Network


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

Hotspot


Dive into the research topics where Linsey Porter is active.

Publication


Featured researches published by Linsey Porter.


Journal of Immunology | 2011

Hypoxia Selectively Inhibits Respiratory Burst Activity and Killing of Staphylococcus aureus in Human Neutrophils

Naomi N. McGovern; Andrew S. Cowburn; Linsey Porter; Sarah R. Walmsley; Charlotte Summers; Alfred A.R. Thompson; Sadia Anwar; Lisa C. Willcocks; Moira K. B. Whyte; Alison M. Condliffe; Edwin R. Chilvers

Neutrophils play a central role in the innate immune response and a critical role in bacterial killing. Most studies of neutrophil function have been conducted under conditions of ambient oxygen, but inflamed sites where neutrophils operate may be extremely hypoxic. Previous studies indicate that neutrophils sense and respond to hypoxia via the ubiquitous prolyl hydroxylase/hypoxia-inducible factor pathway and that this can signal for enhanced survival. In the current study, human neutrophils were shown to upregulate hypoxia-inducible factor (HIF)-1α–dependent gene expression under hypoxic incubation conditions (3 kPa), with a consequent substantial delay in the onset of apoptosis. Despite this, polarization and chemotactic responsiveness to IL-8 and fMLP were entirely unaffected by hypoxia. Similarly, hypoxia did not diminish the ability of neutrophils to phagocytose serum-opsonized heat-killed streptococci. Of the secretory functions examined, IL-8 generation was preserved and elastase release was enhanced by hypoxia. Hypoxia did, however, cause a major reduction in respiratory burst activity induced both by the soluble agonist fMLP and by ingestion of opsonized zymosan, without affecting expression of the NADPH oxidase subunits. Critically, this reduction in respiratory burst activity under hypoxia was associated with a significant defect in the killing of Staphylococcus aureus. In contrast, killing of Escherichia coli, which is predominantly oxidase independent, was fully preserved under hypoxia. In conclusion, these studies suggest that although the NADPH oxidase-dependent bacterial killing mechanism may be compromised by hypoxia, neutrophils overall appear extremely well adapted to operate successfully under severely hypoxic conditions.


PLOS Medicine | 2016

Regulatory T Cell Responses in Participants with Type 1 Diabetes after a Single Dose of Interleukin-2: A Non-Randomised, Open Label, Adaptive Dose-Finding Trial

John A. Todd; Marina Evangelou; Antony J. Cutler; Marcin L. Pekalski; Neil M Walker; Helen Stevens; Linsey Porter; Deborah J. Smyth; Daniel B. Rainbow; Ricardo C. Ferreira; Laura Esposito; Kara Hunter; Kevin W. Loudon; Kathryn Irons; Jennie H. M. Yang; Charles Jm Bell; Helen Schuilenburg; James Heywood; Ben Challis; Sankalpa Neupane; Pamela Clarke; Gillian Coleman; Sarah Dawson; Donna Goymer; Katerina Anselmiova; Jane Kennet; Judy Brown; Sarah Caddy; Jia Lu; Jane Greatorex

Background Interleukin-2 (IL-2) has an essential role in the expansion and function of CD4+ regulatory T cells (Tregs). Tregs reduce tissue damage by limiting the immune response following infection and regulate autoreactive CD4+ effector T cells (Teffs) to prevent autoimmune diseases, such as type 1 diabetes (T1D). Genetic susceptibility to T1D causes alterations in the IL-2 pathway, a finding that supports Tregs as a cellular therapeutic target. Aldesleukin (Proleukin; recombinant human IL-2), which is administered at high doses to activate the immune system in cancer immunotherapy, is now being repositioned to treat inflammatory and autoimmune disorders at lower doses by targeting Tregs. Methods and Findings To define the aldesleukin dose response for Tregs and to find doses that increase Tregs physiologically for treatment of T1D, a statistical and systematic approach was taken by analysing the pharmacokinetics and pharmacodynamics of single doses of subcutaneous aldesleukin in the Adaptive Study of IL-2 Dose on Regulatory T Cells in Type 1 Diabetes (DILT1D), a single centre, non-randomised, open label, adaptive dose-finding trial with 40 adult participants with recently diagnosed T1D. The primary endpoint was the maximum percentage increase in Tregs (defined as CD3+CD4+CD25highCD127low) from the baseline frequency in each participant measured over the 7 d following treatment. There was an initial learning phase with five pairs of participants, each pair receiving one of five pre-assigned single doses from 0.04 × 106 to 1.5 × 106 IU/m2, in order to model the dose-response curve. Results from each participant were then incorporated into interim statistical modelling to target the two doses most likely to induce 10% and 20% increases in Treg frequencies. Primary analysis of the evaluable population (n = 39) found that the optimal doses of aldesleukin to induce 10% and 20% increases in Tregs were 0.101 × 106 IU/m2 (standard error [SE] = 0.078, 95% CI = −0.052, 0.254) and 0.497 × 106 IU/m2 (SE = 0.092, 95% CI = 0.316, 0.678), respectively. On analysis of secondary outcomes, using a highly sensitive IL-2 assay, the observed plasma concentrations of the drug at 90 min exceeded the hypothetical Treg-specific therapeutic window determined in vitro (0.015–0.24 IU/ml), even at the lowest doses (0.040 × 106 and 0.045 × 106 IU/m2) administered. A rapid decrease in Treg frequency in the circulation was observed at 90 min and at day 1, which was dose dependent (mean decrease 11.6%, SE = 2.3%, range 10.0%–48.2%, n = 37), rebounding at day 2 and increasing to frequencies above baseline over 7 d. Teffs, natural killer cells, and eosinophils also responded, with their frequencies rapidly and dose-dependently decreased in the blood, then returning to, or exceeding, pretreatment levels. Furthermore, there was a dose-dependent down modulation of one of the two signalling subunits of the IL-2 receptor, the β chain (CD122) (mean decrease = 58.0%, SE = 2.8%, range 9.8%–85.5%, n = 33), on Tregs and a reduction in their sensitivity to aldesleukin at 90 min and day 1 and 2 post-treatment. Due to blood volume requirements as well as ethical and practical considerations, the study was limited to adults and to analysis of peripheral blood only. Conclusions The DILT1D trial results, most notably the early altered trafficking and desensitisation of Tregs induced by a single ultra-low dose of aldesleukin that resolves within 2–3 d, inform the design of the next trial to determine a repeat dosing regimen aimed at establishing a steady-state Treg frequency increase of 20%–50%, with the eventual goal of preventing T1D. Trial Registration ISRCTN Registry ISRCTN27852285; ClinicalTrials.gov NCT01827735


Thorax | 2016

Hypoxia upregulates neutrophil degranulation and potential for tissue injury

K Hoenderdos; Km Lodge; Robert A. Hirst; Cheng Chen; Stefano G C Palazzo; Annette Emerenciana; Charlotte Summers; Adri Angyal; Linsey Porter; Jatinder K. Juss; Christopher O'Callaghan; Edwin R. Chilvers; Alison M. Condliffe

Background The inflamed bronchial mucosal surface is a profoundly hypoxic environment. Neutrophilic airway inflammation and neutrophil-derived proteases have been linked to disease progression in conditions such as COPD and cystic fibrosis, but the effects of hypoxia on potentially harmful neutrophil functional responses such as degranulation are unknown. Methods and results Following exposure to hypoxia (0.8% oxygen, 3 kPa for 4 h), neutrophils stimulated with inflammatory agonists (granulocyte-macrophage colony stimulating factor or platelet-activating factor and formylated peptide) displayed a markedly augmented (twofold to sixfold) release of azurophilic (neutrophil elastase, myeloperoxidase), specific (lactoferrin) and gelatinase (matrix metalloproteinase-9) granule contents. Neutrophil supernatants derived under hypoxic but not normoxic conditions induced extensive airway epithelial cell detachment and death, which was prevented by coincubation with the antiprotease α-1 antitrypsin; both normoxic and hypoxic supernatants impaired ciliary function. Surprisingly, the hypoxic upregulation of neutrophil degranulation was not dependent on hypoxia-inducible factor (HIF), nor was it fully reversed by inhibition of phospholipase C signalling. Hypoxia augmented the resting and cytokine-stimulated phosphorylation of AKT, and inhibition of phosphoinositide 3-kinase (PI3K)γ (but not other PI3K isoforms) prevented the hypoxic upregulation of neutrophil elastase release. Conclusion Hypoxia augments neutrophil degranulation and confers enhanced potential for damage to respiratory airway epithelial cells in a HIF-independent but PI3Kγ-dependent fashion.


Thorax | 2010

Live and let die: is neutrophil apoptosis defective in severe asthma?

Helen Parfrey; Neda Farahi; Linsey Porter; Edwin R. Chilvers

Patients with severe asthma make up a relatively small proportion of the total population of patients with asthma yet account for a disproportional amount of asthma-related morbidity and healthcare utilisation.1 2 These patients are usually highly symptomatic, difficult to treat and can be extremely refractory to current treatments. As a consequence, understanding the mechanisms underlying this particular form of asthma is of paramount importance. Morphological examination of the asthmatic airway reveals epithelial desquamation, thickening of the reticular basement membrane, mucus gland hyperplasia, goblet cell differentiation, angiogenesis and smooth muscle hypertrophy.3 In addition to these structural changes, an inflammatory cell infiltrate is evident within the airways comprising eosinophils, mast cells, lymphocytes and neutrophils. While eosinophils are the most characteristic inflammatory cell type present in mild to moderate asthma, the neutrophil seems to take centre stage more often in patients with severe disease. In fact, the neutrophil is one of the earliest inflammatory cells recruited to the airways following allergen exposure and is particularly evident in bronchoalveolar lavage samples and bronchial and transbronchial biopsies from patients with corticosteroid-resistant asthma, occupational asthma and fatal asthma as well as those with acute exacerbations.4–6 Furthermore, measures of asthma severity such as forced expiratory volume in 1 s correlate directly with the number of neutrophils present in the sputum and bronchial wall.7 8 There is also evidence of neutrophil activation and impaired function in the asthmatic airway.9 10 Moreover, neutrophil elastase is recognised to be highly histotoxic and results in epithelial and eosinophil activation, increased vascular permeability and promotion of transforming growth factor β release which is linked to airways remodelling. Given the propensity of neutrophils to die quite readily by ‘constitutive’ (ie, inbuilt time-dependent) apoptosis when studied in vitro,11 one major puzzle …


Clinical Immunology | 2017

Human IL-6R(hi)TIGIT(-) CD4(+)CD127(low)CD25(+) T cells display potent in vitro suppressive capacity and a distinct Th17 profile

Ricardo C. Ferreira; Daniel B. Rainbow; Arcadio Rubio García; Marcin L. Pekalski; Linsey Porter; João J. Oliveira; Frank Waldron-Lynch; Linda S. Wicker; John A. Todd

To date many clinical studies aim to increase the number and/or fitness of CD4+CD127lowCD25+ regulatory T cells (Tregs) in vivo to harness their regulatory potential in the context of treating autoimmune disease. Here, we sought to define the phenotype and function of Tregs expressing the highest levels of IL-6 receptor (IL-6R). We have identified a population of CD4+CD127lowCD25+ TIGIT- T cells distinguished by their elevated IL-6R expression that lacked expression of HELIOS, showed higher CTLA-4 expression, and displayed increased suppressive capacity compared to IL-6RhiTIGIT+ Tregs. IL-6RhiTIGIT- CD127lowCD25+ T cells contained a majority of cells demethylated at FOXP3 and displayed a Th17 transcriptional signature, including RORC (RORγt) and the capacity of producing both pro- and anti-inflammatory cytokines, such as IL-17, IL-22 and IL-10. We propose that in vivo, in the presence of IL-6-associated inflammation, the suppressive function of CD4+CD127lowCD25+ FOXP3+IL-6RhiTIGIT- T cells is temporarily disarmed allowing further activation of the effector functions and potential pathogenic tissue damage.


Clinical & Experimental Allergy | 2017

Hypoxia causes IL‐8 secretion, Charcot Leyden crystal formation, and suppression of corticosteroid‐induced apoptosis in human eosinophils

Linsey Porter; Andrew S. Cowburn; Neda Farahi; John Deighton; Stuart N. Farrow; Christine Fiddler; Jatinder K. Juss; Alison M. Condliffe; Edwin R. Chilvers

Inflamed environments are typically hypercellular, rich in pro‐inflammatory cytokines, and profoundly hypoxic. While the effects of hypoxia on neutrophil longevity and function have been widely studied, little is known about the consequences of this stimulus on eosinophils.


Methods of Molecular Biology | 2014

Measurement of eosinophil kinetics in healthy volunteers.

Neda Farahi; Chrystalla Loutsios; Rosalind P. Simmonds; Linsey Porter; Daniel Gillett; Sarah Heard; A. Michael Peters; Alison M. Condliffe; Edwin R. Chilvers

Radiolabelled leukocyte scans are widely used in nuclear medicine to locate sites of infection and inflammation. Radiolabelling of leukocyte subpopulations can also yield valuable information on cell trafficking and kinetics in vivo, but care must be taken to minimize inadvertent cell activation ex vivo. Here, we describe the use of autologous indium(111)-labelled eosinophils to measure eosinophil intravascular life-span and monitor their distribution and fate using gamma camera imaging in healthy non-atopic individuals.


JCI insight | 2018

The DILfrequency study is an adaptive trial to identify optimal IL-2 dosing in patients with type 1 diabetes

Eleonora Seelig; James Howlett; Linsey Porter; Lucy A. Truman; James Heywood; Jane Kennet; Emma L. Arbon; Katerina Anselmiova; Neil Walker; Ravinder Atkar; Marcin L. Pekalski; Ed Rytina; Mark L. Evans; Linda S. Wicker; John A. Todd; Adrian Mander; Simon Bond; Frank Waldron-Lynch

BACKGROUND Type 1 diabetes (T1D) results from loss of immune regulation, leading to the development of autoimmunity to pancreatic β cells, involving autoreactive T effector cells (Teffs). Tregs, which prevent autoimmunity, require IL-2 for maintenance of immunosuppressive functions. Using a response-adaptive design, we aimed to determine the optimal regimen of aldesleukin (recombinant human IL-2) to physiologically enhance Tregs while limiting expansion of Teffs. METHODS DILfrequency is a nonrandomized, open-label, response-adaptive study of participants, aged 18-70 years, with T1D. The initial learning phase allocated 12 participants to 6 different predefined regimens. Then, 3 cohorts of 8 participants were sequentially allocated dose frequencies, based on repeated interim analyses of all accumulated trial data. The coprimary endpoints were percentage change in Tregs and Teffs and CD25 (α subunit of the IL-2 receptor) expression by Tregs, from baseline to steady state. RESULTS Thirty-eight participants were enrolled, with thirty-six completing treatment. The optimal regimen to maintain a steady-state increase in Tregs of 30% and CD25 expression of 25% without Teff expansion is 0.26 × 106 IU/m2 (95% CI -0.007 to 0.485) every 3 days. Tregs and CD25 were dose-frequency responsive, Teffs were not. The commonest adverse event was injection site reaction (464 of 694 events). CONCLUSIONS Using a response-adaptive design, aldesleukin treatment can be optimized. Our methodology can generally be employed to immediately access proof of mechanism, thereby leading to more efficient and safe drug development. TRIAL REGISTRATION International Standard Randomised Controlled Trial Number Register, ISRCTN40319192; ClinicalTrials.gov, NCT02265809. FUNDING Sir Jules Thorn Trust, the Swiss National Science Foundation, Wellcome, JDRF, and NIHR Cambridge Biomedical Research Centre.


Frontiers in Immunology | 2018

Metabolic Profiling of Human Eosinophils

Linsey Porter; Nicole Toepfner; Kathleen Rose Bashant; Jochen Guck; Margaret Ashcroft; Neda Farahi; Edwin R. Chilvers

Immune cells face constant changes in their microenvironment, which requires rapid metabolic adaptation. In contrast to neutrophils, which are known to rely near exclusively on glycolysis, the metabolic profile of human eosinophils has not been characterized. Here, we assess the key metabolic parameters of peripheral blood-derived human eosinophils using real-time extracellular flux analysis to measure extracellular acidification rate and oxygen consumption rate, and compare these parameters to human neutrophils. Using this methodology, we demonstrate that eosinophils and neutrophils have a similar glycolytic capacity, albeit with a minimal glycolytic reserve. However, compared to neutrophils, eosinophils exhibit significantly greater basal mitochondrial respiration, ATP-linked respiration, maximum respiratory capacity, and spare respiratory capacity. Of note, the glucose oxidation pathway is also utilized by eosinophils, something not evident in neutrophils. Furthermore, using a colorimetric enzymatic assay, we show that eosinophils have much reduced glycogen stores compared to neutrophils. We also show that physiologically relevant levels of hypoxia (PO2 3 kPa), by suppressing oxygen consumption rates, have a profound effect on basal and phorbol–myristate–acetate-stimulated eosinophil and neutrophil metabolism. Finally, we compared the metabolic profile of eosinophils purified from atopic and non-atopic subjects and show that, despite a difference in the activation status of eosinophils derived from atopic subjects, these cells exhibit comparable oxygen consumption rates upon priming with IL-5 and stimulation with fMLP. In summary, our findings show that eosinophils display far greater metabolic flexibility compared to neutrophils, with the potential to use glycolysis, glucose oxidation, and oxidative phosphorylation. This flexibility may allow eosinophils to adapt better to diverse roles in host defense, homeostasis, and immunomodulation.


bioRxiv | 2017

Targeting regulatory T cells with Interleukin-2 treatment in type 1 diabetes: a response-adaptive, non-randomised, open-label trial of repeat doses of Aldesleukin (DILfrequency)

Eleonora Seelig; James Howlett; Linsey Porter; Lucy Truman; James Heywood; Jane Kennet; Emma L. Arbon; Katerina Anselmiova; Neil Walker; Ravinder Atkar; Marcin L Pekaiski; Ed Rytina; Mark L. Evans; Linda Wicker; John A. Todd; Adrian P. Mander; Simon Bond; Frank Waldron-Lynch

Background Type 1 diabetes (T1D) results from loss of immune regulation leading to the development of autoimmunity to pancreatic beta-cells, involving autoreactive T effector cells (Teffs). Regulatory T cells (Tregs), that prevent autoimmunity, require Interleukin-2 (IL-2) for maintenance of immunosuppressive functions and, alterations in the IL-2 pathway predispose to T1D. Using an adaptive trial design we aimed to determine the optimal regimen of aldesleukin (recombinant human IL-2) to physiologically enhance Tregs while limiting expansion of autoreactive Teffs. Methods DILfrequency is a single-center, non-randomised, open-label, response-adaptive study of participants aged 18 to 70 years with T1D. The initial learning phase allocated 12 participants to six different predefined dose-frequency regimens. Then, three cohorts of 8 participants were sequentially allocated dose-frequencies, based on repeated interim analyses of all accumulated trial data. The co-primary endpoints were percentage change in Tregs, Teffs and, CD25 (α subunit of the IL-2 receptor) expression by Tregs, from baseline to steady state. Trial registration ISRCTN40319192 and ClinicalTrials.gov (NCT02265809). Findings 115 participants were assessed between November 17th 2014 and May 22nd 2016, 38 participants were enrolled with 36 completing treatment. The optimal regimen to maintain a steady state increase in Tregs of 30% and CD25 expression of 25% without Teff expansion is 0.26 × 106 IU/m2 (95% CI (−0.007 to 0.485)) every 3 days (1.3 to 4.4). Tregs and CD25 were dose-frequency responsive, while Teffs were not. The commonest adverse event was injection site reaction (464/694 events), with a single participant developing transient eosinophilia at the highest dose (0.47 × 106 IU/m2). Interpretation This response-adaptive trial defined a well-tolerated aldesleukin regimen that specifically induces Treg expansion that can now be trialled to treat T1D. Funding Sir Jules Thorn Trust, Wellcome, JDRF, SNSF, NIHR

Collaboration


Dive into the Linsey Porter's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

John A. Todd

Wellcome Trust Centre for Human Genetics

View shared research outputs
Top Co-Authors

Avatar

Neda Farahi

University of Cambridge

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

K Hoenderdos

University of Cambridge

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge