Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
J Cell Biol ; 220(6)2021 06 07.
Article in English | MEDLINE | ID: mdl-33798261

ABSTRACT

EPH/EPHRIN signaling is essential to many aspects of tissue self-organization and morphogenesis, but little is known about how EPH/EPHRIN signaling regulates cell mechanics during these processes. Here, we use a series of approaches to examine how EPH/EPHRIN signaling drives cellular self-organization. Contact angle measurements reveal that EPH/EPHRIN signaling decreases the stability of heterotypic cell:cell contacts through increased cortical actomyosin contractility. We find that EPH/EPHRIN-driven cell segregation depends on actomyosin contractility but occurs independently of directed cell migration and without changes in cell adhesion. Atomic force microscopy and live cell imaging of myosin localization support that EPH/EPHRIN signaling results in increased cortical tension. Interestingly, actomyosin contractility also nonautonomously drives increased EPHB2:EPHB2 homotypic contacts. Finally, we demonstrate that changes in tissue organization are driven by minimization of heterotypic contacts through actomyosin contractility in cell aggregates and by mouse genetics experiments. These data elucidate the biomechanical mechanisms driving EPH/EPHRIN-based cell segregation wherein differences in interfacial tension, regulated by actomyosin contractility, govern cellular self-organization.


Subject(s)
Actin Cytoskeleton/physiology , Actomyosin/physiology , Cell Adhesion , Cell Movement , Ephrins/metabolism , Receptors, Eph Family/metabolism , Animals , Ephrins/genetics , HEK293 Cells , Humans , Mice , Morphogenesis , Protein Binding , Receptors, Eph Family/genetics , Signal Transduction
2.
Genesis ; 57(1): e23271, 2019 01.
Article in English | MEDLINE | ID: mdl-30548771

ABSTRACT

Craniofacial morphogenesis is a highly dynamic process that requires changes in the behaviors and physical properties of cells in order to achieve the proper organization of different craniofacial structures. Boundary formation is a critical process in cellular organization, patterning, and ultimately tissue separation. There are several recurring cellular mechanisms through which boundary formation and cellular organization occur including, transcriptional patterning, cell segregation, cell adhesion and migratory guidance. Disruption of normal boundary formation has dramatic morphological consequences, and can result in human craniofacial congenital anomalies. In this review we discuss boundary formation during craniofacial development, specifically focusing on the cellular behaviors and mechanisms underlying the self-organizing properties that are critical for craniofacial morphogenesis.


Subject(s)
Morphogenesis , Neural Crest/embryology , Skull/embryology , Animals , Cell Differentiation , Humans
3.
Neuropsychopharmacology ; 42(3): 774-784, 2017 02.
Article in English | MEDLINE | ID: mdl-27534267

ABSTRACT

Because of unavoidable confounding variables in the direct study of human subjects, it has been difficult to unravel the effects of prenatal cocaine exposure on the human fetal brain, as well as the cellular and biochemical mechanisms involved. Here, we propose a novel approach using a human pluripotent stem cell (hPSC)-based 3D neocortical organoid model. This model retains essential features of human neocortical development by encompassing a single self-organized neocortical structure, without including an animal-derived gelatinous matrix. We reported previously that prenatal cocaine exposure to rats during the most active period of neural progenitor proliferation induces cytoarchitectural changes in the embryonic neocortex. We also identified a role of CYP450 and consequent oxidative ER stress signaling in these effects. However, because of differences between humans and rodents in neocorticogenesis and brain CYP metabolism, translation of the research findings from the rodent model to human brain development is uncertain. Using hPSC 3D neocortical organoids, we demonstrate that the effects of cocaine are mediated through CYP3A5-induced generation of reactive oxygen species, inhibition of neocortical progenitor cell proliferation, induction of premature neuronal differentiation, and interruption of neural tissue development. Furthermore, knockdown of CYP3A5 reversed these cocaine-induced pathological phenotypes, suggesting CYP3A5 as a therapeutic target to mitigate the deleterious neurodevelopmental effects of prenatal cocaine exposure in humans. Moreover, 3D organoid methodology provides an innovative platform for identifying adverse effects of abused psychostimulants and pharmaceutical agents, and can be adapted for use in neurodevelopmental disorders with genetic etiologies.


Subject(s)
Cocaine/pharmacology , Cytochrome P-450 CYP3A/metabolism , Dopamine Uptake Inhibitors/pharmacology , Neocortex/drug effects , Neurogenesis/drug effects , Pluripotent Stem Cells/drug effects , Cell Line , Humans
4.
J Cell Biol ; 215(2): 217-229, 2016 Oct 24.
Article in English | MEDLINE | ID: mdl-27810913

ABSTRACT

Cell segregation is the process by which cells self-organize to establish developmental boundaries, an essential step in tissue formation. Cell segregation is a common outcome of Eph/ephrin signaling, but the mechanisms remain unclear. In craniofrontonasal syndrome, X-linked mosaicism for ephrin-B1 expression has been hypothesized to lead to aberrant Eph/ephrin-mediated cell segregation. Here, we use mouse genetics to exploit mosaicism to study cell segregation in the mammalian embryo and integrate live-cell imaging to examine the underlying cellular and molecular mechanisms. Our data demonstrate that dramatic ephrin-B1-mediated cell segregation occurs in the early neuroepithelium. In contrast to the paradigm that repulsive bidirectional signaling drives cell segregation, unidirectional EphB kinase signaling leads to cell sorting by the Rho kinase-dependent generation of a cortical actin differential between ephrin-B1- and EphB-expressing cells. These results define mechanisms of Eph/ephrin-mediated cell segregation, implicating unidirectional regulation of cortical actomyosin contractility as a key effector of this fundamental process.


Subject(s)
Actomyosin/metabolism , Cell Differentiation , Ephrin-B1/metabolism , Neuroepithelial Cells/cytology , Receptors, Eph Family/metabolism , Signal Transduction , Actins/metabolism , Animals , Cell Count , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Mice , Models, Biological , Neuroepithelial Cells/metabolism , rho-Associated Kinases/metabolism
5.
Cell Rep ; 10(4): 616-32, 2015 Feb 03.
Article in English | MEDLINE | ID: mdl-25640183

ABSTRACT

Human pluripotent stem cell (hPSC) lines exhibit repeated patterns of genetic variation, which can alter in vitro properties as well as suitability for clinical use. We examined associations between copy-number variations (CNVs) on chromosome 17 and hPSC mesodiencephalic dopaminergic (mDA) differentiation. Among 24 hPSC lines, two karyotypically normal lines, BG03 and CT3, and BG01V2, with trisomy 17, exhibited amplification of the WNT3/WNT9B region and rapid mDA differentiation. In hPSC lines with amplified WNT3/WNT9B, basic fibroblast growth factor (bFGF) signaling through mitogen-activated protein kinase (MAPK)/ERK amplifies canonical WNT signaling by phosphorylating LRP6, resulting in enhanced undifferentiated proliferation. When bFGF is absent, noncanonical WNT signaling becomes dominant due to upregulation of SIAH2, enhancing JNK signaling and promoting loss of pluripotency. When bFGF is present during mDA differentiation, stabilization of canonical WNT signaling causes upregulation of LMX1A and mDA induction. Therefore, CNVs in 17q21.31, a "hot spot" for genetic variation, have multiple and complex effects on hPSC cellular phenotype.


Subject(s)
Neurons/cytology , Neurons/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Wnt Proteins/metabolism , Wnt3 Protein/metabolism , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Line , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Signal Transduction , Wnt Proteins/genetics , Wnt3 Protein/genetics
6.
Dis Model Mech ; 7(12): 1397-405, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25288682

ABSTRACT

Neocortical development involves ordered specification of forebrain cortical progenitors to various neuronal subtypes, ultimately forming the layered cortical structure. Modeling of this process using human pluripotent stem cells (hPSCs) would enable mechanistic studies of human neocortical development, while providing new avenues for exploration of developmental neocortical abnormalities. Here, we show that preserving hPSCs aggregates - allowing embryoid body formation - while adding basic fibroblast growth factor (bFGF) during neuroepithelial development generates neural rosettes showing dorsal forebrain identity, including Mash1(+) dorsal telencephalic GABAergic progenitors. Structures that mirrored the organization of the cerebral cortex formed after rosettes were seeded and cultured for 3 weeks in the presence of FGF18, BDNF and NT3. Neurons migrated along radial glia scaffolding, with deep-layer CTIP2(+) cortical neurons appearing after 1 week and upper-layer SATB2(+) cortical neurons forming during the second and third weeks. At the end of differentiation, these structures contained both glutamatergic and GABAergic neurons, with glutamatergic neurons being most abundant. Thus, this differentiation protocol generated an hPSC-based model that exhibits temporal patterning and a neuronal subtype ratio similar to that of the developing human neocortex. This model was used to examine the effects of cocaine during neocorticogenesis. Cocaine caused premature neuronal differentiation and enhanced neurogenesis of various cortical neuronal subtypes. These cocaine-induced changes were inhibited by the cytochrome P450 inhibitor cimetidine. This in vitro model enables mechanistic studies of neocorticogenesis, and can be used to examine the mechanisms through which cocaine alters the development of the human neocortex.


Subject(s)
Cerebral Cortex/embryology , Cocaine/chemistry , Pluripotent Stem Cells/cytology , Cell Differentiation , Cell Line , Dopamine Uptake Inhibitors/chemistry , Fibroblast Growth Factor 2/metabolism , Humans , Immunohistochemistry , Neurogenesis , Neurons/metabolism , Prosencephalon/embryology , Reactive Oxygen Species , Stem Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...