Network


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

Hotspot


Dive into the research topics where Elizabeth R. Mann is active.

Publication


Featured researches published by Elizabeth R. Mann.


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.


Inflammatory Bowel Diseases | 2010

Homing of immune cells: role in homeostasis and intestinal inflammation.

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

Abstract: Rather like a satellite navigation system directing a vehicle to a particular destination defined by post‐code, immune cells have homing molecules or “immune post‐codes” enabling them to be recruited to specific organs, such as the intestine or skin. An efficient system would be designed such that the site of entry of an antigen influences the homing of effector T cells back to the appropriate organ. For example, to mount an immune response against an intestinal pathogen, T cells with a propensity to home to the gut to clear the infection would be induced. In health, there is such a sophisticated and finely tuned system in operation, enabling an appropriate balance of immune activity in different anatomical compartments. In disease states such as inflammatory bowel disease (IBD), which is characterized by intestinal inflammation and often an inflammatory process involving other organs such as skin, joints, liver, and eye, there is accumulating evidence that there is malfunction of this immune cell trafficking system. The clinical importance of dysregulated immune cell trafficking in IBD is reflected in recently proven efficacious therapies that target trafficking pathways such as natalizumab, an &agr;4 integrin antibody, and Traficet‐EN, a chemokine receptor‐9 (CCR9) antagonist. Here we review the mechanisms involved in the homing of immune cells to different tissues, in particular the intestine, and focus on alterations in immune cell homing pathways in IBD. Unraveling the mechanisms underlying the immune post‐code system would assist in achieving the goal of tissue‐specific immunotherapy. (Inflamm Bowel Dis 2010)


PLOS ONE | 2012

Microbiota/Host Crosstalk Biomarkers: Regulatory Response of Human Intestinal Dendritic Cells Exposed to Lactobacillus Extracellular Encrypted Peptide

David Bernardo; Borja Sánchez; Hafid O. Al-Hassi; Elizabeth R. Mann; Maria C. Urdaci; Stella C. Knight; Abelardo Margolles

The human gastrointestinal tract is exposed to a huge variety of microorganisms, either commensal or pathogenic; at this site, a balance between immunity and immune tolerance is required. Intestinal dendritic cells (DCs) control the mechanisms of immune response/tolerance in the gut. In this paper we have identified a peptide (STp) secreted by Lactobacillus plantarum, characterized by the abundance of serine and threonine residues within its sequence. STp is encoded in one of the main extracellular proteins produced by such species, which includes some probiotic strains, and lacks cleavage sites for the major intestinal proteases. When studied in vitro, STp expanded the ongoing production of regulatory IL-10 in human intestinal DCs from healthy controls. STp-primed DC induced an immunoregulatory cytokine profile and skin-homing profile on stimulated T-cells. Our data suggest that some of the molecular dialogue between intestinal bacteria and DCs may be mediated by immunomodulatory peptides, encoded in larger extracellular proteins, secreted by commensal bacteria. These peptides may be used for the development of nutraceutical products for patients with IBD. In addition, this kind of peptides seem to be absent in the gut of inflammatory bowel disease patients, suggesting a potential role as biomarker of gut homeostasis.


World Journal of Gastroenterology | 2014

Intestinal antigen-presenting cells in mucosal immune homeostasis: crosstalk between dendritic cells, macrophages and B-cells.

Elizabeth R. Mann; Xuhang Li

The intestinal immune system maintains a delicate balance between immunogenicity against invading pathogens and tolerance of the commensal microbiota. Inflammatory bowel disease (IBD) involves a breakdown in tolerance towards the microbiota. Dendritic cells (DC), macrophages (MΦ) and B-cells are known as professional antigen-presenting cells (APC) due to their specialization in presenting processed antigen to T-cells, and in turn shaping types of T-cell responses generated. Intestinal DC are migratory cells, unique in their ability to generate primary T-cell responses in mesenteric lymph nodes or Peyers patches, whilst MΦ and B-cells contribute to polarization and differentiation of secondary T-cell responses in the gut lamina propria. The antigen-sampling function of gut DC and MΦ enables them to sample bacterial antigens from the gut lumen to determine types of T-cell responses generated. The primary function of intestinal B-cells involves their secretion of large amounts of immunoglobulin A, which in turn contributes to epithelial barrier function and limits immune responses towards to microbiota. Here, we review the role of all three types of APC in intestinal immunity, both in the steady state and in inflammation, and how these cells interact with one another, as well as with the intestinal microenvironment, to shape mucosal immune responses. We describe mechanisms of maintaining intestinal immune tolerance in the steady state but also inappropriate responses of APC to components of the gut microbiota that contribute to pathology in IBD.


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.


Inflammatory Bowel Diseases | 2012

Human gut‐specific homeostatic dendritic cells are generated from blood precursors by the gut microenvironment

Elizabeth R. Mann; David Bernardo; Hafid O. Al-Hassi; Nicholas R. English; Susan K. Clark; Neil E. McCarthy; Andrew N. Milestone; Stella A. Cochrane; Ailsa Hart; Andrew J. Stagg; Stella C. Knight

Background: Dendritic cells (DC) dictate not only the type of T‐cell immunity, but also homing patterns of T cells in mice. In humans, we characterized normal human gut DC and tested whether gut‐specific homeostatic DC could be generated from blood precursors by factors in the gut microenvironment. Methods: We characterized the phenotype and function of healthy human gut DC compared with blood and skin DC, and studied whether conditioning of blood DC in the presence of colonic biopsy supernatants (Bx‐SN) induced gut‐like phenotype and functions. Results: Blood DC mostly expressed both gut and skin homing markers, indicating potential to migrate to both major immune surface organs, and induced multi‐homing T cells. However, DC within gut or skin did not demonstrate this multi‐homing phenotype, were tissue‐specific, and induced tissue‐specific T cells. Human gut DC were less stimulatory for allogeneic T cells than their dermal and blood counterparts. Human blood DC cultured in vitro lost homing marker expression. Conditioning of human enriched blood DC with colonic Bx‐SN from healthy controls induced a gut‐homing phenotype and a homeostatic profile. Moreover, Bx‐SN‐conditioned DC demonstrated a restricted T‐cell stimulatory capacity and preferentially induced gut‐specific T cells. Retinoic acid and transforming growth factor beta (TGF‐&bgr;) mediated the acquisition of the gut‐homing and homeostatic properties, respectively, induced by colonic Bx‐SN on blood enriched DC. Conclusions: Tissue‐specific factors manipulate immunity via modulating characteristics of DC and may provide tools to generate tissue‐specific immunotherapy. (Inflamm Bowel Dis 2011;)


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.


Mucosal Immunology | 2013

A mechanistic role for leptin in human dendritic cell migration: differences between ileum and colon in health and Crohn's disease

Hafid O. Al-Hassi; David Bernardo; Aravinth U. Murugananthan; Elizabeth R. Mann; Nicholas R. English; Allison V Jones; Michael A. Kamm; N Arebi; Ailsa Hart; Alexandra I. F. Blakemore; Andrew J. Stagg; Sarah Knight

Dendritic cells (DC) migrate to lymph nodes on expression of C-C motif chemokine receptor 7 (CCR7) and control immune activity. Leptin, an immunomodulatory adipokine, functions via leptin receptors, signaling via the long isoform of receptor, LepRb. Leptin promotes DC maturation and increases CCR7 expression on blood DC. Increased mesenteric fat and leptin occur early in Crohn’s disease (CD), suggesting leptin-mediated change in intestinal CCR7 expression on DC as a pro-inflammatory mechanism. We have demonstrated CCR7 expression and capacity to migrate to its ligand macrophage inflammatory protein 3β in normal human ileal DC but not colonic or blood DC. In CD, functional CCR7 was expressed on DC from all sites. Only DC populations containing CCR7-expressing cells produced LepRb; in vitro exposure to leptin also increased expression of functional CCR7 in intestinal DC in a dose-dependent manner. In conclusion, leptin may regulate DC migration from gut, in homeostatic and inflammatory conditions, providing a link between mesenteric obesity and inflammation.


Scientific Reports | 2015

Variable alterations of the microbiota, without metabolic or immunological change, following faecal microbiota transplantation in patients with chronic pouchitis

J. Landy; Alan W. Walker; Junhua Li; Hafid O. Al-Hassi; E. Ronde; N English; Elizabeth R. Mann; David Bernardo; Simon D. McLaughlin; Julian Parkhill; Paul J. Ciclitira; Susan K. Clark; Stella C. Knight; Ailsa Hart

Faecal microbiota transplantation (FMT) is effective in the treatment of Clostridium difficile infection, where efficacy correlates with changes in microbiota diversity and composition. The effects of FMT on recipient microbiota in inflammatory bowel diseases (IBD) remain unclear. We assessed the effects of FMT on microbiota composition and function, mucosal immune response, and clinical outcome in patients with chronic pouchitis. Eight patients with chronic pouchitis (current PDAI ≥7) were treated with FMT via nasogastric administration. Clinical activity was assessed before and four weeks following FMT. Faecal coliform antibiotic sensitivities were analysed, and changes in pouch faecal and mucosal microbiota assessed by 16S rRNA gene pyrosequencing and 1H NMR spectroscopy. Lamina propria dendritic cell phenotype and cytokine profiles were assessed by flow cytometric analysis and multiplex assay. Following FMT, there were variable shifts in faecal and mucosal microbiota composition and, in some patients, changes in proportional abundance of species suggestive of a “healthier” pouch microbiota. However, there were no significant FMT-induced metabolic or immunological changes, or beneficial clinical response. Given the lack of clinical response following FMT via a single nasogastric administration our results suggest that FMT/bacteriotherapy for pouchitis patients requires further optimisation.

Collaboration


Dive into the Elizabeth R. Mann's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ailsa Hart

Imperial College London

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

S. Peake

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

Andrew J. Stagg

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge