Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Sara Benito-Sanz is active.

Publication


Featured researches published by Sara Benito-Sanz.


American Journal of Medical Genetics Part A | 2010

CDKN1C (p57Kip2) analysis in Beckwith–Wiedemann syndrome (BWS) patients: Genotype–phenotype correlations, novel mutations, and polymorphisms

Valeria Romanelli; Alberta Belinchón; Sara Benito-Sanz; Víctor Martínez-Glez; Ricardo Gracia-Bouthelier; Karen E. Heath; Angel Campos-Barros; Sixto García-Miñaúr; Luis Venancio Oceja Fernández; Heloisa Meneses; Juan Pedro López-Siguero; Encarna Guillén-Navarro; Paulino Gómez-Puertas; Jan-Jaap Wesselink; Graciela Mercado; Rebeca Palomo; Rocío Mena; Aurora Sánchez; Miguel del Campo; Pablo Lapunzina

Beckwith–Wiedemann syndrome (BWS) is an overgrowth syndrome characterized by macroglossia, macrosomia, and abdominal wall defects. It is a multigenic disorder caused in most patients by alterations in growth regulatory genes. A small number of individuals with BWS (5–10%) have mutations in CDKN1C, a cyclin‐dependent kinase inhibitor of G1 cyclin complexes that functions as a negative regulator of cellular growth and proliferation. Here, we report on eight patients with BWS and CDKN1C mutations and review previous reported cases. We analyzed 72 patients (50 BWS, 17 with isolated hemihyperplasia (IH), three with omphalocele, and two with macroglossia) for CDKN1C defects with the aim to search for new mutations and to define genotype–phenotype correlations. Our findings suggest that BWS patients with CDKN1C mutations have a different pattern of clinical malformations than those with other molecular defects. Polydactyly, genital abnormalities, extra nipple, and cleft palate are more frequently observed in BWS with mutations in CDKN1C. The clinical observation of these malformations may help to decide which genetic characterization should be undertaken (i.e., CDKN1C screening), thus optimizing the laboratory evaluation for BWS.


Journal of Medical Genetics | 2012

Identification of the first recurrent PAR1 deletion in Léri-Weill dyschondrosteosis and idiopathic short stature reveals the presence of a novel SHOX enhancer

Sara Benito-Sanz; José Luis Royo; Eva Barroso; Beatriz Paumard-Hernández; Ana Coral Barreda-Bonis; Pengfei Liu; Ricardo Gracia; James R. Lupski; Angel Campos-Barros; José Luis Gómez-Skarmeta; Karen E. Heath

Background SHOX, located in the pseudoautosomal region 1 (PAR1) of the sexual chromosomes, encodes a transcription factor implicated in human growth. Defects in SHOX or its enhancers have been observed in ∼60% of Leri-Weill dyschondrosteosis (LWD) patients, a skeletal dysplasia characterised by short stature and/or the characteristic Madelung deformity, and in 2–5% of idiopathic short stature (ISS). To identify the molecular defect in the remaining genetically undiagnosed LWD and ISS patients, this study screened previously unanalysed PAR1 regions in 124 LWD and 576 ISS probands. Methods PAR1 screening was undertaken by multiplex ligation dependent probe amplification (MLPA). Copy number alterations were subsequently confirmed and delimited by locus-specific custom-designed MLPA, array comparative genomic hybridisation (CGH) and breakpoint junction PCR/sequencing. Results A recurrent PAR1 deletion downstream of SHOX spanning 47543 bp with identical breakpoints was identified in 19 LWD (15.3%) and 11 ISS (1.9%) probands, from 30 unrelated families. Eight evolutionarily conserved regions (ECRs 1–8) identified within the deleted sequence were evaluated for SHOX regulatory activity by means of chromosome conformation capture (3C) in chicken embryo limbs and luciferase reporter assays in human U2OS osteosarcoma cells. The 3C assay indicated potential SHOX regulatory activity by ECR1, which was subsequently confirmed to act as a SHOX enhancer, operating in an orientation and position independent manner, in human U2OS cells. Conclusions This study has identified the first recurrent PAR1 deletion in LWD and ISS, which results in the loss of a previously uncharacterised SHOX enhancer. The loss of this enhancer may decrease SHOX transcription, resulting in LWD or ISS due to SHOX haploinsufficiency.


European Journal of Human Genetics | 2012

Identification of the first PAR1 deletion encompassing upstream SHOX enhancers in a family with idiopathic short stature

Sara Benito-Sanz; Miriam Aza-Carmona; Amaya Rodríguez-Estevez; Ixaso Rica-Etxebarria; Ricardo Gracia; Angel Campos-Barros; Karen E. Heath

Short stature homeobox-containing gene, MIM 312865 (SHOX) is located within the pseudoautosomal region 1 (PAR1) of the sex chromosomes. Mutations in SHOX or its downstream transcriptional regulatory elements represent the underlying molecular defect in ∼60% of Léri-Weill dyschondrosteosis (LWD) and ∼5–15% of idiopathic short stature (ISS) patients. Recently, three novel enhancer elements have been identified upstream of SHOX but to date, no PAR1 deletions upstream of SHOX have been observed that only encompass these enhancers in LWD or ISS patients. We set out to search for genetic alterations of the upstream SHOX regulatory elements in 63 LWD and 100 ISS patients with no known alteration in SHOX or the downstream enhancer regions using a specifically designed MLPA assay, which covers the PAR1 upstream of SHOX. An upstream SHOX deletion was identified in an ISS proband and her affected father. The deletion was confirmed and delimited by array-CGH, to extend ∼286 kb. The deletion included two of the upstream SHOX enhancers without affecting SHOX. The 13.3-year-old proband had proportionate short stature with normal GH and IGF-I levels. In conclusion, we have identified the first PAR1 deletion encompassing only the upstream SHOX transcription regulatory elements in a family with ISS. The loss of these elements may result in SHOX haploinsufficiency because of decreased SHOX transcription. Therefore, this upstream region should be included in the routine analysis of PAR1 in patients with LWD, LMD and ISS.


Human Molecular Genetics | 2011

SHOX interacts with the chondrogenic transcription factors SOX5 and SOX6 to activate the aggrecan enhancer

Miriam Aza-Carmona; Debbie Shears; Patricia Yuste-Checa; Verónica Barca-Tierno; Alberta Belinchón; Sara Benito-Sanz; J. Ignacio Rodríguez; Jesús Argente; Angel Campos-Barros; Peter J. Scambler; Karen E. Heath

SHOX (short stature homeobox-containing gene) encodes a transcription factor implicated in skeletal development. SHOX haploinsufficiency has been demonstrated in Leri-Weill dyschondrosteosis (LWD), a skeletal dysplasia associated with disproportionate short stature, as well as in a variable proportion of cases with idiopathic short stature (ISS). In order to gain insight into the SHOX signalling pathways, we performed a yeast two-hybrid screen to identify SHOX-interacting proteins. Two transcription factors, SOX5 and SOX6, were identified. Co-immunoprecipitation assays confirmed the existence of the SHOX-SOX5 and SHOX-SOX6 interactions in human cells, whereas immunohistochemical studies demonstrated the coexpression of these proteins in 18- and 32-week human fetal growth plates. The SHOX homeodomain and the SOX6 HMG domain were shown to be implicated in the SHOX-SOX6 interaction. Moreover, different SHOX missense mutations, identified in LWD and ISS patients, disrupted this interaction. The physiological importance of these interactions was investigated by studying the effect of SHOX on a transcriptional target of the SOX trio, Agc1, which encodes one of the main components of cartilage, aggrecan. Our results show that SHOX cooperates with SOX5/SOX6 and SOX9 in the activation of the upstream Agc1 enhancer and that SHOX mutations affect this activation. In conclusion, we have identified SOX5 and SOX6 as the first two SHOX-interacting proteins and have shown that this interaction regulates aggrecan expression, an essential factor in chondrogenesis and skeletal development.


American Journal of Human Genetics | 2006

Characterization of SHOX Deletions in Léri-Weill Dyschondrosteosis (LWD) Reveals Genetic Heterogeneity and No Recombination Hotspots

Sara Benito-Sanz; Darya Gorbenko del Blanco; Céline Huber; N. Simon Thomas; Miriam Aza-Carmona; David J. Bunyan; Vivienne Maloney; Jesús Argente; Valérie Cormier-Daire; Angel Campos-Barros; Karen E. Heath

In the July 2005 and March 2006 issues of the Journal, Schneider et al.1 and Zinn et al.,2 respectively, reported mapping studies of SHOX (MIM 312865) deletions in patients with Leri-Weill dyschondrosteosis (LWD [MIM 127300]). In their study, Schneider et al.1 reported that the majority (73%) of patients with LWD who had SHOX deletions shared a 3? deletion breakpoint hotspot located downstream of SHOX. Zinn et al.2 identified a different 3? breakpoint hotspot located several hundred kilobases farther downstream in 86% of Hispanic patients, whereas the recombination hotspot described by Schneider et al.1 was not observed. We characterized the SHOX deletion limits in a cohort of 48 European patients with LWD (n=47) and Langer mesomelic dysplasia (LMD [MIM 249700]) (n=1). SHOX deletions were originally detected by multiplex ligation probe amplification (MLPA) (MRC Holland) or microsatellite analysis (DXYS10092, DXYS201, DYS290, DXYS10093, DXYS233, and DXYS234) and subsequently were finely mapped using a dense panel of microsatellites and SNPs.3 Four newly identified microsatellites (Tandem Repeat Finder), located 133, 54, 31, and 19 kb 5? of SHOX (table 1), and 59 SNPs, 12 of which were previously unreported (table 2), were analyzed.


The Journal of Clinical Endocrinology and Metabolism | 2015

Heterozygous NPR2 Mutations Cause Disproportionate Short Stature, Similar to Léri-Weill Dyschondrosteosis

Ana I. Garre-Vázquez; Fabiola Santaolalla-Caballero; Alberta Belinchón; Ana Coral Barreda-Bonis; Gabriela A. Vasques; Joaquin Ramirez; Cristina Luzuriaga; Gianni Carlone; Isabel González-Casado; Sara Benito-Sanz; Alexander A. L. Jorge; Angel Campos-Barros; Karen E. Heath

CONTEXT SHOX mutations have been detected in approximately 70% of Léri-Weill dyschondrosteosis (LWD) and approximately 2.5% of idiopathic short stature (ISS) cases, suggesting the implication of other genes or loci. The recent identification of NPR2 mutations in ISS suggested that NPR2 mutations may also be involved in disproportionate short stature. OBJECTIVE The objective of the study was to investigate whether NPR2 mutations can account for a proportion of the cases referred for LWD and ISS in whom no SHOX mutation was detected. PATIENTS AND METHODS We undertook NPR2 mutation screening in 173 individuals referred for suspected LWD and 95 for ISS, with no known defect in SHOX or its enhancers. Intracellular localization and natriuretic peptide precursor C-dependent guanylate cyclase activity were determined for the identified NPR2 variants. RESULTS Eight NPR2 variants were identified in nine individuals, seven referred for suspected LWD and two for ISS. Six were demonstrated to affect NPR-B cell trafficking and/or its ability to synthesize cyclic GMP (cGMP) under response to natriuretic peptide precursor C/brain natriuretic peptide stimulation. All pathogenic mutations were detected in the suspected LWD referral group (∼3%). Interestingly, one of these patients is currently being treated with recombinant human GH and in contrast to previous reports is showing a positive response to the treatment. CONCLUSIONS NPR2 mutations account for approximately 3% of patients with disproportionate short stature and/or clinical or radiographic indicators of SHOX deficiency and in whom no SHOX defect has been identified. However, no patient has yet presented with Madelung deformity. Thus, NPR2 should be screened in the SHOX-negative LWD referrals.


European Journal of Human Genetics | 2015

Expanding the mutation spectrum in 182 Spanish probands with craniosynostosis: identification and characterization of novel TCF12 variants

Beatriz Paumard-Hernández; Julia Berges-Soria; Eva Barroso; Carlos I. Rivera-Pedroza; Virginia Pérez-Carrizosa; Sara Benito-Sanz; Eva López-Messa; Fernando Santos; Ignacio I García-Recuero; Ana Romance; Juliana María Ballesta-Martínez; Vanesa López-González; Angel Campos-Barros; Jaime Cruz; Encarna Guillén-Navarro; Jaime Sánchez del Pozo; Pablo Lapunzina; Sixto García-Miñaúr; Karen E. Heath

Craniosynostosis, caused by the premature fusion of one or more of the cranial sutures, can be classified into non-syndromic or syndromic and by which sutures are affected. Clinical assignment is a difficult challenge due to the high phenotypic variability observed between syndromes. During routine diagnostics, we screened 182 Spanish craniosynostosis probands, implementing a four-tiered cascade screening of FGFR2, FGFR3, FGFR1, TWIST1 and EFNB1. A total of 43 variants, eight novel, were identified in 113 (62%) patients: 104 (92%) detected in level 1; eight (7%) in level 2 and one (1%) in level 3. We subsequently screened additional genes in the probands with no detected mutation: one duplication of the IHH regulatory region was identified in a patient with craniosynostosis Philadelphia type and five variants, four novel, were identified in the recently described TCF12, in probands with coronal or multisuture affectation. In the 19 Saethre–Chotzen syndrome (SCS) individuals in whom a variant was detected, 15 (79%) carried a TWIST1 variant, whereas four (21%) had a TCF12 variant. Thus, we propose that TCF12 screening should be included for TWIST1 negative SCS patients and in patients where the coronal suture is affected. In summary, a molecular diagnosis was obtained in a total of 119/182 patients (65%), allowing the correct craniosynostosis syndrome classification, aiding genetic counselling and in some cases provided a better planning on how and when surgical intervention should take place and, subsequently the appropriate clinical follow up.


American Journal of Medical Genetics Part A | 2007

Compound heterozygosity of SHOX-encompassing and downstream PAR1 deletions results in Langer mesomelic dysplasia (LMD).

Angel Campos-Barros; Sara Benito-Sanz; Judith L. Ross; Andrew R. Zinn; Karen E. Heath

We present the clinical and molecular characteristics of a multi‐generation family in which the proband presented with clinical features of Langer mesomelic dysplasia (LMD) whilst different family members had a diagnosis of Léri–Weill dyschondrosteosis (LWD) and/or pseudoachondroplasia (PSACH). In the LMD proband two different deletions were identified in the pseudoautosomal 1 region (PAR1) of the X and Y chromosomes: a SHOX‐encompassing deletion inherited from his father and a downstream PAR1 deletion, which did not include SHOX, inherited from his mother. The individuals with PSACH features presented the previously described G719D mutation in the C‐terminal globular domain of the cartilage oligomeric matrix protein gene (COMP). The LMD proband described here represents the first LMD case due to compound heterozygosity for deletions of the two different PAR1 regions, SHOX‐encompassing and downstream from SHOX, that have been shown to be implicated in the pathogenesis of LWD and LMD.


Human Mutation | 2014

A New Overgrowth Syndrome is Due to Mutations in RNF125.

Jair Tenorio; Alicia Mansilla; María Valencia; Victor Martinez-Glez; Valeria Romanelli; Pedro Arias; Nerea Castrejón; Fernando A. Poletta; Encarna Guillén-Navarro; Gema Gordo; Elena Mansilla; Fe Amalia García-Santiago; Isabel González-Casado; Elena Vallespín; María Palomares; María Ángeles Mori; Fernando Santos-Simarro; Sixto García-Miñaúr; Luis Fernández; Rocío Mena; Sara Benito-Sanz; Angela del Pozo; J.C. Silla; Kristina Ibanez; Eduardo López-Granados; Alex Martin-Trujillo; David Montaner; Karen E. Heath; Angel Campos-Barros; Joaquín Dopazo

Overgrowth syndromes (OGS) are a group of disorders in which all parameters of growth and physical development are above the mean for age and sex. We evaluated a series of 270 families from the Spanish Overgrowth Syndrome Registry with no known OGS. We identified one de novo deletion and three missense mutations in RNF125 in six patients from four families with overgrowth, macrocephaly, intellectual disability, mild hydrocephaly, hypoglycemia, and inflammatory diseases resembling Sjögren syndrome. RNF125 encodes an E3 ubiquitin ligase and is a novel gene of OGS. Our studies of the RNF125 pathway point to upregulation of RIG‐I‐IPS1‐MDA5 and/or disruption of the PI3K‐AKT and interferon signaling pathways as the putative final effectors.


Scientific Reports | 2016

Profiling of conserved non-coding elements upstream of SHOX and functional characterisation of the SHOX cis -regulatory landscape

Hannah Verdin; Ana Fernández-Miñán; Sara Benito-Sanz; Sandra Janssens; Bert Callewaert; Kathleen De Waele; Jean De Schepper; Inge François; Björn Menten; Karen E. Heath; José Luis Gómez-Skarmeta; Elfride De Baere

Genetic defects such as copy number variations (CNVs) in non-coding regions containing conserved non-coding elements (CNEs) outside the transcription unit of their target gene, can underlie genetic disease. An example of this is the short stature homeobox (SHOX) gene, regulated by seven CNEs located downstream and upstream of SHOX, with proven enhancer capacity in chicken limbs. CNVs of the downstream CNEs have been reported in many idiopathic short stature (ISS) cases, however, only recently have a few CNVs of the upstream enhancers been identified. Here, we set out to provide insight into: (i) the cis-regulatory role of these upstream CNEs in human cells, (ii) the prevalence of upstream CNVs in ISS, and (iii) the chromatin architecture of the SHOX cis-regulatory landscape in chicken and human cells. Firstly, luciferase assays in human U2OS cells, and 4C-seq both in chicken limb buds and human U2OS cells, demonstrated cis-regulatory enhancer capacities of the upstream CNEs. Secondly, CNVs of these upstream CNEs were found in three of 501 ISS patients. Finally, our 4C-seq interaction map of the SHOX region reveals a cis-regulatory domain spanning more than 1 Mb and harbouring putative new cis-regulatory elements.

Collaboration


Dive into the Sara Benito-Sanz's collaboration.

Top Co-Authors

Avatar

Karen E. Heath

Hospital Universitario La Paz

View shared research outputs
Top Co-Authors

Avatar

Angel Campos-Barros

Instituto de Salud Carlos III

View shared research outputs
Top Co-Authors

Avatar

Alberta Belinchón

Autonomous University of Madrid

View shared research outputs
Top Co-Authors

Avatar

Miriam Aza-Carmona

Autonomous University of Madrid

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jesús Argente

Instituto de Salud Carlos III

View shared research outputs
Top Co-Authors

Avatar

Encarna Guillén-Navarro

Universidad Católica San Antonio de Murcia

View shared research outputs
Top Co-Authors

Avatar

Eva Barroso

Autonomous University of Madrid

View shared research outputs
Top Co-Authors

Avatar

Pablo Lapunzina

Autonomous University of Madrid

View shared research outputs
Top Co-Authors

Avatar

Ricardo Gracia

Hospital Universitario La Paz

View shared research outputs
Researchain Logo
Decentralizing Knowledge