Network


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

Hotspot


Dive into the research topics where S. Peake is active.

Publication


Featured researches published by S. Peake.


Immunology Letters | 2013

Intestinal dendritic cells: Their role in intestinal inflammation, manipulation by the gut microbiota and differences between mice and men

Elizabeth R. Mann; J. Landy; David Bernardo; S. Peake; Ailsa Hart; Hafid O. Al-Hassi; Stella C. Knight

The intestinal immune system maintains a delicate balance between immunogenicity against invading pathogens and tolerance of the commensal microbiota and food antigens. Dendritic cells (DC) generate primary T-cell responses, and determine whether these responses are immunogenic or tolerogenic. The regulatory role of DC is of particular importance in the gut due to the high antigenic load. Intestinal DC act as sentinels, sampling potentially pathogenic antigens but also harmless antigens including the commensal microbiota. Following antigen acquisition, intestinal DC migrate to secondary lymphoid organs to activate naive T-cells. DC also imprint specific homing properties on T-cells that they stimulate; gut DC specifically induce gut-homing properties on T-cells upon activation, enabling T-cell migration back to intestinal sites. Data regarding properties on gut DC in humans is scarce, although evidence now supports the role of DC as important players in intestinal immunity in humans. Here, we review the role of intestinal DC in shaping mucosal immune responses and directing tissue-specific T-cell responses, with a special focus on the importance of distinguishing DC subsets from macrophages at intestinal sites. We compare and contrast human DC with their murine counterparts, and discuss the ability of the gut microbiota to shape intestinal DC function, and how this may be dysregulated in inflammatory bowel disease (IBD). Lastly, we describe recent advances in the study of probiotics on intestinal DC function, including the use of soluble secreted bacterial products.


Gut | 2011

H1N1 vaccines in a large observational cohort of patients with inflammatory bowel disease treated with immunomodulators and biological therapy

Jean-François Rahier; Pavol Papay; Julia Salleron; Shaji Sebastian; Manuela Marzo; Laurent Peyrin-Biroulet; Valle García-Sánchez; Walter Fries; Dirk P. van Asseldonk; Klaudia Farkas; Nanne de Boer; Taina Sipponen; Pierre Ellul; Edouard Louis; S. Peake; Uri Kopylov; Jochen Maul; Badira Makhoul; Gionata Fiorino; Yazdan Yazdanpanah; Maria Chaparro

Background Safety data are lacking on influenza vaccination in general and on A (H1N1)v vaccination in particular in patients with inflammatory bowel disease (IBD) receiving immmunomodulators and/or biological therapy. Aims and methods The authors conducted a multicentre observational cohort study to evaluate symptoms associated with influenza H1N1 adjuvanted (Pandemrix, Focetria, FluvalP) and non-adjuvanted (Celvapan) vaccines and to assess the risk of flare of IBD after vaccination. Patients with stable IBD treated with immunomodulators and/or biological therapy were recruited from November 2009 until March 2010 in 12 European countries. Harvey–Bradshaw Index and Partial Mayo Score were used to assess disease activity before and 4 weeks after vaccination in Crohns disease (CD) and ulcerative colitis (UC). Vaccination-related events up to 7 days after vaccination were recorded. Results Of 575 patients enrolled (407 CD, 159 UC and nine indeterminate colitis; 53.9% female; mean age 40.3 years, SD 13.9), local and systemic symptoms were reported by 34.6% and 15.5% of patients, respectively. The most common local and systemic reactions were pain in 32.8% and fatigue in 6.1% of subjects. Local symptoms were more common with adjuvanted (39.3%) than non-adjuvanted (3.9%) vaccines (p<0.0001), whereas rates of systemic symptoms were similar with both types (15.0% vs 18.4%, p=0.44). Among the adjuvanted group, Pandemrix more often induced local reactions than FluvalP and Focetria (51.2% vs 27.6% and 15.4%, p<0.0001). Solicited adverse events were not associated with any patient characteristics, specific immunomodulatory treatment, or biological therapy. Four weeks after vaccination, absence of flare was observed in 377 patients with CD (96.7%) and 151 with UC (95.6%). Conclusion Influenza A (H1N1)v vaccines are well tolerated in patients with IBD. Non-adjuvanted vaccines are associated with fewer local reactions. The risk of IBD flare is probably not increased after H1N1 vaccination.


Molecular Nutrition & Food Research | 2014

Altered human gut dendritic cell properties in ulcerative colitis are reversed by Lactobacillus plantarum extracellular encrypted peptide STp

Hafid O. Al-Hassi; Elizabeth R. Mann; Borja Sánchez; Nicholas R. English; S. Peake; J. Landy; R. Man; Maria C. Urdaci; Ailsa Hart; Luis Fernández-Salazar; Gui Han Lee; José Antonio Garrote; Eduardo Arranz; Abelardo Margolles; Andrew J. Stagg; Stella C. Knight; David Bernardo

SCOPE The human/microbiota cross-talk is partially mediated by bacteria-derived peptides like Serine-Threonine peptide (STp), which is resistant to gut proteolysis, is found in the human healthy colon and induces regulatory properties on gut dendritic cells (DCs); here we characterized human gut DC in ulcerative colitis (UC) patients and studied the effect of STp on their properties. METHODS AND RESULTS Human colonic DC from healthy controls and UC patients were isolated, conditioned for 24 h +/- STp and characterized by flow cytometry, immunohistochemistry, and electron microscopy. Expression of immature DC markers DC-SIGN and ILT3, and Toll-like receptors were increased on gut UC-DC. Langerin (involved in phagocytosis), lymph node homing marker CCR7, and activation markers CD40/CD80/CD86 were decreased in UC. Gut DC had restricted stimulatory capacity for T-cells in UC. Conditioning of DC with STp in vitro reduced Toll-like receptor expression, increased CD40 and CD80 expression, and restored their stimulatory capacity. CONCLUSION Colonic DCs display an abnormal immature phenotype in UC, which was partially restored following STp treatment. Bacteria-derived metabolites, like STp, seem to have a role in gut homeostasis that is missing in UC so they might lead a new era of probiotic products setting the basis for nondrug dietary therapy in inflammatory bowel disease.


Inflammatory Bowel Diseases | 2013

Mechanisms of action of anti-tumor necrosis factor α agents in Crohn's disease.

S. Peake; David Bernardo; Elizabeth R. Mann; Hafid O. Al-Hassi; Stella C. Knight; Ailsa Hart

Abstract:Crohn’s disease (CD) is characterized by inflammation that can affect any part of the gastrointestinal tract. It is a chronic destructive condition that follows a relapsing–remitting course and can lead to disability and a poor quality of life. Lifelong pharmacotherapy with systemic immunomodulator therapies remains the cornerstone of CD management. Advances in understanding of the immunopathogenic mechanisms underlying chronic gut inflammation in CD have led to the development of effective biological therapies for patients with CD. Tumor necrosis factor &agr; (TNF-&agr;) is a potent proinflammatory cytokine that plays a pivotal role in the development of Crohn’s inflammation. Therapies designed to target this cytokine have revolutionized treatment of CD since their introduction in the late 1990s, thanks to their ability to induce and maintain remission, heal mucosa, reduce hospital admissions and surgical procedures, and restore quality of life. Despite widespread use of these therapies in CD, their precise mechanism of action remains unclear, although several different mechanisms have been proposed. This review summarizes the biology of the TNF-&agr; cytokine and the development of biological therapies targeting TNF-&agr;, and updates our current understanding of mechanisms of action of the commercially available anti-TNF-&agr; therapies used in the treatment of CD.


Gut | 2016

Compartment-specific immunity in the human gut: properties and functions of dendritic cells in the colon versus the ileum

Elizabeth R. Mann; David Bernardo; Nicholas R. English; Jon Landy; Hafid O. Al-Hassi; S. Peake; R. Man; Timothy R. Elliott; Henning Spranger; Gui Han Lee; Alyssa M. Parian; Steven R. Brant; Mark Lazarev; Ailsa Hart; Xuhang Li; Stella C. Knight

Objective Dendritic cells (DC) mediate intestinal immune tolerance. Despite striking differences between the colon and the ileum both in function and bacterial load, few studies distinguish between properties of immune cells in these compartments. Furthermore, information of gut DC in humans is scarce. We aimed to characterise human colonic versus ileal DC. Design Human DC from paired colonic and ileal samples were characterised by flow cytometry, electron microscopy or used to stimulate T cell responses in a mixed leucocyte reaction. Results A lower proportion of colonic DC produced pro-inflammatory cytokines (tumour necrosis factor-α and interleukin (IL)-1β) compared with their ileal counterparts and exhibited an enhanced ability to generate CD4+FoxP3+IL-10+ (regulatory) T cells. There were enhanced proportions of CD103+Sirpα− DC in the colon, with increased proportions of CD103+Sirpα+ DC in the ileum. A greater proportion of colonic DC subsets analysed expressed the lymph-node-homing marker CCR7, alongside enhanced endocytic capacity, which was most striking in CD103+Sirpα+ DC. Expression of the inhibitory receptor ILT3 was enhanced on colonic DC. Interestingly, endocytic capacity was associated with CD103+ DC, in particular CD103+Sirpα+ DC. However, expression of ILT3 was associated with CD103− DC. Colonic and ileal DC differentially expressed skin-homing marker CCR4 and small-bowel-homing marker CCR9, respectively, and this corresponded to their ability to imprint these homing markers on T cells. Conclusions The regulatory properties of colonic DC may represent an evolutionary adaptation to the greater bacterial load in the colon. The colon and the ileum should be regarded as separate entities, each comprising DC with distinct roles in mucosal immunity and imprinting.


Clinical and Experimental Immunology | 2012

Skin‐ and gut‐homing molecules on human circulating γδ T cells and their dysregulation in inflammatory bowel disease

Elizabeth R. Mann; Neil E. McCarthy; S. Peake; Andrew N. Milestone; Hafid O. Al-Hassi; David Bernardo; Cheng T. Tee; J. Landy; M. C. Pitcher; Stella A. Cochrane; Ailsa Hart; Andrew J. Stagg; Stella C. Knight

Changes in phenotype and function of γδ T cells have been reported in inflammatory bowel disease (IBD), including Crohns disease (CD) and ulcerative colitis (UC). Dysregulation of lymphocyte migration plays a key role in IBD pathogenesis; however, data on migratory properties of γδ T cells are scarce. Human circulating γδ T cells from healthy controls (n = 27), patients with active CD (n = 15), active UC (n = 14) or cutaneous manifestations of IBD (n = 2) were characterized by flow cytometry. Circulating γδ T cells in healthy controls were CD3hi and expressed CD45RO. They expressed gut‐homing molecule β7 but not gut‐homing molecule corresponding chemokine receptors (CCR)9, or skin‐homing molecules cutaneous lymphocyte‐associated antigen (CLA) and CCR4, despite conventional T cells containing populations expressing these molecules. CCR9 expression was increased on γδ T cells in CD and UC, while skin‐homing CLA was expressed aberrantly on γδ T cells in patients with cutaneous manifestations of IBD. Lower levels of CD3 expression were found on γδ T cells in CD but not in UC, and a lower proportion of γδ T cells expressed CD45RO in CD and UC. Enhanced expression of gut‐homing molecules on circulating γδ T cells in IBD and skin‐homing molecules in cutaneous manifestations of IBD may be of clinical relevance.


Cellular and molecular gastroenterology and hepatology | 2016

Chemokine (C-C Motif) Receptor 2 Mediates Dendritic Cell Recruitment to the Human Colon but Is Not Responsible for Differences Observed in Dendritic Cell Subsets, Phenotype, and Function Between the Proximal and Distal Colon

David Bernardo; Lydia R. Durant; Elizabeth R. Mann; Elizabeth Bassity; Enrique Montalvillo; R. Man; Rakesh Vora; D Reddi; Fahri Bayiroglu; Luis Fernández-Salazar; N English; S. Peake; Jon Landy; Gui H. Lee; George Malietzis; Yi Harn Siaw; Aravinth U. Murugananthan; Phil Hendy; Eva Sánchez-Recio; Robin K. S. Phillips; José Antonio Garrote; Paul Scott; Julian Parkhill; Malte Paulsen; Ailsa Hart; Hafid O. Al-Hassi; Eduardo Arranz; Alan W. Walker; Simon R. Carding; Stella C. Knight

Background & Aims Most knowledge about gastrointestinal (GI)-tract dendritic cells (DC) relies on murine studies where CD103+ DC specialize in generating immune tolerance with the functionality of CD11b+/− subsets being unclear. Information about human GI-DC is scarce, especially regarding regional specifications. Here, we characterized human DC properties throughout the human colon. Methods Paired proximal (right/ascending) and distal (left/descending) human colonic biopsies from 95 healthy subjects were taken; DC were assessed by flow cytometry and microbiota composition assessed by 16S rRNA gene sequencing. Results Colonic DC identified were myeloid (mDC, CD11c+CD123−) and further divided based on CD103 and SIRPα (human analog of murine CD11b) expression. CD103-SIRPα+ DC were the major population and with CD103+SIRPα+ DC were CD1c+ILT3+CCR2+ (although CCR2 was not expressed on all CD103+SIRPα+ DC). CD103+SIRPα- DC constituted a minor subset that were CD141+ILT3−CCR2−. Proximal colon samples had higher total DC counts and fewer CD103+SIRPα+ cells. Proximal colon DC were more mature than distal DC with higher stimulatory capacity for CD4+CD45RA+ T-cells. However, DC and DC-invoked T-cell expression of mucosal homing markers (β7, CCR9) was lower for proximal DC. CCR2 was expressed on circulating CD1c+, but not CD141+ mDC, and mediated DC recruitment by colonic culture supernatants in transwell assays. Proximal colon DC produced higher levels of cytokines. Mucosal microbiota profiling showed a lower microbiota load in the proximal colon, but with no differences in microbiota composition between compartments. Conclusions Proximal colonic DC subsets differ from those in distal colon and are more mature. Targeted immunotherapy using DC in T-cell mediated GI tract inflammation may therefore need to reflect this immune compartmentalization.


Inflammatory Bowel Diseases | 2014

Human gut dendritic cells drive aberrant gut-specific t-cell responses in ulcerative colitis, characterized by increased IL-4 production and loss of IL-22 and IFNγ.

Elizabeth R. Mann; David Bernardo; Siew C. Ng; Rachael Rigby; Hafid O. Al-Hassi; Jon Landy; S. Peake; Henning Spranger; Nicholas R. English; Linda V. Thomas; Andrew J. Stagg; Stella C. Knight; Ailsa Hart

Abstract:The pathogenesis of inflammatory bowel disease is incompletely understood but results from a dysregulated intestinal immune response to the luminal microbiota. CD4+ T cells mediate tissue injury in the inflammatory bowel disease–associated immune response. Dendritic cells (DC) generate primary T-cell responses and mediate intestinal immune tolerance to prevent overt inflammation in response to the gut microbiota. However, most information regarding function of intestinal DC has come from mouse models, and information in humans is scarce. We show here that intestinal DC subsets are skewed in ulcerative colitis (UC) in humans, with a loss of CD103+ lymph-node homing DC; this intestinal DC subset preferentially generates regulatory T cells in mice. We show infiltrates of DC negative for myeloid marker CD11c, with enhanced expression of Toll-like receptors for bacterial recognition. After mixed leukocyte reaction, DC from the inflamed UC colon had an enhanced ability to generate gut-specific CD4+ T cells with enhanced production of interleukin-4 but a loss of interferon &ggr; and interleukin-22 production. Conditioning intestinal DC with probiotic strain Lactobacillus casei Shirota in UC partially restored their normal function indicated by reduced Toll-like receptor 2/4 expression and restoration of their ability to imprint homing molecules on T cells and to generate interleukin-22 production by stimulated T cells. This study suggests that T-cell dysfunction in UC is driven by DC. T-cell responses can be manipulated indirectly through effects of bacterial conditioning on gut DC with implications for immunomodulatory effects of the commensal microbiota in vivo. Manipulation of DC to allow generation of DC-specific therapy may be beneficial in inflammatory bowel disease.


Journal of Crohns & Colitis | 2013

Oral tacrolimus as maintenance therapy for refractory ulcerative colitis—an analysis of outcomes in two London tertiary centres ☆

J. Landy; Mahmood Wahed; S. Peake; Mohammed Hussein; Siew C. Ng; James O. Lindsay; Ailsa Hart

BACKGROUND The medical management of refractory ulcerative colitis (UC) remains a significant challenge. Two randomised controlled studies have demonstrated tacrolimus therapy is effective for the induction of remission of moderate to severe UC. However, the long term outcomes of UC patients treated with tacrolimus as maintenance therapy are not certain. AIMS This study aims to assess the efficacy of tacrolimus maintenance therapy for refractory UC. METHODS A retrospective review of patients with UC treated with tacrolimus at two London tertiary centres was performed. Clinical outcomes were assessed at six months, at the end of tacrolimus treatment, or at the last follow-up for patients continuing tacrolimus treatment. Modified Truelove-Witts score (mTW) and Mayo endoscopy subscores were calculated. RESULTS 25 patients with UC, treated with oral tacrolimus between 2005 and 2011, were identified. The median duration of tacrolimus treatment was 9 months (IQR 3.7-18.2 months). The median duration of follow-up was 27 months (range 3-66 months). At six months thirteen (52%) patients had achieved and maintained clinical response and eleven (44%) were in clinical remission. The mean mTW score decreased from 10+/-0.5 before therapy, to 5.8+/-0.8 (p≤0.001 95% CI 2.7-5.8) at cessation of treatment or last follow-up. Mayo endoscopy subscore decreased from 2.6+/-0.1 to 1.2+/-0.2 (p≤0.001 mean reduction 1.4, 95% CI 0.8-1.9). Eight patients (32%) subsequently underwent a colectomy within a mean time of 17 months (range 2-45 months). CONCLUSION Tacrolimus is effective for the maintenance of refractory UC and can deliver sustained improvement in mucosal inflammation.


Gastroenterology | 2013

Tu1985 A Prospective Controlled Pilot Study of Fecal Microbiota Transplantation for Chronic Refractory Pouchitis

J. Landy; Hafid O. Al-Hassi; Elizabeth R. Mann; S. Peake; Simon D. McLaughlin; Paul J. Ciclitira; Susan K. Clark; Stella C. Knight; Ailsa Hart

Introduction Faecal microbiota transplantation (FMT) is an effective therapy for Clostridium difficile and possibly inflammatory bowel diseases (IBD). Published data of FMT for inflammatory bowel diseases are reported in case series and case reports. To our knowledge, there are no controlled studies of FMT for IBD. We aimed to conduct a prospective study of FMT for chronic refractory pouchitis. Methods Patients with clinically, endoscopically and histologically confirmed chronic refractory pouchitis; with a pouch disease activity index (PDAI) > 7 were included. Donors were screened by clinical history and serology for HAV, HBV, HCV, HEV, Treponema, HIV, HTLV I/II and stool for M, C+S, C. difficile toxin and parasites. Fresh donor stool was collected within six hours of nasogasric administration of FMT. Stool samples were also collected from patients for analysis of coliform sensitivities before and 4 weeks after FMT. PDAI and Cleveland global quality of life score (CGQoL ) were recorded prior to FMT and four weeks after FMT. Results Eight patients with chronic refractory pouchitis who had undergone restorative proctocolectomy for ulcerative colitis underwent FMT. Three patients had ESBL resistant coliforms on stool analysis prior toFMT. Two of these patients demonstrated a change to ciprofloxacin sensitive coliform following FMT. The mean PDAI prior to FMT was 12. The mean CGQoL was 0.45. At 4 weeks following FMT, no patient had achieved a clinical remission (mean PDAI 11). No improvement in CGQoL was seen (mean 0.44). Conclusion FMT via nasogastric administration was not effective in achieving clinical remission for chronic refractory pouchitis at 4 weeks after FMT. However, in two patients with ESBL resistant coliform, ciprofloxacin sensitivity was regained following FMT and these patients have subsequently been maintained on ciprofloxacin. This suggests FMT may alter the pouch microbiota. Further molecular microbiological analysis is being undertaken to determine the effect FMT had on these patients’ microbiota. In addition, further studies of FMT are required to assess the effect of different methods of FMT that may be more efficacious for this group of patients. Disclosure of Interest None Declared.

Collaboration


Dive into the S. Peake's collaboration.

Top Co-Authors

Avatar

Ailsa Hart

Imperial College London

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

J. Landy

Imperial College London

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andrew J. Stagg

Queen Mary University of London

View shared research outputs
Researchain Logo
Decentralizing Knowledge