Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Carcinogenesis ; 44(6): 485-496, 2023 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-37463322

RESUMEN

The chromobox-containing protein CBX4 is an important regulator of epithelial cell proliferation and differentiation, and has been implicated in several cancer types. The cancer stem cell (CSC) population is a key driver of metastasis and recurrence. The undifferentiated, plastic state characteristic of CSCs relies on cues from the microenvironment. Cancer-associated fibroblasts (CAFs) are a major component of the microenvironment that can influence the CSC population through the secretion of extracellular matrix and a variety of growth factors. Here we show CBX4 is a critical regulator of the CSC phenotype in squamous cell carcinomas of the skin and hypopharynx. Moreover, CAFs can promote the expression of CBX4 in the CSC population through the secretion of interleukin-6 (IL-6). IL-6 activates JAK/STAT3 signaling to increase ∆Np63α-a key transcription factor that is essential for epithelial stem cell function and the maintenance of proliferative potential that is capable of regulating CBX4. Targeting the JAK/STAT3 axis or CBX4 directly suppresses the aggressive phenotype of CSCs and represents a novel opportunity for therapeutic intervention.


Asunto(s)
Fibroblastos Asociados al Cáncer , Carcinoma de Células Escamosas , Humanos , Fibroblastos Asociados al Cáncer/metabolismo , Interleucina-6/metabolismo , Línea Celular Tumoral , Carcinoma de Células Escamosas/patología , Proliferación Celular/genética , Cromatina/metabolismo , Células Madre Neoplásicas/patología , Fibroblastos/metabolismo , Microambiente Tumoral/genética , Ligasas/genética , Ligasas/metabolismo , Proteínas del Grupo Polycomb/genética , Proteínas del Grupo Polycomb/metabolismo
2.
Stem Cells ; 41(3): 242-251, 2023 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-36636025

RESUMEN

Chromodomain helicase DNA-binding protein 5 (Chd5) is an ATP-dependent chromatin remodeler that promotes neuronal differentiation. However, the mechanism behind the action of Chd5 during neurogenesis is not clearly understood. Here we use transcriptional profiling of cells obtained from Chd5 deficient mice at early and late stages of neuronal differentiation to show that Chd5 regulates neurogenesis by directing stepwise transcriptional changes. During early stages of neurogenesis, Chd5 promotes expression of the proneural transcription factor Six3 to repress Wnt5a, a non-canonical Wnt ligand essential for the maturation of neurons. This previously unappreciated ability of Chd5 to transcriptionally repress neuronal maturation factors is critical for both lineage specification and maturation. Thus, Chd5 facilitates early transcriptional changes in neural stem cells, thereby initiating transcriptional programs essential for neuronal fate specification.


Asunto(s)
Regulación de la Expresión Génica , Factores de Transcripción , Ratones , Animales , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Neuronas/metabolismo , Cromatina/metabolismo , Neurogénesis/genética , Diferenciación Celular/genética
3.
Nature ; 613(7942): 195-202, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36544023

RESUMEN

Inhibition of the tumour suppressive function of p53 (encoded by TP53) is paramount for cancer development in humans. However, p53 remains unmutated in the majority of cases of glioblastoma (GBM)-the most common and deadly adult brain malignancy1,2. Thus, how p53-mediated tumour suppression is countered in TP53 wild-type (TP53WT) GBM is unknown. Here we describe a GBM-specific epigenetic mechanism in which the chromatin regulator bromodomain-containing protein 8 (BRD8) maintains H2AZ occupancy at p53 target loci through the EP400 histone acetyltransferase complex. This mechanism causes a repressive chromatin state that prevents transactivation by p53 and sustains proliferation. Notably, targeting the bromodomain of BRD8 displaces H2AZ, enhances chromatin accessibility and engages p53 transactivation. This in turn enforces cell cycle arrest and tumour suppression in TP53WT GBM. In line with these findings, BRD8 is highly expressed with H2AZ in proliferating single cells of patient-derived GBM, and is inversely correlated with CDKN1A, a canonical p53 target that encodes p21 (refs. 3,4). This work identifies BRD8 as a selective epigenetic vulnerability for a malignancy for which treatment has not improved for decades. Moreover, targeting the bromodomain of BRD8 may be a promising therapeutic strategy for patients with TP53WT GBM.


Asunto(s)
Epigénesis Genética , Glioblastoma , Factores de Transcripción , Proteína p53 Supresora de Tumor , Adulto , Humanos , Puntos de Control del Ciclo Celular , Línea Celular Tumoral , Cromatina/genética , Cromatina/metabolismo , Glioblastoma/genética , Glioblastoma/metabolismo , Glioblastoma/patología , Histonas/metabolismo , Factores de Transcripción/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Proliferación Celular
4.
Trends Cell Biol ; 33(4): 280-292, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36115734

RESUMEN

Our understanding of cancer and the key pathways that drive cancer survival has expanded rapidly over the past several decades. However, there are still important challenges that continue to impair patient survival, including our inability to target cancer stem cells (CSCs), metastasis, and drug resistance. The transcription factor p63 is a p53 family member with multiple isoforms that carry out a wide array of functions. Here, we discuss the critical importance of the ΔNp63α isoform in cancer and potential therapeutic strategies to target ΔNp63α expression to impair the CSC population, as well as to prevent metastasis and drug resistance to improve patient survival.


Asunto(s)
Neoplasias , Proteínas Supresoras de Tumor , Humanos , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Neoplasias/genética , Neoplasias/terapia , Factores de Transcripción/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Regulación Neoplásica de la Expresión Génica , Línea Celular Tumoral
5.
Oncogene ; 41(35): 4130-4144, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35864175

RESUMEN

Enhancer of zeste homolog 2 (EZH2) and SET domain bifurcated 1 (SETDB1, also known as ESET) are oncogenic methyltransferases implicated in a number of human cancers. These enzymes typically function as epigenetic repressors of target genes by methylating histone H3 K27 and H3-K9 residues, respectively. Here, we show that EZH2 and SETDB1 are essential to proliferation in 3 SCC cell lines, HSC-5, FaDu, and Cal33. Additionally, we find both of these proteins highly expressed in an aggressive stem-like SCC sub-population. Depletion of either EZH2 or SETDB1 disrupts these stem-like cells and their associated phenotypes of spheroid formation, invasion, and tumor growth. We show that SETDB1 regulates this SCC stem cell phenotype through cooperation with ΔNp63α, an oncogenic isoform of the p53-related transcription factor p63. Furthermore, EZH2 is upstream of both SETDB1 and ΔNp63α, activating these targets via repression of the tumor suppressor RUNX3. We show that targeting this pathway with inhibitors of EZH2 results in activation of RUNX3 and repression of both SETDB1 and ΔNp63α, antagonizing the SCC cancer stem cell phenotype. This work highlights a novel pathway that drives an aggressive cancer stem cell phenotype and demonstrates a means of pharmacological intervention.


Asunto(s)
Carcinoma de Células Escamosas , Proteína Potenciadora del Homólogo Zeste 2 , Línea Celular Tumoral , Subunidad alfa 3 del Factor de Unión al Sitio Principal , N-Metiltransferasa de Histona-Lisina , Humanos , Células Madre Neoplásicas , Fenotipo , Regiones Promotoras Genéticas , Factores de Transcripción
6.
PLoS One ; 17(1): e0261770, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35077450

RESUMEN

Progressive immune deficiency of aging is characterized by severe thymic atrophy, contracted T cell repertoire, and poor immune function. p63 is critical for the proliferative potential of embryonic and adult stem cells, as well as thymic epithelial cells (TECs). Because p63 null mice experience rapid post-natal lethality due to epidermal and limb morphogenesis defects, studies to define a role for p63 expression in TEC biology focused on embryonic thymus development and in vitro experiments. Since post-natal thymic stromal development and function differs from that of the embryo, we assessed the impact of lineage-restricted p63 loss on pre- and post-natal murine TEC function by generating mice with a loss of p63 function targeted to TEC, termed p63TECko mice. In adult p63TECko mice, severe thymic hypoplasia was observed with a lack in a discernable segregation into medullary and cortical compartments and peripheral T cell lymphopenia. This profound thymic defect was seen in both neonatal as well as embryonic p63TECko mice. In addition to TECs, p63 also plays in important role in the development of stratified epithelium of the skin; lack of p63 results in defects in skin epidermal stratification and differentiation. Interestingly, all adult p63TECko mice lacked hair follicles despite having normal p63 expression in the skin. Together our results show a critical role of TEC p63 in thymic development and maintenance and show that p63 expression is critical for hair follicle formation.


Asunto(s)
Alopecia/genética , Factores de Transcripción Forkhead/genética , Eliminación de Gen , Técnicas de Inactivación de Genes , Regiones Promotoras Genéticas , Timo/crecimiento & desarrollo , Transactivadores/deficiencia , Alopecia/metabolismo , Animales , Factores de Transcripción Forkhead/metabolismo , Ratones , Ratones Noqueados , Transactivadores/metabolismo
7.
Cancer Res ; 81(24): 6246-6258, 2021 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-34697072

RESUMEN

Bromodomain containing protein 4 (BRD4) plays a critical role in controlling the expression of genes involved in development and cancer. Inactivation of BRD4 inhibits cancer growth, making it a promising anticancer drug target. The cancer stem cell (CSC) population is a key driver of recurrence and metastasis in patients with cancer. Here we show that cancer stem-like cells can be enriched from squamous cell carcinomas (SCC), and that these cells display an aggressive phenotype with enhanced stem cell marker expression, migration, invasion, and tumor growth. BRD4 is highly elevated in this aggressive subpopulation of cells, and its function is critical for these CSC-like properties. Moreover, BRD4 regulates ΔNp63α, a key transcription factor that is essential for epithelial stem cell function that is often overexpressed in cancers. BRD4 regulates an EZH2/STAT3 complex that leads to increased ΔNp63α-mediated transcription. Targeting BRD4 in human SCC reduces ΔNp63α, leading to inhibition of spheroid formation, migration, invasion, and tumor growth. These studies identify a novel BRD4-regulated signaling network in a subpopulation of cancer stem-like cells, elucidating a possible avenue for effective therapeutic intervention. SIGNIFICANCE: This study identifies a signaling cascade driven by BRD4 that upregulates ΔNp63α to promote cancer stem-like properties, which has potential therapeutic implications for the treatment of squamous cell carcinomas.


Asunto(s)
Carcinoma de Células Escamosas/patología , Proteínas de Ciclo Celular/metabolismo , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Regulación Neoplásica de la Expresión Génica , Células Madre Neoplásicas/patología , Factor de Transcripción STAT3/metabolismo , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Apoptosis , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Proteínas de Ciclo Celular/genética , Proliferación Celular , Proteína Potenciadora del Homólogo Zeste 2/genética , Humanos , Ratones , Ratones Desnudos , Células Madre Neoplásicas/metabolismo , Factor de Transcripción STAT3/genética , Factores de Transcripción/genética , Células Tumorales Cultivadas , Proteínas Supresoras de Tumor/genética , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Mol Psychiatry ; 26(6): 1967-1979, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-32099100

RESUMEN

The human 16p11.2 gene locus is a hot spot for copy number variations, which predispose carriers to a range of neuropsychiatric phenotypes. Microduplications of 16p11.2 are associated with autism spectrum disorder (ASD), intellectual disability (ID), and schizophrenia (SZ). Despite the debilitating nature of 16p11.2 duplications, the underlying molecular mechanisms remain poorly understood. Here we performed a comprehensive behavioral characterization of 16p11.2 duplication mice (16p11.2dp/+) and identified social and cognitive deficits reminiscent of ASD and ID phenotypes. 16p11.2dp/+ mice did not exhibit the SZ-related sensorimotor gating deficits, psychostimulant-induced hypersensitivity, or motor impairment. Electrophysiological recordings of 16p11.2dp/+ mice found deficient GABAergic synaptic transmission and elevated neuronal excitability in the prefrontal cortex (PFC), a brain region critical for social and cognitive functions. RNA-sequencing identified genome-wide transcriptional aberrance in the PFC of 16p11.2dp/+ mice, including downregulation of the GABA synapse regulator Npas4. Restoring Npas4 expression in PFC of 16p11.2dp/+ mice ameliorated the social and cognitive deficits and reversed GABAergic synaptic impairment and neuronal hyperexcitability. These findings suggest that prefrontal cortical GABAergic synaptic circuitry and Npas4 are strongly implicated in 16p11.2 duplication pathology, and may represent potential targets for therapeutic intervention in ASD.


Asunto(s)
Trastorno del Espectro Autista , Deleción Cromosómica , Animales , Trastorno del Espectro Autista/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Cromosomas Humanos Par 16/genética , Variaciones en el Número de Copia de ADN/genética , Humanos , Ratones , Sinapsis , Ácido gamma-Aminobutírico
9.
J Cell Sci ; 133(17)2020 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-32917730

RESUMEN

p63 (also known as TP63) is a transcription factor of the p53 family, along with p73. Multiple isoforms of p63 have been discovered and these have diverse functions encompassing a wide array of cell biology. p63 isoforms are implicated in lineage specification, proliferative potential, differentiation, cell death and survival, DNA damage response and metabolism. Furthermore, p63 is linked to human disease states including cancer. p63 is critical to many aspects of cell signaling, and in this Cell science at a glance article and the accompanying poster, we focus on the signaling cascades regulating TAp63 and ΔNp63 isoforms and those that are regulated by TAp63 and ΔNp63, as well the role of p63 in disease.


Asunto(s)
Neoplasias , Factores de Transcripción , Humanos , Neoplasias/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transducción de Señal , Factores de Transcripción/genética
10.
J Assist Reprod Genet ; 37(2): 341-346, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31792669

RESUMEN

PURPOSE: To study the incidence of tumor suppressor gene (TSG) mutations in men and women with impaired gametogenesis. METHODS: Gene association analyses were performed on blood samples in two distinct patient populations: males with idiopathic male infertility and females with unexplained diminished ovarian reserve (DOR). The male study group consisted of men with idiopathic azoospermia, oligozoospermia, asthenozoospermia, or teratozoospermia. Age-matched controls were men with normal semen analyses. The female study group consisted of women with unexplained DOR with anti-Müllerian hormone levels ≤ 1.1 ng/mL. Controls were age-matched women with normal ovarian reserve (> 1.1 ng/mL). RESULTS: Fifty-seven male cases (mean age = 38.4; mean sperm count = 15.7 ± 12.1; mean motility = 38.2 ± 24.7) and 37 age-matched controls (mean age = 38.0; mean sperm count = 89.6 ± 37.5; mean motility = 56.2 ± 14.3) were compared. Variants observed in CHD5 were found to be enriched in the study group (p = 0.000107). The incidence of CHD5 mutation c.*3198_*3199insT in the 3'UTR (rs538186680) was significantly higher in cases compared to controls (p = 0.0255). 72 DOR cases (mean age = 38.7; mean AMH = 0.5 ± 0.3; mean FSH = 11.7 ± 12.5) and 48 age-matched controls (mean age = 37.6; mean AMH = 4.1 ± 3.0; mean FSH = 7.1 ± 2.2) were compared. Mutations in CHD5 (c.-140A>C), RB1 (c.1422-18delT, rs70651121), and TP53 (c.376-161A>G, rs75821853) were found at significantly higher frequencies in DOR cases compared to controls (p ≤ 0.05). In addition, 363 variants detected in the DOR patients were not present in the control group. CONCLUSION: Unexplained impaired gametogenesis in both males and females may be associated with genetic variation in TSGs. TSGs, which play cardinal roles in cell-cycle control, might also be critical for normal spermatogenesis and oogenesis. If validated in larger prospective studies, it is possible that TSGs provide an etiological basis for some patients with impaired gametogenesis.


Asunto(s)
Infertilidad Femenina/genética , Infertilidad Masculina/genética , Reserva Ovárica/genética , Espermatogénesis/genética , Adulto , ADN Helicasas/genética , Femenino , Gametogénesis/genética , Genes Supresores de Tumor , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Humanos , Infertilidad Femenina/patología , Infertilidad Masculina/patología , Masculino , Mutación/genética , Proteínas del Tejido Nervioso/genética , Proteínas de Unión a Retinoblastoma/genética , Recuento de Espermatozoides , Motilidad Espermática/genética , Espermatozoides/patología , Proteína p53 Supresora de Tumor/genética , Ubiquitina-Proteína Ligasas/genética
11.
Sleep ; 42(3)2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30541142

RESUMEN

Sleep abnormalities are common among children with neurodevelopmental disorders. The human chr16p11.2 microdeletion is associated with a range of neurological and neurobehavioral abnormalities. Previous studies of a mouse model of human chr16p11.2 microdeletion (chr16p11.2df/+) have demonstrated pathophysiological changes at the synapses in the hippocampus and striatum; however, the impact of this genetic abnormality on system level brain functions, such as sleep and neural oscillation, has not been adequately investigated. Here, we show that chr16p11.2df/+ mice have altered sleep architecture, with increased wake time and reduced time in rapid eye movement (REM) and non-REM (NREM) sleep. Importantly, several measurements of REM sleep are significantly changed in deletion mice. The REM bout number and the bout number ratio of REM to NREM are decreased in mutant mice, suggesting a deficit in REM-NREM transition. The average REM bout duration is shorter in mutant mice, indicating a defect in REM maintenance. In addition, whole-cell patch clamp recording of the ventrolateral periaqueductal gray (vlPAG)-projecting gamma-aminobutyric acid (GABA)ergic neurons in the lateral paragigantocellular nucleus of ventral medulla of mutant mice reveal that these neurons, which are important for NREM-REM transition and REM maintenance, have hyperpolarized resting membrane potential and increased membrane resistance. These changes in intrinsic membrane properties suggest that these projection-specific neurons of mutant mice are less excitable, and thereby may play a role in deficient NREM-REM transition and REM maintenance. Furthermore, mutant mice exhibit changes in neural oscillation involving multiple frequency classes in several vigilance states. The most significant alterations occur in the theta frequency during wake and REM sleep.


Asunto(s)
Trastorno Autístico/genética , Trastorno Autístico/fisiopatología , Trastornos de los Cromosomas/genética , Trastornos de los Cromosomas/fisiopatología , Modelos Animales de Enfermedad , Electroencefalografía/métodos , Neuronas GABAérgicas/fisiología , Discapacidad Intelectual/genética , Discapacidad Intelectual/fisiopatología , Sueño REM/fisiología , Animales , Deleción Cromosómica , Cromosomas Humanos Par 16/genética , Humanos , Masculino , Bulbo Raquídeo/fisiología , Potenciales de la Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Polisomnografía/métodos
12.
Life Sci Alliance ; 1(4): e201700016, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30456361

RESUMEN

Neural cell fate specification is a multistep process in which stem cells undergo sequential changes in states, giving rise to particular lineages such as neurons and astrocytes. This process is accompanied by dynamic changes of chromatin and in transcription, thereby orchestrating lineage-specific gene expression programs. A pressing question is how these events are interconnected to sculpt cell fate. We show that altered chromatin due to loss of the chromatin remodeler Chd5 causes neural stem cell activation to occur ahead of time. This premature activation is accompanied by transcriptional derepression of ribosomal subunits, enhanced ribosome biogenesis, and increased translation. These untimely events deregulate cell fate decisions, culminating in the generation of excessive numbers of astrocytes at the expense of neurons. By monitoring the proneural factor Mash1, we further show that translational control is crucial for appropriate execution of cell fate specification, thereby providing new insight into the interplay between transcription and translation at the initial stages of neurogenesis.

13.
J Neurophysiol ; 119(3): 1005-1018, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29212915

RESUMEN

The pathophysiology of neurodevelopmental disorders is often observed early in infancy and toddlerhood. Mouse models of syndromic disorders have provided insight regarding mechanisms of action, but most studies have focused on characterization in juveniles and adults. Insight into developmental trajectories, particularly those related to circuit and synaptic function, will likely yield important information regarding disorder pathogenesis that leads to symptom progression. Chromosome 16p11.2 microdeletion is one of the most common copy number variations associated with a spectrum of neurodevelopmental disorders. Yet, how haploinsufficiency of chr16p11.2 affects early synaptic maturation and function is unknown. To address this knowledge gap, the present study focused on three key components of circuit formation and function, basal synaptic transmission, local circuit function, and maturation of glutamatergic synapses, in developing hippocampal CA1 neurons in a chr16p11.2 microdeletion mouse model. The data demonstrate increased excitability, imbalance in excitation and inhibition, and accelerated maturation of glutamatergic synapses in heterozygous deletion mutant CA1 neurons. Given the critical role of early synaptic development in shaping neuronal connectivity and circuitry formation, these newly identified synaptic abnormalities in chr16p11.2 microdeletion mice may contribute to altered developmental trajectory and function of the developing brain. NEW & NOTEWORTHY The synaptic pathophysiology underlying neurodevelopmental disorders often emerges during infancy and toddlerhood. Therefore, identifying initial changes in synaptic function is crucial for gaining a mechanistic understanding of the pathophysiology, which ultimately will facilitate the design of early interventions. Here, we investigated synaptic and local circuit properties of hippocampal CA1 neurons in a human chr16p11.2 microdeletion mouse model during early postnatal development (preweaning). The data demonstrate increased neuronal excitability, excitatory/inhibitory imbalance, and accelerated maturation of glutamatergic synapses. These perturbations in early hippocampal circuit function may underlie the early pathogenesis of the heterozygous chr16p11.2 microdeletion, which is often associated with epilepsy and intellectual disability.


Asunto(s)
Región CA1 Hipocampal/fisiopatología , Cromosomas Humanos Par 16/genética , Eliminación de Gen , Neuronas/fisiología , Transmisión Sináptica , Animales , Región CA1 Hipocampal/crecimiento & desarrollo , Variaciones en el Número de Copia de ADN , Humanos , Masculino , Potenciales de la Membrana , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores AMPA/fisiología
14.
PLoS One ; 12(3): e0174122, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28333962

RESUMEN

The apical ectodermal ridge (AER), located at the distal end of each limb bud, is a key signaling center which controls outgrowth and patterning of the proximal-distal axis of the limb through secretion of various molecules. Fibroblast growth factors (FGFs), particularly Fgf8 and Fgf4, are representative molecules produced by AER cells, and essential to maintain the AER and cell proliferation in the underlying mesenchyme, meanwhile Jag2-Notch pathway negatively regulates the AER and limb development. p63, a transcription factor of the p53 family, is expressed in the AER and indispensable for limb formation. However, the underlying mechanisms and specific roles of p63 variants are unknown. Here, we quantified the expression of p63 variants in mouse limbs from embryonic day (E) 10.5 to E12.5, and found that ΔNp63γ was strongly expressed in limbs at all stages, while TAp63γ expression was rapidly increased in the later stages. Fluorescence-activated cell sorting analysis of limb bud cells from reporter mouse embryos at E11.5 revealed that all variants were abundantly expressed in AER cells, and their expression was very low in mesenchymal cells. We then generated AER-specific p63 knockout mice by mating mice with a null and a flox allele of p63, and Msx2-Cre mice (Msx2-Cre;p63Δ/fl). Msx2-Cre;p63Δ/fl neonates showed limb malformation that was more obvious in distal elements. Expression of various AER-related genes was decreased in Msx2-Cre;p63Δ/fl limb buds and embryoid bodies formed by p63-knockdown induced pluripotent stem cells. Promoter analyses and chromatin immunoprecipitation assays demonstrated Fgf8 and Fgf4 as transcriptional targets of ΔNp63γ, and Jag2 as that of TAp63γ. Furthermore, TAp63γ overexpression exacerbated the phenotype of Msx2-Cre;p63Δ/fl mice. These data indicate that ΔNp63 and TAp63 control limb development through transcriptional regulation of different target molecules with different roles in the AER. Our findings contribute to further understanding of the molecular network of limb development.


Asunto(s)
Esbozos de los Miembros/crecimiento & desarrollo , Fosfoproteínas/fisiología , Transactivadores/fisiología , Animales , Animales Recién Nacidos , Factor 4 de Crecimiento de Fibroblastos/fisiología , Factor 8 de Crecimiento de Fibroblastos/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Esbozos de los Miembros/fisiología , Ratones/embriología , Ratones Endogámicos C57BL , Ratones Noqueados , Fosfoproteínas/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Transactivadores/genética
15.
Artículo en Inglés | MEDLINE | ID: mdl-28096241

RESUMEN

A plethora of mutations in chromatin regulators in diverse human cancers is emerging, attesting to the pivotal role of chromatin dynamics in tumorigenesis. A recurrent theme is inactivation of the chromodomain helicase DNA-binding (CHD) family of proteins-ATP-dependent chromatin remodelers that govern the cellular machinery's access to DNA, thereby controlling fundamental processes, including transcription, proliferation, and DNA damage repair. This review highlights what is currently known about how genetic and epigenetic perturbation of CHD proteins and the pathways that they regulate set the stage for cancer, providing new insight for designing more effective anti-cancer therapies.


Asunto(s)
Ensamble y Desensamble de Cromatina/genética , ADN Helicasas/genética , Proteínas de Unión al ADN/genética , Neoplasias/genética , Reparación del ADN , Humanos , Mutación , Fenotipo , Transcripción Genética
16.
Arthritis Rheumatol ; 69(3): 598-609, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27792866

RESUMEN

OBJECTIVE: Transcription factor p63, of the p53 family, regulates cell proliferation, survival, and apoptosis in various cells and tissues. This study was undertaken to examine the expression and roles of p63 transcript variants in the mouse growth plate and articular chondrocytes. METHODS: For in vivo analyses, we generated Cre-mediated TAp63α-transgenic and TAp63γ-transgenic mice. To induce tissue-specific overexpression or deletion in chondrocytes, chondroprogenitor cells, or early limb bud mesenchymal cells, we used Col2a1-Cre, Sox9-Cre, and Prx1-Cre mice, respectively. We analyzed osteoarthritis (OA) development with aging or surgically induced instability in Prx1-Cre;p63fl/fl (P-conditional knockout) mice. RESULTS: Among major variants, TAp63α and TAp63γ are highly expressed in mouse primary costal and articular chondrocytes. The p63 protein was predominantly localized in the hypertrophic zone of the embryonic limb cartilage, and in the middle zone of articular cartilage. No obvious change was observed in skeletal growth of TAp63α-transgenic mice, Sox9-Cre;p63fl/fl , or P-conditional knockout mice, while that of TAp63γ-transgenic mice was impaired due to ectopic apoptosis and the resulting decreased number of chondrocytes. Expression of proapoptotic genes including bax, noxa, puma, and fas was increased in TAp63γ-transgenic mouse chondrocytes, and their transcription was probably sustained by p53 in p63-conditional knockout mouse chondrocytes because both proteins were coexpressed in the growth plate. In contrast, p53 was expressed in the superficial zone of articular cartilage, differently from p63. Notably, P-conditional knockout mice showed significant resistance to OA development, with suppression of chondrocyte apoptosis in the aging and surgical models. CONCLUSION: We demonstrated regulation of chondrocyte survival in articular cartilage by p63.


Asunto(s)
Cartílago Articular/citología , Condrocitos/fisiología , Placa de Crecimiento/citología , Fosfoproteínas/fisiología , Transactivadores/fisiología , Animales , Supervivencia Celular , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos
17.
Nature ; 531(7595): 471-475, 2016 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-26982726

RESUMEN

Mutations disabling the TP53 tumour suppressor gene represent the most frequent events in human cancer and typically occur through a two-hit mechanism involving a missense mutation in one allele and a 'loss of heterozygosity' deletion encompassing the other. While TP53 missense mutations can also contribute gain-of-function activities that impact tumour progression, it remains unclear whether the deletion event, which frequently includes many genes, impacts tumorigenesis beyond TP53 loss alone. Here we show that somatic heterozygous deletion of mouse chromosome 11B3, a 4-megabase region syntenic to human 17p13.1, produces a greater effect on lymphoma and leukaemia development than Trp53 deletion. Mechanistically, the effect of 11B3 loss on tumorigenesis involves co-deleted genes such as Eif5a and Alox15b (also known as Alox8), the suppression of which cooperates with Trp53 loss to produce more aggressive disease. Our results imply that the selective advantage produced by human chromosome 17p deletion reflects the combined impact of TP53 loss and the reduced dosage of linked tumour suppressor genes.


Asunto(s)
Genes p53/genética , Neoplasias/genética , Neoplasias/patología , Eliminación de Secuencia/genética , Proteína p53 Supresora de Tumor/deficiencia , Alelos , Animales , Transformación Celular Neoplásica/genética , Cromosomas Humanos Par 17/genética , Cromosomas de los Mamíferos/genética , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Heterocigoto , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Linfoma/genética , Linfoma/patología , Masculino , Ratones , Factores de Iniciación de Péptidos/genética , Factores de Iniciación de Péptidos/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Sintenía/genética , Factor 5A Eucariótico de Iniciación de Traducción
18.
BMC Genomics ; 16: 982, 2015 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-26589460

RESUMEN

BACKGROUND: Circular chromosome conformation capture (4C) has provided important insights into three dimensional (3D) genome organization and its critical impact on the regulation of gene expression. We developed a new quantitative framework based on polymer physics for the analysis of paired-end sequencing 4C (PE-4Cseq) data. We applied this strategy to the study of chromatin interaction changes upon a 4.3 Mb DNA deletion in mouse region 4E2. RESULTS: A significant number of differentially interacting regions (DIRs) and chromatin compaction changes were detected in the deletion chromosome compared to a wild-type (WT) control. Selected DIRs were validated by 3D DNA FISH experiments, demonstrating the robustness of our pipeline. Interestingly, significant overlaps of DIRs with CTCF/Smc1 binding sites and differentially expressed genes were observed. CONCLUSIONS: Altogether, our PE-4Cseq analysis pipeline provides a comprehensive characterization of DNA deletion effects on chromatin structure and function.


Asunto(s)
Cromatina/genética , Cromatina/metabolismo , Biología Computacional , Eliminación de Secuencia , Alelos , Animales , Cromosomas de los Mamíferos , Biología Computacional/métodos , Variaciones en el Número de Copia de ADN , Expresión Génica , Genómica/métodos , Genotipo , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Hibridación Fluorescente in Situ , Ratones , Polimorfismo de Nucleótido Simple , Reproducibilidad de los Resultados
19.
PLoS One ; 10(8): e0134572, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26273832

RESUMEN

Autism spectrum disorder comprises several neurodevelopmental conditions presenting symptoms in social communication and restricted, repetitive behaviors. A major roadblock for drug development for autism is the lack of robust behavioral signatures predictive of clinical efficacy. To address this issue, we further characterized, in a uniform and rigorous way, mouse models of autism that are of interest because of their construct validity and wide availability to the scientific community. We implemented a broad behavioral battery that included but was not restricted to core autism domains, with the goal of identifying robust, reliable phenotypes amenable for further testing. Here we describe comprehensive findings from two known mouse models of autism, obtained at different developmental stages, using a systematic behavioral test battery combining standard tests as well as novel, quantitative, computer-vision based systems. The first mouse model recapitulates a deletion in human chromosome 16p11.2, found in 1% of individuals with autism. The second mouse model harbors homozygous null mutations in Cntnap2, associated with autism and Pitt-Hopkins-like syndrome. Consistent with previous results, 16p11.2 heterozygous null mice, also known as Del(7Slx1b-Sept1)4Aam weighed less than wild type littermates displayed hyperactivity and no social deficits. Cntnap2 homozygous null mice were also hyperactive, froze less during testing, showed a mild gait phenotype and deficits in the three-chamber social preference test, although less robust than previously published. In the open field test with exposure to urine of an estrous female, however, the Cntnap2 null mice showed reduced vocalizations. In addition, Cntnap2 null mice performed slightly better in a cognitive procedural learning test. Although finding and replicating robust behavioral phenotypes in animal models is a challenging task, such functional readouts remain important in the development of therapeutics and we anticipate both our positive and negative findings will be utilized as a resource for the broader scientific community.


Asunto(s)
Trastorno del Espectro Autista/genética , Trastorno del Espectro Autista/fisiopatología , Cromosomas de los Mamíferos/genética , Proteínas de la Membrana/genética , Mutación , Proteínas del Tejido Nervioso/genética , Animales , Animales Recién Nacidos , Conducta Animal/fisiología , Cognición/fisiología , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Eliminación de Secuencia , Vocalización Animal/fisiología
20.
Nat Neurosci ; 18(2): 182-4, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25581360

RESUMEN

Human chromosome 16p11.2 microdeletion is the most common gene copy number variation in autism, but the synaptic pathophysiology caused by this mutation is largely unknown. Using a mouse with the same genetic deficiency, we found that metabotropic glutamate receptor 5 (mGluR5)-dependent synaptic plasticity and protein synthesis was altered in the hippocampus and that hippocampus-dependent memory was impaired. Notably, chronic treatment with a negative allosteric modulator of mGluR5 reversed the cognitive deficit.


Asunto(s)
Trastorno Autístico/fisiopatología , Trastornos de los Cromosomas/fisiopatología , Hipocampo/fisiopatología , Imidazoles/farmacología , Discapacidad Intelectual/fisiopatología , Trastornos de la Memoria/fisiopatología , Piridinas/farmacología , Receptor del Glutamato Metabotropico 5/fisiología , Animales , Trastorno Autístico/genética , Trastorno Autístico/metabolismo , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Deleción Cromosómica , Trastornos de los Cromosomas/genética , Trastornos de los Cromosomas/metabolismo , Cromosomas Humanos Par 16/genética , Cromosomas Humanos Par 16/metabolismo , Cromosomas de los Mamíferos , Modelos Animales de Enfermedad , Hipocampo/metabolismo , Discapacidad Intelectual/genética , Discapacidad Intelectual/metabolismo , Masculino , Trastornos de la Memoria/tratamiento farmacológico , Trastornos de la Memoria/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Plasticidad Neuronal/efectos de los fármacos , Plasticidad Neuronal/genética , Plasticidad Neuronal/fisiología , Receptor del Glutamato Metabotropico 5/genética , Receptor del Glutamato Metabotropico 5/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transducción de Señal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...