Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Eur Thyroid J ; 10(6): 533-541, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34956926

RESUMO

INTRODUCTION: Resistance to thyroid hormone ß (RTHß) is an inherited syndrome caused by dominant negative variants in the THRB gene (NM_000461.5). The clinical picture of RTHß is variable, and patients harboring the same variant may display different degrees of disease severity. CASE PRESENTATION: A 30-year-old man presented with thyrotoxicosis and central hyperthyroidism and was found to have a novel variant in the exon 10 of THRB gene (c.C1282G, p.L428V), located within the third hot spot region of the C-terminal of the receptor. Surprisingly, the same variant was found in two other relatives with an apparent normal thyroid function at initial screening. After exclusion of a TSH-secreting adenoma and serum interference in the proband, and the finding that exogenous levothyroxine failed to suppress the TSH in the brother affected by nodular goiter, relatives' thyroid function tests (TFTs) were reassessed with additional analytical method revealing biochemical features consistent with RTHß in all carriers of the p.L428V variant. Functional studies showed a slightly impaired in vitro transcriptional activity of p.L428V. Interestingly' the expression of the human p.L428V thyroid hormone receptor beta in the zebrafish embryo background generated a phenotype consistent with RTHß. CONCLUSION: Variable results of TFTs on some immunoassays can be a cause of RTHß diagnostic delay, but the genotype-phenotype correlation in this family and functional studies support p.L428V as a novel THRB variant expanding the spectrum of gene variants causing RTHß. In vivo, rather than in vitro, functional assays may be required to demonstrate the dominant negative action of THRB variants.

2.
Thyroid ; 30(11): 1681-1684, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32669045

RESUMO

A 23-year-old man and his grandmother with hyperthyroxinemia and hypercortisolemia were heterozygous for an ALB mutation (p. Arg218Pro), known to cause familial dysalbuminemic hyperthyroxinemia (FDH). However, serum-free cortisol levels in these individuals were normal and total cortisol concentrations fell markedly after depletion of albumin from their serum. We conclude that binding of steroid as well as iodothyronines to mutant albumin causes raised circulating cortisol as well as thyroid hormones in euthyroid euadrenal individuals with R218P FDH, with potential for misdiagnosis, unnecessary investigation, and inappropriate treatment.


Assuntos
Hidrocortisona/sangue , Hipertireoxinemia Disalbuminêmica Familiar/complicações , Hipertireoxinemia/complicações , Mutação , Albumina Sérica Humana/genética , Albuminas/química , Genótipo , Heterozigoto , Humanos , Imunoensaio , Masculino , Militares , Ligação Proteica , Albumina Sérica/genética , Esteroides/química , Tironinas/sangue , Tiroxina/sangue , Adulto Jovem
3.
Diabetes ; 67(6): 1086-1092, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29622583

RESUMO

Loss-of-function mutations in PPARG cause familial partial lipodystrophy type 3 (FPLD3) and severe metabolic disease in many patients. Missense mutations in PPARG are present in ∼1 in 500 people. Although mutations are often binarily classified as benign or deleterious, prospective functional classification of all missense PPARG variants suggests that their impact is graded. Furthermore, in testing novel mutations with both prototypic endogenous (e.g., prostaglandin J2 [PGJ2]) and synthetic ligands (thiazolidinediones, tyrosine agonists), we observed that synthetic agonists selectively rescue function of some peroxisome proliferator-activated receptor-γ (PPARγ) mutants. We report on patients with FPLD3 who harbor two such PPARγ mutations (R308P and A261E). Both PPARγ mutants exhibit negligible constitutive or PGJ2-induced transcriptional activity but respond readily to synthetic agonists in vitro, with structural modeling providing a basis for such differential ligand-dependent responsiveness. Concordant with this finding, dramatic clinical improvement was seen after pioglitazone treatment of a patient with R308P mutant PPARγ. A patient with A261E mutant PPARγ also responded beneficially to rosiglitazone, although cardiomyopathy precluded prolonged thiazolidinedione use. These observations indicate that detailed structural and functional classification can be used to inform therapeutic decisions in patients with PPARG mutations.


Assuntos
Hipoglicemiantes/uso terapêutico , Lipodistrofia Parcial Familiar/tratamento farmacológico , Lipodistrofia Parcial Familiar/genética , Modelos Moleculares , Mutação de Sentido Incorreto , PPAR gama/genética , Tiazolidinedionas/uso terapêutico , Adolescente , Adulto , Substituição de Aminoácidos , Sítios de Ligação , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Genes Reporter/efeitos dos fármacos , Células HEK293 , Humanos , Hipoglicemiantes/efeitos adversos , Hipoglicemiantes/química , Hipoglicemiantes/farmacologia , Ligantes , Lipodistrofia Parcial Familiar/metabolismo , Conformação Molecular , Simulação de Acoplamento Molecular , PPAR gama/agonistas , PPAR gama/química , PPAR gama/metabolismo , Farmacogenética/métodos , Pioglitazona , Conformação Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Rosiglitazona , Tiazolidinedionas/efeitos adversos , Tiazolidinedionas/química , Tiazolidinedionas/farmacologia , Adulto Jovem
4.
J Endocr Soc ; 1(9): 1203-1212, 2017 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-29264576

RESUMO

Resistance to thyroid hormone ß (RTHß) due to homozygous THRB defects is exceptionally rare, with only five kindreds reported worldwide. Cardiac dysfunction, which can be life-threatening, is recognized in the disorder. Here we describe the clinical, metabolic, ophthalmic, and cardiac findings in a 9-year-old boy harboring a biallelic THRB mutation (R243Q), along with biochemical, physiologic, and cardiac responses to carbimazole and triiodothyroacetic acid (TRIAC) therapy. The patient exhibits recognized features (goiter, nonsuppressed thyroid-stimulating hormone levels, upper respiratory tract infections, hyperactivity, low body mass index) of heterozygous RTHß, with additional characteristics (dysmorphic facies, winging of scapulae) and more markedly elevated thyroid hormone levels, associated with the homozygous form of the disorder. Notably, an older sibling with similar clinical features and probable homozygous RTHß had died of cardiac failure at age 13 years. Features of early dilated cardiomyopathy in our patient prompted combination treatment with carbimazole and TRIAC. Careful titration of therapy limited elevation in TSH levels and associated increase in thyroid volume. Subsequently, sustained reduction in thyroid hormones with normal TSH levels was reflected in lower basal metabolic rate, gain of lean body mass, and improved growth and cardiac function. A combination of antithyroid drug and TRIAC therapy may prevent thyrotoxic cardiomyopathy and its decompensation in homozygous or even heterozygous RTHß in which life-threatening hyperthyroid features predominate.

5.
Thyroid ; 27(7): 973-982, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28471274

RESUMO

BACKGROUND: Resistance to thyroid hormone alpha (RTHα), a disorder characterized by tissue-selective hypothyroidism and near-normal thyroid function tests due to thyroid receptor alpha gene mutations, is rare but probably under-recognized. This study sought to correlate the clinical characteristics and response to thyroxine (T4) therapy in two adolescent RTHα patients with the properties of the THRA mutation, affecting both TRα1 and TRα2 proteins, they harbored. METHODS: Clinical, auxological, biochemical, and physiological parameters were assessed in each patient at baseline and after T4 therapy. RESULTS: Heterozygous THRA mutations occurring de novo were identified in a 17-year-old male (patient P1; c.788C>T, p.A263V mutation) investigated for mild pubertal delay and in a 15-year-old male (patient P2; c.821T>C, p.L274P mutation) with short stature (0.4th centile), skeletal dysplasia, dysmorphic facies, and global developmental delay. Both individuals exhibited macrocephaly, delayed dentition, and constipation, together with a subnormal T4/triiodothyronine (T3) ratio, low reverse T3 levels, and mild anemia. When studied in vitro, A263V mutant TRα1 was transcriptionally impaired and inhibited the function of its wild-type counterpart at low (0.01-10 nM) T3 levels, with higher T3 concentrations (100 nM-1 µM) reversing dysfunction and such dominant negative inhibition. In contrast, L274P mutant TRα1 was transcriptionally inert, exerting significant dominant negative activity, only overcome with 10 µM of T3. Mirroring this, normal expression of KLF9, a TH-responsive target gene, was achieved in A263V mutation-containing peripheral blood mononuclear cells following 1 µM of T3 exposure, but with markedly reduced expression levels in L274P mutation-containing peripheral blood mononuclear cells, even with 10 µM of T3. Following T4 therapy, growth, body composition, dyspraxia, and constipation improved in P1, whereas growth retardation and constipation in P2 were unchanged. Neither A263V nor L274P mutations exhibited gain or loss of function in the TRα2 background, and no additional phenotype attributable to this was discerned. CONCLUSIONS: This study correlates a milder clinical phenotype and favorable response to T4 therapy in a RTHα patient (P1) with heterozygosity for mutant TRα1 exhibiting partial, T3-reversible, loss of function. In contrast, a more severe clinical phenotype refractory to hormone therapy was evident in another case (P2) associated with severe, virtually irreversible, dysfunction of mutant TRα1.


Assuntos
Mutação , Receptores alfa dos Hormônios Tireóideos/genética , Síndrome da Resistência aos Hormônios Tireóideos/genética , Tiroxina/uso terapêutico , Adolescente , Predisposição Genética para Doença , Genótipo , Humanos , Masculino , Fenótipo , Avaliação de Sintomas , Síndrome da Resistência aos Hormônios Tireóideos/diagnóstico , Síndrome da Resistência aos Hormônios Tireóideos/tratamento farmacológico , Resultado do Tratamento , Adulto Jovem
6.
J Clin Invest ; 126(3): 992-6, 2016 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-26854926

RESUMO

Selenium is a trace element that is essential for human health and is incorporated into more than 25 human selenocysteine-containing (Sec-containing) proteins via unique Sec-insertion machinery that includes a specific, nuclear genome-encoded, transfer RNA (tRNA[Ser]Sec). Here, we have identified a human tRNA[Ser]Sec mutation in a proband who presented with a variety of symptoms, including abdominal pain, fatigue, muscle weakness, and low plasma levels of selenium. This mutation resulted in a marked reduction in expression of stress-related, but not housekeeping, selenoproteins. Evaluation of primary cells from the homozygous proband and a heterozygous parent indicated that the observed deficit in stress-related selenoprotein production is likely mediated by reduced expression and diminished 2'-O-methylribosylation at uridine 34 in mutant tRNA[Ser]Sec. Moreover, this methylribosylation defect was restored by cellular complementation with normal tRNA[Ser]Sec. This study identifies a tRNA mutation that selectively impairs synthesis of stress-related selenoproteins and demonstrates the importance of tRNA modification for normal selenoprotein synthesis.


Assuntos
Doenças Genéticas Inatas/diagnóstico , RNA de Transferência Aminoácido-Específico/genética , Selenoproteínas/genética , Sequência de Bases , Criança , Análise Mutacional de DNA , Estudos de Associação Genética , Doenças Genéticas Inatas/genética , Humanos , Masculino , Dados de Sequência Molecular , Mutação Puntual , Polimorfismo de Nucleotídeo Único , Biossíntese de Proteínas , Selenoproteínas/sangue , Selenoproteínas/deficiência
7.
Lancet Diabetes Endocrinol ; 2(8): 619-26, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24969835

RESUMO

BACKGROUND: The thyroid hormone receptor α gene (THRA) transcript is alternatively spliced to generate either thyroid hormone receptor (TR)α1 or a non-hormone-binding variant protein, TRα2, the function of which is unknown. Here, we describe the first patients identified with a mutation in THRA that affects both TRα1 and TRα2, and compare them with patients who have resistance to thyroid hormone owing to a mutation affecting only TRα1, to delineate the relative roles of TRα1 and TRα2. METHODS: We did clinical, biochemical, and genetic analyses of an index case and her two sons. We assessed physical and radiological features, thyroid function, physiological and biochemical markers of thyroid hormone action, and THRA sequence. FINDINGS: The patients presented in childhood with growth failure, developmental delay, and constipation, which improved after treatment with thyroxine, despite normal concentrations of circulating thyroid hormones. They had similar clinical (macrocephaly, broad faces, skin tags, motor dyspraxia, slow speech), biochemical (subnormal ratio of free thyroxine:free tri-iodothyronine [T3], low concentration of total reverse T3, high concentration of creatine kinase, mild anaemia), and radiological (thickened calvarium) features to patients with TRα1-mediated resistance to thyroid hormone, although our patients had a heterozygous mis-sense mutation (Ala263Val) in both TRα1 and TRα2 proteins. The Ala263Val mutant TRα1 inhibited the transcriptional function of normal receptor in a dominant-negative fashion. By contrast, function of Ala263Val mutant TRα2 matched its normal counterpart. In vitro, high concentrations of T3 restored transcriptional activity of Ala263Val mutant TRα1, and reversed the dominant-negative inhibition of its normal counterpart. High concentrations of T3 restored expression of thyroid hormone-responsive target genes in patient-derived blood cells. INTERPRETATION: TRα1 seems to be the principal functional product of the THRA gene. Thyroxine treatment alleviates hormone resistance in patients with mutations affecting this gene, possibly ameliorating the phenotype. These findings will help the diagnosis and treatment of other patients with resistance to thyroid hormone resulting from mutations in THRA. FUNDING: Wellcome Trust, NIHR Cambridge Biomedical Research Centre, Marie Curie Actions, Foundation for Development of Internal Medicine in Europe.


Assuntos
Processamento Alternativo , Mutação de Sentido Incorreto , Receptores alfa dos Hormônios Tireóideos/genética , Síndrome da Resistência aos Hormônios Tireóideos/genética , Adulto , Substituição de Aminoácidos , Saúde da Família , Feminino , Apraxia da Marcha/etiologia , Heterozigoto , Humanos , Masculino , Megalencefalia/etiologia , Pessoa de Meia-Idade , Pólipos/etiologia , Isoformas de Proteínas/agonistas , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Neoplasias Cutâneas/etiologia , Distúrbios da Fala/etiologia , Receptores alfa dos Hormônios Tireóideos/agonistas , Receptores alfa dos Hormônios Tireóideos/metabolismo , Síndrome da Resistência aos Hormônios Tireóideos/tratamento farmacológico , Síndrome da Resistência aos Hormônios Tireóideos/fisiopatologia , Tiroxina/uso terapêutico , Resultado do Tratamento
8.
J Clin Endocrinol Metab ; 99(7): E1381-6, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24646103

RESUMO

CONTEXT: Familial dysalbuminemic hyperthyroxinemia, characterized by abnormal circulating albumin with increased T4 affinity, causes artefactual elevation of free T4 concentrations in euthyroid individuals. OBJECTIVE: Four unrelated index cases with discordant thyroid function tests in different assay platforms were investigated. DESIGN AND RESULTS: Laboratory biochemical assessment, radiolabeled T4 binding studies, and ALB sequencing were undertaken. (125)I-T4 binding to both serum and albumin in affected individuals was markedly increased, comparable with known familial dysalbuminemic hyperthyroxinemia cases. Sequencing showed heterozygosity for a novel ALB mutation (arginine to isoleucine at codon 222, R222I) in all four cases and segregation of the genetic defect with abnormal biochemical phenotype in one family. Molecular modeling indicates that arginine 222 is located within a high-affinity T4 binding site in albumin, with substitution by isoleucine, which has a smaller side chain predicted to reduce steric hindrance, thereby facilitating T4 and rT3 binding. When tested in current immunoassays, serum free T4 values from R222I heterozygotes were more measurably abnormal in one-step vs two-step assay architectures. Total rT3 measurements were also abnormally elevated. CONCLUSIONS: A novel mutation (R222I) in the ALB gene mediates dominantly inherited dysalbuminemic hyperthyroxinemia. Susceptibility of current free T4 immunoassays to interference by this mutant albumin suggests likely future identification of individuals with this variant binding protein.


Assuntos
Hipertireoxinemia Disalbuminêmica Familiar/genética , Mutação de Sentido Incorreto , Pré-Albumina/genética , Adulto , Substituição de Aminoácidos , Arginina/genética , Pré-Escolar , Feminino , Humanos , Hipertireoxinemia Disalbuminêmica Familiar/sangue , Isoleucina/genética , Masculino , Modelos Moleculares , Pré-Albumina/química , Testes de Função Tireóidea , Adulto Jovem
9.
J Clin Endocrinol Metab ; 98(11): 4254-61, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23940126

RESUMO

CONTEXT: The first human cases (female, age 6 y; father and daughter, ages 47 and 11 y, respectively) with growth retardation/short stature, skeletal dysplasia, constipation, and defective thyroid receptor α (TRα) have been recently described. OBJECTIVE: A 45-year-old, short, overweight female with cognitive impairment, epilepsy, and constipation was investigated. DESIGN AND INTERVENTION: Clinical, biochemical, and radiological assessment and THRA sequencing were undertaken. The patient's thyroid status and her biochemical and physiological parameters were evaluated at baseline and after T4 therapy. RESULTS: The patient exhibits disproportionate short stature, macrocephaly, low free T4/free T3 ratio and rT3 levels, together with subnormal heart and basal metabolic rate. She is heterozygous for a novel frameshift/premature stop (Ala382ProfsX7) THRA mutation, generating a mutant TRα with constitutive corepressor binding and negligible coactivator recruitment, which inhibits its wild-type counterpart in a dominant-negative manner-both in vitro and in mutation-containing patient blood mononuclear cells studied ex vivo. Her alertness and constipation responded to T4 therapy, which readily suppressed TSH levels, raised basal metabolic rate, and normalized elevated muscle creatine kinase, but cardiac parameters (heart rate, contractility) remained relatively refractory. The patient and a previous childhood case showed reduced red cell mass with macrocytosis unresponsive to T4 therapy. CONCLUSIONS: Clinical (short stature, macrocephaly, constipation) and biochemical (low free T4/free T3 ratio, subnormal rT3) findings that are congruent with previous cases and newly recognized features (epilepsy) in this adult female with defective TRα define a shared phenotype in TRα-mediated resistance to thyroid hormone, with differential tissue responses to T4 treatment.


Assuntos
Resistência a Medicamentos/genética , Hipotireoidismo/tratamento farmacológico , Hipotireoidismo/genética , Receptores alfa dos Hormônios Tireóideos/genética , Tiroxina/uso terapêutico , Saúde da Família , Feminino , Humanos , Hipotireoidismo/metabolismo , Pessoa de Meia-Idade , Receptores alfa dos Hormônios Tireóideos/metabolismo , Tiroxina/metabolismo
10.
N Engl J Med ; 366(3): 243-9, 2012 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-22168587

RESUMO

Thyroid hormones exert their effects through alpha (TRα1) and beta (TRß1 and TRß2) receptors. Here we describe a child with classic features of hypothyroidism (growth retardation, developmental retardation, skeletal dysplasia, and severe constipation) but only borderline-abnormal thyroid hormone levels. Using whole-exome sequencing, we identified a de novo heterozygous nonsense mutation in a gene encoding thyroid hormone receptor alpha (THRA) and generating a mutant protein that inhibits wild-type receptor action in a dominant negative manner. Our observations are consistent with defective human TRα-mediated thyroid hormone resistance and substantiate the concept of hormone action through distinct receptor subtypes in different target tissues.


Assuntos
Códon sem Sentido , Transtornos do Crescimento/genética , Hipotireoidismo/genética , Receptores alfa dos Hormônios Tireóideos/genética , Tiroxina/sangue , Tiroxina/uso terapêutico , Tri-Iodotironina/sangue , Criança , Feminino , Transtornos do Crescimento/tratamento farmacológico , Heterozigoto , Humanos , Hipotireoidismo/tratamento farmacológico , Modelos Moleculares , Conformação Proteica , Receptores alfa dos Hormônios Tireóideos/química , Hormônios Tireóideos/sangue
11.
J Clin Invest ; 120(12): 4220-35, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21084748

RESUMO

Selenium, a trace element that is fundamental to human health, is incorporated into some proteins as selenocysteine (Sec), generating a family of selenoproteins. Sec incorporation is mediated by a multiprotein complex that includes Sec insertion sequence-binding protein 2 (SECISBP2; also known as SBP2). Here, we describe subjects with compound heterozygous defects in the SECISBP2 gene. These individuals have reduced synthesis of most of the 25 known human selenoproteins, resulting in a complex phenotype. Azoospermia, with failure of the latter stages of spermatogenesis, was associated with a lack of testis-enriched selenoproteins. An axial muscular dystrophy was also present, with features similar to myopathies caused by mutations in selenoprotein N (SEPN1). Cutaneous deficiencies of antioxidant selenoenzymes, increased cellular ROS, and susceptibility to ultraviolet radiation-induced oxidative damage may mediate the observed photosensitivity. Reduced levels of selenoproteins in peripheral blood cells were associated with impaired T lymphocyte proliferation, abnormal mononuclear cell cytokine secretion, and telomere shortening. Paradoxically, raised ROS in affected subjects was associated with enhanced systemic and cellular insulin sensitivity, similar to findings in mice lacking the antioxidant selenoenzyme glutathione peroxidase 1 (GPx1). Thus, mutation of SECISBP2 is associated with a multisystem disorder with defective biosynthesis of many selenoproteins, highlighting their role in diverse biological processes.


Assuntos
Mutação , Proteínas de Ligação a RNA/genética , Selenoproteínas/deficiência , Adulto , Idoso , Sequência de Aminoácidos , Animais , Azoospermia/genética , Sequência de Bases , Criança , Pré-Escolar , Códon sem Sentido , DNA/genética , Feminino , Perda Auditiva Neurossensorial/genética , Humanos , Resistência à Insulina/genética , Masculino , Camundongos , Pessoa de Meia-Idade , Modelos Moleculares , Dados de Sequência Molecular , Distrofias Musculares/genética , Mutação de Sentido Incorreto , Linhagem , Transtornos de Fotossensibilidade/genética , Proteínas de Ligação a RNA/química , Proteínas de Ligação a RNA/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Selenocisteína/metabolismo , Selenoproteínas/metabolismo , Homologia de Sequência de Aminoácidos , Espermatogênese/genética , Linfócitos T/imunologia
13.
Cell Metab ; 4(4): 303-11, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17011503

RESUMO

PPARgamma is essential for adipogenesis and metabolic homeostasis. We describe mutations in the DNA and ligand binding domains of human PPARgamma in lipodystrophic, severe insulin resistance. These receptor mutants lack DNA binding and transcriptional activity but can translocate to the nucleus, interact with PPARgamma coactivators and inhibit coexpressed wild-type receptor. Expression of PPARgamma target genes is markedly attenuated in mutation-containing versus receptor haploinsufficent primary cells, indicating that such dominant-negative inhibition operates in vivo. Our observations suggest that these mutants restrict wild-type PPARgamma action via a non-DNA binding, transcriptional interference mechanism, which may involve sequestration of functionally limiting coactivators.


Assuntos
DNA/metabolismo , Resistência à Insulina/genética , Lipodistrofia/genética , Lipodistrofia/metabolismo , PPAR gama/genética , DNA Complementar/genética , Perfilação da Expressão Gênica , Humanos , Mutação , PPAR gama/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/fisiologia
14.
Endocrinology ; 145(4): 1527-38, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-14657011

RESUMO

Loss-of-function mutations in the ligand-binding domain of human peroxisome proliferator-activated receptor gamma (PPARgamma) are associated with a novel syndrome characterized by partial lipodystrophy and severe insulin resistance. Here we have further characterized the properties of natural dominant-negative PPARgamma mutants (P467L, V290M) and evaluated the efficacy of putative natural ligands and synthetic thiazolidinedione (TZD) or tyrosine-based (TA) receptor agonists in rescuing mutant receptor function. A range of natural ligands failed to activate the PPARgamma mutants and their transcriptional responses to TZDs (e.g. pioglitazone, rosiglitazone) were markedly attenuated, whereas TAs (e.g. farglitazar) corrected defects in ligand binding and coactivator recruitment by the PPARgamma mutants, restoring transcriptional function comparable with wild-type receptor. Transcriptional silencing via recruitment of corepressor contributes to dominant-negative inhibition of wild type by the P467L and V290M mutants and the introduction of an artificial mutation (L318A) disrupting corepressor interaction abrogated their dominant-negative activity. More complete ligand-dependent corepressor release and reversal of dominant-negative inhibition was achieved with TA than TZD agonists. Modeling suggests a structural basis for these observations: both mutations destabilize helix 12 to favor receptor-corepressor interaction; conversely, farglitazar makes more extensive contacts than rosiglitazone within the ligand-binding pocket, to stabilize helix 12, facilitating corepressor release and transcriptional activation. Farglitazar was a more potent inducer of PPARgamma target gene (aP2) expression in peripheral blood mononuclear cells with the P467L mutation. Having shown that rosiglitazone is of variable and limited efficacy in these subjects, we suggest that TAs may represent a more rational therapeutic approach.


Assuntos
Genes Dominantes , Mutação , Receptores Citoplasmáticos e Nucleares/efeitos dos fármacos , Receptores Citoplasmáticos e Nucleares/genética , Fatores de Transcrição/efeitos dos fármacos , Fatores de Transcrição/genética , Tirosina/agonistas , Sequência de Aminoácidos , Linhagem Celular , Expressão Gênica/efeitos dos fármacos , Humanos , Leucina/genética , Ligantes , Metionina/genética , Modelos Estruturais , Dados de Sequência Molecular , Monócitos/fisiologia , Oxazóis/química , Oxazóis/farmacologia , Pioglitazona , Prolina/genética , Receptores Citoplasmáticos e Nucleares/agonistas , Receptores Citoplasmáticos e Nucleares/metabolismo , Rosiglitazona , Tiazolidinedionas/química , Tiazolidinedionas/farmacologia , Fatores de Transcrição/agonistas , Fatores de Transcrição/metabolismo , Transfecção , Tirosina/análogos & derivados , Tirosina/química , Tirosina/farmacologia , Valina/genética
15.
EMBO J ; 21(19): 5079-87, 2002 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-12356724

RESUMO

Most patients with the syndrome resistance to thyroid hormone (RTH) express a mutant thyroid hormone receptor beta (TRbeta) with transdominant negative transcriptional effects. Since no patient with a mutant TRalpha has been identified, we introduced a point mutation into the mouse thyroid hormone receptor (TRalpha1) locus originally found in the TRbeta gene, that reduces ligand binding 10-fold. Heterozygous 2- to 3-week- old mice exhibit a severe retardation of post-natal development and growth, but only a minor reduction in serum thyroxine levels. Homozygous mice died before 3 weeks of age. Adult heterozygotes overcome most of these defects except for cardiac function abnormalities, suggesting that other factors compensate for the receptor defect. However, the additional deletion of the TRbeta gene in this mouse strain caused a 10-fold increase in serum thyroxine, restored hormonal regulation of target genes for TRs, and rescued the growth retardation. The data demonstrate a novel array of effects mediated by a dominant negative TRalpha1, and may provide important clues for identification of a potentially unrecognized human disorder and its treatment.


Assuntos
Crescimento/genética , Mutação Puntual , Receptores alfa dos Hormônios Tireóideos/genética , Receptores alfa dos Hormônios Tireóideos/fisiologia , Animais , Arginina , Citosina , Modelos Animais de Doenças , Citometria de Fluxo , Genes Dominantes , Genoma , Humanos , Camundongos
16.
Nat Genet ; 31(4): 379-84, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12118251

RESUMO

Impaired insulin action is a key feature of type 2 diabetes and is also found, to a more extreme degree, in familial syndromes of insulin resistance. Although inherited susceptibility to insulin resistance may involve the interplay of several genetic loci, no clear examples of interactions among genes have yet been reported. Here we describe a family in which five individuals with severe insulin resistance, but no unaffected family members, were doubly [corrected] heterozygous with respect to frameshift/premature stop mutations in two unlinked genes, PPARG and PPP1R3A these encode peroxisome proliferator activated receptor gamma, which is highly expressed in adipocytes, and protein phosphatase 1, regulatory subunit 3, the muscle-specific regulatory subunit of protein phosphatase 1, which are centrally involved in the regulation of carbohydrate and lipid metabolism, respectively. That mutant molecules primarily involved in either carbohydrate or lipid metabolism can combine to produce a phenotype of extreme insulin resistance provides a model of interactions among genes that may underlie common human metabolic disorders such as type 2 diabetes.


Assuntos
Resistência à Insulina/genética , Fosfoproteínas Fosfatases/genética , Receptores Citoplasmáticos e Nucleares/genética , Fatores de Transcrição/genética , Adulto , Idoso , Animais , Células CHO , Cricetinae , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Diabetes Mellitus Tipo 2/genética , Feminino , Mutação da Fase de Leitura , Heterozigoto , Humanos , Masculino , Pessoa de Meia-Idade , Linhagem , Fosfoproteínas Fosfatases/metabolismo , Proteína Fosfatase 1 , Receptores Citoplasmáticos e Nucleares/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Fatores de Transcrição/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA