Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Exp Dermatol ; 28(9): 1036-1043, 2019 09.
Article in English | MEDLINE | ID: mdl-31287590

ABSTRACT

We analysed the correlation between the expression of HIF-1α (hypoxia-inducible factor 1 alpha), the nuclear receptors: VDR (vitamin D receptor), RORα (retinoic acid receptor-related orphan receptor alpha), and RORγ and CYP24A1 (cytochrome P450 family 24 subfamily A member 1) and CYP27B1 (cytochrome P450 family 27 subfamily B member 1), enzymes involved in vitamin D metabolism. In primary and metastatic melanomas, VDR negatively correlated with nuclear HIF-1α expression (r = -.2273, P = .0302; r = -.5081, P = .0011). Furthermore, the highest HIF-1α expression was observed in pT3-pT4 VDR-negative melanomas. A comparative analysis of immunostained HIF-1α and CYP27B1 and CYP24A1 showed lack of correlation between these parameters both in primary tumors and melanoma metastases. In contrast, RORα expression correlated positively with nuclear HIF-1α expression in primary and metastatic lesions (r = .2438, P = .0175; r = .3662, P = .0166). Comparable levels of HIF-1α expression pattern was observed in localized and advanced melanomas. RORγ in primary melanomas correlated also positively with nuclear HIF-1α expression (r = .2743, P = .0129). HIF-1α expression was the lowest in localized RORγ-negative melanomas. In addition, HIF-1α expression correlated with RORγ-positive lymphocytes in melanoma metastases. We further found that in metastatic lymph nodes FoxP3 immunostaining correlated positively with HIF-1α and RORγ expression in melanoma cells (r = .3667; P = .0327; r = .4208, P = .0129). In summary, our study indicates that the expression of VDR, RORα and RORγ in melanomas is related to hypoxia and/or HIF1-α activity, which also affects FoxP3 expression in metastatic melanoma. Therefore, the hypoxia can affect tumor biology by changing nuclear receptors expression and molecular pathways regulated by nuclear receptors and immune responses.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Melanoma/metabolism , Neoplasm Proteins/biosynthesis , Nuclear Receptor Subfamily 1, Group F, Member 1/biosynthesis , Nuclear Receptor Subfamily 1, Group F, Member 3/biosynthesis , Receptors, Calcitriol/biosynthesis , Skin Neoplasms/metabolism , 25-Hydroxyvitamin D3 1-alpha-Hydroxylase/analysis , Adult , Aged , Aged, 80 and over , Cell Hypoxia , Cell Nucleus/chemistry , Female , Gene Expression Regulation, Neoplastic , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Lymphocytes/chemistry , Lymphocytes, Tumor-Infiltrating/immunology , Male , Melanoma/genetics , Melanoma/immunology , Melanoma/secondary , Middle Aged , Neoplasm Proteins/genetics , Nuclear Receptor Subfamily 1, Group F, Member 1/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Receptors, Calcitriol/genetics , Single-Blind Method , Skin Neoplasms/genetics , Skin Neoplasms/immunology , T-Lymphocytes, Regulatory/immunology , Vitamin D3 24-Hydroxylase/analysis
2.
Arthritis Res Ther ; 20(1): 205, 2018 09 06.
Article in English | MEDLINE | ID: mdl-30189901

ABSTRACT

BACKGROUND: The orphan nuclear receptors retinoic acid-related receptor α and γt (RORα and RORγt) are critical in the development of T helper 17 (Th17) cells, and ROR-specific synthetic ligands have proven efficacy in several mouse models of autoimmunity. However, the pathological significance of RORα in primary Sjögren's syndrome (pSS) remains to be elucidated. The present study was designed to clarify the significance of RORα in the pathogenesis of pSS. METHODS: RORα expression in the labial salivary gland (LSG) was determined by immunohistochemical analysis using a quantitative scoring system in 34 patients with pSS. The correlation between RORα expression in LSGs and the focus score (FS) was determined, and Th17 and IL-17 receptor A (1L-17RA) levels in LSGs were determined. To investigate the effect of RORs and the therapeutic potential of targeting RORs in pSS, we administered SR1001, a selective RORα/γt inverse agonist, to non-obese diabetic (NOD) mice. RESULTS: The expression of RORα was significantly increased in LSGs of patients with pSS and intensified with disease stage/FS, showing a similar increasing trend with IL-17A and IL-17RA. SR1001 significantly improved salivary gland secretory function and relieved sialadenitis in treated mice. CONCLUSION: Our data reveal the importance of RORα in controlling pathologic lymphocytic infiltration of the salivary glands and suggest that RORα may be a druggable target in treating pSS.


Subject(s)
Nuclear Receptor Subfamily 1, Group F, Member 1/biosynthesis , Saliva/metabolism , Salivary Glands/metabolism , Sjogren's Syndrome/metabolism , Adult , Animals , Female , Humans , Male , Mice , Mice, Inbred NOD , Nuclear Receptor Subfamily 1, Group F, Member 1/genetics , Salivary Glands/pathology , Sjogren's Syndrome/genetics , Sjogren's Syndrome/pathology , Th17 Cells
3.
Gene ; 641: 18-24, 2018 Jan 30.
Article in English | MEDLINE | ID: mdl-29032151

ABSTRACT

Globally, there is an alarming decline in sperm count. Very often hormonal supplementation fails to restore normal sperm count. Sertoli cells (Sc) present within seminiferous tubules provide appropriate niche and factors required for the differentiation of germ cells (Gc) into mature sperm (spermatogenesis). Functionally compromised Sc may be one of the reasons for failure of hormones to facilitate normal spermatogenesis. Although role of secretory proteins and signaling molecules of Sc has been studied well, role of transcription factors regulating sperm count has not been addressed appropriately. Retinoic acid receptor-related orphan receptor (ROR)-alpha is one of such transcription factors reported in testis but its role in testicular function is not yet known. In a separate study, we found abundant ROR-alpha binding sites on promoter regions of several genes upregulated in pubertal rat Sc as compared to infant Sc. Immunostaining studies also revealed presence of ROR alpha in nucleus of pubertal Sc. We generated a transgenic knockdown rat model expressing shRNA targeted to ROR-alpha under Sc specific promoter, which is transcriptionally active only at and after puberty. ROR-alpha knockdown animals were found to have abnormal association of Sc and Gc, including Gc sloughing and restricted release of sperm. The knockdown animals displayed compromised spermatogenesis leading to significant reduction in sperm count. This is the first report describing the Sc specific role of ROR-alpha in maintaining quantitatively normal sperm output. Identification of various such molecules can generate avenues to limit or reverse an alarmingly declining sperm count witnessed globally in men.


Subject(s)
Nuclear Receptor Subfamily 1, Group F, Member 1/biosynthesis , Nuclear Receptor Subfamily 1, Group F, Member 1/genetics , Sertoli Cells/metabolism , Sperm Count , Spermatogenesis/physiology , Animals , Cells, Cultured , Infertility, Male/genetics , Litter Size/genetics , Male , RNA Interference , RNA, Small Interfering/genetics , Rats , Rats, Transgenic , Rats, Wistar , Spermatogenesis/genetics , Testis/metabolism
4.
Bull Exp Biol Med ; 160(5): 656-8, 2016 Mar.
Article in English | MEDLINE | ID: mdl-27021094

ABSTRACT

We studied the ability of melatonin in physiological and pharmacological concentrations to induce and/or regulate differentiation of T cells producing IL-17 (Th17). This hormone produced the opposite effect on CD4+T cells, which depended on their activation status. Melatonin induced the synthesis of IL-17A by intact T cells, but had little effect on activated cells. Melatonin in high (pharmacological) concentration decreased the intracellular expression of this cytokine under conditions of polyclonal activation. Melatonin had a dose-depended effect. Taking into the fact that Th17 cells play an important role in the immune defense, it can be suggested that the regulation of their activity by melatonin contributes to this process.


Subject(s)
Cell Differentiation/drug effects , Interleukin-17/biosynthesis , Melatonin/pharmacology , Th17 Cells/cytology , Adult , Humans , Lymphocyte Activation/immunology , Nuclear Receptor Subfamily 1, Group F, Member 1/biosynthesis , Nuclear Receptor Subfamily 1, Group F, Member 3/biosynthesis , Th17 Cells/immunology
5.
Epilepsy Behav ; 55: 38-46, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26731717

ABSTRACT

It is widely known that there is an increase in the inflammatory responses and oxidative stress in temporal lobe epilepsy (TLE). Further, the seizures follow a circadian rhythmicity. Retinoic acid receptor-related orphan receptor alpha (RORα) is related to anti-inflammatory and antioxidant enzyme expression and is part of the machinery of the biological clock and circadian rhythms. However, the participation of RORα in this neurological disorder has not been studied. The aim of this study was to evaluate the RORα mRNA and protein content profiles in the hippocampus of rats submitted to a pilocarpine-induced epilepsy model at different time points throughout the 24-h light-dark cycle analyzing the influence of the circadian rhythm in the expression pattern during the acute, silent, and chronic phases of the experimental model. Real-time PCR and immunohistochemistry results showed that RORα mRNA and protein expressions were globally reduced in both acute and silent phases of the pilocarpine model. However, 60days after the pilocarpine-induced status epilepticus (chronic phase), the mRNA expression was similar to the control except for the time point 3h after the lights were turned off, and no differences were found in immunohistochemistry. Our results indicate that the status epilepticus induced by pilocarpine is able to change the expression and daily variation of RORα in the rat hippocampal area during the acute and silent phases. These findings enhance our understanding of the circadian pattern present in seizures as well as facilitate strategies for the treatment of seizures.


Subject(s)
Epilepsy/chemically induced , Epilepsy/metabolism , Hippocampus/metabolism , Muscarinic Agonists , Nuclear Receptor Subfamily 1, Group F, Member 1/biosynthesis , Nuclear Receptor Subfamily 1, Group F, Member 1/genetics , Pilocarpine , Animals , Chronic Disease , Circadian Rhythm/genetics , Gene Expression Regulation/drug effects , Hippocampus/drug effects , Male , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Wistar , Status Epilepticus/chemically induced , Status Epilepticus/genetics
6.
J Immunol ; 195(7): 3001-10, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26324771

ABSTRACT

IL-23 is the key cytokine that induces the expansion of Th17 cells. It is composed of p19 and p40 subunits of IL-12. The p40 subunit binds competitively to the receptor of IL-23 and blocks its activity. Our aim was to assess the preventive and therapeutic effect of the IL-12p40 homodimer (p40)2 subunit in autoimmune arthritis animal models. In the current study, using IL-1R antagonist-knockout mice and a collagen-induced arthritis model, we investigated the suppressive effect of (p40)2 on inflammatory arthritis. We demonstrated that the recombinant adenovirus-expressing mouse (p40)2 model prevented the development of arthritis when given before the onset of arthritis. It also decreased the arthritis index and joint erosions in the mouse model if transferred after arthritis was established. (p40)2 inhibited the production of inflammatory cytokines and Ag-specific T cell proliferation. It also induced CD4(+)CD25(+)Foxp3 regulatory T (Treg) cells in vitro and in vivo, whereas the generation of retinoic acid receptor-related organ receptor γt and Th17 cells was suppressed. The induction of Treg cells and the suppression of Th17 cells were mediated via activated STAT5 and suppressed STAT3. Our data suggest that (p40)2 suppressed inflammatory arthritis successfully. This could be a useful therapeutic approach in autoimmune arthritis to regulate the Th17/Treg balance and IL-23 signaling.


Subject(s)
Arthritis, Experimental/prevention & control , Interleukin-12 Subunit p40/pharmacology , Interleukin-23 Subunit p19/immunology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Animals , Arthritis, Experimental/drug therapy , Arthritis, Experimental/immunology , Cell Proliferation/drug effects , Cells, Cultured , Collagen/immunology , Cytokines/biosynthesis , Interleukin-12 Subunit p40/immunology , Interleukin-17/biosynthesis , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred DBA , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 1/biosynthesis , Protein Multimerization , Receptors, Interleukin/immunology , Receptors, Interleukin-1 Type I/genetics , STAT3 Transcription Factor/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction/immunology
7.
Eur J Immunol ; 45(4): 1010-8, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25605286

ABSTRACT

Differentiated CD4(+) T cells preserve plasticity under various conditions. However, the stability of Th1 cells is unclear, as is whether Th1 cells can convert into Th17 cells and thereby contribute to the generation of IFN-γ(+) IL-17(+) CD4(+) T cells, the number of which correlates with severity of colitis. We investigated whether IFN-γ(+) Th1 cells can convert into Th17 cells under intestinal inflammation and the mechanisms involved. IFN-γ(Thy1.1+) Th1 cells were generated by culturing naïve CD4(+) T cells from IFN-γ(Thy1.1) CBir1 TCR-Tg reporter mice, whose TCR is specific for an immunodominant microbiota antigen, CBir1 flagellin, under Th1 polarizing conditions. IFN-γ(Thy1.1+) Th1 cells induced colitis in Rag(-/-) mice after adoptive transfer and converted into IL-17(+) Th17, but not Foxp3(+) Treg cells in the inflamed intestines. TGF-ß and IL-6, but not IL-1ß and IL-23, regulated Th1 conversion into Th17 cells. TGF-ß induction of transcriptional factor Runx1 is crucial for the conversion, since silencing Runx1 by siRNA inhibited Th1 conversion into Th17 cells. Furthermore, TGF-ß enhanced histone H3K9 acetylation but inhibited H3K9 trimethylation of Runx1- and ROR-γt-binding sites on il-17 or rorc gene in Th1 cells. We conclude that Th1 cells convert into Th17 cells under inflammatory conditions in intestines, which is possibly mediated by TGF-ß induction of Runx1.


Subject(s)
Core Binding Factor Alpha 2 Subunit/biosynthesis , Intestinal Mucosa/immunology , Th1 Cells/cytology , Th17 Cells/cytology , Transforming Growth Factor beta/metabolism , Acetylation , Animals , Binding Sites , Cell Differentiation/immunology , Cells, Cultured , Colitis/immunology , Core Binding Factor Alpha 2 Subunit/genetics , Flagellin/immunology , Histones/metabolism , Homeodomain Proteins/genetics , Interferon-gamma/biosynthesis , Interleukin-17/biosynthesis , Interleukin-1beta/metabolism , Interleukin-2/metabolism , Interleukin-23/metabolism , Interleukin-6/metabolism , Lymphocyte Activation/immunology , Lymphocyte Count , Methylation , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 1/biosynthesis , Nuclear Receptor Subfamily 1, Group F, Member 3/biosynthesis , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , RNA Interference , RNA, Small Interfering , Th1 Cells/immunology , Th17 Cells/immunology
8.
Am J Physiol Cell Physiol ; 308(5): C385-96, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25500738

ABSTRACT

Lipid metabolic disturbances are related to many diseases, such as obesity, diabetes, and certain cancers. Notably, lipid metabolic disturbances have been reported to be a risk factor for colorectal cancer. Nuclear receptors act as ligand-dependent transcription regulators and play key roles in the regulation of body lipid metabolism and the development of many cancers. Retinoic acid receptor-related orphan receptor α (RORα) is a nuclear receptor and can regulate several lipid metabolism genes in certain cancers. Herein, we demonstrate that the conditioned medium from adipocytes has a proproliferative and promigratory effect on colorectal cancer cells and enhances angiogenesis in chicken embryonic chorioallantoic membranes. In addition, the conditioned medium leads to a decrease in the expression of RORα and its target genes. Meanwhile, RORα and its target gene expressions are lower in human colorectal tumor tissue compared with control colorectal tissue. Activation of RORα inhibits the effect of conditioned medium on the proliferation and migration of colorectal cancer cells as well as the angiogenesis in chicken embryonic allantoic membranes. In colorectal cancer cells, the putative ligand of RORα, cholesterol sulfate (CS), prevents cell cycle progression at the G1/S boundary and concurrently modulates the expression of cell cycle-regulatory genes in colorectal cancer cell. CS inhibits angiogenesis in chicken embryonic chorioallantoic membranes and concurrently decreases the mRNA expression of vascular endothelial growth factor (VEGF) and hypoxia-inducible factor-1α as well as the secretion of VEGF. In addition, lipogenic gene expression is higher in human colorectal tumor tissue compared with control colorectal tissue. CS inhibits the expression of lipogenic genes in colorectal cancer cells. These results suggest that RORα could represent a direct link between local lipid metabolism of colorectal tissue and colorectal cancer. Therefore, the reduction of the expression of RORα could represent a potential warning sign of colorectal cancer.


Subject(s)
Cell Movement/physiology , Cell Proliferation/physiology , Chorioallantoic Membrane/physiology , Colorectal Neoplasms/metabolism , Culture Media, Conditioned/pharmacology , Nuclear Receptor Subfamily 1, Group F, Member 1/biosynthesis , 3T3 Cells , Adipocytes/physiology , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/physiology , Chick Embryo , Chorioallantoic Membrane/blood supply , Chorioallantoic Membrane/drug effects , Colorectal Neoplasms/pathology , Humans , Mice , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/physiology , Nuclear Receptor Subfamily 1, Group F, Member 1/antagonists & inhibitors
9.
Biochem Biophys Res Commun ; 428(1): 99-104, 2012 Nov 09.
Article in English | MEDLINE | ID: mdl-23063684

ABSTRACT

Cholesterol sulfate is abundant in the human epidermis and is a putative natural ligand for retinoic acid receptor-related orphan receptor alpha (RORα). Although direct binding of cholesterol sulfate is expected to activate RORα, cholesterol sulfate can also induce RORα expression and increase RORα target gene expression. The purpose of this study was to determine whether cholesterol sulfate induces profilaggrin expression, a precursor of the barrier protein filaggrin in the epidermis, through activation of RORα by directly binding to RORα, or through increased RORα expression. Immunohistochemical and polymerase chain reaction (PCR) analyses showed that RORα was expressed in normal human epidermal keratinocytes (NHEKs) and that its expression increased during keratinocyte differentiation in parallel with that of profilaggrin and cholesterol sulfotransferase, which catalyzes the synthesis of cholesterol sulfate. Exogenous cholesterol sulfate significantly increased both RORα and profilaggrin expression in NHEKs, whereas no effect on profilaggrin expression was observed in cells in which RORα was knocked down with small interfering RNA (siRNA). Additionally, a luciferase reporter gene assay revealed that exogenous RORα dose-dependently increased the activity of the profilaggrin gene promoter even in the absence of cholesterol sulfate, and that this response involves activator protein-1. In conclusion, the results of this study indicate that cholesterol sulfate induces filaggrin expression through increased RORα expression. Further studies are required to fully elucidate the mechanisms involved.


Subject(s)
Cholesterol Esters/metabolism , Epidermis/metabolism , Intermediate Filament Proteins/biosynthesis , Nuclear Receptor Subfamily 1, Group F, Member 1/biosynthesis , Cells, Cultured , Cholesterol Esters/pharmacology , Epidermis/drug effects , Filaggrin Proteins , Gene Knockdown Techniques , Genes, Reporter , Humans , Intermediate Filament Proteins/genetics , Keratinocytes/drug effects , Keratinocytes/metabolism , Luciferases/biosynthesis , Luciferases/genetics , Nuclear Receptor Subfamily 1, Group F, Member 1/genetics , Promoter Regions, Genetic , RNA, Small Interfering/genetics , Sulfotransferases/metabolism
10.
J Viral Hepat ; 19(11): 811-22, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23043388

ABSTRACT

T helper cells17 (Th17) have accurate but inconclusive roles in the pathogenesis of acute-on-chronic hepatitis B liver failure (ACHBLF). Retinoic acid-related orphan receptor γ t(RORγt) and RORα are two lineage-specific nuclear receptors directly mediating Th17 differentiation. This study was aimed to evaluate the gene expression of RORα and RORγt and their potential role in ACHBLF. Forty patients with liver failure, 30 with chronic hepatitis B (CHB) and 20 healthy controls were studied. The mRNA levels of RORα and RORγt in peripheral mononuclear cells were determined by quantitative real-time polymerase chain reaction. The frequency of peripheral Th17 cells was determined using flow cytometry. The serum levels of interleukin-6(IL-6), transforming growth factor -ß (TGF-ß), interleukin-17(IL-17), interleukin-23(IL-23) and interferon-γ (IFN-γ) were measured by enzyme-linked immunosorbent assay. The frequency of peripheral Th17 cells in patients with liver failure was significantly increased compared to patients with CHB and controls. The peripheral mRNA levels of RORα and RORγt in hepatitis B-associated acute-on-chronic liver failure were significantly higher than in patients with CHB and controls as were the serum levels of IL-6 and TGF-ß. The serum level of IFN-γ in patients with acute-on-chronic liver failure from HBV was significantly higher than patients with CHB but lower than controls. In patients with acute-on-chronic liver failure associated with HBV, RORγt, IL-6 and IL-23 were positively correlated with the frequency of Th17 cells, while RORα, TGF-ß and IFN-γ had no correlation with the latter. The mRNA level of RORγt was positively correlated with model of end-stage liver disease (MELD) score, but there was no correlation of RORα and MELD score. RORγt plays an important role in the pathogenesis of acute-on-chronic HBV-associated liver failure and might be considered to be a candidate factor consistent with the severity of disease.


Subject(s)
Hepatitis B, Chronic/metabolism , Liver Failure, Acute/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 1/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Adult , Female , Hepatitis B, Chronic/genetics , Hepatitis B, Chronic/immunology , Humans , Interferon-gamma/blood , Interleukin-17/blood , Interleukin-23/blood , Interleukin-6/blood , Liver Failure, Acute/genetics , Liver Failure, Acute/immunology , Lymphocyte Count , Male , Nuclear Receptor Subfamily 1, Group F, Member 1/biosynthesis , Nuclear Receptor Subfamily 1, Group F, Member 3/biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Severity of Illness Index , Th17 Cells/immunology , Transforming Growth Factor beta/blood
11.
Cerebellum ; 11(2): 451-2, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22223133

ABSTRACT

Studies of staggerer mice, in which retinoid-related orphan receptor-alpha (RORα) is mutated, have provided new insights into the critical functions of RORα in various physiological processes in peripheral tissues and in the brain. Staggerer mice present an ataxic phenotype caused by a massive neurodegeneration in the cerebellum. As a result, most of studies have focused on the role of RORα in the development of the cerebellum. Recent studies have expanded the role of RORα to other structures and functions in the brain. RORα was considered to be exclusively expressed in neurons in the brain. Recently, it has been shown that, in addition to its neuronal expression, RORα is expressed in glial cells and particularly in astrocytes in different brain regions. Moreover, RORα has been implicated in the regulation of some astrocyte functions such as the inflammatory function. Several reports have also presented evidence for a role of RORα in diverse pathological processes including oxidative stress-induced apoptosis and cerebral hypoxia. This review therefore focuses on the emerging roles of RORα in the brain and particularly in astrocytes.


Subject(s)
Brain/growth & development , Brain/physiology , Nuclear Receptor Subfamily 1, Group F, Member 1/genetics , Nuclear Receptor Subfamily 1, Group F, Member 1/physiology , Animals , Astrocytes/metabolism , Humans , Mice , Mice, Neurologic Mutants , Neurons/physiology , Nuclear Receptor Subfamily 1, Group F, Member 1/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...