Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros

Base de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Adv Med Sci ; 69(2): 320-330, 2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38901547

RESUMEN

PURPOSE: Nuclear receptor subfamily 2 group E member 1 (Nr2e1) has been regarded as an essential regulator in neural stem cells. However, its function is still not clear in hepatocytes. This study aimed to clarify the effects of Nr2e1-deficiency in hepatocytes in lipotoxic conditions. MATERIALS/METHODS: Nr2e1-knockdown AML12 â€‹cells were generated by lentiviral vector transfection. The influences of Nr2e1-deficiency on hepatocyte survival were determined by cell cycle progression and cell apoptosis rate using flow cytometry. Real-time quantitative PCR and Western blot were used to examine the genes and protein expression related to apoptosis, lipid metabolism, and oxidative stress. Meanwhile, RNA sequencing was adopted in liver samples from Nr2e1-knockout (Nr2e1-KO) mice. RESULTS: Nr2e1 expression was observed with a significant decrease in AML12 â€‹cells after palmitic acid-stimulation. Knockdown of Nr2e1 in AML12 â€‹cells resulted in increased sensitivity to lipotoxicity, evidenced by a partial G0/G1 cell-cycle arrest and higher rates of cell apoptosis. Moreover, Nr2e1-knockdown AML12 â€‹cells presented increased gene expressions relative to lipid synthesis but decreased levels of ß-oxidation related genes. Lack of Nr2e1 augmented palmitate-induced oxidative stress in hepatocytes. In vivo, differential genes in Nr2e1-KO mice liver were enriched in pathways associated with liver regeneration and cell proliferation. CONCLUSIONS: This study indicated that hepatocytes lacking Nr2e1 were more susceptible to lipotoxic-mediated damage. Nr2e1 may serve as a potential target for the development of novel therapies for lipotoxicity-induced liver injury.

2.
Life Sci Space Res (Amst) ; 35: 170-179, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36336363

RESUMEN

Neurogenesis is an essential, lifelong process during which neural stem cells generate new neurons within the hippocampus, a center for learning, memory, and mood control. Neural stem cells are vulnerable to environmental insults spanning from chronic stress to radiation. These insults reduce their numbers and diminish neurogenesis, leading to memory decline, anxiety, and depression. Preserving neural stem cells could thus help prevent these neurogenesis-associated pathologies, an outcome particularly important for long-term space missions where environmental exposure to radiation is significantly higher than on Earth. Multiple developments, from mechanistic discoveries of radiation injury on hippocampal neurogenesis to new platforms for the development of selective, specific, effective, and safe small molecules as neurogenesis-protective agents hold great promise to minimize radiation damage on neurogenesis. In this review, we summarize the effects of space-like radiation on hippocampal neurogenesis. We then focus on current advances in drug discovery and development and discuss the nuclear receptor TLX/NR2E1 (oleic acid receptor) as an example of a neurogenic target that might rescue neurogenesis following radiation.


Asunto(s)
Astronautas , Traumatismos por Radiación , Humanos , Neurogénesis/fisiología , Neurogénesis/efectos de la radiación , Hipocampo/patología , Cognición , Traumatismos por Radiación/prevención & control
3.
Biochim Biophys Acta Mol Basis Dis ; 1868(11): 166515, 2022 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-35932893

RESUMEN

Development of targeted therapies will be a critical step towards reducing the mortality associated with triple-negative breast cancer (TNBC). To achieve this, we searched for targets that met three criteria: (1) pharmacologically targetable, (2) expressed in TNBC, and (3) expression is prognostic in TNBC patients. Since nuclear receptors have a well-defined ligand-binding domain and are thus highly amenable to small-molecule intervention, we focused on this class of protein. Our analysis identified TLX (NR2E1) as a candidate. Specifically, elevated tumoral TLX expression was associated with prolonged recurrence-free survival and overall survival for breast cancer patients with either estrogen receptor alpha (ERα)-negative or basal-like tumors. Using two TNBC cell lines, we found that stable overexpression of TLX impairs in vitro proliferation. RNA-Seq analysis revealed that TLX reduced the expression of genes implicated in epithelial-mesenchymal transition (EMT), a cellular program known to drive metastatic progression. Indeed, TLX overexpression significantly decreased cell migration and invasion, and robustly decreased the metastatic capacity of TNBC cells in murine models. We identify SERPINB2 as a likely mediator of these effects. Taken together, our work indicates that TLX impedes the progression of TNBC. Several ligands have been shown to regulate the transcriptional activity of TLX, providing a framework for the future development of this receptor for therapeutic intervention.


Asunto(s)
Neoplasias de la Mama Triple Negativas , Animales , Transición Epitelial-Mesenquimal/genética , Receptor alfa de Estrógeno/genética , Humanos , Ligandos , Ratones , Receptores Nucleares Huérfanos/uso terapéutico , Receptores Citoplasmáticos y Nucleares/genética , Neoplasias de la Mama Triple Negativas/metabolismo
4.
Proc Natl Acad Sci U S A ; 119(13): e2023784119, 2022 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-35333654

RESUMEN

Neural stem cells, the source of newborn neurons in the adult hippocampus, are intimately involved in learning and memory, mood, and stress response. Despite considerable progress in understanding the biology of neural stem cells and neurogenesis, regulating the neural stem cell population precisely has remained elusive because we have lacked the specific targets to stimulate their proliferation and neurogenesis. The orphan nuclear receptor TLX/NR2E1 governs neural stem and progenitor cell self-renewal and proliferation, but the precise mechanism by which it accomplishes this is not well understood because its endogenous ligand is not known. Here, we identify oleic acid (18:1ω9 monounsaturated fatty acid) as such a ligand. We first show that oleic acid is critical for neural stem cell survival. Next, we demonstrate that it binds to TLX to convert it from a transcriptional repressor to a transcriptional activator of cell-cycle and neurogenesis genes, which in turn increases neural stem cell mitotic activity and drives hippocampal neurogenesis in mice. Interestingly, oleic acid-activated TLX strongly up-regulates cell cycle genes while only modestly up-regulating neurogenic genes. We propose a model in which sufficient quantities of this endogenous ligand must bind to TLX to trigger the switch to proliferation and drive the progeny toward neuronal lineage. Oleic acid thus serves as a metabolic regulator of TLX activity that can be used to selectively target neural stem cells, paving the way for future therapeutic manipulations to counteract pathogenic impairments of neurogenesis.


Asunto(s)
Hipocampo , Neurogénesis , Ácido Oléico , Receptores Citoplasmáticos y Nucleares , Animales , Proliferación Celular , Hipocampo/crecimiento & desarrollo , Hipocampo/metabolismo , Ligandos , Ratones , Neurogénesis/fisiología , Ácido Oléico/metabolismo , Receptores Nucleares Huérfanos , Receptores Citoplasmáticos y Nucleares/metabolismo
5.
Endocrinology ; 162(11)2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34463725

RESUMEN

TLX (NR2E1), an orphan member of the nuclear receptor superfamily, is a transcription factor that has been described to be generally repressive in nature. It has been implicated in several aspects of physiology and disease. TLX is best known for its ability to regulate the proliferation of neural stem cells and retinal progenitor cells. Dysregulation, overexpression, or loss of TLX expression has been characterized in numerous studies focused on a diverse range of pathological conditions, including abnormal brain development, psychiatric disorders, retinopathies, metabolic disease, and malignant neoplasm. Despite the lack of an identified endogenous ligand, several studies have described putative synthetic and natural TLX ligands, suggesting that this receptor may serve as a therapeutic target. Therefore, this article aims to briefly review what is known about TLX structure and function in normal physiology, and provide an overview of TLX in regard to pathological conditions. Particular emphasis is placed on TLX and cancer, and the potential utility of this receptor as a therapeutic target.


Asunto(s)
Fenómenos Fisiológicos Celulares/genética , Enfermedad/genética , Receptores Nucleares Huérfanos/fisiología , Animales , Encéfalo/embriología , Encéfalo/crecimiento & desarrollo , Humanos , Terapia Molecular Dirigida/métodos , Terapia Molecular Dirigida/tendencias , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/terapia , Neurogénesis/genética , Receptores Nucleares Huérfanos/genética
6.
Life Sci ; 278: 119562, 2021 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-33915130

RESUMEN

AIMS: To investigate the nuclear receptor subfamily 2 group E member 1 (Nr2e1) expression in adipose tissues of obese mice and assess the role of Nr2e1 in insulin resistance and chronic inflammation of the adipose tissues. MAIN METHODS: An obese model was established in Nr2e1 knockout (KO) mice and their wild type (WT) littermates through a long-term high-fat diet (HFD) feeding regime. The epididymal fat weight, body weight, and daily food intake were recorded. The blood lipid profile, blood inflammatory factors, and the levels of fasting blood glucose (FBG) and fasting insulin were determined. We estimated insulin resistance by the homeostasis model assessment (HOMA). The expression of inflammatory factors and F4/80 was examined by polymerase chain reaction (PCR) and western blotting to assess adipose tissues inflammation. We also determined the molecules of insulin signaling and the nuclear factor kappa B (NF-κB) pathway by western blotting. KEY FINDINGS: The Nr2e1 expression was upregulated in WT obese mice when compared with that in control mice. Despite a lower body weight and epididymal fat mass in Nr2e1-/- mice, these rats showed increased inflammatory cytokines secretion, more pronounced hyperlipidemia, and impaired insulin sensitivity after HFD treatment. Further investigation revealed that Nr2e1 deletion affected the expression of insulin signaling and NF-κB pathway-related molecules in visceral adipose tissues. SIGNIFICANCE: Nr2e1 may act as a potential target to improve insulin sensitivity and inflammation in obesity and related complications.


Asunto(s)
Resistencia a la Insulina , Grasa Intraabdominal/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Alimentación Animal , Animales , Glucemia/metabolismo , Peso Corporal , Enfermedad Crónica , Cruzamientos Genéticos , Modelos Animales de Enfermedad , Intolerancia a la Glucosa , Prueba de Tolerancia a la Glucosa , Homeostasis , Hiperlipidemias/metabolismo , Inflamación , Insulina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Subunidad p50 de NF-kappa B/metabolismo , Obesidad/metabolismo , Transducción de Señal
7.
Psychiatry Res ; 293: 113386, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32805587

RESUMEN

NR2E1 is implicated in the regulation of neurogenesis and considered as a candidate gene for schizophrenia. We resequenced all the exons of NR2E1 in 547 patients with schizophrenia and 567 controls from Taiwan. We identified five common SNPs with no association with patients with schizophrenia. Further haplotype-based association analysis showed that two haplotypes within NR2E1 were correlated with the schizophrenia risk. Four rare mutations located at untranslated regions were identified in patients with schizophrenia but not in our control sample. The present study suggests that NR2E1 is likely to play a significant role in conferring susceptibility to schizophrenia.


Asunto(s)
Estudios de Asociación Genética/métodos , Predisposición Genética a la Enfermedad/genética , Receptores Citoplasmáticos y Nucleares/genética , Esquizofrenia/diagnóstico , Esquizofrenia/genética , Adulto , Exones/genética , Femenino , Predisposición Genética a la Enfermedad/epidemiología , Haplotipos/genética , Humanos , Masculino , Persona de Mediana Edad , Mutación/genética , Receptores Nucleares Huérfanos , Polimorfismo de Nucleótido Simple/genética , Esquizofrenia/epidemiología , Taiwán/epidemiología
8.
Biomed Pharmacother ; 120: 109503, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31590127

RESUMEN

Nonalcoholic fatty liver disease (NAFLD) is a common and complex metabolic disorder. Despite the widespread concern, there are still few effective treatments except lifestyle interventions. Nuclear receptor subfamily 2 group E member 1 (Nr2e1) is a transcription factor which regulates many biological processes, including development, growth, and differentiation of nerve cells. However, its specific function in hepatocyte is still unknown. In the present study, we found that the expression of Nr2e1 decreased in the livers of high-fat diet-fed mice. We generated Nr2e1 knockout (KO) mice and studied whether Nr2e1 ablation was related to NAFLD. We found that typical pathological features of NAFLD, including insulin resistance, hepatic steatosis, and inflammation, were present in Nr2e1-KO mice or high-fat diet-induced mice models. In conclusion, Nr2e1 ablation promotes liver steatosis and systemic insulin resistance. Nr2e1 may play a protective role in the formation of NAFLD and may serve as a worthy therapeutic target for NAFLD.


Asunto(s)
Dieta Alta en Grasa , Glucosa/metabolismo , Inflamación/etiología , Hígado/metabolismo , Obesidad/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Animales , Hígado Graso/inducido químicamente , Regulación de la Expresión Génica/efectos de los fármacos , Intolerancia a la Glucosa , Resistencia a la Insulina , Metabolismo de los Lípidos/efectos de los fármacos , Ratones , Ratones Noqueados , Receptores Citoplasmáticos y Nucleares/genética
9.
Am J Med Genet B Neuropsychiatr Genet ; 180(2): 138-149, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30468562

RESUMEN

Although the loss of brain laterality is one of the most consistent modalities in schizophrenia (SCZ) and bipolar disorder (BD), its molecular basis remains elusive. Our limited previous studies indicated that epigenetic modifications are key to the asymmetric transcriptomes of brain hemispheres. We used whole-genome expression microarrays to profile postmortem brain samples from subjects with SCZ, psychotic BD [BD[+]] or non-psychotic BD [BD(-)], or matched controls (10/group) and performed whole-genome DNA methylation (DNAM) profiling of the same samples (3-4/group) to identify pathways associated with SCZ or BD[+] and genes/sites susceptible to epigenetic regulation. qRT-PCR and quantitative DNAM analysis were employed to validate findings in larger sample sets (35/group). Gene Set Enrichment Analysis (GSEA) demonstrated that BMP signaling and astrocyte and cerebral cortex development are significantly (FDR q < 0.25) coordinately upregulated in both SCZ and BD[+], and glutamate signaling and TGFß signaling are significantly coordinately upregulated in SCZ. GSEA also indicated that collagens are downregulated in right versus left brain of controls, but not in SCZ or BD[+] patients. Ingenuity Pathway Analysis predicted that TGFB2 is an upstream regulator of these genes (p = .0012). While lateralized expression of TGFB2 in controls (p = .017) is associated with a corresponding change in DNAM (p ≤ .023), lateralized expression and DNAM of TGFB2 are absent in SCZ or BD. Loss of brain laterality in SCZ and BD corresponds to aberrant epigenetic regulation of TGFB2 and changes in TGFß signaling, indicating potential avenues for disease prevention/treatment.


Asunto(s)
Trastorno Bipolar/genética , Encéfalo/patología , Esquizofrenia/genética , Adulto , Autopsia , Metilación de ADN/genética , Epigénesis Genética/genética , Epigenoma/genética , Femenino , Predisposición Genética a la Enfermedad/genética , Humanos , Masculino , Persona de Mediana Edad , Regiones Promotoras Genéticas , Trastornos Psicóticos/genética , Transducción de Señal/genética , Transcriptoma/genética , Factor de Crecimiento Transformador beta/metabolismo , Secuenciación Completa del Genoma/métodos
10.
Cancer Rep (Hoboken) ; 2(5): e1204, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-32721119

RESUMEN

BACKGROUND: The human orphan receptor TLX (NR2E1) is a key regulator of neurogenesis, adult stem cell maintenance, and tumorigenesis. However, little is known about the genetic and transcriptomic events that occur following TLX overexpression in human cell lines. AIMS: Here, we used cytogenetics and RNA sequencing to investigate the effect of TLX overexpression with an inducible vector system in the HEK 293T cell line. METHODS AND RESULTS: Conventional spectral karyotyping was used to identify chromosomal abnormalities, followed by fluorescence in situ hybridization (FISH) analysis on chromosome spreads to assess TLX DNA copy number. Illumina paired-end whole transcriptome sequencing was then performed to characterize recurrent genetic variants (single nucleotide polymorphisms (SNPs) and indels), expressed gene fusions, and gene expression profiles. Lastly, flow cytometry was used to analyze cell cycle distribution. Intriguingly, we show that upon transfection with a vector containing the human TLX gene (eGFP-hTLX), an isochromosome forms on the long arm of chromosome 6, thereby resulting in DNA gain of the TLX locus (6q21) and upregulation of TLX. Induction of the eGFP-hTLX vector further increased TLX expression levels, leading to G0-G1 cell cycle arrest, genetic aberrations, modulation of gene expression patterns, and crosstalk with other nuclear receptors (AR, ESR1, ESR2, NR1H4, and NR3C2). We identified a 49-gene signature associated with central nervous system (CNS) development and carcinogenesis, in addition to potentially cancer-driving gene fusions (LARP1-CNOT8 and NSL1-ZDBF2) and deleterious genetic variants (frameshift insertions in the CTSH, DBF4, POSTN, and WDR78 genes). CONCLUSION: Taken together, these findings illustrate that TLX may play a pivotal role in tumorigenesis via genomic instability and perturbation of cancer-related processes.


Asunto(s)
Transformación Celular Neoplásica/genética , Inestabilidad Genómica , Receptores Nucleares Huérfanos/metabolismo , Proliferación Celular/genética , Mutación del Sistema de Lectura , Puntos de Control de la Fase G1 del Ciclo Celular/genética , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Humanos , Receptores Nucleares Huérfanos/genética , RNA-Seq , Regulación hacia Arriba
11.
Molecules ; 23(11)2018 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-30441799

RESUMEN

Orphan nuclear receptor TLX (NR2E1) plays a critical role in the regulation of neural stem cells (NSC) as well as in the development of NSC-derived brain tumors. In the last years, new data have emerged implicating TLX in prostate and breast cancer. Therefore, inhibitors of TLX transcriptional activity may have a significant impact on the treatment of several critical malignancies. However, the TLX protein possesses a non-canonical ligand-binding domain (LBD), which lacks a ligand-binding pocket (conventionally targeted in case of nuclear receptors) that complicates the development of small molecule inhibitors of TLX. Herein, we utilized a rational structure-based design approach to identify small molecules targeting the Atro-box binding site of human TLX LBD. As a result of virtual screening of ~7 million molecular structures, 97 compounds were identified and evaluated in the TLX-responsive luciferase reporter assay. Among those, three chemicals demonstrated 40⁻50% inhibition of luciferase-detected transcriptional activity of the TLX orphan nuclear receptor at a dose of 35 µM. The identified compounds represent the first class of small molecule inhibitors of TLX transcriptional activity identified via methods of computer-aided drug discovery.


Asunto(s)
Diseño Asistido por Computadora , Diseño de Fármacos , Modelos Moleculares , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Receptores Citoplasmáticos y Nucleares/genética , Activación Transcripcional/efectos de los fármacos , Sitios de Unión , Regulación de la Expresión Génica , Genes Reporteros , Humanos , Conformación Molecular , Estructura Molecular , Receptores Nucleares Huérfanos , Unión Proteica , Relación Estructura-Actividad Cuantitativa
12.
FASEB J ; 32(2): 613-624, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28970252

RESUMEN

Hippocampal neurogenesis is a lifelong process whereby new neurons are produced and integrate into the host circuitry within the hippocampus. It is regulated by a multitude of extrinsic and intrinsic regulators and is believed to contribute to certain hippocampal-dependent cognitive tasks. Hippocampal neurogenesis and associated cognition have been demonstrated to be impaired after increases in the levels of proinflammatory cytokine IL-1ß in the hippocampus, such as that which occurs in various neurodegenerative and psychiatric disorders. IL-1ß also suppresses the expression of TLX (orphan nuclear receptor tailless homolog), which is an orphan nuclear receptor that functions to promote neural progenitor cell (NPC) proliferation and suppress neuronal differentiation; therefore, manipulation of TLX represents a potential strategy with which to prevent the antiproliferative effects of IL-1ß. In this study, we assessed the mechanism that underlies IL-1ß-induced changes in TLX expression and determined the protective capacity of TLX to mitigate the effects of IL-1ß on embryonic rat hippocampal neurosphere expansion. We demonstrate that IL-1ß activated the NF-κB pathway in proliferating NPCs and that this activation was responsible for IL-1ß-induced changes in TLX expression. In addition, we report that enhancing TLX expression prevented the IL-1ß-induced suppression of neurosphere expansion. Thus, we highlight TLX as a potential protective regulator of the antiproliferative effects of IL-1ß on hippocampal neurogenesis.-Ó'Léime, C. S., Kozareva, D. A., Hoban, A. E., Long-Smith, C. M., Cryan, J. F., Nolan, Y. M. TLX is an intrinsic regulator of the negative effects of IL-1ß on proliferating hippocampal neural progenitor cells.


Asunto(s)
Proliferación Celular , Regulación de la Expresión Génica , Hipocampo/metabolismo , Interleucina-1beta/metabolismo , Células-Madre Neurales/metabolismo , Receptores Citoplasmáticos y Nucleares/biosíntesis , Animales , Células Cultivadas , Hipocampo/citología , FN-kappa B/metabolismo , Células-Madre Neurales/citología , Neurogénesis , Ratas , Transducción de Señal , Esferoides Celulares/citología , Esferoides Celulares/metabolismo
13.
Genes Brain Behav ; 17(3): e12357, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-27790850

RESUMEN

The orphan nuclear receptor Tlx (Nr2e1) is a key regulator of both embryonic and adult hippocampal neurogenesis. Several different mouse models have been developed which target Tlx in vivo including spontaneous deletion models (from birth) and targeted and conditional knockouts. Although some conflicting findings have been reported, for the most part studies have demonstrated that Tlx is important in regulating processes that underlie neurogenesis, spatial learning, anxiety-like behaviour and interestingly, aggression. More recent data have demonstrated that disrupting Tlx during early life induces hyperactivity and that Tlx plays a role in emotional regulation. Moreover, there are sex- and age-related differences in some behaviours in Tlx knockout mice during adolescence and adulthood. Here, we discuss the role of Tlx in motor-, cognitive-, aggressive- and anxiety-related behaviours during adolescence and adulthood. We examine current evidence which provides insight into Tlx during neurodevelopment, and offer our thoughts on the function of Tlx in brain and behaviour. We further hypothesize that Tlx is a key target in understanding the emergence of neurobiological disorders during adolescence and early adulthood.


Asunto(s)
Conducta/fisiología , Receptores Citoplasmáticos y Nucleares/fisiología , Animales , Hipocampo/fisiología , Humanos , Neurogénesis/fisiología
14.
Brain Behav Immun ; 66: 394-412, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28751020

RESUMEN

Hippocampal neurogenesis, the process by which new neurons are born and develop into the host circuitry, begins during embryonic development and persists throughout adulthood. Over the last decade considerable insights have been made into the role of hippocampal neurogenesis in cognitive function and the cellular mechanisms behind this process. Additionally, an increasing amount of evidence exists on the impact of environmental factors, such as stress and neuroinflammation on hippocampal neurogenesis and subsequent impairments in cognition. Elevated expression of the pro-inflammatory cytokine interleukin-1ß (IL-1ß) in the hippocampus is established as a significant contributor to the neuronal demise evident in many neurological and psychiatric disorders and is now known to negatively regulate hippocampal neurogenesis. In order to prevent the deleterious effects of IL-1ß on neurogenesis it is necessary to identify signalling pathways and regulators of neurogenesis within neural progenitor cells that can interact with IL-1ß. Nuclear receptors are ligand regulated transcription factors that are involved in modulating a large number of cellular processes including neurogenesis. In this review we focus on the signalling mechanisms of specific nuclear receptors involved in regulating neurogenesis (glucocorticoid receptors, peroxisome proliferator activated receptors, estrogen receptors, and nuclear receptor subfamily 2 group E member 1 (NR2E1 or TLX)). We propose that these nuclear receptors could be targeted to inhibit neuroinflammatory signalling pathways associated with IL-1ß. We discuss their potential to be therapeutic targets for neuroinflammatory disorders affecting hippocampal neurogenesis and associated cognitive function.


Asunto(s)
Encefalitis/metabolismo , Hipocampo/metabolismo , Interleucina-1beta/metabolismo , Neurogénesis , Neuronas/metabolismo , Animales , Encefalitis/terapia , Humanos , Células-Madre Neurales/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Transducción de Señal
15.
Curr Top Dev Biol ; 125: 257-273, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28527574

RESUMEN

The nuclear receptor TLX (NR2E1) is a transcription factor that is critical for neural development and adult neurogenesis through its actions in regulating neural stem cell proliferation, self-renewal, and fate determination. These roles are primarily executed by regulating TLX downstream target genes involved in myriad pathways such as cell cycle progression, RNA processing, angiogenesis, and senescence. Recent studies suggest that dysregulation of TLX pathways plays an important role in the pathogenesis of human neurological disorders and brain tumors. Here, we will highlight recent progress in the roles of TLX in brain development and adult neurogenesis, and the relevance of TLX to neurological diseases and brain tumors. We will also discuss the potential of TLX as a therapeutic target for these disorders.


Asunto(s)
Encéfalo/embriología , Neurogénesis/genética , Receptores Citoplasmáticos y Nucleares/fisiología , Animales , Encéfalo/metabolismo , Neoplasias Encefálicas/genética , Regulación de la Expresión Génica , Humanos , Ratones , Enfermedades del Sistema Nervioso/genética , Enfermedades del Sistema Nervioso/metabolismo , Receptores Nucleares Huérfanos , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/metabolismo
16.
Biomed Environ Sci ; 30(3): 185-193, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28427488

RESUMEN

OBJECTIVE: This study aimed to investigate the expression pattern and function of Nuclear receptor subfamily 2 group E member 1 (Nr2e1) in retinoic acid (RA)-induced brain abnormality. METHODS: The mouse model of brain abnormality was established by administering 28 mg/kg RA, and neural stem cells (NSCs) were isolated from the mouse embryo and cultured in vitro. Nr2e1 expression was detected by whole mount in situ hybridization, RT-PCR, and Western blotting. Nr2e1 function was determined by transducing Nr2e1 shRNA into NSCs, and the effect on the sonic hedgehog (Shh) signaling pathway was assessed in the cells. In addition, the regulation of Nr2e1 expression by RA was also determined in vitro. RESULTS: Nr2e1 expression was significantly downregulated in the brain and NSCs of RA-treated mouse embryos, and knockdown of Nr2e1 affected the proliferation of NSCs in vitro. In addition, a similar expression pattern of Nr2e1 and RA receptor (RAR) α was observed after treatment of NSCs with different concentrations of RA. CONCLUSION: Our study demonstrated that Nr2e1 could be regulated by RA, which would aid a better understanding of the mechanism underlying RA-induced brain abnormality.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Células-Madre Neurales/efectos de los fármacos , Receptores Citoplasmáticos y Nucleares/metabolismo , Tretinoina/farmacología , Animales , Encéfalo/citología , Encéfalo/embriología , Proliferación Celular , Regulación hacia Abajo , Regulación de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , Células-Madre Neurales/fisiología , Receptores Citoplasmáticos y Nucleares/genética
17.
Mol Neurobiol ; 54(1): 272-280, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-26738856

RESUMEN

The orphan nuclear receptor TLX, also called NR2E1, is a factor important in the regulation of neural stem cell (NSC) self-renewal, neurogenesis, and maintenance. As a transcription factor, TLX is vital for the expression of genes implicated in neurogenesis, such as DNA replication, cell cycle, adhesion and migration. It acts by way of repressing or activating target genes, as well as controlling protein-protein interactions. Growing evidence suggests that dysregulated TLX acts in the initiation and progression of human disorders of the nervous system. This review describes recent knowledge about TLX expression, structure, targets, and biological functions, relevant to maintaining adult neural stem cells related to both neuropsychiatric conditions and certain nervous system tumours.


Asunto(s)
Células-Madre Neurales/fisiología , Neurogénesis/fisiología , Receptores Citoplasmáticos y Nucleares/fisiología , Animales , Neoplasias del Sistema Nervioso Central/genética , Neoplasias del Sistema Nervioso Central/metabolismo , Estado de Salud , Humanos , Trastornos Mentales/genética , Trastornos Mentales/metabolismo , Células-Madre Neurales/química , Receptores Nucleares Huérfanos
18.
BMC Genomics ; 17(1): 832, 2016 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-27782803

RESUMEN

BACKGROUND: NR2E1 (Tlx) is an orphan nuclear receptor that regulates the maintenance and self-renewal of neural stem cells, and promotes tumourigenesis. Nr2e1-null mice exhibit reduced cortical and limbic structures and pronounced retinal dystrophy. NR2E1 functions mainly as a repressor of gene transcription in association with the co-repressors atrophin-1, LSD1, HDAC and BCL11A. Recent evidence suggests that NR2E1 also acts as an activator of gene transcription. However, co-activator complexes that interact with NR2E1 have not yet been identified. In order to find potential novel co-regulators for NR2E1, we used a microarray assay for real-time analysis of co-regulator-nuclear receptor interaction (MARCoNI) that contains peptides representing interaction motifs from potential co-regulatory proteins, including known co-activator nuclear receptor box sequences (LxxLL motif). RESULTS: We found that NR2E1 binds strongly to an atrophin-1 peptide (Atro box) used as positive control and to 19 other peptides that constitute candidate NR2E1 partners. Two of these proteins, p300 and androgen receptor (AR), were further validated by reciprocal pull-down assays. The specificity of NR2E1 binding to peptides in the array was evaluated using two single amino acid variants, R274G and R276Q, which disrupted the majority of the binding interactions observed with wild-type NR2E1. The decreased binding affinity of these variants to co-regulators was further validated by pull-down assays using atrophin1 as bait. Despite the high conservation of arginine 274 in vertebrates, its reduced interactions with co-regulators were not significant in vivo as determined by retinal phenotype analysis in single-copy Nr2e1-null mice carrying the variant R274G. CONCLUSIONS: We showed that MARCoNI is a specific assay to test interactions of NR2E1 with candidate co-regulators. In this way, we unveiled 19 potential co-regulator partners for NR2E1, including eight co-activators. All the candidates here identified need to be further validated using in vitro and in vivo models. This assay was sensitive to point mutations in NR2E1 ligand binding domain making it useful to identify mutations and/or small molecules that alter binding of NR2E1 to protein partners.


Asunto(s)
Ligandos , Receptores Nucleares Huérfanos/agonistas , Receptores Citoplasmáticos y Nucleares/agonistas , Secuencia de Aminoácidos , Animales , Proteínas Portadoras , Descubrimiento de Drogas , Variación Genética , Humanos , Ratones , Ratones Noqueados , Receptores Nucleares Huérfanos/química , Receptores Nucleares Huérfanos/metabolismo , Fenotipo , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Mapeo de Interacción de Proteínas , Receptores Citoplasmáticos y Nucleares/química , Receptores Citoplasmáticos y Nucleares/metabolismo
19.
Behav Brain Res ; 306: 36-47, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-26970576

RESUMEN

The nuclear receptor Tlx is a key regulator of embryonic and adult hippocampal neurogenesis and has been genetically linked to bipolar disorder. Mice lacking Tlx (Nr2e1(-/-)) display deficits in adult hippocampal neurogenesis and behavioural abnormalities. However, whether Tlx regulates behaviour during adolescence or in a sex-dependent manner remains unexplored. Therefore, we investigated the role of Tlx in a series of behavioural tasks in adolescent male and female mice with a spontaneous deletion of Tlx (Nr2e1(-/-) mice). Testing commenced at adolescence (postnatal day 28) and continued until adulthood (postnatal day 67). Adolescent male and female Nr2e1(-/-) mice were hyperactive in an open field, an effect that persisted in adulthood. Male but not female Nr2e1(-/-) mice exhibited reduced thigmotaxis during adolescence and adulthood. Impairments in rotarod motor performance developed in male and female Nr2e1(-/-) mice at the onset of adulthood. Spontaneous alternation in the Y-maze, a hippocampus-dependent task, was impaired in adolescent but not adult male and female Nr2e1(-/-) mice. Contextual fear conditioning was impaired in adolescent male Nr2e1(-/-) mice only, but both male and female adolescent Nr2e1(-/-) mice showed impaired cued fear conditioning, a hippocampal-amygdala dependent cognitive process. These deficits persisted into adulthood in males but not females. In conclusion, deletion of Tlx impairs motor, cognitive and anxiety-related behaviours during adolescence and adulthood in male and female mice with most effects occurring during adolescence rather than adulthood, independent of housing conditions. This suggests that Tlx has functions beyond regulation of adult hippocampal neurogenesis, and may be an important target in understanding neurobiological disorders.


Asunto(s)
Envejecimiento , Ansiedad/genética , Trastornos del Conocimiento/genética , Hipercinesia/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Caracteres Sexuales , Envejecimiento/genética , Animales , Animales Recién Nacidos , Peso Corporal/genética , Condicionamiento Clásico/fisiología , Señales (Psicología) , Conducta Exploratoria/fisiología , Miedo/fisiología , Femenino , Locomoción/genética , Masculino , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Transgénicos , Receptores Citoplasmáticos y Nucleares/genética , Reflejo/genética
20.
Biochim Biophys Acta ; 1849(2): 210-6, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24930777

RESUMEN

The orphan nuclear receptor TLX, also known as NR2E1, is an essential regulator of neural stem cell (NSC) self-renewal, maintenance, and neurogenesis. In vertebrates, TLX is specifically localized to the neurogenic regions of the forebrain and retina throughout development and adulthood. TLX regulates the expression of genes involved in multiple pathways, such as the cell cycle, DNA replication, and cell adhesion. These roles are primarily performed through the transcriptional repression or activation of downstream target genes. Emerging evidence suggests that the misregulation of TLX might play a role in the onset and progression of human neurological disorders making this factor an ideal therapeutic target. Here, we review the current understanding of TLX function, expression, regulation, and activity significant to NSC maintenance, adult neurogenesis, and brain plasticity. This article is part of a Special Issue entitled: Nuclear receptors in animal development.


Asunto(s)
Células-Madre Neurales/fisiología , Neurogénesis/genética , Receptores Citoplasmáticos y Nucleares/fisiología , Adulto , Animales , Diferenciación Celular/genética , Humanos , Plasticidad Neuronal/genética , Receptores Nucleares Huérfanos , Receptores Citoplasmáticos y Nucleares/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA