Network


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

Hotspot


Dive into the research topics where Matthew A. Brooke is active.

Publication


Featured researches published by Matthew A. Brooke.


The New England Journal of Medicine | 2011

Inflammatory Skin and Bowel Disease Linked to ADAM17 Deletion

Diana C. Blaydon; Paolo Biancheri; Wei Li Di; Vincent Plagnol; Rita M. Cabral; Matthew A. Brooke; David A. van Heel; Franz Rüschendorf; Mark Toynbee; Amanda J. Walne; Edel A. O'Toole; Joanne E. Martin; Keith J. Lindley; Tom Vulliamy; Dominic Abrams; Thomas T. MacDonald; John I. Harper; David P. Kelsell

We performed genetic and immunohistochemical studies in a sister and brother with autosomal recessive neonatal inflammatory skin and bowel lesions. The girl died suddenly at 12 years of age from parvovirus B19-associated myocarditis; her brother had mild cardiomyopathy. We identified a loss-of-function mutation in ADAM17, which encodes a disintegrin and metalloproteinase 17 (also called tumor necrosis factor α [TNF-α]-converting enzyme, or TACE), as the probable cause of this syndrome. Peripheral-blood mononuclear cells (PBMCs) obtained from the brother at 17 years of age showed high levels of lipopolysaccharide-induced production of interleukin-1β and interleukin-6 but impaired release of TNF-α. Despite repeated skin infections, this young man has led a relatively normal life. (Funded by Barts and the London Charity and the European Commission Seventh Framework Programme.).


The Journal of Pathology | 2012

Cell-cell connectivity: desmosomes and disease.

Matthew A. Brooke; Daniela Nitoiu; David P. Kelsell

Cell–cell connectivity is an absolute requirement for the correct functioning of cells, tissues and entire organisms. At the level of the individual cell, direct cell–cell adherence and communication is mediated by the intercellular junction complexes: desmosomes, adherens, tight and gap junctions. A broad spectrum of inherited, infectious and auto‐immune diseases can affect the proper function of intercellular junctions and result in either diseases affecting specific individual tissues or widespread syndromic conditions. A particularly diverse group of diseases result from direct or indirect disruption of desmosomes—a consequence of their importance in tissue integrity, their extensive distribution, complex structure, and the wide variety of functions their components accomplish. As a consequence, disruption of desmosomal assembly, structure or integrity disrupts not only their intercellular adhesive function but also their functions in cell communication and regulation, leading to such diverse pathologies as cardiomyopathy, epidermal and mucosal blistering, palmoplantar keratoderma, woolly hair, keratosis, epidermolysis bullosa, ectodermal dysplasia and alopecia. Here, as well as describing the importance of the other intercellular junctions, we focus primarily on the desmosome, its structure and its role in disease. We will examine the various pathologies that result from impairment of desmosome function and thereby demonstrate the importance of desmosomes to tissues and to the organism as a whole. Copyright


Gut | 2014

Cryptogenic multifocal ulcerating stenosing enteritis associated with homozygous deletion mutations in cytosolic phospholipase A2-α

Matthew A. Brooke; Hilary J. Longhurst; Vincent Plagnol; Nicholas S. Kirkby; Jane A. Mitchell; Franz Rüschendorf; Timothy D. Warner; David P. Kelsell; Thomas T. MacDonald

Objective Cryptogenic multifocal ulcerating stenosing enteritis (CMUSE) is an extremely rare, but devastating, disease of unknown aetiology. We investigated the genetic basis of this autosomal recessive condition in a pair of affected siblings who have 40-year histories of catastrophic gastrointestinal and extraintestinal disease. Design Genome-wide single-nucleotide polymorphism homozygosity mapping in the two affected family members combined with whole-exome sequencing of one affected sibling. This was followed by confirmatory Sanger sequencing of the likely disease-causing sequence variant and functional studies in affected and unaffected family members. Results Insertion/deletion variation analysis revealed the presence of a homozygous 4 bp deletion (g.155574_77delGTAA) in the PLA2G4A gene, located in the splice donor site directly after exon 17 (the penultimate exon) of the gene in both affected siblings. This introduces a frameshift of 10 amino acids before a premature stop codon (p.V707fsX10), which is predicted to result in the loss of 43 amino acids (residues 707–749) at the C-terminus of cytosolic phospholipase A2-α (cPLA2α). cPLA2α protein expression was undetectable in the gut of both siblings, with platelet aggregation and thromboxane A2 production, as functional assays for cPLA2α activity, grossly impaired. Conclusions We have identified mutations in PLA2G4A as a cause of CMUSE in two affected siblings. Further studies are needed to determine if mutations in this gene are also responsible for disease of a similar phenotype in other cases.


Human Molecular Genetics | 2014

iRHOM2-dependent regulation of ADAM17 in cutaneous disease and epidermal barrier function

Matthew A. Brooke; Sarah L. Etheridge; Nihal Kaplan; Charlotte Simpson; Edel A. O'Toole; Akemi Ishida-Yamamoto; Olivier Marchès; Spiro Getsios; David P. Kelsell

iRHOM2 is a highly conserved, catalytically inactive member of the Rhomboid family, which has recently been shown to regulate the maturation of the multi-substrate ectodomain sheddase enzyme ADAM17 (TACE) in macrophages. Dominant iRHOM2 mutations are the cause of the inherited cutaneous and oesophageal cancer-susceptibility syndrome tylosis with oesophageal cancer (TOC), suggesting a role for this protein in epithelial cells. Here, using tissues derived from TOC patients, we demonstrate that TOC-associated mutations in iRHOM2 cause an increase in the maturation and activity of ADAM17 in epidermal keratinocytes, resulting in significantly upregulated shedding of ADAM17 substrates, including EGF-family growth factors and pro-inflammatory cytokines. This activity is accompanied by increased EGFR activity, increased desmosome processing and the presence of immature epidermal desmosomes, upregulated epidermal transglutaminase activity and heightened resistance to Staphylococcal infection in TOC keratinocytes. Many of these features are consistent with the presence of a constitutive wound-healing-like phenotype in TOC epidermis, which may shed light on a novel pathway in skin repair, regeneration and inflammation.


Science Signaling | 2015

Deletions in the cytoplasmic domain of iRhom1 and iRhom2 promote shedding of the TNF receptor by the protease ADAM17.

Sathish Kumar Maney; David R. McIlwain; Robin Polz; Aleksandra A. Pandyra; Balamurugan Sundaram; Dorit Wolff; Kazuhito Ohishi; Thorsten Maretzky; Matthew A. Brooke; Astrid Evers; Ananda Ayyappan Jaguva Vasudevan; Nima Aghaeepour; Jürgen Scheller; Carsten Münk; Dieter Häussinger; Tak W. Mak; Garry P. Nolan; David P. Kelsell; Carl P. Blobel; Karl S. Lang; Philipp A. Lang

Without the N terminus, iRhom proteins cannot properly limit ADAM17 activity, resulting in impaired cancer cell death. Tumor susceptibility from truncated rhomboids Tumor necrosis factor (TNF) is an extracellular signal that can trigger cell death through its receptor. The protease ADAM17 has a dual role in regulating TNF signaling: ADAM17 promotes TNF signaling by cleaving and releasing TNF from the cell surface, and ADAM17 dampens TNF signaling by cleaving and releasing TNF receptors from the surface. The rhomboid proteins iRhom1 and iRhom2, which lack catalytic activity, mediate the maturation and delivery of ADAM17 to the cell surface. Maney et al. found that deletions in the cytoplasmic region of iRhom1 or iRhom2, which mimic mutations in the N-terminal cytoplasmic tail of iRhom2 in some patients with susceptibility to esophageal cancer, reduced TNF signaling, despite increasing ADAM17 activity. Expression of N-terminally truncated iRhoms in mouse fibrosarcoma cells increased the abundance of ADAM17 at the surface and the subsequent shedding of the TNF receptors, thereby suppressing TNF-induced intracellular signaling and cell death. The protease ADAM17 (a disintegrin and metalloproteinase 17) catalyzes the shedding of various transmembrane proteins from the surface of cells, including tumor necrosis factor (TNF) and its receptors. Liberation of TNF receptors (TNFRs) from cell surfaces can dampen the cellular response to TNF, a cytokine that is critical in the innate immune response and promotes programmed cell death but can also promote sepsis. Catalytically inactive members of the rhomboid family of proteases, iRhom1 and iRhom2, mediate the intracellular transport and maturation of ADAM17. Using a genetic screen, we found that the presence of either iRhom1 or iRhom2 lacking part of their extended amino-terminal cytoplasmic domain (herein referred to as ΔN) increases ADAM17 activity, TNFR shedding, and resistance to TNF-induced cell death in fibrosarcoma cells. Inhibitors of ADAM17, but not of other ADAM family members, prevented the effects of iRhom-ΔN expression. iRhom1 and iRhom2 were functionally redundant, suggesting a conserved role for the iRhom amino termini. Cells from patients with a dominantly inherited cancer susceptibility syndrome called tylosis with esophageal cancer (TOC) have amino-terminal mutations in iRhom2. Keratinocytes from TOC patients exhibited increased TNFR1 shedding compared with cells from healthy donors. Our results explain how loss of the amino terminus in iRhom1 and iRhom2 impairs TNF signaling, despite enhancing ADAM17 activity, and may explain how mutations in the amino-terminal region contribute to the cancer predisposition syndrome TOC.


Cell and Tissue Research | 2013

Rhomboid proteins: a role in keratinocyte proliferation and cancer

Sarah L. Etheridge; Matthew A. Brooke; David P. Kelsell; Diana C. Blaydon

The Rhomboids represent a relatively recently discovered family of proteins, consisting in a variety of intramembrane serine proteases and their inactive homologues, the iRhoms. Rhomboids typically contain six or seven transmembrane domains (TMD) and have been classified into four subgroups: Secretase A and B, Presenilin-Associated-Rhomboid-Like (PARL) and iRhoms. Although the iRhoms, iRhom1 and iRhom2, have lost their protease activity during evolution, they retain key non-protease functions and have been implicated in the regulation of epidermal growth factor (EGF) signalling. EGF is moreover a substrate of RHBDL2, their active Rhomboid relative. Other substrates of RHBDL2 include members of the EphrinB family and thrombomodulin. RHBDL2 has also previously been demonstrated to be important in wound healing in cutaneous keratinocytes through the cleavage of thrombomodulin. Additional roles for these intriguing proteins seem likely to be revealed in the future. This review focuses on our current understanding of Rhomboids and, in particular, on RHBDL2 and iRhom2 and their roles in cellular processes and human disease.


Nature Communications | 2018

p63 is a key regulator of iRHOM2 signalling in the keratinocyte stress response

Paola Arcidiacono; Catherine M. Webb; Matthew A. Brooke; Huiqing Zhou; Paul J. Delaney; Keat-Eng Ng; David P. Kelsell; Anissa Chikh

Hyperproliferative keratinocytes induced by trauma, hyperkeratosis and/or inflammation display molecular signatures similar to those of palmoplantar epidermis. Inherited gain-of-function mutations in RHBDF2 (encoding iRHOM2) are associated with a hyperproliferative palmoplantar keratoderma and squamous oesophageal cancer syndrome (termed TOC). In contrast, genetic ablation of rhbdf2 in mice leads to a thinning of the mammalian footpad, and reduces keratinocyte hyperproliferation and migration. Here, we report that iRHOM2 is a novel target gene of p63 and that both p63 and iRHOM2 differentially regulate cellular stress-associated signalling pathways in normal and hyperproliferative keratinocytes. We demonstrate that p63–iRHOM2 regulates cell survival and response to oxidative stress via modulation of SURVIVIN and Cytoglobin, respectively. Furthermore, the antioxidant compound Sulforaphane downregulates p63–iRHOM2 expression, leading to reduced proliferation, inflammation, survival and ROS production. These findings elucidate a novel p63-associated pathway that identifies iRHOM2 modulation as a potential therapeutic target to treat hyperproliferative skin disease and neoplasia.Mutations in the gene encoding iRHOM2 are associated with hyperproliferative epidermal disorders. Here, the authors show that iRHOM2 is a target gene of p63, that together they regulate inflammation, cell survival and response to oxidative stress, and inhibition of p63-iRHOM2 signalling with an antioxidant reduces epidermal inflammation.


Scientific Reports | 2016

ADAM17/EGFR axis promotes transglutaminase-dependent skin barrier formation through phosholipase C γ1 and protein kinase C pathways.

Cristina Wolf; Yawen Qian; Matthew A. Brooke; David P. Kelsell; Claus-Werner Franzke

The vitally important skin barrier is formed by extensive cross-linking activity of transglutaminases (TGs) during terminal epidermal differentiation. We have previously shown that epidermal deficiency of a disintegrin and metalloproteinase 17 (ADAM17), the principal EGFR ligand sheddase, results in postnatal skin barrier defects in mice due to impeded TG activity. However, the mechanism by which ADAM17/EGFR signalling maintains TG activity during epidermal differentiation remains elusive. Here we demonstrate that ADAM17-dependent EGFR signalling promotes TG activity in keratinocytes committed to terminal differentiation by direct induction of TG1 expression. Restored TG1 expression of EGF-stimulated differentiated Adam17−/− keratinocytes was strongly repressed by inhibitors for PLCγ1 or protein kinase C (PKC) pathways, while treatment with the PKC stimulator 12-O-tetradecanoylphorbol-13-acetate restored TG activity in the epidermis of keratinocyte-specific Adam17−/− (AD17ΔKC) mice. Further investigations emphasized the expression of PKCη, a mediator of TGM1 transcription, to be sensitive to EGFR activation. In agreement, topical skin application of cholesterol sulfate, an activator of PKCη, significantly improved TG activity in epidermis of AD17ΔKC mice. Our results suggest ADAM17/EGFR-driven PLCγ1 and PKC pathways as important promoters of TG1 expression during terminal keratinocyte differentiation. These findings may help to identify new therapeutic targets for inflammatory skin diseases related to epidermal barrier defects.


Journal of Investigative Dermatology | 2014

Exoming into Rare Skin Disease: EGFR Deficiency

Matthew A. Brooke; Edel A. O'Toole; David P. Kelsell

Dermatologists are frequently asked to see patients with pustular eruptions caused by EGFR mAbs or tyrosine kinase inhibitors. In this issue, Campbell et al. describe an infant with severely inflammed skin and bowel and lung disease caused by a homozygous mutation in the EGFR gene. This commentary discusses the power of exome sequencing in disease gene discovery within the rare genodermatoses and the role of aberrant EGFR signaling in a subset of monogenic skin and epithelial syndromes.


Nature Communications | 2017

Rhomboid family member 2 regulates cytoskeletal stress-associated Keratin 16

Thiviyani Maruthappu; Anissa Chikh; Benjamin Fell; Paul J. Delaney; Matthew A. Brooke; Clemence Levet; Angela Moncada-Pazos; Akemi Ishida-Yamamoto; Diana C. Blaydon; Ahmad Waseem; Irene M. Leigh; Matthew Freeman; David P. Kelsell

Collaboration


Dive into the Matthew A. Brooke's collaboration.

Top Co-Authors

Avatar

David P. Kelsell

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar

Diana C. Blaydon

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar

Anissa Chikh

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar

Edel A. O'Toole

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar

Thomas T. MacDonald

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar

Benjamin Fell

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar

Joanne E. Martin

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar

John I. Harper

Great Ormond Street Hospital

View shared research outputs
Top Co-Authors

Avatar

Paolo Biancheri

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar

Paul J. Delaney

Queen Mary University of London

View shared research outputs
Researchain Logo
Decentralizing Knowledge