Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Liver Int ; 44(1): 125-138, 2024 01.
Article in English | MEDLINE | ID: mdl-37872645

ABSTRACT

OBJECTIVE: Progressive hepatic fibrosis can be considered the final stage of chronic liver disease. Hepatic stellate cells (HSC) play a central role in liver fibrogenesis. Thyroid hormones (TH, e.g. thyroxine; T4 and triiodothyronine; T3) significantly affect development, growth, cell differentiation and metabolism through activation of TH receptor α and/or ß (TRα/ß). Here, we evaluated the influence of TH in hepatic fibrogenesis. DESIGN: Human liver tissue was obtained from explanted livers following transplantation. TRα-deficient (TRα-KO) and wild-type (WT) mice were fed a control or a profibrogenic methionine-choline deficient (MCD) diet. Liver tissue was assessed by qRT-PCR for fibrogenic gene expression. In vitro, HSC were treated with TGFß in the presence or absence of T3. HSC with stable TRα knockdown and TRα deficient mouse embryonic fibroblasts (MEF) were used to determine receptor-specific function. Activation of HSC and MEF was assessed using the wound healing assay, Western blotting, and qRT-PCR. RESULTS: TRα and TRß expression is downregulated in the liver during hepatic fibrogenesis in humans and mice. TRα represents the dominant isoform in HSC. In vitro, T3 blunted TGFß-induced expression of fibrogenic genes in HSC and abrogated wound healing by modulating TGFß signalling, which depended on TRα presence. In vivo, TRα-KO enhanced MCD diet-induced liver fibrogenesis. CONCLUSION: These observations indicate that TH action in non-parenchymal cells is highly relevant. The interaction of TRα with TH regulates the phenotype of HSC via the TGFß signalling pathway. Thus, the TH-TR axis may be a valuable target for future therapy of liver fibrosis.


Subject(s)
Fibroblasts , Hepatic Stellate Cells , Animals , Mice , Humans , Hepatic Stellate Cells/metabolism , Thyroid Hormones/metabolism , Thyroid Hormones/pharmacology , Thyroid Hormone Receptors alpha/genetics , Thyroid Hormone Receptors alpha/metabolism , Transforming Growth Factor beta
2.
Int J Mol Sci ; 23(21)2022 Nov 01.
Article in English | MEDLINE | ID: mdl-36362133

ABSTRACT

Hypothyroidism has been shown to reduce infarct size in rats, but the underlying mechanisms are unclear. We used isolated pressure-constant perfused hearts of control, hypothyroid and hyperthyroid mice and measured infarct size, functional parameters and phosphorylation of key molecules in cardioprotective signaling with matched heart rate. Compared with controls, hypothyroidism was cardioprotective, while hyperthyroidism was detrimental with enlarged infarct size. Next, we asked how thyroid hormone receptor α (TRα) affects ischemia/reperfusion (IR) injury. Thus, canonical and noncanonical TRα signaling was investigated in the hearts of (i) mice lacking TRα (TRα0), (ii) with a mutation in TRα DNA-binding domain (TRαGS) and (iii) in hyperthyroid TRα0 (TRα0hyper) and TRαGS mice (TRαGShyper). TRα0 mouse hearts were protected against IR injury. Furthermore, infarct size was reduced in the hearts of TRαGS mice that lack canonical TRα signaling but maintain noncanonical TRα action. Hyperthyroidism did not increase infarct size in TRα0 and TRαGS mouse hearts. These cardioprotective effects were not associated with increased phosphorylation of key proteins of RISK, SAFE and eNOS pathways. In summary, chronic hypothyroidism and the lack of canonical TRα signaling are cardioprotective in IR injury and protection is not due to favorable changes in hemodynamics.


Subject(s)
Hyperthyroidism , Hypothyroidism , Reperfusion Injury , Rats , Mice , Animals , Hypothyroidism/metabolism , Thyroid Hormone Receptors alpha/genetics , Thyroid Hormone Receptors alpha/metabolism , Hyperthyroidism/metabolism , Hemodynamics , Reperfusion Injury/metabolism , Infarction , Myocardium/metabolism
3.
Proc Natl Acad Sci U S A ; 114(52): E11323-E11332, 2017 12 26.
Article in English | MEDLINE | ID: mdl-29229863

ABSTRACT

Thyroid hormone (TH) and TH receptors (TRs) α and ß act by binding to TH response elements (TREs) in regulatory regions of target genes. This nuclear signaling is established as the canonical or type 1 pathway for TH action. Nevertheless, TRs also rapidly activate intracellular second-messenger signaling pathways independently of gene expression (noncanonical or type 3 TR signaling). To test the physiological relevance of noncanonical TR signaling, we generated knockin mice with a mutation in the TR DNA-binding domain that abrogates binding to DNA and leads to complete loss of canonical TH action. We show that several important physiological TH effects are preserved despite the disruption of DNA binding of TRα and TRß, most notably heart rate, body temperature, blood glucose, and triglyceride concentration, all of which were regulated by noncanonical TR signaling. Additionally, we confirm that TRE-binding-defective TRß leads to disruption of the hypothalamic-pituitary-thyroid axis with resistance to TH, while mutation of TRα causes a severe delay in skeletal development, thus demonstrating tissue- and TR isoform-specific canonical signaling. These findings provide in vivo evidence that noncanonical TR signaling exerts physiologically important cardiometabolic effects that are distinct from canonical actions. These data challenge the current paradigm that in vivo physiological TH action is mediated exclusively via regulation of gene transcription at the nuclear level.


Subject(s)
Hypothalamo-Hypophyseal System/metabolism , Myocardium/metabolism , Pituitary-Adrenal System/metabolism , Receptors, Thyroid Hormone/metabolism , Signal Transduction , Thyroid Hormones/metabolism , Animals , Gene Knock-In Techniques , Mice , Mice, Knockout , Receptors, Thyroid Hormone/genetics , Thyroid Hormones/genetics
4.
Nat Genet ; 37(11): 1247-52, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16228000

ABSTRACT

Incorporation of selenocysteine (Sec), through recoding of the UGA stop codon, creates a unique class of proteins. Mice lacking tRNA(Sec) die in utero, but the in vivo role of other components involved in selenoprotein synthesis is unknown, and Sec incorporation defects have not been described in humans. Deiodinases (DIOs) are selenoproteins involved in thyroid hormone metabolism. We identified three of seven siblings with clinical evidence of abnormal thyroid hormone metabolism. Their fibroblasts showed decreased DIO2 enzymatic activity not linked to the DIO2 locus. Systematic linkage analysis of genes involved in DIO2 synthesis and degradation led to the identification of an inherited Sec incorporation defect, caused by a homozygous missense mutation in SECISBP2 (also called SBP2). An unrelated child with a similar phenotype was compound heterozygous with respect to mutations in SECISBP2. Because SBP2 is epistatic to selenoprotein synthesis, these defects had a generalized effect on selenoproteins. Incomplete loss of SBP2 function probably causes the mild phenotype.


Subject(s)
Mutation, Missense/genetics , RNA-Binding Proteins/genetics , Thyroid Hormones/metabolism , Adolescent , Adult , Child , Child, Preschool , Female , Fibroblasts/enzymology , Heterozygote , Homozygote , Humans , Iodide Peroxidase/metabolism , Male , Pedigree , Siblings , Skin/enzymology
5.
Stem Cell Res ; 74: 103275, 2024 02.
Article in English | MEDLINE | ID: mdl-38100912

ABSTRACT

THRB is a nuclear receptor, regulating gene expression dependent on thyroid hormone (TH) binding. The same receptor mediates signaling pathway activation in the cytosol. The challenge is to distinguish which of the two mechanisms is responsible for physiological effects of TH. We established an iPSC cell line with two mutations (E125G_G126S) in the THRB DNA-binding domain, which abrogates nuclear action and, thus, allows to study signaling pathway activation exclusively. We also generated a THRB knockout cell line to abolish all THRB effects. Comparison of WT and these two cell lines allows attribution of thyroid hormone effects to the underlying mechanism.


Subject(s)
Induced Pluripotent Stem Cells , Humans , Induced Pluripotent Stem Cells/metabolism , Thyroid Hormones , Signal Transduction , Mutation/genetics , Cell Line , Thyroid Hormone Receptors beta/genetics , Thyroid Hormone Receptors beta/metabolism
6.
Front Endocrinol (Lausanne) ; 14: 1104388, 2023.
Article in English | MEDLINE | ID: mdl-36755907

ABSTRACT

Background and aims: Non-thyroidal illness syndrome (NTIS) is frequent in critically ill patients and associated with adverse outcomes. We aimed to characterize the evolution of NTIS in patients with acute decompensation (AD) of cirrhosis and acute-on-chronic liver failure (ACLF), since NTIS is not well described in these newly defined syndromes. Methods: Thyroid hormones (TH) were quantified at baseline in consecutive patients with cirrhosis. In addition, 76 inflammatory mediators were quantified by proximity extension analysis assay in a subgroup of patients. Associations between TH, cirrhosis stage, mortality and inflammation were assessed. Results: Overall, 437 patients were included, of whom 165 (37.8%), 211 (48.3%), and 61 (14%) had compensated cirrhosis (CC), AD, and ACLF. FT3 concentrations were lower in AD versus CC, and further decreased in ACLF. Importantly, NTIS was present in 83 (39.3%) patients with AD and in 44 (72.1%) patients with ACLF (P<0.001). Yet, TSH and TSH-based indexes (TSH/FT3-ratio, thyroid index) showed an U-shaped evolution during progression of cirrhosis, suggesting a partially preserved responsiveness of the hypothalamus and pituitary in AD. Infections were associated with lower FT3 concentrations in AD, but not in ACLF. Low FT3 concentrations correlated significantly with 90-day mortality. Both, AD/ACLF and NTIS, were associated with signatures of inflammatory mediators, which were partially non-overlapping. Conclusion: NTIS is frequent already in AD and therefore precedes critically illness in a subgroup of patients with decompensated cirrhosis. This might constitute a new paradigm of TH signaling in cirrhosis, offering opportunities to explore preventive effects of TH in AD.


Subject(s)
Acute-On-Chronic Liver Failure , Euthyroid Sick Syndromes , Humans , Acute-On-Chronic Liver Failure/complications , Euthyroid Sick Syndromes/complications , Euthyroid Sick Syndromes/epidemiology , Prospective Studies , Liver Cirrhosis/complications , Critical Illness , Thyrotropin
7.
Cell Rep ; 42(2): 112088, 2023 02 28.
Article in English | MEDLINE | ID: mdl-36753417

ABSTRACT

The hypothalamic pituitary thyroid axis is a major regulator of many differentiation processes, including adipose tissue. However, it remains unclear whether and how thyroid hormone (TH) signaling contributes to preadipocyte commitment and differentiation into mature adipocytes. Here, we show a cell-autonomous effect of TH on the transcriptional regulation of zinc finger protein 423 (Zfp423), an early adipogenic determination factor, in murine adipose depots. Mechanistically, binding of the unliganded TH receptor to a negative TH responsive element within the Zfp423 promoter activates transcriptional activity that is reversed upon TH binding. Zfp423 upregulation is associated with increased GFP+ preadipocyte recruitment in stromal vascular fraction isolated from white fat of hypothyroid Zfp423GFP reporter mice. RNA sequencing identified Zfp423-driven gene programs that are modulated in response to TH during adipogenic differentiation. Collectively, we identified Zfp423 as a key molecule that integrates TH signaling into the regulation of adipose tissue plasticity.


Subject(s)
Adipocytes , DNA-Binding Proteins , Animals , Mice , Adipocytes/metabolism , Adipogenesis/physiology , Adipose Tissue/metabolism , Cell Differentiation/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Obesity/metabolism , Thyroid Hormones/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
8.
Eur J Endocrinol ; 186(5): R65-R77, 2022 Mar 23.
Article in English | MEDLINE | ID: mdl-35175936

ABSTRACT

Over the past few years, growing evidence suggests direct crosstalk between thyroid hormones (THs) and the immune system. Components of the immune system were proposed to interfere with the central regulation of systemic TH levels. Conversely, THs regulate innate and adaptive immune responses as immune cells are direct target cells of THs. Accordingly, they express different components of local TH action, such as TH transporters or receptors, but our picture of the interplay between THs and the immune system is still incomplete. This review provides a critical overview of current knowledge regarding the interaction of THs and the immune system with the main focus on local TH action within major innate and adaptive immune cell subsets. Thereby, this review aims to highlight open issues which might help to infer the clinical relevance of THs in host defence in the context of different types of diseases such as infection, ischemic organ injury or cancer.


Subject(s)
Carrier Proteins , Thyroid Hormones , Humans , Immune System/metabolism , Thyroid Hormones/metabolism
9.
Thyroid ; 31(2): 327-329, 2021 02.
Article in English | MEDLINE | ID: mdl-32546069

ABSTRACT

Background: Multiple endocrine neoplasia type 2 (MEN2) is an autosomal dominant disorder caused by mutations in the RET proto-oncogene. MEN2 is classified into two subtypes, MEN 2A and 2B. MEN2B is characterized by early-onset and aggressive medullary thyroid carcinoma (MTC), pheochromocytoma, and characteristic physical features. Patient Findings: We present a 39-year-old male with early-onset metastatic MTC diagnosed at the age of 13 years and physical features typical for MEN2B such as marfanoid habitus, mucosal neuromas, and thickened eyelids. The patient has two first-degree relatives (mother and maternal uncle) with MTC and pheochromocytoma. The mother has similar facial features. RET sequencing revealed a novel tandem RET E768D/L790F germline mutation in exon 13. The patient's mother has the same RET variant. For functional in vitro characterization, wild-type RET, RET E768D, RET L790F, the double RET E768D/L790F mutant, and RET M918T were expressed in HEK293 cells. The novel double RET E768D/L790F mutant increased ligand-independent RET phosphorylation, activation of the mitogen-activated protein kinase (MAPK)-pathway, and colony formation similar to the classical MEN2B RET M918T mutation. Summary: In this male patient with a MEN2B-like phenotype, we identified a novel double RET germline mutation, E768D/L790F. Functional characterization of the double mutant shows similar transforming capacity as RET M918T.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma, Neuroendocrine/genetics , Multiple Endocrine Neoplasia Type 2b/genetics , Mutation , Proto-Oncogene Proteins c-ret/genetics , Thyroid Neoplasms/genetics , Adult , Biomarkers, Tumor/metabolism , Carcinoma, Neuroendocrine/diagnosis , Carcinoma, Neuroendocrine/metabolism , Female , Genetic Predisposition to Disease , HEK293 Cells , Heredity , Humans , MAP Kinase Signaling System , Male , Middle Aged , Multiple Endocrine Neoplasia Type 2b/diagnosis , Multiple Endocrine Neoplasia Type 2b/metabolism , Pedigree , Phenotype , Proto-Oncogene Mas , Proto-Oncogene Proteins c-ret/metabolism , Thyroid Neoplasms/diagnosis , Thyroid Neoplasms/metabolism
10.
Endocrinology ; 162(7)2021 07 01.
Article in English | MEDLINE | ID: mdl-33999131

ABSTRACT

CONTEXT: Hypothyroidism impairs cardiovascular health and contributes to endothelial dysfunction with reduced vasodilation. How 3,5,3'-triiodothyronine (T3) and its receptors are involved in the regulation of vasomotion is not yet fully understood. In general, thyroid hormone receptors (TRs) either influence gene expression (canonical action) or rapidly activate intracellular signaling pathways (noncanonical action). OBJECTIVE: Here we aimed to characterize the T3 action underlying the mechanism of arterial vasodilation and blood pressure (BP) regulation. METHODS: Mesenteric arteries were isolated from male rats, wild-type (WT) mice, TRα knockout (TRα 0) mice, and from knockin mice with a mutation in the DNA-binding domain (TRα GS). In this mutant, DNA binding and thus canonical action is abrogated while noncanonical signaling is preserved. In a wire myograph system, the isolated vessels were preconstricted with norepinephrine. The response to T3 was measured, and the resulting vasodilation (Δ force [mN]) was normalized to maximum contraction with norepinephrine and expressed as percentage vasodilation after maximal preconstriction with norepinephrine (%NE). Isolated vessels were treated with T3 (1 × 10-15 to 1 × 10-5 mol/L) alone and in combination with the endothelial nitric oxide-synthase (eNOS) inhibitor L-NG-nitroarginine methyl ester (L-NAME) or the phosphatidylinositol 3-kinase (PI3K) inhibitor wortmannin. The endothelium was removed to determine the contribution of T3 to endothelium-dependent vasodilation. The physiological relevance of T3-induced vasodilation was determined by in vivo arterial BP measurements in male and female mice. RESULTS: T3 treatment induced vasodilation of mesenteric arteries from WT mice within 2 minutes (by 21.5 ±â€…1.7%NE). This effect was absent in arteries from TRα 0 mice (by 5.3 ±â€…0.6%NE, P < .001 vs WT) but preserved in TRα GS arteries (by 17.2 ±â€…1.1%NE, not significant vs WT). Inhibition of either eNOS or PI3K reduced T3-mediated vasodilation from 52.7 ±â€…4.5%NE to 28.5 ±â€…4.1%NE and 22.7 ±â€…2.9%NE, respectively. Removal of the endothelium abolished the T3-mediated vasodilation in rat mesenteric arteries (by 36.7 ±â€…5.4%NE vs 3.5 ±â€…6.2%NE). In vivo, T3 injection led to a rapid decrease of arterial BP in WT (by 13.9 ±â€…1.9 mm Hg) and TRα GS mice (by 12.4 ±â€…1.9 mm Hg), but not in TRα 0 mice (by 4.1 ±â€…1.9 mm Hg). CONCLUSION: These results demonstrate that T3 acting through noncanonical TRα action affects cardiovascular physiology by inducing endothelium-dependent vasodilation within minutes via PI3K and eNOS activation.


Subject(s)
Mesenteric Arteries/physiology , Thyroid Hormone Receptors alpha/physiology , Vasodilation/physiology , Animals , Binding Sites/genetics , Blood Pressure/drug effects , Blood Pressure/physiology , DNA/metabolism , Female , Gene Knock-In Techniques , Male , Mice , Mice, Knockout , Mutation , Nitric Oxide Synthase Type III/physiology , Norepinephrine/pharmacology , Phosphatidylinositol 3-Kinases/physiology , Rats , Signal Transduction/physiology , Thyroid Hormone Receptors alpha/chemistry , Thyroid Hormone Receptors alpha/genetics , Triiodothyronine/pharmacology , Vasodilation/drug effects
11.
Eur Thyroid J ; 10(1): 10-38, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33777817

ABSTRACT

BACKGROUND: Fourteen clinical trials have not shown a consistent benefit of combination therapy with levothyroxine (LT4) and liothyronine (LT3). Despite the publication of these trials, combination therapy is widely used and patients reporting benefit continue to generate patient and physician interest in this area. Recent scientific developments may provide insight into this inconsistency and guide future studies. METHODS: The American Thyroid Association (ATA), British Thyroid Association (BTA), and European Thyroid Association (ETA) held a joint conference on November 3, 2019 (live-streamed between Chicago and London) to review new basic science and clinical evidence regarding combination therapy with presentations and input from 12 content experts. After the presentations, the material was synthesized and used to develop Summary Statements of the current state of knowledge. After review and revision of the material and Summary Statements, there was agreement that there was equipoise for a new clinical trial of combination therapy. Consensus Statements encapsulating the implications of the material discussed with respect to the design of future clinical trials of LT4/LT3 combination therapy were generated. Authors voted upon the Consensus Statements. Iterative changes were made in several rounds of voting and after comments from ATA/BTA/ETA members. RESULTS: Of 34 Consensus Statements available for voting, 28 received at least 75% agreement, with 13 receiving 100% agreement. Those with 100% agreement included studies being powered to study the effect of deiodinase and thyroid hormone transporter polymorphisms on study outcomes, inclusion of patients dissatisfied with their current therapy and requiring at least 1.2 µg/kg of LT4 daily, use of twice daily LT3 or preferably a slow-release preparation if available, use of patient-reported outcomes as a primary outcome (measured by a tool with both relevant content validity and responsiveness) and patient preference as a secondary outcome, and utilization of a randomized placebo-controlled adequately powered double-blinded parallel design. The remaining statements are presented as potential additional considerations. DISCUSSION: This article summarizes the areas discussed and presents Consensus Statements to guide development of future clinical trials of LT4/LT3 combination therapy. The results of such redesigned trials are expected to be of benefit to patients and of value to inform future thyroid hormone replacement clinical practice guidelines treatment recommendations.

12.
Thyroid ; 31(2): 156-182, 2021 02.
Article in English | MEDLINE | ID: mdl-33276704

ABSTRACT

Background: Fourteen clinical trials have not shown a consistent benefit of combination therapy with levothyroxine (LT4) and liothyronine (LT3). Despite the publication of these trials, combination therapy is widely used and patients reporting benefit continue to generate patient and physician interest in this area. Recent scientific developments may provide insight into this inconsistency and guide future studies. Methods: The American Thyroid Association (ATA), British Thyroid Association (BTA), and European Thyroid Association (ETA) held a joint conference on November 3, 2019 (live-streamed between Chicago and London) to review new basic science and clinical evidence regarding combination therapy with presentations and input from 12 content experts. After the presentations, the material was synthesized and used to develop Summary Statements of the current state of knowledge. After review and revision of the material and Summary Statements, there was agreement that there was equipoise for a new clinical trial of combination therapy. Consensus Statements encapsulating the implications of the material discussed with respect to the design of future clinical trials of LT4/LT3 combination therapy were generated. Authors voted upon the Consensus Statements. Iterative changes were made in several rounds of voting and after comments from ATA/BTA/ETA members. Results: Of 34 Consensus Statements available for voting, 28 received at least 75% agreement, with 13 receiving 100% agreement. Those with 100% agreement included studies being powered to study the effect of deiodinase and thyroid hormone transporter polymorphisms on study outcomes, inclusion of patients dissatisfied with their current therapy and requiring at least 1.2 µg/kg of LT4 daily, use of twice daily LT3 or preferably a slow-release preparation if available, use of patient-reported outcomes as a primary outcome (measured by a tool with both relevant content validity and responsiveness) and patient preference as a secondary outcome, and utilization of a randomized placebo-controlled adequately powered double-blinded parallel design. The remaining statements are presented as potential additional considerations. Discussion: This article summarizes the areas discussed and presents Consensus Statements to guide development of future clinical trials of LT4/LT3 combination therapy. The results of such redesigned trials are expected to be of benefit to patients and of value to inform future thyroid hormone replacement clinical practice guidelines treatment recommendations.


Subject(s)
Hypothyroidism/drug therapy , Thyroxine/therapeutic use , Triiodothyronine/therapeutic use , Consensus , Drug Combinations , Evidence-Based Medicine , Humans , Hypothyroidism/blood , Hypothyroidism/diagnosis , Thyroxine/adverse effects , Treatment Outcome , Triiodothyronine/adverse effects
13.
Exp Clin Endocrinol Diabetes ; 128(6-07): 383-387, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32330966

ABSTRACT

Thyroid hormone (TH) is essential for the regulation of many physiological processes, especially growth, organ development, energy metabolism and cardiovascular effects. TH acts via the TH receptors (TR) α and ß. By binding to thyroid hormone responsive elements (TREs) on the DNA, TRs regulate expression of TH target genes. Thus, TRs are mainly characterized as ligand dependent transcription factors and regulation of gene expression and protein synthesis is considered the canonical mode of TH/TR action. The demonstration that the ligand-bound TRs α and ß also mediate activation of the phosphatidylinositol-3-kinase (PI3K) pathway established noncanonical TH/TR action as an additional mode of TH signaling. Recently, TR mutant mouse models allowed to determine the underlying mode of TH/TR action, either canonical or noncanonical TH/TR signaling, for several physiological TH effects in vivo: Regulation of the hypothalamic-pituitary-thyroid axis requires DNA-binding of TRß, whereas hepatic triglyceride content appears to be regulated by noncanonical TRß signaling. TRα mediated effects in bone development are dependent on DNA-binding, whereas several cardiovascular TRα effects are rapid and independent from DNA-binding. Therefore, noncanonical TH/TR action contributes to the overall effects of TH in physiology.


Subject(s)
Phosphatidylinositol 3-Kinase/metabolism , Signal Transduction/physiology , Thyroid Hormone Receptors alpha/metabolism , Thyroid Hormone Receptors beta/metabolism , Thyroid Hormones/metabolism , Animals , Humans
14.
Eur J Haematol ; 83(4): 320-7, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19500131

ABSTRACT

The objective of this study was to confirm previous results regarding the differential expression and prognostic significance of the circadian gene CRY1 in chronic lymphocytic leukemia (CLL) patients and its relationship with the expression of other circadian genes and well-established prognostic markers. We also aimed to investigate whether the peripheral circadian machinery may be deregulated in CLL cells. The expression of CRY1, PER1, and PER2 was determined by real-time reverse transcriptase polymerase chain reaction (RT-PCR) in 116 CLL patients. The expression at sequential time points over a 24-h period was measured in six CLL patients and six normal donors. We confirmed the differential expression of CRY1 in ZAP-70(+)/CD38(+) and ZAP-70(-)/CD38(-) CLL samples. Subgroups formed according to CRY1 expression levels differed significantly in time to treatment. This difference was even more pronounced for subgroups stratified by a CRY1 : PER2 expression ratio and the ratio was an independent prognostic marker in a multivariate model. Furthermore, our data indicate disturbances in the periodic expression of circadian genes in CLL cells. Because of their role in the expression of cell cycle-related and DNA-damage response genes, we suggest that the deregulated expression of circadian genes may be linked to the molecular pathogenesis of CLL.


Subject(s)
Flavoproteins/genetics , Gene Expression Regulation, Neoplastic , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Nuclear Proteins/genetics , Predictive Value of Tests , Transcription Factors/genetics , Adult , Aged , Aged, 80 and over , Case-Control Studies , Chronobiology Disorders/genetics , Circadian Rhythm/genetics , Cryptochromes , DNA Repair/genetics , Female , Humans , Kinetics , Male , Middle Aged , Period Circadian Proteins , Prognosis , Treatment Outcome
15.
Thyroid ; 29(10): 1438-1446, 2019 10.
Article in English | MEDLINE | ID: mdl-31526103

ABSTRACT

Background: Radioiodine refractory dedifferentiated thyroid cancer is a major clinical challenge. Anaplastic lymphoma kinase (ALK) mutations with increased ALK activity, especially fusion genes, have been suggested to promote thyroid carcinogenesis, leading to development of poorly differentiated thyroid carcinoma (PDTC) and anaplastic thyroid carcinoma. To determine the oncogenic potential of increased ALK activity in thyroid carcinogenesis in vivo, we studied mice with thyrocyte-specific expression of a constitutively active ALK mutant. Methods: Mice carrying a Cre-activated allele of a constitutively active ALK mutant (F1174L) were crossed with mice expressing tamoxifen-inducible Cre recombinase (CreERT2) under the control of the thyroglobulin (Tg) gene promoter to achieve thyrocyte-specific expression of the ALK mutant (ALKF1174L mice). Survival, thyroid hormone serum concentration, and tumor development were recorded. Thyroids and lungs were studied histologically. To maintain euthyroidism despite dedifferentiation of the thyroid, a cohort was substituted with levothyroxine (LT4) through drinking water. Results: ALKF1174L mice developed massively enlarged thyroids, which showed an early loss of normal follicular architecture 12 weeks after tamoxifen injection. A significant decrease in Tg and Nkx-2.1 expression as well as impaired thyroid hormone synthesis confirmed dedifferentiation. Histologically, the mice developed a carcinoma resembling human PDTC with a predominantly trabecular/solid growth pattern and an increased mitotic rate. The tumors showed extrathyroidal extension into the surrounding strap muscles and developed lung metastases. Median survival of ALKF1174L mice was significantly reduced to five months after tamoxifen injection. Reduced Tg expression and loss of follicular structure led to hypothyroidism with elevated thyrotropin (TSH). To test whether TSH stimulation played a role in thyroid carcinogenesis, we kept ALKF1174L mice euthyroid by LT4 substitution. These mice developed PDTC with identical histological features compared with hypothyroid mice, demonstrating that PDTC development was due to increased ALK activity and not dependent on TSH stimulation. Conclusion: Expression of a constitutively activated ALK mutant in thyroids of mice leads to development of metastasizing thyroid cancer resembling human PDTC. These results demonstrate in vivo that increased ALK activity is a driver mechanism in thyroid carcinogenesis.


Subject(s)
Anaplastic Lymphoma Kinase/genetics , Carcinoma/genetics , Cell Dedifferentiation/genetics , Hypothyroidism/metabolism , Lung Neoplasms/secondary , Thyroid Neoplasms/genetics , Animals , Carcinoma/pathology , Carcinoma/secondary , Hypothyroidism/etiology , Mice , Neoplasm Invasiveness , Thyroglobulin/metabolism , Thyroid Neoplasms/pathology , Thyroid Nuclear Factor 1/metabolism , Thyrotropin/metabolism
17.
Sci Rep ; 8(1): 13096, 2018 08 30.
Article in English | MEDLINE | ID: mdl-30166557

ABSTRACT

Graves' orbitopathy (GO) is the most common extra thyroidal complication of Graves' disease (GD) and occurs predominantly in women but more severe in men. The reason for this effect of gender on GO is unknown. Herein we studied the manifestation of GO in both sexes of an induced mouse model in absence of additional risk factors present in patients like advanced age, genetic variabilities or smoking. Male and female mice were immunized with human TSHR A-subunit encoding plasmid. Both sexes comparably developed autoimmune hyperthyroidism characterized by TSHR stimulating autoantibodies, elevated T4 values, hyperplastic thyroids and hearts. Autoimmune mice developed inflammatory eye symptoms and proptosis, although males earlier than females. Serial in vivo 1H/19F-magnetic resonance imaging revealed elevated inflammatory infiltration, increased fat volume and glycosaminoglycan deposition in orbits of both sexes but most significantly in female mice. Histologically, infiltration of T-cells, extension of brown fat and overall collagen deposition were characteristics of GO in male mice. In contrast, female mice developed predominately macrophage infiltration in muscle and connective tissue, and muscle hypertrophy. Apart from sex-dependent variabilities in pathogenesis, disease classification revealed minor sex-differences in incidence and total outcome. In conclusion, sex does not predispose for autoimmune hyperthyroidism and associated GO.


Subject(s)
Graves Disease/complications , Graves Ophthalmopathy/complications , Orbit/pathology , Sex Characteristics , Animals , Disease Models, Animal , Female , Graves Disease/physiopathology , Graves Ophthalmopathy/physiopathology , Magnetic Resonance Imaging , Male , Mice, Inbred BALB C , Receptors, Thyrotropin/metabolism , Thyroid Function Tests , Thyroid Gland/pathology , Thyroid Gland/physiopathology
18.
J Clin Endocrinol Metab ; 103(10): 3845-3855, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30085133

ABSTRACT

Context: Insulin autoimmune syndrome (IAS), spontaneous hyperinsulinemic hypoglycemia due to insulin-binding autoantibodies, may be difficult to distinguish from tumoral or other forms of hyperinsulinemic hypoglycemia, including surreptitious insulin administration. No standardized treatment regimen exists. Objectives: To evaluate an analytic approach to IAS and responses to different treatments. Design and Setting: Observational study in the UK Severe Insulin Resistance Service. Patients: Six patients with hyperinsulinemic hypoglycemia and detectable circulating anti-insulin antibody (IA). Main Outcome Measures: Glycemia, plasma insulin, and C-peptide concentrations by immunoassay or mass spectrometry (MS). Immunoreactive insulin was determined in the context of polyethylene glycol (PEG) precipitation and gel filtration chromatography (GFC). IA quantification using ELISA and RIA, and IA were further characterized using radioligand binding studies. Results: All patients were diagnosed with IAS (five IgG, one IgA) based on a high insulin/C-peptide ratio, low insulin recovery after PEG precipitation, and GFC evidence of antibody-bound insulin. Neither ELISA nor RIA result proved diagnostic for every case. MS provided a more robust quantification of insulin in the context of IA. One patient was managed conservatively, four were treated with diazoxide without sustained benefit, and four were treated with immunosuppression with highly variable responses. IA affinity did not appear to influence presentation or prognosis. Conclusions: IAS should be considered in patients with hyperinsulinemic hypoglycemia and a high insulin/C-peptide ratio. Low insulin recovery on PEG precipitation supports the presence of insulin-binding antibodies, with GFC providing definitive confirmation. Immunomodulatory therapy should be customized according to individual needs and clinical response.


Subject(s)
Autoimmune Diseases/diagnosis , Congenital Hyperinsulinism/diagnosis , Insulin Antibodies/blood , Adult , Aged , Autoimmune Diseases/drug therapy , Autoimmune Diseases/immunology , Biomarkers/blood , Blood Glucose/metabolism , C-Peptide/blood , Chromatography, Gel , Congenital Hyperinsulinism/drug therapy , Congenital Hyperinsulinism/immunology , Diazoxide/therapeutic use , Female , Humans , Immunosuppressive Agents/therapeutic use , Insulin/blood , Insulin Resistance/physiology , Male , Middle Aged , Syndrome
19.
Lancet Diabetes Endocrinol ; 6(4): 287-298, 2018 04.
Article in English | MEDLINE | ID: mdl-29396246

ABSTRACT

BACKGROUND: European guidelines recommend intravenous methylprednisolone as first-line treatment for active and severe Graves' orbitopathy; however, it is common for patients to have no response or have relapse after discontinuation of treatment. We aimed to compare the efficacy and safety of add-on mycophenolate to methylprednisolone in comparison with methylprednisolone alone in patients with moderate-to-severe Graves' orbitopathy. METHODS: MINGO was an observer-masked, multicentre, block-randomised, centre-stratified trial done in two centres in Germany and two in Italy. Patients with active moderate-to-severe Graves' orbitopathy were randomly assigned to receive intravenous methylprednisolone (500 mg once per week for 6 weeks followed by 250 mg per week for 6 weeks) either alone or with mycophenolate (one 360 mg tablet twice per day for 24 weeks). The prespecified primary endpoints were rate of response (reduction of at least two parameters of a composite ophthalmic index [eyelid swelling, clinical activity score, proptosis, lid width, diplopia, and eye muscle motility] without deterioration in any other parameter) at 12 weeks and rate of relapse (a worsening of symptoms that occurred after a response) at 24 and 36 weeks. Rates of response at week 24 and sustained response at week 36 were added as post-hoc outcomes. Prespecified primary outcomes and post-hoc outcomes were assessed in the modified intention-to-treat population (defined as all patients assigned to treatment who received at least one infusion of methylprednisolone, when outcome data were available), and safety was assessed in all patients who received at least one dose of study drug. This trial is registered with the EU Clinical Trials Register, EUDRACT number 2008-002123-93. FINDINGS: 164 patients were enrolled and randomised between Nov 29, 2009, and July 31, 2015. 81 were randomly assigned to receive methylprednisolone alone and 83 to receive methylprednisolone with mycophenolate. In the intention-to-treat population at 12 weeks, responses were observed in 36 (49%) of 73 patients in the monotherapy group and 48 (63%) of 76 patients in the combination group, giving an odds ratio (OR) of 1·76 (95% CI 0·92-3·39, p=0·089). At week 24, 38 (53%) of 72 patients remaining in the monotherapy group and 53 (71%) of 75 patients remaining in the combination therapy group had responded to treatment (2·16, 1·09-4·25, p=0·026). At week 24, relapse occurred in four (11%) of 38 patients in the monotherapy group and four (8%) of 53 patients in the combination group (OR 0·71, 0·17-3·03, p=0·72). At week 36, relapse occurred in an additional three (8%) patients in the monotherapy group and two (4%) patients in the combination group (0·65, 0·12-3·44, p=0·61). At week 36, 31 (46%) of 68 patients in the monotherapy group and 49 (67%) of 73 patients in the combination group had a sustained response (OR 2·44, 1·23-4·82, p=0·011). 23 patients had 24 serious adverse events, with 11 events in ten patients in the combination group and 13 events in 13 patients in the monotherapy group. Mild and moderate (grade 1-2) drug-related adverse events occurred in 16 (20%) of 81 patients receiving monotherapy and 21 (25%) of 83 patients receiving combination therapy (p=0·48). INTERPRETATION: Although no significant difference was seen in the rate of response at 12 weeks or rate of relapse at 24 and 36 weeks, post-hoc analysis suggested that addition of mycophenolate to treatment with methylprednisolone improved rate of response to therapy by 24 weeks in patients with active and moderate-to-severe Graves' orbitopathy. FUNDING: Novartis, Germany.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Antibiotics, Antineoplastic/therapeutic use , Graves Ophthalmopathy/drug therapy , Methylprednisolone/therapeutic use , Mycophenolic Acid/therapeutic use , Severity of Illness Index , Adolescent , Adult , Aged , Double-Blind Method , Drug Therapy, Combination , Female , Humans , Italy , Male , Middle Aged , Treatment Outcome , Young Adult
20.
Endocrinology ; 148(10): 4727-33, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17640981

ABSTRACT

The genetic basis for differences in TSH sensitivity between two rat strains was examined using consomic rats generated from original strains salt-sensitive Dahl (SS) (TSH 1.8 +/- 0.1 ng/ml; free T(4) index 4.9 +/- 0.4) and Brown Norwegian (BN) (TSH 5.5 +/- 0.6 ng/ml, P < 0.05; free T(4) index 4.3 +/- 0.1, P not significant). Consomic rats SSBN6 [BN chromosome (CH) 6 placed in SS rat] and SSBN2 (BN CH 2 placed in SS rat) have TSH concentrations intermediate between pure SS and BN strains (2.9 +/- 0.3 and 3.1 +/- 0.3 ng/ml, respectively; P < 0.05). Candidate genes on rat CH 2 included TSH beta-subunit and on CH 6 the TSH receptor (TSHR). TSH from sera of BN, SS, SSBN6, and SSBN2 strains had similar in vitro bioactivity suggesting that the cause for the variable TSH concentrations was not due to an altered TSH. Physiological response to TSH was measured by changes in serum T(4) concentrations upon administration of bovine TSH (bTSH). Rat strain SS had a greater T(4) response to bTSH than BN (change in T(4), 1.3 +/- 0.1 vs. 0.4 +/- 0.1 microg/dl, P < 0.005), suggesting reduced thyrocyte sensitivity to TSH in BN. Sequencing of the TSHR coding region revealed an amino acid difference in BN (Q46R). This substitution is unlikely to contribute to the strain difference in serum TSH because both TSHR variants were equally expressed at the cell surface of transfected cells and responsive to bTSH. Given similar TSH activity and similar TSHR structure, TSHR mRNA expression in thyroid tissue was quantitated by real-time PCR. BN had 54 +/- 5% the total TSHR expression compared to SS (100 +/- 7%, P < 0.0001), when corrected for GAPDH expression, a difference confirmed at the protein level. Therefore, the higher TSH level in the BN strain appears to reflect an adjustment of the feedback loop to reduced thyrocyte sensitivity to TSH secondary to reduced TSHR expression. These strains of rat provide a model to study the cis- and trans-acting factors underlying the difference in TSHR expression.


Subject(s)
Feedback, Physiological/physiology , Pituitary Gland/metabolism , Receptors, Thyrotropin/metabolism , Thyroid Gland/metabolism , Amino Acid Sequence , Animals , Animals, Genetically Modified , Cattle , Chromosome Mapping , Chromosomes/physiology , Introns , Osmolar Concentration , Promoter Regions, Genetic/physiology , RNA, Messenger/metabolism , Rats , Rats, Inbred BN , Rats, Inbred Dahl , Receptors, Thyrotropin/genetics , Thyroid Function Tests , Thyrotropin/genetics , Thyrotropin/pharmacology , Thyrotropin/physiology , Thyrotropin, beta Subunit/genetics , Thyroxine/blood
SELECTION OF CITATIONS
SEARCH DETAIL