Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
1.
Indian J Otolaryngol Head Neck Surg ; 76(1): 1224-1228, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38440545

ABSTRACT

Epidermoid cysts are infrequent lesions occupying the intracranial space, comprising approximately 1-2% of all intracranial tumors. Brainstem epidermoids are exceptionally uncommon in children; up until now, only a few scattered case reports have been documented in the literature regarding this unique location. These cysts commonly arise from the inclusion of ectodermal elements during neural tube closure. Complete excision of these cysts is challenging due to their close proximity and adherence to the brainstem, which makes it difficult to achieve. As a result, recurrence of the cysts is not uncommon. We have reported a rare case of a 3-year-old with a 5-month history of progressive headache, imbalance while walking and progressive weakness in his right upper limb and lower limb along with difficulty in swallowing. On MRI Brain imaging study he had a pre-pontine epidermoid with intra-axial extension in the pons. The patient underwent retro-sigmoid/suboccipital craniotomy and microsurgical excision of the tumor, including the tumor capsule. After completing the surgery, the cavity was irrigated using a solution containing hydrocortisone and Ringer lactate to prevent the occurrence of aseptic meningitis. In the postoperative, the patient recovered without any complications, as all symptoms showed immediate improvement, and the lower cranial nerves returned to normal functioning.

2.
Methods Mol Biol ; 2024 Mar 06.
Article in English | MEDLINE | ID: mdl-38441725

ABSTRACT

Linking phenotypes to genetic components has been an essential part of novel drug discovery, and screening methods have been widely employed to achieve such a goal. Screens can be conducted in either pooled or arrayed formats. Although arrayed screenings provide a better and cheaper alternative in small scale, the larger-scale screenings are conducted in pooled manner. With its adaptability to various models and conditions, CRISPR/Cas9 technology provides an invaluable alternative to classical and RNAi-based screening methods. Combined with high-throughput sequencing and bioinformatics, CRISPR-/Cas9-based pooled screening methods provide unbiased and robust data. In this protocol, we employed CRISPR-/Cas9-based pooled screening for a non-binary and non-immediate readout.

3.
Stem Cell Reports ; 19(4): 562-578, 2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38552632

ABSTRACT

Human pluripotent stem cell (hPSC) cultures are prone to genetic drift, because cells that have acquired specific genetic abnormalities experience a selective advantage in vitro. These abnormalities are highly recurrent in hPSC lines worldwide, but their functional consequences in differentiating cells are scarcely described. In this work, we show that the loss of chromosome 18q impairs neuroectoderm commitment and that downregulation of SALL3, a gene located in the common 18q loss region, is responsible for this failed neuroectodermal differentiation. Knockdown of SALL3 in control lines impaired differentiation in a manner similar to the loss of 18q, and transgenic overexpression of SALL3 in hESCs with 18q loss rescued the differentiation capacity of the cells. Finally, we show that loss of 18q and downregulation of SALL3 leads to changes in the expression of genes involved in pathways regulating pluripotency and differentiation, suggesting that these cells are in an altered state of pluripotency.


Subject(s)
Human Embryonic Stem Cells , Pluripotent Stem Cells , Humans , Cell Differentiation/genetics , Pluripotent Stem Cells/metabolism , Chromosomes
4.
Front Cell Dev Biol ; 11: 1240767, 2023.
Article in English | MEDLINE | ID: mdl-37655161

ABSTRACT

Acetylcholine, a vital neurotransmitter, plays a multifarious role in the brain and peripheral nervous system of various organisms. Previous research has demonstrated the proximity of cholinergic neurons to serotonergic neurons in the apical organ of sea urchin embryos. While several transcription factors have been identified as playing a role in the development of serotonergic neurons in this region of a sea urchin, Hemicentrotus pulcherrimus, comparatively little is known about the specific transcription factors and their spatiotemporal expression patterns that regulate the development of cholinergic neurons. In this study, we establish the requirement of the transcription factor Rx for the development of cholinergic neurons in the apical organ of the species. Furthermore, we investigate the role of the RNA-binding protein Musashi1, known to be involved in neurogenesis, including cholinergic neurons in other organisms, and demonstrate that it is a downstream factor of Rx, and that choline acetyltransferase expression is suppressed in Musashi1 downregulated embryos. Our research also highlights the intricate network formed by neurons and other cells in and around the apical organ of sea urchin larvae through axons and dendrites, providing possibility for a systematic and complexed neural pattern like those of the brain in other organisms.

5.
Cell Rep ; 42(9): 113146, 2023 09 26.
Article in English | MEDLINE | ID: mdl-37725511

ABSTRACT

The retinoblastoma family proteins (RBs) and E2F transcription factors are cell-autonomous regulators of cell-cycle progression, but they also impact fate choice in addition to tumor suppression. The range of mechanisms involved remains to be uncovered. Here, we show that RBs, particularly RBL2/p130, repress WNT ligands such as WNT4 and WNT8A, thereby directing ectoderm specification between neural crest to neuroepithelium. RBL2 achieves this function through cell-cycle-dependent cooperation with E2Fs and GCN5 on the regulatory regions of WNT loci, which direct neuroepithelial versus neural crest specification by temporal fluctuations of WNT/ß-catenin and DLL/NOTCH signaling activity. Thus, the RB-E2F bona fide cell-autonomous axis controls cell fate decisions, and RBL2 regulates field effects via WNT ligands. This reveals a non-cell-autonomous function of RBL2-E2F in stem cell and tissue progenitor differentiation that has broader implications for cell-cycle-dependent cell fate specification in organogenesis, adult stem cells, tissue homeostasis, and tumorigenesis.


Subject(s)
Body Patterning , Retinoblastoma Protein , Signal Transduction , Humans , Cell Cycle , Cell Differentiation , Cell Division , E2F Transcription Factors/genetics , E2F Transcription Factors/metabolism , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism
6.
Stem Cell Res Ther ; 14(1): 50, 2023 03 23.
Article in English | MEDLINE | ID: mdl-36959625

ABSTRACT

BACKGROUND: Three-dimensional (3D) human brain spheroids are instrumental to study central nervous system (CNS) development and (dys)function. Yet, in current brain spheroid models the limited variety of cell types hampers an integrated exploration of CNS (disease) mechanisms. METHODS: Here we report a 5-month culture protocol that reproducibly generates H9 embryonic stem cell-derived human cortical spheroids (hCSs) with a large cell-type variety. RESULTS: We established the presence of not only neuroectoderm-derived neural progenitor populations, mature excitatory and inhibitory neurons, astrocytes and oligodendrocyte (precursor) cells, but also mesoderm-derived microglia and endothelial cell populations in the hCSs via RNA-sequencing, qPCR, immunocytochemistry and transmission electron microscopy. Transcriptomic analysis revealed resemblance between the 5-months-old hCSs and dorsal frontal rather than inferior regions of human fetal brains of 19-26 weeks of gestational age. Pro-inflammatory stimulation of the generated hCSs induced a neuroinflammatory response, offering a proof-of-principle of the applicability of the spheroids. CONCLUSIONS: Our protocol provides a 3D human brain cell model containing a wide variety of innately developing neuroectoderm- as well as mesoderm-derived cell types, furnishing a versatile platform for comprehensive examination of intercellular CNS communication and neurological disease mechanisms.


Subject(s)
Brain , Neurons , Humans , Infant , Brain/metabolism , Neurons/metabolism , Cells, Cultured , Spheroids, Cellular , Astrocytes
7.
Front Cell Neurosci ; 16: 878351, 2022.
Article in English | MEDLINE | ID: mdl-35783089

ABSTRACT

Retinal organoids (ROs) derived from human pluripotent stem cells (hPSCs) recapitulate key features of retinogenesis and provide a promising platform to study retinal development and disease in a human context. Although multiple protocols are currently in use, hPSCs exhibit tremendous variability in differentiation efficiency, with some cell lines consistently yielding few or even no ROs, limiting their utility in research. We report here that early nicotinamide (NAM) treatment significantly improves RO yield across 8 hPSC lines from different donors, including some that would otherwise fail to generate a meaningful number of ROs. NAM treatment promotes neural commitment of hPSCs at the expense of non-neural ectodermal cell fate, which in turn increases eye field progenitor generation. Further analysis suggests that this effect is partially mediated through inhibition of BMP signaling. Our data encourage a broader use of human ROs for disease modeling applications that require the use of multiple patient-specific cell lines.

8.
J Mol Histol ; 53(4): 623-634, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35870072

ABSTRACT

Differentiated cells can be reprogrammed to embryonic stem cell-like cells called induced pluripotent stem cells (iPSCs), in which the natural developmental differentiation process is reversed. It is unclear whether the multi-lineage cells can be isolated and identified during reprogramming. In the current study, we detected the expression of lineage markers, isolated neural lineages, and identified the related microRNAs during iPSC formation. Our results demonstrated that a neuroectoderm appeared earlier than mesoderm and definitive endoderm before forming colonies when mouse embryonic fibroblasts were subjected to iPSC formation using transcription factors (TFs). On day 3, the cells expressed Sox1 and Nestin and had ultrastructure consistent with the transition to identity neural germ layer lineage. Fluorescence-activated cell sorting analysis revealed a peak (40%) in neural progenitor marker-positive cells. When subsequently cultured in a neural precursor cell medium, these cells proliferated slowly, became round and aggregated, generating into neurons and glia. Genome-wide microRNA (miRNA) analysis identified 45 differentially regulated miRNAs. Molecular network analysis demonstrated that these miRNAs validated 6,047 experimental mRNA targets. The GO functional annotation analysis of mRNA targets revealed that most genes were related to neurogenesis, such as growth cone, neuronal cell body, neuron projection, and cell junction synapse. The network of protein-protein interactions was observed, which demonstrated that key nodes of neural lineage reprogramming-associated targets were Sall1, Foxa2, Nf2, Ctnnb1, Shh, and Bmpr1a. Therefore, these data suggested that TFs can drive the reprogramming of somatic cells towards a pluripotent state via neuroectoderm. Moreover, the neural lineage reprogramming system can address how miRNAs influence their target sites.


Subject(s)
Cellular Reprogramming , MicroRNAs , Animals , Cell Differentiation/genetics , Cellular Reprogramming/genetics , Fibroblasts/metabolism , Mesoderm , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Messenger/metabolism
9.
Eur J Cancer ; 173: 59-70, 2022 09.
Article in English | MEDLINE | ID: mdl-35863107

ABSTRACT

Ovarian immature teratoma is a rare subtype of germ cell tumour that can be pure or associated with non-teratomatous germ cell tumour elements and is graded based on extent of the immature neuroectodermal component. Immature teratoma (IT) can also be associated with somatic differentiation in the form of sarcoma, carcinoma, or extensive immature neuroectodermal elements and may produce low levels of serum alpha-fetoprotein. Variable interpretation of these issues underlies diagnostic and management dilemmas, resulting in substantial practice differences between paediatric and adult women with IT. The Malignant Germ Cell International Consortium (MaGIC) convened oncologists, surgeons, and pathologists to address the following crucial clinicopathologic issues related to IT: (1) grading of IT, (2) definition and significance of 'microscopic' yolk sac tumour, (3) transformation to a somatic malignancy, and (4) interpretation of serum tumour biomarkers. This review highlights the discussion, conclusions, and suggested next steps from this clinicopathologic conference.


Subject(s)
Endodermal Sinus Tumor , Neoplasms, Germ Cell and Embryonal , Ovarian Neoplasms , Teratoma , Adult , Child , Consensus Development Conferences as Topic , Endodermal Sinus Tumor/drug therapy , Endodermal Sinus Tumor/therapy , Female , Humans , Neoplasms, Germ Cell and Embryonal/diagnosis , Neoplasms, Germ Cell and Embryonal/therapy , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/therapy , Teratoma/drug therapy , Teratoma/therapy
10.
Prog Retin Eye Res ; 91: 101093, 2022 11.
Article in English | MEDLINE | ID: mdl-35817658

ABSTRACT

The development of the vertebrate eyes is a complex process starting from anterior-posterior and dorso-ventral patterning of the anterior neural tube, resulting in the formation of the eye field. Symmetrical separation of the eye field at the anterior neural plate is followed by two symmetrical evaginations to generate a pair of optic vesicles. Next, reciprocal invagination of the optic vesicles with surface ectoderm-derived lens placodes generates double-layered optic cups. The inner and outer layers of the optic cups develop into the neural retina and retinal pigment epithelium (RPE), respectively. In vitro produced retinal tissues, called retinal organoids, are formed from human pluripotent stem cells, mimicking major steps of retinal differentiation in vivo. This review article summarizes recent progress in our understanding of early eye development, focusing on the formation the eye field, optic vesicles, and early optic cups. Recent single-cell transcriptomic studies are integrated with classical in vivo genetic and functional studies to uncover a range of cellular mechanisms underlying early eye development. The functions of signal transduction pathways and lineage-specific DNA-binding transcription factors are dissected to explain cell-specific regulatory mechanisms underlying cell fate determination during early eye development. The functions of homeodomain (HD) transcription factors Otx2, Pax6, Lhx2, Six3 and Six6, which are required for early eye development, are discussed in detail. Comprehensive understanding of the mechanisms of early eye development provides insight into the molecular and cellular basis of developmental ocular anomalies, such as optic cup coloboma. Lastly, modeling human development and inherited retinal diseases using stem cell-derived retinal organoids generates opportunities to discover novel therapies for retinal diseases.


Subject(s)
Retinal Diseases , Transcription Factors , Humans , Transcription Factors/genetics , Transcription Factors/metabolism , Gene Expression Regulation, Developmental , Cell Differentiation/physiology , Eye , Retina/metabolism , Signal Transduction , Retinal Diseases/metabolism
11.
Methods Mol Biol ; 2429: 57-72, 2022.
Article in English | MEDLINE | ID: mdl-35507155

ABSTRACT

Human pluripotent stem cells have a wide variety of potential applications, ranging from clinical translation to in vitro disease modeling. However, there is significant variation in the potential of individual cell lines to differentiate towards each of the three germ layers as a result of (epi)genetic background, culture conditions, and other factors. We describe here in detail a methodology to evaluate this bias using short directed differentiation towards neuroectoderm, mesendoderm, and definitive endoderm in combination with quantification by RT-qPCR and immunofluorescent stains.


Subject(s)
Endoderm , Pluripotent Stem Cells , Cell Differentiation , Germ Layers , Humans , Neural Plate
12.
Development ; 149(12)2022 06 15.
Article in English | MEDLINE | ID: mdl-35593440

ABSTRACT

Planar cell polarity (PCP) is the aligned cell polarity within a tissue plane. Mechanical signals are known to act as a global cue for PCP, yet their exact role is still unclear. In this study, we focused on PCP in the posterior neuroectoderm of Xenopus laevis and investigated how mechanical signals regulate polarity. We reveal that the neuroectoderm is under a greater tension in the anterior-posterior direction and that perturbation of this tension causes PCP disappearance. We show that application of uniaxial stretch to explant tissues can control the orientation of PCP and that cells sense the tissue stretch indirectly through a change in their shape, rather than directly through detection of anisotropic tension. Furthermore, we reveal that PCP is most strongly established when the orientation of tissue stretch coincides with that of diffusion of locally expressed Wnt ligands, suggesting a cooperative relationship between these two PCP regulators.


Subject(s)
Cell Polarity , Wnt Signaling Pathway , Animals , Cell Polarity/physiology , Xenopus laevis
14.
J Cell Sci ; 134(24)2021 12 15.
Article in English | MEDLINE | ID: mdl-34806749

ABSTRACT

The orientation of epithelial cells in the plane of the tissue, known as planar cell polarity (PCP), is regulated by interactions of asymmetrically localized PCP protein complexes. In the Xenopus neural plate, Van Gogh-like2 (Vangl2) and Prickle3 (Pk3) proteins form a complex at the anterior cell boundaries, but how this complex is regulated in vivo remains largely unknown. Here, we use proximity biotinylation and crosslinking approaches to show that Vangl2-Pk3 association is inhibited by Frizzled3 (Fz3, also known as Fzd3), a core PCP protein that is specifically expressed in the neuroectoderm and is essential for the establishment of PCP in this tissue. This inhibition required Fz3-dependent Vangl2 phosphorylaton. Consistent with our observations, the complex of Pk3 with nonphosphorylatable Vangl2 did not polarize in the neural plate. These findings provide evidence for in vivo regulation of Vangl2-Pk3 complex formation and localization by a Frizzled receptor.


Subject(s)
Cell Polarity , Frizzled Receptors , Intercellular Signaling Peptides and Proteins , Membrane Proteins , Neural Plate , Xenopus Proteins , Animals , Transcription Factors , Xenopus laevis
15.
Stem Cell Reports ; 16(11): 2642-2658, 2021 11 09.
Article in English | MEDLINE | ID: mdl-34715053

ABSTRACT

p53 alterations occur during culture of pluripotent stem cells (PSCs), but the significance of these events on epigenetic control of PSC fate determination remains poorly understood. Wdr5 deletion in p53-null (DKO) mouse ESCs (mESCs) leads to impaired self-renewal, defective retinal neuroectoderm differentiation, and de-repression of germ cell/meiosis (GCM)-specific genes. Re-introduction of a WDR5 mutant with defective H3K4 methylation activity into DKO ESCs restored self-renewal and suppressed GCM gene expression but failed to induce retinal neuroectoderm differentiation. Mechanistically, mutant WDR5 targets chromatin that is largely devoid of H3K4me3 and regulates gene expression in p53-null mESCs. Furthermore, MAX and WDR5 co-target lineage-specifying chromatin and regulate chromatin accessibility of GCM-related genes. Importantly, MAX and WDR5 are core subunits of a non-canonical polycomb repressor complex 1 responsible for gene silencing. This function, together with canonical, pro-transcriptional WDR5-dependent MLL complex H3K4 methyltransferase activity, highlight how WDR5 mediates crosstalk between transcription and repression during mESC fate choice.


Subject(s)
Cell Differentiation/genetics , Cell Self Renewal/genetics , Histones/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Mouse Embryonic Stem Cells/metabolism , Pluripotent Stem Cells/metabolism , Tumor Suppressor Protein p53/genetics , Animals , Cell Line , Chromatin/genetics , Chromatin/metabolism , Chromatin Immunoprecipitation Sequencing/methods , Gene Expression Profiling/methods , Intracellular Signaling Peptides and Proteins/metabolism , Methylation , Mice , Mice, Knockout , Mice, Transgenic , RNA-Seq/methods , Tumor Suppressor Protein p53/metabolism
16.
Mol Cells ; 44(10): 723-735, 2021 Oct 31.
Article in English | MEDLINE | ID: mdl-34711690

ABSTRACT

Spemann organizer is a center of dorsal mesoderm and itself retains the mesoderm character, but it has a stimulatory role for neighboring ectoderm cells in becoming neuroectoderm in gastrula embryos. Goosecoid (Gsc) overexpression in ventral region promotes secondary axis formation including neural tissues, but the role of gsc in neural specification could be indirect. We examined the neural inhibitory and stimulatory roles of gsc in the same cell and neighboring cells contexts. In the animal cap explant system, Gsc overexpression inhibited expression of neural specific genes including foxd4l1.1, zic3, ncam, and neurod. Genome-wide chromatin immunoprecipitation sequencing (ChIP-seq) and promoter analysis of early neural genes of foxd4l1.1 and zic3 were performed to show that the neural inhibitory mode of gsc was direct. Site-directed mutagenesis and serially deleted construct studies of foxd4l1.1 promoter revealed that Gsc directly binds within the foxd4l1.1 promoter to repress its expression. Conjugation assay of animal cap explants was also performed to demonstrate an indirect neural stimulatory role for gsc. The genes for secretory molecules, Chordin and Noggin, were up-regulated in gsc injected cells with the neural fate only achieved in gsc uninjected neighboring cells. These experiments suggested that gsc regulates neuroectoderm formation negatively when expressed in the same cell and positively in neighboring cells via soluble factors. One is a direct suppressive circuit of neural genes in gsc expressing mesoderm cells and the other is an indirect stimulatory circuit for neurogenesis in neighboring ectoderm cells via secreted BMP antagonizers.


Subject(s)
Goosecoid Protein/metabolism , Neural Plate/embryology , Xenopus Proteins/metabolism , Xenopus laevis/embryology , Animals
17.
Front Cell Dev Biol ; 9: 702462, 2021.
Article in English | MEDLINE | ID: mdl-34568320

ABSTRACT

Epigenetic modifications play a crucial role in neurogenesis, learning, and memory, but the study of their role in early neuroectoderm commitment from pluripotent inner cell mass is relatively lacking. Here we utilized the system of directed neuroectoderm differentiation from human embryonic stem cells and identified that KDM6B, an enzyme responsible to erase H3K27me3, was the most upregulated enzyme of histone methylation during neuroectoderm differentiation by transcriptome analysis. We then constructed KDM6B-null embryonic stem cells and found strikingly that the pluripotent stem cells with KDM6B knockout exhibited much higher neuroectoderm induction efficiency. Furthermore, we constructed a series of embryonic stem cell lines knocking out the other H3K27 demethylase KDM6A, and depleting both KDM6A and KDM6B, respectively. These cell lines together confirmed that KDM6 impeded early neuroectoderm commitment. By RNA-seq, we found that the expression levels of a panel of WNT genes were significantly affected upon depletion of KDM6. Importantly, the result that WNT agonist and antagonist could abolish the differential neuroectoderm induction due to manipulating KDM6 further demonstrated that WNT was the major downstream of KDM6 during early neural induction. Moreover, we found that the chemical GSK-J1, an inhibitor of KDM6, could enhance neuroectoderm induction from both embryonic stem cells and induced pluripotent stem cells. Taken together, our findings not only illustrated the important role of the histone methylation modifier KDM6 in early neurogenesis, providing insights into the precise epigenetic regulation in cell fate determination, but also showed that the inhibitor of KDM6 could facilitate neuroectoderm differentiation from human pluripotent stem cells.

18.
Biol Open ; 10(7)2021 07 15.
Article in English | MEDLINE | ID: mdl-34259326

ABSTRACT

Coordinated polarization of cells in the tissue plane, known as planar cell polarity (PCP), is associated with a signaling pathway critical for the control of morphogenetic processes. Although the segregation of PCP components to opposite cell borders is believed to play a critical role in this pathway, whether PCP derives from egg polarity or preexistent long-range gradient, or forms in response to a localized cue, remains a challenging question. Here we investigate the Xenopus neural plate, a tissue that has been previously shown to exhibit PCP. By imaging Vangl2 and Prickle3, we show that PCP is progressively acquired in the neural plate and requires a signal from the posterior region of the embryo. Tissue transplantations indicated that PCP is triggered in the neural plate by a planar cue from the dorsal blastopore lip. The PCP cue did not depend on the orientation of the graft and was distinct from neural inducers. These observations suggest that neuroectodermal PCP is not instructed by a preexisting molecular gradient but induced by a signal from the dorsal blastopore lip.


Subject(s)
Cell Polarity/physiology , Gastrula/embryology , Morphogenesis/physiology , Neural Plate/embryology , Xenopus/embryology , Animals , Signal Transduction
19.
Curr Top Dev Biol ; 145: 41-60, 2021.
Article in English | MEDLINE | ID: mdl-34074535

ABSTRACT

Planar cell polarity (PCP) refers to the coordinated polarization of cells within the plane of a tissue. PCP is a controlled by a group of conserved proteins organized in a specific signaling pathway known as the PCP pathway. A hallmark of PCP signaling is the asymmetric localization of "core" PCP protein complexes at the cell cortex, although endogenous PCP cues needed to establish this asymmetry remain unknown. While the PCP pathway was originally discovered as a mechanism directing the planar organization of Drosophila epithelial tissues, subsequent studies in Xenopus and other vertebrates demonstrated a critical role for this pathway in the regulation of actomyosin-dependent morphogenetic processes, such as neural tube closure. Large size and external development of amphibian embryos allows live cell imaging, placing Xenopus among the best models of vertebrate neurulation at the molecular, cellular and organismal level. This review describes cross-talk between core PCP proteins and actomyosin contractility that ultimately leads to tissue-scale movement during neural tube closure.


Subject(s)
Actomyosin/metabolism , Cell Polarity , Models, Animal , Neural Tube/embryology , Neurulation , Xenopus laevis/embryology , Animals , Humans
20.
Cell Mol Life Sci ; 78(9): 4435-4450, 2021 May.
Article in English | MEDLINE | ID: mdl-33796894

ABSTRACT

During early neural development, changes in signalling inform the expression of transcription factors that in turn instruct changes in cell identity. At the same time, switches in adhesion molecule expression result in cellular rearrangements that define the morphology of the emerging neural tube. It is becoming increasingly clear that these two processes influence each other; adhesion molecules do not simply operate downstream of or in parallel with changes in cell identity but rather actively feed into cell fate decisions. Why are differentiation and adhesion so tightly linked? It is now over 60 years since Conrad Waddington noted the remarkable "Constancy of the Wild Type" (Waddington in Nature 183: 1654-1655, 1959) yet we still do not fully understand the mechanisms that make development so reproducible. Conversely, we do not understand why directed differentiation of cells in a dish is sometimes unpredictable and difficult to control. It has long been suggested that cells make decisions as 'local cooperatives' rather than as individuals (Gurdon in Nature 336: 772-774, 1988; Lander in Cell 144: 955-969, 2011). Given that the cadherin family of adhesion molecules can simultaneously influence morphogenesis and signalling, it is tempting to speculate that they may help coordinate cell fate decisions between neighbouring cells in the embryo to ensure fidelity of patterning, and that the uncoupling of these processes in a culture dish might underlie some of the problems with controlling cell fate decisions ex-vivo. Here we review the expression and function of cadherins during early neural development and discuss how and why they might modulate signalling and differentiation as neural tissues are formed.


Subject(s)
Cadherins/metabolism , Neural Crest/metabolism , Animals , Cadherins/chemistry , Cadherins/classification , Cell Adhesion , Cell Differentiation , Evolution, Molecular , Humans , Neural Crest/cytology , Neural Crest/growth & development , Neurogenesis , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...