Network


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

Hotspot


Dive into the research topics where Adrian J. L. Clark is active.

Publication


Featured researches published by Adrian J. L. Clark.


Circulation Research | 2007

Epigenetic Modification of the Renin-Angiotensin System in the Fetal Programming of Hypertension

Irina Bogdarina; Simon J. M. Welham; Peter King; Shamus P. Burns; Adrian J. L. Clark

Hypertension is a major risk factor for cardiovascular and cerebrovascular disease. Lifelong environmental factors (eg, salt intake, obesity, alcohol) and genetic factors clearly contribute to the development of hypertension, but it has also been established that stress in utero may program the later development of the disease. This phenomenon, known as fetal programming can be modeled in a range of experimental animal models. In maternal low protein diet rat models of programming, administration of angiotensin converting enzyme inhibitors or angiotensin receptor antagonists in early life can prevent development of hypertension, thus implicating the renin-angiotensin system in this process. Here we show that in this model, expression of the AT1b angiotensin receptor gene in the adrenal gland is upregulated by the first week of life resulting in increased receptor protein expression consistent with the increased adrenal angiotensin responsiveness observed by others. Furthermore, we show that the proximal promoter of the AT1b gene in the adrenal is significantly undermethylated, and that in vitro, AT1b gene expression is highly dependent on promoter methylation. These data suggest a link between fetal insults to epigenetic modification of genes and the resultant alteration of gene expression in adult life leading ultimately to the development of hypertension. It seems highly probable that similar influences may be involved in the development of human hypertension.


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.


Trends in Endocrinology and Metabolism | 2000

DNA Methylation and Silencing of Gene Expression

John Newell-Price; Adrian J. L. Clark; Peter King

DNA methylation is associated with the silencing of gene expression. The predominant mechanism involves the methylation of DNA and the subsequent recruitment of binding proteins that preferentially recognize methylated DNA. In turn, these proteins associate with histone deacetylase and chromatin remodelling complexes to cause the stabilization of condensed chromatin. Recent studies have indicated that the opposite might also hold; namely, that targeting of methylation might depend on altered chromatin structure. The family of methyltransferases and methyl-binding proteins is expanding and becoming better characterized. This review will focus on the mechanisms of methylation-associated silencing of gene expression.


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.


Science | 2013

Loss of function of the melanocortin 2 receptor accessory protein 2 is associated with mammalian obesity.

Masato Asai; Shwetha Ramachandrappa; Maria Joachim; Yuan Shen; Rong Zhang; Nikhil Nuthalapati; Visali Ramanathan; David E. Strochlic; P. R. Ferket; Kirsten Linhart; Caroline Ho; Tatiana V. Novoselova; Sumedha Garg; Martin Ridderstråle; Claude Marcus; Joel N. Hirschhorn; Julia M. Keogh; Stephen O’Rahilly; Li F. Chan; Adrian J. L. Clark; I. Sadaf Farooqi; Joseph A. Majzoub

Accessory to Obesity? Melanocortin receptors are a family of cell membrane receptors that control diverse physiological functions. Mutations in the gene encoding melanocortin 4 receptor (MC4R) are a cause of familial early-onset obesity. Asai et al. (p. 275) studied the function of an accessory protein for MC4R signaling, MRAP2, and found that mice genetically deficient in MRAP2 develop severe obesity. Sequencing of MRAP2 in unrelated, severely obese humans revealed one individual with a clearly disruptive genetic variant, suggesting that MRAP2 mutations might also be a rare cause of human obesity. In a zebrafish model, Sebag et al. (p. 278) studied two paralogs of the MRAP2 accessory protein, one of which enhanced MC4R responsiveness to α–melanocyte-stimulating hormone, which regulates feeding and growth. Disruption of a protein required for effective signaling by a melanocortin receptor causes severe obesity in mice. Melanocortin receptor accessory proteins (MRAPs) modulate signaling of melanocortin receptors in vitro. To investigate the physiological role of brain-expressed melanocortin 2 receptor accessory protein 2 (MRAP2), we characterized mice with whole-body and brain-specific targeted deletion of Mrap2, both of which develop severe obesity at a young age. Mrap2 interacts directly with melanocortin 4 receptor (Mc4r), a protein previously implicated in mammalian obesity, and it enhances Mc4r-mediated generation of the second messenger cyclic adenosine monophosphate, suggesting that alterations in Mc4r signaling may be one mechanism underlying the association between Mrap2 disruption and obesity. In a study of humans with severe, early-onset obesity, we found four rare, potentially pathogenic genetic variants in MRAP2, suggesting that the gene may also contribute to body weight regulation in humans.


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

CONTEXT Familial 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. OBJECTIVE A locus on chromosome 8 has previously been linked to the disease in three families, but no underlying gene defect has to date been identified. DESIGN The study design comprised single-nucleotide polymorphism genotyping and mutation detection. SETTING The study was conducted at secondary and tertiary referral centers. PATIENTS Eighty probands from families referred for investigation of the genetic cause of FGD participated in the study. INTERVENTIONS There were no interventions. RESULTS Analysis 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. CONCLUSIONS Mutations 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.


Molecular Endocrinology | 2010

Minireview: The melanocortin 2 receptor accessory proteins

Tom R. Webb; Adrian J. L. Clark

The melanocortin 2 receptor (MC2R) accessory protein, MRAP, is one of a growing number of G protein-coupled receptor accessory proteins that have been identified in recent years that add control and complexity to G protein-coupled receptor functional expression and signal transduction. MRAP interacts directly with MC2R and is essential for its trafficking from the endoplasmic reticulum to the cell surface, where it acts as the receptor for the pituitary hormone ACTH. In addition, MRAP2, a newly described homolog of MRAP, is also able to support the cell surface expression of MC2R. Although it is clear that MRAP is required for MC2R function, the mechanism of MRAP action is only beginning to be understood. Recent work has started to reveal some of these mechanisms and the MRAP domains involved in MC2R functional expression, and new data have shown a potential role for both MRAP and MRAP2 in the regulation of the other melanocortin receptors.

Collaboration


Dive into the Adrian J. L. Clark's collaboration.

Top Co-Authors

Avatar

Louise A. Metherell

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar

Li F. Chan

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Peter King

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Helen L. Storr

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Leonardo Guasti

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar

Linda B. Johnston

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar

Sadani N. Cooray

Queen Mary University of London

View shared research outputs
Researchain Logo
Decentralizing Knowledge