Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
J Clin Endocrinol Metab ; 102(3): 776-785, 2017 03 01.
Article in English | MEDLINE | ID: mdl-27768856

ABSTRACT

Context: The sources and biological impact of 3,3',5,5' tetraiodothyroacetic acid (TA4) are uncertain. CD34+ fibrocytes express several proteins involved in the production of thyroid hormones. They infiltrate the orbit in Graves disease (GD), an autoimmune process known as thyroid-associated ophthalmopathy. It appears that the thyrotropin receptor plays an important role in the pathogenesis of thyroid-associated ophthalmopathy. Objective: To quantify levels of TA4 in healthy participants and those with GD, determine whether fibrocytes generate this thyroid hormone analogue, and determine whether TA4 influences the actions of thyroid-stimulating hormone and thyroid-stimulating immunoglobulins in orbital fibroblasts. Design/Setting/Participants: Patients with GD and healthy donors in an academic medical center clinical practice were recruited. Main Outcome Measures: Liquid chromatography-tandem mass spectrometry, autoradiography, real-time polymerase chain reaction, hyaluronan immunoassay. Results: Serum levels of TA4 are elevated in GD. TA4 levels are positively correlated with those of thyroxine and negatively correlated with serum levels of triiodothyronine. Several cell types in culture generate TA4 from ambient thyroxine, including fibrocytes, HELA cells, human Müller stem cells, and retinal pigmented epithelial cells. Propylthiouracil inhibits TA4 generation. TA4 enhances the induction by thyrotropin and thyroid-stimulating immunoglobulins of several participants in the pathogenesis of thyroid-associated ophthalmopathy, including interleukin 6, hyaluronan synthase 1, prostaglandin endoperoxide H synthase 2, and haluronan production. Conclusion: TA4 may be ubiquitously generated in many tissues and enhances the biological impact of thyrotropin and thyroid-stimulating immunoglobulins in orbital connective tissue. These findings may identify a physiologically important determinant of extrathyroidal thyroid-stimulating hormone action.


Subject(s)
Graves Disease/blood , Graves Ophthalmopathy/blood , Thyroxine/analogs & derivatives , Adult , Aged , Aged, 80 and over , Autoradiography , Case-Control Studies , Cells, Cultured , Chromatography, Liquid , Ependymoglial Cells/metabolism , Female , Fibroblasts/metabolism , Flow Cytometry , Glucuronosyltransferase/metabolism , Graves Disease/complications , Graves Ophthalmopathy/etiology , HeLa Cells , Humans , Hyaluronan Synthases , Hyaluronic Acid/metabolism , Immunoassay , Immunoglobulins, Thyroid-Stimulating/metabolism , Interleukin-6/metabolism , Male , Middle Aged , Orbit , Prostaglandin-Endoperoxide Synthases/metabolism , Real-Time Polymerase Chain Reaction , Receptors, Thyrotropin/metabolism , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/metabolism , Sex Factors , Tandem Mass Spectrometry , Thyrotropin/metabolism , Thyroxine/blood , Triiodothyronine/blood , Young Adult
2.
PLoS One ; 11(9): e0162994, 2016.
Article in English | MEDLINE | ID: mdl-27631497

ABSTRACT

CONTEXT: Fibrocytes appear to participate in inflammation and tissue remodeling in patients with thyroid-associated ophthalmopathy (TAO). These patients have increased frequencies of circulating TSH receptor (TSHR)- and CD40-positive fibrocytes, suggesting TSHR and CD40 may play roles in proinflammatory cytokine production, which ultimately leads to orbital inflammation and tissue remodeling. OBJECTIVE: To investigate the potential interactions between the TSHR and CD40 signaling pathways and their roles in IL-6 and TNF-α production. DESIGN AND OUTCOME MEASURES: CD40 expression on fibrocytes was assessed using flow cytometry; IL-6 and TNF-α protein release using Luminex technology; increased IL-6 and TNF-α mRNA abundance, using real-time PCR; TSH- and CD40 ligand (CD40L)-stimulated Akt phosphorylation in fibrocytes, by western blot analysis; TSHR-CD40 protein-protein interaction, using co-immunoprecipitation, and CD40-TSHR co-localization, using immunocytochemistry. RESULTS: TSH enhances CD40 expression at a pre-translational level in fibrocytes. Production of IL-6 and TNF-α after costimulation with TSH and CD40L was greater than that after TSH or CD40L stimulation alone. TSH and CD40L costimulation also resulted in greater Akt phosphorylation. Akt and nuclear factor (NF)-κB inhibitors significantly reduced cytokine production after TSH and CD40L costimulation. TSHR and CD40L are colocalized on the cell surface and form a complex. CONCLUSIONS: TSHR and CD40 in fibrocytes appear to be physically and functionally related. TSH stimulates CD40 production on the fibrocyte surface. Cytokine expression upon simultaneous stimulation of TSHR and CD40 is greater than levels achieved with TSH or CD40L alone. Increased expression of CD40 by TSH is a potential mechanism for this process.


Subject(s)
CD40 Antigens/metabolism , Fibroblasts/immunology , Thyrotropin/physiology , Cytokines/biosynthesis , Humans , Receptors, Thyrotropin/metabolism , Signal Transduction
3.
J Nutr Biochem ; 25(10): 1066-76, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25086780

ABSTRACT

Early life nutritional intervention causes adult-onset insulin resistance and obesity in rats. Thyroid hormone receptor (TR), in turn, transcriptionally enhances skeletal muscle Glut4 expression. We tested the hypothesis that reduced circulating thyroid-stimulating hormone and T4 concentrations encountered in postnatal (PN4-PN24) high-carbohydrate (HC) milk formula-fed versus the mother-fed controls (MF) would epigenetically interfere with TR induction of adult (100 days) male rat skeletal muscle Glut4 expression, thereby providing a molecular mechanism mediating insulin resistance. We observed increased DNA methylation of the CpG island with enhanced recruitment of Dnmt3a, Dnmt3b and MeCP2 in the glut4 promoter region along with reduced acetylation of histone (H)2A.Z and H4 particularly at the H4.lysine (K)16 residue, which was predominantly mediated by histone deacetylase 4 (HDAC4). This was followed by enhanced recruitment of heterochromatin protein 1ß to the glut4 promoter with increased Suv39H1 methylase concentrations. These changes reduced TR binding of the T3 response element of the glut4 gene (TREs; -473 to -450 bp) detected qualitatively in vivo (electromobility shift assay) and quantified ex vivo (chromatin immunoprecipitation). In addition, the recruitment of steroid receptor coactivator and CREB-binding protein to the glut4 promoter-protein complex was reduced. Co-immunoprecipitation experiments confirmed the interaction between TR and CBP to be reduced and HDAC4 to be enhanced in HC versus MF groups. These molecular changes were associated with diminished skeletal muscle Glut4 mRNA and protein concentrations. We conclude that early postnatal exposure to HC diet epigenetically reduced TR induction of adult male skeletal muscle Glut4 expression, uncovering novel molecular mechanisms contributing to adult insulin resistance and obesity.


Subject(s)
Dietary Carbohydrates/administration & dosage , Epigenesis, Genetic , Glucose Transporter Type 4/metabolism , Muscle, Skeletal/metabolism , Receptors, Thyroid Hormone/genetics , Animals , Animals, Newborn , CREB-Binding Protein/genetics , CREB-Binding Protein/metabolism , Cells, Cultured , CpG Islands , DNA Methylation , Glucose Transporter Type 4/genetics , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Histones/genetics , Histones/metabolism , Immunoprecipitation , Insulin Resistance , Male , Muscle, Skeletal/cytology , Postnatal Care , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Receptors, Thyroid Hormone/blood , Thyroxine/blood , Triiodothyronine/blood
4.
J Clin Endocrinol Metab ; 99(9): E1635-40, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24878056

ABSTRACT

CONTEXT: Thyroid-associated ophthalmopathy (TAO) is the component of Graves' disease characterized by orbital inflammation and connective tissue remodeling. The IGF-1 receptor (IGF-1R) and TSH receptor (TSHR) form a physical and functional complex in orbital fibroblasts. A subset of these fibroblasts is derived from infiltrating CD34(+) fibrocytes. Teprotumumab (RV 001, R1507) is a human monoclonal anti-IGF-1R blocking antibody currently undergoing a phase 2 clinical trial in patients with active TAO. OBJECTIVE: To determine whether teprotumumab inhibits the induction by TSH of IL-6 and IL-8 in fibrocytes. DESIGN: Fibrocytes were treated without or with teprotumumab in combination with IGF-1 or TSH. MAIN OUTCOME MEASURES: IL-6 and IL-8 mRNA expression and protein production were analyzed by real-time PCR and Luminex, respectively. Phosphorylated Akt (S473) levels were analyzed by Western blot. TSHR and IGF-1R display was assessed by flow cytometry. RESULTS: Fibrocyte display of IGF-1R and TSHR was reduced with teprotumumab, as were IGF-1- and TSH-dependent phosphorylated Akt levels. TSH induction of IL-6 and IL-8 mRNA and protein was also reduced by the monoclonal antibody. CONCLUSIONS: Teprotumumab attenuates the actions of both IGF-1 and TSH in fibrocytes. Specifically, it blocks the induction of proinflammatory cytokines by TSH. These results provide, at least in part, the molecular rationale for interrogating the therapeutic efficacy of this antibody in TAO.


Subject(s)
Antibodies, Blocking/pharmacology , Antibodies, Monoclonal/pharmacology , Fibroblasts/drug effects , Insulin-Like Growth Factor I/antagonists & inhibitors , Thyrotropin/antagonists & inhibitors , Antibodies, Blocking/immunology , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Humanized , Cells, Cultured , Fibroblasts/immunology , Fibroblasts/metabolism , Graves Disease/genetics , Graves Disease/immunology , Graves Disease/metabolism , Humans , Insulin-Like Growth Factor I/pharmacology , Interleukin-6/genetics , Interleukin-6/metabolism , Interleukin-8/genetics , Interleukin-8/metabolism , Protein Modification, Translational/immunology , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/metabolism , Receptor, IGF Type 1/antagonists & inhibitors , Receptor, IGF Type 1/metabolism , Thyrotropin/pharmacology
5.
PLoS One ; 8(9): e75100, 2013.
Article in English | MEDLINE | ID: mdl-24086448

ABSTRACT

IL-6 plays diverse roles in normal and disease-associated immunity such as that associated with Graves' disease (GD). In that syndrome, the orbit undergoes remodeling during a process known as thyroid-associated ophthalmopathy (TAO). Recently, CD34(+) fibrocytes were found to infiltrate the orbit in TAO where they transition into CD34(+) orbital fibroblasts. Surprisingly, fibrocytes display high levels of functional thyrotropin receptor (TSHR), the central antigen in GD. We report here that TSH and the pathogenic anti-TSHR antibodies that drive hyperthyroidism in GD induce IL-6 expression in fibrocytes and orbital fibroblasts. Unlike TSHR signaling in thyroid epithelium, that occurring in fibrocytes is completely independent of adenylate cyclase activation and cAMP generation. Instead TSH activates PDK1 and both AKT/PKB and PKC pathways. Expression and use of PKCßII switches to that of PKCµ as fibrocytes transition to TAO orbital fibroblasts. This shift is imposed by CD34(-) orbital fibroblasts but reverts when CD34(+) fibroblasts are isolated. The up-regulation of IL-6 by TSH results from coordinately enhanced IL-6 gene promoter activity and increased IL-6 mRNA stability. TSH-dependent IL-6 expression requires activity at both CREB (-213 to -208 nt) and NF-κB (-78 to -62 nt) binding sites. These results provide novel insights into the molecular action of TSH and signaling downstream for TSHR in non-thyroid cells. Fibrocytes neither express adenylate cyclase nor generate cAMP and thus these findings are free from any influence of cAMP-related signaling. They identify potential therapeutic targets for TAO.


Subject(s)
Fibroblasts/metabolism , Gene Expression Regulation/drug effects , Graves Disease/physiopathology , Graves Ophthalmopathy/physiopathology , Interleukin-6/metabolism , Thyrotropin/pharmacology , Antigens, CD34/metabolism , Blotting, Western , Cyclic AMP/metabolism , DNA Primers/genetics , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Mutagenesis, Site-Directed , Protein Serine-Threonine Kinases/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Real-Time Polymerase Chain Reaction
6.
Am J Physiol Endocrinol Metab ; 304(6): E583-98, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23321477

ABSTRACT

We have shown in vitro a hypoxia-induced time-dependent increase in facilitative glucose transporter isoform 3 (GLUT3) expression in N2A murine neuroblasts. This increase in GLUT3 expression is partially reliant on a transcriptional increase noted in actinomycin D and cycloheximide pretreatment experiments. Transient transfection assays in N2A neuroblasts using murine glut3-luciferase reporter constructs mapped the hypoxia-induced enhancer activities to -857- to -573-bp and -203- to -177-bp regions. Hypoxia-exposed N2A nuclear extracts demonstrated an increase in HIF-1α and p-Creb binding to HRE (-828 to -824 bp) and AP-1 (-187 to -180 bp) cis-elements, respectively, in electromobility shift and supershift assays, which was confirmed by chromatin immunoprecipitation assays. In addition, the interaction of CBP with Creb and HIF-1α and CREST with CBP in hypoxia was detected by coimmunoprecipitation. Furthermore, small interference (si)RNA targeting Creb in these cells decreased endogenous Creb concentrations that reduced by twofold hypoxia-induced glut3 gene transcription. Thus, in N2A neuroblasts, phosphorylated HIF-1α and Creb mediated the hypoxia-induced increase in glut3 transcription. Coactivation by the Ca⁺⁺-dependent CREST and CBP proteins may enhance cross-talk between p-Creb-AP-1 and HIF-1α/HRE of the glut3 gene. Collectively, these processes can facilitate an adaptive response to hypoxic energy depletion targeted at enhancing glucose transport and minimizing injury while fueling the proliferative potential of neuroblasts.


Subject(s)
CREB-Binding Protein/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Glucose Transporter Type 3/biosynthesis , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Neural Stem Cells/metabolism , Trans-Activators/metabolism , Transcriptional Activation , Animals , Cell Hypoxia , Cell Line , Cell Nucleus/metabolism , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/genetics , Genes, Reporter , Glucose Transporter Type 3/genetics , Glucose Transporter Type 3/metabolism , Mice , Mice, Inbred BALB C , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Stem Cells/cytology , Neurons/cytology , Neurons/metabolism , Protein Transport , RNA Interference , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/metabolism , Signal Transduction , Up-Regulation
7.
Invest Ophthalmol Vis Sci ; 53(12): 7746-53, 2012 Nov 21.
Article in English | MEDLINE | ID: mdl-23092922

ABSTRACT

PURPOSE: CD40-CD40 ligand (CD40L) interactions appear to play pathogenic roles in autoimmune disease. Here we quantify CD40 expression on fibrocytes, circulating, and bone marrow-derived progenitor cells. The functional consequences of CD40 ligation are determined since these may promote tissue remodeling linked with thyroid-associated ophthalmopathy (TAO). METHODS: CD40 levels on cultivated fibrocytes and orbital fibroblasts (GOFB) from patients with Graves' disease (GD), as well as fibrocyte abundance, were determined by flow cytometry. CD40 mRNA expression was evaluated by real-time PCR, whereas response to CD40 ligation was measured by Luminex and RT-PCR. Protein kinase B (Akt) and nuclear factor (NF)-kappa B (NF-κB) signaling were determined by Western blot and immunofluorescence. RESULTS: Basal CD40 expression on fibrocytes is greater than that on GOFB. IFN-γ upregulates CD40 in both cell types and its actions are mediated at the pretranslational level. Fibrocytes produce high levels of cytokines, including interleukin-6 (IL-6), TNF-α, IL-8, MCP-1, and RANTES (Regulated on Activation, Normal T Cell Expressed and Secreted) in response to CD40L. IL-6 induction results from an increase in steady state IL-6 mRNA, and is mediated through Akt and NF-κB activation. Circulating CD40(+)CD45(+)Col1(+) fibrocytes are far more frequent in vivo in donors with TAO compared with healthy subjects. CONCLUSIONS: Particularly high levels of functional CD40 are displayed by fibrocytes. CD40L-provoked signaling results in the production of several cytokines. Among these, IL-6 expression is mediated through Akt and NF-κB pathways. The frequency of circulating CD40(+) fibrocytes is markedly increased in patients with TAO, suggesting that this receptor might represent a therapeutic target for TAO.


Subject(s)
CD40 Antigens/genetics , Gene Expression Regulation , Graves Ophthalmopathy/genetics , Interleukin-6/genetics , NF-kappa B/genetics , Proto-Oncogene Proteins c-akt/genetics , RNA, Messenger/genetics , Adult , Blotting, Western , CD40 Antigens/biosynthesis , Cells, Cultured , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Flow Cytometry , Follow-Up Studies , Graves Ophthalmopathy/metabolism , Graves Ophthalmopathy/pathology , Humans , Interleukin-6/biosynthesis , Male , Microscopy, Fluorescence , Middle Aged , NF-kappa B/biosynthesis , Orbit/pathology , Proto-Oncogene Proteins c-akt/biosynthesis , Real-Time Polymerase Chain Reaction , Signal Transduction
8.
Proc Natl Acad Sci U S A ; 109(19): 7427-32, 2012 May 08.
Article in English | MEDLINE | ID: mdl-22517745

ABSTRACT

Thyroglobulin (Tg) is the macromolecular precursor of thyroid hormones and is thought to be uniquely expressed by thyroid epithelial cells. Tg and the thyroid-stimulating hormone receptor (TSHR) are targets for autoantibody generation in the autoimmune disorder Graves disease (GD). Fully expressed GD is characterized by thyroid overactivity and orbital tissue inflammation and remodeling. This process is known as thyroid-associated ophthalmopathy (TAO). Early reports suggested that in TAO, both Tg and TSHR become overexpressed in orbital tissues. Previously, we found that CD34(+) progenitor cells, known as fibrocytes, express functional TSHR, infiltrate the orbit, and comprise a large subset of orbital fibroblasts in TAO. We now report that fibrocytes also express Tg, which resolves as a 305-kDa protein on Western blots. It can be immunoprecipitated with anti-Tg Abs. Further, (125)iodine and [(35)S]methionine are incorporated into Tg expressed by fibrocytes. De novo Tg synthesis is attenuated with a specific small interfering RNA targeting the protein. A fragment of the Tg gene promoter fused to a luciferase reporter exhibits substantial activity when transfected into fibrocytes. Unlike fibrocytes, GD orbital fibroblasts, which comprise a mixture of CD34(+) and CD34(-) cells, express much lower levels of Tg and TSHR. When sorted into pure CD34(+) and CD34(-) subsets, Tg and TSHR mRNA levels become substantially higher in CD34(+) cells. These findings indicate that human fibrocytes express multiple "thyroid-specific" proteins, the levels of which are reduced after they infiltrate tissue. Our observations establish the basis for Tg accumulation in orbital GD.


Subject(s)
Fibroblasts/metabolism , Mesenchymal Stem Cells/metabolism , Receptors, Thyrotropin/metabolism , Thyroglobulin/metabolism , Antigens, CD34/metabolism , Blotting, Western , Cells, Cultured , Connective Tissue Cells/metabolism , Gene Expression , Graves Ophthalmopathy/genetics , Graves Ophthalmopathy/metabolism , Graves Ophthalmopathy/pathology , Humans , Immunohistochemistry , Leukocyte Common Antigens/metabolism , Orbit/metabolism , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Thyrotropin/genetics , Reverse Transcriptase Polymerase Chain Reaction , Thyroglobulin/genetics , Thyroid Gland/cytology , Thyroid Gland/metabolism
9.
J Clin Endocrinol Metab ; 97(5): E740-6, 2012 May.
Article in English | MEDLINE | ID: mdl-22399514

ABSTRACT

CONTEXT: The molecular basis for anatomically dispersed clinical manifestations in Graves' disease (GD) eludes our understanding. Bone marrow-derived, pluripotent fibrocytes represent a subset of peripheral blood mononuclear cells and infiltrate the orbital and thyroid tissues in GD. These cells may be involved in the pathogenesis of thyroid-associated ophthalmopathy (TAO). OBJECTIVE: The objective of the study was to quantify fibrocyte display of functional cell surface TSH receptor (TSHR), identify the profile of chemokines they express after TSHR activation, and determine whether circulating TSHR(+) peripheral blood fibrocytes are more frequent in situ in patients with TAO. DESIGN/SETTING/PARTICIPANTS: Using a newly developed technique, fibrocytes were directly identified in peripheral blood from 31 patients with TAO and 19 healthy subjects receiving care at a multidisciplinary academic center. MAIN OUTCOME MEASURES: The frequency in situ of fibrocytes (collagen 1(+), CD45(+), CD34(+), CD14(+), CD86(+) peripheral blood mononuclear cells) was assessed by multiparameter flow cytometry and correlated to clinical disease activity and smoking status. Levels of TSHR-displaying fibrocytes and their response to TSH and TSHR-activating antibody, M22, were measured by flow cytometry, Luminex, and real-time PCR. RESULTS: The levels of TSHR expression by fibrocytes are substantially higher than those found in orbital fibroblasts. Moreover, the frequency of TSHR(+) fibrocytes in patients with TAO was greater than that in healthy subjects in situ. Their abundance is not influenced by disease activity or smoking history. These cells produce high levels of several cytokines and chemokines including IL-8, regulated upon activation, normal T cell expressed and secreted, and monocyte chemoattractant protein-1 when treated with TSH or M22. TSH induces IL-8 production at the pretranslational level. This induced cytokine can be detected in intact fibrocytes ex vivo. CONCLUSIONS: Frequency of circulating TSHR(+) fibrocytes is markedly increased in patients with TAO, and they express proinflammatory chemokines in response to TSH. Because they infiltrate both orbit and thyroid in GD, they may represent the link between systemic immunoreactivity and organ-specific autoimmunity.


Subject(s)
Chemokines/biosynthesis , Graves Ophthalmopathy/metabolism , Leukocytes, Mononuclear/metabolism , Receptors, Thyrotropin/metabolism , Adult , Cells, Cultured , Chemokine CCL2/metabolism , Female , Flow Cytometry , Graves Ophthalmopathy/genetics , Graves Ophthalmopathy/immunology , Humans , Interleukin-8/metabolism , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Male , Receptors, Thyrotropin/genetics , Thyrotropin/metabolism , Thyrotropin/pharmacology
10.
J Clin Endocrinol Metab ; 96(12): 3827-37, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21956421

ABSTRACT

CONTEXT: Graves' disease (GD) is a systemic autoimmune syndrome comprising manifestations in thyroid and orbital connective tissue. The link between these two tissues in GD eludes our understanding. Patients with GD have increased frequency of circulating monocyte lineage cells known as fibrocytes. These fibrocytes infiltrate orbital connective tissues in thyroid-associated ophthalmopathy and express functional TSH receptor (TSHR). OBJECTIVE: The aim of the study was to identify and characterize CD34(+) fibrocytes in thyroid tissue. DESIGN/SETTING/PARTICIPANTS: Patients undergoing surgical thyroidectomy at two academic medical centers were recruited to the study. MAIN OUTCOME MEASURES: We performed immunohistochemistry, flow cytometry, real-time PCR, cytokine-specific ELISA, and cell differentiation. RESULTS: CD34(+)ColI(+)CXCR4(+)TSHR(+) cells can be identified in situ in thyroid tissue from donors with GD, Hashimoto's thyroiditis, or in normal-appearing tissue. Thyroid fibroblasts cultivated from these glands express a CD34(-)ColI(+)CXCR4(+)TSHR(+) phenotype. TSHR levels are higher than those in orbital fibroblasts. When treated with TSH, thyroid fibroblasts generate IL-6 and IL-8. The induction of IL-6 can be blocked by dexamethasone, a chemical inhibitor of Akt/Pkb, and by knocking down Akt with a specific small interfering RNA. When treated with TGF-ß or rosiglitazone, thyroid fibroblasts differentiate into myofibrocytes or adipocytes, respectively. CONCLUSIONS: ColI(+)CXCR4(+)TSHR(+) thyroid fibroblasts resemble orbital fibroblasts and circulating fibrocytes. CD34(+) fibrocytes appear to infiltrate both tissues in GD. Thyroid fibroblasts lose CD34 display in culture, unlike orbital fibroblasts and circulating fibrocytes. Fibrocytes and their fibroblast derivatives may participate in the pathogenesis of thyroid autoimmunity after TSHR activation. They could represent a therapeutic target for these diseases.


Subject(s)
Fibroblasts/metabolism , Graves Disease/metabolism , Orbit/metabolism , Receptors, Thyrotropin/metabolism , Thyroid Gland/metabolism , Adult , Cell Differentiation , Cells, Cultured , Female , Graves Disease/genetics , Graves Disease/surgery , Humans , Interleukin-6/genetics , Interleukin-6/metabolism , Interleukin-8/genetics , Interleukin-8/metabolism , Male , Middle Aged , Receptors, Thyrotropin/genetics , Thyroid Gland/surgery , Thyroidectomy
11.
PLoS One ; 5(12): e15296, 2010 Dec 23.
Article in English | MEDLINE | ID: mdl-21209948

ABSTRACT

BACKGROUND: IL-6 plays an important role in the pathogenesis of Graves' disease and its orbital component, thyroid-associated ophthalmopathy (TAO). Orbital tissues become inflamed in TAO, a process in which prostanoids have been implicated. Orbital fibroblasts both generate and respond to PGE(2), underlying the inflammatory phenotype of these cells. METHODOLOGY/PRINCIPAL FINDINGS: Using cultured orbital and dermal fibroblasts, we characterized the effects of PGE(2) on IL-6 expression. We found that the prostanoid provokes substantially greater cytokine synthesis in orbital fibroblasts, effects that are mediated through cell-surface EP(2) receptors and increased steady-state IL-6 mRNA levels. The pre-translational up-regulation of IL-6 results from increased gene promoter activity and can be reproduced with the PKA agonist, Sp-cAMP and blocked by interrupting the PKA pathway. PGE(2)-induced production of cAMP in orbital fibroblasts was far greater than that in dermal fibroblasts, resulting from higher levels of adenylate cyclase. PGE(2) provokes CREB phosphorylation, increases the pCREB/CREB ratio, and initiates nuclear localization of the pCREB/CREB binding protein/p300 complex (CBP) preferentially in orbital fibroblasts. Transfection with siRNAs targeting either CREB or CBP blunts the induction of IL-6 gene expression. PGE(2) promotes the binding of pCREB to its target DNA sequence which is substantially greater in orbital fibroblasts. CONCLUSION/SIGNIFICANCE: These results identify the mechanism underlying the exaggerated induction of IL-6 in orbital fibroblasts and tie together two proinflammatory pathways involved in the pathogenesis of TAO. Moreover, they might therefore define an attractive therapeutic target for the treatment of TAO.


Subject(s)
Dinoprostone/metabolism , Graves Disease/metabolism , Graves Ophthalmopathy/metabolism , Interleukin-6/metabolism , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Cyclic AMP/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Dose-Response Relationship, Drug , Fibroblasts/metabolism , Humans , Phenotype , Phosphorylation , Prostaglandins/metabolism , RNA, Small Interfering/metabolism , p300-CBP Transcription Factors/metabolism
12.
J Biol Chem ; 283(20): 13611-26, 2008 May 16.
Article in English | MEDLINE | ID: mdl-18326493

ABSTRACT

We examined transcriptional and epigenetic mechanism(s) behind diminished skeletal muscle GLUT4 mRNA in intrauterine growth-restricted (IUGR) female rat offspring. An increase in MEF2D (inhibitor) with a decline in MEF2A (activator) and MyoD (co-activator) binding to the glut4 promoter in IUGR versus control was observed. The functional role of MEF2/MyoD-binding sites and neighboring three CpG clusters in glut4 gene transcription was confirmed in C2C12 muscle cells. No differential methylation of these three and other CpG clusters in the glut4 promoter occurred. DNA methyltransferase 1 (DNMT1) in postnatal, DNMT3a, and DNMT3b in adult was differentially recruited with increased MeCP2 (methyl CpG-binding protein) concentrations to bind the IUGR glut4 gene. Covalent modifications of the histone (H) code consisted of H3.K14 de-acetylation by recruitment of histone deacetylase (HDAC) 1 and enhanced association of HDAC4 enzymes. This set the stage for Suv39H1 methylase-mediated di-methylation of H3.K9 and increased recruitment of heterochromatin protein 1alpha, which partially inactivates postnatal and adult IUGR glut4 gene transcription. Further increased interactions in the adult IUGR between DNMT3a/DNMT3b and HDAC1 and MEF2D and HDAC1/HDAC4 and decreased association between MyoD and MEF2A existed. We conclude that epigenetic mechanisms consisting of histone code modifications repress skeletal muscle glut4 transcription in the postnatal period and persist in the adult female IUGR offspring.


Subject(s)
CpG Islands , Glucose Transporter Type 4/genetics , Glucose Transporter Type 4/metabolism , Histone Code , Histones/metabolism , Animals , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/metabolism , Epigenesis, Genetic , Female , Fetal Growth Retardation/genetics , MEF2 Transcription Factors , Mice , Myogenic Regulatory Factors/metabolism , Pregnancy , Rats , Rats, Sprague-Dawley
13.
Pol J Microbiol ; 54(1): 37-41, 2005.
Article in English | MEDLINE | ID: mdl-16209093

ABSTRACT

Mutants of Mesorhizobium ciceri BICC 651 were generated by N-methyl-N'-nitro-N-nitrosoguanidine mutagenesis. Siderophore overproducing mutants were identified on Chrome azurol S agar plates. One of them determined as N15 was examined for symbiotic efficiency and compared to its wild type parent i.e. BICC 651 strain under sterile conditions using Leonard jars in growth chamber and also in pots containing nonsterile alluvial field soil. It was observed that the strain N15 produced about 30% higher number of nodules per plant, fixed 25% more nitrogen per gram of nodule and caused more than 30% increased dry weight of plant shoots.


Subject(s)
Alphaproteobacteria/growth & development , Cicer/microbiology , Mutation , Siderophores/metabolism , Symbiosis , Agar , Alphaproteobacteria/genetics , Alphaproteobacteria/metabolism , Cicer/growth & development , Culture Media , Iron/metabolism , Methylnitronitrosoguanidine/pharmacology , Mutagenesis , Nitrogen Fixation , Seeds/growth & development , Seeds/microbiology
14.
J Biol Chem ; 279(25): 26768-79, 2004 Jun 18.
Article in English | MEDLINE | ID: mdl-15054091

ABSTRACT

The murine facilitative glucose transporter isoform 3 is developmentally regulated and is predominantly expressed in neurons. By employing the primer extension assay, the transcription start site of the murine Glut 3 gene in the brain was localized to -305 bp 5' to the ATG translation start codon. Transient transfection assays in N2A neuroblasts using murine GLUT3-luciferase reporter constructs mapped enhancer activities to two regions located at -203 to -177 and -104 to -29 bp flanking a previously described repressor element (-137 to -130 bp). Dephosphorylated Sp1 and Sp3 proteins from the 1- and 21-day-old mouse brain nuclear extracts bound the repressor elements, whereas both dephosphorylated and phosphorylated cAMP-response element-binding protein (CREB) in N2A, 1- and 21-day-old mouse brain nuclear extracts bound the 5'-enhancer cis-elements (-187 to -180 bp) of the Glut 3 gene, and the Y box protein MSY-1 bound the sense strand of the -83- to -69-bp region. Sp3, CREB, and MSY-1 binding to the GLUT 3 DNA was confirmed by the chromatin immunoprecipitation assay, whereas CREB and MSY-1 interaction was detected by the co-immunoprecipitation assay. Furthermore, small interference RNA targeted at CREB in N2A cells decreased endogenous CREB concentrations, and CREB mediated GLUT 3 transcription. Thus, in the murine brain similar to the N2A cells, phosphorylated CREB and MSY-1 bound the Glut 3 gene trans-activating the expression in neurons, whereas Sp1/Sp3 bound the repressor elements. We speculate that phosphorylated CREB and Sp3 also interacted to bring about GLUT 3 expression in response to development/cell differentiation and neurotransmission.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Monosaccharide Transport Proteins/biosynthesis , Monosaccharide Transport Proteins/chemistry , Nerve Tissue Proteins , Neurons/metabolism , Transcription Factors/metabolism , Transcriptional Activation , Animals , Brain/embryology , Brain/metabolism , Cell Differentiation , Cell Line , Cell Line, Tumor , Cell Nucleus/metabolism , Chromatin/metabolism , Codon, Initiator , DNA Primers/metabolism , Gene Deletion , Genes, Reporter , Glucose Transporter Type 3 , Mice , Mice, Inbred BALB C , Phosphorylation , Precipitin Tests , Promoter Regions, Genetic , Protein Binding , RNA/metabolism , RNA, Small Interfering/metabolism , Sp1 Transcription Factor/metabolism , Sp3 Transcription Factor , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...