and validated by quantitative RT-PCR. Using immunofluorescence staining, the TFAP2B protein expression was found to be localized only at the E11.5 epithelium and E14.5 mesenchyme. CONCLUSIONS: Thus, our empirical findings in the present study may provide a new perspective into the characterization of the molecules responsible for the odontogenic potential and may have an implication in the cell-based whole tooth regeneration strategy.


Asunto(s)
Diente Molar/crecimiento & desarrollo , Odontogénesis/genética , Germen Dentario/crecimiento & desarrollo , Transcriptoma/genética , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Epitelio/crecimiento & desarrollo , Epitelio/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Estudios de Asociación Genética , Proteínas de Homeodominio/genética , Humanos , Proteínas con Homeodominio LIM/genética , Linfocinas/genética , Mesodermo/crecimiento & desarrollo , Mesodermo/metabolismo , Ratones , Diente Molar/metabolismo , Factor de Crecimiento Derivado de Plaquetas/genética , RNA-Seq , Proteínas Represoras/genética , Semaforina-3A/genética , Germen Dentario/metabolismo , Factor de Transcripción AP-2/genética , Factores de Transcripción/genética
13.
Genes (Basel) ; 12(4)2021 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-33920662

RESUMEN

Ascidians are invertebrate chordates and the closest living relative to vertebrates. In ascidian embryos a large part of the central nervous system arises from cells associated with mesoderm rather than ectoderm lineages. This seems at odds with the traditional view of vertebrate nervous system development which was thought to be induced from ectoderm cells, initially with anterior character and later transformed by posteriorizing signals, to generate the entire anterior-posterior axis of the central nervous system. Recent advances in vertebrate developmental biology, however, show that much of the posterior central nervous system, or spinal cord, in fact arises from cells that share a common origin with mesoderm. This indicates a conserved role for bi-potential neuromesoderm precursors in chordate CNS formation. However, the boundary between neural tissue arising from these distinct neural lineages does not appear to be fixed, which leads to the notion that anterior-posterior patterning and neural fate formation can evolve independently.


Asunto(s)
Sistema Nervioso Central/crecimiento & desarrollo , Urocordados/embriología , Animales , Tipificación del Cuerpo , Linaje de la Célula , Ectodermo/crecimiento & desarrollo , Regulación del Desarrollo de la Expresión Génica , Mesodermo/crecimiento & desarrollo , Urocordados/crecimiento & desarrollo
14.
Development ; 148(9)2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33757991

RESUMEN

In the face, symmetry is established when bilateral streams of neural crest cells leave the neural tube at the same time, follow identical migration routes and then give rise to the facial prominences. However, developmental instability exists, particularly surrounding the steps of lip fusion. The causes of instability are unknown but inability to cope with developmental fluctuations are a likely cause of congenital malformations, such as non-syndromic orofacial clefts. Here, we tracked cell movements over time in the frontonasal mass, which forms the facial midline and participates in lip fusion, using live-cell imaging of chick embryos. Our mathematical examination of cell velocity vectors uncovered temporal fluctuations in several parameters, including order/disorder, symmetry/asymmetry and divergence/convergence. We found that treatment with a Rho GTPase inhibitor completely disrupted the temporal fluctuations in all measures and blocked morphogenesis. Thus, we discovered that genetic control of symmetry extends to mesenchymal cell movements and that these movements are of the type that could be perturbed in asymmetrical malformations, such as non-syndromic cleft lip. This article has an associated 'The people behind the papers' interview.


Asunto(s)
Movimiento Celular , Cara/fisiología , Mesodermo/crecimiento & desarrollo , Cresta Neural/fisiología , Actomiosina , Animales , Encéfalo/anatomía & histología , Encéfalo/crecimiento & desarrollo , División Celular , Proliferación Celular , Embrión de Pollo , Pollos , Labio Leporino/genética , Fisura del Paladar/genética , Ojo/anatomía & histología , Ojo/crecimiento & desarrollo , Cara/anomalías , Regulación del Desarrollo de la Expresión Génica , Mesodermo/anatomía & histología , Morfogénesis/genética , Cresta Neural/anatomía & histología
15.
Development ; 148(6)2021 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-33658223

RESUMEN

The anteroposterior axial identity of motor neurons (MNs) determines their functionality and vulnerability to neurodegeneration. Thus, it is a crucial parameter in the design of strategies aiming to produce MNs from human pluripotent stem cells (hPSCs) for regenerative medicine/disease modelling applications. However, the in vitro generation of posterior MNs corresponding to the thoracic/lumbosacral spinal cord has been challenging. Although the induction of cells resembling neuromesodermal progenitors (NMPs), the bona fide precursors of the spinal cord, offers a promising solution, the progressive specification of posterior MNs from these cells is not well defined. Here, we determine the signals guiding the transition of human NMP-like cells toward thoracic ventral spinal cord neurectoderm. We show that combined WNT-FGF activities drive a posterior dorsal pre-/early neural state, whereas suppression of TGFß-BMP signalling pathways promotes a ventral identity and neural commitment. Based on these results, we define an optimised protocol for the generation of thoracic MNs that can efficiently integrate within the neural tube of chick embryos. We expect that our findings will facilitate the comparison of hPSC-derived spinal cord cells of distinct axial identities.


Asunto(s)
Diferenciación Celular/genética , Mesodermo/crecimiento & desarrollo , Células-Madre Neurales/metabolismo , Médula Espinal/crecimiento & desarrollo , Animales , Tipificación del Cuerpo/genética , Proteínas Morfogenéticas Óseas/genética , Linaje de la Célula/genética , Embrión de Pollo , Factores de Crecimiento de Fibroblastos/genética , Regulación del Desarrollo de la Expresión Génica/genética , Humanos , Mesodermo/metabolismo , Neuronas Motoras/metabolismo , Células-Madre Neurales/citología , Células Madre Pluripotentes/citología , Transducción de Señal/genética , Médula Espinal/metabolismo , Factor de Crecimiento Transformador beta/genética , Proteínas Wnt/genética
16.
Cell Prolif ; 54(4): e13012, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33656760

RESUMEN

OBJECTIVES: Vitronectin (VTN) has been widely used for the maintenance and expansion of human pluripotent stem cells (hPSCs) as feeder-free conditions. However, the effect of VTN on hPSC differentiation remains unclear. Here, we investigated the role of VTN in early haematopoietic development of hPSCs. MATERIALS AND METHODS: A chemically defined monolayer system was applied to study the role of different matrix or basement membrane proteins in haematopoietic development of hPSCs. The role of integrin signalling in VTN-mediated haematopoietic differentiation was investigated by integrin antagonists. Finally, small interfering RNA was used to knock down integrin gene expression in differentiated cells. RESULTS: We found that the haematopoietic differentiation of hPSCs on VTN was far more efficient than that on Matrigel that is also often used for hPSC culture. VTN promoted the fate determination of endothelial-haematopoietic lineage during mesoderm development to generate haemogenic endothelium (HE). Moreover, we demonstrated that the signals through αvß3 and αvß5 integrins were required for VTN-promoted haematopoietic differentiation. Blocking αvß3 and αvß5 integrins by the integrin antagonists impaired the development of HE, but not endothelial-to-haematopoietic transition (EHT). Finally, both αvß3 and αvß5 were confirmed acting synergistically for early haematopoietic differentiation by knockdown the expression of αv, ß3 or ß5. CONCLUSION: The established VTN-based monolayer system of haematopoietic differentiation of hPSCs presents a valuable platform for further investigating niche signals involved in human haematopoietic development.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Integrina alfaVbeta3/metabolismo , Receptores de Vitronectina/metabolismo , Vitronectina/farmacología , Adhesión Celular/efectos de los fármacos , Línea Celular , Regulación de la Expresión Génica , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Integrina alfaVbeta3/antagonistas & inhibidores , Integrina alfaVbeta3/genética , Mesodermo/citología , Mesodermo/crecimiento & desarrollo , Mesodermo/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Receptores de Vitronectina/antagonistas & inhibidores , Receptores de Vitronectina/genética , Transducción de Señal/efectos de los fármacos , Venenos de Serpiente/farmacología
17.
Mol Biol Rep ; 48(1): 395-403, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33387197

RESUMEN

High vascularization is a biological characteristic of glioblastoma (GBM); however, an in-vitro experimental model to verify the mechanism and physiological role of vasculogenesis in GBM is not well-established. Recently, we established a self-organizing vasculogenic model using human umbilical vein endothelial cells (HUVECs) co-cultivated with human lung fibroblasts (hLFs). Here, we exploited this system to establish a realistic model of vasculogenesis in GBM. We developed two polydimethylsiloxane (PDMS) devices, a doughnut-hole dish and a 5-lane microfluidic device to observe the contact-independent effects of glioblastoma cells on HUVECs. We tested five patient-derived and five widely used GBM cell lines. Confocal fluorescence microscopy was used to observe the morphological changes in Red Fluorescent Protein (RFP)-HUVECs and fluorescein isothiocyanate (FITC)-dextran perfusion. The genetic and expression properties of GBM cell lines were analyzed. The doughnut-hole dish assay revealed KNS1451 as the only cells to induce HUVEC transformation to vessel-like structures, similar to hLFs. The 5-lane device assay demonstrated that KNS1451 promoted the formation of a vascular network that was fully perfused, revealing the functioning luminal construction. Microarray analysis revealed that KNS1451 is a mesenchymal subtype of GBM. Using a patient-derived mesenchymal GBM cell line, mature de-novo vessel formation could be induced in HUVECs by contact-independent co-culture with GBM in a microfluidic device. These results support the development of a novel in vitro research model and provide novel insights in the neovasculogenic mechanism of GBM and may potentially facilitate the future detection of unknown molecular targets.


Asunto(s)
Neoplasias Encefálicas/genética , Diferenciación Celular/genética , Glioblastoma/genética , Neovascularización Patológica/genética , Vasos Sanguíneos/crecimiento & desarrollo , Vasos Sanguíneos/patología , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Proliferación Celular/genética , Técnicas de Cocultivo , Células Endoteliales/metabolismo , Células Endoteliales/patología , Glioblastoma/metabolismo , Glioblastoma/patología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Dispositivos Laboratorio en un Chip , Proteínas Luminiscentes/metabolismo , Células Madre Mesenquimatosas/metabolismo , Mesodermo/crecimiento & desarrollo , Mesodermo/patología , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Proteína Fluorescente Roja
18.
Development ; 148(22)2021 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-35020873

RESUMEN

The dynamics of multipotent neural crest cell differentiation and invasion as cells travel throughout the vertebrate embryo remain unclear. Here, we preserve spatial information to derive the transcriptional states of migrating neural crest cells and the cellular landscape of the first four chick cranial to cardiac branchial arches (BA1-4) using label-free, unsorted single-cell RNA sequencing. The faithful capture of branchial arch-specific genes led to identification of novel markers of migrating neural crest cells and 266 invasion genes common to all BA1-4 streams. Perturbation analysis of a small subset of invasion genes and time-lapse imaging identified their functional role to regulate neural crest cell behaviors. Comparison of the neural crest invasion signature to other cell invasion phenomena revealed a shared set of 45 genes, a subset of which showed direct relevance to human neuroblastoma cell lines analyzed after exposure to the in vivo chick embryonic neural crest microenvironment. Our data define an important spatio-temporal reference resource to address patterning of the vertebrate head and neck, and previously unidentified cell invasion genes with the potential for broad impact.


Asunto(s)
Región Branquial/crecimiento & desarrollo , Cabeza/crecimiento & desarrollo , Cuello/crecimiento & desarrollo , Cresta Neural/crecimiento & desarrollo , Animales , Tipificación del Cuerpo/genética , Región Branquial/embriología , Diferenciación Celular/genética , Movimiento Celular/genética , Microambiente Celular/genética , Embrión de Pollo , Embrión de Mamíferos , Embrión no Mamífero , Desarrollo Embrionario/genética , Cabeza/embriología , Humanos , Mesodermo/crecimiento & desarrollo , Células Madre Multipotentes/citología , Cuello/embriología , Cresta Neural/metabolismo , Neuroblastoma/genética , Neuroblastoma/patología , Organogénesis/genética , Microambiente Tumoral/genética , Vertebrados/genética , Vertebrados/crecimiento & desarrollo
19.
Methods Mol Biol ; 2179: 7-12, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32939708

RESUMEN

The epithelial-mesenchymal transition (EMT) is a key process required for building the early body plan of metazoa. It involves coordinated and precisely timed changes in multiple cell processes such as de-adhesion, motility, invasion, and cell polarity. While much has been learned about how embryos deploy epithelial-mesenchymal transitions since Betty Hay named the process decades ago, a number of things are still not well understood. Here I will discuss some of the big questions that remain, including how is all of this controlled, how does each of the cell biological events work, and how are they so nicely coordinated with one another?


Asunto(s)
Adhesión Celular/genética , Transición Epitelial-Mesenquimal/genética , Gastrulación/genética , Mesodermo/crecimiento & desarrollo , Animales , Movimiento Celular/genética , Polaridad Celular/genética , Embrión no Mamífero , Epitelio/metabolismo , Epitelio/patología , Humanos , Mesodermo/metabolismo
20.
Methods Mol Biol ; 2179: 29-33, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32939711

RESUMEN

As our understanding of Epithelial Mesenchymal Transition (EMT) increases, the original binary concept of E versus M no longer fits with experimental evidence. Re-definition of the EMT paradigm as spectral transitions between a full epithelium and a full mesenchyme suggests the existence of a virtual infinity of intermediate cellular states. The new challenge is to develop technical tools needed to contextualize each of these states and identify biologically significant cellular mechanisms that could be targeted in combatting EMT-related diseases.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Transición Epitelial-Mesenquimal/genética , Epitelio/crecimiento & desarrollo , Epitelio/metabolismo , Humanos , Mesodermo/crecimiento & desarrollo , Mesodermo/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...