Network


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

Hotspot


Dive into the research topics where Susanne Heck is active.

Publication


Featured researches published by Susanne Heck.


European Journal of Immunology | 2013

CD161 expression characterizes a subpopulation of human regulatory T cells that produces IL‐17 in a STAT3‐dependent manner

Behdad Afzali; Peter Mitchell; Francis C. Edozie; Giovanni A.M. Povoleri; Sophie E. Dowson; Laura Demandt; Gina J. Walter; James B. Canavan; Cristiano Scottà; Bina Menon; Prabhjoat Chana; Wafa Khamri; Shahram Kordasti; Susanne Heck; Bodo Grimbacher; Timothy Tree; Andrew P. Cope; Leonie S. Taams; Robert I. Lechler; Susan John; Giovanna Lombardi

Treg cells are critical for the prevention of autoimmune diseases and are thus prime candidates for cell‐based clinical therapy. However, human Treg cells are “plastic”, and are able to produce IL‐17 under inflammatory conditions. Here, we identify and characterize the human Treg subpopulation that can be induced to produce IL‐17 and identify its mechanisms. We confirm that a subpopulation of human Treg cells produces IL‐17 in vitro when activated in the presence of IL‐1β, but not IL‐6. “IL‐17 potential” is restricted to population III (CD4+CD25hiCD127loCD45RA−) Treg cells expressing the natural killer cell marker CD161. We show that these cells are functionally as suppressive and have similar phenotypic/molecular characteristics to other subpopulations of Treg cells and retain their suppressive function following IL‐17 induction. Importantly, we find that IL‐17 production is STAT3 dependent, with Treg cells from patients with STAT3 mutations unable to make IL‐17. Finally, we show that CD161+ population III Treg cells accumulate in inflamed joints of patients with inflammatory arthritis and are the predominant IL‐17‐producing Treg‐cell population at these sites. As IL‐17 production from this Treg‐cell subpopulation is not accompanied by a loss of regulatory function, in the context of cell therapy, exclusion of these cells from the cell product may not be necessary.


Scientific Reports | 2016

Standardizing Flow Cytometry Immunophenotyping Analysis from the Human ImmunoPhenotyping Consortium

Greg Finak; Marc Langweiler; Maria Jaimes; Mehrnoush Malek; Jafar Taghiyar; Yael Korin; Lesley Devine; Gerlinde Obermoser; Marcin L. Pekalski; Nikolas Pontikos; Alain Diaz; Susanne Heck; Federica Villanova; Nadia Terrazzini; Florian Kern; Yu Qian; Rick Stanton; Kui Wang; Aaron Brandes; John Ramey; Nima Aghaeepour; Tim R. Mosmann; Richard H. Scheuermann; Elaine F. Reed; Karolina Palucka; Virginia Pascual; Bonnie B. Blomberg; Frank O. Nestle; Robert B. Nussenblatt; Ryan R. Brinkman

Standardization of immunophenotyping requires careful attention to reagents, sample handling, instrument setup, and data analysis, and is essential for successful cross-study and cross-center comparison of data. Experts developed five standardized, eight-color panels for identification of major immune cell subsets in peripheral blood. These were produced as pre-configured, lyophilized, reagents in 96-well plates. We present the results of a coordinated analysis of samples across nine laboratories using these panels with standardized operating procedures (SOPs). Manual gating was performed by each site and by a central site. Automated gating algorithms were developed and tested by the FlowCAP consortium. Centralized manual gating can reduce cross-center variability, and we sought to determine whether automated methods could streamline and standardize the analysis. Within-site variability was low in all experiments, but cross-site variability was lower when central analysis was performed in comparison with site-specific analysis. It was also lower for clearly defined cell subsets than those based on dim markers and for rare populations. Automated gating was able to match the performance of central manual analysis for all tested panels, exhibiting little to no bias and comparable variability. Standardized staining, data collection, and automated gating can increase power, reduce variability, and streamline analysis for immunophenotyping.


Journal of Virology | 2006

Antigenic and Immunogenic Characterization of Recombinant Baculovirus-Expressed Severe Acute Respiratory Syndrome Coronavirus Spike Protein: Implication for Vaccine Design

Yuxian He; Jingjing Li; Susanne Heck; Sara Lustigman; Shibo Jiang

ABSTRACT The spike (S) glycoprotein of severe acute respiratory syndrome coronavirus (SARS-CoV) mediates the receptor interaction and immune recognition and is considered a major target for vaccine design. However, its antigenic and immunogenic properties remain to be elucidated. In this study, we immunized mice with full-length S protein (FL-S) or its extracellular domain (EC-S) expressed by recombinant baculoviruses in insect cells. We found that the immunized mice developed high titers of anti-S antibodies with potent neutralizing activities against SARS pseudoviruses constructed with the S proteins of Tor2, GD03T13, and SZ3, the representative strains of 2002 to 2003 and 2003 to 2004 human SARS-CoV and palm civet SARS-CoV, respectively. These data suggest that the recombinant baculovirus-expressed S protein vaccines possess excellent immunogenicity, thereby inducing highly potent neutralizing responses against human and animal SARS-CoV variants. The antigenic structure of the S protein was characterized by a panel of 38 monoclonal antibodies (MAbs) isolated from the immunized mice. The epitopes of most anti-S MAbs (32 of 38) were localized within the S1 domain, and those of the remaining 6 MAbs were mapped to the S2 domain. Among the anti-S1 MAbs, 17 MAbs targeted the N-terminal region (amino acids [aa] 12 to 327), 9 MAbs recognized the receptor-binding domain (RBD; aa 318 to 510), and 6 MAbs reacted with the C-terminal region of S1 domain that contains the major immunodominant site (aa 528 to 635). Strikingly, all of the RBD-specific MAbs had potent neutralizing activity, 6 of which efficiently blocked the receptor binding, confirming that the RBD contains the main neutralizing epitopes and that blockage of the receptor association is the major mechanism of SARS-CoV neutralization. Five MAbs specific for the S1 N-terminal region exhibited moderate neutralizing activity, but none of the MAbs reacting with the S2 domain and the major immunodominant site in S1 showed neutralizing activity. All of the neutralizing MAbs recognize conformational epitopes. These data provide important information for understanding the antigenicity and immunogenicity of S protein and for designing SARS vaccines. This panel of anti-S MAbs can be used as tools for studying the structure and function of the SARS-CoV S protein.


Diabetes | 2015

β-cell-specific CD8 T cell phenotype in type 1 diabetes reflects chronic autoantigen exposure

Ania Skowera; Kristin Ladell; James Edward McLaren; Garry Dolton; Katherine K. Matthews; Emma Gostick; Deborah Kronenberg-Versteeg; Martin Eichmann; Robin R. Knight; Susanne Heck; Jake Powrie; Polly J. Bingley; Colin Mark Dayan; John J. Miles; Andrew K. Sewell; David A. Price; Mark Peakman

Autoreactive CD8 T cells play a central role in the destruction of pancreatic islet β-cells that leads to type 1 diabetes, yet the key features of this immune-mediated process remain poorly defined. In this study, we combined high-definition polychromatic flow cytometry with ultrasensitive peptide–human leukocyte antigen class I tetramer staining to quantify and characterize β-cell–specific CD8 T cell populations in patients with recent-onset type 1 diabetes and healthy control subjects. Remarkably, we found that β-cell–specific CD8 T cell frequencies in peripheral blood were similar between subject groups. In contrast to healthy control subjects, however, patients with newly diagnosed type 1 diabetes displayed hallmarks of antigen-driven expansion uniquely within the β-cell–specific CD8 T cell compartment. Molecular analysis of selected β-cell–specific CD8 T cell populations further revealed highly skewed oligoclonal T cell receptor repertoires comprising exclusively private clonotypes. Collectively, these data identify novel and distinctive features of disease-relevant CD8 T cells that inform the immunopathogenesis of type 1 diabetes.


Journal of Immunology | 2015

Phenotypic Complexity of the Human Regulatory T Cell Compartment Revealed by Mass Cytometry

Gavin M. Mason; Katie Lowe; Rossella Melchiotti; Richard Ellis; Emanuele de Rinaldis; Mark Peakman; Susanne Heck; Giovanna Lombardi; Timothy Tree

Regulatory T cells (Tregs) are an essential component of the cellular immune response, occupying a key role in maintaining immunological tolerance and present an attractive therapeutic target in a range of immunopathologies. Comprehensive analysis of the human Treg compartment has been restricted due to technical limitations. The advent of mass cytometry enables simultaneous assessment of vastly increased phenotypic parameters at single-cell resolution. In this study, we used mass cytometry to examine the complexity of human Tregs using an extensive panel of surface markers associated with Treg function and phenotype. We applied unsupervised clustering analysis, revealing 22 distinct subpopulations of Tregs, representing previously identified and novel subpopulations. Our data represent the most in-depth phenotypic description of the human Treg compartment at single-cell resolution and show a hitherto unrecognized degree of phenotypic complexity among cells of the regulatory lineage.


Blood | 2016

Deep phenotyping of Tregs identifies an immune signature for idiopathic aplastic anemia and predicts response to treatment

Shahram Kordasti; Benedetta Costantini; Thomas Seidl; Pilar Perez Abellan; Marc Martinez Llordella; Donal McLornan; Kirsten E. Diggins; Austin Kulasekararaj; Cinzia Benfatto; Xingmin Feng; Alexander E. Smith; Syed A. Mian; Rossella Melchiotti; Emanuele de Rinaldis; Richard Ellis; Nedyalko Petrov; Giovanni A.M. Povoleri; Sun Sook Chung; N. Shaun B. Thomas; Farzin Farzaneh; Jonathan M. Irish; Susanne Heck; Neal S. Young; Judith Marsh; Ghulam J. Mufti

Idiopathic aplastic anemia (AA) is an immune-mediated and serious form of bone marrow failure. Akin to other autoimmune diseases, we have previously shown that in AA regulatory T cells (Tregs) are reduced in number and function. The aim of this study was to further characterize Treg subpopulations in AA and investigate the potential correlation between specific Treg subsets and response to immunosuppressive therapy (IST) as well as their in vitro expandability for potential clinical use. Using mass cytometry and an unbiased multidimensional analytical approach, we identified 2 specific human Treg subpopulations (Treg A and Treg B) with distinct phenotypes, gene expression, expandability, and function. Treg B predominates in IST responder patients, has a memory/activated phenotype (with higher expression of CD95, CCR4, and CD45RO within FOXP3(hi), CD127(lo) Tregs), expresses the interleukin-2 (IL-2)/STAT5 pathway and cell-cycle commitment genes. Furthermore, in vitro-expanded Tregs become functional and take on the characteristics of Treg B. Collectively, this study identifies human Treg subpopulations that can be used as predictive biomarkers for response to IST in AA and potentially other autoimmune diseases. We also show that Tregs from AA patients are IL-2-sensitive and expandable in vitro, suggesting novel therapeutic approaches such as low-dose IL-2 therapy and/or expanded autologous Tregs and meriting further exploration.


Science Translational Medicine | 2017

Metabolic and immune effects of immunotherapy with proinsulin peptide in human new-onset type 1 diabetes*

Mohammad Alhadj Ali; Yuk-Fun Liu; Sefina Arif; Danijela Tatovic; Hina Shariff; Vivienne Gibson; Norkhairin Yusuf; Roman Baptista; Martin Eichmann; Nedyalko Petrov; Susanne Heck; Jennie H. M. Yang; Timothy Tree; Irma Pujol-Autonell; Lorraine Yeo; Lucas R. Baumard; Rachel Stenson; Alex Howell; Alison Clark; Zoe Boult; Jake Powrie; Laura Adams; Florence Susan Wong; Stephen Luzio; Gareth Dunseath; Kate Green; Alison O'Keefe; Graham Bayly; Natasha Thorogood; Rob C Andrews

Immunotherapy using peptide from the self-antigen proinsulin is safe in type 1 diabetes and is associated with changes in immune regulation. Peptide therapy prompts responses in diabetes Immunotherapy using peptides has been successful for some patients with allergies, but has not yet been deployed in autoimmune diseases, which may involve greater safety risks. Alhadj Ali et al. designed a placebo-controlled trial to determine whether a proinsulin peptide could safely elicit immune and metabolic responses in people recently diagnosed with type 1 diabetes without accelerating disease. This small trial showed that treatment seemed to modify T cell responses and did not interfere with residual β cell function. In contrast to subjects in the placebo arm, treated subjects did not need to increase their insulin use. These encouraging results support a larger trial to investigate efficacy of the peptide therapy for treating disease. Immunotherapy using short immunogenic peptides of disease-related autoantigens restores immune tolerance in preclinical disease models. We studied safety and mechanistic effects of injecting human leukocyte antigen–DR4(DRB1*0401)–restricted immunodominant proinsulin peptide intradermally every 2 or 4 weeks for 6 months in newly diagnosed type 1 diabetes patients. Treatment was well tolerated with no systemic or local hypersensitivity. Placebo subjects showed a significant decline in stimulated C-peptide (measuring insulin reserve) at 3, 6, 9, and 12 months versus baseline, whereas no significant change was seen in the 4-weekly peptide group at these time points or the 2-weekly group at 3, 6, and 9 months. The placebo group’s daily insulin use increased by 50% over 12 months but remained unchanged in the intervention groups. C-peptide retention in treated subjects was associated with proinsulin-stimulated interleukin-10 production, increased FoxP3 expression by regulatory T cells, low baseline levels of activated β cell–specific CD8 T cells, and favorable β cell stress markers (proinsulin/C-peptide ratio). Thus, proinsulin peptide immunotherapy is safe, does not accelerate decline in β cell function, and is associated with antigen-specific and nonspecific immune modulation.


Journal of Immunology | 2009

Recombinant Ov-ASP-1, a Th1-Biased Protein Adjuvant Derived from the Helminth Onchocerca volvulus, Can Directly Bind and Activate Antigen-Presenting Cells

Yuxian He; Sophie J. Barker; Angus J. MacDonald; Yu Yu; Long Cao; Jingjing Li; Ranjit Parhar; Susanne Heck; Susanne Hartmann; Douglas T. Golenbock; Shibo Jiang; Nathan A. Libri; Amanda E. Semper; William Rosenberg; Sara Lustigman

We previously reported that rOv-ASP-1, a recombinant Onchocerca volvulus activation associated protein-1, was a potent adjuvant for recombinant protein or synthetic peptide-based Ags. In this study, we further evaluated the adjuvanticity of rOv-ASP-1 and explored its mechanism of action. Consistently, recombinant full-length spike protein of SARS-CoV or its receptor-binding domain in the presence of rOv-ASP-1 could effectively induce a mixed but Th1-skewed immune response in immunized mice. It appears that rOv-ASP-1 primarily bound to the APCs among human PBMCs and triggered Th1-biased proinflammatory cytokine production probably via the activation of monocyte-derived dendritic cells and the TLR, TLR2, and TLR4, thus suggesting that rOv-ASP-1 is a novel potent innate adjuvant.


PLOS ONE | 2013

Integration of Lyoplate Based Flow Cytometry and Computational Analysis for Standardized Immunological Biomarker Discovery

Federica Villanova; Paola Di Meglio; Margaret Inokuma; Nima Aghaeepour; Esperanza Perucha; Jennifer Mollon; Laurel Nomura; Maria P. Hernandez-Fuentes; Andrew P. Cope; A Toby Prevost; Susanne Heck; Vernon C. Maino; Graham M. Lord; Ryan R. Brinkman; Frank O. Nestle

Discovery of novel immune biomarkers for monitoring of disease prognosis and response to therapy in immune-mediated inflammatory diseases is an important unmet clinical need. Here, we establish a novel framework for immunological biomarker discovery, comparing a conventional (liquid) flow cytometry platform (CFP) and a unique lyoplate-based flow cytometry platform (LFP) in combination with advanced computational data analysis. We demonstrate that LFP had higher sensitivity compared to CFP, with increased detection of cytokines (IFN-γ and IL-10) and activation markers (Foxp3 and CD25). Fluorescent intensity of cells stained with lyophilized antibodies was increased compared to cells stained with liquid antibodies. LFP, using a plate loader, allowed medium-throughput processing of samples with comparable intra- and inter-assay variability between platforms. Automated computational analysis identified novel immunophenotypes that were not detected with manual analysis. Our results establish a new flow cytometry platform for standardized and rapid immunological biomarker discovery with wide application to immune-mediated diseases.


Journal of Clinical Investigation | 2018

Autoreactive T effector memory differentiation mirrors β cell function in type 1 diabetes

Lorraine Yeo; Alyssa Woodwyk; Sanjana Sood; Anna Lorenc; Martin Eichmann; Irma Pujol-Autonell; Rossella Melchiotti; Ania Skowera; Efthymios Fidanis; Garry Dolton; Katie Tungatt; Andrew K. Sewell; Susanne Heck; Alka Saxena; Craig A. Beam; Mark Peakman

In type 1 diabetes, cytotoxic CD8+ T cells with specificity for &bgr; cell autoantigens are found in the pancreatic islets, where they are implicated in the destruction of insulin-secreting &bgr; cells. In contrast, the disease relevance of &bgr; cell–reactive CD8+ T cells that are detectable in the circulation, and their relationship to &bgr; cell function, are not known. Here, we tracked multiple, circulating &bgr; cell–reactive CD8+ T cell subsets and measured &bgr; cell function longitudinally for 2 years, starting immediately after diagnosis of type 1 diabetes. We found that change in &bgr; cell–specific effector memory CD8+ T cells expressing CD57 was positively correlated with C-peptide change in subjects below 12 years of age. Autoreactive CD57+ effector memory CD8+ T cells bore the signature of enhanced effector function (higher expression of granzyme B, killer-specific protein of 37 kDa, and CD16, and reduced expression of CD28) compared with their CD57– counterparts, and network association modeling indicated that the dynamics of &bgr; cell–reactive CD57+ effector memory CD8+ T cell subsets were strongly linked. Thus, coordinated changes in circulating &bgr; cell–specific CD8+ T cells within the CD57+ effector memory subset calibrate to functional insulin reserve in type 1 diabetes, providing a tool for immune monitoring and a mechanism-based target for immunotherapy.

Collaboration


Dive into the Susanne Heck's collaboration.

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
Top Co-Authors

Avatar

Nedyalko Petrov

Guy's and St Thomas' NHS Foundation Trust

View shared research outputs
Top Co-Authors

Avatar

Ryan R. Brinkman

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge