Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
J Biol Chem ; 300(3): 105732, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38336290

ABSTRACT

The manganese (Mn) export protein SLC30A10 is essential for Mn excretion via the liver and intestines. Patients with SLC30A10 deficiency develop Mn excess, dystonia, liver disease, and polycythemia. Recent genome-wide association studies revealed a link between the SLC30A10 variant T95I and markers of liver disease. The in vivo relevance of this variant has yet to be investigated. Using in vitro and in vivo models, we explore the impact of the T95I variant on SLC30A10 function. While SLC30A10 I95 expressed at lower levels than T95 in transfected cell lines, both T95 and I95 variants protected cells similarly from Mn-induced toxicity. Adeno-associated virus 8-mediated expression of T95 or I95 SLC30A10 using the liver-specific thyroxine binding globulin promoter normalized liver Mn levels in mice with hepatocyte Slc30a10 deficiency. Furthermore, Adeno-associated virus-mediated expression of T95 or I95 SLC30A10 normalized red blood cell parameters and body weights and attenuated Mn levels and differential gene expression in livers and brains of mice with whole body Slc30a10 deficiency. While our in vivo data do not indicate that the T95I variant significantly compromises SLC30A10 function, it does reinforce the notion that the liver is a key site of SLC30A10 function. It also supports the idea that restoration of hepatic SLC30A10 expression is sufficient to attenuate phenotypes in SLC30A10 deficiency.


Subject(s)
Amino Acid Substitution , Cation Transport Proteins , Dependovirus , Liver , Manganese , Mutation , Animals , Mice , Body Weight , Brain/metabolism , Cation Transport Proteins/deficiency , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Cell Line , Dependovirus/genetics , Erythrocytes , Genome-Wide Association Study , Hepatocytes/metabolism , Liver/cytology , Liver/metabolism , Liver Diseases/genetics , Liver Diseases/metabolism , Manganese/metabolism , Manganese Poisoning/metabolism , Phenotype , Promoter Regions, Genetic , Thyroxine-Binding Globulin/genetics
2.
Endocrinol Metab (Seoul) ; 37(6): 870-878, 2022 12.
Article in English | MEDLINE | ID: mdl-36475360

ABSTRACT

BACKGRUOUND: Thyroxine-binding globulin (TBG) is a major transporter protein for thyroid hormones. The serpin family A member 7 (SERPINA7) gene codes for TBG, and mutations of the SERPINA7 gene result in TBG deficiency. Although more than 40 mutations have been reported in several countries, only a few studies of TBG deficiency and SERPINA7 gene mutation have been performed in Korea. The aim of this study is to review the clinical presentations and laboratory findings of patients with TBG deficiency and to investigate the types of SERPINA7 gene mutation. METHODS: Five unrelated Korean adults with TBG deficiency attending endocrinology clinic underwent SERPINA7 gene sequencing. Four patients harbored a SERPINA7 gene mutation. Serum thyroid hormones, anti-microsomal antibodies, and TBG were measured. Genomic DNA was extracted from whole blood. All exons and intron-exon boundaries of the TBG gene were amplified and sequencing was performed. RESULTS: Two patients were heterozygous females, and the other two were hemizygous males. One heterozygous female had coexisting hypothyroidism. The other heterozygous female was erroneously prescribed levothyroxine at a local clinic. One hemizygous male harbored a novel mutation, p.Phe269Cysfs*18, which caused TBG partial deficiency. Three patients had the p.Leu372Phefs*23 mutation, which is known as TBG-complete deficiency Japan (TBG-CDJ) and was also presented in previous mutation analyses in Korea. CONCLUSION: This study presents four patients diagnosed with TBG deficiency and provides the results of SERPINA7 gene sequencing. One novel mutation, p.Phe269Cysfs*18, causing TBD-partial deficiency and three cases of TBG-CDJ were demonstrated. It is necessary to identify TBG deficiency to prevent improper treatment. Also, sequencing of the SERPINA7 gene would provide valuable information about the TBG variants in Korea.


Subject(s)
Thyroxine-Binding Globulin , Thyroxine-Binding Proteins , Adult , Humans , Male , Female , Thyroxine-Binding Globulin/genetics , Thyroxine-Binding Globulin/metabolism , Thyroxine-Binding Proteins/genetics , Thyroxine-Binding Proteins/metabolism , Mutation , Republic of Korea/epidemiology
3.
Int J Mol Sci ; 23(20)2022 Oct 21.
Article in English | MEDLINE | ID: mdl-36293546

ABSTRACT

Adeno-associated virus (AAV) vector-based therapies can effectively correct some disease pathology in murine models with mucopolysaccharidoses. However, immunogenicity can limit therapeutic effect as immune responses target capsid proteins, transduced cells, and gene therapy products, ultimately resulting in loss of enzyme activity. Inherent differences in male versus female immune response can significantly impact AAV gene transfer. We aim to investigate sex differences in the immune response to AAV gene therapies in mice with mucopolysaccharidosis IVA (MPS IVA). MPS IVA mice, treated with different AAV vectors expressing human N-acetylgalactosamine 6-sulfate sulfatase (GALNS), demonstrated a more robust antibody response in female mice resulting in subsequent decreased GALNS enzyme activity and less therapeutic efficacy in tissue pathology relative to male mice. Under thyroxine-binding globulin promoter, neutralizing antibody titers in female mice were approximately 4.6-fold higher than in male mice, with GALNS enzyme activity levels approximately 6.8-fold lower. Overall, male mice treated with AAV-based gene therapy showed pathological improvement in the femur and tibial growth plates, ligaments, and articular cartilage as determined by contrasting differences in pathology scores compared to females. Cardiac histology revealed a failure to normalize vacuolation in females, in contrast, to complete correction in male mice. These findings promote the need for further determination of sex-based differences in response to AAV-mediated gene therapy related to developing treatments for MPS IVA.


Subject(s)
Chondroitinsulfatases , Mucopolysaccharidoses , Mucopolysaccharidosis IV , Humans , Female , Mice , Male , Animals , Thyroxine-Binding Globulin/genetics , Thyroxine-Binding Globulin/metabolism , Disease Models, Animal , Sex Characteristics , Capsid Proteins/genetics , Genetic Therapy , Antibodies, Neutralizing/therapeutic use , Gene Expression , Chondroitinsulfatases/genetics
4.
J Endocrinol Invest ; 45(4): 731-739, 2022 Apr.
Article in English | MEDLINE | ID: mdl-34761328

ABSTRACT

PURPOSE: This study presents a case of familial transmission of thyroxine-binding globulin (TBG) deficiency. The SERPINA7-gene which codes for TBG is located on the X-chromosome (Xq21-22). More than 45 mutations have been reported to cause TBG- deficiency from various countries, but none from India so far. Genetic analysis of SERPINA7 gene was carried out to determine the cause of low TBG levels in one family. METHODS: DNA samples of the propositus and the family members were subjected to Polymerase Chain Reaction (PCR) followed by direct sequencing. Allele-specific PCR and Next-gen sequencing (NGS) were employed to confirm the site of the mutation. Thyroid function tests were estimated by Radioimmunoassay (RIA) and Immunoradiometric assay (IRMA) kits. X-chromosomal inactivation status was analyzed in the female members harboring the mutation. RESULTS: A mutational screening in this family revealed a novel frame-shift mutation S353Q, 354fs3X in the exon 4 of the SERPINA7 gene which will be referred to as TBG-complete deficiency-India (TBG-CD-Ind). One out of four female family members harboring the mutation showed selective X-chromosomal inactivation. The affected family members were clinically euthyroid initially, showed changes in the thyroid function when tested after a long time span. However, the changes in the thyroid function in the affected family members had an autoimmune etiology. CONCLUSION: This study presents the first report of TBG-CD from India wherein a novel frameshift mutation referred to as TBG-CD-Ind (S353Q, 354fs3X) in the SERPINA7 gene was detected. No apparent association was identified between thyroid function and the TBG-mutation in the affected subjects. A detailed biochemical and genomic testing to determine the exact cause of discordant TFT in the patients would certainly aid in the unequivocal diagnosis of the thyroid function and for the precise individualized treatment.


Subject(s)
Thyroxine-Binding Globulin/analysis , Thyroxine-Binding Globulin/deficiency , Thyroxine-Binding Globulin/genetics , Adult , Female , Genetic Diseases, X-Linked/blood , Genetic Diseases, X-Linked/complications , Genetic Diseases, X-Linked/genetics , Humans , India , Male , Polymerase Chain Reaction/methods , Polymerase Chain Reaction/statistics & numerical data , Thyroid Function Tests/methods , Thyroid Function Tests/statistics & numerical data , Exome Sequencing/methods , Exome Sequencing/statistics & numerical data
5.
Mol Med Rep ; 25(1)2022 01.
Article in English | MEDLINE | ID: mdl-34779500

ABSTRACT

The recombinant adeno­associated virus 8 (rAAV8) vector is a widely used tool in basic research and clinical trials. The cytomegalovirus immediate­early enhancer/chicken ß­actin (CAG) promoter is a synthetic promoter used in adenoviral constructs with a wide spectrum and notable efficiency. The thyroxine binding globulin (TBG) promoter is a liver­specific promoter, which directs transgene expression in hepatocytes. However, the transduction efficiency of the rAAV vector is dependent on both the administration routes and the promoter elements. In the present study, the transduction efficiency in the liver following intraperitoneal (IP) and intravenous (IV) injections of rAAV8 with the CAG, TBG669 and TBG410 promoters was compared. Enhanced green fluorescent protein (EGFP) expression was used as the biomarker to indicate efficiency. Among the three different promoters, CAG exhibited the highest efficiency from both IV and IP injections. Following IV administration, EGFP expression, induced by the CAG promoter, was 67­fold higher compared with that in the TBG410 promoter group and 26­fold higher compared with that in the TBG669 promoter group. EGFP protein expression was higher with IV injection compared with that for IP injection for both the CAG and TBG669 promoters (P<0.05). With the CAG promoter, EGFP protein expression was 1.5­fold higher with the use of IV injection than with IP injection. With the TBG410 promoter, no differences were observed between the two administrations. In conclusion, these findings demonstrated that the CAG promoter was much more efficient at driving gene expression in the liver compared with that for the TBG promoters in rAAV8. In addition, IP administration produced comparable efficiency for gene delivery via the rAAV8 vector, particularly with the promoter TBG410.


Subject(s)
Dependovirus/genetics , Promoter Regions, Genetic/genetics , Transduction, Genetic/methods , Actins/genetics , Animals , Antigens, Viral/genetics , China , Dependovirus/metabolism , Gene Expression/genetics , Gene Expression Regulation/genetics , Genetic Vectors/genetics , Hepatocytes/metabolism , Immediate-Early Proteins/genetics , Liver/metabolism , Male , Mice , Mice, Inbred ICR , Thyroxine-Binding Globulin/genetics , Transgenes/genetics
6.
Pharmazie ; 76(9): 428-430, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34481533

ABSTRACT

To explore an unusual cause of the decrease of T3/T4 through a new mutation of TBG gene in a family, so as to avoid habitual thinking and reduce subsequent over treatment. TSH, free total T4, T3 and free T4, T3 were determined by automatic chemiluminescence immunoassay. The TBG mutation was identified by direct DNA sequencing. A frameshift mutation of p. l372ffs * 32 was found in the TBG gene (c.1114delc) of the patient by direct DNA sequencing, and the proband of the family was heterozygous. In vitro expression showed that the affinity of TBG for T4 decreased. Further examination of the family members showed that T3 and T4 were decreased, while FT3, FT4 and TSH were normal. If the patients with low TT4 and TT3 but normal TSH are found, the serum TBG level and related genes should be detected to determine whether it is TBG deficiency and avoid wrong treatment.


Subject(s)
Thyroglobulin , Triiodothyronine , Base Sequence , Humans , Mutation/genetics , Thyroglobulin/genetics , Thyrotropin/genetics , Thyroxine/genetics , Thyroxine-Binding Globulin/genetics , Triiodothyronine/genetics
7.
Eur J Endocrinol ; 185(5): 743-753, 2021 Oct 11.
Article in English | MEDLINE | ID: mdl-34524976

ABSTRACT

OBJECTIVE: Genetic factors underpin the narrow intraindividual variability of thyroid function, although precise contributions of environmental vs genetic factors remain uncertain. We sought to clarify the heritability of thyroid function traits and thyroid peroxidase antibody (TPOAb) positivity and identify single nucleotide polymorphisms (SNPs) contributing to the trait variance. METHODS: Heritability of thyroid-stimulating hormone (TSH), free T4 (fT4), free T3 (fT3) and TPOAb in a cohort of 2854 euthyroid, dizygous and monozygous twins (age range 11.9-16.9 years) from the Brisbane Longitudinal Twin Study (BLTS) was assessed using structural equation modelling. A genome-wide analysis was conducted on 2832 of these individuals across 7 522 526 SNPs as well as gene-based association analyses. Replication analysis of the association results was performed in the Raine Study (n = 1115) followed by meta-analysis to maximise power for discovery. RESULTS: Heritability of thyroid function parameters in the BLTS was 70.8% (95% CI: 66.7-74.9%) for TSH, 67.5% (59.8-75.3%) for fT4, 59.7% (54.4-65.0%) for fT3 and 48.8% (40.6-56.9%) for TPOAb. The genome-wide association study (GWAS) in the discovery cohort identified a novel association between rs2026401 upstream of NCOA3 and TPOAb. GWAS meta-analysis found associations between TPOAb and rs445219, also near NCOA3, and fT3 and rs12687280 near SERPINA7. Gene-based association analysis highlighted SERPINA7 for fT3 and NPAS3 for fT4. CONCLUSION: Our findings resolve former contention regarding heritability estimates of thyroid function traits and TPOAb positivity. GWAS and gene-based association analysis identified variants accounting for a component of this heritability.


Subject(s)
Genome-Wide Association Study , Nuclear Receptor Coactivator 3/genetics , Thyroid Function Tests , Thyroid Gland/physiology , Thyroxine-Binding Globulin/genetics , Adolescent , Australia/epidemiology , Cohort Studies , Female , Humans , Iodide Peroxidase/analysis , Iodide Peroxidase/immunology , Longitudinal Studies , Male , Polymorphism, Single Nucleotide , Thyrotropin/blood , Thyroxine/blood , Triiodothyronine/blood , Twins, Monozygotic
8.
Horm Res Paediatr ; 94(1-2): 76-80, 2021.
Article in English | MEDLINE | ID: mdl-34126618

ABSTRACT

INTRODUCTION: Neonatal screening programs for congenital hypothyroidism (CH) have been implemented worldwide to facilitate early diagnosis and treatment. The Dutch neonatal CH screening is primarily based on the measurement of thyroxine (T4). When T4 is low, an additional thyroxine-binding globulin (TBG) measurement is performed to reduce the number of false-positive screening results due to harmless TBG deficiency. Here, we present a case of a rare functional TBG deficiency leading to a false suspicion of CH. CASE PRESENTATION: Neonatal screening in this patient revealed a decreased T4, normal TSH, and normal TBG concentration, suggesting central CH. However, free T4 was normal. DNA sequencing analysis revealed a novel, hemizygous mutation (c.139G>A) in SERPINA7, the gene encoding TBG, resulting in the substitution of the conserved amino acid alanine to threonine at position 27. Crystal structure analyses showed that this substitution has a detrimental effect on binding of T4 to TBG. CONCLUSIONS: The novel SERPINA7 variant in this patient led to a false suspicion of central hypothyroidism in the Dutch T4-based neonatal screening program. It is important to recognize patients with such TBG defects to prevent unnecessary additional testing and treatment.


Subject(s)
Congenital Hypothyroidism/diagnosis , Genetic Diseases, X-Linked/diagnosis , Mutation, Missense , Thyroxine-Binding Globulin/deficiency , Thyroxine-Binding Globulin/genetics , Congenital Hypothyroidism/genetics , Diagnostic Errors , Genetic Diseases, X-Linked/genetics , High-Throughput Nucleotide Sequencing , Humans , Infant, Newborn , Male , Neonatal Screening , Thyroid Function Tests
9.
Mol Genet Genomic Med ; 9(2): e1571, 2021 02.
Article in English | MEDLINE | ID: mdl-33554479

ABSTRACT

SUB-HEADING: Compound hemizygous variants in SERPINA7 gene. BACKGROUND: Thyroxine-binding globulin (TBG) is encoded by SERPINA7 (OMIM. 314200) which is located on Xq22.3. SERPINA7 variants caused TBG deficiency which does not require treatment, but the decreased thyroxine may be misdiagnosed as hypothyroidism. We discovered some variants of TBG caused by alterations that differ from previously reported. MATERIALS AND METHODS: In this study, we enrolled 32 subjects from 10 families and sequenced the SERPINA7 genes of TBG-deficient subjects. Then, variants were analyzed to assess their effect on TBG expression and secretion. Bioinformatics database, protein structure, and dynamics simulation were used to evaluate the deleterious effects. Finally, we identified 2 novel and 4 known variants, and found 26 of 30 subjects carried the p.L303F. The DynaMut predictions indicated the variants (p.E91K, p.I92T, p.R294C, and p.L303F) exhibited decreased stability. CONCLUSION: Analyses revealed the p.L303F change the protein stability and flexibility, and it had an impact on the function of TBG, but when coexisted with other variants it might change the conformational structure of the protein and aggravate the damage to the protein. We speculated that the existence of a higher number of variants resulted in lower TBG secretion.


Subject(s)
Congenital Hypothyroidism/genetics , Polymorphism, Single Nucleotide , Thyroxine-Binding Globulin/genetics , Adult , Child , Congenital Hypothyroidism/pathology , Female , Gene Frequency , Hemizygote , Humans , Male , Mutation , Pedigree , Protein Stability , Thyroxine-Binding Globulin/chemistry , Thyroxine-Binding Globulin/deficiency
10.
J Endocrinol Invest ; 43(12): 1703-1710, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32266677

ABSTRACT

PURPOSE: T4-binding globulin (TBG) is the main thyroid hormone (TH) transporter present in human serum. Inherited thyroxine-binding globulin (TBG) deficiency is caused by mutations in the TBG (SERPINA7) gene, which is located on the X chromosome. This study was performed to report and evaluate coding region mutations in TBG gene for partial thyroxine-binding globulin deficiency. METHODS: A pedigree spanning four generations is described in this study. The proband is a female with partial TBG deficiency. All members of this pedigree underwent thyroid function tests, while Sanger sequencing was used to identify the TBG gene mutations. Bioinformatics databases were used to evaluate the deleterious effects of the mutation(s). Two hundred and seven unrelated individuals were used to evaluate the thyroid function of individuals with different TBG mutations. A one-way ANOVA was used to analyze the impact of the TBG mutations on thyroid function. RESULTS: TBG gene sequencing results revealed that the proband had a novel mutation in codon 27 leading to alanine to valine substitution (p.A27V). This mutation was associated with lower serum T4 levels (p < 0.0001) when compared to the groups that did not carry the mutation. The previously reported p.L283F mutation was also found in the proband. The hemizygous p.L283F individuals presenting with lower T4 serum and TBG levels (p < 0.001) when compared to wildtype males and females. Both mutations were deleterious upon SIFT and PolyPhen-2 evaluation. CONCLUSION: Associated with partial thyroxine-binding globulin deficiency, this study reports a novel p.A27V mutation in the TBG gene.


Subject(s)
Abortion, Habitual/genetics , Genetic Diseases, X-Linked/genetics , Thyroxine-Binding Globulin/deficiency , Adult , China , Family , Female , Genetic Diseases, X-Linked/complications , Humans , Mutation, Missense , Open Reading Frames/genetics , Pedigree , Pregnancy , Thyroid Function Tests , Thyroxine-Binding Globulin/genetics
11.
Environ Pollut ; 254(Pt B): 112925, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31454572

ABSTRACT

Polychlorinated biphenyls (PCBs), polybrominated diphenyl ethers (PBDEs) and new flame retardants (NFRs) are known thyroid hormone (TH) disruptors, but their disrupting mechanisms in humans are not completely understood. In this study, we aimed to explore the disrupting mechanisms of the aforementioned chemicals via examining TH-regulated proteins and gene expression in human serum. Adult participants from an e-waste dismantling (exposed group) and a control region (control group) in South China provided blood samples for the research. Some compounds of PCBs, PBDEs, and NFRs showed strong binding affinity to the thyroid-stimulating hormone (TSH), thyroglobulin, thyroxine-binding globulin (TBG), gene expression of TH receptor α (TRα) and ß, and iodothyronine deiodinase I (ID1). The highly exposed individuals had lower levels of TBG, TSH, and expression of TRα, but higher expression of ID1 than those of the control group. The disruption of TH-regulated proteins and gene expression suggested the exertion of different and, at times, even contradictory effects on TH disruption. However, no statistically significant difference was found in the TH levels between the exposed and the control group, implying that the TH disruption induced by these chemicals depends on the combined influence of multiple mechanisms. Gene expression appears to be an effective approach for investigations of TH disruption and the potential health effects.


Subject(s)
Electronic Waste/adverse effects , Flame Retardants/analysis , Halogenated Diphenyl Ethers/blood , Polychlorinated Biphenyls/blood , Thyroid Hormones/blood , Adult , Aged , China , Female , Flame Retardants/adverse effects , Gene Expression/drug effects , Halogenated Diphenyl Ethers/adverse effects , Humans , Male , Middle Aged , Polychlorinated Biphenyls/adverse effects , Receptors, Thyroid Hormone/genetics , Receptors, Thyroid Hormone/metabolism , Thyrotropin/blood , Thyroxine-Binding Globulin/genetics , Thyroxine-Binding Globulin/metabolism
12.
Gene ; 666: 58-63, 2018 Aug 05.
Article in English | MEDLINE | ID: mdl-29733970

ABSTRACT

OBJECTIVE: Thyroxine-binding globulin (TBG) is the major human thyroid hormone transport protein, encoded by the SERPINA7 gene (Xq22.2). We aim to investigate the molecular basis of partial TBG deficiency (TBG-PD) in a female, by evaluating the X-chromosome inactivation pattern as well as the mutant protein structural modeling. DESIGN AND METHODS: Sequencing of the coding region of the SERPINA7 gene was performed in a female with a TBG-PD phenotype and her first-degree relatives. The proband presented with low serum levels of total T3 (TT3) and total T4 (TT4), serum TSH level of 5.4 µUI/mL (normal range, 0.35-5.5), and serum TBG level of 5.5 mg/L (normal range, 13.6-27.2). X-chromosome inactivation pattern was evaluated by methylation analysis of the androgen receptor gene (Xq11.2). Structural analysis of the SERPIN family was performed using Pymol and Areaimol, and PFSTATS for conservation analysis and family-wide investigation of equivalent positions in human homologs. Modeller was used for point mutation structural modeling. RESULTS: A novel missense SERPINA7 mutation (p.R35W; c.163C > T) was found in heterozygosity in the proband, and in hemizygosity in her affected siblings. The proband X-chromosome inactivation ratio was 20:80. The substitution of an arginine by a tryptophan is predicted to disrupt the protein surface and main electrostatic interactions. Tryptophans are extremely rare (0.1%) in this position. CONCLUSIONS: We report a new SERPINA7 variant associated with TBG-PD in three siblings. We named this variant TBG-Brasilia. The X-chromosome inactivation pattern may have accounted for the rare phenotypic expression in a female. The hydrophobic nature of the mutant is predicted to create an apolar patch at the surface, which results in protein aggregation and/or misfolding, potentially responsible for thyroid hormone transport defect.


Subject(s)
Genetic Diseases, X-Linked/genetics , Thyroxine-Binding Globulin/deficiency , Adult , Base Sequence , DNA Mutational Analysis , Female , Genetic Association Studies , Humans , Hydrophobic and Hydrophilic Interactions , Male , Models, Molecular , Mutation, Missense , Pedigree , Point Mutation , Protein Conformation, alpha-Helical , Protein Domains , Thyroxine-Binding Globulin/chemistry , Thyroxine-Binding Globulin/genetics , X Chromosome Inactivation
13.
Nucleic Acids Res ; 45(W1): W453-W457, 2017 07 03.
Article in English | MEDLINE | ID: mdl-28460062

ABSTRACT

Many analyses for the detection of biological phenomena rely on a multiple sequence alignment as input. The results of such analyses are often further studied through parametric bootstrap procedures, using sequence simulators. One of the problems with conducting such simulation studies is that users currently have no means to decide which insertion and deletion (indel) parameters to choose, so that the resulting sequences mimic biological data. Here, we present SpartaABC, a web server that aims to solve this issue. SpartaABC implements an approximate-Bayesian-computation rejection algorithm to infer indel parameters from sequence data. It does so by extracting summary statistics from the input. It then performs numerous sequence simulations under randomly sampled indel parameters. By computing a distance between the summary statistics extracted from the input and each simulation, SpartaABC retains only parameters behind simulations close to the real data. As output, SpartaABC provides point estimates and approximate posterior distributions of the indel parameters. In addition, SpartaABC allows simulating sequences with the inferred indel parameters. To this end, the sequence simulators, Dawg 2.0 and INDELible were integrated. Using SpartaABC we demonstrate the differences in indel dynamics among three protein-coding genes across mammalian orthologs. SpartaABC is freely available for use at http://spartaabc.tau.ac.il/webserver.


Subject(s)
Algorithms , INDEL Mutation , Sequence Analysis/methods , Software , Bayes Theorem , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Internet , Receptor, Parathyroid Hormone, Type 1/genetics , Thyroxine-Binding Globulin/genetics
14.
Mol Cell Endocrinol ; 459: 43-52, 2017 Dec 25.
Article in English | MEDLINE | ID: mdl-28249735

ABSTRACT

Thyroid hormones (THs) are evolutionarily old hormones, having effects on metabolism in bacteria, invertebrates and vertebrates. THs bind specific distributor proteins (THDPs) to ensure their efficient distribution through the blood and cerebrospinal fluid in vertebrates. Albumin is a THDP in the blood of all studied species of vertebrates, so may be the original vertebrate THDP. However, albumin has weak affinity for THs. Transthyretin (TTR) has been identified in the blood across different lineages in adults vs juveniles. TTR has intermediate affinity for THs. Thyroxine-binding globulin has only been identified in mammals and has high affinity for THs. Of these THDPs, TTR is the only one known to be synthesised in the brain and is involved in moving THs from the blood into the cerebrospinal fluid. We analysed the rates of evolution of these three THDPs: TTR has been most highly conserved and albumin has had the highest rate of divergence.


Subject(s)
Albumins/chemistry , Evolution, Molecular , Prealbumin/chemistry , Thyroid Hormones/metabolism , Thyroxine-Binding Globulin/chemistry , Albumins/genetics , Albumins/metabolism , Animals , Conserved Sequence , Gene Expression , Humans , Models, Molecular , Phylogeny , Prealbumin/genetics , Prealbumin/metabolism , Protein Binding , Protein Structure, Secondary , Protein Transport , Selection, Genetic , Thyroid Hormones/genetics , Thyroxine-Binding Globulin/genetics , Thyroxine-Binding Globulin/metabolism
15.
Mol Cell Endocrinol ; 458: 16-21, 2017 Dec 15.
Article in English | MEDLINE | ID: mdl-28257828

ABSTRACT

Thyroid hormones are bound to three major serum transport proteins, thyroxin-binding globulin (TBG), transthyretin (TTR) and human serum albumin (HSA). TBG has the strongest affinity for thyroid hormones, TTR is also found in the cerebrospinal fluid and HSA is the most abundant protein in plasma. Combination defects of either a high affinity TTR or HSA variant do not compensate TBG deficiency, underscoring the dominant role of TBG among the thyroid hormone transport proteins. On the other hand, coexistence of raised affinity TTR and HSA variants causes an augmented hyperthyroxinemia. Variations in thyroid hormone transport proteins may alter thyroid function tests to mimic hypo- or hyperthyroidism. As affected individuals are clinically euthyroid and do not require treatment, identification of thyroid hormone transport protein defects is important to avoid unnecessary diagnostic and therapeutic interventions. Mammals share the multilayered system of thyroid hormone binding proteins with humans. Some of them, especially carnivores, do not express TBG. In dogs, this defect has been shown to be caused by a defective hepatocyte nuclear factor-1 binding site in the TBG promoter, preventing TBG synthesis in the liver. The major endogenous thyroid hormone metabolite 3-iodothyronamine (3-T1AM) exerts marked cryogenic, metabolic, cardiac and central nervous system actions. It is bound to apolipoproteinB-100 (ApoB100), possibly facilitating its cellular uptake via interaction with the low density lipoprotein-receptor. This review summarizes the handling of hydrophobic charged thyroid hormone signaling molecules and their metabolite 3-T1AM in aqueous body fluids and the advantages and limits of their serum distributor proteins.


Subject(s)
Prealbumin/metabolism , Serum Albumin, Human/metabolism , Thyroid Hormones/metabolism , Thyroxine-Binding Globulin/metabolism , Animals , Binding Sites , Dogs , Humans , Mammals/metabolism , Prealbumin/cerebrospinal fluid , Promoter Regions, Genetic , Protein Transport , Thyroid Hormones/blood , Thyronines/metabolism , Thyroxine-Binding Globulin/chemistry , Thyroxine-Binding Globulin/genetics
16.
Toxicology ; 366-367: 32-42, 2016 07 29.
Article in English | MEDLINE | ID: mdl-27528273

ABSTRACT

Perfluoroalkyl substances (PFASs) have been shown to cause abnormal levels of thyroid hormones (THs) in experimental animals, but the molecular mechanism is poorly understood. Here, a fluorescence displacement assay was used to determine the binding affinities of 16 PFASs with two major TH transport proteins, transthyretin (TTR) and thyroxine-binding globulin (TBG). Most of the tested PFASs bound TTR with relative potency (RP) values of 3×10(-4) to 0.24 when compared with that of the natural ligand thyroxine, whereas fluorotelomer alcohols did not bind. Only perfluorotridecanoic acid and perfluorotetradecanoic acid bound TBG, with RP values of 2×10(-4) when compared with that of thyroxine. Based on these results, it was estimated that displacement of T4 from TTR by perfluorooctane sulfonate and perfluorooctanoic acids would be significant for the occupationally exposed workers but not the general population. Structure-binding analysis revealed that PFASs with a medium chain length and a sulfonate acid group are optimal for TTR binding, and PFASs with lengths longer than 12 carbons are optimal for TBG binding. Three mutant proteins were prepared to examine crucial residues involved in the binding of PFASs to TH transport proteins. TTR with a K15G mutation and TBG with either a R378G or R381G mutation showed decreased binding affinity to PFASs, indicating that these residues play key roles in the interaction with the compounds. Molecular docking showed that the PFASs bind to TTR with their acid group forming a hydrogen bond with K15 and the hydrophobic chain towards the interior. PFASs were modeled to bind TBG with their acid group forming a hydrogen bond with R381 and the hydrophobic chain extending towards R378. The findings aid our understanding of the behavior and toxicity of PFASs on the thyroid hormone system.


Subject(s)
Alkanesulfonic Acids/toxicity , Caprylates/toxicity , Carrier Proteins/metabolism , Fluorocarbons/toxicity , Prealbumin/metabolism , Thyroxine-Binding Globulin/metabolism , Alkanesulfonic Acids/blood , Caprylates/blood , Carrier Proteins/genetics , Fluorocarbons/blood , Humans , Inhibitory Concentration 50 , Molecular Docking Simulation , Prealbumin/genetics , Protein Conformation , Structure-Activity Relationship , Thyroid Hormones/blood , Thyroid Hormones/metabolism , Thyroxine/metabolism , Thyroxine-Binding Globulin/genetics
17.
Zhonghua Er Ke Za Zhi ; 54(6): 428-32, 2016 Jun 02.
Article in Chinese | MEDLINE | ID: mdl-27256229

ABSTRACT

OBJECTIVE: To investigate the clinical characteristics of twins with thyroxine binding globulin (TBG) deficiency and to find SERPINA7 gene mutations. METHOD: Data(2015) related to clinical characteristics, serum biochemistry, gene mutations and pedigree of two children with TBG deficiency were collected in the First Affiliated Hospital of College of Medicine, Zhejiang University. The related literature was searched form China National Knowledge Infrastructure, Wanfang Data Knowledge Service Platform, National Center for Biotechnology Information and PubMed (up to December 2015) by using search terms "Thyroxine binding globulin deficiency, gene, mutation" . RESULT: Both patients were diagnosed as central hypothyroidism at the beginning and treated with L-thyroxine. Both of the identical twins of the triplet were observed for mutation in exon3, c. 631Gï¹¥A(p.A211T), a new mutation had not been reported, but their parents and another non-identical triplet brother were normal. Literature review showed that 23 foreign cases with SERPINA7 gene mutation had been reported, however, no Chinese with SERPINA7 gene mutation had been reported. Among reported cases it was shown that SERPINA7 gene mutations located in exon, intron, promoter and enhancer. Up to now, 49 variants had been identified, 41 of them located in the mutated genes. Including these two cases, patients with thyroxine binding globulin deficiency were characterized by reduced serum TH levels, but normal free TH and TSH and absence of clinical manifestations. CONCLUSION: The new mutation of SERPINA7 gene c. 631Gï¹¥A(p.A211T)is not transmitted via the known X chromosome linked heredity, and as the cases were test tube triplet infants, it is a de novo mutation. The serum thyroid function tests of TBG deficiency showed decreased TT4, TT3 and normal TSH and TBG deficiency is often misdiagnosed as central hypothyroidism.


Subject(s)
Genetic Diseases, X-Linked/diagnosis , Thyroxine-Binding Globulin/deficiency , Child , China , Exons , Humans , Infant , Introns , Male , Mutation , Pedigree , Promoter Regions, Genetic , Thyroid Function Tests , Thyroxine/therapeutic use , Thyroxine-Binding Globulin/genetics , Triplets , Twins, Monozygotic
18.
J Genet ; 95(2): 311-6, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27350674

ABSTRACT

The defect of low density lipoprotein receptor disturbs cholesterol metabolism and causes familial hypercholesterolaemia (FH). In this study, we directly delivered exogenous Ldlr gene into the liver of FH model mice (Ldlr(-/-)) by lentiviral gene transfer system. The results showed that the Ldlr gene controlled by hepatocyte-specific human thyroxine-binding globulin (TBG) promoter successfully and exclusively expressed in livers.We found that, although, the content of high density lipoprotein in serum was not significantly affected by the Ldlr gene expression, the serum low density lipoprotein level was reduced by 46%, associated with a 30% and 28% decrease in triglyceride and total cholesterol, respectively, compared to uninjected Ldlr(-/-) mice. Moreover, the TBG directed expression of Ldlr significantly decreased the lipid accumulation in liver and reduced plaque burden in aorta (32%). Our results indicated that the hepatocyte-specific expression of Ldlr gene strikingly lowered serum lipid levels and resulted in amelioration of hypercholesterolaemia.


Subject(s)
Genetic Therapy/methods , Hyperlipoproteinemia Type II/therapy , Liver/metabolism , Plaque, Atherosclerotic/therapy , Receptors, LDL/genetics , Transgenes , Animals , Aorta/metabolism , Aorta/pathology , Cholesterol, HDL/blood , Cholesterol, LDL/blood , Disease Models, Animal , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Hyperlipoproteinemia Type II/genetics , Hyperlipoproteinemia Type II/metabolism , Hyperlipoproteinemia Type II/pathology , Lentivirus/genetics , Lentivirus/metabolism , Liver/pathology , Mice , Mice, Transgenic , Plaque, Atherosclerotic/genetics , Plaque, Atherosclerotic/metabolism , Plaque, Atherosclerotic/pathology , Promoter Regions, Genetic , Receptors, LDL/metabolism , Thyroxine-Binding Globulin/genetics , Thyroxine-Binding Globulin/metabolism , Transfection , Triglycerides/blood
19.
Scand J Rheumatol ; 45(5): 394-6, 2016 Oct.
Article in English | MEDLINE | ID: mdl-26885894

ABSTRACT

OBJECTIVES: Systemic lupus erythematosus (SLE) is a chronic disease occurring up to 15 times more frequently in females than males. This bias extends to possible differences in disease flares and response to therapy. This study was initiated to investigate the differences between girls and boys with childhood-onset SLE (cSLE) at the molecular level. METHOD: We analysed the Gene Expression Omnibus National Center for Biotechnology Information (GEO NCBI) microarray data available for 88 girls and 16 boys with treatment-naïve cSLE and compared the results to those from healthy controls. Transcriptional profiles were generated using the platforms of Affymetrix U133A and U133B gene chips and Bioconductor/R programming packages were used to process and compare the data. RESULTS: Girls with cSLE overexpressed an interferon (IFN)-α signature that was absent in boys. Boys with cSLE were observed to overexpress tumour necrosis factor-related genes that were absent in girls. Both boys and girls were observed to overexpress several genes related to granulopoeisis. CONCLUSIONS: Our results suggest a potential application of genomics to differentially predict response to therapy between females and males with SLE.


Subject(s)
Interferons/genetics , Lupus Erythematosus, Systemic/genetics , Sex Factors , Transcriptome , Adaptor Proteins, Signal Transducing , Adolescent , Antigens/genetics , Carrier Proteins/genetics , Case-Control Studies , Child , Cytoskeletal Proteins/genetics , Female , G-Protein-Coupled Receptor Kinase 1/genetics , Gene Expression , Genomics , Humans , Intracellular Signaling Peptides and Proteins/genetics , Linear Models , Male , Membrane Proteins/genetics , Microarray Analysis , Mitochondrial Proteins/genetics , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , RNA-Binding Proteins , Receptor-Like Protein Tyrosine Phosphatases, Class 2/genetics , Sialic Acid Binding Ig-like Lectin 1/genetics , Thyroxine-Binding Globulin/genetics , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics
20.
J Clin Endocrinol Metab ; 100(1): E173-81, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25361180

ABSTRACT

CONTEXT: T4-binding globulin (TBG), a protein secreted by the liver, is the main thyroid hormone (TH) transporter in human serum. TBG deficiency is characterized by reduced serum TH levels, but normal free TH and TSH and absent clinical manifestations. The inherited form of TBG deficiency is usually due to a mutation in the TBG gene located on the X-chromosome. OBJECTIVE: Among the 75 families with X-chromosome-linked TBG deficiency identified in our laboratory, no mutations in the TBG gene were found in four families. The aim of the study was to identify the mechanism of TBG deficiency in these four families using biochemical and genetic studies. DESIGN: Observational cohort, prospective. SETTING: University research center. PATIENTS: Four families with inherited TBG deficiency and no mutations in the TBG gene. INTERVENTION: Clinical evaluation, thyroid function tests, and targeted resequencing of 1 Mb of the X-chromosome. RESULTS: Next-generation sequencing identified a novel G to A variant 20 kb downstream of the TBG gene in all four families. In silico analysis predicted that the variant resides within a liver-specific enhancer. In vitro studies confirmed the enhancer activity of a 2.2-kb fragment of genomic DNA containing the novel variant and showed that the mutation reduces the activity of this enhancer. The affected subjects share a haplotype of 8 Mb surrounding the mutation, and the most recent common ancestor among the four families was estimated to be 19.5 generations ago (95% confidence intervals, 10.4-37). CONCLUSIONS: To our knowledge, the present study is the first report of an inherited endocrine disorder caused by a mutation in an enhancer region.


Subject(s)
Enhancer Elements, Genetic , Liver/metabolism , Mutation , Thyroxine-Binding Globulin/genetics , Adolescent , Adult , Child , Female , Haplotypes , Humans , Male , Middle Aged , Pedigree , Thyroid Diseases/genetics , Thyroid Diseases/metabolism , Thyroxine-Binding Globulin/metabolism , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...