Network


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

Hotspot


Dive into the research topics where David Bernardo is active.

Publication


Featured researches published by David Bernardo.


Mediators of Inflammation | 2009

High Levels of Proinflammatory Cytokines, but Not Markers of Tissue Injury, in Unaffected Intestinal Areas from Patients with IBD

Alberto J. Leon; Emma Gómez; José Antonio Garrote; David Bernardo; Asterio Barrera; Jose L. Marcos; Luis Fernández-Salazar; Benito Velayos; Alfredo Blanco-Quirós; Eduardo Arranz

Intestinal alterations in IBD are triggered and maintained by an overexpression of proinflammatory cytokines. Additionally, increased immune activation has been found in the adjacent intestinal areas without displaying any apparent histological alterations, however, the regulatory environment is not well established. Biopsy specimens from patients with ulcerative colitis (UC) and Crohns disease (CD), from both affected and unaffected areas, and also from a group of colonic biopsies from healthy controls, were included in our study. Cytokines and markers of mucosal damage were analyzed by real-time PCR, and some of the results confirmed by western-blot and ELISA. Levels of IFNγ, TNFα, IL-6, IL-15, IL-18, and IL-23 were increased (above healthy controls) in both affected and unaffected areas from IBD. IL-1β, IL-6, IL-12, and IL-27 were higher in affected areas compared to unaffected ones in UC but not CD. In general, a correlation was observed between mRNA levels of these cytokines and both iNOS and Granzyme B. SOCS-2 and SOCS-3 were also increased in the affected areas. In conclusion, the unaffected areas from IBD show increased levels of a restricted set of cytokines that may exert immune activating roles in these areas without being able to trigger tissue damage.


Ejso | 2015

Is right-sided colon cancer different to left-sided colorectal cancer? – A systematic review

Gui Han Lee; George Malietzis; Alan Askari; David Bernardo; Hafid O. Al-Hassi; Susan K. Clark

Colorectal cancer (CRC) exhibits differences in incidence, pathogenesis, molecular pathways and outcome depending on the location of the tumor. This review focuses on the latest developments in epidemiological and scientific studies, which have enhanced our understanding on the underlying genetic and immunological differences between the proximal (right-sided) colon and the distal (left-sided) colorectum. The different ways in which environmental risk factors influence the pathogenesis of CRC depending on its location and the variations in surgical and oncological outcomes are also discussed in this review. In the current era of personalized medicine, we aim to reiterate the importance of tumor location in management of CRC and the implication on future clinical and scientific research.


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.


Journal of Pediatric Gastroenterology and Nutrition | 2008

Celiac disease pathogenesis: the proinflammatory cytokine network.

José Antonio Garrote; Emma Gómez-González; David Bernardo; Eduardo Arranz; Fernando G. Chirdo

In susceptible individuals, the adaptive response, mediated by the activation of antigen-specific T lymphocytes, drives a proinflammatory response, which ends in an immune-mediated enteropathy characterized by villous atrophy, crypt hyperplasia, and recruitment of intraepithelial lymphocytes. In addition, some gluten peptides are able to induce an innate immune response in intestinal mucosa. The molecular mechanisms and the cells involved in the initial stages of the gluten–intestinal mucosa interaction are poorly understood to date. There is evidence of a direct toxic effect of gluten peptides in several biological models. However, the failure to control the inflammatory response may be one of the factors underlying gluten intolerance in these individuals. The cytokine network involved in celiac disease is characterized by abundant interferon-γ in the intestinal mucosa. In addition, the production of interleukin (IL)-15, IL-18, and IL-21 is linked to gluten intake, which can drive the inflammatory response probably sustained by IL-18, IL-21, and perhaps IL-27 through STAT1 and STAT5 pathways, whereas neither IL-12 nor IL-23 plays a significant role in pathogenic mechanisms. Herein we describe the involvement of these activation pathways in the context of the pathogenesis of celiac disease.


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.


Clinical and Experimental Immunology | 2008

Higher constitutive IL15Rα expression and lower IL-15 response threshold in coeliac disease patients

David Bernardo; José Antonio Garrote; Yessica Lorena Allegretti; A.J. León; E. Gómez; J. F. Bermejo-Martin; C. Calvo; S. Riestra; Luis Fernández-Salazar; A. Blanco-Quirós; F. Chirdo; Eduardo Arranz

The IL‐15 triggering effect of gliadin is not exclusive to coeliac disease (CD) patients, whereas the secondary response is CD specific. We have studied the expression of the IL‐15 receptor, and the IL‐15 response upon stimulation, in non‐CD and CD patients, and the possible existence of a lower immunological threshold in the latter. Forty‐two CD patients (20 on a gluten‐containing diet, GCD, and 22 on gluten‐free diet, GFD) and 24 non‐CD healthy individuals were studied. IL15Rα mRNA expression, and tissue characterization, were assayed in the duodenum. Biopsies from six CD patients on GFD and 10 non‐CD individuals were studied in vitro using organ culture in basal conditions, as well as after IL‐15 stimulation discarding basal IL‐15 production. Secretion of immune mediators was measured in the culture supernatants. IL15Rα mRNA expression was increased in CD patients, as compared with non‐CD controls (on GFD P = 0·0334, on GCD P = 0·0062, respectively), and confirmed also by immunofluorescence. No differences were found between CD patients on GFD and on GCD. After in vitro IL‐15 stimulation, IL15Rα expression was only triggered in non‐CD controls (P = 0·0313), though it remained increased in CD patients. Moreover, IL‐15 induced a more intense immunological response in CD patients after triggering the production of both nitrites and IFNγ (P = 0·0313, P = 0·0313, respectively). Gliadin‐induced IL15 has a lower response threshold in CD patients, leading to the production of other immune mediators and the development of the intestinal lesion, and thus magnifying its effects within the CD intestine.


Gut | 2007

Is gliadin really safe for non-coeliac individuals? Production of interleukin 15 in biopsy culture from non-coeliac individuals challenged with gliadin peptides

David Bernardo; José Antonio Garrote; Luis Fernández-Salazar; S Riestra; Eduardo Arranz

Nowadays it is assumed that an innate immunity to gluten plays a key role in the development of coeliac disease (CD).1 This innate response, mediated by interleukin (IL) 15 and elicited by “toxic peptides”, like the 19-mer, through a DQ2-independent mechanism, induces epithelial stress and reprogrammes intraepithelial lymphocytes into natural killer (NK)-like cells2 leading to enterocyte apoptosis and an increase in epithelium permeability. Thus, immunodominant peptides, like the 33-mer, can reach the lamina propria to trigger adaptive immunity. However, although an innate specific response in CD has been reported,3 no differential factors between patients with and without CD have been described controlling the innate immune response. Thus, since the toxic 19-mer elicits its harmful effect through a DQ2-independent mechanism, we hypothesise that the innate response is common in patients with and without CD, whereas the adaptive response is exclusive of susceptible patients with CD. To test the hypothesis, biopsy cultures were taken from at least three patients with CD who are on a gluten-free diet (GFD) and three patients without …


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.

Collaboration


Dive into the David Bernardo's collaboration.

Top Co-Authors

Avatar
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

José Antonio Garrote

Spanish National Research Council

View shared research outputs
Top Co-Authors

Avatar

Eduardo Arranz

University of Valladolid

View shared research outputs
Top Co-Authors

Avatar

J. Landy

Imperial College London

View shared research outputs
Top Co-Authors

Avatar

Gui Han Lee

Imperial College London

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge