Network


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

Hotspot


Dive into the research topics where Laura C. Hernández-Ramírez is active.

Publication


Featured researches published by Laura C. Hernández-Ramírez.


PLOS ONE | 2012

Structure of the TPR Domain of AIP: Lack of Client Protein Interaction with the C-Terminal α-7 Helix of the TPR Domain of AIP Is Sufficient for Pituitary Adenoma Predisposition

Rhodri M. L. Morgan; Laura C. Hernández-Ramírez; Giampaolo Trivellin; Lihong Zhou; S. Mark Roe; Márta Korbonits; Chrisostomos Prodromou

Mutations of the aryl hydrocarbon receptor interacting protein (AIP) have been associated with familial isolated pituitary adenomas predisposing to young-onset acromegaly and gigantism. The precise tumorigenic mechanism is not well understood as AIP interacts with a large number of independent proteins as well as three chaperone systems, HSP90, HSP70 and TOMM20. We have determined the structure of the TPR domain of AIP at high resolution, which has allowed a detailed analysis of how disease-associated mutations impact on the structural integrity of the TPR domain. A subset of C-terminal α-7 helix (Cα-7h) mutations, R304* (nonsense mutation), R304Q, Q307* and R325Q, a known site for AhR and PDE4A5 client-protein interaction, occur beyond those that interact with the conserved MEEVD and EDDVE sequences of HSP90 and TOMM20. These C-terminal AIP mutations appear to only disrupt client-protein binding to the Cα-7h, while chaperone binding remains unaffected, suggesting that failure of client-protein interaction with the Cα-7h is sufficient to predispose to pituitary adenoma. We have also identified a molecular switch in the AIP TPR-domain that allows recognition of both the conserved HSP90 motif, MEEVD, and the equivalent sequence (EDDVE) of TOMM20.


The Journal of Clinical Endocrinology and Metabolism | 2017

Somatic USP8 Gene Mutations Are a Common Cause of Pediatric Cushing Disease

Fabio R. Faucz; Amit Tirosh; Christina Tatsi; Annabel Berthon; Laura C. Hernández-Ramírez; Nikolaos Settas; Anna Angelousi; Ricardo Correa; Georgios Z. Papadakis; Prashant Chittiboina; Martha Quezado; Nathan Pankratz; John Lane; Aggeliki Dimopoulos; James L. Mills; Maya Lodish; Constantine A. Stratakis

Context: Somatic mutations in the ubiquitin-specific protease 8 (USP8) gene have been recently identified as the most common genetic alteration in patients with Cushing disease (CD). However, the frequency of these mutations in the pediatric population has not been extensively assessed. Objective: We investigated the status of the USP8 gene at the somatic level in a cohort of pediatric patients with corticotroph adenomas. Design and Methods: The USP8 gene was fully sequenced in both germline and tumor DNA samples from 42 pediatric patients with CD. Clinical, biochemical, and imaging data were compared between patients with and without somatic USP8 mutations. Results: Five different USP8 mutations (three missense, one frameshift, and one in-frame deletion) were identified in 13 patients (31%), all of them located in exon 14 at the previously described mutational hotspot, affecting the 14-3-3 binding motif of the protein. Patients with somatic mutations were older at disease presentation [mean 5.1 ± 2.1 standard deviation (SD) vs 13.1 ± 3.6 years, P = 0.03]. Levels of urinary free cortisol, midnight serum cortisol, and adrenocorticotropic hormone, as well as tumor size and frequency of invasion of the cavernous sinus, were not significantly different between the two groups. However, patients harboring somatic USP8 mutations had a higher likelihood of recurrence compared with patients without mutations (46.2% vs 10.3%, P = 0.009). Conclusion: Somatic USP8 gene mutations are a common cause of pediatric CD. Patients harboring a somatic mutation had a higher likelihood of tumor recurrence, highlighting the potential importance of this molecular defect for the disease prognosis and the development of targeted therapeutic options.


Human Mutation | 2017

Increased Population Risk of AIP-related Acromegaly and Gigantism in Ireland.

Serban Radian; Yoan Diekmann; Plamena Gabrovska; Brendan Holland; Lisa Bradley; Helen Wallace; Karen Stals; Anna Marie Bussell; Karen McGurren; Martin Cuesta; Anthony W. Ryan; Maria Herincs; Laura C. Hernández-Ramírez; Aidan Holland; Jade Samuels; Elena Daniela Aflorei; Sayka Barry; Judit Dénes; Ida Pernicova; Craig E Stiles; Giampaolo Trivellin; Ronan McCloskey; Michal Ajzensztejn; Noina Abid; Moisés Mercado; Mark Cohen; Rajesh V. Thakker; Stephanie Baldeweg; Ariel L. Barkan; Madalina Musat

The aryl hydrocarbon receptor interacting protein (AIP) founder mutation R304* (or p.R304*; NM_003977.3:c.910C>T, p.Arg304Ter) identified in Northern Ireland (NI) predisposes to acromegaly/gigantism; its population health impact remains unexplored. We measured R304* carrier frequency in 936 Mid Ulster, 1,000 Greater Belfast (both in NI) and 2,094 Republic of Ireland (ROI) volunteers and in 116 NI or ROI acromegaly/gigantism patients. Carrier frequencies were 0.0064 in Mid Ulster (95%CI = 0.0027–0.013; P = 0.0005 vs. ROI), 0.001 in Greater Belfast (0.00011–0.0047) and zero in ROI (0–0.0014). R304* prevalence was elevated in acromegaly/gigantism patients in NI (11/87, 12.6%, P < 0.05), but not in ROI (2/29, 6.8%) versus non‐Irish patients (0–2.41%). Haploblock conservation supported a common ancestor for all the 18 identified Irish pedigrees (81 carriers, 30 affected). Time to most recent common ancestor (tMRCA) was 2550 (1,275–5,000) years. tMRCA‐based simulations predicted 432 (90–5,175) current carriers, including 86 affected (18–1,035) for 20% penetrance. In conclusion, R304* is frequent in Mid Ulster, resulting in numerous acromegaly/gigantism cases. tMRCA is consistent with historical/folklore accounts of Irish giants. Forward simulations predict many undetected carriers; geographically targeted population screening improves asymptomatic carrier identification, complementing clinical testing of patients/relatives. We generated disease awareness locally, necessary for early diagnosis and improved outcomes of AIP‐related disease.


Journal of the Endocrine Society | 2017

Corticotropinoma as a Component of Carney Complex

Laura C. Hernández-Ramírez; Christina Tatsi; Maya Lodish; Fabio R. Faucz; Nathan Pankratz; Prashant Chittiboina; John Lane; Denise M. Kay; Nuria Valdés; Aggeliki Dimopoulos; James L. Mills; Constantine A. Stratakis

Known germline gene abnormalities cause one-fifth of the pituitary adenomas in children and adolescents, but, in contrast with other pituitary tumor types, the genetic causes of corticotropinomas are largely unknown. In this study, we report a case of Cushing disease (CD) due to a loss-of-function mutation in PRKAR1A, providing evidence for association of this gene with a corticotropinoma. A 15-year-old male presenting with hypercortisolemia was diagnosed with CD. Remission was achieved after surgical resection of a corticotropin (ACTH)-producing pituitary microadenoma, but recurrence 3 years later prompted reoperation and radiotherapy. Five years after the original diagnosis, the patient developed ACTH-independent Cushing syndrome, and a diagnosis of primary pigmented nodular adrenocortical disease was confirmed. A PRKAR1A mutation (c.671delG, p.G225Afs*16) was detected in a germline DNA sample from the patient, which displayed loss of heterozygosity in the corticotropinoma. No other germline or somatic mutations of interest were found. As corticotropinomas are not a known component of Carney complex (CNC), we performed loss of heterozygosity and messenger RNA stability studies in the patient’s tissues, and analyzed the effect of Prkar1a silencing on AtT-20/D16v-F2 mouse corticotropinoma cells. No PRKAR1A defects were found among 97 other pediatric CD patients studied. Our clinical case and experimental data support a role for PRKAR1A in the pathogenesis of a corticotroph cell tumor. This is a molecularly confirmed report of a corticotropinoma presenting in association with CNC. We conclude that germline PRKAR1A mutations are a novel, albeit apparently infrequent, cause of CD.


Endocrine-related Cancer | 2017

Loss-of-function mutations in the CABLES1 gene are a novel cause of Cushing’s disease

Laura C. Hernández-Ramírez; Nuria Valdés; Maya Lodish; Nathan Pankratz; Aurelio Balsalobre; Yves Gauthier; Fabio R. Faucz; Giampaolo Trivellin; Prashant Chittiboina; John Lane; Denise M. Kay; Aggeliki Dimopoulos; Stephan Gaillard; M. Neou; Jérôme Bertherat; Guillaume Assié; Chiara Villa; James L. Mills; Jacques Drouin; Constantine A. Stratakis

The CABLES1 cell cycle regulator participates in the adrenal–pituitary negative feedback, and its expression is reduced in corticotropinomas, pituitary tumors with a largely unexplained genetic basis. We investigated the presence of CABLES1 mutations/copy number variations (CNVs) and their associated clinical, histopathological and molecular features in patients with Cushing’s disease (CD). Samples from 146 pediatric (118 germline DNA only/28 germline and tumor DNA) and 35 adult (tumor DNA) CD patients were screened for CABLES1 mutations. CNVs were assessed in 116 pediatric CD patients (87 germline DNA only/29 germline and tumor DNA). Four potentially pathogenic missense variants in CABLES1 were identified, two in young adults (c.532G > A, p.E178K and c.718C > T, p.L240F) and two in children (c.935G > A, p.G312D and c.1388A > G, and p.D463G) with CD; no CNVs were found. The four variants affected residues within or close to the predicted cyclin-dependent kinase-3 (CDK3)-binding region of the CABLES1 protein and impaired its ability to block cell growth in a mouse corticotropinoma cell line (AtT20/D16v-F2). The four patients had macroadenomas. We provide evidence for a role of CABLES1 as a novel pituitary tumor-predisposing gene. Its function might link two of the main molecular mechanisms altered in corticotropinomas: the cyclin-dependent kinase/cyclin group of cell cycle regulators and the epidermal growth factor receptor signaling pathway. Further studies are needed to assess the prevalence of CABLES1 mutations among patients with other types of pituitary adenomas and to elucidate the pituitary-specific functions of this gene.


European Journal of Endocrinology | 2014

Sequence analysis of the catalytic subunit of PKA in somatotroph adenomas.

Sarah Larkin; Francesco Ferraù; Niki Karavitaki; Laura C. Hernández-Ramírez; Olaf Ansorge; Ashley B. Grossman; Márta Korbonits

OBJECTIVE The pathogenetic mechanisms of sporadic somatotroph adenomas are not well understood, but derangements of the cAMP pathway have been implicated. Recent studies have identified L206R mutations in the alpha catalytic subunit of protein kinase A (PRKACA) in cortisol-producing adrenocortical adenomas and amplification of the beta catalytic subunit of protein kinase A PRKACB in acromegaly associated with Carney complex. Given that both adrenocortical adenomas and somatotroph adenomas are known to be reliant on the cAMP signalling pathway, we sought to determine the relevance of the L206R mutation in both PRKACA and PRKACB for the pathogenesis of sporadic somatotroph adenomas. DESIGN Somatotroph adenoma specimens, both frozen and formalin-fixed, from patients who underwent surgery for their acromegaly between 1995 and 2012, were used in the study. METHODS The DNA sequence at codon 206 of PRKACA and PRKACB was determined by PCR amplification and sequencing. The results were compared with patient characteristics, the mutational status of the GNAS complex locus and the tumour granulation pattern. RESULTS No mutations at codon 206 of PRKACA or PRKACB were found in a total of 92 specimens, comprising both WT and mutant GNAS cases, and densely, sparsely and mixed granulation patterns. CONCLUSIONS It is unlikely that mutation at this locus is involved in the pathogenesis of sporadic somatotroph adenoma; however, gene amplification or mutations at other loci or in other components of the cAMP signalling pathway, while unlikely, cannot be ruled out.


Oncotarget | 2018

Multi-chaperone function modulation and association with cytoskeletal proteins are key features of the function of AIP in the pituitary gland

Laura C. Hernández-Ramírez; Rhodri M. L. Morgan; Sayka Barry; Fulvio D’Acquisto; Chrisostomos Prodromou; Márta Korbonits

Despite the well-recognized role of loss-of-function mutations of the aryl hydrocarbon receptor interacting protein gene (AIP) predisposing to pituitary adenomas, the pituitary-specific function of this tumor suppressor remains an enigma. To determine the repertoire of interacting partners for the AIP protein in somatotroph cells, wild-type and variant AIP proteins were used for pull-down/quantitative mass spectrometry experiments against lysates of rat somatotropinoma-derived cells; relevant findings were validated by co-immunoprecipitation and co-localization. Global gene expression was studied in AIP mutation positive and negative pituitary adenomas via RNA microarrays. Direct interaction with AIP was confirmed for three known and six novel partner proteins. Novel interactions with HSPA5 and HSPA9, together with known interactions with HSP90AA1, HSP90AB1 and HSPA8, indicate that the function/stability of multiple chaperone client proteins could be perturbed by a deficient AIP co-chaperone function. Interactions with TUBB, TUBB2A, NME1 and SOD1 were also identified. The AIP variants p.R304* and p.R304Q showed impaired interactions with HSPA8, HSP90AB1, NME1 and SOD1; p.R304* also displayed reduced binding to TUBB and TUBB2A, and AIP-mutated tumors showed reduced TUBB2A expression. Our findings suggest that cytoskeletal organization, cell motility/adhesion, as well as oxidative stress responses, are functions that are likely to be involved in the tumor suppressor activity of AIP.


Journal of Medical Genetics | 2018

Risk category system to identify pituitary adenoma patients with AIP mutations

Francisca Caimari; Laura C. Hernández-Ramírez; Mary N. Dang; Plamena Gabrovska; Donato Iacovazzo; Karen Stals; Sian Ellard; Márta Korbonits

Background Predictive tools to identify patients at risk for gene mutations related to pituitary adenomas are very helpful in clinical practice. We therefore aimed to develop and validate a reliable risk category system for aryl hydrocarbon receptor-interacting protein (AIP) mutations in patients with pituitary adenomas. Methods An international cohort of 2227 subjects were consecutively recruited between 2007 and 2016, including patients with pituitary adenomas (familial and sporadic) and their relatives. All probands (n=1429) were screened for AIP mutations, and those diagnosed with a pituitary adenoma prospectively, as part of their clinical screening (n=24), were excluded from the analysis. Univariate analysis was performed comparing patients with and without AIP mutations. Based on a multivariate logistic regression model, six potential factors were identified for the development of a risk category system, classifying the individual risk into low-risk, moderate-risk and high-risk categories. An internal cross-validation test was used to validate the system. Results 1405 patients had a pituitary tumour, of which 43% had a positive family history, 55.5% had somatotrophinomas and 81.5% presented with macroadenoma. Overall, 134 patients had an AIP mutation (9.5%). We identified four independent predictors for the presence of an AIP mutation: age of onset providing an odds ratio (OR) of 14.34 for age 0-18 years, family history (OR 10.85), growth hormone excess (OR 9.74) and large tumour size (OR 4.49). In our cohort, 71% of patients were identified as low risk (<5% risk of AIP mutation), 9.2% as moderate risk and 20% as high risk (≥20% risk). Excellent discrimination (c-statistic=0.87) and internal validation were achieved. Conclusion We propose a user-friendly risk categorisation system that can reliably group patients into high-risk, moderate-risk and low-risk groups for the presence of AIP mutations, thus providing guidance in identifying patients at high risk of carrying an AIP mutation. This risk score is based on a cohort with high prevalence of AIP mutations and should be applied cautiously in other populations.


Expert Review of Endocrinology & Metabolism | 2017

Sporadic pituitary adenomas: the role of germline mutations and recommendations for genetic screening

Donato Iacovazzo; Laura C. Hernández-Ramírez; Márta Korbonits

ABSTRACT Introduction: Although most pituitary adenomas occur sporadically, these common tumors can present in a familial setting in approximately 5% of cases. Germline mutations in several genes with autosomal dominant (AIP, MEN1, CDKN1B, PRKAR1A, SDHx) or X-linked dominant (GPR101) inheritance are causative of familial pituitary adenomas. Due to variable disease penetrance and occurrence of de novo mutations, some patients harboring germline mutations have no family history of pituitary adenomas (simplex cases). Areas covered: We summarize the recent findings on the role of germline mutations associated with familial pituitary adenomas in patients with sporadic clinical presentation. Expert commentary: Up to 12% of patients with young onset pituitary adenomas (age at diagnosis/onset ≤30 years) and up to 25% of simplex patients with gigantism carry mutations in the AIP gene, while most cases of X-linked acrogigantism (XLAG) due to GPR101 duplication are simplex female patients with very early disease onset (<5 years). With regard to the syndromes of multiple endocrine neoplasia (MEN), MEN1 mutations can be identified in a significant proportion of patients with childhood onset prolactinomas. Somatotroph and lactotroph adenomas are the most common pituitary adenomas associated with germline predisposing mutations. Genetic screening should be considered in patients with young onset pituitary adenomas.


Clinical Endocrinology | 2018

Children with MEN1 gene mutations may present first (and at a young age) with Cushing disease

Angeliki Makri; Maria Belen Bonella; Margaret F. Keil; Laura C. Hernández-Ramírez; Gabriella Paluch; Amit Tirosh; Carolina Saldarriaga; Prashant Chittiboina; Stephen J. Marx; Constantine A. Stratakis; Maya Lodish

Cushing disease (CD) is a rare entity caused by ACTH‐secreting pituitary tumours, leading to prolonged hypercortisolism. Most cases are sporadic but can rarely occur in the context of familial predisposition, due to germline mutations in genes such as MEN1, leading to multiple endocrine neoplasia type 1, MEN1. We have reported previously that CD can be the first and only presenting manifestation of MEN1. In this report, we describe a cohort of paediatric patients who presented with CD as the first manifestation of MEN1.

Collaboration


Dive into the Laura C. Hernández-Ramírez's collaboration.

Top Co-Authors

Avatar

Giampaolo Trivellin

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Márta Korbonits

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Plamena Gabrovska

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar

Donato Iacovazzo

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar

Karen Stals

National Health Service

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Francesco Ferraù

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar

Judit Dénes

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar

Sayka Barry

Queen Mary University of London

View shared research outputs
Researchain Logo
Decentralizing Knowledge