Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
1.
Arerugi ; 70(10): 1383-1390, 2021.
Artigo em Japonês | MEDLINE | ID: mdl-34911891

RESUMO

BACKGROUND: Atopic dermatitis (AD) in early infancy can lead to severe protein-loss in atopic dermatitis (SPLAD). The aim of this study was to elucidate the prognosis of SPLAD. METHODS: This was a single-center, retrospective, observational study based on medical records. Participants comprised 61 children with SPLAD hospitalized at the Allergy Center, National Center for Child Health and Development, from 2002 to 2017. We examined patient characteristics, blood test results, and prognoses up to 3 years, including frequency of topical corticosteroid-(TCS) use and food intake status. RESULTS: All participants improved hypoproteinemia and electrolyte abnormalities with AD treatment alone, without intravenous fluids. We performed proactive therapy to maintain remission by gradually decreasing the frequency of TCS-use. After 1, 2, and 3 years, 77%, 92%, and 95%, respectively, remission was maintained by using TCS 2 days a week or less, whereas 39% did not require TCS after 3 years. No participants received systemic therapy, including systemic steroids, immunosuppressants, or biologics. We observed that 29% of infants younger than 1 year at admission had eliminated one or more egg, milk, or wheat component after 3 years. CONCLUSIONS: Even in patients with SPLAD, the most severe AD, TCS-use may be reduced to 2 days per week or less after 3 years with appropriate skin treatment.


Assuntos
Dermatite Atópica , Fármacos Dermatológicos , Criança , Dermatite Atópica/tratamento farmacológico , Fármacos Dermatológicos/uso terapêutico , Humanos , Lactente , Prognóstico , Estudos Retrospectivos , Resultado do Tratamento
2.
Circ J ; 83(11): 2282-2291, 2019 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-31527337

RESUMO

BACKGROUND: Treatment of myocardial infarction (MI) includes inhibition of the sympathetic nervous system (SNS). Cell-based therapy using adipose-derived stem cells (ASCs) has emerged as a novel therapeutic approach to treat heart failure in MI. The purpose of this study was to determine whether a combination of ASC transplantation and SNS inhibition synergistically improves cardiac functions after MI.Methods and Results:ASCs were isolated from fat tissues of Lewis rats. In in vitro studies using cultured ASC cells, mRNA levels of angiogenic factors under normoxia or hypoxia, and the effects of norepinephrine and a ß-blocker, carvedilol, on the mRNA levels were determined. Hypoxia increased vascular endothelial growth factor (VEGF) mRNA in ASCs. Norepinephrine further increased VEGF mRNA; this effect was unaffected by carvedilol. VEGF promoted VEGF receptor phosphorylation and tube formation of human umbilical vein endothelial cells, which were inhibited by carvedilol. In in vivo studies using a rat MI model, transplanted ASC sheets improved contractile functions of MI hearts; they also facilitated neovascularization and suppressed fibrosis after MI. These beneficial effects of ASC sheets were abolished by carvedilol. The effects of ASC sheets and carvedilol on MI heart functions were confirmed by Langendorff perfusion experiments using isolated hearts. CONCLUSIONS: ASC sheets prevented cardiac dysfunctions and remodeling after MI in a rat model via VEGF secretion. Inhibition of VEGF effects by carvedilol abolished their beneficial effects.


Assuntos
Antagonistas Adrenérgicos beta/farmacologia , Carvedilol/farmacologia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/efeitos dos fármacos , Contração Miocárdica/efeitos dos fármacos , Infarto do Miocárdio/cirurgia , Gordura Subcutânea/citologia , Função Ventricular Esquerda/efeitos dos fármacos , Animais , Hipóxia Celular , Células Cultivadas , Modelos Animais de Doenças , Fibrose , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Masculino , Células-Tronco Mesenquimais/metabolismo , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Neovascularização Fisiológica/efeitos dos fármacos , Fosforilação , Ratos Endogâmicos Lew , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Recuperação de Função Fisiológica , Fator A de Crescimento do Endotélio Vascular/metabolismo , Remodelação Ventricular/efeitos dos fármacos
4.
Endocr J ; 63(8): 755-64, 2016 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-27349268

RESUMO

Graves' ophthalmopathy (GO) is a common manifestation of Graves' disease (GD); however, its pathogenesis is not well understood. Recently, the dysregulation of regulatory T cells (Tregs) has been thought to be closely associated with the pathogenesis and clinical symptoms of autoimmune disease. We therefore evaluated whether T cell subsets, including Tregs, are associated with GO pathogenesis and clinical symptoms. In this observational study we evaluated 35 GD patients with overt ophthalmopathy (GOs) and 28 patients without ophthalmopathy (non-GOs). Fifteen healthy euthyroid patients served as healthy controls (HCs). Peripheral blood mononuclear cells from GOs, non-GOs and HCs were analyzed for CD4, CD25, and FoxP3 expression using flow cytometry. We also evaluated their correlation with disease activity according to the clinical activity score (CAS) and magnetic resonance imaging (MRI) findings. Disease severity was evaluated using the NOSPECS score, and clinical progression of GO was followed for 24 weeks. The main outcome measures were the frequencies of FoxP3-positive and -negative CD4(+) CD25(+) T cells at study outset, namely Tregs and effector T cells (Teffs), respectively. GOs had higher frequencies of Teffs (30.8±8.4%) than non-GOs (19.4±7.1%) and HCs (22.7±7.9%). Notably, patients with improved GOs had lower frequencies of Tregs (5.8±1.1%) than patients with stable or deteriorated GOs (7.3±1.2%), although ophthalmic and radiological parameters were not significantly different at the start of the study. In conclusion, an expanded Teff population may be associated with GO pathogenesis. Additionally, decreased Tregs in peripheral blood may predict a good clinical outcome.


Assuntos
Linfócitos T CD4-Positivos/fisiologia , Fatores de Transcrição Forkhead/metabolismo , Oftalmopatia de Graves/imunologia , Subunidade alfa de Receptor de Interleucina-2/metabolismo , Adulto , Antígenos CD4/metabolismo , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/patologia , Feminino , Citometria de Fluxo , Oftalmopatia de Graves/sangue , Oftalmopatia de Graves/diagnóstico por imagem , Humanos , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Linfócitos T Reguladores/metabolismo , Linfócitos T Reguladores/fisiologia
5.
J Mol Cell Cardiol ; 86: 138-46, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26232501

RESUMO

Kv1.5 confers ultra-rapid delayed-rectifier potassium channel current (IKur) which contributes to repolarization of the atrial action potential. Kv1.5 proteins, degraded via the ubiquitin-proteasome pathway, decreased in some atrial fibrillation patients. Carboxyl-terminus heat shock cognate 70-interacting protein (CHIP), an E3 ubiquitin ligase, is known to ubiquitinate short-lived proteins. Here, we investigated the roles of CHIP in Kv1.5 degradation to provide insights into the mechanisms of Kv1.5 decreases and treatments targeting Kv1.5 for atrial fibrillation. Coexpression of CHIP with Kv1.5 in HEK293 cells increased Kv1.5 protein ubiquitination and decreased the protein level. Immunofluorescence revealed decreases of Kv1.5 proteins in the endoplasmic reticulum and on the cell membrane. A siRNA against CHIP suppressed Kv1.5 protein ubiquitination and increased its protein level. CHIP mutants, lacking either the N-terminal tetratricopeptide region domain or the C-terminal U-box domain, failed to exert these effects on Kv1.5 proteins. Immunoprecipitation showed that CHIP formed complexes with Kv1.5 proteins and heat shock cognate protein 70 (Hsc70). Effects of Hsc70 on Kv1.5 were similar to CHIP by altering interaction of CHIP with Kv1.5 protein. Coexpression of CHIP and Hsc70 with Kv1.5 additionally enhanced Kv1.5 ubiquitination. Kv1.5 currents were decreased by overexpression of CHIP or Hsc70 but were increased by knockdown of CHIP or Hsc70 in HEK 293 cells stably expressing Kv1.5. These effects of CHIP and Hsc70 were also observed on endogenous Kv1.5 in HL-1 mouse cardiomyocytes, decreasing IKur and prolonging action potential duration. These results indicate that CHIP decreases the Kv1.5 protein level and functional channel by facilitating its degradation in concert with chaperone Hsc70.


Assuntos
Fibrilação Atrial/genética , Proteínas de Choque Térmico HSC70/genética , Canal de Potássio Kv1.5/genética , Ubiquitina-Proteína Ligases/genética , Animais , Fibrilação Atrial/patologia , Regulação da Expressão Gênica , Células HEK293 , Proteínas de Choque Térmico HSC70/biossíntese , Proteínas de Choque Térmico HSC70/metabolismo , Humanos , Canal de Potássio Kv1.5/biossíntese , Canal de Potássio Kv1.5/metabolismo , Camundongos , Ligação Proteica , Estrutura Terciária de Proteína , RNA Interferente Pequeno , Transdução de Sinais , Ubiquitina-Proteína Ligases/biossíntese , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação/genética
6.
Clin Exp Nephrol ; 19(4): 576-84, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25239792

RESUMO

BACKGROUND: Familial juvenile hyperuricemic nephropathy (FJHN) is an autosomal dominant disorder caused by mutations in UMOD that encodes uromodulin. Topiroxostat, a novel non-purine analog, selectively inhibits xanthine oxidase and reduces the serum uric acid levels and the urinary albuminuria. METHODS: Genomic DNA of a patient was extracted from peripheral white blood. Exons and flanking sequences of UMOD were amplified by PCR with primers. Mutation analysis was performed by direct sequencing of the PCR products. The wild-type and mutant uromodulin were expressed in HEK293 cells and analyzed by western blotting, immunoprecipitation, immunofluorescence, and flow cytometry. RESULTS: We identified an FJHN patient who carried a novel UMOD mutation G335A (C112Y). The levels of both cytosolic and secreted C112Y protein were significantly decreased compared with the wild-type, whereas the level of ubiquitination was higher in C112Y than that in the wild type. The half-life of C112Y was shortened and it was restored by a proteasome inhibitor MG132. Immunofluorescence revealed decreased levels of C112Y in the Golgi apparatus and on the plasma membrane. Expression of C112Y induced cellular apoptosis as revealed by flow cytometry. Apoptosis induced by C112Y was suppressed by topiroxostat. CONCLUSION: C112Y causes its protein instability resulting cellular apoptosis which could be suppressed with topiroxostat.


Assuntos
Apoptose/efeitos dos fármacos , Gota/genética , Hiperuricemia/genética , Nefropatias/genética , Nitrilas/uso terapêutico , Piridinas/uso terapêutico , Uromodulina/genética , Adulto , Gota/tratamento farmacológico , Células HEK293 , Humanos , Hiperuricemia/tratamento farmacológico , Nefropatias/tratamento farmacológico , Masculino , Mutação , Nitrilas/farmacologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Piridinas/farmacologia
7.
Endocr J ; 62(11): 1007-15, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26370556

RESUMO

We have previously shown that follicular thyroglobulin (Tg) has an unexpected function as an autocrine negative-feedback regulator of thyroid hormone (TH) biosynthesis. Tg significantly suppressed the expression of genes necessary for iodide transport and TH synthesis by counteracting stimulation by TSH. However, whether follicular Tg also regulates intracellular TH transport and its secretion from thyrocytes is not known. In the present study, we examined the potential effect of follicular Tg on TH transport and secretion by quantifying the expression of two TH transporters: monocarboxylate transporter 8 (MCT8) and µ-crystallin (CRYM). Our results showed that follicular Tg at physiologic concentrations enhanced both the mRNA and protein expression levels of MCT8 and CRYM in a time- and dose-dependent manner in rat thyroid FRTL-5 cells. Although both the sodium/iodide symporter (NIS), an essential transporter of iodide from blood into the thyroid, and MCT8, a transporter of synthesized TH from the gland, were co-localized on the basolateral membrane of rat thyrocytes in vivo, Tg decreased NIS expression and increased the expression of MCT8 by counteracting TSH action. Thus, the effect of Tg on TH secretion opposed its previously described negative-feedback suppression of TH synthesis. Our results indicate that Tg mediates a complex intrinsic regulation of gene expression that is necessary to balance two opposing vectorial transport systems: the inflow of newly synthesized TH and the outflow of TH by external secretion.


Assuntos
Cristalinas/metabolismo , Transportadores de Ácidos Monocarboxílicos/metabolismo , Tireoglobulina/farmacologia , Glândula Tireoide/efeitos dos fármacos , Animais , Linhagem Celular , Cristalinas/genética , Relação Dose-Resposta a Droga , Expressão Gênica/efeitos dos fármacos , Transportadores de Ácidos Monocarboxílicos/genética , Ratos , Glândula Tireoide/metabolismo , Fatores de Tempo , Cristalinas mu
8.
BMC Endocr Disord ; 14: 52, 2014 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-24990570

RESUMO

BACKGROUND: Promyelocytic leukaemia zinc finger (PLZF) is a transcriptional repressor that was originally isolated from a patient with promyelocytic leukaemia. PLZF also affects key elements for cell cycle progression, such as cyclin A, and can affect the tumourigenicity of various cancers. Thus far, the behaviour of PLZF in thyroid carcinoma remains unclear. METHODS: We analysed the expression profile of PLZF in different types of benign and malignant thyroid lesions as well as in normal thyroid tissue. Specifically, we examined PLZF expression in normal thyroid (N; n = 4), adenomatous lesion (AL; n = 5), follicular adenoma (FA; n = 2), papillary thyroid carcinoma (PTC; n = 20), and anaplastic thyroid carcinoma (ATC; n = 3) samples. PLZF expression was estimated by western blotting and immunohistochemical (IHC) staining. RESULTS: PLZF was expressed in all samples of thyroid lesions examined. In N, AL, and FA, PLZF was mainly localized in the nucleus. In contrast, in PTC and ATC, PLZF was mainly expressed in the cytosol with high intensity. In more detail, the cytoplasmic IHC scores in PTC with capsular invasion (CI) and lymph node (LN) metastasis were higher than those in PTC without CI and LN metastasis. CONCLUSIONS: PLZF shows different subcellular localizations among PTC, ATC, and other thyroid lesions. Furthermore, high cytoplasmic expression of PLZF may be correlated with CI and LN metastasis in thyroid carcinoma. The present report is the first to describe the implications of intracellular PLZF expression in thyroid carcinomas.


Assuntos
Adenoma/metabolismo , Carcinoma Papilar/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , Glândula Tireoide/metabolismo , Neoplasias da Glândula Tireoide/metabolismo , Adenoma/patologia , Adulto , Idoso , Western Blotting , Carcinoma Papilar/secundário , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Feminino , Humanos , Técnicas Imunoenzimáticas , Metástase Linfática , Masculino , Pessoa de Meia-Idade , Invasividade Neoplásica , Estadiamento de Neoplasias , Prognóstico , Proteína com Dedos de Zinco da Leucemia Promielocítica , Neoplasias da Glândula Tireoide/patologia
9.
Pacing Clin Electrophysiol ; 37(7): 853-63, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24499369

RESUMO

BACKGROUND: A KCNE1 polymorphism, D85N, causes long QT syndrome (LQTS) with a decrease in the slowly activating delayed-rectifier K(+) channel current (IKs ). We examined impacts of D85N polymorphism on KCNE1 protein stability and functions, and tested the ability of various drugs to modify them. METHODS: KCNE1-D85N or the wild-type protein was coexpressed in COS7 cells with KCNQ1 to form K(+) channels. Expression, degradation, and intracellular localization of KCNE1 proteins, as well as the currents conferred by KCNQ1/KCNE1 complexes, were determined using immunoblots, immunofluorescence, and patch-clamp techniques. RESULTS: The protein level of KCNE1-D85N was lower than that of the wild-type, in spite of the comparable levels of their mRNA. KCNE1-D85N was highly ubiquitinated and rapidly degraded as compared to the wild-type; a proteasome inhibitor, MG132, inhibited its degradation and increased its steady-state level. Both KCNE1-D85N and the wild-type proteins were co-immunoprecipitated with KCNQ1. Immunofluorescent signals of KCNE1-D85N accumulated in the endoplasmic reticulum and Golgi apparatus, with reduced levels on the cell membrane. Patch-clamp experiments demonstrated that the membrane current corresponding to IKs was much smaller in cells expressing KCNE1-D85N than in those expressing the wild-type. Verapamil (0.5-10 µM) increased the protein level of KCNE1-D85N, decreased its ubiquitination, slowed its degradation, and enhanced KCNQ1/KCNE1-D85N channel currents. Pretreatment with amiodarone abolished these effects of verapamil. CONCLUSION: KCNE1-D85N is less stable than the wild-type protein, and is rapidly degraded through the ubiquitin-proteasome system. Verapamil may be of a therapeutic value in LQTS patients via preventing degradation of KCNE1-D85N.


Assuntos
Bloqueadores dos Canais de Cálcio/farmacologia , Bloqueadores dos Canais de Cálcio/uso terapêutico , Síndrome do QT Longo/tratamento farmacológico , Síndrome do QT Longo/genética , Polimorfismo Genético , Canais de Potássio de Abertura Dependente da Tensão da Membrana/efeitos dos fármacos , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Verapamil/farmacologia , Verapamil/uso terapêutico , Células Cultivadas , Humanos
10.
Clin Exp Hypertens ; 36(7): 447-53, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24433018

RESUMO

PURPOSE: To examine effects of a long-acting calcium channel blocker (CCB) azelnidipine on uric acid metabolism in hypertensive patients. METHODS: Azelnidipine was administered to 72 patients at a daily dose of 8 mg or 16 mg. In 22 cases out of the 72 patients, a different CCB was switched to azelnidipine. Blood pressure was measured and biochemical parameters of blood and urine were evaluated before and 2-3 months after the administration. RESULTS: Azelnidipine significantly decreased both systolic and diastolic blood pressure and the heart rate. It decreased both serum urate levels and the urinary uric acid to creatinine ratio (Uur/Ucr), but did not affect the uric acid clearance to creatinine clearance ratio (Cur/Ccr). Azelnidipine decreased both Uur/Ucr and Cur/Ccr in patients with Uur/Ucr ≥ 0.5 or ≥ 0.34, although it did not change these clearance parameters in patients with Uur/Ucr <0.5 or <0.34. Azelnidipine decreased the serum urate levels and Uur/Ucr in hyperuricemic patients with uric acid levels ≥ 7.0 mg/dL in males and ≥ 6.0 mg/dL in females. It did not change these parameters in normouricemic patients with serum urate levels <7.0 mg/dL in males and <6.0 mg/dL in females. Azelnidipine decreased Uur/Ucr and Cur/Ccr in hyperuricemic patients with normal or over excretion of uric acid, although it did not change these clearance parameters in hyperuricemic patients with uric acid hypoexcretion. CONCLUSIONS: Azelnidipine decreased the serum urate acid levels and Uur/Ucr, and this response was most prominent in hyperuricemic patients or patients with normal and over excretion of uric acid.


Assuntos
Ácido Azetidinocarboxílico/análogos & derivados , Bloqueadores dos Canais de Cálcio/uso terapêutico , Di-Hidropiridinas/uso terapêutico , Hipertensão/tratamento farmacológico , Hipertensão/metabolismo , Hiperuricemia/tratamento farmacológico , Ácido Úrico/metabolismo , Idoso , Idoso de 80 Anos ou mais , Ácido Azetidinocarboxílico/uso terapêutico , Pressão Sanguínea/efeitos dos fármacos , Creatinina/metabolismo , Hipertensão Essencial , Feminino , Humanos , Hipertensão/complicações , Hiperuricemia/complicações , Hiperuricemia/metabolismo , Masculino , Ácido Úrico/sangue , Ácido Úrico/urina
11.
Biochem Biophys Res Commun ; 434(1): 131-6, 2013 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-23542464

RESUMO

Remote reperfusion lung injury following skeletal muscle ischemia and reperfusion accounts for high morbidity and mortality. AMP deaminase (AMPD), a key enzyme for nucleotide cycle, has been implicated in the regulation of this phenomenon. However, the function of Ampd2 and Ampd3 subtype has not been elucidated in remote reperfusion rodent lung injury. We utilized AMPD3 and AMPD2-deficient mice. The two types of AMPD-deficient mice and wild-type (WT) littermates were subjected to ischemia-reperfusion injury. After 3h bilateral hind-limb ischemia and reperfusion, AMPD3 mRNA, AMPD activity and inosine monophosphate (IMP) increased significantly in WT and AMPD2-deficient mice lungs, while they did not show significant alterations in AMPD3-deficient mice lungs. Genetic inactivation of Ampd3 resulted in markedly accelerated myeloperoxidase (MPO) activity along with exaggerated neutrophils infiltration and hemorrhage in the lungs compared to WT and AMPD2-deficient mice, furthermore, IMP treatment significantly attenuated MPO activity and neutrophils infiltration in WT and the two types of AMPD-deficient mice lungs after 3h reperfusion. These findings demonstrate for the first time in AMP-deficient mice models that AMPD3 plays a critical role in remote reperfusion lung injury via generation of IMP and validate the potential to use IMP into the clinical arena to attenuate remote ischemia-reperfusion lung injury.


Assuntos
AMP Desaminase/fisiologia , Lesão Pulmonar/enzimologia , Traumatismo por Reperfusão/enzimologia , AMP Desaminase/deficiência , AMP Desaminase/genética , Animais , Modelos Animais de Doenças , Inosina Monofosfato/administração & dosagem , Inosina Monofosfato/biossíntese , Lesão Pulmonar/genética , Lesão Pulmonar/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/patologia
12.
Clin Endocrinol (Oxf) ; 79(1): 35-42, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23215816

RESUMO

BACKGROUND: Thyroid nodules are common among adults, and accurate diagnosis is critical in for management decisions. Ultrasound and fine needle aspiration cytology are the most common methods to evaluate nodules, but they are not practical for screening large numbers of patients because of cost and time considerations. OBJECTIVE: The aim of this study was to isolate an autoantibody to tumour antigen, WD repeat domain 1 (WDR1), and evaluate its diagnostic sensitivity and specificity for thyroid neoplasms. PATIENTS AND METHODS: We investigated serological biomarkers in patients with thyroid carcinoma who had a poor prognosis. Using a serological analysis of recombinant cDNA expression cloning (SEREX) strategy, we isolated WDR1 and its specific autoantibody in the sera of patients with undifferentiated thyroid carcinoma (UTC). We examined using indirect ELISA, the titre of the anti-WDR1 antibody (AWA) in 54 study patients: 10 with UTC, 20 with papillary thyroid carcinoma (PTC), 17 with benign thyroid nodule (BTN), 7 with autoimmune thyroid disease (AITD), as well as 38 controls (N). RESULTS: WDR1 was ubiquitously expressed in various types of thyroid tissues. However, the titre of AWA in UTC and PTC was significantly higher than that in BTN, AITD and N (P < 0·001). No significant correlation was observed between thyroid function, serum thyroglobulin and tumour diameter. The cut-off value estimated using ROC to differentiate malignancies from others was 0·95 (sensitivity 96·7%, specificity 91·9%, AUC 0·969, P < 0·001). CONCLUSIONS: Anti-WDR1 antibody could be a novel approach for serological screening of PTC and UTC, and could be an efficient and inexpensive biomarker.


Assuntos
Autoanticorpos/imunologia , Biomarcadores Tumorais/imunologia , Proteínas dos Microfilamentos/imunologia , Neoplasias da Glândula Tireoide/imunologia , Animais , Autoanticorpos/sangue , Biomarcadores Tumorais/sangue , Biomarcadores Tumorais/genética , Northern Blotting , Carcinoma/diagnóstico , Carcinoma/genética , Carcinoma/imunologia , Carcinoma Papilar , Linhagem Celular , DNA Complementar/química , DNA Complementar/genética , Diagnóstico Diferencial , Ensaio de Imunoadsorção Enzimática , Feminino , Regulação Neoplásica da Expressão Gênica , Biblioteca Gênica , Humanos , Masculino , Proteínas dos Microfilamentos/sangue , Proteínas dos Microfilamentos/genética , Curva ROC , Análise de Sequência de DNA , Tireoglobulina/sangue , Câncer Papilífero da Tireoide , Neoplasias da Glândula Tireoide/diagnóstico , Neoplasias da Glândula Tireoide/genética , Nódulo da Glândula Tireoide/diagnóstico , Nódulo da Glândula Tireoide/genética , Nódulo da Glândula Tireoide/imunologia
13.
Circ Res ; 108(4): 458-68, 2011 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-21183741

RESUMO

RATIONALE: The human ether-a-go-go-related gene (hERG) encodes the α subunit of the potassium current I(Kr). It is highly expressed in cardiomyocytes and its mutations cause long QT syndrome type 2. Heat shock protein (Hsp)70 is known to promote maturation of hERG. Hsp70 and heat shock cognate (Hsc70) 70 has been suggested to play a similar function. However, Hsc70 has recently been reported to counteract Hsp70. OBJECTIVE: We investigated whether Hsc70 counteracts Hsp70 in the control of wild-type and mutant hERG stability. METHODS AND RESULTS: Coexpression of Hsp70 with hERG in HEK293 cells suppressed hERG ubiquitination and increased the levels of both immature and mature forms of hERG. Immunocytochemistry revealed increased levels of hERG in the endoplasmic reticulum and on the cell surface. Electrophysiological studies showed increased I(Kr). All these effects of Hsp70 were abolished by Hsc70 coexpression. Heat shock treatment of HL-1 mouse cardiomyocytes induced endogenous Hsp70, switched mouse ERG associated with Hsc70 to Hsp70, increased I(Kr), and shortened action potential duration. Channels with disease-causing missense mutations in intracellular domains had a higher binding capacity to Hsc70 than wild-type channels and channels with mutations in the pore region. Knockdown of Hsc70 by small interfering RNA or heat shock prevented degradation of mutant hERG proteins with mutations in intracellular domains. CONCLUSIONS: These results indicate reciprocal control of hERG stability by Hsp70 and Hsc70. Hsc70 is a potential target in the treatment of LQT2 resulting from missense hERG mutations.


Assuntos
Canais de Potássio Éter-A-Go-Go/genética , Canais de Potássio Éter-A-Go-Go/metabolismo , Proteínas de Choque Térmico HSC70/metabolismo , Proteínas de Choque Térmico HSP70/metabolismo , Síndrome do QT Longo/genética , Síndrome do QT Longo/metabolismo , Mutação de Sentido Incorreto/genética , Potenciais de Ação/fisiologia , Animais , Membrana Celular/metabolismo , Células Cultivadas , Modelos Animais de Doenças , Técnicas Eletrofisiológicas Cardíacas , Retículo Endoplasmático/metabolismo , Canais de Potássio Éter-A-Go-Go/farmacologia , Células HEK293 , Resposta ao Choque Térmico/fisiologia , Humanos , Camundongos , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , RNA Interferente Pequeno/farmacologia
14.
Circ J ; 76(12): 2875-83, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22972200

RESUMO

BACKGROUND: The prion protein (PrP) has been reported to serve as a surface maker for isolation of cardiomyogenic progenitors from murine embryonic stem (ES) cells. Although PrP-positive cells exhibited automaticity, their electrophysiological characteristics remain unresolved. The aim of the present study was therefore to investigate the electrophysiological properties of PrP-positive cells in comparison with those of HCN4p-or Nkx2.5-positive cells. METHODS AND RESULTS: Differentiation of AB1, HCN5p-EGFP and hcgp7 ES cells into cardiac progenitors was induced by embryoid body (EB) formation. EBs were dissociated and cells expressing PrP, HCN4-EGFP and/or Nkx2.5-GFP were collected via flow cytometry. Sorted cells were subjected to reverse transcriptase-polymerase chain reaction, immunostaining and patch-clamp experiments. PrP-positive cells expressed mRNA of undifferentiation markers, first and second heart field markers, and cardiac-specific genes and ion channels, indicating their commitment to cardiomyogenic progenitors. PrP-positive cells with automaticity showed positive and negative chronotropic responses to isoproterenol and carbamylcholine, respectively. Hyperpolarization-activated cation current (I(f)) was barely detectable, whereas Na(+) and L-type Ca(2+) channel currents were frequently observed. Their spontaneous activity was slowed by inhibition of sarcoplasmic reticulum Ca(2+) uptake and release but not by blocking I(f). The maximum diastolic potential of their spontaneous firings was more depolarized than that of Nkx2.5-GFP-positive cells. CONCLUSIONS: PrP-positive cells contained cardiac progenitors that separated from the lineage of sinoatrial node cells. PrP can be used as a marker to enrich nascent cardiac progenitors.


Assuntos
Potenciais de Ação , Células-Tronco Embrionárias/metabolismo , Miócitos Cardíacos/metabolismo , Príons/metabolismo , Animais , Biomarcadores/metabolismo , Diferenciação Celular , Linhagem Celular , Linhagem da Célula , Separação Celular/métodos , Técnicas de Cocultura , Canais de Cátion Regulados por Nucleotídeos Cíclicos/genética , Canais de Cátion Regulados por Nucleotídeos Cíclicos/metabolismo , Citometria de Fluxo , Regulação da Expressão Gênica no Desenvolvimento , Imuno-Histoquímica , Camundongos , Camundongos da Linhagem 129 , Contração Miocárdica , Técnicas de Patch-Clamp , Periodicidade , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transfecção
15.
Clin Exp Hypertens ; 34(7): 470-3, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22681507

RESUMO

The effects of cilnidipine on the serum uric acid level and urinary NO excretion in hypertensive patients were investigated. Blood and urine samples of 16 hypertensive outpatients were collected before and 2 months after cilnidipine therapy (10 mg). The serum uric acid level decreased significantly after cilnidipine treatment, while the uric acid-creatinine clearance ratio was unaffected. The cilnidipine medication produced a significant increase in urinary NO excretion, although amlodipine did not change it significantly. Therefore, cilnidipine has a profound antihypertensive effect and may reduce the serum uric acid level and increase NO production in the kidney.


Assuntos
Bloqueadores dos Canais de Cálcio/uso terapêutico , Di-Hidropiridinas/uso terapêutico , Hipertensão/tratamento farmacológico , Óxido Nítrico/urina , Ácido Úrico/sangue , Idoso , Idoso de 80 Anos ou mais , Anlodipino/uso terapêutico , Feminino , Humanos , Hipertensão/sangue , Hipertensão/urina , Rim/fisiopatologia , Masculino , Pessoa de Meia-Idade , Resultado do Tratamento
16.
Biochem Biophys Res Commun ; 393(2): 286-90, 2010 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-20132794

RESUMO

Although it is well known that an excess of iodide suppresses thyroid function and blood flow in vivo, the underlying molecular mechanisms are not fully known. The functional effect of iodide occurs at multiple steps, which include inhibition of sodium/iodide symporter (NIS) expression, transient block of organification, and inhibition of hormonal release. The vascular effect likely involves suppression of the vascular endothelial growth factor (VEGF) gene. In this report, we show that excess iodide coordinately suppresses the expression of the NIS and VEGF genes in FRTL-5 thyroid cells. We also demonstrate that the mechanism of iodide suppression of NIS gene expression is transcriptional, which is synergized by the addition of thyroglobulin. Based on the findings of reporter gene assays and electrophoretic gel mobility shift analysis, we also report two novel DNA binding proteins that responded specifically to iodide and modulated NIS promoter activity. The results suggest that excess iodide affects thyroid vascular function in addition to iodide uptake. This study provides additional insights into the mechanism of action of excess iodide on thyroid function.


Assuntos
Iodetos/farmacologia , Simportadores/genética , Glândula Tireoide/efeitos dos fármacos , Transcrição Gênica/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/genética , Animais , Linhagem Celular , Ensaio de Desvio de Mobilidade Eletroforética , Iodetos/metabolismo , Ratos , Simportadores/antagonistas & inibidores , Tireoglobulina/metabolismo , Tireoglobulina/farmacologia , Glândula Tireoide/metabolismo , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores
17.
Circ Rep ; 2(8): 425-432, 2020 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-33693264

RESUMO

Background: Monocarboxylate transporter 9 (MCT9), an orphan transporter member of the solute carrier family 16 (SLC16), possibly reabsorbs uric acid in the renal tubule and has been suggested by genome-wide association studies to be involved in the development of hyperuricemia and gout. In this study we investigated the mechanisms regulating the expression of human (h) MCT9, its degradation, and physiological functions. Methods and Results: hMCT9-FLAG was stably expressed in HEK293 cells and its degradation, intracellular localization, and urate uptake activities were assessed by pulse-chase analysis, immunofluorescence, and [14C]-urate uptake experiments, respectively. hMCT9-FLAG was localized on the plasma membrane as well as in the endoplasmic reticulum and Golgi apparatus. The proteasome inhibitors MG132 and lactacystine increased levels of hMCT9-FLAG protein expression with enhanced ubiquitination, prolonged their half-life, and decreased [14C]-urate uptake. [14C]-urate uptake was increased by both heat shock (HS) and the HS protein inducer geranylgeranylacetone (GGA). Both HS and GGA restored the [14C]-urate uptake impaired by MG132. Conclusions: hMCT9 does transport urate and is degraded by a proteasome, inhibition of which reduces hMCT9 expression on the cell membrane and urate uptake. HS enhanced urate uptake through hMCT9.

18.
J Clin Endocrinol Metab ; 94(2): 442-8, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19050049

RESUMO

CONTEXT: Pendrin is an apical protein of thyroid follicular cells, responsible for the efflux of iodide into the follicular lumen via an iodide-chloride transport mechanism. It is unknown whether pendrin is recognized by autoantibodies. OBJECTIVE: Our objective was to examine the prevalence of pendrin antibodies in autoimmune thyroid diseases and compare with that of thyroglobulin, thyroperoxidase, TSH receptor, and sodium iodide symporter antibodies. DESIGN: In a prevalent case-control study, we analyzed the sera of 140 autoimmune thyroid disease cases (100 with Graves' disease and 40 with Hashimoto's thyroiditis) and 80 controls (50 healthy subjects, 10 patients with papillary thyroid cancer, 10 with systemic lupus erythematosus, and 10 with rheumatoid arthritis). Pendrin antibodies were measured by immunoblotting using extract of COS-7 cells transfected with pendrin and a rabbit polyclonal pendrin antibody. RESULTS: Pendrin antibodies were found in 81% of the cases and 9% of controls (odds ratio = 44; P < 0.0001). Among cases, pendrin antibodies were more frequent and of higher titers in Hashimoto's thyroiditis than in Graves' disease. Pendrin antibodies correlated significantly with thyroglobulin, thyroperoxidase, and sodium iodide symporter antibodies but not with TSH receptor antibodies. Pendrin antibodies were equally effective as thyroglobulin and thyroperoxidase antibodies in diagnosis of autoimmune thyroid diseases, especially Hashimoto's thyroiditis. CONCLUSIONS: The study identifies pendrin as a novel autoantigen recognized by patients with autoimmune thyroid diseases and proposes the use of pendrin antibodies as an accurate diagnostic tool.


Assuntos
Autoanticorpos/sangue , Autoantígenos/imunologia , Proteínas de Membrana Transportadoras/imunologia , Tireoidite Autoimune/sangue , Tireoidite Autoimune/imunologia , Animais , Reações Antígeno-Anticorpo , Autoantígenos/sangue , Autoantígenos/isolamento & purificação , Células COS , Estudos de Casos e Controles , Chlorocebus aethiops , Humanos , Iodeto Peroxidase/imunologia , Proteínas de Ligação ao Ferro/imunologia , Proteínas de Membrana Transportadoras/genética , Sensibilidade e Especificidade , Estudos Soroepidemiológicos , Transportadores de Sulfato , Simportadores/imunologia , Tireoglobulina/imunologia , Tireoidite Autoimune/diagnóstico
19.
Am J Physiol Heart Circ Physiol ; 297(4): H1329-36, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19666845

RESUMO

Autologous cell implantation and angiogenic gene therapy have been evaluated in critical limb ischemic patients. Here, we compared the features of these strategies individually and in combination. C57BL/6J mice with ischemic hindlimbs were injected with adherent mononuclear cells (aMNCs) from bone marrow or adenovirus encoding the hepatocyte growth factor (HGF) gene (Ad-HGF). Under comparable angiogenic conditions, 10 x 10(5) aMNCs produced significantly higher amounts of VEGF and FGF-2 and stimulated the number of arterioles in ischemic muscle compared with 1 x 10(8) plaque-forming units (pfu) of Ad-HGF. Ad-HGF produced 10 times more HGF in ischemic muscle compared with aMNCs. Injection of 0.3 x 10(5) aMNCs previously transfected with Ad-HGF (aMNC/Ad-HGF) increased blood flow and elevated the numbers of capillaries and arterioles to levels comparable with that seen with 10 x 10(5) aMNCs or 1 x 10(8) pfu of Ad-HGF. Hypoxic conditions induced the apoptotic death of aMNCs. However, coincubation with HGF or aMNC/Ad-HGF protected cells against apoptosis. HGF stimulated the migration of aMNCs, and the migration capacity of the aMNC/Ad-HGF group was significantly higher than that in the aMNC/Ad-LacZ group. In conclusion, cell-based HGF gene therapy decreased the number of cells required for neovascularization. This strategy can be an effective angiogenic therapy.


Assuntos
Transplante de Medula Óssea , Terapia Genética/métodos , Fator de Crescimento de Hepatócito/biossíntese , Isquemia/terapia , Leucócitos Mononucleares/transplante , Músculo Esquelético/irrigação sanguínea , Neovascularização Fisiológica , Adenoviridae/genética , Animais , Apoptose , Hipóxia Celular , Movimento Celular , Células Cultivadas , Terapia Combinada , Modelos Animais de Doenças , Fator 2 de Crescimento de Fibroblastos/metabolismo , Vetores Genéticos , Fator de Crescimento de Hepatócito/genética , Membro Posterior , Isquemia/genética , Isquemia/metabolismo , Isquemia/fisiopatologia , Leucócitos Mononucleares/metabolismo , Leucócitos Mononucleares/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Recuperação de Função Fisiológica , Fluxo Sanguíneo Regional , Fatores de Tempo , Transfecção , Transplante Autólogo , Fator A de Crescimento do Endotélio Vascular/metabolismo
20.
Biochem Biophys Res Commun ; 389(2): 269-73, 2009 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-19716808

RESUMO

Thyroglobulin (Tg) is an essential substrate for thyroid hormone biosynthesis whose production is primarily limited to the thyroid follicular cell. We have previously identified an approximately 1.2 kb fragment of Tg mRNA in cultured mouse mesangial cells, and in the present study provide evidence showing that this transcript is transcribed and translated into a unique protein (kTg) in the kidney, but not the thyroid gland. Cloning of kTg from a mouse kidney cDNA library showed that transcription starts in the middle of intron 41 of the Tg gene and continues in-frame with the remaining coding sequence of thyroid-derived Tg beginning with exon 42. Translation of this mRNA is predicted to yield a protein of 367 amino acids (40 kDa) containing a unique 13 amino acid sequence serving as a signal peptide followed by a 354 amino acid segment identical to the carboxy-terminal end of thyroid Tg. Western blot analysis with an antibody directed against the C-terminus of thyroid Tg detected a 40 kDa protein expressed in the kidney. Immunohistochemistry with this antibody showed that immunoreactive Tg was localized in podocytes and the mesangial area of the renal glomerulus. A part of a homologous transcript was also detected in human kidney, and the kTg protein was recognized by sera from Hashimoto's thyroiditis but not from controls. Together these results suggest that a unique low molecular weight variant of Tg is expressed in the kidney, where it could serve both physiological and pathological roles, including that of an autoantigen.


Assuntos
Glomérulos Renais/imunologia , Tireoglobulina/imunologia , Sequência de Aminoácidos , Animais , Autoantígenos/genética , Autoantígenos/imunologia , Clonagem Molecular , Biblioteca Gênica , Humanos , Camundongos , Dados de Sequência Molecular , Tireoglobulina/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA