Matthew Arno
King's College London
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Matthew Arno.
Science | 2016
Giuseppina Arbore; Erin E. West; Rosanne Spolski; Avril A. B. Robertson; Andreas Klos; Claudia Rheinheimer; Pavel Dutow; Trent M. Woodruff; Zu Xi Yu; Luke A. J. O'Neill; Rebecca C. Coll; Alan Sher; Warren J. Leonard; Jörg Köhl; Peter N. Monk; Matthew A. Cooper; Matthew Arno; Behdad Afzali; Helen J. Lachmann; Andrew P. Cope; Katrin D. Mayer-Barber; Claudia Kemper
Innate immune crosstalk in T cells The classical view of immune activation is that innate immune cells, such as macrophages and dendritic cells, recognize invading microbes and then alert adaptive immune cells, such as T cells, to respond. Arbore et al. now show that innate and adaptive immunity converge in human and mouse T cells. Activated T cells express components of the complement cascade, which in turn leads to the assembly of NLRP3 inflammasomes—both critical components of innate immunity that help hosts detect and eliminate microbes. In T cells, complement and inflammasomes work together to push T cells to differentiate into a specialized subset of T cells important for eliminating intracellular bacteria. Science, this issue p. 10.1126/science.aad1210 Complement and NLRP3 inflammasomes work together to promote T helper 1 cell differentiation. INTRODUCTION The inflammasomes and the complement system are traditionally viewed as quintessential components of innate immunity required for the detection and elimination of pathogens. Assembly of the NLRP3 inflammasome in innate immune cells controls the maturation of interleukin (IL)–1β, a proinflammatory cytokine critical to host defense, whereas activation of the liver-derived complement key components C3 and C5 in serum leads to opsonization and removal of microbes and induction of the inflammatory reaction. Recent studies, however, have highlighted an unanticipated direct role for complement C3 also in human T cell immunity: The anaphylatoxin C3a receptor (C3aR) and the complement regulator CD46 (which binds C3b) are critical checkpoints in human T cell lineage commitment, and they control initiation and resolution of T helper 1 (TH1) responses in an autocrine fashion via T cell–derived and intracellularly activated C3. We explored a novel functional cross-talk of complement with the NLRP3 inflammasome within CD4+ T cells and determined how the cooperation between these two “classically” innate systems directly affects interferon-γ (IFN-γ) production by adaptive immune cells. RATIONALE Given the critical role of intracellular C3 activation in human TH1 responses and the importance of C5 activation products in inflammation, we investigated whether human CD4+ T cells also harbor an “intracellular C5 activation system” and by what means this system may contribute to effector responses by using C5aR1 and C5aR2 agonists and antagonists, T cells from patients with cryopyrin-associated periodic syndromes (CAPS), and mouse models of infection and autoimmunity. RESULTS Human CD4+ T cells expressed C5 and generated increased intracellular C5a upon T cell receptor activation and CD46 autocrine costimulation. Subsequent engagement of the intracellular C5aR1 by C5a induced the generation of reactive oxygen species (ROS) and the unexpected assembly of a functional NLRP3 inflammasome in CD4+ T cells, whereas the surface-expressed C5aR2 negatively controlled this process. NLRP3 inflammasome–dependent autocrine IL-1β secretion and activity were required for optimal IFN-γ production by T cells; consequently, dysregulation of NLRP3 function in these cells affected their normal effector responses. For example, mutated, constitutively active NLRP3 in T cells from patients with CAPS induced hyperactive TH1 responses that could be normalized with a NLRP3 inhibitor. The in vivo importance of a T cell–intrinsic NLRP3 inflammasome was further supported by the finding that IFN-γ production by Nlrp3–/– CD4+ T cells was significantly reduced during viral infections in mice and that diminished TH1 induction due to lack of NLRP3 function in a CD4+ T cell transfer model of colitis led to uncontrolled TH17 infiltration and/or expansion in the intestine and aggravated disease. CONCLUSION Our results demonstrate that the regulated cross-talk between intracellularly activated complement components (the “complosome”) and the NLRP3 inflammasome is fundamental to human TH1 induction and regulation. The finding that established innate immune pathways are also operative in adaptive immune cells and orchestrate immunological responses contributes to our understanding of immunobiology and immune system evolution. In addition, the results suggest that the complement-NLRP3 axis in T cells represents a novel therapeutic target for the modulation of TH1 activity in autoimmunity and infection. An intrinsic complement-NLRP3 axis regulates human TH1 responses. T cell receptor activation and CD46 costimulation trigger NLRP3 expression and intracellular C5a generation. Subsequent intracellular C5aR1 engagement induces ROS production (and possibly IL1B gene transcription) and NLRP3 assembly, which in turn mediates IL-1β maturation. Autocrine IL-1β promotes TH1 induction (IFN-γ production) but restricts TH1 contraction (IL-10 coexpression). C5aR2 cell surface activation by secreted C5a negatively controls these events via undefined mechanisms. Dysfunction of this system contributes to impaired TH1 responses in infection or increased TH17 responses during intestinal inflammation. The NLRP3 inflammasome controls interleukin-1β maturation in antigen-presenting cells, but a direct role for NLRP3 in human adaptive immune cells has not been described. We found that the NLRP3 inflammasome assembles in human CD4+ T cells and initiates caspase-1–dependent interleukin-1β secretion, thereby promoting interferon-γ production and T helper 1 (TH1) differentiation in an autocrine fashion. NLRP3 assembly requires intracellular C5 activation and stimulation of C5a receptor 1 (C5aR1), which is negatively regulated by surface-expressed C5aR2. Aberrant NLRP3 activity in T cells affects inflammatory responses in human autoinflammatory disease and in mouse models of inflammation and infection. Our results demonstrate that NLRP3 inflammasome activity is not confined to “innate immune cells” but is an integral component of normal adaptive TH1 responses.
Free Radical Biology and Medicine | 2011
Alison C. Brewer; Thomas V.A. Murray; Matthew Arno; Min Zhang; Narayana Anilkumar; Giovanni E. Mann; Ajay M. Shah
NADPH oxidase-4 (Nox4) is an important modulator of redox signaling that is inducible at the level of transcriptional expression in multiple cell types. By contrast to other Nox enzymes, Nox4 is continuously active without requiring stimulation. We reported recently that expression of Nox4 is induced in the adult heart as an adaptive stress response to pathophysiological insult. To elucidate the potential downstream target(s) regulated by Nox4, we performed a microarray screen to assess the transcriptomes of transgenic (tg) mouse hearts in which Nox4 was overexpressed. The screen revealed a significant increase in the expression of many antioxidant and detoxifying genes regulated by Nrf2 in tg compared to wild-type (wt) mouse hearts, and this finding was subsequently confirmed by Q-PCR. Expression of glutathione biosynthetic and recycling enzymes was increased in tg hearts and associated with higher levels of both GSH and the ratio of reduced:oxidised GSH, compared to wt hearts. The increases in expression of the antioxidant genes and the changes in glutathione redox effected by Nox4 were ablated in an Nrf2-null genetic background. These data therefore demonstrate that Nox4 can activate the Nrf2-regulated pathway, and suggest a potential role for Nox4 in the regulation of GSH redox in cardiomyocytes.
Leukemia | 2006
W Ingram; Nicholas Lea; José Cervera; Ulrich Germing; Pierre Fenaux; Bruno Cassinat; Jean-Jacques Kiladjian; Judit Várkonyi; Petar Antunovic; Nigel Westwood; Matthew Arno; Azim Mohamedali; Joop Gaken; T. Kontou; Barbara Czepulkowski; N A Twine; J Tamaska; J Csomer; S Benedek; Norbert Gattermann; E Zipperer; A. Giagounidis; Zaida Garcia-Casado; Guillermo Sanz; Ghulam J. Mufti
The JAK2 V617F mutation identifies a subgroup of MDS patients with isolated deletion 5q and a proliferative bone marrow
Immunity | 2015
Chrysothemis Brown; Daria Esterházy; Aurelien Sarde; Mariya London; Venu Pullabhatla; Ines Osma-Garcia; Raya al-Bader; Carla Ortiz; Raul Elgueta; Matthew Arno; Emanuele de Rinaldis; Daniel Mucida; Graham M. Lord; Randolph J. Noelle
Summary CD4+ T cells differentiate into phenotypically distinct T helper cells upon antigenic stimulation. Regulation of plasticity between these CD4+ T-cell lineages is critical for immune homeostasis and prevention of autoimmune disease. However, the factors that regulate lineage stability are largely unknown. Here we investigate a role for retinoic acid (RA) in the regulation of lineage stability using T helper 1 (Th1) cells, traditionally considered the most phenotypically stable Th subset. We found that RA, through its receptor RARα, sustains stable expression of Th1 lineage specifying genes, as well as repressing genes that instruct Th17-cell fate. RA signaling is essential for limiting Th1-cell conversion into Th17 effectors and for preventing pathogenic Th17 responses in vivo. Our study identifies RA-RARα as a key component of the regulatory network governing maintenance and plasticity of Th1-cell fate and defines an additional pathway for the development of Th17 cells.
Stem cell reports | 2014
Anastasia Petrova; Anna Celli; Laureen Jacquet; Dimitra Dafou; Debra Crumrine; Melanie Hupe; Matthew Arno; Carl Hobbs; Aleksandra Cvoro; Panagiotis Karagiannis; Liani Devito; Richard Sun; Lillian C. Adame; Robert Vaughan; John A. McGrath; Theodora M. Mauro; Dusko Ilic
Summary Cornification and epidermal barrier defects are associated with a number of clinically diverse skin disorders. However, a suitable in vitro model for studying normal barrier function and barrier defects is still lacking. Here, we demonstrate the generation of human epidermal equivalents (HEEs) from human embryonic stem cells (hESCs) and induced pluripotent stem cells (iPSCs). HEEs are structurally similar to native epidermis, with a functional permeability barrier. We exposed a pure population of hESC/iPSC-derived keratinocytes, whose transcriptome corresponds to the gene signature of normal primary human keratinocytes (NHKs), to a sequential high-to-low humidity environment in an air/liquid interface culture. The resulting HEEs had all of the cellular strata of the human epidermis, with skin barrier properties similar to those of normal skin. Such HEEs generated from disease-specific iPSCs will be an invaluable tool not only for dissecting molecular mechanisms that lead to epidermal barrier defects but also for drug development and screening.
Nutrition | 2010
Alex K. Lee; Marjan Mojtahed-Jaberi; Theodosios Kyriakou; Estibaliz Aldecoa-Otalora Astarloa; Matthew Arno; Nichola J. Marshall; Susan D. Brain; Sandra D. O'Dell
Objective Hypothalamic centers integrate external signals of nutrient availability and energy status and initiate responses to maintain homeostasis. Quantifying changes in hypothalamic gene expression in the presence of nutrient excess may identify novel responsive elements. Methods Affymetrix Mouse Genome 430 2.0 oligonucleotide microarrays containing 45 102 probe sets were used to interrogate differential expression of genes in dietary-induced obesity model C57BL6 inbred mice fed a high-fat (35% fat; n = 8) or standard (4% fat; n = 6) diet from 3 to 15 wk of age. Ontologies of regulated genes were examined and expression of selected genes was validated by quantitative real-time polymerase chain reaction. Results One thousand two hundred twelve unique gene transcripts showed altered expression on the microarrays. Gene ontology analysis revealed changes in neuropeptide genes responding to leptin, Pomc, Cart, Npy, and Agrp, compatible with a homeostatic response to high-fat intake, although mean weight increased 2.3-fold compared with standard fed mice (P < 0.001). Neurotransmitter system ontologies revealed upregulation of five genes controlling availability of dopamine. Changes in Th tyrosine hydroxylase (2.1-fold) and Slc18a2 solute carrier family 18 (vesicular monoamine), member 2 (4.4-fold) controlling synthesis and release, and Slc6a3 solute carrier family 6 (neurotransmitter transporter, dopamine), member 3 (4.8-fold), Snca α-synuclein (1.3-fold), and Maoa monoamine oxidase (1.9-fold) limiting availability were confirmed by quantitative real-time polymerase chain reaction. Conclusion Expression of five genes involved in availability of dopamine was increased after a high-fat diet. Failure to reduce dopamine availability sufficiently, to counter the feeding reward effect, could contribute to diet-induced obesity in these mice.
Journal of Endocrinology | 2007
Min Zhao; Stephanie A. Amiel; Michael R. Christie; Paolo Muiesan; Parthi Srinivasan; Wendy Littlejohn; Mohamed Rela; Matthew Arno; Nigel Heaton; Guo Cai Huang
The origin of cells replacing ageing beta-cells in adult life is unknown. This study assessed the expression of classic stem cell markers: Oct4, Sox2 and CD34 in islet-enriched fractions versus exocrine cell-enriched fractions from 25 adult human pancreases following human islet isolation. Expression of Oct4, Sox2 and CD34 mRNAs was found in all cell samples, with no significant differences between endocrine and exocrine cell fractions. Immunohistochemical staining for Oct4, Sox2, CD133, CD34, CK19, insulin and nestin on human pancreas sections showed that the majority of Oct4(+ve) cells were found in the walls of small ducts. Similar localisations were observed for Sox2(+ve) cells. The majority of Sox2(+ve) cells were found to co-express Oct4 proteins, but not vice versa. Cells positive for Oct4 and Sox2 appeared to be a unique cell population in the adult human pancreases without co-expression for CK19, CD34, CD133, insulin and nestin proteins. The numbers of Oct4(+ve) and Sox2(+ve) cells varied among donors and were approximately 1-200 and 1-30 per 100 000 pancreatic cells respectively.
The Journal of Infectious Diseases | 2014
David L. Moyes; Chengguo Shen; Celia Murciano; Manohursingh Runglall; Jonathan P. Richardson; Matthew Arno; Estibaliz Aldecoa-Otalora; Julian R. Naglik
Background. The ability of epithelial cells (ECs) to discriminate between commensal and pathogenic microbes is essential for healthy living. Key to these interactions are mucosal epithelial responses to pathogen-induced damage. Methods. Using reconstituted oral epithelium, we assessed epithelial gene transcriptional responses to Candida albicans infection by microarray. Signal pathway activation was monitored by Western blotting and transcription factor enzyme-linked immunosorbent assay, and the role of these pathways in C. albicans–induced damage protection was determined using chemical inhibitors. Results. Transcript profiling demonstrated early upregulation of epithelial genes involved in immune responses. Many of these genes constituted components of signaling pathways, but only NF-κB, MAPK, and PI3K/Akt pathways were functionally activated. We demonstrate that PI3K/Akt signaling is independent of NF-κB and MAPK signaling and plays a key role in epithelial immune activation and damage protection via mammalian target of rapamycin (mTOR) activation. Conclusions. PI3K/Akt/mTOR signaling may play a critical role in protecting epithelial cells from damage during mucosal fungal infections independent of NF-κB or MAPK signaling.
Molecular Pharmacology | 2006
Chunyan Liao; Bin Hu; Matthew Arno; Barry Panaretou
Screening the Saccharomyces cerevisiae homozygous diploid deletion library against a sublethal concentration of cisplatin revealed 76 strains sensitive to the drug. As expected, the largest category of deletions, representing 40% of the sensitive strains, was composed of strains lacking genes involved in DNA replication and damage repair. Deletions lacking function of the highly conserved vacuolar H+ translocating ATPase (V-ATPase) composed the category representing the second largest number of sensitive strains. The effect on cell death exhibited by V-ATPase mutants was found to be a general response to various DNA damaging agents as opposed to being specific to cisplatin, as evidenced by sensitivity of the mutants to hydroxyurea (a DNA-alkylating agent) and UV irradiation. Loss of V-ATPase does not affect DNA repair, because double mutants defective for V-ATPase function and DNA repair pathways were more sensitive to cisplatin than the single mutants. V-ATPase mutants are more prone to DNA damage than wild-type cells, indicated by enhanced activation of the DNA damage checkpoint. Vacuole function per se is not cisplatin-sensitive, because vacuolar morphology and vacuolar acidification were unaffected by cisplatin in wild-type cells. V-ATPase also controls cytoplasmic pH, so the enhanced sensitivity to DNA damage may be associated with the drop in pHi associated with V-ATPase mutants. The increased loss in cell viability induced by cisplatin at lower pH in V-ATPase mutants supports this hypothesis. The loss in viability seen in wild-type cells under the same conditions was far less dramatic.
Mechanisms of Development | 2006
Alexander Alexandrovich; Matthew Arno; Roger Patient; Ajay M. Shah; John Pizzey; Alison C. Brewer
The GATA4, 5 and 6 subfamily of transcription factors are potent transactivators of transcription expressed within the precardiac mesoderm. However, little is known of the immediate downstream targets of GATA-factor regulation during the earliest stages of cardiogenesis. Using the P19-CL6 embryonal carcinoma (EC) cell line as an in vitro model of cardiogenesis, we show that GATA6 is the most abundantly expressed of the GATA factors in presumptive cardiac cells. Consequently, we performed a microarray screen comparing mRNA from control EC cells, early in the cardiac differentiation pathway, with those in which GATA6 had been overexpressed. These studies identified 103 genes whose expression changed significantly and this was verified in a representative array of these genes by real-time RT-PCR. We show that early cardiac expression of one of these genes, Wnt2, mirrors that of GATA6 in vitro and in vivo. In addition, its upregulation by GATA6 in differentiating EC cells is mediated by the direct binding of GATA-factor(s) to the cognate Wnt2 promoter, suggesting Wnt2 is an immediate downstream target of GATA-factor regulation during early cardiogenesis.