Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Nature ; 532(7598): 250-4, 2016 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-27042933

RESUMO

Cancer is a disease of ageing. Clinically, aged cancer patients tend to have a poorer prognosis than young. This may be due to accumulated cellular damage, decreases in adaptive immunity, and chronic inflammation. However, the effects of the aged microenvironment on tumour progression have been largely unexplored. Since dermal fibroblasts can have profound impacts on melanoma progression, we examined whether age-related changes in dermal fibroblasts could drive melanoma metastasis and response to targeted therapy. Here we find that aged fibroblasts secrete a Wnt antagonist, sFRP2, which activates a multi-step signalling cascade in melanoma cells that results in a decrease in ß-catenin and microphthalmia-associated transcription factor (MITF), and ultimately the loss of a key redox effector, APE1. Loss of APE1 attenuates the response of melanoma cells to DNA damage induced by reactive oxygen species, rendering the cells more resistant to targeted therapy (vemurafenib). Age-related increases in sFRP2 also augment both angiogenesis and metastasis of melanoma cells. These data provide an integrated view of how fibroblasts in the aged microenvironment contribute to tumour progression, offering new possibilities for the design of therapy for the elderly.


Assuntos
Envelhecimento/metabolismo , Resistencia a Medicamentos Antineoplásicos , Melanoma/tratamento farmacológico , Melanoma/patologia , Proteínas de Membrana/metabolismo , Metástase Neoplásica , Microambiente Tumoral , Adulto , Animais , Linhagem Celular Tumoral , Meios de Cultivo Condicionados/farmacologia , Dano ao DNA , DNA Liase (Sítios Apurínicos ou Apirimidínicos)/metabolismo , Progressão da Doença , Fibroblastos/metabolismo , Humanos , Indóis/farmacologia , Indóis/uso terapêutico , Masculino , Melanoma/irrigação sanguínea , Melanoma/genética , Camundongos , Fator de Transcrição Associado à Microftalmia/metabolismo , Pessoa de Meia-Idade , Terapia de Alvo Molecular , Neovascularização Patológica , Estresse Oxidativo , Fenótipo , Espécies Reativas de Oxigênio/metabolismo , Sulfonamidas/farmacologia , Sulfonamidas/uso terapêutico , Vemurafenib , Via de Sinalização Wnt , Proteína Wnt1/antagonistas & inibidores , beta Catenina/metabolismo
3.
Genet Med ; 19(11): 1276-1279, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28541281

RESUMO

PurposeNonclassic 21-hydroxylase deficiency, a mild form of congenital adrenal hyperplasia (CAH), is estimated to be the most common autosomal recessive condition, with an especially high prevalence in Ashkenazi Jews (3.7% affected, 30.9% carriers), based on a 1985 HLA-B linkage study of affected families. Affected individuals, especially women, may suffer from hyperandrogenism and infertility. State-of-the-art genetic studies have not been done to confirm these remarkable rates.MethodsCYP21A2 genotyping was performed in 200 unrelated healthy Ashkenazi Jewish subjects and 200 random US Caucasians who did not self-identify as a specific ethnicity using multiplex minisequencing, real-time polymerase chain reaction and junction site analysis.ResultsNonclassic CAH carriership was found similarly in 15% (95% confidence interval (CI): 10.4-20.7) of Ashkenazi Jews and 9.5% (95% CI: 5.8-14.4) of Caucasians (P=0.13). The proportion of Ashkenazi Jewish nonclassic CAH carriers (0.15 versus 0.309, P<0.0001) and disease affected (0.005 versus 0.037, P=0.009) was not as high as previously reported. The estimated prevalence of nonclassic CAH in Caucasians was 1 in 200 (0.5%, 95% CI: 0.01-2.8).ConclusionNonclassic CAH is a common condition, regardless of ethnicity, and should be considered with preconception and infertility counseling.


Assuntos
Hiperplasia Suprarrenal Congênita/epidemiologia , Judeus , Hiperplasia Suprarrenal Congênita/genética , Frequência do Gene , Técnicas de Genotipagem , Heterozigoto , Humanos , Mutação , Prevalência , Esteroide 21-Hidroxilase/genética , Estados Unidos/epidemiologia , População Branca/genética
4.
Arch Biochem Biophys ; 633: 58-67, 2017 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-28893510

RESUMO

Glycosylphosphatidylinositol transamidase (GPI-T) catalyzes the post-translational addition of the GPI anchor to the C-terminus of some proteins. In most eukaryotes, Gpi8, the active site subunit of GPI-T, is part of a hetero-pentameric complex containing Gpi16, Gaa1, Gpi17, and Gab1. Gpi8, Gaa1, and Gpi16 co-purify as a heterotrimer from Saccharomyces cerevisiae, suggesting that they form the core of the GPI-T. Details about the assembly and organization of these subunits have been slow to emerge. We have previously shown that the soluble domain of S. cerevisiae Gpi8 (Gpi823-306) assembles as a homodimer, similar to the caspases with which it shares weak sequence homology (Meitzler, J. L. et al., 2007). Here we present the characterization of a complex between the soluble domains of Gpi8 and Gaa1. The complex between GST-Gpi823-306 (α) and His6-Gaa150-343 (ß) was characterized by native gel analysis and size exclusion chromatography (SEC) and results are most consistent with an α2ß2 stoichiometry. These results demonstrate that Gpi8 and Gaa1 interact specifically without a requirement for other subunits, bring us closer to determining the stoichiometry of the core subunits of GPI-T, and lend further credence to the hypothesis that these three subunits assemble into a dimer of a trimer.


Assuntos
Aminoaciltransferases/química , Glicoproteínas de Membrana/química , Proteínas de Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/química , Motivos de Aminoácidos , Aminoaciltransferases/genética , Aminoaciltransferases/metabolismo , Sítios de Ligação , Clonagem Molecular , Escherichia coli/genética , Escherichia coli/metabolismo , Expressão Gênica , Cinética , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Modelos Moleculares , Ligação Proteica , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Domínios e Motivos de Interação entre Proteínas , Multimerização Proteica , Estrutura Terciária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Saccharomyces cerevisiae/enzimologia , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Solubilidade , Homologia Estrutural de Proteína , Especificidade por Substrato , Vibrionaceae/química , Vibrionaceae/enzimologia
6.
Hum Mutat ; 37(9): 893-7, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27297501

RESUMO

Some variants that cause autosomal-recessive congenital adrenal hyperplasia (CAH) also cause hypermobility type Ehlers-Danlos syndrome (EDS) due to the monoallelic presence of a chimera disrupting two flanking genes: CYP21A2, encoding 21-hydroxylase, necessary for cortisol and aldosterone biosynthesis, and TNXB, encoding tenascin-X, an extracellular matrix protein. Two types of CAH tenascin-X (CAH-X) chimeras have been described with a total deletion of CYP21A2 and characteristic TNXB variants. CAH-X CH-1 has a TNXB exon 35 120-bp deletion resulting in haploinsufficiency, and CAH-X CH-2 has a TNXB exon 40 c.12174C>G (p.Cys4058Trp) variant resulting in a dominant-negative effect. We present here three patients with biallelic CAH-X and identify a novel dominant-negative chimera termed CAH-X CH-3. Compared with monoallelic CAH-X, biallelic CAH-X results in a more severe phenotype with skin features characteristic of classical EDS. We present evidence for disrupted tenascin-X function and computational data linking the type of TNXB variant to disease severity.


Assuntos
Hiperplasia Suprarrenal Congênita/genética , Síndrome de Ehlers-Danlos/genética , Deleção de Genes , Esteroide 21-Hidroxilase/genética , Tenascina/genética , Adolescente , Hiperplasia Suprarrenal Congênita/metabolismo , Adulto , Alelos , Colágeno/metabolismo , Síndrome de Ehlers-Danlos/metabolismo , Feminino , Fibrilina-1/metabolismo , Humanos , Masculino , Linhagem , Tenascina/metabolismo , Adulto Jovem
7.
FASEB J ; 28(8): 3313-24, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24732132

RESUMO

Fibromuscular dysplasia (FMD) is a rare, nonatherosclerotic arterial disease for which the molecular basis is unknown. We comprehensively studied 47 subjects with FMD, including physical examination, spine magnetic resonance imaging, bone densitometry, and brain magnetic resonance angiography. Inflammatory biomarkers in plasma and transforming growth factor ß (TGF-ß) cytokines in patient-derived dermal fibroblasts were measured by ELISA. Arterial pathology other than medial fibrodysplasia with multifocal stenosis included cerebral aneurysm, found in 12.8% of subjects. Extra-arterial pathology included low bone density (P<0.001); early onset degenerative spine disease (95.7%); increased incidence of Chiari I malformation (6.4%) and dural ectasia (42.6%); and physical examination findings of a mild connective tissue dysplasia (95.7%). Screening for mutations causing known genetically mediated arteriopathies was unrevealing. We found elevated plasma TGF-ß1 (P=0.009), TGF-ß2 (P=0.004) and additional inflammatory markers, and increased TGF-ß1 (P=0.0009) and TGF-ß2 (P=0.0001) secretion in dermal fibroblast cell lines from subjects with FMD compared to age- and gender-matched controls. Detailed phenotyping of patients with FMD allowed us to demonstrate that FMD is a systemic disease with alterations in common with the spectrum of genetic syndromes that involve altered TGF-ß signaling and offers TGF-ß as a marker of FMD.


Assuntos
Fibroblastos/metabolismo , Displasia Fibromuscular/metabolismo , Fator de Crescimento Transformador beta1/biossíntese , Adulto , Idoso , Malformação de Arnold-Chiari/complicações , Biomarcadores/sangue , Densidade Óssea , Doenças Ósseas Metabólicas/etiologia , Estudos de Casos e Controles , Ciclo Celular , Linhagem Celular , Tecido Conjuntivo/patologia , Derme/patologia , Dilatação Patológica , Dura-Máter/patologia , Feminino , Displasia Fibromuscular/complicações , Displasia Fibromuscular/patologia , Humanos , Inflamação/sangue , Inflamação/etiologia , Mediadores da Inflamação/sangue , Instabilidade Articular/etiologia , Masculino , Pessoa de Meia-Idade , Fenótipo , Artéria Renal/patologia , Método Simples-Cego , Coluna Vertebral/patologia , Fator de Crescimento Transformador beta1/sangue , Fator de Crescimento Transformador beta1/metabolismo , Fator de Crescimento Transformador beta2/sangue , Fator de Crescimento Transformador beta2/metabolismo , Adulto Jovem
8.
Am J Med Genet A ; 161A(8): 2040-6, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23824657

RESUMO

The transforming growth factor ß (TGF-ß) family of growth factors are key regulators of mammalian development and their dysregulation is implicated in human disease, notably, heritable vasculopathies including Marfan (MFS, OMIM #154700) and Loeys-Dietz syndromes (LDS, OMIM #609192). We described a syndrome presenting at birth with distal arthrogryposis, hypotonia, bifid uvula, a failure of normal post-natal muscle development but no evidence of vascular disease; some of these features overlap with MFS and LDS. A de novo mutation in TGFB3 was identified by exome sequencing. Several lines of evidence indicate the mutation is hypomorphic suggesting that decreased TGF-ß signaling from a loss of TGFB3 activity is likely responsible for the clinical phenotype. This is the first example of a mutation in the coding portion of TGFB3 implicated in a clinical syndrome suggesting TGFB3 is essential for both human palatogenesis and normal muscle growth.


Assuntos
Artrogripose/genética , Transtornos do Crescimento/genética , Síndrome de Loeys-Dietz/genética , Síndrome de Marfan/genética , Debilidade Muscular/genética , Mutação/genética , Fator de Crescimento Transformador beta3/genética , Adulto , Animais , Artrogripose/diagnóstico , Células Cultivadas , Criança , Embrião não Mamífero/citologia , Embrião não Mamífero/metabolismo , Feminino , Transtornos do Crescimento/diagnóstico , Humanos , Síndrome de Loeys-Dietz/diagnóstico , Masculino , Síndrome de Marfan/diagnóstico , Debilidade Muscular/diagnóstico , Fenótipo , Transdução de Sinais , Fator de Crescimento Transformador beta3/metabolismo , Xenopus laevis/metabolismo
9.
Food Chem Toxicol ; 178: 113876, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37286029

RESUMO

While human milk is considered the optimal source of nutrition for infants for the first six and twelve months of age, with continued benefit of breastfeeding with complementary foods, a safe alternative, nutritionally adequate to support infant growth and development, is necessary. In the United States, the Food and Drug Administration (FDA) establishes the requirements necessary to demonstrate the safety of infant formula within the framework of the Federal Food, Drug, and Cosmetic Act. FDA's Center for Food Safety and Applied Nutrition/Office of Food Additive Safety evaluates the safety and lawfulness of individual ingredients used in infant formula, whereas the Office of Nutrition and Food Labeling oversees the safety of infant formula. Most infant formula ingredients are either from sources with history of safe consumption by infants or are like components in human milk. Information demonstrating the regulatory status of all ingredients is required in submissions for new infant formulas, and ingredient manufacturers often use the Generally Recognized as Safe (GRAS) Notification program to establish ingredient regulatory status. We provide an overview of ingredients used in infant formula evaluated through the GRAS Notification program to highlight trends and discuss the data and information used to reach these GRAS conclusions.


Assuntos
Aditivos Alimentares , Fórmulas Infantis , Lactente , Humanos , Estados Unidos , Inocuidade dos Alimentos , Rotulagem de Alimentos , Estado Nutricional , United States Food and Drug Administration
10.
J Clin Endocrinol Metab ; 100(8): E1143-52, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26075496

RESUMO

CONTEXT: The contiguous gene deletion syndrome (CAH-X) was described in a subset (7%) of congenital adrenal hyperplasia (CAH) patients with a TNXA/TNXB chimera, resulting in deletions of CYP21A2, encoding 21-hydroxylase necessary for cortisol biosynthesis, and TNXB, encoding the extracellular matrix glycoprotein tenascin-X (TNX). This TNXA/TNXB chimera is characterized by a 120-bp deletion in exon 35 and results in TNXB haploinsufficiency, disrupted TGF-ß signaling, and an Ehlers Danlos syndrome phenotype. OBJECTIVE: The objective of the study was to determine the genetic status of TNXB and resulting protein defects in CAH patients with a CAH-X phenotype but not the previously described TNXA/TNXB chimera. Design, Settings, Participants, and Intervention: A total of 246 unrelated CAH patients were screened for TNXB defects. Genetic defects were investigated by Southern blotting, multiplex ligation-dependent probe amplification, Sanger, and next-generation sequencing. Dermal fibroblasts and tissue were used for immunoblotting, immunohistochemical, and coimmunoprecipitation experiments. MAIN OUTCOME MEASURES: The genetic and protein status of tenascin-X in phenotypic CAH-X patients was measured. RESULTS: Seven families harbor a novel TNXB missense variant c.12174C>G (p.C4058W) and a clinical phenotype consistent with hypermobility-type Ehlers Danlos syndrome. Fourteen CAH probands carry previously described TNXA/TNXB chimeras, and seven unrelated patients carry the novel TNXB variant, resulting in a CAH-X prevalence of 8.5%. This highly conserved pseudogene-derived variant in the TNX fibrinogen-like domain is predicted to be deleterious and disulfide bonded, results in reduced dermal elastin and fibrillin-1 staining and altered TGF-ß1 binding, and represents a novel TNXA/TNXB chimera. Tenascin-X protein expression was normal in dermal fibroblasts, suggesting a dominant-negative effect. CONCLUSIONS: CAH-X syndrome is commonly found in CAH due to 21-hydroxylase deficiency and may result from various etiological mechanisms.


Assuntos
Hiperplasia Suprarrenal Congênita/complicações , Síndrome de Ehlers-Danlos/classificação , Síndrome de Ehlers-Danlos/complicações , Adolescente , Hiperplasia Suprarrenal Congênita/epidemiologia , Hiperplasia Suprarrenal Congênita/genética , Adulto , Idoso , Criança , Pré-Escolar , Estudos de Coortes , Síndrome de Ehlers-Danlos/epidemiologia , Síndrome de Ehlers-Danlos/genética , Feminino , Humanos , Lactente , Masculino , Pessoa de Meia-Idade , Fenótipo , Adulto Jovem
11.
Eur J Med Genet ; 57(2-3): 95-102, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24380766

RESUMO

Patients with congenital adrenal hyperplasia (CAH) with tenascin-X deficiency (CAH-X syndrome) have both endocrine imbalances and characteristic Ehlers Danlos syndrome phenotypes. Unlike other subtypes, tenascin-X-related Ehlers Danlos syndrome is caused by an extracellular matrix protein deficiency rather than a defect in fibrillar collagen or a collagen-modifying enzyme, and the understanding of the disease mechanisms is limited. We hypothesized that transforming growth factor-ß pathway dysregulation may, in part, be responsible for connective tissue phenotypes observed in CAH-X, due to this pathway's known role in connective tissue disorders. Fibroblasts and direct tissue from human skin biopsies from CAH-X probands and age- and sex-matched controls were screened for transforming growth factor-ß biomarkers known to be dysregulated in other hereditary disorders of connective tissue. In CAH-X fibroblast lines and dermal tissue, pSmad1/5/8 was significantly upregulated compared to controls, suggesting involvement of the bone morphogenetic protein pathway. Additionally, CAH-X samples compared to controls exhibited significant increases in fibroblast-secreted TGF-ß3, a cytokine important in secondary palatal development, and in plasma TGF-ß2, a cytokine involved in cardiac function and development, as well as palatogenesis. Finally, MMP-13, a matrix metalloproteinase important in secondary palate formation and tissue remodeling, had significantly increased mRNA and protein expression in CAH-X fibroblasts and direct tissue. Collectively, these results demonstrate that patients with CAH-X syndrome exhibit increased expression of several transforming growth factor-ß biomarkers and provide a novel link between this signaling pathway and the connective tissue dysplasia phenotypes associated with tenascin-X deficiency.


Assuntos
Hiperplasia Suprarrenal Congênita/genética , Hiperplasia Suprarrenal Congênita/metabolismo , Síndrome de Ehlers-Danlos/genética , Síndrome de Ehlers-Danlos/metabolismo , Transdução de Sinais , Tenascina/deficiência , Fator de Crescimento Transformador beta/metabolismo , Adolescente , Hiperplasia Suprarrenal Congênita/diagnóstico , Adulto , Proteínas Morfogenéticas Ósseas/genética , Proteínas Morfogenéticas Ósseas/metabolismo , Criança , Pré-Escolar , Síndrome de Ehlers-Danlos/diagnóstico , Feminino , Fibroblastos/metabolismo , Regulação da Expressão Gênica , Humanos , Imuno-Histoquímica , Masculino , Metaloproteinase 13 da Matriz/genética , Metaloproteinase 13 da Matriz/metabolismo , Fator de Crescimento Transformador beta2/sangue , Fator de Crescimento Transformador beta2/metabolismo , Fator de Crescimento Transformador beta3/biossíntese , Adulto Jovem
12.
Circ Cardiovasc Genet ; 7(1): 80-8, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24399159

RESUMO

BACKGROUND: Vascular Ehlers-Danlos syndrome (VEDS) causes reduced life expectancy because of arterial dissections/rupture and hollow organ rupture. Although the causative gene, COL3A1, was identified >20 years ago, there has been limited progress in understanding the disease mechanisms or identifying treatments. METHODS AND RESULTS: We studied inflammatory and transforming growth factor-ß (TGF-ß) signaling biomarkers in plasma and from dermal fibroblasts from patients with VEDS. Analyses were done in terms of clinical disease severity, genotype-phenotype correlations, and body composition and fat deposition alterations. VEDS subjects had increased circulating TGF-ß1, TGF-ß2, monocyte chemotactic protein-1, C-reactive protein, intercellular adhesion molecule-1, vascular cell adhesion molecule-1, and leptin and decreased interleukin-8 versus controls. VEDS dermal fibroblasts secreted more TGF-ß2, whereas downstream canonical/noncanonical TGF-ß signaling was not different. Patients with COL3A1 exon skipping mutations had higher plasma intercellular adhesion molecule-1 and vascular cell adhesion molecule-1, and VEDS probands had abnormally high plasma C-reactive protein versus affected patients identified through family members before any disease manifestations. Patients with VEDS had higher mean platelet volumes, suggesting increased platelet turnover because of ongoing vascular damage, as well as increased regional truncal adiposity. CONCLUSIONS: These findings suggest that VEDS is a systemic disease with a major inflammatory component. C-reactive protein is linked to disease state and may be a disease activity marker. No changes in downstream TGF-ß signaling and increased platelet turnover suggest that chronic vascular damage may partially explain increased plasma TGF-ß1. Finally, we found a novel role for dysregulated TGF-ß2, as well as adipocyte dysfunction, as demonstrated through reduced interleukin-8 and elevated leptin in VEDS.


Assuntos
Síndrome de Ehlers-Danlos/sangue , Inflamação/sangue , Fator de Crescimento Transformador beta/sangue , Adipocinas/sangue , Adolescente , Adulto , Biomarcadores/análise , Biomarcadores/sangue , Composição Corporal , Proteína C-Reativa/análise , Criança , Colágeno Tipo III/antagonistas & inibidores , Colágeno Tipo III/genética , Colágeno Tipo III/metabolismo , Síndrome de Ehlers-Danlos/etiologia , Síndrome de Ehlers-Danlos/genética , Feminino , Fibroblastos/citologia , Fibroblastos/metabolismo , Estudos de Associação Genética , Humanos , Inflamação/genética , Masculino , Pessoa de Meia-Idade , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta/análise , Fator de Crescimento Transformador beta1/análise , Fator de Crescimento Transformador beta1/sangue , Fator de Crescimento Transformador beta2/análise , Fator de Crescimento Transformador beta2/sangue , Adulto Jovem
13.
J Clin Endocrinol Metab ; 98(2): E379-87, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23284009

RESUMO

CONTEXT: The gene for congenital adrenal hyperplasia (CAH) due to 21-hydroxylase deficiency, CYP21A2, is flanked by the gene encoding tenascin-X (TNXB), a connective tissue extracellular matrix protein that has been linked to both autosomal dominant and autosomal recessive Ehlers-Danlos syndrome (EDS). A contiguous deletion of CYP21A2 and TNXB has been described. OBJECTIVE: The objective of the study was to determine the frequency and clinical significance of TNXB haploinsufficiency in CAH patients. DESIGN, SETTING, AND PARTICIPANTS: A total of 192 consecutive unrelated CAH patients being seen as part of an observational study at the National Institutes of Health Clinical Center (Bethesda, MD) were prospectively studied during 2006-2010. Patients were evaluated for clinical evidence of EDS, including cardiac evaluation. DNA was analyzed by PCR, multiplex ligation-dependent probe amplification, Southern blot, and TNXB sequencing. Tenascin-X expression was evaluated by Western blot analysis of fibroblasts and immunostaining of the skin. CAH patients with TNXB haploinsufficiency were compared with age-matched CAH patients with normal TNXB (controls). Phenotyping of 7 parents with TNXB haploinsufficiency was performed. MAIN OUTCOME MEASURES: The frequency of TNXB haploinsufficiency among CAH patients and the frequency of EDS symptomatology among CAH patients with TNXB haploinsufficiency and controls. RESULTS: TNXB haploinsufficiency, here termed CAH-X syndrome, was present in 7% of CAH patients. Twelve of 91 patients carrying a CYP21A2 deletion (13%) carried a contiguous deletion that extended into TNXB. One patient carried a TNXB premature stop codon. Twelve of 13 patients with CAH-X had EDS clinical features. Patients with CAH-X were more likely than age-matched controls to have joint hypermobility (P < .001), chronic joint pain (P = .003), multiple joint dislocations (P = .004), a structural cardiac valve abnormality by echocardiography (P = .02), and reduced tenascin-X expression by Western blot and immunostaining. A subset of parents had clinical findings. CONCLUSIONS: Clinical evaluation for connective tissue dysplasia should be routinely performed in CAH patients, especially those harboring a CYP21A2 deletion.


Assuntos
Hiperplasia Suprarrenal Congênita/genética , Síndrome de Ehlers-Danlos/genética , Haploinsuficiência , Esteroide 21-Hidroxilase/genética , Tenascina/genética , Adolescente , Hiperplasia Suprarrenal Congênita/fisiopatologia , Adulto , Idoso , Criança , Pré-Escolar , Síndrome de Ehlers-Danlos/fisiopatologia , Feminino , Humanos , Lactente , Luxações Articulares/genética , Luxações Articulares/fisiopatologia , Instabilidade Articular/genética , Instabilidade Articular/fisiopatologia , Masculino , Pessoa de Meia-Idade , Mutação , Dor/genética , Dor/fisiopatologia
14.
Biosci Rep ; 32(6): 577-86, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22938202

RESUMO

In eukaryotes, GPI (glycosylphosphatidylinositol) lipid anchoring of proteins is an abundant post-translational modification. The attachment of the GPI anchor is mediated by GPI-T (GPI transamidase), a multimeric, membrane-bound enzyme located in the ER (endoplasmic reticulum). Upon modification, GPI-anchored proteins enter the secretory pathway and ultimately become tethered to the cell surface by association with the plasma membrane and, in yeast, by covalent attachment to the outer glucan layer. This work demonstrates a novel in vivo assay for GPI-T. Saccharomyces cerevisiae INV (invertase), a soluble secreted protein, was converted into a substrate for GPI-T by appending the C-terminal 21 amino acid GPI-T signal sequence from the S. cerevisiae Yapsin 2 [Mkc7p (Y21)] on to the C-terminus of INV. Using a colorimetric assay and biochemical partitioning, extracellular presentation of GPI-anchored INV was shown. Two human GPI-T signal sequences were also tested and each showed diminished extracellular INV activity, consistent with lower levels of GPI anchoring and species specificity. Human/fungal chimaeric signal sequences identified a small region of five amino acids that was predominantly responsible for this species specificity.


Assuntos
Aminoaciltransferases/metabolismo , Ensaios Enzimáticos , Glicosilfosfatidilinositóis/metabolismo , Saccharomyces cerevisiae/enzimologia , beta-Frutofuranosidase/metabolismo , Sequência de Aminoácidos , Aminoaciltransferases/análise , Ácido Aspártico Endopeptidases/química , Ácido Aspártico Endopeptidases/metabolismo , Ensaios Enzimáticos/métodos , Glicosilfosfatidilinositóis/análise , Humanos , Dados de Sequência Molecular , Sinais Direcionadores de Proteínas , Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/citologia , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/metabolismo , Especificidade da Espécie , beta-Frutofuranosidase/química
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa