Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Eur Thyroid J ; 13(2)2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38417253

ABSTRACT

Thyroid hormones play an important role during the development and functioning of the different sensory systems. In order to exert their actions, thyroid hormones need to access their target cells through transmembrane transporter proteins, among which the monocarboxylate transporter 8 (MCT8) stands out for its pathophysiological relevance. Mutations in the gene encoding for MCT8 lead to the Allan-Herndon-Dudley syndrome (AHDS), a rare disease characterised by severe neuromotor and cognitive impairments. The impact of MCT8 deficiency in the neurosensory capacity of AHDS patients is less clear, with only a few patients displaying visual and auditory impairments. In this review we aim to gather data from different animal models regarding thyroid hormone transport and action in the different neurosensory systems that could aid to identify potential neurosensorial alterations in MCT8-deficient patients.


Subject(s)
Mental Retardation, X-Linked , Muscular Atrophy , Thyroid Hormones , Animals , Humans , Thyroid Hormones/metabolism , Mental Retardation, X-Linked/genetics , Biological Transport , Muscle Hypotonia/genetics , Monocarboxylic Acid Transporters/genetics
2.
Front Endocrinol (Lausanne) ; 14: 1235614, 2023.
Article in English | MEDLINE | ID: mdl-38107517

ABSTRACT

Introduction: Pluripotent stem cells can be generated from somatic cells by the Yamanaka factors Oct4, Sox2, Klf4 and c-Myc. Methods: Mouse embryonic fibroblasts (MEFs) were transduced with the Yamanaka factors and generation of induced pluripotent stem cells (iPSCs) was assessed by formation of alkaline phosphatase positive colonies, pluripotency gene expression and embryod bodies formation. Results: The thyroid hormone triiodothyronine (T3) enhances MEFs reprogramming. T3-induced iPSCs resemble embryonic stem cells in terms of the expression profile and DNA methylation pattern of pluripotency genes, and of their potential for embryod body formation and differentiation into the three major germ layers. T3 induces reprogramming even though it increases expression of the cyclin kinase inhibitors p21 and p27, which are known to oppose acquisition of pluripotency. The actions of T3 on reprogramming are mainly mediated by the thyroid hormone receptor beta and T3 can enhance iPSC generation in the absence of c-Myc. The hormone cannot replace Oct4 on reprogramming, but in the presence of T3 is possible to obtain iPSCs, although with low efficiency, without exogenous Klf4. Furthermore, depletion of the corepressor NCoR (or Nuclear Receptor Corepressor 1) reduces MEFs reprogramming in the absence of the hormone and strongly decreases iPSC generation by T3 and also by 9cis-retinoic acid, a well-known inducer of reprogramming. NCoR depletion also markedly antagonizes induction of pluripotency gene expression by both ligands. Conclusions: Inclusion of T3 on reprogramming strategies has a potential use in enhancing the generation of functional iPSCs for studies of cell plasticity, disease and regenerative medicine.


Subject(s)
Cellular Reprogramming , Nuclear Receptor Co-Repressor 1 , Pluripotent Stem Cells , Animals , Mice , Co-Repressor Proteins/genetics , Fibroblasts/metabolism , Hormones/metabolism , Pluripotent Stem Cells/metabolism , Thyroid Hormones/metabolism , Nuclear Receptor Co-Repressor 1/genetics
3.
Fluids Barriers CNS ; 20(1): 79, 2023 Nov 03.
Article in English | MEDLINE | ID: mdl-37924081

ABSTRACT

BACKGROUND: The monocarboxylate transporter 8 (MCT8) plays a vital role in maintaining brain thyroid hormone homeostasis. This transmembrane transporter is expressed at the brain barriers, as the blood-brain barrier (BBB), and in neural cells, being the sole known thyroid hormone-specific transporter to date. Inactivating mutations in the MCT8 gene (SLC16A2) cause the Allan-Herndon-Dudley Syndrome (AHDS) or MCT8 deficiency, a rare X-linked disease characterized by delayed neurodevelopment and severe psychomotor disorders. The underlying pathophysiological mechanisms of AHDS remain unclear, and no effective treatments are available for the neurological symptoms of the disease. METHODS: Neurovascular unit ultrastructure was studied by means of transmission electron microscopy. BBB permeability and integrity were evaluated by immunohistochemistry, non-permeable dye infiltration assays and histological staining techniques. Brain blood-vessel density was evaluated by immunofluorescence and magnetic resonance angiography. Finally, angiogenic-related factors expression was evaluated by qRT-PCR. The studies were carried out both in an MCT8 deficient subject and Mct8/Dio2KO mice, an AHDS murine model, and their respective controls. RESULTS: Ultrastructural analysis of the BBB of Mct8/Dio2KO mice revealed significant alterations in neurovascular unit integrity and increased transcytotic flux. We also found functional alterations in the BBB permeability, as shown by an increased presence of peripheral IgG, Sodium Fluorescein and Evans Blue, along with increased brain microhemorrhages. We also observed alterations in the angiogenic process, with reduced blood vessel density in adult mice brain and altered expression of angiogenesis-related factors during brain development. Similarly, AHDS human brain samples showed increased BBB permeability to IgG and decreased blood vessel density. CONCLUSIONS: These findings identify for the first time neurovascular alterations in the MCT8-deficient brain, including a disruption of the integrity of the BBB and alterations in the neurovascular unit ultrastructure as a new pathophysiological mechanism for AHDS. These results open a new field for potential therapeutic targets for the neurological symptoms of these patients and unveils magnetic resonance angiography as a new non-invasive in vivo technique for evaluating the progression of the disease.


Subject(s)
Mental Retardation, X-Linked , Symporters , Animals , Humans , Mice , Blood-Brain Barrier/metabolism , Immunoglobulin G , Mental Retardation, X-Linked/diagnosis , Mental Retardation, X-Linked/genetics , Mental Retardation, X-Linked/pathology , Monocarboxylic Acid Transporters/genetics , Monocarboxylic Acid Transporters/metabolism , Muscle Hypotonia/diagnosis , Muscle Hypotonia/genetics , Muscle Hypotonia/metabolism , Muscular Atrophy/diagnosis , Muscular Atrophy/genetics , Muscular Atrophy/metabolism , Symporters/genetics , Symporters/metabolism , Symporters/therapeutic use , Thyroid Hormones/metabolism , Thyroid Hormones/therapeutic use
4.
Int J Mol Sci ; 24(11)2023 Jun 01.
Article in English | MEDLINE | ID: mdl-37298594

ABSTRACT

Monocarboxylate transporter 8 (MCT8) and organic anion-transporting polypeptide 1C1 (OATP1C1) are thyroid hormone (TH) transmembrane transporters relevant for the availability of TH in neural cells, crucial for their proper development and function. Mutations in MCT8 or OATP1C1 result in severe disorders with dramatic movement disability related to alterations in basal ganglia motor circuits. Mapping the expression of MCT8/OATP1C1 in those circuits is necessary to explain their involvement in motor control. We studied the distribution of both transporters in the neuronal subpopulations that configure the direct and indirect basal ganglia motor circuits using immunohistochemistry and double/multiple labeling immunofluorescence for TH transporters and neuronal biomarkers. We found their expression in the medium-sized spiny neurons of the striatum (the receptor neurons of the corticostriatal pathway) and in various types of its local microcircuitry interneurons, including the cholinergic. We also demonstrate the presence of both transporters in projection neurons of intrinsic and output nuclei of the basal ganglia, motor thalamus and nucleus basalis of Meynert, suggesting an important role of MCT8/OATP1C1 for modulating the motor system. Our findings suggest that a lack of function of these transporters in the basal ganglia circuits would significantly impact motor system modulation, leading to clinically severe movement impairment.


Subject(s)
Basal Ganglia , Organic Anion Transporters , Symporters , Adult , Humans , Basal Ganglia/metabolism , Brain/metabolism , Interneurons/metabolism , Monocarboxylic Acid Transporters/genetics , Monocarboxylic Acid Transporters/metabolism , Neurons/metabolism , Organic Anion Transporters/metabolism , Symporters/genetics , Symporters/metabolism , Thalamus/metabolism , Thyroid Hormones/metabolism
5.
Int J Mol Sci ; 24(4)2023 Feb 06.
Article in English | MEDLINE | ID: mdl-36834621

ABSTRACT

Monocarboxylate transporter 8 (MCT8) and organic anion transporter polypeptide 1C1 (OATP1C1) are thyroid hormone (TH) transmembrane transporters that play an important role in the availability of TH for neural cells, allowing their proper development and function. It is important to define which cortical cellular subpopulations express those transporters to explain why MCT8 and OATP1C1 deficiency in humans leads to dramatic alterations in the motor system. By means of immunohistochemistry and double/multiple labeling immunofluorescence in adult human and monkey motor cortices, we demonstrate the presence of both transporters in long-projection pyramidal neurons and in several types of short-projection GABAergic interneurons in both species, suggesting a critical position of these transporters for modulating the efferent motor system. MCT8 is present at the neurovascular unit, but OATP1C1 is only present in some of the large vessels. Both transporters are expressed in astrocytes. OATP1C1 was unexpectedly found, only in the human motor cortex, inside the Corpora amylacea complexes, aggregates linked to substance evacuation towards the subpial system. On the basis of our findings, we propose an etiopathogenic model that emphasizes these transporters' role in controlling excitatory/inhibitory motor cortex circuits in order to understand some of the severe motor disturbances observed in TH transporter deficiency syndromes.


Subject(s)
Motor Cortex , Organic Anion Transporters , Symporters , Adult , Humans , Brain/metabolism , Interneurons/metabolism , Monocarboxylic Acid Transporters/metabolism , Motor Cortex/metabolism , Organic Anion Transporters/metabolism , Peptides , Pyramidal Cells/metabolism , Thyroid Hormones
6.
Thyroid ; 33(4): 501-510, 2023 04.
Article in English | MEDLINE | ID: mdl-36565029

ABSTRACT

Introduction: Patients lacking functional monocarboxylate transporter 8 (MCT8), a highly specific thyroid hormone (TH) transporter, present severe psychomotor disabilities. MCT8 deficiency leads to peripheral hyperthyroidism and brain hypothyroidism, the latter due to impaired transport of TH across brain barriers. Available treatments for patients are limited and aim to overcome the limited TH transport across brain barriers. The use of TH analogues such as 3,3',5-triiodothyroacetic acid (TRIAC) that do not require MCT8 to cross the cellular membranes is considered a potential therapy for MCT8 deficiency. Previous studies have shown that systemic administration of TRIAC at therapeutic doses does not increase TRIAC content in the brain, while intracerebroventricular (ICV) administration of therapeutic doses of TRIAC increases TRIAC content in the brain but does not mediate thyromimetic effects. In view of this, we hypothesize that ICV administration of high doses of TRIAC can mediate thyromimetic effects in the brain without worsening the brain hypothyroidism or peripheral hyperthyroidism of patients. Methods: We administered 400 ng/g of body weight per day of ICV TRIAC in a mouse model of MCT8 deficiency: Mct8-/y and deiodinase 2 (Dio2)-/- double knockout mice. The effects of this treatment on TH and TRIAC levels/content in blood and tissues were determined by radioimmunoassay and effects on TH-regulated genes were assessed by real time-quantitative polymerase chain reaction in peripheral and central tissues. Results: ICV administration of high doses of TRIAC ameliorated the peripheral hyperthyroidism. In the brain, this treatment did not further aggravate brain hypothyroidism and increased TRIAC content in several brain regions; however, only moderate thyromimetic activity was observed in restricted brain areas. Conclusion: Administration of high doses of TRIAC by ICV delivery at juvenile stages in a mouse model of MCT8 deficiency is effective in normalizing peripheral hyperthyroidism but exerts minimal thyromimetic activity in the brain.


Subject(s)
Hyperthyroidism , Hypothyroidism , Symporters , Animals , Mice , Symporters/genetics , Triiodothyronine , Thyroid Hormones , Brain , Hyperthyroidism/drug therapy , Hypothyroidism/drug therapy , Mice, Knockout , Disease Models, Animal , Monocarboxylic Acid Transporters/genetics
7.
Neurobiol Dis ; 174: 105896, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36243247

ABSTRACT

Inactivating mutations in the specific thyroid hormone transporter monocarboxylate transporter 8 (MCT8) lead to an X-linked rare disease named MCT8 deficiency or Allan-Herndon-Dudley Syndrome. Patients exhibit a plethora of severe endocrine and neurological alterations, with no effective treatment for the neurological symptoms. An optimal mammalian model is essential to explore the pathological mechanisms and potential therapeutic approaches. Here we have generated by CRISPR/Cas9 an avatar mouse model for MCT8 deficiency with a point mutation found in two MCT8-deficient patients (P253L mice). We have predicted by in silico studies that this mutation alters the substrate binding pocket being the probable cause for impairing thyroid hormone transport. We have characterized the phenotype of MCT8-P253L mice and found endocrine alterations similar to those described in patients and in MCT8-deficient mice. Importantly, we detected brain hypothyroidism, structural and functional neurological alterations resembling the patient's neurological impairments. Thus, the P253L mouse provides a valuable model for studying the pathophysiology of MCT8 deficiency and in the future will allow to test therapeutic alternatives such as in vivo gene therapy and pharmacological chaperone therapy to improve the neurological impairments in MCT8 deficiency.


Subject(s)
Monocarboxylic Acid Transporters , Symporters , Animals , Mice , Monocarboxylic Acid Transporters/genetics , Monocarboxylic Acid Transporters/metabolism , Symporters/genetics , Symporters/metabolism , CRISPR-Cas Systems , Thyroid Hormones/metabolism , Disease Models, Animal , Mammals/metabolism
8.
Sci Adv ; 8(14): eabj7110, 2022 04 08.
Article in English | MEDLINE | ID: mdl-35385300

ABSTRACT

The modulation of the host's metabolism to protect tissue from damage induces tolerance to infections increasing survival. Here, we examined the role of the thyroid hormones, key metabolic regulators, in the outcome of malaria. Hypothyroidism confers protection to experimental cerebral malaria by a disease tolerance mechanism. Hypothyroid mice display increased survival after infection with Plasmodium berghei ANKA, diminishing intracranial pressure and brain damage, without altering pathogen burden, blood-brain barrier disruption, or immune cell infiltration. This protection is reversed by treatment with a Sirtuin 1 inhibitor, while treatment of euthyroid mice with a Sirtuin 1 activator induces tolerance and reduces intracranial pressure and lethality. This indicates that thyroid hormones and Sirtuin 1 are previously unknown targets for cerebral malaria treatment, a major killer of children in endemic malaria areas.


Subject(s)
Hypothyroidism , Malaria, Cerebral , Sirtuin 1 , Animals , Brain/metabolism , Disease Models, Animal , Hypothyroidism/metabolism , Malaria, Cerebral/drug therapy , Malaria, Cerebral/metabolism , Mice , Mice, Inbred C57BL , Plasmodium berghei , Sirtuin 1/antagonists & inhibitors , Sirtuin 1/metabolism
9.
Mol Metab ; 42: 101097, 2020 12.
Article in English | MEDLINE | ID: mdl-33049408

ABSTRACT

OBJECTIVE: Metainflammation is a chronic low-grade inflammatory state induced by obesity and associated comorbidities, including peripheral insulin resistance. Brown adipose tissue (BAT), a therapeutic target against obesity, is an insulin target tissue sensitive to inflammation. Therefore, it is necessary to find strategies to protect BAT against the effects of inflammation in energy balance. In this study, we explored the impact of moderate sirtuin 1 (SIRT1) overexpression on insulin sensitivity and ß-adrenergic responses in BAT and brown adipocytes (BA) under pro-inflammatory conditions. METHODS: The effect of inflammation on BAT functionality was studied in obese db/db mice and lean wild-type (WT) mice or mice with moderate overexpression of SIRT1 (SIRT1Tg+) injected with a low dose of bacterial lipopolysaccharide (LPS) to mimic endotoxemia. We also conducted studies on differentiated BA (BA-WT and BA-SIRT1Tg+) exposed to a macrophage-derived pro-inflammatory conditioned medium (CM) to evaluate the protection of SIRT1 overexpression in insulin signaling and glucose uptake, mitochondrial respiration, fatty acid oxidation (FAO), and norepinephrine (NE)-mediated-modulation of uncoupling protein-1 (UCP-1) expression. RESULTS: BAT from the db/db mice was susceptible to metabolic inflammation manifested by the activation of pro-inflammatory signaling cascades, increased pro-inflammatory gene expression, tissue-specific insulin resistance, and reduced UCP-1 expression. Impairment of insulin and noradrenergic responses were also found in the lean WT mice upon LPS injection. In contrast, BAT from the mice with moderate overexpression of SIRT1 (SIRT1Tg+) was protected against LPS-induced activation of pro-inflammatory signaling, insulin resistance, and defective thermogenic-related responses upon cold exposure. Importantly, the decline in triiodothyronine (T3) levels in the circulation and intra-BAT after exposure of the WT mice to LPS and cold was markedly attenuated in the SIRT1Tg+ mice. In vitro BA experiments in the two genotypes revealed that upon differentiation with a T3-enriched medium and subsequent exposure to a macrophage-derived pro-inflammatory CM, only BA-SIRT1Tg+ fully recovered insulin and noradrenergic responses. CONCLUSIONS: This study has ascertained the benefit of the moderate overexpression of SIRT1 to confer protection against defective insulin and ß-adrenergic responses caused by BAT inflammation. Our results have potential therapeutic value in combinatorial therapies for BAT-specific thyromimetics and SIRT1 activators to combat metainflammation in this tissue.


Subject(s)
Adipose Tissue, Brown/metabolism , Sirtuin 1/metabolism , Adipocytes/metabolism , Adipocytes/physiology , Adipocytes, Brown/metabolism , Adipocytes, Brown/physiology , Adipose Tissue/metabolism , Adipose Tissue, Brown/physiology , Animals , Energy Metabolism , Female , Gene Expression/genetics , Gene Expression Regulation/genetics , Inflammation/prevention & control , Insulin/metabolism , Insulin Resistance/physiology , Male , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Obesity/metabolism , Receptors, Adrenergic, beta/metabolism , Sirtuin 1/genetics , Sirtuin 1/physiology , Thermogenesis/drug effects , Uncoupling Protein 1/metabolism
10.
Thyroid ; 29(11): 1669-1682, 2019 11.
Article in English | MEDLINE | ID: mdl-31359845

ABSTRACT

Background: Mutations in the thyroid hormone (TH) transporter monocarboxylate transporter 8 (MCT8) lead to peripheral hyperthyroidism and profound psychomotor alterations in humans. Mice lacking Mct8 present peripheral hyperthyroidism but no gross neurological abnormalities due to brain compensatory mechanisms involving the enzyme deiodinase type 2 (Dio2). Methods: Here we have analyzed the endocrine and neurologic phenotype of mice lacking both Mct8 and Dio2 at three and six months of age. Thyroxine (T4) and 3,5,3' triiodothyronine (T3) levels/content were measured by specific radioimmunoassays; motor skill performance was evaluated by the footprint, rotarod, four limb hanging wire, and balance beam tests; and brain histological analysis was performed by immunostaining for neurofilament and parvalbumin. Results: We have found that this mouse model presents peripheral hyperthyroidism and brain hypothyroidism. Interestingly, the severity of the brain hypothyroidism seems permanent and varies across regions, with the striatum being a particularly affected area. We have also found brain alterations at the histological level compatible with TH deficiency and impaired motor skills. Conclusions: These findings indicate the potential of Mct8/Dio2-deficient mice to represent a model for human MCT8 deficiency, to understand the mechanisms underlying its pathophysiology, and ultimately design therapeutic interventions for human patients.


Subject(s)
Brain Diseases/genetics , Iodide Peroxidase/genetics , Monocarboxylic Acid Transporters/genetics , Motor Skills , Nervous System Diseases/genetics , Symporters/genetics , Thyroid Hormones/metabolism , Animals , Brain Diseases/pathology , Brain Diseases/psychology , Disease Models, Animal , Female , Iodide Peroxidase/deficiency , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocarboxylic Acid Transporters/deficiency , Psychomotor Performance , Symporters/deficiency , Thyroid Gland/pathology , Thyroxine/blood , Triiodothyronine/blood , Iodothyronine Deiodinase Type II
11.
Psychoneuroendocrinology ; 84: 51-60, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28654773

ABSTRACT

A euthyroid state in the brain is crucial for its adequate development and function. Impairments in thyroid hormones (THs; T3 or 3,5,3'-triiodothyronine and T4 or thyroxine) levels and availability in brain can lead to neurological alterations and to psychiatric disorders, particularly mood disorders. The thyroid gland synthetizes mainly T4, which is secreted to circulating blood, however, most actions of THs are mediated by T3, the transcriptionally active form. In the brain, intracellular concentrations of T3 are modulated by the activity of type 2 (D2) and type 3 (D3) deiodinases. In the present work, we evaluated learning and memory capabilities and anxiety-like behavior at adult stages in mice lacking D2 (D2KO) and we analyzed the impact of D2-deficiency on TH content and on the expression of T3-dependent genes in the amygdala and the hippocampus. We found that D2KO mice do not present impairments in spatial learning and memory, but they display emotional alterations with increased anxiety-like behavior as well as enhanced auditory-cued fear memory and spontaneous recovery of fear memory following extinction. D2KO mice also presented reduced T3 content in the hippocampus and decreased expression of the T3-dependent gene Dio3 in the amygdala suggesting a hypothyroid status in this structure. We propose that the emotional dysfunctions found in D2KO mice can arise from the reduced T3 content in their brain, which consequently leads to alterations in gene expression with functional consequences. We found a downregulation in the gene encoding for the calcium-binding protein calretinin (Calb2) in the amygdala of D2KO mice that could affect the GABAergic transmission. The current findings in D2KO mice can provide insight into emotional disorders present in humans with DIO2 polymorphisms.


Subject(s)
Anxiety/metabolism , Iodide Peroxidase/deficiency , Triiodothyronine/metabolism , Amygdala/physiopathology , Animals , Anxiety/genetics , Anxiety Disorders/metabolism , Calbindin 2/genetics , Calbindin 2/metabolism , Calcium-Binding Proteins/genetics , Fear , Female , Gene Expression , Hippocampus/physiopathology , Iodide Peroxidase/genetics , Learning/physiology , Male , Memory/physiology , Mice , Mice, Knockout , Thyroid Hormones/metabolism , Thyroxine/blood , Thyroxine/metabolism , Triiodothyronine/blood
12.
Cereb Cortex ; 27(5): 2809-2819, 2017 05 01.
Article in English | MEDLINE | ID: mdl-27166167

ABSTRACT

TaqIA is a polymorphism associated with addictions and dopamine-related traits. It is located in the ankyrin repeat and kinase domain containing 1 gene (ANKK1) nearby the gene for the dopamine D2 receptor (D2R). Since ANKK1 function is unknown, TaqIA-associated traits have been explained only by differences in D2R. Here we report ANKK1 studies in mouse and human brain using quantitative real-time PCR, Western blot, immunohistochemistry, and flow cytometry. ANKK1 mRNA and protein isoforms vary along neurodevelopment in the human and mouse brain. In mouse adult brain ANKK1 is located in astrocytes, nuclei of postmitotic neurons and neural precursors from neurogenic niches. In both embryos and adults, nuclei of neural precursors show significant variation of ANKK1 intensity. We demonstrate a correlation between ANKK1 and the cell cycle. Cell synchronization experiments showed a significant increment of ANKK1-kinase in mitotic cells while ANKK1-kinase overexpression affects G1 and M phase that were found to be modulated by ANKK1 alleles and apomorphine treatment. Furthermore, during embryonic neurogenesis ANKK1 was expressed in slow-dividing neuroblasts and rapidly dividing precursors which are mitotic cells. These results suggest a role of ANKK1 during the cell cycle in neural precursors thus providing biological support to brain structure involvement in the TaqIA-associated phenotypes.


Subject(s)
Brain/metabolism , Cell Cycle/physiology , Gene Expression Regulation, Developmental/genetics , Neural Stem Cells/physiology , Protein Serine-Threonine Kinases/metabolism , Adolescent , Age Factors , Animals , Animals, Newborn , Brain/embryology , Brain/growth & development , Cell Differentiation/physiology , Cell Line, Tumor , Embryo, Mammalian , Fetus , Gestational Age , Glial Fibrillary Acidic Protein/metabolism , Humans , Infant , Mice , Middle Aged , Neurogenesis/physiology , Protein Serine-Threonine Kinases/genetics , RNA, Messenger/metabolism , Tubulin/genetics , Tubulin/metabolism
13.
Proc Natl Acad Sci U S A ; 113(24): E3451-60, 2016 06 14.
Article in English | MEDLINE | ID: mdl-27247403

ABSTRACT

TGF-ß, the most potent profibrogenic factor, acts by activating SMAD (mothers against decapentaplegic) transcription factors, which bind to SMAD-binding elements in target genes. Here, we show that the thyroid hormone triiodothyronine (T3), through binding to its nuclear receptors (TRs), is able to antagonize transcriptional activation by TGF-ß/SMAD. This antagonism involves reduced phosphorylation of SMADs and a direct interaction of the receptors with SMAD3 and SMAD4 that is independent of T3-mediated transcriptional activity but requires residues in the receptor DNA binding domain. T3 reduces occupancy of SMAD-binding elements in response to TGF-ß, reducing histone acetylation and inhibiting transcription. In agreement with this transcriptional cross-talk, T3 is able to antagonize fibrotic processes in vivo. Liver fibrosis induced by carbon tetrachloride is attenuated by thyroid hormone administration to mice, whereas aged TR knockout mice spontaneously accumulate collagen. Furthermore, skin fibrosis induced by bleomycin administration is also reduced by the thyroid hormones. These findings define an important function of the thyroid hormone receptors and suggest TR ligands could have beneficial effects to block the progression of fibrotic diseases.


Subject(s)
Liver Cirrhosis/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism , Triiodothyronine/metabolism , Animals , Bleomycin/adverse effects , Bleomycin/pharmacology , Carbon Tetrachloride Poisoning/genetics , Carbon Tetrachloride Poisoning/metabolism , Carbon Tetrachloride Poisoning/pathology , Liver Cirrhosis/chemically induced , Liver Cirrhosis/genetics , Liver Cirrhosis/pathology , Mice , Mice, Knockout , Smad3 Protein/genetics , Smad3 Protein/metabolism , Smad4 Protein/genetics , Smad4 Protein/metabolism , Transforming Growth Factor beta/genetics , Triiodothyronine/genetics
14.
PLoS One ; 9(8): e103857, 2014.
Article in English | MEDLINE | ID: mdl-25083788

ABSTRACT

BACKGROUND: Thyroid hormones have a key role in both the developing and adult central nervous system and skeletal muscle. The thyroid gland produces mainly thyroxine (T4) but the intracellular concentrations of 3,5,3'-triiodothyronine (T3; the transcriptionally active hormone) in the central nervous system and skeletal muscle are modulated by the activity of type 2 deiodinase (D2). To date no neurological syndrome has been associated with mutations in the DIO2 gene and previous studies in young and juvenile D2-knockout mice (D2KO) did not find gross neurological alterations, possibly due to compensatory mechanisms. AIM: This study aims to analyze the motor phenotype of 3-and-6-month-old D2KO mice to evaluate the role of D2 on the motor system at adult stages in which compensatory mechanisms could have failed. RESULTS: Motor abilities were explored by validated tests. In the footprint test, D2KO showed an altered global gait pattern (mice walked slower, with shorter strides and with a hindlimb wider base of support than wild-type mice). No differences were detected in the balance beam test. However, a reduced latency to fall was found in the rotarod, coat-hanger and four limb hanging wire tests indicating impairment on coordination and prehensile reflex and a reduction of muscle strength. In histological analyses of cerebellum and skeletal muscle, D2KO mice did not present gross structural abnormalities. Thyroid hormones levels and deiodinases activities were also determined. In D2KO mice, despite euthyroid T3 and high T4 plasma levels, T3 levels were significantly reduced in cerebral cortex (48% reduction) and skeletal muscle (33% reduction), but not in the cerebellum where other deiodinase (type 1) is expressed. CONCLUSIONS: The motor alterations observed in D2KO mice indicate an important role for D2 in T3 availability to maintain motor function and muscle strength. Our results suggest a possible implication of D2 in motor disorders.


Subject(s)
Iodide Peroxidase/metabolism , Animals , Cerebral Cortex/enzymology , Cerebral Cortex/metabolism , Gait/genetics , Gait/physiology , Iodide Peroxidase/genetics , Locomotion/genetics , Locomotion/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/enzymology , Muscle, Skeletal/metabolism , Thyroxine/metabolism , Triiodothyronine/metabolism
15.
PLoS One ; 6(10): e26582, 2011.
Article in English | MEDLINE | ID: mdl-22039511

ABSTRACT

Hypothyroidism is the most common hormonal disease in adults, which is frequently accompanied by learning and memory impairments and emotional disorders. However, the deleterious effects of thyroid hormones deficiency on emotional memory are poorly understood and often underestimated. To evaluate the consequences of hypothyroidism on emotional learning and memory, we have performed a classical Pavlovian fear conditioning paradigm in euthyroid and adult-thyroidectomized Wistar rats. In this experimental model, learning acquisition was not impaired, fear memory was enhanced, memory extinction was delayed and spontaneous recovery of fear memory was exacerbated in hypothyroid rats. The potentiation of emotional memory under hypothyroidism was associated with an increase of corticosterone release after fear conditioning and with higher expression of glucocorticoid and mineralocorticoid receptors in the lateral and basolateral nuclei of the amygdala, nuclei that are critically involved in the circuitry of fear memory. Our results demonstrate for the first time that adult-onset hypothyroidism potentiates fear memory and also increases vulnerability to develop emotional memories. Furthermore, our findings suggest that enhanced corticosterone signaling in the amygdala is involved in the pathophysiological mechanisms of fear memory potentiation. Therefore, we recommend evaluating whether inappropriate regulation of fear in patients with post-traumatic stress and other mental disorders is associated with abnormal levels of thyroid hormones, especially those patients refractory to treatment.


Subject(s)
Amygdala/metabolism , Fear , Hypothyroidism/psychology , Memory , Receptors, Glucocorticoid/metabolism , Receptors, Mineralocorticoid/metabolism , Adult , Age of Onset , Animals , Humans , Hypothyroidism/metabolism , Rats
16.
Eur J Neurosci ; 30(4): 679-92, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19686470

ABSTRACT

Activity-dependent changes taking place at the hippocampal perforant pathway-dentate gyrus synapse during classical eyeblink conditioning were recorded in adult thyroidectomized (hypothyroid) and control (euthyroid) rats, and in animals treated with thyroid hormones 20 days after thyroidectomy (recovery rats). The aim was to determine the contribution of thyroid hormones and the consequences of adult-onset hypothyroidism to both associative learning and the physiological potentiation of hippocampal synapses during the actual learning process in alert behaving animals. Control and recovery rats presented similar learning curves, whereas hypothyroid animals presented lower values. A single pulse presented to the perforant pathway during the conditioned-unconditioned inter-stimulus interval evoked a monosynaptic field excitatory postsynaptic potential in dentate granule cells (whose slope was linearly related to the rate of acquisition in the control group), but not in hypothyroid and recovery animals. Input-output relationships and long-term potentiation evoked by train stimulation of the perforant pathway were significantly depressed in hypothyroid animals. Thyroid hormone treatment failed to normalize these two neurophysiological abnormalities observed in hypothyroid animals. In contrast, paired-pulse facilitation was not affected by thyroidectomy. The results indicate that thyroid hormone treatment after a short period of adult hypothyroidism helps to restore some hippocampally dependent functions, such as classical conditioning, but not other hippocampal properties, such as the synaptic plasticity evoked during associative learning and during experimentally induced long-term potentiation. The present results have important clinical implications for the handling of patients with adult-onset thyroid diseases.


Subject(s)
Conditioning, Eyelid/drug effects , Hippocampus/drug effects , Neuronal Plasticity/drug effects , Neurons/drug effects , Synaptic Potentials/drug effects , Thyroid Hormones/pharmacology , Acoustic Stimulation , Analysis of Variance , Animals , Conditioning, Eyelid/physiology , Electric Stimulation , Electrodes, Implanted , Electromyography , GABA Plasma Membrane Transport Proteins/metabolism , Hippocampus/metabolism , Hippocampus/physiology , Immunohistochemistry , Liver/chemistry , Long-Term Potentiation/drug effects , Long-Term Potentiation/physiology , Male , Neuronal Plasticity/physiology , Neurons/metabolism , Neurons/physiology , Parvalbumins/metabolism , Rats , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/metabolism , Signal Processing, Computer-Assisted , Synapses/drug effects , Synapses/physiology , Synaptic Potentials/physiology , Thyroidectomy , Thyroxine/analysis
17.
Mol Cell Biol ; 26(6): 2317-26, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16508007

ABSTRACT

dickkopf (dkk) genes encode a small family of secreted Wnt antagonists, except for dkk3, which is divergent and whose function is poorly understood. Here, we describe the generation and characterization of dkk3 mutant mice. dkk3-deficient mice are viable and fertile. Phenotypic analysis shows no major alterations in organ morphology, physiology, and most clinical chemistry parameters. Since Dkk3 was proposed to function as thyroid hormone binding protein, we have analyzed deiodinase activities, as well as thyroid hormone levels. Mutant mice are euthyroid, and the data do not support a relationship of dkk3 with thyroid hormone metabolism. Altered phenotypes in dkk3 mutant mice were observed in the frequency of NK cells, immunoglobulin M, hemoglobin, and hematocrit levels, as well as lung ventilation. Furthermore, dkk3-deficient mice display hyperactivity.


Subject(s)
Behavior, Animal/physiology , Immune System/physiology , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Pulmonary Ventilation/genetics , Adaptor Proteins, Signal Transducing , Animals , Antigens, Helminth/genetics , Antigens, Helminth/immunology , Erythrocytes/pathology , Female , Immunoglobulin M/blood , Intercellular Signaling Peptides and Proteins/immunology , Iodide Peroxidase/metabolism , Lung/physiopathology , Male , Mice , Mice, Mutant Strains , Thyroxine/metabolism , Triiodothyronine/metabolism , Iodothyronine Deiodinase Type II
18.
Endocrinology ; 144(3): 1045-52, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12586781

ABSTRACT

Deiodinases (D1, D2, and D3) are selenoproteins involved in thyroid hormone metabolism. Generation of the active hormone T(3), from T(4), is carried out by D1 and D2, whereas D3 degrades both hormones. The identity of the cloned D2 as a selenoprotein is well supported by biochemical and physiological data. However, an alternative view has proposed that type 2 deiodinase is a nonselenoprotein complex containing a putative T(4) binding subunit called p29, with an almost identity in sequence with the Dickkopf protein Dkk3. To explore a possible functional relationship between p29 and D2, we have compared their mRNA expression patterns in the rat brain. In brain, parenchyma p29 was expressed in neurons. High expression levels were found in all the regions of the blood-cerebrospinal fluid (CSF) barrier. p29 was present in different types of cells than D2, with the exception of the tanycytes. Our data do not support that p29 has a functional relationship with D2. On the other hand, expression of p29 in the blood-CSF barrier suggests that it might be involved in T(4) transport to and from the CSF, but further studies are needed to substantiate this hypothesis.


Subject(s)
Brain Chemistry , Gene Expression , Iodide Peroxidase/genetics , Thyroxine-Binding Proteins/genetics , Animals , Biological Transport , Blood-Brain Barrier , Blotting, Northern , In Situ Hybridization , Iodide Peroxidase/cerebrospinal fluid , Male , Neuroglia/chemistry , Neurons/chemistry , RNA, Messenger/analysis , RNA, Messenger/cerebrospinal fluid , Rats , Rats, Wistar , Thyroxine/metabolism , Thyroxine-Binding Proteins/cerebrospinal fluid , Thyroxine-Binding Proteins/physiology , Tissue Distribution , Iodothyronine Deiodinase Type II
SELECTION OF CITATIONS
SEARCH DETAIL
...