Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Bioessays ; 46(8): e2300245, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38778437

RESUMEN

Entosis, a form of cell cannibalism, is a newly discovered pathogenic mechanism leading to the development of small brains, termed microcephaly, in which P53 activation was found to play a major role. Microcephaly with entosis, found in Pals1 mutant mice, displays P53 activation that promotes entosis and apoptotic cell death. This previously unappreciated pathogenic mechanism represents a novel cellular dynamic in dividing cortical progenitors which is responsible for cell loss. To date, various recent models of microcephaly have bolstered the importance of P53 activation in cell death leading to microcephaly. P53 activation caused by mitotic delay or DNA damage manifests apoptotic cell death which can be suppressed by P53 removal in these animal models. Such genetic studies attest P53 activation as quality control meant to eliminate genomically unfit cells with minimal involvement in the actual function of microcephaly associated genes. In this review, we summarize the known role of P53 activation in a variety of microcephaly models and introduce a novel mechanism wherein entotic cell cannibalism in neural progenitors is triggered by P53 activation.


Asunto(s)
Apoptosis , Entosis , Microcefalia , Proteína p53 Supresora de Tumor , Microcefalia/genética , Microcefalia/metabolismo , Microcefalia/patología , Proteína p53 Supresora de Tumor/metabolismo , Proteína p53 Supresora de Tumor/genética , Animales , Humanos , Ratones , Modelos Animales de Enfermedad
2.
Dev Biol ; 2024 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-39284539

RESUMEN

Combined removal of Crb1 and Crb2 from the developing optic vesicle evokes cellular and laminar disorganization by disrupting the apical cell-cell adhesion in developing retinal epithelium. As a result, at postnatal stages, affected mouse retinas show temporarily thickened, coarsely laminated retinas in addition to functional deficits, including a severely abnormal electroretinogram and decreased visual acuity. These features are reminiscent of Leber congenital amaurosis 8, which is caused in humans by subsets of Crb1 mutations. However, the cellular basis of the abnormalities in retinal progenitor cells (RPCs) that lead to retinal disorganization is largely unknown. In this study, we analyze specific features of RPCs in mutant retinas, including maintenance of the progenitor pool, cell cycle progression, cell cycle phase-dependent nuclear positioning, cell survival, and generation of mature retinal cell types. We find crucial defects in the mutant RPCs. Upon removal of CRB1 and CRB2, apical structures of the RPCs, determined by markers of cilia and centrosomes, are basally shifted. In addition, the positioning of the somata of the M-phase cells, normally localized at the apical surface of the retinal epithelium, is basally shifted in a nearly randomized pattern along the apico-basal axis. Consequently, we propose that positioning of RPCs is desynchronized from cell cycle phase and largely randomized during embryonic development at E17.5. Because the resultant postmitotic cells inevitably lose positional information, the outer and inner nuclear layers (ONL and INL) fail to form from ONBL during neonatal development and retinal cells become mixed locally and globally. Additional results of the lost tissue polarity in Crb1/Crb2 dKO retinas include atypical formation of heterotopic cell patches containing photoreceptor cells in the ganglion cell layer and acellular patches filled with neural processes. Collectively, these changes lead to a mouse model of LCA8-like pathology. LCA8-like pathology differs substantially from the well-characterized, broad range of degeneration phenotypes that arise during the differentiation of photoreceptor and Muller glial cells in retinitis pigmentosa 12, a closely related disease caused by mutated human Crb1. Importantly, the present results suggest that Crb1/Crb2 serve indispensable functions in maintaining cell-cycle phase-dependent positioning of RPCs along the apico-basal axis, regulating cell cycle progression, and maintaining structural laminar integrity without significantly affecting the size of the RPC pools, generation of the subsets of the retinal cell types, or the distribution of cell cycle phases during RPC division. Taken together, these findings provide the crucial cellular basis of the thickening and severely disorganized lamination that are the unique features of the retinal abnormalities in LCA8 patients.

3.
Hum Mol Genet ; 31(12): 1979-1996, 2022 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-34999833

RESUMEN

The multi-systemic genetic disorder tuberous sclerosis complex (TSC) impacts multiple neurodevelopmental processes including neuronal morphogenesis, neuronal migration, myelination and gliogenesis. These alterations contribute to the development of cerebral cortex abnormalities and malformations. Although TSC is caused by mTORC1 hyperactivation, cognitive and behavioral impairments are not improved through mTORC1 targeting, making the study of the downstream effectors of this complex important for understanding the mechanisms underlying TSC. As mTORC1 has been shown to promote the activity of the transcriptional co-activator Yap, we hypothesized that altered Yap/Taz signaling contributes to the pathogenesis of TSC. We first observed that the levels of Yap/Taz are increased in human cortical tuber samples and in embryonic cortices of Tsc2 conditional knockout (cKO) mice. Next, to determine how abnormal upregulation of Yap/Taz impacts the neuropathology of TSC, we deleted Yap/Taz in Tsc2 cKO mice. Importantly, Yap/Taz/Tsc2 triple conditional knockout (tcKO) animals show reduced cortical thickness and cortical neuron cell size, despite the persistence of high mTORC1 activity, suggesting that Yap/Taz play a downstream role in cytomegaly. Furthermore, Yap/Taz/Tsc2 tcKO significantly restored cortical and hippocampal lamination defects and reduced hippocampal heterotopia formation. Finally, the loss of Yap/Taz increased the distribution of myelin basic protein in Tsc2 cKO animals, consistent with an improvement in myelination. Overall, our results indicate that targeting Yap/Taz lessens the severity of neuropathology in a TSC animal model. This study is the first to implicate Yap/Taz as contributors to cortical pathogenesis in TSC and therefore as potential novel targets in the treatment of this disorder.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Esclerosis Tuberosa , Proteínas Señalizadoras YAP , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones , Neuronas/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Esclerosis Tuberosa/patología , Proteína 1 del Complejo de la Esclerosis Tuberosa/genética , Proteína 1 del Complejo de la Esclerosis Tuberosa/metabolismo , Proteínas Señalizadoras YAP/genética
4.
Dev Dyn ; 251(7): 1094-1106, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35150033

RESUMEN

Mutations in the CRB1 (Crumbs homolog 1) cause rare retinal diseases like retinitis pigmentosa type 12 (RP12) and Leber congenital amaurosis type 8 (LCA8). RP12 results in progressively worsening peripheral vision, whereas LCA8 causes severe visual impairment at birth or in early life. While several mouse models have been proposed for RP12, few replicate the full spectrum of human LCA8 pathology, such as disorganized retinal layering, abnormal retinal thickening, pigmentary defects, hyperreflective lesions, and severely attenuated electroretinogram responses at birth. Six models have been proposed utilizing the Cre-loxP system to delete candidate genes in specific retinal cell types and developmental stages. The model ablating Crb1 and its homolog Crb2 (using mRx-Cre) from the beginning of the eye development is the most complete as it shows blindness during the eye-opening stage, pigmentary defects in the RPE, ganglion cell layer heterotopia, disruption of retinal lamination, and acellular patches. LCA8 represents a unique type of retinal dystrophy among LCA subtypes, driven by dysfunctional retinal progenitor cells during eye development. In contrast, other LCA types and RP12 are caused by photoreceptor defects. Therefore, the most accurate LCA8-like mouse model must target both alleles of the Crb1 and Crb2 genes in the optic vesicle or earlier.


Asunto(s)
Amaurosis Congénita de Leber , Retinitis Pigmentosa , Animales , Modelos Animales de Enfermedad , Proteínas del Ojo/genética , Amaurosis Congénita de Leber/genética , Amaurosis Congénita de Leber/patología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Mutación , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Retina/metabolismo , Retinitis Pigmentosa/genética
5.
Hum Mol Genet ; 29(20): 3388-3401, 2020 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-33073849

RESUMEN

Membrane Protein Palmitoylated 5 (MPP5) is a highly conserved apical complex protein essential for cell polarity, fate and survival. Defects in cell polarity are associated with neurologic disorders including autism and microcephaly. MPP5 is essential for neurogenesis in animal models, but human variants leading to neurologic impairment have not been described. We identified three patients with heterozygous MPP5 de novo variants (DNV) and global developmental delay (GDD) and compared their phenotypes and magnetic resonance imaging (MRI) to ascertain how MPP5 DNV leads to GDD. All three patients with MPP5 DNV experienced GDD with language delay/regression and behavioral changes. MRI ranged from normal to decreased gyral folding and microcephaly. The effects of MPP5 depletion on the developing brain were assessed by creating a heterozygous conditional knock out (het CKO) murine model with central nervous system (CNS)-specific Nestin-Cre drivers. In the het CKO model, Mpp5 depletion led to microcephaly, decreased cerebellar volume and cortical thickness. Het CKO mice had decreased ependymal cells and Mpp5 at the apical surface of cortical ventricular zone compared with wild type. Het CKO mice also failed to maintain progenitor pools essential for neurogenesis. The proportion of cortical cells undergoing apoptotic cell death increased, suggesting that cell death reduces progenitor population and neuron number. Het CKO mice also showed behavioral changes, similar to our patients. To our knowledge, this is the first report to show that variants in MPP5 are associated with GDD, behavioral abnormalities and language regression/delay. Murine modeling shows that neurogenesis is likely altered in these individuals, with cell death and skewed cellular composition playing significant roles.


Asunto(s)
Discapacidades del Desarrollo/etiología , Proteínas de la Membrana/genética , Proteínas de la Membrana/fisiología , Mutación , Enfermedades del Sistema Nervioso/etiología , Nucleósido-Fosfato Quinasa/genética , Nucleósido-Fosfato Quinasa/fisiología , Adolescente , Adulto , Animales , Niño , Discapacidades del Desarrollo/metabolismo , Discapacidades del Desarrollo/patología , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Enfermedades del Sistema Nervioso/metabolismo , Enfermedades del Sistema Nervioso/patología , Adulto Joven
6.
Dev Biol ; 457(1): 150-162, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31586559

RESUMEN

Yap/Taz are well-established downstream effectors of the Hippo pathway, known to regulate organ size by directing proliferation and apoptosis. Although the functions of Yap/Taz have been extensively studied, little is known about their role in brain development. Here, through genetic ablation, we show that Yap/Taz are required for cerebellar morphogenesis. Yap/Taz deletion in neural progenitors causes defects in secondary fissure formation, leading to abnormal folia development. Although they seemed very likely to serve an important function in the development of cerebellar granule cell precursors, Yap/Taz are dispensable for their proliferation. Furthermore, Yap/Taz loss does not rescue the medulloblastoma phenotype caused by constitutively active Smoothened. Importantly, Yap/Taz are highly expressed in radial glia and play a crucial role in establishing the radial scaffold and cellular polarity of neural progenitors during embryogenesis. We found that Yap/Taz are necessary to establish and maintain junctional integrity of cerebellar neuroepithelium as prominent junction proteins are not maintained at the apical junction in the absence of Yap/Taz. Our study identifies a novel function of Yap/Taz in cerebellar foliation and finds that they are required to establish the radial glia scaffold and junctional stability.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de Ciclo Celular/metabolismo , Cerebelo/embriología , Organogénesis , Transactivadores/metabolismo , Animales , Proliferación Celular , Cerebelo/citología , Cerebelo/metabolismo , Células Ependimogliales , Ratones , Tamaño de los Órganos , Células Madre/metabolismo , Proteínas Señalizadoras YAP
7.
Hum Mol Genet ; 28(11): 1822-1836, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-30668728

RESUMEN

BUB-related 1 (BubR1) encoded by Budding Uninhibited by Benzimidazole 1B (BUB1B) is a crucial mitotic checkpoint protein ensuring proper segregation of chromosomes during mitosis. Mutations of BUB1B are responsible for mosaic variegated aneuploidy (MVA), a human congenital disorder characterized by extensive abnormalities in chromosome number. Although microcephaly is a prominent feature of MVA carrying the BUB1B mutation, how BubR1 deficiency disturbs neural progenitor proliferation and neuronal output and leads to microcephaly is unknown. Here we show that conditional loss of BubR1 in mouse cerebral cortex recapitulates microcephaly. BubR1-deficient cortex includes a strikingly reduced number of late-born, but not of early-born, neurons, although BubR1 expression is substantially reduced from an early stage. Importantly, absence of BubR1 decreases the proportion of neural progenitors in mitosis, specifically in metaphase, suggesting shortened mitosis owing to premature chromosome segregation. In the BubR1 mutant, massive apoptotic cell death, which is likely due to the compromised genomic integrity that results from aberrant mitosis, depletes progenitors and neurons during neurogenesis. There is no apparent alteration in centrosome number, spindle formation or primary cilia, suggesting that the major effect of BubR1 deficiency on neural progenitors is to impair the mitotic checkpoint. This finding highlights the importance of the mitotic checkpoint in the pathogenesis of microcephaly. Furthermore, the ependymal cell layer does not form in the conditional knockout, revealing an unrecognized role of BubR1 in assuring the integrity of the ventricular system, which may account for the presence of hydrocephalus in some patients.


Asunto(s)
Proteínas de Ciclo Celular/genética , Microcefalia/genética , Mitosis/genética , Neurogénesis/genética , Proteínas Serina-Treonina Quinasas/genética , Alelos , Animales , Apoptosis/genética , Proteínas de Ciclo Celular/deficiencia , Proliferación Celular/genética , Corteza Cerebral/crecimiento & desarrollo , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Trastornos de los Cromosomas/genética , Trastornos de los Cromosomas/fisiopatología , Segregación Cromosómica/genética , Modelos Animales de Enfermedad , Humanos , Ratones , Microcefalia/metabolismo , Microcefalia/fisiopatología , Mosaicismo , Mutación/genética , Neuronas/metabolismo , Neuronas/patología , Proteínas Serina-Treonina Quinasas/deficiencia , Huso Acromático/genética , Huso Acromático/patología
8.
Dev Biol ; 453(2): 141-154, 2019 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-31145883

RESUMEN

The Crb1 and 2 (Crumbs homolog 1 & 2) genes encode large, single-pass transmembrane proteins essential for the apicobasal polarity and adhesion of epithelial cells. Crb1 mutations cause degenerative retinal diseases in humans, including Leber congenital amaurosis type 8 (LCA8) and retinitis pigmentosa type 12 (RP12). In LCA8, impaired photoreceptor development and/or survival is thought to cause blindness during early infancy, whereas, in RP12, progressive photoreceptor degeneration damages peripheral vision later in life. There are multiple animal models of RP12 pathology, but no experimental model of LCA8 recapitulates the full spectrum of its pathological features. To generate a mouse model of LCA8 and identify the functions of Crb1/2 in developing ocular tissues, we used an mRx-Cre driver to generate allelic combinations that enabled conditional gene ablation from the optic vesicle stage. In this series only Crb1/2 double knockout (dKO) mice exhibited characteristics of human LCA8 disease: locally thickened retina with spots devoid of cells, aberrant positioning of retinal cells, severely disrupted lamination, and depigmented retinal-pigmented epithelium. Retinal defects antedated E12.5, which is far earlier than the stage at which photoreceptor cells mainly differentiate. Most remarkably, Crb1/Crb2 dKO showed a severely attenuated electroretinogram at the eye opening stage. These results suggest that human LCA8 can be modeled in the mouse by simultaneously ablating Crb1/2 from the beginning of eye development. Importantly, they also indicate that LCA8 is caused by malfunction of retinal progenitor cells during early ocular development rather than by defective photoreceptor-Muller glial interaction, a mechanism proposed for RP12.


Asunto(s)
Ojo/metabolismo , Ojo/patología , Eliminación de Gen , Amaurosis Congénita de Leber/genética , Proteínas de la Membrana/genética , Proteínas del Tejido Nervioso/genética , Adulto , Animales , Modelos Animales de Enfermedad , Electrorretinografía , Humanos , Amaurosis Congénita de Leber/patología , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación/genética , Proteínas del Tejido Nervioso/metabolismo , Especificidad de Órganos , Células Fotorreceptoras de Vertebrados/metabolismo , Pigmentación , Epitelio Pigmentado de la Retina/metabolismo , Epitelio Pigmentado de la Retina/patología
9.
Development ; 143(1): 133-46, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26657772

RESUMEN

Through their biased localization and function within the cell, polarity complex proteins are necessary to establish the cellular asymmetry required for tissue organization. Well-characterized germinal zones, mitogenic signals and cell types make the cerebellum an excellent model for addressing the crucial function of polarity complex proteins in the generation and organization of neural tissues. Deletion of the apical polarity complex protein Pals1 in the developing cerebellum results in a remarkably undersized cerebellum with disrupted layers in poorly formed folia and strikingly reduced granule cell production. We demonstrate that Pals1 is not only essential for cerebellum organogenesis, but also for preventing premature differentiation and thus maintaining progenitor pools in cerebellar germinal zones, including cerebellar granule neuron precursors in the external granule layer. In the Pals1 mouse mutants, the expression of genes that regulate the cell cycle was diminished, correlating with the loss of the proliferating cell population of germinal zones. Furthermore, enhanced Shh signaling through activated Smo cannot overcome impaired cerebellar cell generation, arguing for an epistatic role of Pals1 in proliferation capacity. Our study identifies Pals1 as a novel intrinsic factor that regulates the generation of cerebellar cells and Pals1 deficiency as a potential inhibitor of overactive mitogenic signaling.


Asunto(s)
Proliferación Celular/genética , Cerebelo/embriología , Proteínas de la Membrana/metabolismo , Neurogénesis/fisiología , Nucleósido-Fosfato Quinasa/metabolismo , Células Madre/citología , Animales , Ciclo Celular/genética , Cerebelo/citología , Regulación del Desarrollo de la Expresión Génica , Proteínas Hedgehog/metabolismo , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Neuroglía/citología , Nucleósido-Fosfato Quinasa/genética , Organogénesis/genética , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/genética , Receptor Smoothened
10.
Dev Dyn ; 247(6): 794-806, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29532607

RESUMEN

The Hippo-Yes associated protein (Yap) pathway plays an important role in organ size control by regulating cell proliferation, apoptosis, and stem cell renewal. Hippo-Yap signaling also functions at the level of cellular development in a variety of organs through its effects on cell cycle control, cell survival, cell polarity, and cell fate. Because of its important roles in normal development and homeostasis, abnormal regulation of this pathway has been shown to lead to pathological outcomes such as tissue overgrowth, tumor formation, and abnormal organogenesis, including ocular-specific disorders. In this review, we summarize how normal and perturbed control of Yap signaling is implicated in ocular development and disease Developmental Dynamics 247:794-806, 2018. © 2018 Wiley Periodicals, Inc.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Oftalmopatías/metabolismo , Fosfoproteínas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal/fisiología , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Polaridad Celular/genética , Polaridad Celular/fisiología , Oftalmopatías/genética , Vía de Señalización Hippo , Humanos , Fosfoproteínas/genética , Proteínas Serina-Treonina Quinasas/genética , Factores de Transcripción , Proteínas Señalizadoras YAP
11.
Dev Biol ; 419(2): 336-347, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27616714

RESUMEN

Yap functions as a transcriptional regulator by acting together with sequence-specific DNA binding factors and transcription cofactors to mediate cell proliferation in developing epithelial tissues and tumors. An upstream kinase cascade controls nuclear localization and function in response to partially identified exogenous signals, including cell-to-cell contact. Nevertheless, its role in CNS development is poorly understood. In order to investigate Yap function in developing CNS, we characterized the cellular outcomes after selective Yap gene ablation in developing ocular tissues. When Yap was lost, presumptive retinal pigment epithelium acquired anatomical and molecular characteristics resembling those of the retinal epithelium rather than of RPE, including loss of pigmentation, pseudostratified epithelial morphology and ectopic induction of markers for retinal progenitor cells, like Chx10, and neurons, like ß-Tubulin III. In addition, developing retina showed signs of progressive degeneration, including laminar folding, thinning and cell loss, which resulted from multiple defects in cell proliferation and survival, and in junction integrity. Furthermore, Yap-deficient retinal progenitors displayed decreased S-phase cells and altered cell cycle progression. Altogether, our studies not only illustrate the canonical function of Yap in promoting the proliferation of progenitors, but also shed new light on its evolutionarily conserved, instructive role in regional specification, maintenance of junctional integrity and precise regulation of cell proliferation during neuroepithelial development.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Proteínas del Ojo/fisiología , Ojo/embriología , Fosfoproteínas/fisiología , Epitelio Pigmentado de la Retina/citología , Proteínas Adaptadoras Transductoras de Señales/biosíntesis , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Adhesión Celular , Ciclo Celular/fisiología , Proteínas de Ciclo Celular , División Celular , Linaje de la Célula , Polaridad Celular , Transdiferenciación Celular , Ojo/metabolismo , Proteínas del Ojo/biosíntesis , Proteínas del Ojo/genética , Regulación del Desarrollo de la Expresión Génica , Técnicas de Inactivación de Genes , Proteínas de Homeodominio/análisis , Ratones , Microscopía Fluorescente , Placa Neural/citología , Placa Neural/metabolismo , Orgánulos/metabolismo , Fosfoproteínas/biosíntesis , Fosfoproteínas/genética , Retina/embriología , Epitelio Pigmentado de la Retina/metabolismo , Células Madre/citología , Factores de Transcripción/análisis , Proteínas Señalizadoras YAP
12.
BMC Ophthalmol ; 16(1): 193, 2016 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-27809828

RESUMEN

BACKGROUND: Mutations of Crb1 gene cause irreversible and incurable visual impairment in humans. This study aims to use an LCA8-like mouse model to identify host-mediated responses that might interfere with survival, retinal integration and differentiation of grafted cells during neonatal cell therapy. METHODS: Mixed retinal donor cells (1 ~ 2 × 104) isolated from neural retinas of neonatal eGFP transgenic mice were injected into the subretinal space of LCA8-like model neonatal mice. Markers of specific cell types were used to analyze microglial attraction, CSPG induction and retinal cell differentiation. The positions of host retinal cells were traced according to their laminar location during disease progression to look for host cell rearrangements that might inhibit retinal integration of the transplanted cells. RESULTS: Transplanted retinal cells showed poor survival and attracted microglial cells, but CSPG was not greatly induced. Retinas of the LCA8 model hosts underwent significant cellular rearrangement, including rosette formation and apical displacement of inner retinal cells. CONCLUSIONS: Local disease environment, particularly host immune responses to injected cells and formation of a physical barrier caused by apical migration of host retinal cells upon disruption of outer limiting membrane, may impose two major barriers in LCAs cell transplantation therapy.


Asunto(s)
Trasplante de Células/métodos , Retina/citología , Degeneración Retiniana/cirugía , Animales , Supervivencia Celular , Modelos Animales de Enfermedad , Ratones , Ratones Transgénicos , Retina/patología , Degeneración Retiniana/patología
13.
Dev Biol ; 386(2): 281-90, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-24384391

RESUMEN

Hippo-Yap signaling has been implicated in organ size determination via its regulation of cell proliferation, growth and apoptosis (Pan, 2007). The vertebrate lens comprises only two major cell types, lens progenitors and differentiated fiber cells, thereby providing a relatively simple system for studying size-controlling mechanisms. In order to investigate the role of Hippo-Yap signaling in lens size regulation, we conditionally ablated Yap in the developing mouse lens. Lens progenitor-specific deletion of Yap led to near obliteration of the lens primarily due to hypocellularity in the lens epithelium (LE) and accompanying lens fiber (LF) defects. A significantly reduced LE progenitor pool resulted mainly from failed self-renewal and increased apoptosis. Additionally, Yap-deficient lens progenitor cells precociously exited the cell cycle and expressed the LF marker, ß-Crystallin. The mutant progenitor cells also exhibited multiple cellular and subcellular alterations including cell and nuclear shape change, organellar polarity disruption, and disorganized apical polarity complex and junction proteins such as Crumbs, Pals1, Par3 and ZO-1. Yap-deficient LF cells failed to anchor to the overlying LE layer, impairing their normal elongation and packaging. Furthermore, our localization study results suggest that, in the developing LE, Yap participates in the cell context-dependent transition from the proliferative to differentiation-competent state by integrating cell density information. Taken together, our results shed new light on Yap's indispensable and novel organizing role in mammalian organ size control by coordinating multiple events including cell proliferation, differentiation, and polarity.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Polaridad Celular/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Cristalino/embriología , Fosfoproteínas/metabolismo , Transducción de Señal/fisiología , Células Madre/fisiología , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Proteínas de Ciclo Celular , Forma de la Célula/fisiología , Cartilla de ADN/genética , Células Epiteliales/citología , Técnica del Anticuerpo Fluorescente , Vía de Señalización Hippo , Hibridación in Situ , Cristalino/citología , Ratones , Fosfoproteínas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal/genética , Proteínas Señalizadoras YAP , beta-Cristalinas/metabolismo
14.
Hum Mol Genet ; 21(12): 2663-76, 2012 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-22398208

RESUMEN

Mutation of the polarity gene Crumbs homolog 1 (CRB1) is responsible for >10% of Leber congenital amaurosis (LCA) cases worldwide; LCA is characterized by early-onset degenerative retinal dystrophy. The role of CRB1 in LCA8 pathogenesis remains elusive since Crb1 mouse mutants, including a null allele, have failed to mimic the early-onset of LCA, most likely due to functional compensation by closely related genes encoding Crb2 and Crb3. Crb proteins form an evolutionarily conserved, apical polarity complex with the scaffolding protein associated with lin-seven 1 (Pals1), also known as MAGUK p55 subfamily member 5 (MPP5). Pals1 and Crbs are functionally inter-dependent in establishing and maintaining epithelial polarity. Pals1 is a single gene in the mouse and human genomes; therefore, we ablated Pals1 to establish a mouse genetic model mimicking human LCA. In our study, the deletion of Pals1 leads to the disruption of the apical localization of Crb proteins in retinal progenitors and the adult retina, validating their mutual interaction. Remarkably, the Pals1 mutant mouse exhibits the critical features of LCA such as early visual impairment as assessed by electroretinogram, disorganization of lamination and apical junctions and retinal degeneration. Our data uncover the indispensible role of Pals1 in retinal development, likely involving the maintenance of retinal polarity and survival of retinal neurons, thus providing the basis for the pathologic mechanisms of LCA8.


Asunto(s)
Amaurosis Congénita de Leber/metabolismo , Proteínas de la Membrana/metabolismo , Nucleósido-Fosfato Quinasa/metabolismo , Retina/metabolismo , Células Madre/metabolismo , Animales , Animales Recién Nacidos , Proliferación Celular , Electrorretinografía , Femenino , Regulación del Desarrollo de la Expresión Génica , Humanos , Inmunohistoquímica , Hibridación in Situ , Amaurosis Congénita de Leber/genética , Amaurosis Congénita de Leber/patología , Masculino , Glicoproteínas de Membrana , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Microscopía Electrónica , Proteínas del Tejido Nervioso/metabolismo , Nucleósido-Fosfato Quinasa/genética , Retina/embriología , Retina/crecimiento & desarrollo , Células Madre/patología , Células Madre/ultraestructura , Agudeza Visual
15.
Neurobiol Dis ; 51: 93-103, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23123587

RESUMEN

Tuberous sclerosis complex (TSC) is a dominant tumor suppressor disorder caused by mutations in either TSC1 or TSC2. TSC causes substantial neuropathology, often leading to autism spectrum disorders (ASDs) in up to 60% of patients. The anatomic and neurophysiologic links between these two disorders are not well understood. We have generated and characterized a novel TSC mouse model with Purkinje cell specific Tsc2 loss. These Tsc2f/-;Cre mice exhibit progressive Purkinje cell degeneration. Since loss of Purkinje cells is a well reported postmortem finding in patients with ASD, we conducted a series of behavior tests to asses if Tsc2f/-;Cre mice displayed autistic-like deficits. Tsc2f/-;Cre mice demonstrated increased repetitive behavior as assessed with marble burying activity. Using the three chambered apparatus to asses social behavior, we found that Tsc2f/-;Cre mice showed behavioral deficits, exhibiting no preference between a stranger mouse and an inanimate object, or between a novel and a familiar mouse. We also detected social deficits in Tsc2f/f;Cre mice, suggesting that Purkinje cell pathology is sufficient to induce ASD-like behavior. Importantly, social behavior deficits were prevented with rapamycin treatment. Altogether, these results demonstrate that loss of Tsc2 in Purkinje cells in a Tsc2-haploinsufficient background leads to autistic-like behavioral deficits. These studies provide compelling evidence that Purkinje cell loss and/or dysfunction may be an important link between TSC and ASD as well as a general anatomic phenomenon that contributes to the ASD phenotype.


Asunto(s)
Conducta Animal/fisiología , Células de Purkinje/metabolismo , Esclerosis Tuberosa/fisiopatología , Proteínas Supresoras de Tumor/deficiencia , Animales , Niño , Trastornos Generalizados del Desarrollo Infantil/genética , Trastornos Generalizados del Desarrollo Infantil/metabolismo , Trastornos Generalizados del Desarrollo Infantil/fisiopatología , Modelos Animales de Enfermedad , Femenino , Humanos , Inmunohistoquímica , Masculino , Ratones , Células de Purkinje/patología , Esclerosis Tuberosa/genética , Esclerosis Tuberosa/metabolismo , Proteína 2 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/genética
16.
FASEB J ; 26(2): 503-12, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21990376

RESUMEN

Pulmonary fibrosis, characterized by excess deposition of extracellular matrix by myofibroblasts, is a serious component of chronic lung diseases. Cadherin-11 (CDH11) is increased in wound healing and fibrotic skin. We hypothesized that CDH11 is increased in pulmonary fibrosis and contributes its development. CDH11 expression was assessed in lung tissue from idiopathic pulmonary fibrosis patients. The role of CDH11 in lung fibrosis was determined using the bleomycin model of pulmonary fibrosis, and in vitro analyses were performed on A549 cells during the process of epithelial to mesenchymal transition (EMT). Immunohistochemical studies demonstrated CDH11 expression on fibroblasts, epithelial cells, and alveolar macrophages of patients with pulmonary fibrosis and mice given bleomycin. Interestingly, CDH11-deficient mice had decreased fibrotic endpoints in the bleomycin model of pulmonary fibrosis compared to wild-type mice. Furthermore, anti-CDH11-neutralizing monoclonal antibodies successfully treated established pulmonary fibrosis induced by bleomycin. TGF-ß levels were reduced in bronchoalveolar lavage (BAL) fluid, BAL cells, and primary alveolar macrophages from CDH11-deficient mice. Mechanistic studies demonstrated that TGF-ß up-regulated CDH11 expression on A549 cells, and inhibition of CDH11 expression using siRNA reduced TGF-ß-induced EMT. Together, these results identify CDH11 as a novel therapeutic target for pulmonary fibrosis.


Asunto(s)
Cadherinas/fisiología , Transición Epitelial-Mesenquimal/fisiología , Fibrosis Pulmonar/etiología , Factor de Crecimiento Transformador beta/biosíntesis , Animales , Bleomicina/toxicidad , Cadherinas/antagonistas & inhibidores , Cadherinas/deficiencia , Cadherinas/genética , Línea Celular , Modelos Animales de Enfermedad , Femenino , Humanos , Fibrosis Pulmonar Idiopática/etiología , Fibrosis Pulmonar Idiopática/patología , Fibrosis Pulmonar Idiopática/fisiopatología , Macrófagos Alveolares/metabolismo , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Fibrosis Pulmonar/patología , Fibrosis Pulmonar/fisiopatología
17.
Nat Commun ; 14(1): 82, 2023 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-36604424

RESUMEN

Entosis is cell cannibalism utilized by tumor cells to engulf live neighboring cells for pro- or anti-tumorigenic purposes. It is unknown whether this extraordinary cellular event can be pathogenic in other diseases such as microcephaly, a condition characterized by a smaller than normal brain at birth. We find that mice mutant for the human microcephaly-causing gene Pals1, which exhibit diminished cortices due to massive cell death, also exhibit nuclei enveloped by plasma membranes inside of dividing cells. These cell-in-cell (CIC) structures represent a dynamic process accompanied by lengthened mitosis and cytokinesis abnormalities. As shown in tumor cells, ROCK inhibition completely abrogates CIC structures and restores the normal length of mitosis. Moreover, genetic elimination of Trp53 produces a remarkable rescue of cortical size along with substantial reductions of CIC structures and cell death. These results provide a novel pathogenic mechanism by which microcephaly is produced through entotic cell cannibalism.


Asunto(s)
Microcefalia , Humanos , Animales , Ratones , Microcefalia/genética , Entosis/fisiología , Carcinogénesis , Mitosis/genética , Núcleo Celular
18.
Dev Dyn ; 238(9): 2327-9, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19449303

RESUMEN

Specification of the peripheral optic cup by Wnt signaling is critical for formation of the ciliary body/iris. Identification of marker genes for this region during development provides a starting point for functional analyses. During transcriptional profiling of single cells from the developing eye, two cells were identified that expressed genes not found in most other single cell profiles. In situ hybridizations demonstrated that many of these genes were expressed in the peripheral optic cup in both early mouse and chicken development, and in the ciliary body/iris at subsequent developmental stages. These analyses indicate that the two cells probably originated from the developing ciliary body/iris. Changes in expression of these genes were assayed in embryonic chicken retinas when canonical Wnt signaling was ectopically activated by CA-beta-catenin. Twelve ciliary body/iris genes were identified as upregulated following induction, suggesting they are excellent candidates for downstream effectors of Wnt signaling in the optic cup.


Asunto(s)
Cuerpo Ciliar/metabolismo , Iris/metabolismo , Animales , Embrión de Pollo , Cuerpo Ciliar/embriología , Regulación del Desarrollo de la Expresión Génica/genética , Regulación del Desarrollo de la Expresión Génica/fisiología , Inmunohistoquímica , Hibridación in Situ , Iris/embriología , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas Wnt/genética , beta Catenina/genética , beta Catenina/metabolismo
19.
PLoS Biol ; 2(9): E247, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15226823

RESUMEN

The vertebrate retina is comprised of seven major cell types that are generated in overlapping but well-defined intervals. To identify genes that might regulate retinal development, gene expression in the developing retina was profiled at multiple time points using serial analysis of gene expression (SAGE). The expression patterns of 1,051 genes that showed developmentally dynamic expression by SAGE were investigated using in situ hybridization. A molecular atlas of gene expression in the developing and mature retina was thereby constructed, along with a taxonomic classification of developmental gene expression patterns. Genes were identified that label both temporal and spatial subsets of mitotic progenitor cells. For each developing and mature major retinal cell type, genes selectively expressed in that cell type were identified. The gene expression profiles of retinal Müller glia and mitotic progenitor cells were found to be highly similar, suggesting that Müller glia might serve to produce multiple retinal cell types under the right conditions. In addition, multiple transcripts that were evolutionarily conserved that did not appear to encode open reading frames of more than 100 amino acids in length ("noncoding RNAs") were found to be dynamically and specifically expressed in developing and mature retinal cell types. Finally, many photoreceptor-enriched genes that mapped to chromosomal intervals containing retinal disease genes were identified. These data serve as a starting point for functional investigations of the roles of these genes in retinal development and physiology.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Genoma , Retina/embriología , Retina/crecimiento & desarrollo , Retina/fisiología , Animales , Bromodesoxiuridina/farmacología , Linaje de la Célula , Mapeo Cromosómico , Análisis por Conglomerados , Biología Computacional , Bases de Datos Genéticas , Etiquetas de Secuencia Expresada , Regulación de la Expresión Génica , Biblioteca de Genes , Hibridación in Situ , Interneuronas/metabolismo , Ratones , Mitosis , Datos de Secuencia Molecular , Neuroglía/citología , Neuroglía/metabolismo , Sistemas de Lectura Abierta , ARN Mensajero/metabolismo , Retina/metabolismo , Células Madre/citología , Factores de Tiempo
20.
Elife ; 62017 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-28124973

RESUMEN

Nuclear exclusion of the transcriptional regulators and potent oncoproteins, YAP/TAZ, is considered necessary for adult tissue homeostasis. Here we show that nuclear YAP/TAZ are essential regulators of peripheral nerve development and myelin maintenance. To proliferate, developing Schwann cells (SCs) require YAP/TAZ to enter S-phase and, without them, fail to generate sufficient SCs for timely axon sorting. To differentiate, SCs require YAP/TAZ to upregulate Krox20 and, without them, completely fail to myelinate, resulting in severe peripheral neuropathy. Remarkably, in adulthood, nuclear YAP/TAZ are selectively expressed by myelinating SCs, and conditional ablation results in severe peripheral demyelination and mouse death. YAP/TAZ regulate both developmental and adult myelination by driving TEAD1 to activate Krox20. Therefore, YAP/TAZ are crucial for SCs to myelinate developing nerve and to maintain myelinated nerve in adulthood. Our study also provides a new insight into the role of nuclear YAP/TAZ in homeostatic maintenance of an adult tissue.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Vaina de Mielina/metabolismo , Fosfoproteínas/metabolismo , Células de Schwann/fisiología , Factores de Transcripción/metabolismo , Aciltransferasas , Animales , Proteínas de Ciclo Celular , Diferenciación Celular , Proliferación Celular , Ratones , Proteínas Señalizadoras YAP
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA