Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
J Invest Dermatol ; 142(7): 1845-1857, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-34958806

RESUMEN

Phenotypic plasticity drives cancer progression, impacts treatment response, and is a major driver of therapeutic resistance. In melanoma, a regulatory axis between the MITF and BRN2 transcription factors has been reported to promote tumor heterogeneity by mediating switching between proliferative and invasive phenotypes, respectively. Despite strong evidence that subpopulations of cells that exhibit a BRN2high/MITFlow expression profile switch to a predominantly invasive phenotype, the mechanisms by which this switch is propagated and promotes invasion remain poorly defined. We have found that a reciprocal relationship between BRN2 and NOTCH1/2 signaling exists in melanoma cells in vitro, within patient datasets, and in in vivo primary and metastatic human tumors that bolsters acquisition of invasiveness. Working through the epigenetic modulator EZH2, the BRN2‒NOTCH1/2 axis is potentially a key mechanism by which the invasive phenotype is maintained. Given the emergence of agents targeting both EZH2 and NOTCH, understanding the mechanism through which BRN2 promotes heterogeneity may provide crucial biomarkers to predict treatment response to prevent metastasis.


Asunto(s)
Proteínas de Homeodominio , Melanoma , Factores del Dominio POU , Receptor Notch1 , Receptor Notch2 , Línea Celular Tumoral , Movimiento Celular , Regulación Neoplásica de la Expresión Génica , Proteínas de Homeodominio/genética , Humanos , Melanoma/patología , Factor de Transcripción Asociado a Microftalmía/genética , Invasividad Neoplásica/genética , Factores del Dominio POU/genética , Receptor Notch1/genética , Receptor Notch2/genética
2.
J Biomed Opt ; 26(10)2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34689443

RESUMEN

SIGNIFICANCE: Peripheral pitting edema is a clinician-administered measure for grading edema. Peripheral edema is graded 0, 1 + , 2 + , 3 + , or 4 + , but subjectivity is a major limitation of this technique. A pilot clinical study for short-wave infrared (SWIR) molecular chemical imaging (MCI) effectiveness as an objective, non-contact quantitative peripheral edema measure is underway. AIM: We explore if SWIR MCI can differentiate populations with and without peripheral edema. Further, we evaluate the technology for correctly stratifying subjects with peripheral edema. APPROACH: SWIR MCI of shins from healthy subjects and heart failure (HF) patients was performed. Partial least squares discriminant analysis (PLS-DA) was used to discriminate the two populations. PLS regression (PLSR) was applied to assess the ability of MCI to grade edema. RESULTS: Average spectra from edema exhibited higher water absorption than non-edema spectra. SWIR MCI differentiated healthy volunteers from a population representing all pitting edema grades with 97.1% accuracy (N = 103 shins). Additionally, SWIR MCI correctly classified shin pitting edema levels in patients with 81.6% accuracy. CONCLUSIONS: Our study successfully achieved the two primary endpoints. Application of SWIR MCI to monitor patients while actively receiving HF treatment is necessary to validate SWIR MCI as an HF monitoring technology.


Asunto(s)
Insuficiencia Cardíaca , Imagen Molecular , Análisis Discriminante , Edema/diagnóstico por imagen , Insuficiencia Cardíaca/diagnóstico por imagen , Humanos , Análisis de los Mínimos Cuadrados
3.
FASEB J ; 33(5): 6412-6430, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30779881

RESUMEN

Growth hormone (GH) has an important function as an insulin antagonist with elevated insulin sensitivity evident in humans and mice lacking a functional GH receptor (GHR). We sought the molecular basis for this sensitivity by utilizing a panel of mice possessing specific deletions of GHR signaling pathways. Metabolic clamps and glucose homeostasis tests were undertaken in these obese adult C57BL/6 male mice, which indicated impaired hepatic gluconeogenesis. Insulin sensitivity and glucose disappearance rate were enhanced in muscle and adipose of mice lacking the ability to activate the signal transducer and activator of transcription (STAT)5 via the GHR (Ghr-391-/-) as for GHR-null (GHR-/-) mice. These changes were associated with a striking inhibition of hepatic glucose output associated with altered glycogen metabolism and elevated hepatic glycogen content during unfed state. The enhanced hepatic insulin sensitivity was associated with increased insulin receptor ß and insulin receptor substrate 1 activation along with activated downstream protein kinase B signaling cascades. Although phosphoenolpyruvate carboxykinase (Pck)-1 expression was unchanged, its inhibitory acetylation was elevated because of decreased sirtuin-2 expression, thereby promoting loss of PCK1. Loss of STAT5 signaling to defined chromatin immunoprecipitation targets would further increase lipogenesis, supporting hepatosteatosis while lowering glucose output. Finally, up-regulation of IL-15 expression in muscle, with increased secretion of adiponectin and fibroblast growth factor 1 from adipose tissue, is expected to promote insulin sensitivity.-Chhabra, Y., Nelson, C. N., Plescher, M., Barclay, J. L., Smith, A. G., Andrikopoulos, S., Mangiafico, S., Waxman, D. J., Brooks, A. J., Waters, M. J. Loss of growth hormone-mediated signal transducer and activator of transcription 5 (STAT5) signaling in mice results in insulin sensitivity with obesity.


Asunto(s)
Proteínas Portadoras , Hígado Graso , Resistencia a la Insulina/genética , Hígado , Obesidad , Factor de Transcripción STAT5/deficiencia , Transducción de Señal/genética , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Hígado Graso/genética , Hígado Graso/metabolismo , Hígado Graso/patología , Glucosa/genética , Glucosa/metabolismo , Glucógeno/genética , Glucógeno/metabolismo , Proteínas Sustrato del Receptor de Insulina/genética , Proteínas Sustrato del Receptor de Insulina/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Hígado/metabolismo , Hígado/patología , Masculino , Ratones , Ratones Noqueados , Obesidad/genética , Obesidad/metabolismo , Obesidad/patología , Fosfoenolpiruvato Carboxiquinasa (GTP)/genética , Fosfoenolpiruvato Carboxiquinasa (GTP)/metabolismo , Receptor de Insulina/genética , Receptor de Insulina/metabolismo , Factor de Transcripción STAT5/metabolismo
4.
Pigment Cell Melanoma Res ; 32(1): 9-24, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-29781575

RESUMEN

The POU domain family of transcription factors play a central role in embryogenesis and are highly expressed in neural crest cells and the developing brain. BRN2 is a class III POU domain protein that is a key mediator of neuroendocrine and melanocytic development and differentiation. While BRN2 is a central regulator in numerous developmental programs, it has also emerged as a major player in the biology of tumourigenesis. In melanoma, BRN2 has been implicated as one of the master regulators of the acquisition of invasive behaviour within the phenotype switching model of progression. As a mediator of melanoma cell phenotype switching, it coordinates the transition to a dedifferentiated, slow cycling and highly motile cell type. Its inverse expression relationship with MITF is believed to mediate tumour progression and metastasis within this model. Recent evidence has now outlined a potential epigenetic switching mechanism in melanoma cells driven by BRN2 expression that induces melanoma cell invasion. We summarize the role of BRN2 in tumour cell dissemination and metastasis in melanoma, while also examining it as a potential metastatic regulator in other tumour models.


Asunto(s)
Melanoma/patología , Factores del Dominio POU/metabolismo , Animales , Humanos , Modelos Biológicos , Invasividad Neoplásica , Metástasis de la Neoplasia , Fenotipo
5.
Pigment Cell Melanoma Res ; 31(1): 51-63, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28755520

RESUMEN

A SNP within intron4 of the interferon regulatory factor4 (IRF4) gene, rs12203592*C/T, has been independently associated with pigmentation and age-specific effects on naevus count in European-derived populations. We have characterized the cis-regulatory activity of this intronic region and using human foreskin-derived melanoblast strains, we have explored the correlation between IRF4 rs12203592 homozygous C/C and T/T genotypes with TYR enzyme activity, supporting its association with pigmentation traits. Further, higher IRF4 protein levels directed by the rs12203592*C allele were associated with increased basal proliferation but decreased cell viability following UVR, an etiological factor in melanoma development. Since UVR, and accompanying IFNγ-mediated inflammatory response, is associated with melanomagenesis, we evaluated its effects in the context of IRF4 status. Manipulation of IRF4 levels followed by IFNγ treatment revealed a subset of chemokines and immuno-evasive molecules that are sensitive to IRF4 expression level and genotype including CTLA4 and PD-L1.


Asunto(s)
Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/metabolismo , Interferón gamma/farmacología , Melanocitos/patología , Melanoma/patología , Monofenol Monooxigenasa/metabolismo , Polimorfismo de Nucleótido Simple , Antivirales/farmacología , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Regulación de la Expresión Génica , Predisposición Genética a la Enfermedad , Genotipo , Humanos , Melanocitos/efectos de los fármacos , Melanocitos/metabolismo , Melanoma/tratamiento farmacológico , Melanoma/genética , Melanoma/metabolismo , Rayos Ultravioleta
6.
Mol Cancer Res ; 15(9): 1184-1196, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28607006

RESUMEN

Exposure of melanocytes to ultraviolet radiation (UVR) induces the formation of UV lesions that can produce deleterious effects in genomic DNA. Encounters of replication forks with unrepaired UV lesions can lead to several complex phenomena, such as the formation of DNA double-strand breaks (DSBs). The NR4A family of nuclear receptors are transcription factors that have been associated with mediating DNA repair functions downstream of the MC1R signaling pathway in melanocytes. In particular, emerging evidence shows that upon DNA damage, the NR4A2 receptor can translocate to sites of UV lesion by mechanisms requiring post-translational modifications within the N-terminal domain and at a serine residue in the DNA-binding domain at position 337. Following this, NR4A2 aids in DNA repair by facilitating chromatin relaxation, allowing accessibility for DNA repair machinery. Using A2058 and HT144 melanoma cells engineered to stably express wild-type or mutant forms of the NR4A2 proteins, we reveal that the expression of functional NR4A2 is associated with elevated cytoprotection against UVR. Conversely, knockdown of NR4A2 expression by siRNA results in a significant loss of cell viability after UV insult. By analyzing the kinetics of the ensuing 53BP1 and RAD51 foci following UV irradiation, we also reveal that the expression of mutant NR4A2 isoforms, lacking the ability to translocate, transactivate, or undergo phosphorylation, display compromised repair capacity.Implications: These data expand the understanding of the mechanism by which the NR4A2 nuclear receptor can facilitate DNA DSB repair. Mol Cancer Res; 15(9); 1184-96. ©2017 AACR.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN , ADN de Neoplasias/efectos de la radiación , Melanoma/genética , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Muerte Celular/efectos de la radiación , Línea Celular Tumoral , ADN de Neoplasias/genética , Humanos , Melanocitos/metabolismo , Melanocitos/efectos de la radiación , Melanoma/metabolismo , Melanoma/patología , Melanoma/radioterapia , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/biosíntesis , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal , Transfección , Rayos Ultravioleta
7.
Int J Cancer ; 140(12): 2634-2641, 2017 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-28076901

RESUMEN

Tumour heterogeneity poses a distinct obstacle to therapeutic intervention. While the initiation of tumours across various physiological systems is frequently associated with signature mutations in genes that drive proliferation and bypass senescence, increasing evidence suggests that tumour progression and clonal diversity is driven at an epigenetic level. The tumour microenvironment plays a key role in driving diversity as cells adapt to demands imposed during tumour growth, and is thought to drive certain subpopulations back to a stem cell-like state. This stem cell-like phenotype primes tumour cells to react to external cues via the use of developmental pathways that facilitate changes in proliferation, migration and invasion. Because the dynamism of this stem cell-like state requires constant chromatin remodelling and rapid alterations at regulatory elements, it is of great therapeutic interest to identify the cell-intrinsic factors that confer these epigenetic changes that drive tumour progression. The nuclear factor one (NFI) family are transcription factors that play an important role in the development of many mammalian organ systems. While all four family members have been shown to act as either oncogenes or tumour suppressors across various cancer models, evidence has emerged implicating them as key epigenetic regulators during development and within tumours. Notably, NFIs have also been shown to regulate chromatin accessibility at distal regulatory elements that drive tumour cell dissemination and metastasis. Here we summarize the role of the NFIs in cancer, focusing largely on the potential mechanisms associated with chromatin remodelling and epigenetic modulation of gene expression.


Asunto(s)
Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Factores de Transcripción NFI/genética , Neoplasias/genética , Progresión de la Enfermedad , Estudio de Asociación del Genoma Completo/métodos , Humanos , Factores de Transcripción NFI/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Células Madre Neoplásicas/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Microambiente Tumoral/genética
8.
EBioMedicine ; 16: 63-75, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28119061

RESUMEN

While invasion and metastasis of tumour cells are the principle factor responsible for cancer related deaths, the mechanisms governing the process remain poorly defined. Moreover, phenotypic divergence of sub-populations of tumour cells is known to underpin alternative behaviors linked to tumour progression such as proliferation, survival and invasion. In the context of melanoma, heterogeneity between two transcription factors, BRN2 and MITF, has been associated with phenotypic switching between predominantly invasive and proliferative behaviors respectively. Epigenetic changes, in response to external cues, have been proposed to underpin this process, however the mechanism by which the phenotypic switch occurs is unclear. Here we report the identification of the NFIB transcription factor as a novel downstream effector of BRN2 function in melanoma cells linked to the migratory and invasive characteristics of these cells. Furthermore, the function of NFIB appears to drive an invasive phenotype through an epigenetic mechanism achieved via the upregulation of the polycomb group protein EZH2. A notable target of NFIB mediated up-regulation of EZH2 is decreased MITF expression, which further promotes a less proliferative, more invasive phenotype. Together our data reveal that NFIB has the ability to promote dynamic changes in the chromatin state of melanoma cells to facilitate migration, invasion and metastasis.


Asunto(s)
Movimiento Celular/genética , Proteína Potenciadora del Homólogo Zeste 2/genética , Proteínas de Homeodominio/genética , Melanoma/genética , Factor de Transcripción Asociado a Microftalmía/genética , Factores de Transcripción NFI/genética , Factores del Dominio POU/genética , Animales , Western Blotting , Línea Celular Tumoral , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Humanos , Masculino , Melanoma/metabolismo , Melanoma/patología , Ratones Endogámicos BALB C , Ratones Noqueados , Factor de Transcripción Asociado a Microftalmía/metabolismo , Microscopía Fluorescente , Factores de Transcripción NFI/metabolismo , Invasividad Neoplásica , Factores del Dominio POU/metabolismo , Unión Proteica , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Trasplante Heterólogo
9.
Cell Mol Life Sci ; 73(20): 3789-800, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27544210

RESUMEN

The skin forms a vital barrier between an organism's external environment, providing protection from pathogens and numerous physical and chemical threats. Moreover, the intact barrier is essential to prevent water and electrolyte loss without which terrestrial life could not be maintained. Accordingly, acute disruption of the skin through physical or chemical trauma needs to be repaired timely and efficiently as sustained skin pathologies ranging from mild irritations and inflammation through to malignancy impact considerably on morbidity and mortality. The Nuclear Hormone Receptor Family of transcriptional regulators has proven to be highly valuable targets for addressing a range of pathologies, including metabolic syndrome and cancer. Indeed members of the classic endocrine sub-group, such as the glucocorticoid, retinoid, and Vitamin D receptors, represent mainstay treatment strategies for numerous inflammatory skin disorders, though side effects from prolonged use are common. Emerging evidence has now highlighted important functional roles for nuclear receptors belonging to the adopted and orphan subgroups in skin physiology and patho-physiology. This review will focus on these subgroups and explore the current evidence that suggests these nuclear receptor hold great promise as future stand-alone or complementary drug targets in treating common skin diseases and maintaining skin homeostasis.


Asunto(s)
Salud , Terapia Molecular Dirigida , Receptores Citoplasmáticos y Nucleares/metabolismo , Enfermedades de la Piel/metabolismo , Animales , Humanos
10.
Methods Mol Biol ; 1314: 207-17, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26139270

RESUMEN

Protein screening/detection is an essential tool in many laboratories. Owing to the relatively large time investments that are required by standard protocols, the development of methods with higher throughput while maintaining an at least comparable signal-to-noise ratio is highly beneficial in many research areas. This chapter describes how cold microwave technology can be used to enhance the rate of molecular interactions and provides protocols for dot blots, Western blots, and ELISA procedures permitting a completion of all incubation steps (blocking and antibody steps) within 24-45 min.


Asunto(s)
Western Blotting/métodos , Ensayo de Inmunoadsorción Enzimática/métodos , Immunoblotting/métodos , Microondas , Proteínas/análisis , Animales , Western Blotting/instrumentación , Electroforesis en Gel de Poliacrilamida/métodos , Ensayo de Inmunoadsorción Enzimática/instrumentación , Diseño de Equipo , Humanos , Immunoblotting/instrumentación , Relación Señal-Ruido
11.
Brain Res ; 1616: 71-87, 2015 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-25960350

RESUMEN

Nuclear factor one X (NFIX) has been shown to play a pivotal role during the development of many regions of the brain, including the neocortex, the hippocampus and the cerebellum. Mechanistically, NFIX has been shown to promote neural stem cell differentiation through the activation of astrocyte-specific genes and via the repression of genes central to progenitor cell self-renewal. Interestingly, mice lacking Nfix also exhibit other phenotypes with respect to development of the central nervous system, and whose underlying causes have yet to be determined. Here we examine one of the phenotypes displayed by Nfix(-/-) mice, namely hydrocephalus. Through the examination of embryonic and postnatal Nfix(-/-) mice we reveal that hydrocephalus is first seen at around postnatal day (P) 10 in mice lacking Nfix, and is fully penetrant by P20. Furthermore, we examined the subcommissural organ (SCO), the Sylvian aqueduct and the ependymal layer of the lateral ventricles, regions that when malformed and functionally perturbed have previously been implicated in the development of hydrocephalus. SOX3 is a factor known to regulate SCO development. Although we revealed that NFIX could repress Sox3-promoter-driven transcriptional activity in vitro, SOX3 expression within the SCO was normal within Nfix(-/-) mice, and Nfix mutant mice showed no abnormalities in the structure or function of the SCO. Moreover, these mutant mice exhibited no overt blockage of the Sylvian aqueduct. However, the ependymal layer of the lateral ventricles was frequently absent in Nfix(-/-) mice, suggesting that this phenotype may underlie the development of hydrocephalus within these knockout mice.


Asunto(s)
Epéndimo/patología , Regulación del Desarrollo de la Expresión Génica/genética , Hidrocefalia/patología , Ventrículos Laterales/patología , Factores de Transcripción NFI/deficiencia , Factores de Edad , Animales , Animales Recién Nacidos , Biología Computacional , Modelos Animales de Enfermedad , Embrión de Mamíferos , Epéndimo/embriología , Epéndimo/crecimiento & desarrollo , Hidrocefalia/genética , Ventrículos Laterales/embriología , Ventrículos Laterales/crecimiento & desarrollo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Factores de Transcripción NFI/genética , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo
12.
Mol Endocrinol ; 28(7): 1166-85, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24911119

RESUMEN

Protein arginine methyltransferases (PRMTs) methylate arginine residues on histones and target transcription factors that play critical roles in many cellular processes, including gene transcription, mRNA splicing, proliferation, and differentiation. Recent studies have linked PRMT-dependent epigenetic marks and modifications to carcinogenesis and metastasis in cancer. However, the role of PRMT2-dependent signaling in breast cancer remains obscure. We demonstrate PRMT2 mRNA expression was significantly decreased in breast cancer relative to normal breast. Gene expression profiling, Ingenuity and protein-protein interaction network analysis after PRMT2-short interfering RNA transfection into MCF-7 cells, revealed that PRMT2-dependent gene expression is involved in cell-cycle regulation and checkpoint control, chromosomal instability, DNA repair, and carcinogenesis. For example, PRMT2 depletion achieved the following: 1) increased p21 and decreased cyclinD1 expression in (several) breast cancer cell lines, 2) decreased cell migration, 3) induced an increase in nucleotide excision repair and homologous recombination DNA repair, and 4) increased the probability of distance metastasis free survival (DMFS). The expression of PRMT2 and retinoid-related orphan receptor-γ (RORγ) is inversely correlated in estrogen receptor-positive breast cancer and increased RORγ expression increases DMFS. Furthermore, we found decreased expression of the PRMT2-dependent signature is significantly associated with increased probability of DMFS. Finally, weighted gene coexpression network analysis demonstrated a significant correlation between PRMT2-dependent genes and cell-cycle checkpoint, kinetochore, and DNA repair circuits. Strikingly, these PRMT2-dependent circuits are correlated with pan-cancer metagene signatures associated with epithelial-mesenchymal transition and chromosomal instability. This study demonstrates the role and significant correlation between a histone methyltransferase (PRMT2)-dependent signature, RORγ, the cell-cycle regulation, DNA repair circuits, and breast cancer survival outcomes.


Asunto(s)
Neoplasias de la Mama/mortalidad , Regulación Neoplásica de la Expresión Génica/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Proteína-Arginina N-Metiltransferasas/genética , Neoplasias de la Mama/genética , Puntos de Control del Ciclo Celular/genética , Línea Celular Tumoral , Movimiento Celular/genética , Inestabilidad Cromosómica/genética , Ciclina D1/biosíntesis , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/biosíntesis , Roturas del ADN de Doble Cadena , Reparación del ADN/genética , Transición Epitelial-Mesenquimal/genética , Femenino , Perfilación de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intracelular/biosíntesis , Cinetocoros/metabolismo , Células MCF-7 , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/biosíntesis , Mapas de Interacción de Proteínas , Proteína-Arginina N-Metiltransferasas/biosíntesis , Interferencia de ARN , ARN Mensajero/biosíntesis , ARN Interferente Pequeño , Receptores de Estrógenos/metabolismo , Transducción de Señal
13.
Exp Dermatol ; 23(7): 449-52, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24758341

RESUMEN

Ultraviolet radiation (UVR) is the most common mutagen that melanocytes are exposed to. UVR causes a diverse range of DNA photolesions contributing to genome instability and promotes melanoma and non-melanoma development. Melanocytes are pigment-producing cells that synthesise the photoprotective melanins when the melanocortin-1 receptor (MC1R) is activated. MC1R is a G-protein-coupled receptor expressed predominantly in melanocytes. Its signalling pathway has been directly linked to melanogenesis, enhanced cytoprotection against UV damage and augmented DNA repair response. Interestingly, previous studies have revealed that MC1R signalling induces the transcription of the NR4A subfamily of orphan nuclear receptors in response to UV. In line with this, studies have also observed that NR4A receptors are recruited to distinct nuclear foci in response to cellular stress, independent of their transcriptional roles. Here, we review the regulated expression of NR4A2 and its potential roles upon cellular stress conditions. Current work in developing synthetic NR4A2 agonists further provides exciting avenues for exploring the potential role of NR4A2 as an antiskin cancer drug target.


Asunto(s)
Reparación del ADN , Regulación de la Expresión Génica , Melanocitos/citología , Melanocitos/efectos de la radiación , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Receptor de Melanocortina Tipo 1/metabolismo , ADN/efectos de la radiación , Daño del ADN , Predisposición Genética a la Enfermedad , Humanos , Luz , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/metabolismo , Rayos Ultravioleta
14.
J Neurosci ; 34(8): 2921-30, 2014 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-24553933

RESUMEN

Epigenetic mechanisms are essential in regulating neural progenitor cell self-renewal, with the chromatin-modifying protein Enhancer of zeste homolog 2 (EZH2) emerging as a central player in promoting progenitor cell self-renewal during cortical development. Despite this, how Ezh2 is itself regulated remains unclear. Here, we demonstrate that the transcription factor nuclear factor IB (NFIB) plays a key role in this process. Nfib(-/-) mice exhibit an increased number of proliferative ventricular zone cells that express progenitor cell markers and upregulation of EZH2 expression within the neocortex and hippocampus. NFIB binds to the Ezh2 promoter and overexpression of NFIB represses Ezh2 transcription. Finally, key downstream targets of EZH2-mediated epigenetic repression are misregulated in Nfib(-/-) mice. Collectively, these results suggest that the downregulation of Ezh2 transcription by NFIB is an important component of the process of neural progenitor cell differentiation during cortical development.


Asunto(s)
Corteza Cerebral/crecimiento & desarrollo , Epigénesis Genética/fisiología , Factores de Transcripción NFI/genética , Factores de Transcripción NFI/fisiología , Complejo Represivo Polycomb 2/genética , Complejo Represivo Polycomb 2/fisiología , Animales , Recuento de Células , Corteza Cerebral/citología , Corteza Cerebral/fisiología , Ensayo de Cambio de Movilidad Electroforética , Proteína Potenciadora del Homólogo Zeste 2 , Femenino , Hipocampo/citología , Hipocampo/crecimiento & desarrollo , Inmunohistoquímica , Masculino , Ratones , Ratones Noqueados , Análisis por Micromatrices , Mutación/genética , Mutación/fisiología , Células-Madre Neurales/fisiología , Cultivo Primario de Células , Regiones Promotoras Genéticas/genética , Reacción en Cadena en Tiempo Real de la Polimerasa
15.
Cereb Cortex ; 24(1): 261-79, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23042739

RESUMEN

Neural progenitor cells have the ability to give rise to neurons and glia in the embryonic, postnatal and adult brain. During development, the program regulating whether these cells divide and self-renew or exit the cell cycle and differentiate is tightly controlled, and imbalances to the normal trajectory of this process can lead to severe functional consequences. However, our understanding of the molecular regulation of these fundamental events remains limited. Moreover, processes underpinning development of the postnatal neurogenic niches within the cortex remain poorly defined. Here, we demonstrate that Nuclear factor one X (NFIX) is expressed by neural progenitor cells within the embryonic hippocampus, and that progenitor cell differentiation is delayed within Nfix(-/-) mice. Moreover, we reveal that the morphology of the dentate gyrus in postnatal Nfix(-/-) mice is abnormal, with fewer subgranular zone neural progenitor cells being generated in the absence of this transcription factor. Mechanistically, we demonstrate that the progenitor cell maintenance factor Sry-related HMG box 9 (SOX9) is upregulated in the hippocampus of Nfix(-/-) mice and demonstrate that NFIX can repress Sox9 promoter-driven transcription. Collectively, our findings demonstrate that NFIX plays a central role in hippocampal morphogenesis, regulating the formation of neuronal and glial populations within this structure.


Asunto(s)
Diferenciación Celular/fisiología , Hipocampo/embriología , Factores de Transcripción NFI/fisiología , Células-Madre Neurales/fisiología , Animales , Recuento de Células , Colorantes , Biología Computacional , Giro Dentado/embriología , Giro Dentado/crecimiento & desarrollo , Giro Dentado/fisiología , Ensayo de Cambio de Movilidad Electroforética , Electroporación , Femenino , Hematoxilina , Hipocampo/citología , Hipocampo/metabolismo , Inmunohistoquímica , Hibridación in Situ , Luciferasas/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis por Micromatrices , Factores de Transcripción NFI/genética , Células-Madre Neurales/metabolismo , Adhesión en Parafina , Embarazo , Regiones Promotoras Genéticas/genética , Reacción en Cadena en Tiempo Real de la Polimerasa
16.
PLoS One ; 8(11): e78075, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24223135

RESUMEN

Ultraviolet radiation (UVR) is one of the most common mutagens encountered by humans and induces the formation of cyclobutane pyrimidine dimers (CPDs) and pyrimidine-(6-4)-pyrimidone photoproduct (6-4PP) lesions in the genomic DNA. To prevent the accumulation of deleterious mutations these lesions must be efficiently repaired, primarily by nucleotide excision repair. We have previously demonstrated that the NR4A family of nuclear receptors are crucial mediators of the DNA repair function of the MC1R signalling pathway in melanocytes. Here we explore the role of the NR4A2 protein in the DNA repair process further. Using EYFP tagged-NR4A2 we have demonstrated a UVR induced recruitment to distinct nuclear foci where they co-localise with known DNA repair proteins. We reveal that the N-terminal domain of the receptor is required for this translocation and identify a role for p38 and PARP signalling in this process. Moreover disruption of the functional integrity of the Ligand Binding Domain of the receptor by deleting the terminal helix 12 effectively blocks co-localisation of the receptor with DNA repair factors. Restored co-localisation of the mutant receptor with DNA repair proteins in the presence of a Histone Deacetylase Inhibitor suggests that impaired chromatin accessibility underpins the mis-localisation observed. Finally NR4A2 over-expression facilitated a more efficient clearance of UVR induced CPD and 6-4PP lesions. Taken together these data uncover a novel role for the NR4A nuclear receptors as direct facilitators of nucleotide excision repair.


Asunto(s)
Núcleo Celular/metabolismo , Reparación del ADN , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Rayos Ultravioleta , Sitios de Unión , Línea Celular Tumoral , Núcleo Celular/efectos de la radiación , Enzimas Reparadoras del ADN/metabolismo , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Ácidos Hidroxámicos/farmacología , Sistema de Señalización de MAP Quinasas , Poli(ADP-Ribosa) Polimerasa-1 , Poli(ADP-Ribosa) Polimerasas/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Transporte de Proteínas
17.
Cell ; 155(5): 1022-33, 2013 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-24267888

RESUMEN

Sequence polymorphisms linked to human diseases and phenotypes in genome-wide association studies often affect noncoding regions. A SNP within an intron of the gene encoding Interferon Regulatory Factor 4 (IRF4), a transcription factor with no known role in melanocyte biology, is strongly associated with sensitivity of skin to sun exposure, freckles, blue eyes, and brown hair color. Here, we demonstrate that this SNP lies within an enhancer of IRF4 transcription in melanocytes. The allele associated with this pigmentation phenotype impairs binding of the TFAP2A transcription factor that, together with the melanocyte master regulator MITF, regulates activity of the enhancer. Assays in zebrafish and mice reveal that IRF4 cooperates with MITF to activate expression of Tyrosinase (TYR), an essential enzyme in melanin synthesis. Our findings provide a clear example of a noncoding polymorphism that affects a phenotype by modulating a developmental gene regulatory network.


Asunto(s)
Factores Reguladores del Interferón/metabolismo , Polimorfismo de Nucleótido Simple , Animales , Secuencia de Bases , Elementos de Facilitación Genéticos , Humanos , Factores Reguladores del Interferón/química , Factores Reguladores del Interferón/genética , Melanocitos/metabolismo , Ratones , Datos de Secuencia Molecular , Pigmentación , Transducción de Señal , Factor de Transcripción AP-2/química , Factor de Transcripción AP-2/metabolismo , Pez Cebra
18.
Cell Mol Neurobiol ; 33(7): 867-73, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23852417

RESUMEN

The transcription factor nuclear factor one X (NFIX) plays a central role during the development of the neocortex and hippocampus, through the activation of astrocyte-specific gene expression and the repression of progenitor-specific pathways. However, our understanding of transcriptional targets of NFIX during cortical development remains limited. Here, we identify the transcription factor Bobby sox (Bbx) as a target for NFI-mediated transcriptional control. BBX is expressed within ventricular zone progenitor cells within the developing neocortex and hippocampus, and its expression is upregulated in Nfix (-/-) mice. Moreover, we reveal that NFIX can repress Bbx promoter-driven expression. Collectively, these data suggest that Bbx is a downstream target of NFIX during development of the forebrain.


Asunto(s)
Factores de Transcripción NFI/metabolismo , Prosencéfalo/embriología , Prosencéfalo/metabolismo , Transactivadores/metabolismo , Animales , Secuencia de Bases , Hipocampo/citología , Hipocampo/metabolismo , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Regiones Promotoras Genéticas/genética , Prosencéfalo/citología , Transcripción Genética , Regulación hacia Arriba
19.
Microsc Microanal ; 19(3): 507-12, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23578755

RESUMEN

Replication of chloroplast in plant cells is an essential process that requires co-assembly of the tubulin-like plastid division proteins FtsZ1 and FtsZ2 at mid-chloroplast to form a ring structure called the Z-ring. The Z-ring is stabilized via its interaction with the transmembrane protein ARC6 on the inner envelope membrane of chloroplasts. Plants lacking ARC6 are defective in plastid division and contain only one or two enlarged chloroplasts per cell with abnormal localization of FtsZ: instead of a single Z-ring, many short FtsZ filaments are distributed throughout the chloroplast. ARC6 is thought to be the anchoring point for FtsZ assemblies. To investigate the role of ARC6 in FtsZ anchoring, the mobility of green fluorescent protein-tagged FtsZ assemblies was assessed by single particle tracking in mutant plants lacking the ARC6 protein. Mean square displacement analysis showed that the mobility of FtsZ assemblies is to a large extent characterized by anomalous diffusion behavior (indicative of intermittent binding) and restricted diffusion suggesting that besides ARC6-mediated anchoring, an additional FtsZ-anchoring mechanism is present in chloroplasts.


Asunto(s)
Proteínas de Arabidopsis/metabolismo , Proteínas de Cloroplastos/metabolismo , Cloroplastos/fisiología , Replicación del ADN , Arabidopsis , Genes Reporteros , Proteínas Fluorescentes Verdes/análisis , Proteínas Fluorescentes Verdes/genética , Microscopía Fluorescente , Unión Proteica , Mapeo de Interacción de Proteínas , Coloración y Etiquetado/métodos
20.
Arch Biochem Biophys ; 513(2): 94-101, 2011 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-21781955

RESUMEN

FtsZ was identified in bacteria as the first protein to localize mid-cell prior to division and homologs have been found in many plant species. Bacterial studies demonstrated that FtsZ forms a ring structure that is dynamically exchanged with a soluble pool of FtsZ. Our previous work established that Arabidopsis FtsZ1 and FtsZ2-1 are capable of in vitro self-assembly into two distinct filament types, termed type-I and type-II and noted the presence of filament precursor molecules which prompted this investigation. Using a combination of electron microscopy, gel chromatography and native PAGE revealed that (i) prior to FtsZ assembly initiation the pool consists solely of dimers and (ii) during assembly of the Arabidopsis FtsZ type-II filaments the most common intermediate between the dimer and filament state is a tetramer. Three-dimensional reconstructions of the observed dimer and tetramer suggest these oligomeric forms may represent consecutive steps in type-II filament assembly and a mechanism is proposed, which is expanded to include FtsZ assembly into type-I filaments. Finally, the results permit a discussion of the oligomeric nature of the soluble pool in plants.


Asunto(s)
Proteínas de Arabidopsis/química , Arabidopsis/química , Arabidopsis/ultraestructura , Proteínas de Arabidopsis/ultraestructura , Dimerización , Imagenología Tridimensional , Microscopía Electrónica de Transmisión , Modelos Moleculares , Plastidios/química , Multimerización de Proteína , Estructura Cuaternaria de Proteína
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...