Network


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

Hotspot


Dive into the research topics where Louise A. Metherell is active.

Publication


Featured researches published by Louise A. Metherell.


Nature Genetics | 2005

Mutations in MRAP, encoding a new interacting partner of the ACTH receptor, cause familial glucocorticoid deficiency type 2

Louise A. Metherell; J. Paul Chapple; Sadani N. Cooray; Alessia David; Christian F. W. Becker; Franz Rüschendorf; Danielle Naville; Martine Begeot; Bernard Khoo; Peter Nürnberg; Angela Huebner; Michael E. Cheetham; Adrian J. L. Clark

Familial glucocorticoid deficiency (FGD), or hereditary unresponsiveness to adrenocorticotropin (ACTH; OMIM 202200), is an autosomal recessive disorder resulting from resistance to the action of ACTH on the adrenal cortex, which stimulates glucocorticoid production. Affected individuals are deficient in cortisol and, if untreated, are likely to succumb to hypoglycemia or overwhelming infection in infancy or childhood. Mutations of the ACTH receptor (melanocortin 2 receptor, MC2R) account for ∼25% of cases of FGD. FGD without mutations of MC2R is called FGD type 2. Using SNP array genotyping, we mapped a locus involved in FGD type 2 to chromosome 21q22.1. We identified mutations in a gene encoding a 19-kDa single–transmembrane domain protein, now known as melanocortin 2 receptor accessory protein (MRAP). We show that MRAP interacts with MC2R and may have a role in the trafficking of MC2R from the endoplasmic reticulum to the cell surface.


Journal of Clinical Investigation | 2012

MCM4 mutation causes adrenal failure, short stature, and natural killer cell deficiency in humans.

Claire Hughes; Leonardo Guasti; Eirini Meimaridou; Chen-Hua Chuang; John Schimenti; Peter King; Colm Costigan; Adrian J. L. Clark; Louise A. Metherell

An interesting variant of familial glucocorticoid deficiency (FGD), an autosomal recessive form of adrenal failure, exists in a genetically isolated Irish population. In addition to hypocortisolemia, affected children show signs of growth failure, increased chromosomal breakage, and NK cell deficiency. Targeted exome sequencing in 8 patients identified a variant (c.71-1insG) in minichromosome maintenance-deficient 4 (MCM4) that was predicted to result in a severely truncated protein (p.Pro24ArgfsX4). Western blotting of patient samples revealed that the major 96-kDa isoform present in unaffected human controls was absent, while the presence of the minor 85-kDa isoform was preserved. Interestingly, histological studies with Mcm4-depleted mice showed grossly abnormal adrenal morphology that was characterized by non-steroidogenic GATA4- and Gli1-positive cells within the steroidogenic cortex, which reduced the number of steroidogenic cells in the zona fasciculata of the adrenal cortex. Since MCM4 is one part of a MCM2-7 complex recently confirmed as the replicative helicase essential for normal DNA replication and genome stability in all eukaryotes, it is possible that our patients may have an increased risk of neoplastic change. In summary, we have identified what we believe to be the first human mutation in MCM4 and have shown that it is associated with adrenal insufficiency, short stature, and NK cell deficiency.


Proceedings of the National Academy of Sciences of the United States of America | 2009

MRAP and MRAP2 are bidirectional regulators of the melanocortin receptor family

Li F. Chan; Tom R. Webb; Teng-Teng Chung; Eirini Meimaridou; Sadani N. Cooray; Leonardo Guasti; Jp Chapple; Michaela Egertová; Maurice R. Elphick; Michael E. Cheetham; Louise A. Metherell; Adrian J. L. Clark

The melanocortin receptor (MCR) family consists of 5 G protein-coupled receptors (MC1R–MC5R) with diverse physiologic roles. MC2R is a critical component of the hypothalamic–pituitary–adrenal axis, whereas MC3R and MC4R have an essential role in energy homeostasis. Mutations in MC4R are the single most common cause of monogenic obesity. Investigating the way in which these receptors signal and traffic to the cell membrane is vital in understanding disease processes related to MCR dysfunction. MRAP is an MC2R accessory protein, responsible for adrenal MC2R trafficking and function. Here we identify MRAP2 as a unique homologue of MRAP, expressed in brain and the adrenal gland. We report that MRAP and MRAP2 can interact with all 5 MCRs. This interaction results in MC2R surface expression and signaling. In contrast, MRAP and MRAP2 can reduce MC1R, MC3R, MC4R, and MC5R responsiveness to [Nle4,D-Phe7]alpha-melanocyte-stimulating hormone (NDP-MSH). Collectively, our data identify MRAP and MRAP2 as unique bidirectional regulators of the MCR family.


Nature Genetics | 2012

Mutations in NNT encoding nicotinamide nucleotide transhydrogenase cause familial glucocorticoid deficiency

Eirini Meimaridou; Julia Kowalczyk; Leonardo Guasti; Claire Hughes; F Wagner; Peter Frommolt; Peter Nürnberg; Np Mann; R Banerjee; Hn Saka; Jp Chapple; Peter King; Adrian J. L. Clark; Louise A. Metherell

Using targeted exome sequencing, we identified mutations in NNT, an antioxidant defense gene, in individuals with familial glucocorticoid deficiency. In mice with Nnt loss, higher levels of adrenocortical cell apoptosis and impaired glucocorticoid production were observed. NNT knockdown in a human adrenocortical cell line resulted in impaired redox potential and increased reactive oxygen species (ROS) levels. Our results suggest that NNT may have a role in ROS detoxification in human adrenal glands.


Endocrine Reviews | 2011

Evidence for a Continuum of Genetic, Phenotypic, and Biochemical Abnormalities in Children with Growth Hormone Insensitivity

Alessia David; Vivian Hwa; Louise A. Metherell; Irene Netchine; Cecilia Camacho-Hübner; Adrian J. L. Clark; Ron G. Rosenfeld; Martin O. Savage

GH insensitivity (GHI) presents in childhood as growth failure and in its severe form is associated with dysmorphic and metabolic abnormalities. GHI may be caused by genetic defects in the GH-IGF-I axis or by acquired states such as chronic illness. This article discusses the former category. The field of GHI due to mutations affecting GH action has evolved considerably since the original description of the extreme phenotype related to homozygous GH receptor (GHR) mutations over 40 yr ago. A continuum of genetic, phenotypic, and biochemical abnormalities can be defined associated with clinically relevant defects in linear growth. The role and mechanisms of the GH-IGF-I axis in normal human growth is discussed, followed by descriptions of mutations in GHR, STAT5B, PTPN11, IGF1, IGFALS, IGF1R, and GH1 defects causing bioinactive GH or anti-GH antibodies. These defects are associated with a range of genetic, clinical, and hormonal characteristics. Genetic abnormalities causing growth failure that is less severe than the extreme phenotype are emphasized, together with an analysis of height and serum IGF-I across the spectrum of different types of GHR defects. An overall view of genotype and phenotype relationships is presented, together with an updated approach to the assessment of the patient with GHI, focusing on investigation of the GH-IGF-I axis and relevant molecular studies contributing to this diagnosis.


American Journal of Human Genetics | 2001

Pseudoexon Activation as a Novel Mechanism for Disease Resulting in Atypical Growth-Hormone Insensitivity

Louise A. Metherell; Patricia B. Munroe; Stephen Rose; Mark J. Caulfield; Martin O. Savage; Shern L Chew; Adrian Clark

Inherited growth-hormone insensitivity (GHI) is a heterogeneous disorder that is often caused by mutations in the coding exons or flanking intronic sequences of the growth-hormone receptor gene (GHR). Here we describe a novel point mutation, in four children with GHI, that leads to activation of an intronic pseudoexon resulting in inclusion of an additional 108 nt between exons 6 and 7 in the majority of GHR transcripts. This mutation lies within the pseudoexon (A(-1)-->G(-1) at the 5 pseudoexon splice site) and, under in vitro splicing conditions, results in inclusion of the mutant pseudoexon, whereas the wild-type pseudoexon is skipped. The presence of the pseudoexon results in inclusion of an additional 36-amino acid sequence in a region of the receptor known to be involved in homo-dimerization, which is essential for signal transduction.


The Journal of Clinical Endocrinology and Metabolism | 2009

Nonclassic lipoid congenital adrenal hyperplasia masquerading as familial glucocorticoid deficiency.

Louise A. Metherell; Danielle Naville; George Halaby; Martine Begeot; Angela Huebner; Gudrun Nürnberg; Peter Nürnberg; Jane Green; Jeremy W. Tomlinson; Nils Krone; Lin Lin; Michael S. Racine; Daniel M. Berney; John C. Achermann; Wiebke Arlt; Adrian J. L. Clark

CONTEXTnFamilial glucocorticoid deficiency (FGD) is an autosomal recessive disorder resulting from resistance to the action of ACTH on the adrenal cortex. Affected individuals are deficient in cortisol and, if untreated, are likely to succumb to hypoglycemia and/or overwhelming infection. Mutations of the ACTH receptor (MC2R) and the melanocortin 2 receptor accessory protein (MRAP), FGD types 1 and 2 respectively, account for approximately 45% of cases.nnnOBJECTIVEnA locus on chromosome 8 has previously been linked to the disease in three families, but no underlying gene defect has to date been identified.nnnDESIGNnThe study design comprised single-nucleotide polymorphism genotyping and mutation detection.nnnSETTINGnThe study was conducted at secondary and tertiary referral centers.nnnPATIENTSnEighty probands from families referred for investigation of the genetic cause of FGD participated in the study.nnnINTERVENTIONSnThere were no interventions.nnnRESULTSnAnalysis by single-nucleotide polymorphism array of the genotype of one individual with FGD previously linked to chromosome 8 revealed a large region of homozygosity encompassing the steroidogenic acute regulatory protein gene, STAR. We identified homozygous STAR mutations in this patient and his affected siblings. Screening of our total FGD patient cohort revealed homozygous STAR mutations in a further nine individuals from four other families.nnnCONCLUSIONSnMutations in STAR usually cause lipoid congenital adrenal hyperplasia, a disorder characterized by both gonadal and adrenal steroid deficiency. Our results demonstrate that certain mutations in STAR (R192C and the previously reported R188C) can present with a phenotype indistinguishable from that seen in FGD.


Nature Clinical Practice Endocrinology & Metabolism | 2006

Endocrine assessment, molecular characterization and treatment of growth hormone insensitivity disorders.

Martin O. Savage; Kenneth M. Attie; Alessia David; Louise A. Metherell; Adrian J. L. Clark; Cecilia Camacho-Hübner

Advances in the diagnosis and treatment of growth hormone insensitivity disorders have occurred in the past 15 years. We discuss the current status of endocrine and molecular evaluation, focusing on the pediatric age range. All the identified mutations of the growth hormone receptor are included. Treatment with recombinant human insulin-like growth factor (rhIGF) 1 in classical cases is summarized and new targets for treatment are discussed, together with therapy using the complex formed between rhIGF1 and rhIGF-binding protein 3.


Trends in Endocrinology and Metabolism | 2005

Inherited ACTH insensitivity illuminates the mechanisms of ACTH action

Adrian J. L. Clark; Louise A. Metherell; Michael E. Cheetham; Angela Huebner

Adrenocorticotrophin (ACTH) insensitivity is a potentially lethal inherited disorder of ACTH signalling in the adrenal. Inactivating mutations of the ACTH receptor account for approximately 25% of these cases. A second genetic cause for this syndrome has recently been identified in the MRAP gene. The MRAP protein appears to function in the trafficking and cell surface expression of the ACTH receptor, and might indicate the existence of more widespread G-protein-coupled receptor trafficking mechanisms. Molecular defects underlying other causes of ACTH insensitivity syndromes will probably contribute further to our understanding of these pathways.


Clinical Endocrinology | 2007

Severe loss-of-function mutations in the adrenocorticotropin receptor (ACTHR, MC2R) can be found in patients diagnosed with salt-losing adrenal hypoplasia

Lin Lin; Peter C. Hindmarsh; Louise A. Metherell; Mahmoud Alzyoud; Maryam Alali; Caroline Brain; Adrian J. L. Clark; Mehul T. Dattani; John C. Achermann

Objectiveu2002 Familial glucocorticoid deficiency type I (FGD1) is a rare form of primary adrenal insufficiency resulting from recessive mutations in the ACTH receptor (MC2R, MC2R). Individuals with this condition typically present in infancy or childhood with signs and symptoms of cortisol insufficiency, but disturbances in the renin‐angiotensin system, aldosterone synthesis or sodium homeostasis are not a well‐documented association of FGD1. As ACTH stimulation has been shown to stimulate aldosterone release in normal controls, and other causes of hyponatraemia can occur in children with cortisol deficiency, we investigated whether MC2R changes might be identified in children with primary adrenal failure who were being treated for mineralocorticoid insufficiency.

Collaboration


Dive into the Louise A. Metherell's collaboration.

Top Co-Authors

Avatar

Adrian J. L. Clark

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Li F. Chan

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Eirini Meimaridou

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar

Claire Hughes

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar

Helen L. Storr

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar

Leonardo Guasti

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar

Julia Kowalczyk

Queen Mary University of London

View shared research outputs
Researchain Logo
Decentralizing Knowledge