Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
FASEB J ; 36(3): e22185, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35133032

RESUMO

FGF19/FGF15 is an endocrine regulator of hepatic bile salt and lipid metabolism, which has shown promising effects in the treatment of NASH in clinical trials. FGF19/15 is transcribed and released from enterocytes of the small intestine into enterohepatic circulation in response to bile-induced FXR activation. Previously, the TSS of FGF19 was identified to bind Wnt-regulated TCF7L2/encoded transcription factor TCF4 in colorectal cancer cells. Impaired Wnt signaling and specifical loss of function of its coreceptor LRP6 have been associated with NASH. We, therefore, examined if TCF7L2/TCF4 upregulates Fgf19 in the small intestine and restrains NASH through gut-liver crosstalk. We examined the mice globally overexpressing, haploinsufficient, and conditional knockout models of TCF7L2 in the intestinal epithelium. The TCF7L2+/- mice exhibited increased plasma bile salts and lipids and developed diet-induced fatty liver disease while mice globally overexpressing TCF7L2 were protected against these traits. Comprehensive in vivo analysis revealed that TCF7L2 transcriptionally upregulates FGF15 in the gut, leading to reduced bile synthesis and diminished intestinal lipid uptake. Accordingly, VilinCreert2 ; Tcf7L2fl/fl mice showed reduced Fgf19 in the ileum, and increased plasma bile. The global overexpression of TCF7L2 in mice with metabolic syndrome-linked LRP6R611C substitution rescued the fatty liver and fibrosis in the latter. Strikingly, the hepatic levels of TCF4 were reduced and CYP7a1 was increased in human NASH, indicating the relevance of TCF4-dependent regulation of bile synthesis to human disease. These studies identify the critical role of TCF4 as an upstream regulator of the FGF15-mediated gut-liver crosstalk that maintains bile and liver triglyceride homeostasis.


Assuntos
Ácidos e Sais Biliares/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Íleo/metabolismo , Metabolismo dos Lipídeos , Fígado/metabolismo , Hepatopatia Gordurosa não Alcoólica/metabolismo , Proteína 2 Semelhante ao Fator 7 de Transcrição/metabolismo , Animais , Colesterol 7-alfa-Hidroxilase/genética , Colesterol 7-alfa-Hidroxilase/metabolismo , Fatores de Crescimento de Fibroblastos/genética , Homeostase , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteína 2 Semelhante ao Fator 7 de Transcrição/genética
2.
Arterioscler Thromb Vasc Biol ; 39(2): 250-262, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30567484

RESUMO

Objective- TCF7L2 (transcription factor 7-like 2) is a Wnt-regulated transcription factor that maintains stemness and promotes proliferation in embryonic tissues and adult stem cells. Mice with a coronary artery disease-linked mutation in Wnt-coreceptor LRP6 (LDL receptor-related protein 6) exhibit vascular smooth muscle cell dedifferentiation and obstructive coronary artery disease, which are paradoxically associated with reduced TCF7L2 expression. We conducted a comprehensive study to explore the role of TCF7L2 in vascular smooth muscle cell differentiation and protection against intimal hyperplasia. Approach and Results- Using multiple mouse models, we demonstrate here that TCF7L2 promotes differentiation and inhibits proliferation of vascular smooth muscle cells. TCF7L2 accomplishes these effects by stabilization of GATA6 (GATA-binding protein 6) and upregulation of SM-MHC (smooth muscle cell myosin heavy chain) and cell cycle inhibitors. Accordingly, TCF7L2 haploinsufficient mice exhibited increased susceptibility to injury-induced hyperplasia, while mice overexpressing TCF7L2 were protected against injury-induced intimal hyperplasia compared with wild-type littermates. Consequently, the overexpression of TCF7L2 in LRP6 mutant mice rescued the injury-induced intimal hyperplasia. Conclusions- Our novel findings imply cell type-specific functional role of TCF7L2 and provide critical insight into mechanisms underlying the pathogenesis of intimal hyperplasia.


Assuntos
Plasticidade Celular , Fator de Transcrição GATA6/fisiologia , Músculo Liso Vascular/fisiologia , Miócitos de Músculo Liso/fisiologia , Proteína 2 Semelhante ao Fator 7 de Transcrição/fisiologia , Túnica Íntima/patologia , Animais , Células Cultivadas , Hiperplasia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Músculo Liso Vascular/citologia , Fator de Crescimento Derivado de Plaquetas/farmacologia
3.
Blood ; 121(13): 2553-62, 2013 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-23361909

RESUMO

B-cell lymphoma 11A (BCL11A) downregulation in human primary adult erythroid progenitors results in elevated expression of fetal γ-globin. Recent reports showed that BCL11A expression is activated by KLF1, leading to γ-globin repression. To study regulation of erythropoiesis and globin expression by KLF1 and BCL11A in an in vivo model, we used mice carrying a human ß-globin locus transgene with combinations of Klf1 knockout, Bcl11a floxed, and EpoR(Cre) knockin alleles. We found a higher percentage of reticulocytes in adult Klf1(wt/ko) mice and a mild compensated anemia in Bcl11a(cko/cko) mice. These phenotypes were more pronounced in compound Klf1(wt/ko)::Bcl11a(cko/cko) mice. Analysis of Klf1(wt/ko), Bcl11a(cko/cko), and Klf1(wt/ko)::Bcl11a(cko/cko) mutant embryos demonstrated increased expression of mouse embryonic globins during fetal development. Expression of human γ-globin remained high in Bcl11a(cko/cko) embryos during fetal development, and this was further augmented in Klf1(wt/ko)::Bcl11a(cko/cko) embryos. After birth, expression of human γ-globin and mouse embryonic globins decreased in Bcl11a(cko/cko) and Klf1(wt/ko)::Bcl11a(cko/cko) mice, but the levels remained much higher than those observed in control animals. Collectively, our data support an important role for the KLF1-BCL11A axis in erythroid maturation and developmental regulation of globin expression.


Assuntos
Proteínas de Transporte/genética , Eritropoese/genética , Genes de Troca/genética , Globinas/genética , Fatores de Transcrição Kruppel-Like/genética , Proteínas Nucleares/genética , Animais , Proteínas de Ligação a DNA , Embrião de Mamíferos , Eritropoese/fisiologia , Desenvolvimento Fetal/genética , Regulação da Expressão Gênica no Desenvolvimento , Rearranjo Gênico/genética , Rearranjo Gênico/fisiologia , Genes de Troca/fisiologia , Humanos , Camundongos , Camundongos Mutantes , Camundongos Transgênicos , Proteínas Repressoras , Reticulocitose/genética , Reticulocitose/fisiologia , Baço/citologia , Baço/embriologia , Baço/metabolismo
4.
Haematologica ; 100(11): 1396-406, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26294724

RESUMO

Expansion of erythroblasts from human peripheral blood mononuclear cells is 4- to 15-fold more efficient than that of CD34(+) cells purified from peripheral blood mononuclear cells. In addition, purified CD34(+) and CD34(-) populations from blood do not reconstitute this erythroid yield, suggesting a role for feeder cells present in blood mononuclear cells that increase hematopoietic output. Immunodepleting peripheral blood mononuclear cells for CD14(+) cells reduced hematopoietic stem and progenitor cell expansion. Conversely, the yield was increased upon co-culture of CD34(+) cells with CD14(+) cells (full contact or transwell assays) or CD34(+) cells re-constituted in conditioned medium from CD14(+) cells. In particular, CD14(++)CD16(+) intermediate monocytes/macrophages enhanced erythroblast outgrowth from CD34(+) cells. No effect of CD14(+) cells on erythroblasts themselves was observed. However, 2 days of co-culturing CD34(+) and CD14(+) cells increased CD34(+) cell numbers and colony-forming units 5-fold. Proliferation assays suggested that CD14(+) cells sustain CD34(+) cell survival but not proliferation. These data identify previously unrecognized erythroid and non-erythroid CD34(-) and CD34(+) populations in blood that contribute to the erythroid yield. A flow cytometry panel containing CD34/CD36 can be used to follow specific stages during CD34(+) differentiation to erythroblasts. We have shown modulation of hematopoietic stem and progenitor cell survival by CD14(+) cells present in peripheral blood mononuclear cells which can also be found near specific hematopoietic niches in the bone marrow.


Assuntos
Células Eritroides/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Receptores de Lipopolissacarídeos/metabolismo , Macrófagos/metabolismo , Monócitos/metabolismo , Sobrevivência Celular , Técnicas de Cocultura , Células Eritroides/citologia , Células-Tronco Hematopoéticas/citologia , Humanos , Macrófagos/citologia , Monócitos/citologia
5.
Mol Cell Proteomics ; 11(11): 1263-73, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22872859

RESUMO

Chromatin target of Prmt1 (Chtop) is a vertebrate-specific chromatin-bound protein that plays an important role in transcriptional regulation. As its mechanism of action remains unclear, we identified Chtop-interacting proteins using a biotinylation-proteomics approach. Here we describe the identification and initial characterization of Five Friends of Methylated Chtop (5FMC). 5FMC is a nuclear complex that can only be recruited by Chtop when the latter is arginine-methylated by Prmt1. It consists of the co-activator Pelp1, the Sumo-specific protease Senp3, Wdr18, Tex10, and Las1L. Pelp1 functions as the core of 5FMC, as the other components become unstable in the absence of Pelp1. We show that recruitment of 5FMC to Zbp-89, a zinc-finger transcription factor, affects its sumoylation status and transactivation potential. Collectively, our data provide a mechanistic link between arginine methylation and (de)sumoylation in the control of transcriptional activity.


Assuntos
Arginina/metabolismo , Cromatina/metabolismo , Proteínas Correpressoras/metabolismo , Cisteína Endopeptidases/metabolismo , Complexos Multiproteicos/metabolismo , Proteínas Nucleares/metabolismo , Proteína-Arginina N-Metiltransferases/metabolismo , Sumoilação , Fatores de Transcrição/metabolismo , Animais , Núcleo Celular/metabolismo , Regulação da Expressão Gênica , Humanos , Metilação , Camundongos , Modelos Biológicos , Peptídeo Hidrolases/metabolismo , Ligação Proteica , Estabilidade Proteica
6.
Haematologica ; 98(5): 696-704, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23100274

RESUMO

ß-thalassemia is caused by mutations in the ß-globin locus resulting in loss of, or reduced, hemoglobin A (adult hemoglobin, HbA, α2ß2) production. Hydroxyurea treatment increases fetal γ-globin (fetal hemoglobin, HbF, α2γ2) expression in postnatal life substituting for the missing adult ß-globin and is, therefore, an attractive therapeutic approach. Patients treated with hydroxyurea fall into three categories: i) 'responders' who increase hemoglobin to therapeutic levels; (ii) 'moderate-responders' who increase hemoglobin levels but still need transfusions at longer intervals; and (iii) 'non-responders' who do not reach adequate hemoglobin levels and remain transfusion-dependent. The mechanisms underlying these differential responses remain largely unclear. We generated RNA expression profiles from erythroblast progenitors of 8 responder and 8 non-responder ß-thalassemia patients. These profiles revealed that hydroxyurea treatment induced differential expression of many genes in cells from non-responders while it had little impact on cells from responders. Part of the gene program up-regulated by hydroxyurea in non-responders was already highly expressed in responders before hydroxyurea treatment. Baseline HbF expression was low in non-responders, and hydroxyurea treatment induced significant cell death. We conclude that cells from responders have adapted well to constitutive stress conditions and display a propensity to proceed to the erythroid differentiation program.


Assuntos
Adaptação Biológica , Células Precursoras Eritroides/citologia , Células Precursoras Eritroides/metabolismo , Hidroxiureia/uso terapêutico , Estresse Fisiológico , Talassemia beta/tratamento farmacológico , Talassemia beta/metabolismo , Fatores de Ribosilação do ADP/genética , Adaptação Biológica/genética , Apoptose/genética , Diferenciação Celular , Análise por Conglomerados , Inibidor de Quinase Dependente de Ciclina p15/genética , Inibidor p16 de Quinase Dependente de Ciclina/genética , Células Precursoras Eritroides/efeitos dos fármacos , Hemoglobina Fetal/genética , Hemoglobina Fetal/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Loci Gênicos , Hemoglobina A/metabolismo , Humanos , Estresse Fisiológico/genética , Resultado do Tratamento , Talassemia beta/genética , gama-Globinas/genética
7.
Am J Med Genet A ; 158A(8): 1857-64, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22736430

RESUMO

MYO15A is located at the DFNB3 locus on chromosome 17p11.2, and encodes myosin-XV, an unconventional myosin critical for the formation of stereocilia in hair cells of cochlea. Recessive mutations in this gene lead to profound autosomal recessive nonsyndromic hearing loss (ARNSHL) in humans and the shaker2 (sh2) phenotype in mice. Here, we performed a study on 140 Iranian families in order to determine mutations causing ARNSHL. The families, who were negative for mutations in GJB2, were subjected to linkage analysis. Eight of these families showed linkage to the DFNB3 locus, suggesting a MYO15A mutation frequency of 5.71% in our cohort of Iranian population. Subsequent sequencing of the MYO15A gene led to identification of 7 previously unreported mutations, including 4 missense mutations, 1 nonsense mutation, and 2 deletions in different regions of the myosin-XV protein.


Assuntos
Surdez/genética , Genes Recessivos , Mutação , Miosinas/genética , Mapeamento Cromossômico , Cromossomos Humanos Par 17 , Conexina 26 , Conexinas , Feminino , Humanos , Irã (Geográfico) , Masculino , Linhagem
8.
Clin Exp Pharmacol Physiol ; 39(6): 510-7, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22469229

RESUMO

1. Hydroxyurea (HU) is a drug used for the treatment of haemoglobinopathies. Hydroxyurea functions by upregulating γ-globin transcription and fetal haemoglobin (HbF) production in erythroid cells. The K562 erythroleukaemia cell line is widely used as a model system in which to study the mechanism of γ-globin induction by HU. However, the transcription factors required for the upregulation of γ-globin expression by HU in K562 cells have not been identified. Similarities between the HU and sodium butyrate (SB) pathways suggest cAMP response element-binding protein (CREB) 1 as a potential candidate. Thus, the aim of the present study was to investigate the possible role of CREB1 in the HU pathway. 2. Experiments were performed using transient and stable RNA interference (RNAi) to show that CREB1 is necessary for HU-mediated induction of γ-globin expression and haemoglobin production in K562 cells. 3. Furthermore, western blot analyses demonstrated that CREB1 becomes phosphorylated in a dose-dependent manner after HU (100-400 µmol/L) treatment of K562 cells for 72 h. 4. We also investigated role of a Gγ promoter CREB1 response element (G-CRE) in this pathway. Quantitative amplification refractory mutation system-polymerase chain reaction experiments were performed to demonstrate that HU induces the expression of both Gγ and Aγ in this cell line. In addition, electrophoretic mobility shift assays were used to show that levels of CREB1 complexes binding to the G-CRE site are increased following HU treatment and are decreased in CREB1-knockdown cells. 5. The results suggest that CREB1 is necessary for γ-globin induction by HU in K562 cells, a role that may be mediated, in part, through the G-CRE element.


Assuntos
Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/fisiologia , Regulação Neoplásica da Expressão Gênica , Hidroxiureia/farmacologia , gama-Globinas/biossíntese , gama-Globinas/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/fisiologia , Hemoglobinas/biossíntese , Humanos , Células K562 , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/fisiologia
9.
Haematologica ; 96(9): 1252-60, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21606163

RESUMO

BACKGROUND: In erythroblasts, the CoREST repressor complex is recruited to target promoters by the transcription factor Gfi1b, leading to repression of genes mainly involved in erythroid differentiation. Hmg20b is a subunit of CoREST, but its role in erythropoiesis has not yet been established. DESIGN AND METHODS: To study the role of Hmg20b in erythropoiesis, we performed knockdown experiments in a differentiation-competent mouse fetal liver cell line, and in primary mouse fetal liver cells. The effects on globin gene expression were determined. We used microarrays to investigate global gene expression changes induced by Hmg20b knockdown. Functional analysis was carried out on Hrasls3, an Hmg20b target gene. RESULTS: We show that Hmg20b depletion induces spontaneous differentiation. To identify the target genes of Hmg20b, microarray analysis was performed on Hmg20b knockdown cells and controls. In line with its association to the CoREST complex, we found that 85% (527 out of 620) of the deregulated genes are up-regulated when Hmg20b levels are reduced. Among the few down-regulated genes was Gfi1b, a known repressor of erythroid differentiation. Among the consistently up-regulated targets were embryonic ß-like globins and the phospholipase HRAS-like suppressor 3 (Hrasls3). We show that Hrasls3 expression is induced during erythroid differentiation and that knockdown of Hrasls3 inhibits terminal differentiation of proerythroblasts. CONCLUSIONS: We conclude that Hmg20b acts as an inhibitor of erythroid differentiation, through the down-regulation of genes involved in differentiation such as Hrasls3, and activation of repressors of differentiation such as Gfi1b. In addition, Hmg20b suppresses embryonic ß-like globins.


Assuntos
Células Eritroides/citologia , Células Eritroides/metabolismo , Eritropoese/genética , Proteínas de Grupo de Alta Mobilidade/metabolismo , Proteínas Repressoras/metabolismo , Animais , Proteínas de Ciclo Celular , Linhagem Celular Tumoral , Células Cultivadas , Proteínas Correpressoras , Proteínas de Ligação a DNA , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Células HeLa , Proteínas de Grupo de Alta Mobilidade/genética , Humanos , Camundongos , Complexos Multiproteicos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Fosfolipases A2 Independentes de Cálcio/genética , Proteínas Proto-Oncogênicas/genética , Proteínas Repressoras/genética
10.
Am J Med Genet A ; 155A(10): 2453-8, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21910243

RESUMO

Mutations in GJB2 are a major cause of autosomal recessive non-syndromic hearing loss (ARNSHL) in many populations. A single mutation of this gene (35delG) accounts for approximately 70% of GJB2 mutations that are associated with ARNSHL in Caucasians in many European countries and also in Iranian. In this study, we used PCR and restriction digestion to genotype five single nucleotide polymorphisms (SNPs) that define the genetic background of the 35delG mutation over an interval of 98 Kbp that includes the coding and flanking regions of GJB2. Two microsatellite markers, D13S175 and D13S141, were also analyzed in patients and controls. These data suggest that the 35delG mutation originated in northern Iran.


Assuntos
Conexinas/genética , Emigração e Imigração/história , Perda Auditiva/etnologia , Perda Auditiva/genética , Deleção de Sequência/genética , Conexina 26 , Feminino , Genes Recessivos/genética , Genética Populacional , História Antiga , Humanos , Irã (Geográfico)/epidemiologia , Masculino , Repetições de Microssatélites/genética , Polimorfismo de Nucleotídeo Único/genética
11.
Pediatr Hematol Oncol ; 27(5): 405-12, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20670167

RESUMO

alpha-Thalassemia is a common genetic disorder in Iran. However, no comprehensive data on epidemiology of severe forms of alpha-thalassemia, including hemoglobin H (HbH) or hydrops fetalis, is available in this population. This is a first case report of an Iranian family with large number of HbH individuals. The proband is a 48-year-old woman, referred to our center with anemia and no history of previous blood transfusions. Similar clinical phenotype has been observed in all of her 5 siblings, 2 of her 4 children, and her granddaughter, whose parents are first cousins. A reverse hybridization assay covering 21 alpha globin mutations was performed to determine the genotype in 11 members of this family and a fetus. HbH genotype was identified in 9 individuals, representing 3 generations, including a fetus. The high prevalence of alpha-thalassemia carriers together with the high rate of consanguineous marriages could lead to a large number of individuals with HbH or even hydrops fetalis in Iranian families. Therefore, to avoid the risk of having affected offspring, carrier detection, genetic counseling, and prenatal diagnosis would be of vital importance for individuals with low red blood cell (RBC) indices, normal iron status, and normal HbA(2) level, who are suspected to be alpha-thalassemia carriers.


Assuntos
Hemoglobina H/genética , Talassemia alfa/diagnóstico , Análise Mutacional de DNA , Família , Feminino , Testes Genéticos , Genótipo , Humanos , Hidropisia Fetal/diagnóstico , Irã (Geográfico) , Pessoa de Meia-Idade , Fenótipo , Gravidez , Diagnóstico Pré-Natal , Talassemia alfa/genética
12.
PLoS One ; 14(3): e0208659, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30921348

RESUMO

Krüppel-like factor 1 (KLF1) is an essential transcription factor for erythroid development, as demonstrated by Klf1 knockout mice which die around E14 due to severe anemia. In humans, >140 KLF1 variants, causing different erythroid phenotypes, have been described. The KLF1 Nan variant, a single amino acid substitution (p.E339D) in the DNA binding domain, causes hemolytic anemia and is dominant over wildtype KLF1. Here we describe the effects of the KLF1 Nan variant during fetal development. We show that Nan embryos have defects in erythroid maturation. RNA-sequencing of the KLF1 Nan fetal liver cells revealed that Exportin 7 (Xpo7) was among the 782 deregulated genes. This nuclear exportin is implicated in terminal erythroid differentiation; in particular it is involved in nuclear condensation. Indeed, KLF1 Nan fetal liver cells had larger nuclei and reduced chromatin condensation. Knockdown of XPO7 in wildtype erythroid cells caused a similar phenotype. We propose that reduced expression of XPO7 is partially responsible for the erythroid defects observed in KLF1 Nan erythroid cells.


Assuntos
Anemia Hemolítica/genética , Células Eritroides/citologia , Fatores de Transcrição Kruppel-Like/genética , Proteína ran de Ligação ao GTP/genética , Substituição de Aminoácidos , Animais , Diferenciação Celular , Células Cultivadas , Cromatina/metabolismo , Modelos Animais de Doenças , Embrião não Mamífero/metabolismo , Células Eritroides/metabolismo , Eritropoese , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Masculino , Camundongos , Análise de Sequência de RNA/métodos , Proteína ran de Ligação ao GTP/metabolismo
13.
Nat Genet ; 51(8): 1233-1243, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31358993

RESUMO

Factors that underlie the clustering of metabolic syndrome traits are not fully known. We performed whole-exome sequence analysis in kindreds with extreme phenotypes of early-onset atherosclerosis and metabolic syndrome, and identified novel loss-of-function mutations in the gene encoding the pancreatic elastase chymotrypsin-like elastase family member 2A (CELA2A). We further show that CELA2A is a circulating enzyme that reduces platelet hyperactivation, triggers both insulin secretion and degradation, and increases insulin sensitivity. CELA2A plasma levels rise postprandially and parallel insulin levels in humans. Loss of these functions by the mutant proteins provides insight into disease mechanisms and suggests that CELA2A could be an attractive therapeutic target.


Assuntos
Aterosclerose/patologia , Insulina/sangue , Ilhotas Pancreáticas/patologia , Síndrome Metabólica/patologia , Mutação , Elastase Pancreática/sangue , Elastase Pancreática/genética , Serina Endopeptidases/genética , Adulto , Idade de Início , Aterosclerose/sangue , Aterosclerose/etiologia , Estudos de Casos e Controles , Feminino , Predisposição Genética para Doença , Humanos , Resistência à Insulina , Ilhotas Pancreáticas/metabolismo , Desequilíbrio de Ligação , Masculino , Síndrome Metabólica/sangue , Síndrome Metabólica/etiologia , Pessoa de Meia-Idade , Linhagem , Ativação Plaquetária
14.
Genet Test ; 12(4): 549-56, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18939939

RESUMO

To improve the differentiation of thalassemia intermedia from other hemoglobinopathies in Iran, four known genetic mechanisms-XmnI (G)gamma polymorphism, inheritance of mild and silent beta-thalassemia alleles, delta beta deletion, and coinheritance of alpha- and beta-thalassemia-were investigated in 52 Iranian individuals suspected to have thalassemia intermedia based on clinical and hematological characteristics. Beta-globin mutations were studied using a reverse-hybridization assay and sequencing of the total beta-globin gene. The XmnI (G)gamma polymorphism, the Sicilian delta beta deletion, and four alpha-globin mutations (-a(3.7), -a(4.2), -(MED), aaa(anti-3.7)) were studied using PCR-based techniques. The inheritance of the XmnI (G)gamma polymorphism with severe beta-thalassemia alleles in the homozygous or compound heterozygous state was the predominant mechanism observed in 27 individuals (55.3%). In five cases, this status overlapped with the -a(3.7)/aa genotype. The second most frequent cause for thalassemia intermedia (14.8%) was the inheritance of mild beta-thalassemia alleles, including IVS-I-6 (T > C), -88 (C > A), and + 113 (A > G). In three subjects (4.3%) the Sicilian delta beta deletion was identified. HbS in association with beta-zero-thalassemia was found in three patients with thalassemia intermedia phenotype. In 11 cases (21.3%) no causative genetic alteration could be identified. Our results reflect the diversity underlying thalassemia intermedia, and the limitations of the applied clinical, hematological, and molecular approaches for correct diagnosis. Some of the unresolved cases will offer an opportunity to discover additional molecular mechanisms leading to thalassemia intermedia.


Assuntos
Subunidades de Hemoglobina/genética , Mutação , Talassemia/genética , Adolescente , Adulto , Alelos , Sequência de Bases , Criança , Análise Mutacional de DNA , Primers do DNA/genética , Feminino , Genótipo , Humanos , Irã (Geográfico) , Masculino , Pessoa de Meia-Idade , Fenótipo , Talassemia/sangue , Talassemia/classificação , Adulto Jovem , alfa-Globinas/genética , Talassemia alfa/sangue , Talassemia alfa/genética , Globinas beta/genética , Talassemia beta/sangue , Talassemia beta/genética , Globinas delta/genética , Talassemia delta/sangue , Talassemia delta/genética
15.
Hepatol Res ; 32(3): 154-7, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15963758

RESUMO

UNLABELLED: Autoimmune hepatitis (AIH) is a rare frequent, multiplex disorder with undefined etiology. Susceptibility to autoimmune hepatitis results from complex interactions between genetic and environmental factors. In this study, we investigated the involvement of C77G mutation in CD45 gene in patients with autoimmune hepatitis among Iranian population by genotyping this mutation in 70 patients and 140 healthy individuals. METHODS: : After amplifying exon 4 by Polymerase chain reaction, we genotyped this mutation with MspI restriction endonuclease among the studied population. RESULTS: : None of the cases with AIH was hetero or homozygote for C77G mutation. Controls had normal genotype except one of them who was heterozygote for C77G mutation. CONCLUSION: : Our results do not confirm the genetic link between C77G mutation and autoimmune hepatitis in Iranian population.

17.
Am J Physiol Endocrinol Metab ; 295(4): E929-37, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18682534

RESUMO

Fibroblast growth factor 23 (FGF23) is a hormone required for normal renal phosphate reabsorption. FGF23 gain-of-function mutations result in autosomal dominant hypophosphatemic rickets (ADHR), and FGF23 loss-of-function mutations cause familial hyperphosphatemic tumoral calcinosis (TC). In this study, we identified a novel recessive FGF23 TC mutation, a lysine (K) substitution for glutamine (Q) (160 C>A) at residue 54 (Q54K). To understand the molecular consequences of all known FGF23-TC mutants (H41Q, S71G, M96T, S129F, and Q54K), these proteins were stably expressed in vitro. Western analyses revealed minimal amounts of secreted intact protein for all mutants, and ELISA analyses demonstrated high levels of secreted COOH-terminal FGF23 fragments but low amounts of intact protein, consistent with TC patients' FGF23 serum profiles. Mutant protein function was tested and showed residual, yet decreased, bioactivity compared with wild-type protein. In examining the role of the FGF23 COOH-terminal tail (residues 180-251) in protein processing and activity, truncated mutants revealed that the majority of the residues downstream from the known FGF23 SPC protease site ((176)RXXR(179)/S(180)) were not required for protein secretion. However, residues adjacent to the RXXR site (between residues 188 and 202) were required for full bioactivity. In summary, we report a novel TC mutation and demonstrate a common defect of reduced FGF23 stability for all known FGF23-TC mutants. Finally, the majority of the COOH-terminal tail of FGF23 is not required for protein secretion but is required for full bioactivity.


Assuntos
Calcinose/genética , Fatores de Crescimento de Fibroblastos/genética , Fatores de Crescimento de Fibroblastos/metabolismo , Hiperfosfatemia/genética , Mutação/fisiologia , Adulto , Western Blotting , Calcinose/metabolismo , Calcinose/cirurgia , Calcitriol/sangue , Criança , Pré-Escolar , Análise Mutacional de DNA , Proteína 1 de Resposta de Crescimento Precoce/biossíntese , Proteína 1 de Resposta de Crescimento Precoce/genética , Éxons/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fator de Crescimento de Fibroblastos 23 , Humanos , Hiperfosfatemia/metabolismo , Masculino , Biologia Molecular , Mutagênese Sítio-Dirigida , N-Acetilgalactosaminiltransferases/genética , Hormônio Paratireóideo/sangue , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Polipeptídeo N-Acetilgalactosaminiltransferase
18.
Hemoglobin ; 31(4): 463-9, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17994380

RESUMO

Deltabeta-thalassemia (thal) is a disorder, characterized by increased levels of fetal hemoglobin (Hb F) in adult life. A considerable number of deletions of variable size and position in the beta-globin gene cluster are associated with the clinical manifestation of deltabeta-thal. In this study we have determined the presence of the eight most common deletions in Iranian patients. Thirty-two patients from 19 families were referred to the Kariminejad-Najmabadi Pathology and Genetics Center, Tehran, Iran (a private genetics center), within the past 3 years with elevated levels of Hb F and low mean corpuscular volume (MCV). After obtaining their informed consent, DNA was extracted from whole blood by the salting-out method. Detection of eight deletions was performed using polymerase chain reaction (PCR). These deletions included the hereditary persistence of fetal Hb (HPFH) 1 (Black) and 3 (Indian), Spanish (-114 kb), Sicilian (-13,377 bp), Chinese (G)gamma((A)gammadeltabeta)(0)-thal (-100 kb), Asian-Indian inversion-deletion (G)gamma((A)gammadeltabeta)(0)-thal, and the Turkish form of inversion-deletion (deltabeta)(0)-thal, as well as the Hbs Lepore, which are characterized by unequal crossovers between the delta- and beta-globin genes. We found the Sicilian (-13,377 bp) and Hb Lepore deletions as well as the Asian-Indian (G)gamma((A)gammadeltabeta)(0)-thal in 11 (57.89%), three (15.78%) and five (26.31%) families, respectively. None of the aforementioned deletions were found in one of the patients. This is the first study of the deletions involved in deltabeta-thal in Iranian patients. Our study highlights the importance of detecting these mutations for prenatal diagnosis carrier detection and genotype/phenotype prediction.


Assuntos
Testes Genéticos , Globinas/genética , Mutação , Deleção de Sequência , Talassemia beta/genética , Adolescente , Adulto , Pré-Escolar , Estudos de Coortes , Análise Mutacional de DNA , Família , Feminino , Hemoglobina Fetal/genética , Genótipo , Hemoglobinas Anormais/genética , Humanos , Irã (Geográfico) , Masculino , Pessoa de Meia-Idade , Família Multigênica , Fenótipo
19.
Am J Med Genet A ; 143A(20): 2390-6, 2007 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-17853462

RESUMO

Familial tumoral calcinosis (TC) is characterized by elevated serum phosphate concentrations, normal or elevated 1,25(OH)2 vitamin D, as well as periarticular and vascular calcifications. Recessive mutations in the mucin-like glycosyltransferase GalNAc transferase-3 (GALNT3) and the phosphaturic hormone fibroblast growth factor-23 (FGF23) have been shown to result in TC. In the present study, mutational analyses were performed on two patients with TC to determine the molecular basis of their diseases. Analysis of the first patient revealed a novel, homozygous base insertion (1102_1103insT) in GALNT3 exon 5 that results in a frameshift and premature stop codon (E375X). The second patient had a novel homozygous transition (1460 g>a) in GALNT3 exon 7, which caused a nonsense mutation (W487X). Both mutations are predicted to markedly truncate the mature GALNT3 protein product. Although the patients carry GALNT3 mutations, these individuals presented with low-normal serum concentrations of intact biologically active FGF23 and high levels of C-terminal FGF23. In order to discern a possible relationship between GALNT3 and FGF23 in TC, a comprehensive assessment of the reported TC mutations was also performed. In summary, we have detected novel GALNT3 mutations that result in familial TC, and show that disturbed serum FGF23 concentrations are present in our TC cases as well as in previously reported cases. These studies expand our current genetic understanding of familial TC, and support a pathophysiological association between GALNT3 and FGF23.


Assuntos
Calcinose/genética , Mutação , N-Acetilgalactosaminiltransferases/genética , Sequência de Aminoácidos , Sequência de Bases , Calcinose/patologia , Análise Mutacional de DNA , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/sangue , Fatores de Crescimento de Fibroblastos/genética , Fatores de Crescimento de Fibroblastos/metabolismo , Humanos , Dados de Sequência Molecular , N-Acetilgalactosaminiltransferases/sangue , N-Acetilgalactosaminiltransferases/metabolismo , Polipeptídeo N-Acetilgalactosaminiltransferase
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA