Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 183
Filter
1.
Neurogastroenterol Motil ; 33(6): e14073, 2021 06.
Article in English | MEDLINE | ID: mdl-33382180

ABSTRACT

BACKGROUND: Calcitonin gene-related peptide (CGRP) is possibly involved in recruitment of mucosal mast cells (MCs) in the gut that may be associated with the development of irritable bowel syndrome (IBS), but the role of CGRP on the activation of MCs is still unknown. METHODS: Using RNA sequencing (RNA-seq), we examined differentially expressed genes (DEGs) in mouse MCs following CGRP treatment. The expression of key genes in colonic MCs and their relationship with CGRP-containing fibers were examined by immunofluorescence in chronic water-avoidance stress (WAS)-induced visceral hyperalgesia mice. KEY RESULTS: A total of 29 DEGs were found significantly changed with 28 upregulated and 1 downregulated following treatment of MCs with CGRP. Bioinformatics analysis showed that key higher DEGs included those associated with response to corticotropin-releasing hormone (CRH), regulation of transcription, MC activation, and proliferation. These processes are enriched for genes associated with stress-induced MC activation in IBS. Western blot verified changes in representative DEGs (Nr4a3, Crem, Gpr35, FosB, Sphlk1) and real-time cell analysis (RTCA) verified the MC proliferation. The vast majority of colonic MCs nearly CGRP-containing fibers in WAS mice overexpressed only Nr4a3 with little to no FosB, Gpr35, Sphlk1, or Crem expression. Nr4a3 knockdown may attenuate the promotion effect of CGRP on MC viability. CONCLUSIONS & INFERENCES: Our results suggest that CGRP is a critical regulator of key expressed genes in MC activation. Nr4a3 as a novel regulator of MC function may have an effect on stress-induced visceral hyperalgesia, and this may represent the novel target for drug development.


Subject(s)
Calcitonin Gene-Related Peptide/biosynthesis , Colon/pathology , Gene Expression Regulation , Hyperalgesia/pathology , Mast Cells/pathology , Visceral Pain/pathology , Animals , Calcitonin Gene-Related Peptide/genetics , Cell Proliferation , Computational Biology , Corticotropin-Releasing Hormone/metabolism , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Female , Intestinal Mucosa/cytology , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Receptors, Steroid/biosynthesis , Receptors, Steroid/genetics , Receptors, Thyroid Hormone/biosynthesis , Receptors, Thyroid Hormone/genetics , Stress, Psychological
2.
Toxicol Appl Pharmacol ; 394: 114960, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32201330

ABSTRACT

During pregnancy, fetal thyroid hormones (THs) are dependent on maternal placental transport and their physiological level is crucial for normal fetal neurodevelopment. Earlier research has shown that Di-(2-ethylhexyl) phthalate (DEHP) disrupts thyroid function and THs homeostasis in pregnant women and fetuses, and affects placental THs transport. However, the underlying mechanisms are poorly understood. The present study, therefore, aimed to systematically investigate the potential mechanisms of DEHP-induced disruption in the placental THs transport using two human placental trophoblastic cells, HTR-8/SVneo cells and JEG-3 cells. While the exposure of DEHP at the doses of 0-400 µM for 24 h did not affect cell viability, we found reduced consumption of T3 and T4 in the culture medium of HTR-8/Svneo cells treated with DEHP at 400 µM. DEHP treatment did not affect T3 uptake and the expression of monocarboxylate transporters 8 (MCT8) and organic anion transporters 1C1 (OATP1C1). However, DEHP significantly inhibited transthyretin (TTR) internalization, down-regulated TTR, deiodinase 2 (DIO2), and thyroid hormone receptors mRNA expression and protein levels, and up-regulated deiodinase 3 (DIO3) protein levels in a dose-dependent manner. These results indicate that DEHP acts on placental trophoblast cells, inhibits its TTR internalization, down-regulates TTR expression and affects the expression of DIO2, DIO3, and thyroid hormone receptor. These may be the mechanisms by which PAEs affects THs transport through placental.


Subject(s)
Diethylhexyl Phthalate/toxicity , Placenta/metabolism , Prealbumin/metabolism , Trophoblasts/metabolism , Adult , Cell Line, Tumor , Dose-Response Relationship, Drug , Female , Humans , Iodide Peroxidase/antagonists & inhibitors , Monocarboxylic Acid Transporters/antagonists & inhibitors , Organic Anion Transporters/antagonists & inhibitors , Placenta/cytology , Placenta/drug effects , Prealbumin/biosynthesis , Pregnancy , Receptors, Thyroid Hormone/biosynthesis , Receptors, Thyroid Hormone/drug effects , Symporters/antagonists & inhibitors , Thyroid Hormones/metabolism , Thyroxine/metabolism , Triiodothyronine/biosynthesis , Trophoblasts/drug effects , Iodothyronine Deiodinase Type II
3.
Med Oncol ; 36(8): 66, 2019 Jun 10.
Article in English | MEDLINE | ID: mdl-31183633

ABSTRACT

Nuclear receptor subfamily 4, group A, member 3 (NR4A3) is a member of the NR4A subgroup of orphan nuclear receptors, implicated in the regulation of diverse biological functions, including metabolism, angiogenesis, inflammation, cell proliferation, and apoptosis. Although many reports have suggested the involvement of NR4A3 in the development and/or progression of tumors, its role varies among tumor types. Previously, we reported that DNA hypomethylation at NR4A3 exon 3 is associated with lower survival rate of neuroblastoma (NB) patients. As hypomethylation of this region results in reduced expression of NR4A3, our observations suggested that NR4A3 functions as a tumor suppressor in NB. However, the exact mechanisms underlying its functions have not been clarified. In the present study, we analyzed public databases and showed that reduced NR4A3 expression was associated with shorter survival period of NB in two out of three datasets. An in vitro study revealed that forced expression of NR4A3 in human NB-derived cell line NB1 resulted in elongation of neurites along with overexpression of GAP43, one of the differentiation markers of NB. On the other hand, siRNA-mediated knockdown of NR4A3 suppressed the expression level of GAP43. Interestingly, the forced expression of NR4A3 induced only the GAP43 but not the other molecules involved in NB cell differentiation, such as MYCN, TRKA, and PHOX2B. These results indicated that NR4A3 directly activates the expression of GAP43 and induces differentiated phenotypes of NB cells, without affecting the upstream signals regulating GAP43 expression and NB differentiation.


Subject(s)
DNA-Binding Proteins/biosynthesis , Neuroblastoma/metabolism , Receptors, Steroid/biosynthesis , Receptors, Thyroid Hormone/biosynthesis , Cell Differentiation/physiology , Cell Line, Tumor , DNA-Binding Proteins/genetics , Disease Progression , GAP-43 Protein/biosynthesis , Gene Knockdown Techniques , Humans , Neurites/metabolism , Neurites/pathology , Neuroblastoma/genetics , Neuroblastoma/pathology , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Receptors, Steroid/genetics , Receptors, Thyroid Hormone/genetics , Up-Regulation
4.
Am J Surg Pathol ; 43(9): 1264-1272, 2019 09.
Article in English | MEDLINE | ID: mdl-31094928

ABSTRACT

Recently, we discovered the recurrent genomic rearrangement [t(4;9)(q13;q31)] enabling upregulation of the transcription factor Nuclear Receptor Subfamily 4 Group A Member 3 (NR4A3) through enhancer hijacking as the oncogenic driver event in acinic cell carcinoma (AciCC) of the salivary glands. In the current study, we evaluated the usefulness of NR4A3 immunostaining and NR4A3 fluorescence in situ hybridization (FISH) in the differential diagnosis of AciCC, comparing a total of 64 AciCCs including 17% cases with high-grade transformation, 29 secretory (mammary analog) carcinomas (MASC), and 70 other salivary gland carcinomas. Nuclear NR4A3 immunostaining was a highly specific (100%) and sensitive (98%) marker for AciCC with only 1 negative case, whereas NR4A3 FISH was less sensitive (84%). None of the MASCs or other salivary gland carcinomas displayed any nuclear NR4A3 immunostaining. The recently described HTN3-MSANTD3 gene fusion was observed in 4 of 49 (8%) evaluable AciCCs, all with nuclear NR4A3 immunostaining. In summary, NR4A3 immunostaining is a highly specific and sensitive marker for AciCC, which may be especially valuable in cases with high-grade transformation and in "zymogen granule"-poor examples within the differential diagnostic spectrum of AciCC and MASC.


Subject(s)
Biomarkers, Tumor/analysis , Carcinoma, Acinar Cell/diagnosis , DNA-Binding Proteins/biosynthesis , Receptors, Steroid/biosynthesis , Receptors, Thyroid Hormone/biosynthesis , Salivary Gland Neoplasms/diagnosis , Adult , Aged , Aged, 80 and over , DNA-Binding Proteins/analysis , Diagnosis, Differential , Female , Humans , Immunohistochemistry , Male , Middle Aged , Receptors, Steroid/analysis , Receptors, Thyroid Hormone/analysis , Sensitivity and Specificity , Young Adult
5.
J Biol Chem ; 294(13): 4889-4897, 2019 03 29.
Article in English | MEDLINE | ID: mdl-30696767

ABSTRACT

The Nr4a subfamily of nuclear receptor comprises three members in mammalian cells: Nur77/Nr4a1, Nurr1/Nr4a2, and Nor1/Nr4a3. Nr4a proteins play key roles in the regulation of glucose homeostasis in peripheral metabolic tissues. However, their biological functions in ß-cells remain relatively uncharacterized. Here we sought to investigate the potential role of Nor1 in the regulation of ß-cell mass and, in particular, ß-cell survival/apoptosis. We used histological analysis to examine the consequences of genetic deletion of either Nur77 and Nor1 on ß-cell mass, investigated the expression patterns of Nr4as in human islets and INS cells and performed gain- and loss-of-function experiments to further characterize the role of Nor1 in ß-cell apoptosis. Surprisingly, Nor1 knockout mice displayed increased ß-cell mass, whereas mice with genetic deletion of Nur77 did not exhibit any significant differences compared with their WT littermates. The increase in ß-cell mass in Nor1 knockout mice was accompanied by improved glucose tolerance. A gene expression study performed in both human islets and INS cells revealed that Nor1 expression is significantly increased by pro-inflammatory cytokines and, to a lesser extent, by elevated concentrations of glucose. Nor1 overexpression in both INS and human islet cells caused apoptosis, whereas siRNA-mediated Nor1 knockdown prevented cytokine-induced ß-cell death. Finally, Nor1 expression was up-regulated in islets of individuals with type 2 diabetes. Altogether, our results uncover that Nor1 negatively regulates ß-cell mass. Nor1 represents a promising molecular target in diabetes treatment to prevent ß-cell destruction.


Subject(s)
Apoptosis , DNA-Binding Proteins/biosynthesis , Diabetes Mellitus, Type 2/metabolism , Insulin-Secreting Cells/metabolism , Nerve Tissue Proteins/biosynthesis , Receptors, Steroid/biosynthesis , Receptors, Thyroid Hormone/biosynthesis , Up-Regulation , Animals , Cytokines , DNA-Binding Proteins/genetics , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/pathology , Humans , Insulin-Secreting Cells/pathology , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Receptors, Steroid/genetics , Receptors, Thyroid Hormone/genetics
6.
Mol Cell Biochem ; 448(1-2): 251-263, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29446045

ABSTRACT

Dietary administration of orotic acid (OA), an intermediate in the pyrimidine biosynthetic pathway, is considered to provide a wide range of beneficial effects, including cardioprotection and exercise adaptation. Its mechanisms of action, when applied extracellularly, however, are barely understood. In this study, we evaluated potential effects of OA on skeletal muscle using an in vitro contraction model of electrically pulse-stimulated (EPS) C2C12 myotubes. By analyzing a subset of genes representing inflammatory, metabolic, and structural adaptation pathways, we could show that OA supplementation diminishes the EPS-provoked expression of inflammatory transcripts (interleukin 6, Il6; chemokine (C-X-C Motif) ligand 5, Cxcl5), and attenuated transcript levels of nuclear receptor subfamily 4 group A member 3 (Nr4A3), early growth response 1 (Egr1), activating transcription factor 3 (Atf3), and fast-oxidative MyHC-IIA isoform (Myh2). By contrast, OA had no suppressive effect on the pathogen-provoked inflammatory gene response in skeletal muscle cells, as demonstrated by stimulation of C2C12 myotubes with bacterial LPS. In addition, we observed a suppressive effect of OA on EPS-induced phosphorylation of AMP-activated protein kinase (AMPK), whereas EPS-triggered phosphorylation/activation of the mammalian target of rapamycin (mTOR) was not affected. Finally, we demonstrate that OA positively influences glycogen levels in EP-stimulated myotubes. Taken together, our results suggest that in skeletal muscle cells, OA modulates both the inflammatory and the metabolic reaction provoked by acute contraction. These results might have important clinical implications, specifically in cardiovascular and exercise medicine.


Subject(s)
Muscle Contraction/drug effects , Myoblasts, Skeletal/metabolism , Orotic Acid/pharmacology , Activating Transcription Factor 3/biosynthesis , Animals , Chemokine CXCL5/biosynthesis , DNA-Binding Proteins/biosynthesis , Early Growth Response Protein 1/biosynthesis , Electric Stimulation , Gene Expression Regulation/drug effects , Interleukin-6/biosynthesis , Mice , Myoblasts, Skeletal/cytology , Nerve Tissue Proteins/biosynthesis , Receptors, Steroid/biosynthesis , Receptors, Thyroid Hormone/biosynthesis , TOR Serine-Threonine Kinases/biosynthesis
7.
J Cell Biochem ; 119(8): 6514-6526, 2018 08.
Article in English | MEDLINE | ID: mdl-29363789

ABSTRACT

The somatic component of seminiferous epithelium, the Sertoli cells (Sc) respond to Follicle stimulating hormone (FSH), and Testosterone (T) to produce factors which are necessary for germ cell (Gc) survival and differentiation. Infant Sc do not support spermatogenesis in spite of sufficient hormonal milieu, a situation similar to that found in male idiopathic infertility. Sc maturation during pubertal period involves expression of some genes which may be important for initiation of spermatogenesis. Analysis of differentially expressed genes, one by one, in infant and pubertal Sc might provide useful information about the regulation of spermatogenesis. DNA microarray based analysis of mRNA from 5-days (infant) and 12-days (pubertal) old rat Sc revealed increased expression of Nor-1 by pubertal Sc. NOR-1 is an orphan nuclear receptor involved in maintaining cellular homeostasis and disease. We generated transgenic mice using shRNA cloned under Pem (Rhox5) promoter which is activated at puberty in Sc. Such transgenic mice had reduced Nor-1 expression and increased Tgfß1, Tgfß3, and Smad3 expression. Moreover, an increase in ß-catenin expression was observed in NOR-1 knockdown testes. High ß-catenin in such transgenic mice was found to be associated with disruption of Sc maturation characterized by elevated expression of Anti Mullerian hormone, Cytokeratin 18, and Sox9. This disruption of Sc maturation resulted in Gc apoptosis. Such NOR-1 knockdown mice showed reduced sperm count and litter size. We report for the first time that NOR-1 plays a crucial role in regulating sperm count and male fertility.


Subject(s)
DNA-Binding Proteins/biosynthesis , Fertility , Nerve Tissue Proteins/biosynthesis , Receptors, Steroid/biosynthesis , Receptors, Thyroid Hormone/biosynthesis , Sertoli Cells/metabolism , Spermatogenesis , Spermatozoa/metabolism , Animals , DNA-Binding Proteins/genetics , Male , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Rats , Receptors, Steroid/genetics , Receptors, Thyroid Hormone/genetics , Sertoli Cells/cytology , Sperm Count , Spermatozoa/cytology
8.
FASEB J ; 31(11): 4821-4831, 2017 11.
Article in English | MEDLINE | ID: mdl-28739643

ABSTRACT

Histone modifications are associated with transcriptional regulation by diverse transcription factors. Genome-wide correlation studies have revealed that histone activation marks and repression marks are associated with activated and repressed gene expression, respectively. Among the histone activation marks is histone H3 K79 methylation, which is carried out by only a single methyltransferase, disruptor of telomeric silencing-1-like (DOT1L). We have been studying thyroid hormone (T3)-dependent amphibian metamorphosis in two highly related species, the pseudo-tetraploid Xenopus laevis and diploid Xenopus tropicalis, as a model for postembryonic development, a period around birth in mammals that is difficult to study. We previously showed that H3K79 methylation levels are induced at T3 target genes during natural and T3-induced metamorphosis and that Dot1L is itself a T3 target gene. These suggest that T3 induces Dot1L expression, and Dot1L in turn functions as a T3 receptor (TR) coactivator to promote vertebrate development. We show here that in cotransfection studies or in the reconstituted frog oocyte in vivo transcription system, overexpression of Dot1L enhances gene activation by TR in the presence of T3. Furthermore, making use of the ability to carry out transgenesis in X. laevis and gene knockdown in X. tropicalis, we demonstrate that endogenous Dot1L is critical for T3-induced activation of endogenous TR target genes while transgenic Dot1L enhances endogenous TR function in premetamorphic tadpoles in the presence of T3. Our studies thus for the first time provide complementary gain- and loss-of functional evidence in vivo for a cofactor, Dot1L, in gene activation by TR during vertebrate development.-Wen, L., Fu, L., Shi, Y.-B. Histone methyltransferase Dot1L is a coactivator for thyroid hormone receptor during Xenopus development.


Subject(s)
Gene Expression Regulation, Developmental/physiology , Gene Expression Regulation, Enzymologic/physiology , Metamorphosis, Biological/physiology , Methyltransferases/biosynthesis , Receptors, Thyroid Hormone/biosynthesis , Xenopus Proteins/biosynthesis , Animals , Histones/genetics , Histones/metabolism , Methyltransferases/genetics , Receptors, Thyroid Hormone/genetics , Xenopus Proteins/genetics , Xenopus laevis
9.
Cancer Res ; 77(9): 2375-2386, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28249906

ABSTRACT

Nuclear orphan receptor NR4A1 exerts an essential tumor suppressor function in aggressive lymphomas. In this study, we investigated the hypothesized contribution of the related NR4A family member NR4A3 to lymphomagenesis. In aggressive lymphoma patients, low expression of NR4A3 was associated with poor survival. Ectopic expression or pharmacological activation of NR4A3 in lymphoma cell lines led to a significantly higher proportion of apoptotic cells. In a mouse NSG xenograft model of lymphoma (stably transduced SuDHL4 cells), NR4A3 expression abrogated tumor growth, compared with vector control and uninduced cells that formed massive tumors. Transcript analysis of four different aggressive lymphoma cell lines overexpressing either NR4A3 or NR4A1 revealed that apoptosis was driven similarly by induction of BAK, Puma, BIK, BIM, BID, and Trail. Overall, our results showed that NR4A3 possesses robust tumor suppressor functions of similar impact to NR4A1 in aggressive lymphomas. Cancer Res; 77(9); 2375-86. ©2017 AACR.


Subject(s)
Carcinogenesis/genetics , Cell Proliferation/genetics , DNA-Binding Proteins/genetics , Lymphoma/genetics , Receptors, Steroid/genetics , Receptors, Thyroid Hormone/genetics , Animals , Apoptosis/genetics , Cell Line, Tumor , DNA-Binding Proteins/biosynthesis , Disease-Free Survival , Female , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Lymphoma/pathology , Male , Mice , Receptors, Steroid/biosynthesis , Receptors, Thyroid Hormone/biosynthesis , Xenograft Model Antitumor Assays
10.
Neurourol Urodyn ; 35(8): 895-901, 2016 11.
Article in English | MEDLINE | ID: mdl-26250619

ABSTRACT

AIM: To evaluate the morphometry and thyroid-hormone receptor (TR) expression in pelvic (pubococcygeus, Pcm) and perineal (bulbospongiosus, Bsm) muscles of control and hypothyroid female rabbits. METHODS: Hypothyroidism was induced administering 0.02% methimazole in the drinking water for one month. Hematoxylin-eosin stained muscle sections were used to evaluate the fiber cross-sectional area (CSA) and the number of peripheral myonuclei per fiber. Immunohistochemistry was used to calculate the proportion of TR immunoreactive nuclei per fiber. Significant differences were considered at a P ≤ 0.05. RESULTS: As compared to control rabbits, hypothyroidism increased the averaged fiber CSA and the myonuclei per fiber in the Bsm. Although the myonuclei number per fiber was also increased in the Pcm, the effect concerning the fiber CSA was only observed in a fraction of the Pcm fibers. Both TRα and TRß were similarly expressed in the Pcm and Bsm. Hypothyroidism increased the expression of the TRα in the Bsm. Meanwhile, the expression of TR isoforms in the Pcm was not altered. CONCLUSION: Our findings support that the TR signaling is directly involved in morphometrical changes induced by hypothyroidism in the Pcm and Bsm. The effect of hypothyroidism on the Pcm and Bsm could be related to the different type of fiber and metabolism that these muscles have. Neurourol. Urodynam. 35:895-901, 2016. © 2015 Wiley Periodicals, Inc.


Subject(s)
Hypothyroidism/metabolism , Hypothyroidism/pathology , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Receptors, Thyroid Hormone/biosynthesis , Urethra/metabolism , Urethra/pathology , Anatomy, Cross-Sectional , Animals , Antithyroid Agents , Chinchilla , Female , Hypothyroidism/chemically induced , Immunohistochemistry , Methimazole , Muscle Fibers, Skeletal/pathology , Pelvic Floor/pathology , Rabbits , Thyroid Hormone Receptors alpha/metabolism , Thyroid Hormone Receptors beta/metabolism
11.
J Hazard Mater ; 303: 76-82, 2016 Feb 13.
Article in English | MEDLINE | ID: mdl-26513566

ABSTRACT

Arsenic (As) contamination in aquatic environment adversely impacts aquatic organisms. The present study assessed the toxicity of different As species and concentrations on bighead carp (Hypophthalmichthys nobilis) at early life stage, a major fish in Yangtze River, China. We measured the changes in embryo and larvae survival rate, larvae aberration, concentrations of thyroid hormone thyroxine, and transcription levels of thyroid hormone receptors (TRs) in fish larvae after exposing to arsenite (AsIII) or arsenate (AsV) at 0, 10, 30, 50, 100, or 150 µg L(-1) for 78 h. As concentrations ≤ 150 µg L(-1) had limited effect on embryo survival rate (6-8% inhibition), but larvae survival rate decreased to 53-57% and larvae aberration rate increased to 20-24% after As exposure. Moreover, thyroxine levels elevated by 23% and 50% at 100 µg L(-1) AsIII and 150 µg L(-1) AsV. Besides, AsIII and AsV decreased the transcriptional levels of TRα by 72 and 53%, and TRß by 91 and 81% at 150 µg L(-1) As. Our data showed that AsIII and AsV had limited effect on carp embryo survival, but they were both toxic to carp larvae, with AsIII showing more effect than AsV. As concentrations <150µg L(-1) adversely influenced the development of bighead carp larvae and disturbed their thyroid hormone homeostasis.


Subject(s)
Arsenic/toxicity , Carps/metabolism , Growth/drug effects , Receptors, Thyroid Hormone/biosynthesis , Thyroid Hormones/biosynthesis , Transcription, Genetic/drug effects , Animals , Carps/growth & development , Embryo, Nonmammalian , Larva/growth & development , Larva/metabolism
12.
Cell Tissue Res ; 364(2): 299-308, 2016 05.
Article in English | MEDLINE | ID: mdl-26662056

ABSTRACT

In the developing limb, differentiation of skeletal progenitors towards distinct connective tissues of the digits is correlated with the establishment of well-defined domains of Btg1 gene expression. Zones of high expression of Btg1 include the earliest digit blastemas, the condensing mesoderm at the tip of the growing digits, the peritendinous mesenchyme, and the chondrocytes around the developing interphalangeal joints. Gain- and loss-of function experiments in micromass cultures of skeletal progenitors reveal a negative influence of Btg1 in cartilage differentiation accompanied by up-regulation of Ccn1, Scleraxis and PTHrP. Previous studies have assigned a role to these factors in the aggregation of progenitors in the digit tips (Ccn1), in the differentiation of tendon blastemas (Scleraxis) and repressing hypertrophic cartilage differentiation (PTHrP). Overexpression of Btg1 up-regulates the expression of retinoic acid and thyroid hormone receptors, but, different from other systems, the influence of BTG1 in connective tissue differentiation appears to be independent of retinoic acid and thyroid hormone signaling.


Subject(s)
Cartilage/cytology , Chondrogenesis/physiology , Extremities/embryology , Mesoderm/metabolism , Neoplasm Proteins/metabolism , Toes/embryology , Animals , Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Cell Differentiation/physiology , Cell Proliferation/physiology , Cells, Cultured , Chick Embryo , Chondrocytes/cytology , Chondrocytes/metabolism , Cysteine-Rich Protein 61/biosynthesis , Gene Expression Regulation, Developmental , Mice , Mice, Inbred C57BL , Neoplasm Proteins/biosynthesis , Parathyroid Hormone-Related Protein/biosynthesis , Receptors, Thyroid Hormone/biosynthesis , Signal Transduction/physiology , Tretinoin/metabolism
13.
Neuropathology ; 35(6): 503-9, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26096603

ABSTRACT

Nuclear receptor subfamily 4, group A, member 3 (NR4A3), also known as neuron-derived orphan receptor-1, is a nuclear receptor which plays key roles in cell cycle, neuronal differentiation, apoptosis and metabolism. These processes may be involved in the pathogenesis of certain neurodegenerative diseases. Previous studies have shown that there are high levels of NR4A3 mRNA in the CNS. Moreover, NR4A2, a transcription factor with homology to NR4A3, has been reported to contribute to the pathogenesis of Parkinson's disease. However, it is uncertain whether NR4A3 is also involved in diseases such as dementia with Lewy bodies, multiple system atrophy, and other neurodegenerative disorders such as tauopathies, TDP-43 proteinopathies and polyglutamine diseases. In the present study we used immunohistochemistry to examine the brain and spinal cord from patients with various neurodegenerative diseases and normal control subjects using two polyclonal anti-NR4A3 antibodies. In controls, the cytoplasm of neurons and glial cells was faintly immunostained with anti-NR4A3 antibodies. In tissues from patients with neurodegenerative diseases, immunoreactivity for NR4A3 was observed in cortical and brainstem-type Lewy bodies in Parkinson's disease and in dementia with Lewy bodies, as well as in neuronal and glial cytoplasmic inclusions in multiple system atrophy. A double-labeled immunofluorescence study showed co-localization of NR4A3 and phosphorylated α-synuclein in these inclusions. Neuronal and glial inclusions in other neurodegenerative disorders were NR4A3 negative. These findings suggest that accumulation of NR4A3 is specific to α-synucleinopathy.


Subject(s)
DNA-Binding Proteins/biosynthesis , Lewy Body Disease/metabolism , Multiple System Atrophy/metabolism , Receptors, Steroid/biosynthesis , Receptors, Thyroid Hormone/biosynthesis , Adult , Aged , Aged, 80 and over , Brain/metabolism , Brain/pathology , DNA-Binding Proteins/analysis , Female , Fluorescent Antibody Technique , Humans , Lewy Body Disease/pathology , Male , Middle Aged , Multiple System Atrophy/pathology , Neurons/metabolism , Neurons/pathology , Receptors, Steroid/analysis , Receptors, Thyroid Hormone/analysis
14.
Thyroid ; 24(6): 1040-50, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24684177

ABSTRACT

BACKGROUND: Progesterone (P4) is the main steroid secreted by the corpora lutea (CL) and is required for successful implantation and maintenance of pregnancy. Although adequate circulating levels of thyroid hormone (TH) are needed to support formation and maintenance of CL during pregnancy, TH signaling had not been described in this gland. We determined luteal thyroid hormone receptor isoforms (TR) expression and regulation throughout pregnancy and under the influence of thyroid status, and in vitro effects of triiodothyronine (T3) exposure on luteal P4 synthesis. METHODS: Euthyroid female Wistar rats were sacrificed by decapitation on gestational day (G) 5, G10, G15, G19, or G21 of pregnancy or on day 2 postpartum (L2). Hyperthyroidism and hypothyroidism were induced in female Wistar rats by daily administration of thyroxine (T4; 0.25 mg/kg subcutaneously) or 6-propyl-2-thiouracil (PTU; 0.1 g/L in drinking water), respectively. Luteal TR expression of mRNA was determined using real-time reverse-transcription quantitative polymerase chain reaction, and of protein using Western blot and immunohistochemistry. Primary cultures of luteal cells and of luteinized granulosa cells were used to study in vitro effects of T3 on P4 synthesis. In addition, the effect of T3 on P4 synthesis under basal conditions and under stimulation with luteinizing hormone (LH), prolactin (PRL), and prostaglandin E2 (PGE2) was evaluated. RESULTS: TRα1, TRα2, and TRß1 mRNA were present in CL, increasing during the first half and decreasing during the second half of pregnancy. At the protein level, TRß1 was abundantly expressed during gestation reaching a peak at G19 and decreasing afterwards. TRα1 was barely expressed during early gestation, peaked at G19, and diminished thereafter. Expression of TRß1 and TRα1 at the protein and mRNA level were not influenced by thyroid status. T3 neither modified P4 secretion from CL of pregnancy nor its synthesis in luteinized granulosa cells in culture. CONCLUSIONS: This study confirms for the first time the presence of TR isoforms in the CL during pregnancy and postpartum, identifying this gland as a TH target during gestation. TR expression is modulated in this tissue in accordance with the regulation of P4 metabolism, and the abrupt peripartum changes suggest a role of TH during luteolysis. However, TH actions on the CL do not seem to be related to a direct regulation of P4 synthesis.


Subject(s)
Corpus Luteum/metabolism , Postpartum Period/metabolism , Receptors, Thyroid Hormone/biosynthesis , Animals , Female , Luteinizing Hormone , Pregnancy/metabolism , Progesterone/biosynthesis , Prolactin , Propylthiouracil/pharmacology , Protein Isoforms/biosynthesis , RNA, Messenger/metabolism , Rats, Wistar , Thyrotropin/biosynthesis , Triiodothyronine/pharmacology
15.
Exp Biol Med (Maywood) ; 239(6): 707-14, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24676904

ABSTRACT

Protein disulfide isomerase (PDI) is a multifunctional protein and plays important roles in protein folding, triglyceride transfer, insulin degradation, and thyroid hormone transportation. This study examined the modulation of PDI expression by soy consumption using rat as a model. Sprague-Dawley male and female rats at 50 days (d) of age were fed diets containing either 20% casein or alcohol-washed soy protein isolate (SPI, containing 50 mg isoflavones (ISFs)/kg diet) or SPI plus ISF (250 mg/kg diet) and mated at age of 120 d. The offspring (F1) were fed the same diets as their parents. Addition of ISF to SPI diet markedly increased PDI protein content in the liver and testis of the adult rats compared with the casein or SPI diet. PDI mRNA abundance in the liver and protein content in the brain, thyroid, heart, and uterus were unchanged by the diets. Two-dimensional Western blot showed that the rats fed diets containing SPI had a diminished hepatic PDI protein with an isoelectric point (pI) of 6.12, a dephosphorylated form, compared with the rats fed diets containing either casein or SPI with supplemental ISF. Soy ISF added into SPI diet remarkably suppressed hepatic PDI activity of the rats compared with the casein diet. Moreover, soy ISF dose-dependently increased PDI and thyroid hormone receptor (TR) ß protein content, whereas reduced TR DNA binding ability in human hepatocytes. Overall, this study shows that soy ISF increased hepatic PDI protein content, but addition of ISF into SPI diet inhibited its enzymatic activities and this effect may be mediated through a post-transcriptional mechanism.


Subject(s)
Gene Expression Regulation, Enzymologic/drug effects , Glycine max/chemistry , Isoflavones/pharmacology , Liver/enzymology , Plant Proteins, Dietary/pharmacology , Protein Disulfide-Isomerases/biosynthesis , Soybean Proteins/pharmacology , Animals , Female , Humans , Isoflavones/chemistry , Male , Plant Proteins, Dietary/chemistry , Rats , Rats, Sprague-Dawley , Receptors, Thyroid Hormone/biosynthesis , Soybean Proteins/chemistry
16.
J Ovarian Res ; 7: 23, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24528603

ABSTRACT

BACKGROUND: The standard treatment of ovarian cancer with chemotherapy often leads to drug resistance and relapse of the disease, and the need for development of novel therapy alternatives is obvious. The MOC31PE immunotoxin binds to the cell surface antigen EpCAM, which is expressed by the majority of epithelial cancers including ovarian carcinomas, and we studied the cytotoxic effects of MOC31PE in ovarian cancer cells. METHODS: Investigation of the effects of MOC31PE treatment on protein synthesis, cell viability, proliferation and gene expression of the ovarian cancer cell lines B76 and HOC7. RESULTS: MOC31PE treatment for 24 h caused a dose-dependent reduction of protein synthesis with ID50 values of less than 10 ng/ml, followed by reduced cell viability. In a gene expression array monitoring the expression of 84 key genes in cancer pathways, 13 of the genes were differentially expressed by MOC31PE treatment in comparison to untreated cells. By combining MOC31PE and the immune suppressor cyclosporin A (CsA) the MOC31PE effect on protein synthesis inhibition and cell viability increased tenfold. Cell migration was also reduced, both in the individual MOC31PE and CsA treatment, but even more when combining MOC31PE and CsA. In tumor metastasis PCR arrays, 23 of 84 genes were differentially expressed comparing CsA versus MOC31PE + CsA treatment. Increased expression of the tumor suppressor KISS1 and the nuclear receptor NR4A3 was observed, and the differential candidate gene expression was confirmed in complementary qPCR analyses. For NR4A3 this was not accompanied by increased protein expression. However, a subcellular fractionation assay revealed increased mitochondrial NR4A3 in MOC31PE treated cells, suggesting a role for this protein in MOC31PE-induced apoptotic cell death. CONCLUSION: The present study demonstrates that MOC31PE may become a new targeted therapy for ovarian cancer and that the MOC31PE anti-cancer effect is potentiated by CsA.


Subject(s)
Cell Movement/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Immunoconjugates/pharmacology , Immunotoxins/pharmacology , Ovarian Neoplasms/pathology , Antineoplastic Agents/pharmacology , Cell Death/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cyclosporine/pharmacology , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Dose-Response Relationship, Drug , Drug Synergism , Female , Gene Expression Profiling/methods , Humans , Inhibitory Concentration 50 , Kisspeptins/biosynthesis , Kisspeptins/genetics , Oligonucleotide Array Sequence Analysis , Ovarian Neoplasms/genetics , Ovarian Neoplasms/immunology , Ovarian Neoplasms/metabolism , Polymerase Chain Reaction , Receptors, Steroid/biosynthesis , Receptors, Steroid/genetics , Receptors, Thyroid Hormone/biosynthesis , Receptors, Thyroid Hormone/genetics , Time Factors , Up-Regulation
17.
Virology ; 446(1-2): 173-9, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24074579

ABSTRACT

Erythrovirus B19 (B19V) is a member of the family Parvoviridae. Infection with B19V has been linked to a variety of diseases including erythroid, thyroid, neurological and autoimmune diseases. Here we show that infection of primary CD36+ cells with B19V coincides with downregulation of thyroid, retinoid, and estrogen hormone receptors. In addition we show changes in expression of a variety of related downstream signaling genes participating in cancer and cardiac-related diseases in B19V-infected erythroid primary cells.


Subject(s)
Host-Pathogen Interactions , Myeloid Progenitor Cells/virology , Parvovirus B19, Human/physiology , Receptors, Estrogen/biosynthesis , Receptors, Retinoic Acid/biosynthesis , Receptors, Thyroid Hormone/biosynthesis , Virus Replication , CD36 Antigens/analysis , Cells, Cultured , Down-Regulation , Humans , Myeloid Progenitor Cells/chemistry , Signal Transduction
18.
J Biol Chem ; 288(22): 16155-66, 2013 May 31.
Article in English | MEDLINE | ID: mdl-23595988

ABSTRACT

One goal of diabetic regenerative medicine is to instructively convert mature pancreatic exocrine cells into insulin-producing cells. We recently reported that ligand-bound thyroid hormone receptor α (TRα) plays a critical role in expansion of the ß-cell mass during postnatal development. Here, we used an adenovirus vector that expresses TRα driven by the amylase 2 promoter (AdAmy2TRα) to induce the reprogramming of pancreatic acinar cells into insulin-producing cells. Treatment with l-3,5,3-triiodothyronine increases the association of TRα with the p85α subunit of phosphatidylinositol 3-kinase (PI3K), leading to the phosphorylation and activation of Akt and the expression of Pdx1, Ngn3, and MafA in purified acinar cells. Analyses performed with the lectin-associated cell lineage tracing system and the Cre/loxP-based direct cell lineage tracing system indicate that newly synthesized insulin-producing cells originate from elastase-expressing pancreatic acinar cells. Insulin-containing secretory granules were identified in these cells by electron microscopy. The inhibition of p85α expression by siRNA or the inhibition of PI3K by LY294002 prevents the expression of Pdx1, Ngn3, and MafA and the reprogramming to insulin-producing cells. In immunodeficient mice with streptozotocin-induced hyperglycemia, treatment with AdAmy2TRα leads to the reprogramming of pancreatic acinar cells to insulin-producing cells in vivo. Our findings suggest that ligand-bound TRα plays a critical role in ß-cell regeneration during postnatal development via activation of PI3K signaling.


Subject(s)
Acinar Cells/metabolism , Cell Dedifferentiation , Insulin-Secreting Cells/metabolism , Receptors, Thyroid Hormone/biosynthesis , Triiodothyronine/pharmacology , Acinar Cells/cytology , Adenoviridae , Animals , Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Basic Helix-Loop-Helix Transcription Factors/genetics , Chromones/pharmacology , Enzyme Inhibitors/pharmacology , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Insulin-Secreting Cells/cytology , Maf Transcription Factors, Large/biosynthesis , Maf Transcription Factors, Large/genetics , Mice , Mice, Inbred BALB C , Mice, Nude , Mice, Transgenic , Morpholines/pharmacology , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Pancreatic alpha-Amylases/genetics , Pancreatic alpha-Amylases/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Thyroid Hormone/genetics , Signal Transduction/drug effects , Signal Transduction/physiology , Trans-Activators/biosynthesis , Trans-Activators/genetics , Transduction, Genetic
19.
Endocr Res ; 38(2): 112-8, 2013.
Article in English | MEDLINE | ID: mdl-23545000

ABSTRACT

BACKGROUND: Tetraiodothyroacetic acid (tetrac) and its nanoparticulate formulation (Nanotetrac) act at a cell surface receptor to block angiogenesis and tumor cell proliferation. OBJECTIVE: The complex anti-angiogenic properties of tetrac and Nanotetrac caused us to search in the literature and in certain of our unpublished mRNA experiments for evidence that these agents affect the early inflammatory response, perhaps through actions on specific cytokines and chemokines. RESULTS AND DISCUSSION: Tetrac and Nanotetrac inhibit expression in tumor cells of cytokine genes, e.g., specific interleukins, and chemokine genes, such as fractalkine (CX3CL1), and chemokine receptor genes (CX3CR1) that have been identified as high priority targets in the development of inflammation-suppressant drugs. The possibility is also examined that tetrac formulations have an effect on the function of inflammatory cells.


Subject(s)
Cytokines/metabolism , Inflammation/immunology , Inflammation/pathology , Thyroxine/analogs & derivatives , Cytokines/biosynthesis , Cytokines/genetics , Humans , Inflammation/metabolism , Interleukins/biosynthesis , Nanoparticles/administration & dosage , Nanoparticles/therapeutic use , Receptors, Thyroid Hormone/biosynthesis , Thyroxine/pharmacology
20.
Hum Mol Genet ; 22(10): 1949-59, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23390133

ABSTRACT

We have previously shown that NOR-1 (NR4A3) modulates the proliferation and survival of vascular cells in culture. However, in genetically modified animal models, somewhat conflicting results have been reported concerning the involvement of NOR-1 in neointimal formation after vascular injury. The aim of this study was to generate a transgenic mouse model over-expressing NOR-1 in smooth muscle cells (SMCs) and assess the consequence of a gain of function of this receptor on intimal hyperplasia after vascular injury. The transgene construct (SM22-NOR1) was prepared by ligating the full-length human NOR-1 cDNA (hNOR-1) and a mouse SM22α minimal promoter able to drive NOR-1 expression to SMC. Two founders were generated and two stable transgenic mouse lines (TgNOR-1) were established by backcrossing the transgene-carrying founders with C57BL/6J mice. Real-time PCR and immunohistochemistry confirmed that hNOR-1 was mainly targeted to vascular beds such as aorta and carotid arteries, and was similar in both transgenic lines. Vascular SMC from transgenic animals exhibit increased NOR-1 transcriptional activity (assessed by electrophoretic mobility shift assay and luciferase assays), increased mitogenic activity (determined by [(3)H]-thymidine incorporation; 1.58-fold induction, P < 0.001) and increased expression of embryonic smooth muscle myosin heavy chain (SMemb) than wild-type cells from control littermates. Using the carotid artery ligation model, we show that neointima formation was increased in transgenic versus wild-type mice (2.36-fold induction, P < 0.01). Our in vivo data support a role for NOR-1 in VSMC proliferation and vascular remodelling. This NOR-1 transgenic mouse could be a useful model to study fibroproliferative vascular diseases.


Subject(s)
Carotid Arteries/metabolism , Carotid Artery Injuries/metabolism , DNA-Binding Proteins/biosynthesis , Neointima/metabolism , Nerve Tissue Proteins/biosynthesis , Receptors, Steroid/biosynthesis , Receptors, Thyroid Hormone/biosynthesis , Animals , Carotid Arteries/pathology , Carotid Artery Injuries/genetics , Carotid Artery Injuries/pathology , DNA-Binding Proteins/genetics , Humans , Hyperplasia/genetics , Hyperplasia/metabolism , Hyperplasia/pathology , Mice , Mice, Transgenic , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Neointima/genetics , Neointima/pathology , Nerve Tissue Proteins/genetics , Rats , Receptors, Steroid/genetics , Receptors, Thyroid Hormone/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...