Network


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

Hotspot


Dive into the research topics where Martin Brockington is active.

Publication


Featured researches published by Martin Brockington.


American Journal of Human Genetics | 2001

Mutations in the fukutin-related protein gene (FKRP) cause a form of congenital muscular dystrophy with secondary laminin alpha2 deficiency and abnormal glycosylation of alpha-dystroglycan.

Martin Brockington; Derek J. Blake; Paola Prandini; Susan C. Brown; Silvia Torelli; Matthew A. Benson; Chris P. Ponting; Brigitte Estournet; Norma B. Romero; Eugenio Mercuri; Thomas Voit; C. Sewry; Pascale Guicheney; Francesco Muntoni

The congenital muscular dystrophies (CMD) are a heterogeneous group of autosomal recessive disorders presenting in infancy with muscle weakness, contractures, and dystrophic changes on skeletal-muscle biopsy. Structural brain defects, with or without mental retardation, are additional features of several CMD syndromes. Approximately 40% of patients with CMD have a primary deficiency (MDC1A) of the laminin alpha2 chain of merosin (laminin-2) due to mutations in the LAMA2 gene. In addition, a secondary deficiency of laminin alpha2 is apparent in some CMD syndromes, including MDC1B, which is mapped to chromosome 1q42, and both muscle-eye-brain disease (MEB) and Fukuyama CMD (FCMD), two forms with severe brain involvement. The FCMD gene encodes a protein of unknown function, fukutin, though sequence analysis predicts it to be a phosphoryl-ligand transferase. Here we identify the gene for a new member of the fukutin protein family (fukutin related protein [FKRP]), mapping to human chromosome 19q13.3. We report the genomic organization of the FKRP gene and its pattern of tissue expression. Mutations in the FKRP gene have been identified in seven families with CMD characterized by disease onset in the first weeks of life and a severe phenotype with inability to walk, muscle hypertrophy, marked elevation of serum creatine kinase, and normal brain structure and function. Affected individuals had a secondary deficiency of laminin alpha2 expression. In addition, they had both a marked decrease in immunostaining of muscle alpha-dystroglycan and a reduction in its molecular weight on western blot analysis. We suggest these abnormalities of alpha-dystroglycan are caused by its defective glycosylation and are integral to the pathology seen in MDC1C.


Journal of Medical Genetics | 2005

POMT2 mutations cause alpha-dystroglycan hypoglycosylation and Walker-Warburg syndrome

J. van Reeuwijk; M.H.M. Janssen; C. van der Elzen; D. Beltran Valero de Bernabe; P. Sabatelli; Luciano Merlini; M. Boon; H. Scheffer; Martin Brockington; Francesco Muntoni; Martijn A. Huynen; Aad Verrips; Christopher A. Walsh; Peter G. Barth; Han G. Brunner; J.H.L.M. van Bokhoven

Background: Walker-Warburg syndrome (WWS) is an autosomal recessive condition characterised by congenital muscular dystrophy, structural brain defects, and eye malformations. Typical brain abnormalities are hydrocephalus, lissencephaly, agenesis of the corpus callosum, fusion of the hemispheres, cerebellar hypoplasia, and neuronal overmigration, which causes a cobblestone cortex. Ocular abnormalities include cataract, microphthalmia, buphthalmos, and Peters anomaly. WWS patients show defective O-glycosylation of α-dystroglycan (α-DG), which plays a key role in bridging the cytoskeleton of muscle and CNS cells with extracellular matrix proteins, important for muscle integrity and neuronal migration. In 20% of the WWS patients, hypoglycosylation results from mutations in either the protein O-mannosyltransferase 1 (POMT1), fukutin, or fukutin related protein (FKRP) genes. The other genes for this highly heterogeneous disorder remain to be identified. Objective: To look for mutations in POMT2 as a cause of WWS, as both POMT1 and POMT2 are required to achieve protein O-mannosyltransferase activity. Methods: A candidate gene approach combined with homozygosity mapping. Results: Homozygosity was found for the POMT2 locus at 14q24.3 in four of 11 consanguineous WWS families. Homozygous POMT2 mutations were present in two of these families as well as in one patient from another cohort of six WWS families. Immunohistochemistry in muscle showed severely reduced levels of glycosylated α-DG, which is consistent with the postulated role for POMT2 in the O-mannosylation pathway. Conclusions: A fourth causative gene for WWS was uncovered. These genes account for approximately one third of the WWS cases. Several more genes are anticipated, which are likely to play a role in glycosylation of α-DG.


Nature Genetics | 2001

Mutations in SEPN1 cause congenital muscular dystrophy with spinal rigidity and restrictive respiratory syndrome.

Behzad Moghadaszadeh; Nathalie Petit; Céline Jaillard; Martin Brockington; Susana Quijano Roy; Luciano Merlini; Norma B. Romero; Brigitte Estournet; Isabelle Desguerre; Denys Chaigne; Francesco Muntoni; Haluk Topaloglu; Pascale Guicheney

One form of congenital muscular dystrophy, rigid spine syndrome (MIM 602771), is a rare neuromuscular disorder characterized by early rigidity of the spine and respiratory insufficiency. A locus on 1p35–36 (RSMD1) was recently found to segregate with rigid spine muscular dystrophy 1 (ref. 1). Here we refine the locus and find evidence of linkage disequilibrium associated with SEPN1, which encodes the recently described selenoprotein N (ref. 2). Our identification and analysis of mutations in SEPN1 is the first description of a selenoprotein implicated in a human disease.


Journal of Medical Genetics | 2004

Mutations in the FKRP gene can cause muscle-eye-brain disease and Walker–Warburg syndrome

D. Beltran Valero de Bernabe; Thomas Voit; Cheryl Longman; Alice Steinbrecher; Volker Straub; Yeliz Yuva; Ralf Herrmann; J. Sperner; C.G. Korenke; Diesen C; William B. Dobyns; Han G. Brunner; J.H.L.M. van Bokhoven; Martin Brockington; F. Muntoni

The hypoglycosylation of α-dystroglycan is a new disease mechanism recently identified in four congenital muscular dystrophies (CMDs): Walker–Warburg syndrome (WWS), muscle-eye-brain disease (MEB), Fukuyama CMD (FCMD), and CMD type 1C (MDC1C).1 The underlying genetic defects in these disorders are mutations in known or putative glycosyltransferase enzymes, which among their targets probably include α-dystroglycan. FCMD (MIM: 253800) is caused by mutations in fukutin2; MEB (MEB [MIM 236670]) is due to mutations in POMGnT13; and in WWS (WWS [MIM: 236670]) POMT1 is mutated.4 In addition to the brain abnormalities, both MEB and WWS have structural eye involvement. In FCMD, eye involvement is more variable, ranging from myopia to retinal detachment, persistent primary vitreous body, persistent hyaloid artery, or microphthalmos.5 WWS, MEB, and FCMD display type II or cobblestone lissencephaly, in which the main abnormality is different degrees of brain malformation secondary at least in part to the overmigration of heterotopic neurones into the leptominenges through gaps in the external (pial) basement membrane.6,7 Whereas there are broad similarities between WWS and MEB, clear diagnostic criteria differentiating between these two conditions have been proposed8 and are shown as clinical features in table 1. A similar combination of muscular dystrophy and cobblestone lissencephaly is also found in the myodystrophy mouse (myd, renamed Largemyd), in which the Large gene is mutated.6,9,10 Our group has very recently identified mutations in the human LARGE gene in a patient with a novel form of CMD (MDC1D).11 View this table: Table 1 Clinical features of patients 1 and 2, compared with MEB and WWS patients with confirmed mutations in POGnT1 and POMT1, respectively The gene encoding the fukutin related protein (FKRP, [MIM 606612]) is mutated in a severe form of CMD (MDC1C, [OMIM 606612]).12 Clinical features of MDC1C are …


Nature Genetics | 2005

Mutations in SIL1 cause Marinesco-Sjogren syndrome, a cerebellar ataxia with cataract and myopathy

Jan Senderek; M. Krieger; Claudia Stendel; Carsten Bergmann; Markus Moser; N. Breitbach-Faller; Sabine Rudnik-Schöneborn; A. Blaschek; N. I. Wolf; I. Harting; Kathryn N. North; Janine Smith; Francesco Muntoni; Martin Brockington; Susana Quijano-Roy; F. Renault; Ralf Herrmann; L. M. Hendershot; J. M. Schroder; Hanns Lochmüller; Haluk Topaloglu; Thomas Voit; Joachim Weis; F. Ebinger; Klaus Zerres

SIL1 (also called BAP) acts as a nucleotide exchange factor for the Hsp70 chaperone BiP (also called GRP78), which is a key regulator of the main functions of the endoplasmic reticulum. We found nine distinct mutations that would disrupt the SIL1 protein in individuals with Marinesco-Sjögren syndrome, an autosomal recessive cerebellar ataxia complicated by cataracts, developmental delay and myopathy. Identification of SIL1 mutations implicates Marinesco-Sjögren syndrome as a disease of endoplasmic reticulum dysfunction and suggests a role for this organelle in multisystem disorders.


Annals of Neurology | 2003

Phenotypic spectrum associated with mutations in the fukutin-related protein gene

Eugenio Mercuri; Martin Brockington; Volker Straub; Susana Quijano-Roy; Yeliz Yuva; Ralf Herrmann; Susan C. Brown; Silvia Torelli; Victor Dubowitz; Derek J. Blake; Norma B. Romero; Brigitte Estournet; C. Sewry; Pascale Guicheney; Thomas Voit; Francesco Muntoni

We describe 22 patients with mutations in the fukutin‐related protein (FKPR) gene. Four patients had congenital muscular dystrophy (MDC1C), with presentation at birth, severe weakness and inability to stand unsupported. The other 18 had limb girdle muscular dystrophy (LGMD2I). Eleven showed a Duchenne‐like course with loss of ambulation in the early teens while 7 had a milder phenotype. Muscle biopsy invariably showed abnormal expression of a‐dystroglycan. MDC1C patients either carried 2 missense or 1 missense and 1 nonsense mutations. Patients with LGMD2I shared a common mutation (C826A,Leu276Ileu) and their phenotypic severity was correlated with the second allelic mutation. Ann Neurol 2003;53:537–542


American Journal of Pathology | 2004

Abnormalities in α-dystroglycan expression in MDC1C and LGMD2I muscular dystrophies

Susan C. Brown; Silvia Torelli; Martin Brockington; Yeliz Yuva; Cecilia Jimenez; L. Feng; Louise V. B. Anderson; Isabella Ugo; Stephan Kröger; Kate Bushby; Thomas Voit; Caroline Sewry; Francesco Muntoni

We recently identified mutations in the fukutin related protein (FKRP) gene in patients with congenital muscular dystrophy type 1C (MDC1C) and limb girdle muscular dystrophy type 2I (LGMD2I). The sarcolemma of these patients typically displays an immunocytochemical reduction of α-dystroglycan. In this report we extend these observations and report a clear correlation between the residual expression of α-dystroglycan and the phenotype. Three broad categories were identified. Patients at the severe end of the clinical spectrum (MDC1C) were compound heterozygote between a null allele and a missense mutation or carried two missense mutations and displayed a profound depletion of α-dystroglycan. Patients with LGMD with a Duchenne-like severity typically had a moderate reduction in α-dystroglycan and were compound heterozygotes between a common C826A (Leu276Ileu) FKRP mutation and either a missense or a nonsense mutation. Individuals with the milder form of LGMD2I were almost invariably homozygous for the Leu276Ile FKRP mutation and showed a variable but subtle alteration in α-dystroglycan immunolabeling. Our data therefore suggest a correlation between a reduction in α-dystroglycan, the mutation and the clinical phenotype in MDC1C and LGMD2I which supports the hypothesis that dystroglycan plays a central role in the pathogenesis of these disorders.


Neurology | 2003

The phenotype of limb-girdle muscular dystrophy type 2I

Maja Poppe; Lynsey Cree; John P. Bourke; M. Eagle; Lvb Anderson; D Birchall; Martin Brockington; M Buddles; M Busby; F. Muntoni; A Wills; Kate Bushby

Background: Mutations in the fukutin-related protein gene FKRP cause limb-girdle muscular dystrophy (LGMD2I) as well as a form of congenital muscular dystrophy (MDC1C). Objective: To define the phenotype in LGMD2I. Methods: The authors assessed 16 patients from 14 families with FKRP gene mutations and LGMD and collected the results of mutation analysis, protein studies, and respiratory and cardiac investigations. Results: Thirteen patients, most with adult presentation, were homozygous for the common C826A mutation in FKRP. The three other cases were compound heterozygotes for C826A and two of them presented in childhood, with more progressive disease. The pattern of muscle involvement, frequently including calf hypertrophy, was similar to dystrophinopathy. Complications in patients with LGMD2I were common and sometimes out of proportion to the skeletal muscle involvement. Six patients had cardiac involvement, and 10 had respiratory impairment: five required nocturnal respiratory support. All patients had serum creatine kinase at least 5 to 70 times normal. The most consistent protein abnormality found on muscle biopsy was a reduction of laminin α2 immunolabeling, either on muscle sections or immunoblotting alone. Conclusions: LGMD2I due to FKRP mutations appears to be a relatively common cause of LGMD, with respiratory and cardiac failure as prominent complications.


The Lancet | 2002

Defective glycosylation in muscular dystrophy

Francesco Muntoni; Martin Brockington; Derek J. Blake; Silvia Torelli; Susan C. Brown

CONTEXT Over the past 15 years the causative genes of several inherited muscular dystrophies have been identified. These genes encode sarcolemmal, extracellular matrix, sarcomeric, and nuclear envelope proteins. Although the post-translational processing of muscle proteins has a significant role in their correct assembly and function, these processes have not been shown to be primarily involved in the pathogenesis of muscular dystrophies until recently. In the past 18 months, four different forms of inherited muscular dystrophy in human beings have been associated with mutations in genes encoding for putative glycosyltransferases. Aberrant glycosylation of alpha-dystroglycan, an external membrane protein expressed in muscle, brain, and other tissues, is a common feature in these disorders. alpha-dystroglycan is highly glycosylated, its sugar components varying in different tissues and controlling its interaction with extracellular matrix partners. Disrupted glycosylation of alpha-dystroglycan results in a loss of these interactions, giving rise to both progressive muscle degeneration and abnormal neuronal migration in the brain. STARTING POINT Kevin Campbell and colleagues have recently demonstrated that patients with muscle-eye-brain disease (MEB) and Fukuyama congenital muscular dystrophy (FCMD), as well as the myodystrophy (myd) mouse, have an abnormally glycosyated form of alpha-dystroglycan (Nature 2002; 418: 417-22 and 422-25). The abnormally glycosylated protein did not bind to three of its extracellular matrix ligands, laminin alpha2 chain, agrin, and neurexin. The investigators also showed that a neuronal migration disorder occurs in both the myd mouse and in a brain-restricted alpha-dystroglycan knock-out mouse that is similar to that seen in patients with MEB and FCMD. These results identify alpha-dystroglycan as having an essential role in both muscle and brain development and function. WHERE NEXT Emphasis is moving away from identifying the protein components of the muscle fibre that are involved in muscular dystrophies towards the post-translational processing of proteins and the enzymes involved in these modifications. This opens up new avenues of research. Abnormal glycosylation of alpha-dystroglycan may underlie other as yet uncharacterised forms of muscular dystrophy and neuronal migration disorders.


Current Opinion in Neurology | 2004

Defective glycosylation in congenital muscular dystrophies

Francesco Muntoni; Martin Brockington; Silvia Torelli; Susan C. Brown

Purpose of reviewThe recent identification of mutations in five genes coding for proteins with putative or demonstrated glycosyltransferase activity has shed light on a novel mechanism responsible for muscular dystrophy. Abnormal glycosylation of α-dystroglycan appears to be a common finding in all these conditions. Surprisingly, the disease severity due to mutations in several of these genes is extremely variable. This article provides an overview of the clinical, biochemical and genetic advances that have been made over the last year in this field. Recent findingsMutations in the human LARGE gene, a putative glycosyltransferase mutated in the myodystrophy mouse, have now been identified in a form of human muscular dystrophy. In addition, the clinical variability of patients with mutations in the genes encoding fukutin, protein O-linked mannose β1,2-N-acetylglucosaminyltransferase 1 and the fukutin-related protein has been significantly expanded. Disease severity in patients with mutations in the gene encoding the fukutin-related protein varies from a severe prenatal form of congenital muscular dystrophy with cobblestone lissencephaly and structural eye defects to a mild form of limb-girdle muscular dystrophy with onset in adult life and neither brain nor eye involvement. SummaryGlycosylation disorders represent a rapidly growing and common group of muscular dystrophies. Accurate genetic diagnosis can now be made for five forms, and it is anticipated that several other variants will eventually fall into these categories.

Collaboration


Dive into the Martin Brockington's collaboration.

Top Co-Authors

Avatar

Francesco Muntoni

Great Ormond Street Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Silvia Torelli

UCL Institute of Child Health

View shared research outputs
Top Co-Authors

Avatar

C. Sewry

Great Ormond Street Hospital

View shared research outputs
Top Co-Authors

Avatar

Thomas Voit

University College London

View shared research outputs
Top Co-Authors

Avatar

L. Feng

UCL Institute of Child Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Cheryl Longman

Southern General Hospital

View shared research outputs
Top Co-Authors

Avatar

Eugenio Mercuri

The Catholic University of America

View shared research outputs
Researchain Logo
Decentralizing Knowledge