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

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Hum Reprod ; 36(2): 506-518, 2021 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-33313884

RESUMEN

STUDY QUESTION: Is there an (epi)genetic basis in patients with central precocious puberty (CPP) associated with multiple anomalies that unmasks underlying mechanisms or reveals novel genetic findings related to human pubertal control? SUMMARY ANSWER: In a group of 36 patients with CPP associated with multiple phenotypes, pathogenic or likely pathogenic (epi)genetic defects were identified in 12 (33%) patients, providing insights into the genetics of human pubertal control. WHAT IS KNOWN ALREADY: A few studies have described patients with CPP associated with multiple anomalies, but without making inferences on causalities of CPP. Genetic-molecular studies of syndromic cases may reveal disease genes or mechanisms, as the presentation of such patients likely indicates a genetic disorder. STUDY DESIGN, SIZE, DURATION: This translational study was based on a genetic-molecular analysis, including genome-wide high throughput methodologies, for searching structural or sequence variants implicated in CPP and DNA methylation analysis of candidate regions. PARTICIPANTS/MATERIALS, SETTING, METHODS: A cohort of 197 patients (188 girls) with CPP without structural brain lesions was submitted to a detailed clinical evaluation, allowing the selection of 36 unrelated patients (32 girls) with CPP associated with multiple anomalies. Pathogenic allelic variants of genes known to cause monogenic CPP (KISS1R, KISS1, MKRN3 and DLK1) had been excluded in the entire cohort (197 patients). All selected patients with CPP associated with multiple anomalies (n = 36) underwent methylation analysis of candidate regions and chromosomal microarray analysis. A subset (n = 9) underwent whole-exome sequencing, due to presenting familial CPP and/or severe congenital malformations and neurocognitive abnormalities. MAIN RESULTS AND THE ROLE OF CHANCE: Among the 36 selected patients with CPP, the more prevalent associated anomalies were metabolic, growth and neurocognitive conditions. In 12 (33%) of them, rare genetic abnormalities were identified: six patients presented genetic defects in loci known to be involved with CPP (14q32.2 and 7q11.23), whereas the other six presented defects in candidate genes or regions. In detail, three patients presented hypomethylation of DLK1/MEG3:IG-DMR (14q32.2 disruption or Temple syndrome), resulting from epimutation (n = 1) or maternal uniparental disomy of chromosome 14 (n = 2). Seven patients presented pathogenic copy number variants: three with de novo 7q11.23 deletions (Williams-Beuren syndrome), three with inherited Xp22.33 deletions, and one with de novo 1p31.3 duplication. Exome sequencing revealed potential pathogenic variants in two patients: a sporadic female case with frameshift variants in TNRC6B and AREL1 and a familial male case with a missense substitution in UGT2B4 and a frameshift deletion in MKKS. LIMITATIONS, REASONS FOR CAUTION: The selection of patients was based on a retrospective clinical characterization, lacking a longitudinal inclusion of consecutive patients. In addition, future studies are needed, showing the long-term (mainly reproductive) outcomes in the included patients, as most of them are not in adult life yet. WIDER IMPLICATIONS OF THE FINDINGS: The results highlighted the relevance of an integrative clinical-genetic approach in the elucidation of mechanisms and factors involved in pubertal control. Chromosome 14q32.2 disruption indicated the loss of imprinting of DLK1 as a probable mechanism of CPP. Two other chromosomal regions (7q11.23 and Xp22.33) represented new candidate loci potentially involved in this disorder of pubertal timing. STUDY FUNDING/COMPETING INTEREST(S): This work was supported by grant number 2018/03198-0 (to A.P.M.C.) and grant number 2013/08028-1 (to A.C.V.K) from the São Paulo Research Foundation (FAPESP), and grant number 403525/2016-0 (to A.C.L.) and grant number 302849/2015-7 (to A.C.L.) and grant number 141625/2016-3 (to A.C.V.K) from the National Council for Scientific and Technological Development (CNPq). The authors have nothing to disclose. TRIAL REGISTRATION NUMBER: N/A.


Asunto(s)
Pubertad Precoz , Adulto , Brasil , Femenino , Pruebas Genéticas , Humanos , Masculino , Pubertad , Pubertad Precoz/genética , Proteínas de Unión al ARN , Estudios Retrospectivos , Ubiquitina-Proteína Ligasas
2.
Hum Mutat ; 40(11): 2033-2043, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31231873

RESUMEN

Isolated growth hormone deficiency (IGHD) is a rare condition mainly caused by mutations in GH1. The aim of this study was to assess the contribution of GHRHR mutations to IGHD in an unusually large group of patients. All GHRHR coding exons and flanking intronic regions were sequenced in 312 unrelated patients with nonsyndromic IGHD. Functional consequences of all newly identified missense variants were assessed in vitro (i.e., study of the expression of recombinant GHRHRs and their ability to activate the cyclic adenosine monophosphate (cAMP) signaling pathway). Genotype-phenotype correlation analyses were performed according to the nature of the identified mutation. We identified 20 different disease-causing GHRHR mutations (truncating and missense loss-of-function mutations), among which 15 are novel, in 24 unrelated patients. Of note, about half (13/24) of those patients represent sporadic cases. The clinical phenotype of patients with at least one missense GHRHR mutation was found to be indistinguishable from that of patients with bi-allelic truncating mutations. This study, which unveils disease-causing GHRHR mutations in 8% (24/312) of IGHD cases, identifies GHRHR as the second IGHD gene most frequently involved after GH1. The finding that 8% of IGHD cases without GH1 mutations are explained by GHRHR molecular defects (including missense mutations), together with the high proportion of sporadic cases among those patients, has important implications for genetic counseling.


Asunto(s)
Enanismo Hipofisario/genética , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Mutación , Receptores de Neuropéptido/genética , Receptores de Hormona Reguladora de Hormona Hipofisaria/genética , Alelos , Secuencia de Aminoácidos , Sustitución de Aminoácidos , AMP Cíclico , Análisis Mutacional de ADN , Enanismo Hipofisario/diagnóstico , Femenino , Genotipo , Hormona de Crecimiento Humana/genética , Humanos , Masculino , Linaje , Receptores de Neuropéptido/química , Receptores de Hormona Reguladora de Hormona Hipofisaria/química
3.
Hum Mol Genet ; 25(3): 472-83, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26612202

RESUMEN

POU1F1, a pituitary-specific POU-homeo domain transcription factor, plays an essential role in the specification of the somatotroph, lactotroph and thyrotroph lineages and in the activation of GH1, PRL and TSHß transcription. Individuals with mutations in POU1F1 present with combined deficiency of GH, PRL and TSH. Here, we identified a heterozygous missense mutation with evidence of pathogenicity, at the POU1F1 locus, in a large family in which an isolated growth hormone deficiency segregates as an autosomal dominant trait. The corresponding p.Pro76Leu mutation maps to a conserved site within the POU1F1 transactivation domain. Bandshift assays revealed that the mutation alters wild-type POU1F1 binding to cognate sites within the hGH-LCR and hGH1 promoter, but not to sites within the PRL promoter, and it selectively increases binding affinity to sites within the hGH-LCR. Co-immunoprecipitation studies reveal that this substitution enhances interactions of POU1F1 with three of its cofactors, PITX1, LHX3a and ELK1, and that residue 76 plays a critical role in these interactions. The insertion of the mutation at the mouse Pou1f1 locus results in a dramatic loss of protein expression despite normal mRNA concentrations. Mice heterozygous for the p.Pro76Leu mutation were phenotypically normal while homozygotes demonstrated a dwarf phenotype. Overall, this study unveils the involvement of POU1F1 in dominantly inherited isolated GH deficiency and demonstrates a significant impact of the Pro76Leu mutation on DNA-binding activities, alterations in transactivating functions and interactions with cofactors. Our data further highlight difficulties in modeling human genetic disorders in the mouse despite apparent conservation of gene expression pathways and physiologic functions.


Asunto(s)
Enanismo Hipofisario/genética , Mutación Missense , Carácter Cuantitativo Heredable , Factor de Transcripción Pit-1/genética , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Secuencia de Bases , Enanismo Hipofisario/metabolismo , Enanismo Hipofisario/patología , Femenino , Regulación de la Expresión Génica , Genes Dominantes , Sitios Genéticos , Hormona del Crecimiento/genética , Hormona del Crecimiento/metabolismo , Heterocigoto , Humanos , Proteínas con Homeodominio LIM/genética , Proteínas con Homeodominio LIM/metabolismo , Masculino , Ratones , Datos de Secuencia Molecular , Factores de Transcripción Paired Box/genética , Factores de Transcripción Paired Box/metabolismo , Linaje , Hipófisis/metabolismo , Hipófisis/patología , Prolactina/genética , Prolactina/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , Transducción de Señal , Factor de Transcripción Pit-1/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteína Elk-1 con Dominio ets/genética , Proteína Elk-1 con Dominio ets/metabolismo
4.
Clin Endocrinol (Oxf) ; 82(6): 876-84, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25557026

RESUMEN

BACKGROUND/OBJECTIVES: Congenital hypopituitarism is a rare disease which, for most patients, has no identified molecular cause. We aimed to document the molecular basis of growth retardation in a Moroccan cohort. DESIGN/PATIENTS: 80 index cases [54 with isolated growth hormone deficiency (IGHD), 26 with combined pituitary hormone deficiency (CPHD)] were screened for molecular defects in GH1 (including LCR-GH1), GHRHR, GHSR, GHRH, PROP1, POU1F1, HESX1, LHX3, LHX4 and SOX3. RESULTS: Five different deleterious mutations were identified in 14 patients from eight families. In the IGHD group, three genes were found to be involved: GH1, GHRHR and GHSR. In the CPHD group, PROP1 was the only mutated gene. In addition, two heterozygous variations whose deleterious effect remains to be demonstrated were identified (in GH1 and LHX4), and two polymorphisms (missense variations) were detected (in LHX3 and in GHSR). The prevalence of mutations in this Moroccan GHD cohort was 10% (8/80), 11·1% (6/54) in the IGHD group and 7·7% (2/26) in the CPHD group. CONCLUSION: This is the first molecular screening of congenital GHD in a Moroccan population and, like other studies, mutations were preferentially identified in familial cases (75%); mutations in genes such as POU1F1, HESX1, SOX3, LHX3 and LHX4 are extremely rare. The p.R73C PROP1 mutation was the most frequent mutation in CPHD; this should be the first one to screen in this population. Our results should contribute to a better diagnosis and management of this heterogeneous disease condition.


Asunto(s)
Proteínas de Homeodominio/genética , Hormona de Crecimiento Humana , Hipopituitarismo , Adolescente , Estatura/genética , Niño , Estudios de Cohortes , Femenino , Predisposición Genética a la Enfermedad , Hormona de Crecimiento Humana/sangre , Hormona de Crecimiento Humana/deficiencia , Humanos , Hipopituitarismo/sangre , Hipopituitarismo/genética , Masculino , Marruecos , Mutación , Prevalencia
5.
Cells ; 11(12)2022 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-35741015

RESUMEN

In the 30 years since the first report of parental imprinting in insulin-like growth factor 2 (Igf2) knockout mouse models, we have learnt much about the structure of this protein, its role and regulation. Indeed, many animal and human studies involving innovative techniques have shed light on the complex regulation of IGF2 expression. The physiological roles of IGF-II have also been documented, revealing pleiotropic tissue-specific and developmental-stage-dependent action. Furthermore, in recent years, animal studies have highlighted important interspecies differences in IGF-II function, gene expression and regulation. The identification of human disorders due to impaired IGF2 gene expression has also helped to elucidate the major role of IGF-II in growth and in tumor proliferation. The Silver-Russell and Beckwith-Wiedemann syndromes are the most representative imprinted disorders, as they constitute both phenotypic and molecular mirrors of IGF2-linked abnormalities. The characterization of patients with either epigenetic or genetic defects altering IGF2 expression has confirmed the central role of IGF-II in human growth regulation, particularly before birth, and its effects on broader body functions, such as metabolism or tumor susceptibility. Given the long-term health impact of these rare disorders, it is important to understand the consequences of IGF2 defects in these patients.


Asunto(s)
Síndrome de Beckwith-Wiedemann , Síndrome de Silver-Russell , Animales , Síndrome de Beckwith-Wiedemann/genética , Epigenómica , Impresión Genómica , Humanos , Factor II del Crecimiento Similar a la Insulina/genética , Factor II del Crecimiento Similar a la Insulina/metabolismo , Ratones , Síndrome de Silver-Russell/genética
6.
Int J Oral Sci ; 14(1): 19, 2022 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-35368018

RESUMEN

Parental imprinting is an epigenetic process leading to monoallelic expression of certain genes depending on their parental origin. Imprinting diseases are characterized by growth and metabolic issues starting from birth to adulthood. They are mainly due to methylation defects in imprinting control region that drive the abnormal expression of imprinted genes. We currently lack relevant animal or cellular models to unravel the pathophysiology of growth failure in these diseases. We aimed to characterize the methylation of imprinting regions in dental pulp stem cells and during their differentiation in osteogenic cells (involved in growth regulation) to assess the interest of this cells in modeling imprinting diseases. We collected dental pulp stem cells from five controls and four patients (three with Silver-Russell syndrome and one with Beckwith-Wiedemann syndrome). Methylation analysis of imprinting control regions involved in these syndromes showed a normal profile in controls and the imprinting defect in patients. These results were maintained in dental pulp stem cells cultured under osteogenic conditions. Furthermore, we confirmed the same pattern in six other loci involved in imprinting diseases in humans. We also confirmed monoallelic expression of H19 (an imprinted gene) in controls and its biallelic expression in one patient. Extensive imprinting control regions methylation analysis shows the strong potential of dental pulp stem cells in modeling imprinting diseases, in which imprinting regions are preserved in culture and during osteogenic differentiation. This will allow to perform in vitro functional and therapeutic tests in cells derived from dental pulp stem cells and generate other cell-types.


Asunto(s)
Metilación de ADN , Osteogénesis , Adulto , Animales , Pulpa Dental , Impresión Genómica , Humanos , Osteogénesis/genética , Células Madre
7.
Front Endocrinol (Lausanne) ; 13: 836731, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35295988

RESUMEN

Detecting SGA (small for gestational age) during pregnancy improves the fetal and neonatal prognosis. To date, there is no valid antenatal biomarker of SGA used in clinical practice. Maternal circulating DLK1 (delta-like non-canonical notch ligand 1) levels have been shown to be significantly lower in pregnant women at 36 weeks of gestation (WG) who delivered a SGA newborn than in controls. Data in the literature are contradictory on the association between maternal circulating DLK1 levels and placental vascular dysfunction. The objective was to determine if maternal DLK1 levels in the second trimester of pregnancy are predictive of SGA, and to assess whether the measurement of DLK1 levels in maternal blood could be a means to distinguish SGA with placental vascular dysfunction from that due to other causes. We conducted a nested cased-control study within the EDEN mother-child cohort. 193 SGA (birth weight < 10th percentile) and 370 mother-child control (birth weight between the 25th and 75th percentile) matched pairs were identified in the EDEN cohort. Maternal circulating DLK1 levels at 26 WG were significantly lower for children born SGA than for controls (27.7 ± 8.7 ng/mL vs 30.4 ± 10.6 ng/mL, p = 0.001). Maternal blood DLK1 levels in the first quartile (DLK1 < 22.85 ng/mL) were associated with an odds ratio for SGA of 1.98 [1.15 - 3.37]. DLK1 was less predictive of SGA than ultrasound, with an area under the curve of 0.578. Maternal circulating DLK1 levels were not significantly different in cases of SGA with signs of placental vascular dysfunction (n = 63, 27.1 ± 9.2 ng/mL) than in those without placental dysfunction (n = 129, 28.0 ± 8.5 ng/mL, p = 0.53). The level of circulating DLK1 is reduced in the second trimester of pregnancy in cases of SGA at birth, independently of signs of placental vascular dysfunction. However, DLK1 alone cannot predict the risk of SGA.


Asunto(s)
Placenta , Ultrasonografía Prenatal , Peso al Nacer , Proteínas de Unión al Calcio , Femenino , Retardo del Crecimiento Fetal , Edad Gestacional , Humanos , Recién Nacido , Proteínas de la Membrana , Embarazo , Tercer Trimestre del Embarazo , Estudios Prospectivos
8.
Clin Endocrinol (Oxf) ; 75(2): 214-9, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21521297

RESUMEN

CONTEXT: POU1F1 encodes a pituitary-specific homeodomain transcription factor that is crucial for development and differentiation of anterior pituitary cell types producing GH, TSH and PRL. Although the first mutations in humans were reported in 1992, to date, less than 25 different mutations of POU1F1 have been identified worldwide. OBJECTIVES: To describe the long-term follow-up of a 22-year-old male of Israeli Arab Muslim origin, born to a consanguineous union, with congenital hypothyroidism, who presented with life-threatening hypoglycaemic episodes and severe growth retardation from infancy. To identify the molecular basis of this severe disease. MAIN OUTCOME MEASURES: Endocrine investigations, neuroimaging, sequencing of POU1F1 and assessment of the identified mutated POU1F1's ability to transactivate three specific targets (POU1F1, TSHß and PRL). RESULTS: Central hypothyroidism was diagnosed at the age of 2 months and GH and PRL deficiencies were documented at 9 months. MRI at 14 years revealed a hypoplastic adenohypophysis. The patient underwent spontaneous but delayed puberty. A novel disease-causing mutation (c.502insT) was identified in the homozygous state in exon 4 of POU1F1. This insertion results in a frameshift introducing an early termination codon at position 174 (p.Thr168IlefsX7), leading to a severely truncated protein lacking the entire homeodomain. This mutation abolishes POU1F1's transactivation properties on three target promoters. CONCLUSION: This study, which identifies a novel loss-of-function mutation in POU1F1, describes the phenotype of a rare condition in a patient followed from the first weeks of life to adulthood. The severity of the central hypothyroidism should alert clinicians to assess other pituitary axes, in particular GH and prolactin.


Asunto(s)
Hipopituitarismo/genética , Mutación , Factor de Transcripción Pit-1/genética , Adolescente , Niño , Preescolar , Estudios de Seguimiento , Trastornos del Crecimiento , Hormona del Crecimiento , Humanos , Hipoglucemia , Hipopituitarismo/metabolismo , Hipopituitarismo/patología , Hipotiroidismo , Lactante , Masculino , Neuroimagen , Prolactina , Adulto Joven
9.
Eur J Hum Genet ; 29(12): 1756-1761, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34276055

RESUMEN

Silver-Russell syndrome (SRS) is a rare imprinting disorder associated with prenatal and postnatal growth retardation. Loss of methylation (LOM) on chromosome 11p15 is observed in 40 to 60% of patients and maternal uniparental disomy (mUPD) for chromosome 7 (upd(7)mat) in ~5 to 10%. Patients with LOM or mUPD 14q32 can present clinically as SRS. Delta like non-canonical Notch ligand 1 (DLK1) is one of the imprinted genes expressed from chromosome 14q32. Dlk1-null mice display fetal growth restriction (FGR) but no genetic defects of DLK1 have been described in human patients born small for gestational age (SGA). We screened a cohort of SGA patients with a SRS phenotype for DLK1 variants using a next-generation sequencing (NGS) approach to search for new molecular defects responsible for SRS. Patients born SGA with a clinical suspicion of SRS and normal methylation by molecular testing at the 11p15 or 14q32 loci and upd(7)mat were screened for DLK1 variants using targeted NGS. Among 132 patients, only two rare variants of DLK1 were identified (NM_003836.6:c.103 G > C (p.(Gly35Arg) and NM_003836.6: c.194 A > G p.(His65Arg)). Both variants were inherited from the mother of the patients, which does not favor a role in pathogenicity, as the mono-allelic expression of DLK1 is from the paternal-inherited allele. We did not identify any pathogenic variants in DLK1 in a large cohort of SGA patients with a SRS phenotype. DLK1 variants are not a common cause of SGA.


Asunto(s)
Proteínas de Unión al Calcio/genética , Recién Nacido Pequeño para la Edad Gestacional , Proteínas de la Membrana/genética , Síndrome de Silver-Russell/genética , Femenino , Humanos , Recién Nacido , Mutación , Fenotipo , Síndrome de Silver-Russell/diagnóstico
10.
PLoS Genet ; 2(9): e138, 2006 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-16948532

RESUMEN

Splice site selection is a key element of pre-mRNA splicing. Although it is known to involve specific recognition of short consensus sequences by the splicing machinery, the mechanisms by which 5' splice sites are accurately identified remain controversial and incompletely resolved. The human F7 gene contains in its seventh intron (IVS7) a 37-bp VNTR minisatellite whose first element spans the exon7-IVS7 boundary. As a consequence, the IVS7 authentic donor splice site is followed by several cryptic splice sites identical in sequence, referred to as 5' pseudo-sites, which normally remain silent. This region, therefore, provides a remarkable model to decipher the mechanism underlying 5' splice site selection in mammals. We previously suggested a model for splice site selection that, in the presence of consecutive splice consensus sequences, would stimulate exclusively the selection of the most upstream 5' splice site, rather than repressing the 3' following pseudo-sites. In the present study, we provide experimental support to this hypothesis by using a mutational approach involving a panel of 50 mutant and wild-type F7 constructs expressed in various cell types. We demonstrate that the F7 IVS7 5' pseudo-sites are functional, but do not compete with the authentic donor splice site. Moreover, we show that the selection of the 5' splice site follows a scanning-type mechanism, precluding competition with other functional 5' pseudo-sites available on immediate sequence context downstream of the activated one. In addition, 5' pseudo-sites with an increased complementarity to U1snRNA up to 91% do not compete with the identified scanning mechanism. Altogether, these findings, which unveil a cell type-independent 5'-3'-oriented scanning process for accurate recognition of the authentic 5' splice site, reconciliate apparently contradictory observations by establishing a hierarchy of competitiveness among the determinants involved in 5' splice site selection.


Asunto(s)
Mamíferos/genética , Sitios de Empalme de ARN/genética , Empalme del ARN/genética , Animales , Secuencia de Bases , Células CHO , Cricetinae , Cricetulus , Exones/genética , Humanos , Intrones/genética , Datos de Secuencia Molecular , Mutación/genética , ARN Nuclear Pequeño/metabolismo
11.
Sci Adv ; 5(2): eaau9425, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30801013

RESUMEN

Imprinting disorders (IDs) often affect growth in humans, leading to diseases with overlapping features, regardless of the genomic region affected. IDs related to hypomethylation of the human 14q32.2 region and its DLK1/MEG3 domain are associated with Temple syndrome (TS14). TS14 is a rare type of growth retardation, the clinical signs of which overlap considerably with those of Silver-Russell syndrome (SRS), another ID related to IGF2 down-regulation at 11p15.5 region. We show that 14q32.2 hypomethylation affects expression, not only for genes at this locus but also for other imprinted genes, and especially lowers IGF2 levels at 11p15.5. Furthermore, expression of nonimprinted genes is also affected, some of which are also deregulated in SRS patients. These findings highlight the epigenetic regulation of gene expression at the DLK1/MEG3 domain. Expression profiling of TS14 and SRS patients highlights common signatures, which may account for the clinical overlap observed between TS14 and SRS.


Asunto(s)
Proteínas de Unión al Calcio , Cromosomas Humanos Par 11 , Cromosomas Humanos Par 14 , Metilación de ADN , Epigénesis Genética , Impresión Genómica , Proteínas de la Membrana , Síndrome de Silver-Russell , Proteínas de Unión al Calcio/biosíntesis , Proteínas de Unión al Calcio/genética , Cromosomas Humanos Par 11/genética , Cromosomas Humanos Par 11/metabolismo , Cromosomas Humanos Par 14/genética , Cromosomas Humanos Par 14/metabolismo , Femenino , Humanos , Masculino , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/genética , Síndrome de Silver-Russell/genética , Síndrome de Silver-Russell/metabolismo
12.
J Clin Endocrinol Metab ; 102(1): 290-301, 2017 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-27820671

RESUMEN

Context: LHX4 encodes a LIM-homeodomain transcription factor that is implicated in early pituitary development. In humans, only 13 heterozygous LHX4 mutations have been associated with congenital hypopituitarism. Objective: The aims of this study were to evaluate the prevalence of LHX4 mutations in patients with hypopituitarism, to define the associated phenotypes, and to characterize the functional impact of the identified variants and the respective role of the 2 LIM domains of LHX4. Design and Patients: We screened 417 unrelated patients with isolated growth hormone deficiency or combined pituitary hormone deficiency associated with ectopic posterior pituitary and/or sella turcica anomalies for LHX4 mutations (Sanger sequencing). In vitro studies were performed to assess the functional consequences of the identified variants. Results: We identified 7 heterozygous variations, including p.(Tyr131*), p.(Arg48Thrfs*104), c.606+1G>T, p.Arg65Val, p.Thr163Pro, p.Arg221Gln, and p.Arg235Gln), that were associated with variable expressivity; 5 of the 7 were also associated with incomplete penetrance. The p.(Tyr131*), p.(Arg48Thrfs*104), p.Ala65Val, p.Thr163Pro, and p.Arg221Gln LHX4 variants are unable to transactivate the POU1F1 and GH promoters. As suggested by transactivation, subcellular localization, and protein-protein interaction studies, p.Arg235Gln is probably a rare polymorphism. Coimmunoprecipitation studies identified LHX3 as a potential protein partner of LHX4. As revealed by functional studies of LIM-defective recombinant LHX4 proteins, the LIM1 and LIM2 domains are not redundant. Conclusion: This study, performed in the largest cohort of patients screened so far for LHX4 mutations, describes 6 disease-causing mutations that are responsible for congenital hypopituitarism. LHX4 mutations were found to be associated with variable expressivity, and most of them with incomplete penetrance; their contribution to pituitary deficits that are associated with an ectopic posterior pituitary and/or a sella turcica defect is ∼1.4% in the 417 probands tested.


Asunto(s)
Hipopituitarismo/genética , Proteínas con Homeodominio LIM/genética , Mutación/genética , Factores de Transcripción/genética , Adolescente , Secuencia de Aminoácidos , Biomarcadores/metabolismo , Western Blotting , Niño , Preescolar , Estudios de Cohortes , Femenino , Estudios de Seguimiento , Humanos , Inmunoprecipitación , Lactante , Recién Nacido , Masculino , Linaje , Pronóstico , Homología de Secuencia de Aminoácido
13.
J Clin Endocrinol Metab ; 91(11): 4528-36, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16940453

RESUMEN

CONTEXT: Hesx1 is one of the earliest homeodomain transcription factors expressed during pituitary development. Very few HESX1 mutations have been identified in humans; although in those cases the disease phenotype shows considerable variability, all but one of the patients display an ectopic posterior pituitary and/or optic nerve abnormalities. OBJECTIVE: The objectives of the study were to describe the complex phenotype associated with the panhypopituitarism of two unrelated Italian patients who, at birth, presented with hypoglycemic seizures and respiratory distress complicated by shock, in a familial context of neonatal death in one family and spontaneous miscarriage in both families and to identify the molecular basis of this unusual syndrome. MAIN OUTCOME MEASURES: Magnetic resonance imaging of the pituitary region, study of HESX1 gene and transcripts, and assessment of the ability of mutated HESX1 proteins to repress transcription were measured. RESULTS: Magnetic resonance imaging examination showed an anterior pituitary aplasia in a flat sella turcica and a normally located posterior pituitary in both patients. A constellation of extrapituitary developmental defects were found in the two patients, but without any optic nerve abnormalities. Sequencing of HESX1 exons and their flanking intronic regions revealed two different homozygous mutations. A frameshift (c.449_450delAC) was identified in one case, whereas the other patient carried a splice defect (c.357 + 2Tb > C) confirmed by the study of HESX1 transcripts. If translated, these mutations would lead to the synthesis of truncated proteins partly or entirely lacking the homeodomain, with no transcriptional repression, as shown by their inability to inhibit PROP1 activity. CONCLUSIONS: These observations reveal two novel HESX1 mutations in a so-far-undescribed disease phenotype characterized by a life-threatening neonatal condition associated with anterior pituitary aplasia, in the absence of ectopic posterior pituitary and optic nerve abnormalities, two features classically associated with HESX1 defects.


Asunto(s)
Proteínas de Homeodominio/genética , Nervio Óptico/anatomía & histología , Fenotipo , Enfermedades de la Hipófisis/genética , Neurohipófisis/anatomía & histología , Células Cultivadas , Niño , Preescolar , Análisis Mutacional de ADN , Femenino , Humanos , Lactante , Recién Nacido , Proteínas Mutantes/metabolismo , Mutación , Linaje , Procesamiento Postranscripcional del ARN , Factores de Transcripción/metabolismo , Transcripción Genética , Transfección
14.
Hum Mutat ; 25(5): 503, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15841484

RESUMEN

The pathophysiology of combined pituitary hormone deficiency is just beginning to be elucidated. None of the genes known to be necessary for pituitary development has so far been involved in pituitary gland aplasia in humans. Among these, Hesx1/HESX1, which encodes a homeobox transcription factor, has been shown to be essential for normal forebrain development in mice, and HESX1 mutations in humans have been associated with various pituitary hormone deficiencies usually combined with optic nerve anomalies. Here we have investigated a consanguineous family in which two siblings displayed a complete absence of the anterior pituitary revealed by a deficit in all anterior pituitary hormones. One patient, who also has retinal coloboma, carries a HESX1 defect in the homozygous state: an Alu insertion in exon 3, a sequence that encodes the major part of the homeodomain. The Alu-containing HESX1 allele generates a major transcript lacking this exon, and a minor one in which exons 2 and 3 are skipped, predicting severely truncated proteins. This observation, which combines pituitary aplasia and retinal coloboma, further illustrates the heterogeneity of HESX1-associated disease phenotypes. Anterior pituitary aplasia is a new example of a human disease caused by a germline retrotransposition event involving an Alu sequence.


Asunto(s)
Elementos Alu/genética , Proteínas de Homeodominio/genética , Adenohipófisis/anomalías , Hormonas Hipofisarias/deficiencia , Adolescente , Alelos , Secuencia de Bases , Coloboma/complicaciones , Coloboma/patología , Consanguinidad , Análisis Mutacional de ADN , Femenino , Humanos , Recién Nacido , Masculino , Datos de Secuencia Molecular , Nervio Óptico/anomalías , Nervio Óptico/patología , Linaje , Hipófisis/patología
17.
J Clin Endocrinol Metab ; 97(3): E503-9, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22238406

RESUMEN

CONTEXT: Only 11 mutations have been reported in the transcription factor LHX3, known to be important for the development of the pituitary and motor neurons. All patients were homozygous, with various syndromic forms of combined pituitary hormone deficiency (CPHD), hampering to allocate, in these consanguineous patients, the respective contribution of LHX3 and additional genes to each symptom. OBJECTIVE: The aim of the study was to report the family history and the molecular basis of a nonconsanguineous patient with syndromic CPHD. PATIENT: The patient, who presented at birth with respiratory distress, had a syndromic CPHD, including severe scoliosis, and normal intelligence. His father and paternal grandmother displayed limited head rotation. RESULTS: Two new LHX3 defects were identified. The paternally inherited c.252-3C>G mutation, which disrupts an acceptor splice site, would lead to severely truncated proteins containing a single LIM domain, resembling LIM-only proteins. Coexpression studies revealed the dominant-negative effect of this LIM-only protein over the wild-type LHX3. The maternally inherited p.Cys118Tyr mutation results in partial loss of transcriptional activity and synergy with POU1F1. Given the severity of the patient's phenotype, two prenatal diagnoses were performed: the first led to pregnancy interruption, the second to the birth of a healthy boy. CONCLUSIONS: This study of the first nonconsanguineous patient with LHX3 mutations demonstrates the pleiotropic roles of LHX3 during development and its full involvement in the complex disease phenotype. Isolated limitation of head rotation may exist in heterozygous carriers and would result from a dominant-negative effect. These data allowed the first prenatal diagnoses of this severe condition to be performed.


Asunto(s)
Hipopituitarismo/genética , Proteínas con Homeodominio LIM/genética , Hormonas Hipofisarias/deficiencia , Factores de Transcripción/genética , Niño , Preescolar , Heterocigoto , Humanos , Hipopituitarismo/diagnóstico , Lactante , Recién Nacido , Masculino , Mutación , Diagnóstico Prenatal , Síndrome
18.
Eur J Endocrinol ; 167(1): 85-91, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22535646

RESUMEN

BACKGROUND: In humans, pituitary hormone deficiency may be part of a syndrome including extra-pituitary defects like ocular abnormalities. Very few genes have been linked to this particular phenotype. In the mouse, Lhx2, which encodes a member of the LIM (Lin-11, Isl-1, and Mec-3) class of homeodomain proteins, was shown to be expressed during early development in the posterior pituitary, eye, and liver, and its expression persists in adulthood in the central nervous system Lhx2(-/-) mice display absence of posterior pituitary and intermediate lobes, malformation of the anterior lobe, anophthalmia, and they die from anemia. METHODS: We tested the implication of the LHX2 gene in patients presenting pituitary hormone deficiency associated with ectopic or nonvisible posterior pituitary and developmental ocular defects. A cohort of 59 patients, including two familial cases, was studied. Direct sequencing of the LHX2 coding sequence and intron/exon boundaries was performed. LHX2 transcriptional activity on several pituitary promoters (AGSU, PRL, POU1F1, and TSHB) was tested in vitro. RESULTS: Six heterozygous sequence variations were identified, among which two are novel missense changes (p.Ala203Thr and p.Val333Met). In vitro, LHX2 activates transcription of TSHB, PRL, and POU1F1 promoters in the HEK293 cell line. A synergistic action of POU1F1 and LHX2 was also shown on these promoters. The two missense variations were tested and no significant difference was observed, leading to the conclusion that they are not deleterious. CONCLUSIONS: These results suggest that if LHX2 is involved in pituitary hormone deficiency associated with posterior pituitary and ocular defects, it would be a rare cause of this disease condition.


Asunto(s)
Ceguera/genética , Trastornos del Crecimiento/genética , Proteínas con Homeodominio LIM/genética , Enfermedades del Nervio Óptico/genética , Neurohipófisis/anomalías , Displasia Septo-Óptica/genética , Factores de Transcripción/genética , Adulto , Niño , Preescolar , Femenino , Pruebas Genéticas , Humanos , Lactante , Masculino , Mutación Missense , Nervio Óptico/anomalías , Regiones Promotoras Genéticas
19.
Mol Med ; 14(5-6): 286-92, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18297129

RESUMEN

Isolated growth hormone deficiency (IGHD) may be of genetic origin. One of the few genes involved in that condition encodes the growth hormone releasing hormone receptor (GHRHR) that, through its ligand (GHRH), plays a pivotal role in the GH synthesis and secretion by the pituitary. Our objective is to describe the phenotype of two siblings born to a consanguineous union presenting with short stature (IGHD) and Magnetic Resonance Imaging (MRI) abnormalities, and to identify the molecular basis of this condition. Our main outcome measures were clinical and endocrinological investigations, MRI of the pituitary region, study of the GHRHR gene sequence and transcripts. In both patients, the severe growth retardation (-5SD) was combined with anterior pituitary hypoplasia. In addition to these classical phenotypic features for IGHD, one of the patients had a Chiari I malformation, an arachnoid cyst, and a dysmorphic anterior pituitary. A homozygous sequence variation in the consensus donor splice site of intron 1 (IVS1 + 2T > G) of the GHRHR gene was identified in both patients. Using in vitro transcription assay, we showed that this mutation results in abnormal splicing of GHRHR transcripts. In this report, which broadens the phenotype associated with GHRHR defects, we discuss the possible role of the GHRHR in the proper development of extrapituitary structures, through a mechanism that could be direct or secondary to severe GH deficiency.


Asunto(s)
Enanismo Hipofisario/genética , Mutación , Sitios de Empalme de ARN/genética , Receptores de Neuropéptido/genética , Receptores de Hormona Reguladora de Hormona Hipofisaria/genética , Empalme Alternativo , Niño , Análisis Mutacional de ADN , Enanismo Hipofisario/tratamiento farmacológico , Enanismo Hipofisario/patología , Femenino , Hormona del Crecimiento/uso terapéutico , Humanos , Imagen por Resonancia Magnética , Masculino , Linaje , Fenotipo , Receptores de Neuropéptido/fisiología , Receptores de Hormona Reguladora de Hormona Hipofisaria/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
20.
Blood ; 102(2): 561-3, 2003 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-12676783

RESUMEN

In a patient with lethal factor VII (FVII) deficiency, 2 homozygous nucleotide substitutions were identified in the F7 gene: a IVS7+2T>G transversion involving the IVS7 donor splice site, followed by a mutation at nucleotide 10588 that would result in a missense variation (Arg224Gln). The mutated splice site, located within the first repeat of a minisatellite, is followed by a variable number of pseudo-sites, normally silent. To investigate the consequences of this mutation on F7 splicing, we designed normal and mutant minigenes, spanning exons 5 to 8. In cells transfected with the mutant construct, no normal splicing occurred. Only spliced transcripts including the first minisatellite repeat were observed, resulting from the activation of the most proximal wild-type pseudo-site, which would generate a truncated protein (stop codon upstream of nucleotide 10588). These findings, which suggest the existence of a mechanism selecting one single splice site among multiple cryptic sites, explain the patient's phenotype.


Asunto(s)
Deficiencia del Factor VII/genética , Factor VII/genética , Intrones/genética , Sitios de Empalme de ARN/genética , Empalme del ARN/fisiología , Animales , Células CHO , Cricetinae , Cricetulus , Exones/genética , Genes Letales , Humanos , Modelos Genéticos , Fenotipo , Empalme del ARN/genética , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA