Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
J Comp Neurol ; 529(12): 3206-3221, 2021 08.
Article in English | MEDLINE | ID: mdl-33855704

ABSTRACT

The genetic profile of vertebrate pallia has long driven debate on homology across distantly related clades. Based on an expression profile of the orphan nuclear receptor NR4A2 in mouse and chicken brains, Puelles et al. (The Journal of Comparative Neurology, 2016, 524, 665-703) concluded that the avian lateral mesopallium is homologous to the mammalian claustrum, and the medial mesopallium homologous to the insula cortex. They argued that their findings contradict conclusions by Jarvis et al. (The Journal of Comparative Neurology, 2013, 521, 3614-3665) and Chen et al. (The Journal of Comparative Neurology, 2013, 521, 3666-3701) that the hyperpallium densocellare is instead a mesopallium cell population, and by Suzuki and Hirata (Frontiers in Neuroanatomy, 2014, 8, 783) that the avian mesopallium is homologous to mammalian cortical layers 2/3. Here, we find that NR4A2 is an activity-dependent gene and cannot be used to determine brain organization or species relationships without considering behavioral state. Activity-dependent NR4A2 expression has been previously demonstrated in the rodent brain, with the highest induction occurring within the claustrum, amygdala, deep and superficial cortical layers, and hippocampus. In the zebra finch, we find that NR4A2 is constitutively expressed in the arcopallium, but induced in parts of the mesopallium, and in sparse cells within the hyperpallium, depending on animal stimulus or behavioral state. Basal and induced NR4A2 expression patterns do not discount the previously named avian hyperpallium densocellare as dorsal mesopallium and conflict with proposed homology between the avian mesopallium and mammalian claustrum/insula at the exclusion of other brain regions. Broadly, these findings highlight the importance of controlling for behavioral state and neural activity to genetically define brain cell population relationships within and across species.


Subject(s)
Brain Chemistry/physiology , Brain/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 2/biosynthesis , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Vocalization, Animal/physiology , Animals , Chickens , Finches , Male , Mice , Species Specificity
2.
J Neurosci ; 40(18): 3675-3691, 2020 04 29.
Article in English | MEDLINE | ID: mdl-32238479

ABSTRACT

The transcription factor Nurr1 has been identified to be ectopically induced in the striatum of rodents expressing l-DOPA-induced dyskinesia (LID). In the present study, we sought to characterize Nurr1 as a causative factor in LID expression. We used rAAV2/5 to overexpress Nurr1 or GFP in the parkinsonian striatum of LID-resistant Lewis or LID-prone Fischer-344 (F344) male rats. In a second cohort, rats received the Nurr1 agonist amodiaquine (AQ) together with l-DOPA or ropinirole. All rats received a chronic DA agonist and were evaluated for LID severity. Finally, we performed single-unit recordings and dendritic spine analyses on striatal medium spiny neurons (MSNs) in drug-naïve rAAV-injected male parkinsonian rats. rAAV-GFP injected LID-resistant hemi-parkinsonian Lewis rats displayed mild LID and no induction of striatal Nurr1 despite receiving a high dose of l-DOPA. However, Lewis rats overexpressing Nurr1 developed severe LID. Nurr1 agonism with AQ exacerbated LID in F344 rats. We additionally determined that in l-DOPA-naïve rats striatal rAAV-Nurr1 overexpression (1) increased cortically-evoked firing in a subpopulation of identified striatonigral MSNs, and (2) altered spine density and thin-spine morphology on striatal MSNs; both phenomena mimicking changes seen in dyskinetic rats. Finally, we provide postmortem evidence of Nurr1 expression in striatal neurons of l-DOPA-treated PD patients. Our data demonstrate that ectopic induction of striatal Nurr1 is capable of inducing LID behavior and associated neuropathology, even in resistant subjects. These data support a direct role of Nurr1 in aberrant neuronal plasticity and LID induction, providing a potential novel target for therapeutic development.SIGNIFICANCE STATEMENT The transcription factor Nurr1 is ectopically induced in striatal neurons of rats exhibiting levodopa-induced dyskinesia [LID; a side-effect to dopamine replacement strategies in Parkinson's disease (PD)]. Here we asked whether Nurr1 is causing LID. Indeed, rAAV-mediated expression of Nurr1 in striatal neurons was sufficient to overcome LID-resistance, and Nurr1 agonism exacerbated LID severity in dyskinetic rats. Moreover, we found that expression of Nurr1 in l-DOPA naïve hemi-parkinsonian rats resulted in the formation of morphologic and electrophysiological signatures of maladaptive neuronal plasticity; a phenomenon associated with LID. Finally, we determined that ectopic Nurr1 expression can be found in the putamen of l-DOPA-treated PD patients. These data suggest that striatal Nurr1 is an important mediator of the formation of LID.


Subject(s)
Corpus Striatum/metabolism , Dyskinesia, Drug-Induced/metabolism , Levodopa/toxicity , Nuclear Receptor Subfamily 4, Group A, Member 2/biosynthesis , Parkinsonian Disorders/metabolism , Aged , Animals , Corpus Striatum/drug effects , Dyskinesia, Drug-Induced/pathology , Female , Humans , Male , Oxidopamine/toxicity , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/pathology , Rats , Rats, Inbred F344 , Rats, Inbred Lew , Rats, Sprague-Dawley
3.
CNS Neurosci Ther ; 26(1): 55-65, 2020 01.
Article in English | MEDLINE | ID: mdl-31087449

ABSTRACT

BACKGROUND: Neural stem cells (NSCs) transplantation is considered a promising treatment for Parkinson's disease. But most NSCs are differentiated into glial cells rather than neurons, and only a few of them survive after transplantation due to the inflammatory environment. METHODS: In this study, neural stem cells (NSCs) and microglial cells both forced with the Nurr1 gene were transplanted into the striatum of the rat model of PD. The results were evaluated through reverse transcription polymerase chain reaction (RT-PCR), Western blot, and immunofluorescence analysis. RESULTS: The behavioral abnormalities of PD rats were improved by combined transplantation of NSCs and microglia, both forced with Nurr1. The number of tyrosine hydroxylase+ cells in the striatum of PD rats increased, and the number of Iba1+ cells decreased compared with the other groups. Moreover, the dopamine neurons differentiated from grafted NSCs could still be detected in the striatum of PD rats after 5 months. CONCLUSIONS: The results suggested that transplantation of Nurr1-overexpressing NSCs and microglia could improve the inhospitable host brain environments, which will be  a new potential strategy for the cell replacement therapy in PD.


Subject(s)
Genetic Therapy/methods , Microglia/transplantation , Neural Stem Cells/transplantation , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Parkinsonian Disorders/therapy , Stem Cell Transplantation/methods , Amphetamine , Animals , Behavior, Animal , Calcium-Binding Proteins/genetics , Cell Differentiation , Corpus Striatum/surgery , Dopaminergic Neurons/transplantation , Encephalitis/therapy , Female , Hydroxydopamines , Male , Microfilament Proteins/genetics , Nuclear Receptor Subfamily 4, Group A, Member 2/biosynthesis , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/psychology , Rats , Rats, Sprague-Dawley
4.
Mol Med Rep ; 21(1): 107-114, 2020 01.
Article in English | MEDLINE | ID: mdl-31746417

ABSTRACT

Nuclear receptor related 1 protein (Nurr1), a member of the nuclear receptor 4 family of orphan nuclear receptors, has been reported to display anti­inflammatory properties. The present study investigated the alteration of Nurr1 immunoreactivity in the gerbil hippocampus proper following 5 min of transient global cerebral ischemia. In sham operated gerbils, Nurr1 immunoreactivity was observed in pyramidal neurons in all cornu ammonis 1­3 (CA1­3) subfields of the hippocampus proper. In ischemia­operated gerbils, Nurr1 immunoreactivity was altered in the CA1 subfield. Nurr1 immunoreactivity in CA1 pyramidal neurons gradually decreased until 2 days post­ischemia, and, at 4 days post­ischemia, Nurr1 immunoreactivity was concentrated in CA1 pyramidal neurons. Additionally, Nurr1 immunoreactivity was newly expressed in microglia in the CA1 subfield at 4 days post­ischemia. Conversely, in the CA2/3 subfield, time­dependent alteration of Nurr1 immunoreactivity was not identified at any time following ischemia. These results indicated that the alteration of Nurr1 expression in the CA1 subfield in the hippocampus may be associated with the death of CA1 pyramidal neurons.


Subject(s)
Brain Ischemia/metabolism , CA1 Region, Hippocampal/metabolism , Gene Expression Regulation , Nuclear Receptor Subfamily 4, Group A, Member 2/biosynthesis , Pyramidal Cells/metabolism , Animals , Brain Ischemia/pathology , CA1 Region, Hippocampal/pathology , Gerbillinae , Male , Pyramidal Cells/pathology
5.
Med Hypotheses ; 131: 109302, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31443765

ABSTRACT

Parkinson's disease (PD) patients have higher rates of melanoma and vice versa, observations suggesting that the two conditions may share common pathogenic pathways. ß-Catenin is a transcriptional cofactor that, when concentrated in the nucleus, upregulates the expression of canonical Wnt target genes, such as Nurr1, many of which are important for neuronal survival. ß-Catenin-mediated activity is decreased in sporadic PD as well as in leucine-rich repeat kinase 2 (LRRK2) and ß-glucosidase (GBA) mutation cellular models of PD, which is the most common genetic cause of and risk for PD, respectively. In addition, ß-catenin expression is significantly decreased in more aggressive and metastatic melanoma. Multiple observational studies have shown smokers to have significantly lower rates of PD as well as melanoma implying that tobacco may contain one or more elements that protect against both conditions. In support, smoker's brains have significantly reduced levels of α-synuclein, a pathological intracellular protein found in PD brain and melanoma cells. Tobacco contains very high lithium levels compared to other plants. Lithium has a broad array of neuroprotective actions, including enhancing autophagy and reducing intracellular α-synuclein levels, and is effective in both neurotoxin and transgenic preclinical PD models. One of lithium's neuroprotective actions is enhancement of ß-catenin-mediated activity leading to increased Nurr1 expression through its ability to inhibit glycogen synthase kinase-3 ß (GSK-3ß). Lithium also has anti-proliferative effects on melanoma cells and the clinical use of lithium is associated with a reduced incidence of melanoma as well as reduced melanoma-associated mortality. This is the first known report hypothesizing that inhaled lithium from smoking may account for the associated reduced rates of both PD and melanoma and that this protection may be mediated, in part, through lithium-induced GSK-3ß inhibition and consequent enhanced ß-catenin-mediated activity. This hypothesis could be directly tested in clinical trials assessing lithium therapy's ability to affect ß-catenin-mediated activity and slow disease progression in patients with PD or melanoma.


Subject(s)
Lithium/pharmacology , Melanoma/prevention & control , Models, Biological , Neuroprotective Agents/pharmacology , Nicotiana/chemistry , Parkinson Disease/prevention & control , Smokers , Wnt Signaling Pathway/drug effects , beta Catenin/physiology , Alzheimer Disease/drug therapy , Alzheimer Disease/epidemiology , Alzheimer Disease/prevention & control , Autophagy/drug effects , Brain Chemistry/drug effects , Drug Evaluation, Preclinical , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta/physiology , Humans , Incidence , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Lithium/analysis , Lithium/therapeutic use , Lithium Carbonate/therapeutic use , Melanoma/epidemiology , Mutation , Neuroprotective Agents/analysis , Nuclear Receptor Subfamily 4, Group A, Member 2/biosynthesis , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Parkinson Disease/epidemiology , Parkinsonian Disorders/drug therapy , Water/chemistry , Wnt Signaling Pathway/physiology , alpha-Synuclein/metabolism , beta-Glucosidase/genetics
6.
Am J Physiol Cell Physiol ; 317(3): C513-C524, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31188636

ABSTRACT

Sustained elevation of sympathetic activity is an important contributor to pathological cardiac hypertrophy, ventricular arrhythmias, and left ventricular contractile dysfunction in chronic heart failure. The orphan nuclear receptor NR4A2 is an immediate early-response gene activated in the heart under ß-adrenergic stimulation. The goal of this study was to identify the transcriptional remodeling events induced by increased NR4A2 expression in cardiomyocytes and their impact on the physiological response of those cells to sustained ß-adrenergic stimulation. Treatment of adult rat ventricular myocytes with isoproterenol induced a rapid (<4 h) increase in NR4A2 levels that was accompanied by a transient (<24 h) increase in nuclear localization of the transcription factor. Adenovirus-mediated overexpression of NR4A2 to similar levels modulated the expression of genes linked to adrenoceptor signaling, calcium signaling, cell growth and proliferation and counteracted the increase in protein synthesis rate and cell surface area mediated by chronic isoproterenol stimulation. Consistent with those findings, NR4A2 overexpression also blocked the phosphorylative activation of growth-related kinases ERK1/2, Akt, and p70 S6 kinase. Prominent among the transcriptional changes induced by NR4A2 was the upregulation of the dual-specificity phosphatases DUSP2 and DUSP14, two known inhibitors of ERK1/2. Pretreatment of NR4A2-overexpressing cardiomyocytes with the DUSP inhibitor BCI [(E)-2-benzylidene-3-(cyclohexylamino)-2,3-dihydro-1H-inden-1-one] prevented the inhibition of ERK1/2 following isoproterenol stimulation. In conclusion, our results suggest that NR4A2 acts as a novel negative feedback regulator of the ß-adrenergic receptor-mediated growth response in cardiomyocytes and this at least partly through DUSP-mediated inhibition of ERK1/2 signaling.


Subject(s)
Adrenergic beta-Agonists/pharmacology , Cell Proliferation/physiology , MAP Kinase Signaling System/physiology , Myocytes, Cardiac/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 2/biosynthesis , Receptors, Adrenergic, beta/metabolism , Age Factors , Animals , Cell Proliferation/drug effects , Cells, Cultured , MAP Kinase Signaling System/drug effects , Male , Myocytes, Cardiac/drug effects , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Rats , Rats, Sprague-Dawley
7.
Biomed Res Int ; 2019: 6843265, 2019.
Article in English | MEDLINE | ID: mdl-30949504

ABSTRACT

The aim of this study was to investigate the effect of minocycline in rats with rotenone-induced Parkinson's disease (PD). The open field test was performed to determine the motor ability of the rats. Double immunofluorescence staining was used to detect the expression of tyrosine hydroxylase (TH) and Nurr1 in the substantia nigra (SN) of rats. The relative protein levels of TH, Nurr1, and the total- and phosphorylated-cAMP-response element binding protein (CREB) were determined by western blot analysis. The production of reactive oxygen species (ROS) and nitric oxide (NO) was detected by commercial kits. After exposure to rotenone for 28 days, rats exhibited decreased ambulation and rearing frequency and prolonged immobility time with loss of TH positive neurons in the SN. The phosphorylation levels of CREB and Nurr1 expression decreased significantly accompanied with the release of ROS and NO. Minocycline alleviated the motor deficits of rats lesioned by rotenone and elevated the expression of TH, as well as suppressing the release of ROS and NO in the SN. That was in line with the elevated phosphorylation levels of CREB and Nurr1 expression. In conclusion, our present study showed minocycline protected against neurotoxicity in a rotenone-induced rat model of PD, which was correlated with upregulation of Nurr1.


Subject(s)
Minocycline/pharmacology , Nuclear Receptor Subfamily 4, Group A, Member 2/biosynthesis , Parkinsonian Disorders , Rotenone/administration & dosage , Substantia Nigra/metabolism , Up-Regulation/drug effects , Animals , Male , Nitric Oxide/metabolism , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/metabolism , Parkinsonian Disorders/pathology , Parkinsonian Disorders/prevention & control , Rats , Rats, Wistar , Rotenone/pharmacology , Substantia Nigra/pathology
8.
J Neuroimmunol ; 330: 48-54, 2019 05 15.
Article in English | MEDLINE | ID: mdl-30825859

ABSTRACT

Inflammatory responses are considered to play pivotal roles in the pathogenesis of intracerebral hemorrhage (ICH). Here we show that a nuclear receptor Nurr1 (NR4A2) was expressed prominently in microglia/macrophages and astrocytes in the perihematomal region in the striatum of mice after ICH. Daily administration of a Nurr1 agonist amodiaquine (40 mg/kg, i.p.) from 3 h after ICH induction diminished perihematomal activation of microglia/macrophages and astrocytes. Amodiaquine also suppressed ICH-induced mRNA expression of IL-1ß, CCL2 and CXCL2, and ameliorated motor dysfunction of mice. These results suggest that Nurr1 serves a novel target for ICH therapy.


Subject(s)
Amodiaquine/therapeutic use , Cerebral Hemorrhage/drug therapy , Disease Models, Animal , Inflammation Mediators/antagonists & inhibitors , Nervous System Diseases/prevention & control , Nuclear Receptor Subfamily 4, Group A, Member 2/agonists , Amodiaquine/pharmacology , Animals , Cerebral Hemorrhage/metabolism , Inflammation Mediators/metabolism , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Inbred ICR , Microglia/drug effects , Microglia/metabolism , Nervous System Diseases/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 2/biosynthesis
9.
CNS Neurosci Ther ; 24(9): 790-800, 2018 09.
Article in English | MEDLINE | ID: mdl-29450981

ABSTRACT

INTRODUCTION: Neural stem cells (NSCs) are the most promising cells for cell replacement therapy for Parkinson's disease (PD). However, a majority of the transplanted NSCs differentiated into glial cells, thereby limiting the clinical application. Previous studies indicated that chronic neuroinflammation plays a vital role in the degeneration of midbrain DA (mDA) neurons, which suggested the developing potential of therapies for PD by targeting the inflammatory processes. Thus, Nurr1 (nuclear receptor-related factor 1), a transcription factor, has been referred to play a pivotal role in both the differentiation of dopaminergic neurons in embryonic stages and the maintenance of the dopaminergic phenotype throughout life. AIM: This study investigated the effect of Nurr1 on neuroinflammation and differentiation of NSCs cocultured with primary microglia in the transwell coculture system. RESULTS: The results showed that Nurr1 exerted anti-inflammatory effects and promoted the differentiation of NSCs into dopaminergic neurons. CONCLUSIONS: The results suggested that Nurr1 protects dopaminergic neurons from neuroinflammation insults by limiting the production of neurotoxic mediators by microglia and maintain the survival of transplanted NSCs. These phenomena provided a new theoretical and experimental foundation for the transplantation of Nurr1-overexpressed NSCs as a potential treatment of PD.


Subject(s)
Dopaminergic Neurons/metabolism , Inflammation Mediators/metabolism , Microglia/metabolism , Neural Stem Cells/metabolism , Neurogenesis/physiology , Nuclear Receptor Subfamily 4, Group A, Member 2/biosynthesis , Animals , Animals, Newborn , Cell Differentiation/physiology , Cells, Cultured , Coculture Techniques , HEK293 Cells , Humans , Inflammation Mediators/antagonists & inhibitors , Rats , Rats, Sprague-Dawley
10.
Mol Neurobiol ; 55(3): 2443-2453, 2018 03.
Article in English | MEDLINE | ID: mdl-28365874

ABSTRACT

Developmental vitamin D (DVD) deficiency has been proposed as an important risk factor for schizophrenia. Our previous study using Sprague Dawley rats found that DVD deficiency disrupted the ontogeny of mesencephalic dopamine neurons by decreasing the mRNA level of a crucial differentiation factor of dopamine cells, the nuclear receptor related 1 protein (Nurr1). However, it remains unknown whether this reflects a reduction in dopamine cell number or in Nurr1 expression. It is also unclear if any particular subset of developing dopamine neurons in the mesencephalon is selectively affected. In this study, we employed state-of-the-art spinning disk confocal microscopy optimized for the imaging of tissue sections and 3D segmentation to assess post-mitotic dopamine cells on a single-cell basis in the rat mesencephalon at embryonic day 15. Our results showed that DVD deficiency did not alter the number, morphology, or positioning of post-mitotic dopamine cells. However, the ratio of Nurr1+TH+ cells in the substantia nigra pars compacta (SNc) compared with the ventral tegmental area (VTA) was increased in DVD-deficient embryos. In addition, the expression of Nurr1 in immature dopamine cells and mature dopamine neurons in the VTA was decreased in DVD-deficient group. Tyrosine hydroxylase was selectively reduced in SNc of DVD-deficient mesencephalon. We conclude that DVD deficiency induced early alterations in mesencephalic dopamine development may in part explain the abnormal dopamine-related behaviors found in this model. Our findings may have broader implications for how certain environmental risk factors for schizophrenia may shape the ontogeny of dopaminergic systems and by inference increase the risk of schizophrenia.


Subject(s)
Dopaminergic Neurons/metabolism , Mesencephalon/metabolism , Mitosis/physiology , Nuclear Receptor Subfamily 4, Group A, Member 2/biosynthesis , Tyrosine 3-Monooxygenase/biosynthesis , Vitamin D Deficiency/metabolism , Animals , Dopaminergic Neurons/pathology , Gene Expression , Mesencephalon/pathology , Nuclear Receptor Subfamily 4, Group A, Member 2/antagonists & inhibitors , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Rats , Rats, Sprague-Dawley , Tyrosine 3-Monooxygenase/antagonists & inhibitors , Tyrosine 3-Monooxygenase/genetics , Vitamin D Deficiency/genetics , Vitamin D Deficiency/pathology
11.
J Neurol Sci ; 379: 58-63, 2017 Aug 15.
Article in English | MEDLINE | ID: mdl-28716280

ABSTRACT

INTRODUCTION: The NR4A2 transcription factor is important in the development, survival and phenotype of dopaminergic neurons and it is postulated as a possible biomarker for Parkinson's disease (PD). Therefore, our aim was to analyze in a sample of a Mexican population with idiopathic PD, mutations (in two hotspot mutation regions) and two polymorphisms (rs34884856 in promotor and rs35479735 intronic regions) of the NR4A2 gene. We also evaluate the levels of NR4A2 gene expression in peripheral blood for a Mexican population, and identify whether they are associated with NR4A2 gene polymorphisms. METHODS: We conducted a case-control study, which included 227 idiopathic PD cases and 454 unrelated controls. Genetic variants of the NR4A2 gene were genotyped by high-resolution melting (HRM) and validated by an automated sequencing method. The gene expression was performed in peripheral blood using a real-time polymerase chain reaction. RESULTS: The rs35479735 polymorphism was associated with a higher risk of developing PD. In addition, NR4A2 gene expression was significantly decreased in patients with PD. Linkage disequilibrium analysis showed a haplotype H4 (3C-3G) that showed lower levels of expression, and contained the risk alleles for both polymorphisms. CONCLUSIONS: In summary, this is the first study in a Mexican population that considers the analysis of NR4A2 in patients with PD. An association was identified between genotype and mRNA expression levels of NR4A2 in patients with PD. These results suggest that polymorphisms and expression of the NR4A2 gene could play an important role in the risk of developing PD in Mexican populations.


Subject(s)
Genetic Association Studies/methods , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Parkinson Disease/epidemiology , Parkinson Disease/genetics , Polymorphism, Genetic/genetics , Population Surveillance , Aged , Case-Control Studies , Cohort Studies , Female , Gene Expression , Genetic Predisposition to Disease/epidemiology , Genetic Predisposition to Disease/genetics , Genetic Variation/genetics , Genotype , Humans , Male , Mexico/epidemiology , Middle Aged , Nuclear Receptor Subfamily 4, Group A, Member 2/biosynthesis , Parkinson Disease/diagnosis , RNA, Messenger/biosynthesis , RNA, Messenger/genetics
12.
Mol Cancer Res ; 15(9): 1184-1196, 2017 09.
Article in English | MEDLINE | ID: mdl-28607006

ABSTRACT

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.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair , DNA, Neoplasm/radiation effects , Melanoma/genetics , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Cell Death/radiation effects , Cell Line, Tumor , DNA, Neoplasm/genetics , Humans , Melanocytes/metabolism , Melanocytes/radiation effects , Melanoma/metabolism , Melanoma/pathology , Melanoma/radiotherapy , Nuclear Receptor Subfamily 4, Group A, Member 2/biosynthesis , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Transfection , Ultraviolet Rays
13.
ACS Chem Neurosci ; 8(7): 1455-1458, 2017 07 19.
Article in English | MEDLINE | ID: mdl-28452458

ABSTRACT

Nuclear receptor related 1 (NURR1) is an essential protein for maintenance of dopaminergic neurons in adult midbrain of which deficiency leads to Parkinson's disease. To enhance the NURR1 production of neural cells, various approaches are under investigation. Here we report that NURR1 is highly expressed in stem cells by exposure to an L-polarized blue light emitting diode (LED). Compared to stem cells cultured in the absence of a LED, under polarized green and red LEDs, the stem cells exposed to a polarized blue LED significantly enhanced neuronal biomarkers such as neurofilament M (NFM) and neuron specific enolase (NSE) at both mRNA and protein levels. In particular, NURR1 was selectively enhanced by the stem cells exposed to the L-polarized blue LED. Stem cells exposed to the L-polarized blue LED increased mitochondrial ATP and intracellular calcium ions, which support neuronal differentiation of the stem cells. This study suggests that chiro-optical treatments by using polarized light with a specific wavelength can be used for engineering of stem cells with enhanced specific biochemicals, which may open a new method for a specific disease.


Subject(s)
Light , Mesenchymal Stem Cells/metabolism , Neurogenesis , Nuclear Receptor Subfamily 4, Group A, Member 2/biosynthesis , Adenosine Triphosphate/metabolism , Calcium/metabolism , Cell Culture Techniques/instrumentation , Cell Survival , Child , Female , Fluorescent Antibody Technique , Gene Expression/radiation effects , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/radiation effects , Mitochondria/metabolism , Mitochondria/radiation effects , Neurofilament Proteins/biosynthesis , Neurofilament Proteins/genetics , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Palatine Tonsil , Phosphopyruvate Hydratase/biosynthesis , Phosphopyruvate Hydratase/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction
14.
Oncotarget ; 8(14): 23289-23302, 2017 Apr 04.
Article in English | MEDLINE | ID: mdl-28423575

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the most malignant cancers. Conventional therapies are limited due to the human liver being such a unique organ and easily showing side-effects. The unclear molecular mechanisms are tough challenges for scientists searching for new and effective anti-HCC targeting drugs. We identified that the nuclear receptor NR4A2 is a novel oncogene in HCC progression. In this study, we show that NR4A2 and the notch recceptor Notch1 were expressed highly in primary HCC tissues and immortal HCC cells by using qPCR, western blot and immuno-histochemistry assays. Both genes were observed to stimulate HCC cell proliferation, anti-apoptosis and cell cycle arrest by using cell proliferation assays and FACS assays. We also observed that the four notch receptor subtypes (Notch1-4) displayed different effects on HCC cell growth. The over-expression of Notch1 by transiently transfecting the intracellular domain of Notch1 (ICN1, Notch1 active form) increased the expression of NR4A2, with the knockdown of Notch1 decreasing NR4A2. This indicates that NR4A2 is one of the Notch-mediated downstream genes. Moreover, both NR4A2 and Notch1 suppressed the expression of tumor suppressors p21 and p63. These findings support that Notch1/NR4A2 co-regulate HCC cell functions by playing oncogenic roles and regulating the associated downstream signaling pathways. Novel Notch1/NR4A2-mediated oncogenic signaling may provide us a great opportunity for anti-HCC drug development.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Receptor, Notch1/metabolism , Apoptosis/physiology , Carcinoma, Hepatocellular/genetics , Cell Differentiation/physiology , Cell Line, Tumor , Gene Knockdown Techniques , Hep G2 Cells , Humans , Liver Neoplasms/genetics , Nuclear Receptor Subfamily 4, Group A, Member 2/biosynthesis , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Receptor, Notch1/biosynthesis , Receptor, Notch1/genetics , Signal Transduction , Transfection , Up-Regulation
15.
J Comp Neurol ; 525(8): 1962-1979, 2017 Jun 01.
Article in English | MEDLINE | ID: mdl-28177524

ABSTRACT

The Nuclear receptor subfamily 4 group A member 2 (Nr4a2) is crucial for the formation or maintenance of dopaminergic neurons in the central nervous system including the retina, where dopaminergic amacrine cells contribute to visual function. Little is known about which cells express Nr4a2 at which developmental stage. Furthermore, whether Nr4a2 functions in combination with other genes is poorly understood. Thus, we generated a novel transgenic to visualize Nr4a2 expression in vivo during zebrafish retinogenesis. A 4.1 kb fragment of the nr4a2a promoter was used to drive green fluorescent protein expression in this Tg(nr4a2a:eGFP) line. In situ hybridization showed that transgene expression follows endogenous RNA expression at a cellular level. Temporal expression and lineages were quantified using in vivo time-lapse imaging in embryos. Nr4a2 expressing retinal subtypes were characterized immunohistochemically. Nr4a2a:eGFP labeled multiple neuron subtypes including 24.5% of all amacrine interneurons. Nr4a2a:eGFP labels all tyrosine hydroxylase labeled dopaminergic amacrine cells, and other nondopaminergic GABAergic amacrine populations. Nr4a2a:eGFP is confined to a specific progenitor lineage identified by sequential expression of the bhlh transcription factor Atonal7 (Atoh7) and Pancreas transcription factor 1a (Ptf1a), and labels postmitotic postmigratory amacrine cells. Thus, developmental Nr4a2a expression indicates a role during late differentiation of specific amacrine interneurons. Tg(nr4a2a:eGFP) is an early marker of distinct neurons including dopaminergic amacrine cells. It can be utilized to assess consequences of gene manipulations and understand whether Nr4a2 only carries out its role in the presence of specific coexpressed genes. This will allow Nr4a2 use to be refined for regenerative approaches.


Subject(s)
Amacrine Cells/cytology , Amacrine Cells/metabolism , Neurogenesis/physiology , Nuclear Receptor Subfamily 4, Group A, Member 2/biosynthesis , Zebrafish Proteins/biosynthesis , Animals , Animals, Genetically Modified , Cell Differentiation/physiology , Gene Expression Profiling , Gene Expression Regulation, Developmental , Immunohistochemistry , In Situ Hybridization , Transcriptome , Zebrafish
16.
Mol Neurobiol ; 54(2): 1119-1128, 2017 03.
Article in English | MEDLINE | ID: mdl-26803497

ABSTRACT

Parkinson's disease (PD) is a progressive neurological disorder characterized by a large number of motor and non-motor features and is known as the second most common neurodegenerative disorder after Alzheimer's disease. The hallmark pathology of PD is the damage and death of dopamine-producing neurons in the substantia-nigra of midbrain. Intrastriatal transplants of fetal mesencephalon derived DAergic neurons have provided proof-of-principle for the cell replacement strategy and have demonstrated reinnervation of the denervated striatum. However, ethical, technical, and practical limitations of deploying fetal DAergic neurons as the source for cell therapy in PD have ceased the spread of this procedure into clinical practice. Embryonic stem (ES) cells have emerged as a therapeutic alternative that can proliferate extensively and generate dopamine-producing neurons. To this extent and to surmount the obstacles related to embryonic neural cells, many investigations have focused on using pluripotent stem cells for the derivation of DAergic neurons. In the present study, a mouse embryonic stem (mES) R1 cell line was generated which could stably co-express Nurr1 (an essential transcription factor in DAergic neuron development) and GPX-1 (a neuroprotective enzyme against oxidative stress). The Nurr1/GPX-1-expressing ES cells (Nurr1/GPX-1-ES) were differentiated into DAergic-like cells via a three-dimensional culture environment consisting of Poly-ε-Caprolactone (PCL) nanofibrous scaffolds embedded by Matrigel (Mtg) in the presence of specific signaling molecules. DAergic neuron-specific genes were highly expressed in ES-derived DAergic neurons cultured and differentiated on PCL/Mtg scaffolds. Reverse-phase HPLC confirmed that the Nurr1/GPX-1-ES-cells differentiated on PCL/Mtg electrospun scaffolds could efficiently and exclusively secrete dopamine in response to stimulus. In conclusion, our results demonstrated that PCL/Matrigel nanofibrous scaffolds could efficiently support and promote the generation of functional DAergic-like cells from Nurr1/GPX-1-ES cells. The results of this study may have an impact on future tissue engineering for cell therapy of PD.


Subject(s)
Collagen/administration & dosage , Dopaminergic Neurons/metabolism , Embryonic Stem Cells/metabolism , Glutathione Peroxidase/biosynthesis , Laminin/administration & dosage , Nuclear Receptor Subfamily 4, Group A, Member 2/biosynthesis , Polyesters/administration & dosage , Proteoglycans/administration & dosage , Animals , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cells, Cultured , Dopamine/metabolism , Dopaminergic Neurons/drug effects , Drug Combinations , Embryonic Stem Cells/drug effects , Glutathione Peroxidase/genetics , Humans , Mice , Mice, Knockout , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Tissue Scaffolds , Glutathione Peroxidase GPX1
17.
Mol Med Rep ; 14(5): 4309-4314, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27667480

ABSTRACT

Down syndrome (DS) is the most common birth defect in children. To investigate the mechanisms of DS, the present study analyzed the bisulfite­sequencing (seq) data GSE42144, which was downloaded from the Gene Expression Omnibus. GSE42144 included DNA methylation data of three DS samples and three control samples, and RNA­seq data of two DS samples and five control samples. The methylated sites in the bisulfite­seq data were detected using Bismark and Bowtie2. The BiSeq tool was applied to determine differentially methylated regions and to identify adjacent genes. Using the Database for Annotation, Visualization and Integrated Discovery, the functions of the abnormal demethylated genes were predicted by functional enrichment analyses. Differentially expressed genes (DEGs) were then screened using a paired t­test. Furthermore, the interactions of the proteins encoded by selected genes were determined using the Search Tool for the Retrieval of Interacting Genes, and a protein­protein interaction (PPI) network was constructed using Cytoscape. A total of 74 CpG regions showed significant differential DNA methylation between the DS and normal samples. There were five abnormal demethylated DNA regions in chromosome 21. In the DS samples, a total of 43 adjacent genes were identified with demethylation in their promoter regions and one adjacent gene was identified with upregulated methylation in its promoter regions. In addition, 584 upregulated genes were identified, including 24 genes with transcriptional regulatory function. In particular, upregulated Runt­related transcription factor 1 (RUNX1) was located on chromosome 21. Functional enrichment analysis indicated that inhibitor of DNA binding 4 (ID4) was involved in neuronal differentiation and transcriptional suppression. In the PPI network, genes may be involved in DS by interacting with others, including nuclear receptor subfamily 4 group A member 2 (NR4A2)­early growth response (EGR)2 and NR4A2­EGR3. Therefore, RUNX1, NR4A2, EGR2, EGR3 and ID4 may be key genes associated with the pathogenesis of DS.


Subject(s)
Core Binding Factor Alpha 2 Subunit/genetics , Down Syndrome/genetics , Early Growth Response Protein 2/genetics , Early Growth Response Protein 3/genetics , Inhibitor of Differentiation Proteins/genetics , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Computational Biology , Core Binding Factor Alpha 2 Subunit/biosynthesis , DNA Methylation/genetics , Databases, Genetic , Down Syndrome/pathology , Early Growth Response Protein 2/biosynthesis , Early Growth Response Protein 3/biosynthesis , Gene Expression Regulation , Gene Regulatory Networks , Humans , Inhibitor of Differentiation Proteins/biosynthesis , Nuclear Receptor Subfamily 4, Group A, Member 2/biosynthesis , Protein Interaction Maps/genetics , Sequence Analysis, RNA , Signal Transduction
18.
Med Sci Monit ; 22: 2831-8, 2016 Aug 11.
Article in English | MEDLINE | ID: mdl-27513680

ABSTRACT

BACKGROUND Simazine is a triazine herbicide used worldwide in both agricultural and non-agricultural fields that is frequently detected in surface water and groundwater. Due to its widespread use, an increasing amount of research has focused on the potentially serious environmental and health risks. MATERIAL AND METHODS We used Western blotting and real-time quantitative PCR to analyze the effects of simazine on dopamine neuronal development-related factors in MN9D dopaminergic cells. RESULTS The expression of tyrosine hydroxylase (TH) mRNA was significantly increased after treatment with 300 and 600 µmol L-1 simazine after 24 and 48 h. Levels of nuclear-related receptor 1 (Nurr1) mRNA after 24- and 48-h exposure were decreased with 50 µmol L-1 simazine, but increased with 600 µmol L-1 simazine. Significant increases in TH and Nurr1 protein were observed in all simazine-treated groups at 24 and 48 h. The expression of neurogenin 2 and LIM homeobox transcription factor 1 beta (Lmx1b) mRNA were significantly increased after exposure to 600 µmol L-1 simazine for 48 h, while the expression of wingless-type MMTV integration site family member 1 (Wnt1) mRNA was increased by all doses of simazine. CONCLUSIONS Simazine may have an impact on TH in MN9D cells through 2 mechanisms; one mechanism is through the Lmx1a/Ngn2 pathway, and the other mechanism is through the Lmx1b-pitx3/Wnt1-Nurr1 pathway. These 2 pathways likely do not operate in isolation, but rather together, during the cellular response to simazine exposure.


Subject(s)
Dopaminergic Neurons/drug effects , Neurogenesis/drug effects , Simazine/toxicity , Blotting, Western , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Line , Dopamine/metabolism , Dopaminergic Neurons/cytology , Dopaminergic Neurons/metabolism , Homeodomain Proteins/genetics , Humans , LIM-Homeodomain Proteins , Nuclear Receptor Subfamily 4, Group A, Member 2/biosynthesis , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Transcription Factors , Tyrosine 3-Monooxygenase/biosynthesis , Tyrosine 3-Monooxygenase/genetics , Wnt1 Protein
19.
BMC Cancer ; 16: 257, 2016 Mar 31.
Article in English | MEDLINE | ID: mdl-27036119

ABSTRACT

BACKGROUND: Nurr1, a member of the orphan receptor family, plays an important role in several types of cancer. Our previous work demonstrated that increased expression of Nurr1 plays a significant role in the initiation and progression of prostate cancer (PCa), though the mechanisms for regulation of Nurr1 expression remain unknown. In this study, we investigated the hypothesis that Nemo-like kinase (NLK) is a key regulator of Nurr1 expression in PCa. METHODS: Immunohistochemistry and Western blot analysis were used to evaluate levels of NLK and Nurr1 in prostatic tissues and cell lines. The effects of overexpression or knockdown of Nurr1 were evaluated in PCa cells through use of PCR, Western blots and promoter reporter assays. The role of Nurr1 promoter cis element was studied by creation of two mutant Nurr1 promoter luciferase constructs, one with a mutated NF-κB binding site and one with a mutated CREB binding site. In addition, three specific inhibitors were used to investigate the roles of these proteins in transcriptional activation of Nurr1, including BAY 11-7082 (NF-κB inhibitor), KG-501 (CREB inhibitor) and ICG-001 (CREB binding protein, CBP, inhibitor). The function of CBP in NLK-mediated regulation of Nurr1 expression was investigated using immunofluorescence, co-immunoprecipitation (Co-IP) and chromatin immunoprecipitation assays (ChIPs). RESULTS: NLK expression was inversely correlated with Nurr1 expression in prostate cancer tissues and cell lines. Overexpression of NLK suppressed Nurr1 promoter activity, leading to downregulation of Nurr1 expression. In contrast, knockdown of NLK demonstrated opposite results, leading to upregulation of Nurr1. When compared with the wild-type Nurr1 promoter, mutation of NF-κB- and CREB-binding sites of the Nurr1 promoter region significantly reduced the upregulation of Nurr1 induced by knockdown of NLK in LNCaP cells; treatment with inhibitors of CREB, CBP and NF-κB led to similar results. We also found that NLK directly interacts with CBP, that knockdown of NLK significantly increases the recruitment of CBP to both NF-κB- and CREB-binding sites, and that regulation of NLK on Nurr1 expression is abrogated by knockdown of CBP. CONCLUSIONS: Our results suggest that NLK inhibits transcriptional activation of Nurr1 gene by impeding CBP's role as a co-activator of NF-κB and CREB in prostate cancer.


Subject(s)
DNA-Binding Proteins/genetics , Intracellular Signaling Peptides and Proteins/genetics , Nuclear Receptor Subfamily 4, Group A, Member 2/biosynthesis , Prostatic Neoplasms/genetics , Protein Serine-Threonine Kinases/genetics , Binding Sites , CREB-Binding Protein/antagonists & inhibitors , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cyclic AMP Response Element-Binding Protein/antagonists & inhibitors , DNA-Binding Proteins/biosynthesis , Gene Expression Regulation, Neoplastic , Humans , Intracellular Signaling Peptides and Proteins/biosynthesis , Male , NF-kappa B/genetics , Nuclear Receptor Subfamily 4, Group A, Member 2/antagonists & inhibitors , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Promoter Regions, Genetic , Prostatic Neoplasms/pathology , Protein Serine-Threonine Kinases/biosynthesis , Transcriptional Activation/genetics
20.
Neurotox Res ; 30(1): 14-31, 2016 07.
Article in English | MEDLINE | ID: mdl-26678495

ABSTRACT

NURR1 is an essential transcription factor for the differentiation, maturation, and maintenance of midbrain dopaminergic neurons (DA neurons) as it has been demonstrated using knock-out mice. DA neurons of the substantia nigra pars compacta degenerate in Parkinson's disease (PD) and mutations in the Nurr1 gene have been associated with this human disease. Thus, the study of NURR1 actions in vivo is fundamental to understand the mechanisms of neuron generation and degeneration in the dopaminergic system. Here, we present and discuss findings indicating that NURR1 is a valuable molecular tool for the in vitro generation of DA neurons which could be used for modeling and studying PD in cell culture and in transplantation approaches. Transduction of Nurr1 alone or in combination with other transcription factors such as Foxa2, Ngn2, Ascl1, and Pitx3, induces the generation of DA neurons, which upon transplantation have the capacity to survive and restore motor behavior in animal models of PD. We show that the survival of transplanted neurons is increased when the Nurr1-transduced olfactory bulb stem cells are treated with GDNF. The use of these and other factors with the induced pluripotent stem cell (iPSC)-based technology or the direct reprogramming of astrocytes or fibroblasts into human DA neurons has produced encouraging results for the study of the cellular and molecular mechanisms of neurodegeneration in PD and for the search of new treatments for this disease.


Subject(s)
Dopaminergic Neurons/physiology , Neurogenesis/physiology , Nuclear Receptor Subfamily 4, Group A, Member 2/physiology , Animals , Cells, Cultured , Humans , Mice, Knockout , Nuclear Receptor Subfamily 4, Group A, Member 2/biosynthesis , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Olfactory Bulb/cytology , Stem Cell Transplantation/methods , Stem Cells/physiology , Transcription Factors/biosynthesis , Transcription Factors/genetics , Transcription Factors/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...