Network


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

Hotspot


Dive into the research topics where Alana Keyser is active.

Publication


Featured researches published by Alana Keyser.


American Journal of Respiratory and Critical Care Medicine | 2010

Specific T Cell Frequency and Cytokine Expression Profile Do Not Correlate with Protection against Tuberculosis after Bacillus Calmette-Guérin Vaccination of Newborns

Benjamin M. Kagina; Brian Abel; Thomas J. Scriba; Elizabeth J. Hughes; Alana Keyser; Andreia Soares; Hoyam Gamieldien; Mzwandile Sidibana; Mark Hatherill; Sebastian Gelderbloem; Hassan Mahomed; Anthony Hawkridge; Gregory D. Hussey; Gilla Kaplan; Willem A. Hanekom

RATIONALE Immunogenicity of new tuberculosis (TB) vaccines is commonly assessed by measuring the frequency and cytokine expression profile of T cells. OBJECTIVES We tested whether this outcome correlates with protection against childhood TB disease after newborn vaccination with bacillus Calmette-Guérin (BCG). METHODS Whole blood from 10-week-old infants, routinely vaccinated with BCG at birth, was incubated with BCG for 12 hours, followed by cryopreservation for intracellular cytokine analysis. Infants were followed for 2 years to identify those who developed culture-positive TB-these infants were regarded as not protected against TB. Infants who did not develop TB disease despite exposure to TB in the household, and another group of randomly selected infants who were never evaluated for TB, were also identified-these groups were regarded as protected against TB. Cells from these groups were thawed, and CD4, CD8, and γδ T cell-specific expression of IFN-γ, TNF-α, IL-2, and IL-17 measured by flow cytometry. MEASUREMENTS AND MAIN RESULTS A total of 5,662 infants were enrolled; 29 unprotected and two groups of 55 protected infants were identified. There was no difference in frequencies of BCG-specific CD4, CD8, and γδ T cells between the three groups of infants. Although BCG induced complex patterns of intracellular cytokine expression, there were no differences between protected and unprotected infants. CONCLUSIONS The frequency and cytokine profile of mycobacteria-specific T cells did not correlate with protection against TB. Critical components of immunity against Mycobacterium tuberculosis, such as CD4 T cell IFN-γ production, may not necessarily translate into immune correlates of protection against TB disease.


European Journal of Immunology | 2009

Modified vaccinia Ankara-expressing Ag85A, a novel tuberculosis vaccine, is safe in adolescents and children, and induces polyfunctional CD4+ T cells

Thomas J. Scriba; Michele Tameris; Nazma Mansoor; Erica Smit; Linda van der Merwe; Fatima Isaacs; Alana Keyser; Sizulu Moyo; Nathaniel Brittain; Alison M. Lawrie; Sebastian Gelderbloem; Ashley Veldsman; Mark Hatherill; Anthony Hawkridge; Adrian V. S. Hill; Gregory D. Hussey; Hassan Mahomed; Helen McShane; Willem A. Hanekom

Modified vaccinia Ankara‐expressing Ag85A (MVA85A) is a new tuberculosis (TB) vaccine aimed at enhancing immunity induced by BCG. We investigated the safety and immunogenicity of MVA85A in healthy adolescents and children from a TB endemic region, who received BCG at birth. Twelve adolescents and 24 children were vaccinated and followed up for 12 or 6 months, respectively. Adverse events were documented and vaccine‐induced immune responses assessed by IFN‐γ ELISpot and intracellular cytokine staining. The vaccine was well tolerated and there were no vaccine‐related serious adverse events. MVA85A induced potent and durable T‐cell responses. Multiple CD4+ T‐cell subsets, based on expression of IFN‐γ, TNF‐α, IL‐2, IL‐17 and GM‐CSF, were induced. Polyfunctional CD4+ T cells co‐expressing IFN‐γ, TNF‐α and IL‐2 dominated the response in both age groups. A novel CD4+ cell subset co‐expressing these three Th1 cytokines and IL‐17 was induced in adolescents, while a novel CD4+ T‐cell subset co‐expressing Th1 cytokines and GM‐CSF was induced in children. Ag‐specific CD8+ T cells were not detected. We conclude that in adolescents and children MVA85A safely induces the type of immunity thought to be important in protection against TB. This includes induction of novel Th1‐cell populations that have not been previously described in humans.


PLOS Pathogens | 2011

Association of human TLR1 and TLR6 deficiency with altered immune responses to BCG vaccination in South African infants

April Kaur Randhawa; Muki Shey; Alana Keyser; Blas Peixoto; Richard D. Wells; Marwou de Kock; Lesedi Lerumo; Jane Hughes; Gregory D. Hussey; Anthony Hawkridge; Gilla Kaplan; Willem A. Hanekom; Thomas R. Hawn

The development of effective immunoprophylaxis against tuberculosis (TB) remains a global priority, but is hampered by a partially protective Bacillus Calmette-Guérin (BCG) vaccine and an incomplete understanding of the mechanisms of immunity to Mycobacterium tuberculosis. Although host genetic factors may be a primary reason for BCGs variable and inadequate efficacy, this possibility has not been intensively examined. We hypothesized that Toll-like receptor (TLR) variation is associated with altered in vivo immune responses to BCG. We examined whether functionally defined TLR pathway polymorphisms were associated with T cell cytokine responses in whole blood stimulated ex vivo with BCG 10 weeks after newborn BCG vaccination of South African infants. In the primary analysis, polymorphism TLR6_C745T (P249S) was associated with increased BCG-induced IFN-γ in both discovery (n = 240) and validation (n = 240) cohorts. In secondary analyses of the combined cohort, TLR1_T1805G (I602S) and TLR6_G1083C (synonymous) were associated with increased IFN-γ, TLR6_G1083C and TLR6_C745T were associated with increased IL-2, and TLR1_A1188T was associated with increased IFN-γ and IL-2. For each of these polymorphisms, the hypo-responsive allele, as defined by innate immunity signaling assays, was associated with increased production of TH1-type T cell cytokines (IFN-γ or IL-2). After stimulation with TLR1/6 lipopeptide ligands, PBMCs from TLR1/6-deficient individuals (stratified by TLR1_T1805G and TLR6_C745T hyporesponsive genotypes) secreted lower amounts of IL-6 and IL-10 compared to those with responsive TLR1/6 genotypes. In contrast, no IL-12p70 was secreted by PBMCs or monocytes. These data support a mechanism where TLR1/6 polymorphisms modulate TH1 T-cell polarization through genetic regulation of monocyte IL-10 secretion in the absence of IL-12. These studies provide evidence that functionally defined innate immune gene variants are associated with the development of adaptive immune responses after in vivo vaccination against a bacterial pathogen in humans. These findings could potentially guide novel adjuvant vaccine strategies as well as have implications for IFN-γ-based diagnostic testing for TB.


BMC Medical Genomics | 2009

Transcriptional profiling of mycobacterial antigen-induced responses in infants vaccinated with BCG at birth

Helen A. Fletcher; Alana Keyser; Mark Bowmaker; Peter C. Sayles; Gilla Kaplan; Greg Hussey; Adrian V. S. Hill; Willem A. Hanekom

BackgroundNovel tuberculosis (TB) vaccines recently tested in humans have been designed to boost immunity induced by the current vaccine, Mycobacterium bovis Bacille Calmette-Guérin (BCG). Because BCG vaccination is used extensively in infants, this population group is likely to be the first in which efficacy trials of new vaccines will be conducted. However, our understanding of the complexity of immunity to BCG in infants is inadequate, making interpretation of vaccine-induced immune responses difficult.MethodsTo better understand BCG-induced immunity, we performed gene expression profiling in five 10-week old infants routinely vaccinated with BCG at birth. RNA was extracted from 12 hour BCG-stimulated or purified protein derivative of tuberculin (PPD)-stimulated PBMC, isolated from neonatal blood collected 10 weeks after vaccination. RNA was hybridised to the Sentrix® HumanRef-8 Expression BeadChip (Illumina) to measure expression of >16,000 genes.ResultsWe found that ex vivo stimulation of PBMC with PPD and BCG induced largely similar gene expression profiles, except that BCG induced greater macrophage activation. The peroxisome proliferator-activated receptor (PPAR) signaling pathway, including PPAR-γ, involved in activation of the alternative, anti-inflammatory macrophage response was down-regulated following stimulation with both antigens. In contrast, up-regulation of genes associated with the classic, pro-inflammatory macrophage response was noted. Further analysis revealed a decrease in the expression of cell adhesion molecules (CAMs), including integrin alpha M (ITGAM), which is known to be important for entry of mycobacteria into the macrophage. Interestingly, more leukocyte genes were down-regulated than up-regulated.ConclusionOur results suggest that a combination of suppressed and up-regulated genes may be key in determining development of protective immunity to TB induced by vaccination with BCG.


Journal of Immunological Methods | 2015

Qualification of a whole blood intracellular cytokine staining assay to measure mycobacteria-specific CD4 and CD8 T cell immunity by flow cytometry.

Benjamin M. Kagina; Nazma Mansoor; Eloi P. Kpamegan; Adam Penn-Nicholson; Elisa Nemes; Erica Smit; Sebastian Gelderbloem; Andreia Soares; Brian Abel; Alana Keyser; Mzwandile Sidibana; Jane Hughes; Gilla Kaplan; Gregory D. Hussey; Willem A. Hanekom; Thomas J. Scriba

BACKGROUND Qualified or validated assays are essential in clinical trials. Short-term stimulation of whole blood and intracellular cytokine staining assay is commonly used to measure immunogenicity in tuberculosis vaccine clinical trials. Previously, the short-term stimulation process of whole blood with BCG was optimized. We aimed to qualify the intracellular cytokine staining process and assess the effects of long-term cryopreservation. Our hypotheses were that the assay is robust in the measurement of the mycobacteria-specific T cells, and long-term cryopreservation of fixed cells from stimulated whole blood would not compromise reliable measurement of mycobacteria induced CD4 T cell immunity. METHODS Whole blood from healthy adults was collected in sodium heparinized tubes. The blood was left unstimulated or stimulated with mycobacterial antigens or mitogens for 12h. Cells were harvested, fixed and multiple aliquots from each participant cryopreserved. Later, mycobacteria-specific CD4 and CD8 T cells expressing IFN-γ, TNF-α, IL-2 and IL-17 were quantitated by flow cytometry. Assay performance characteristics evaluated included limit of quantification and detection, reproducibility, precision, robustness, specificity and sensitivity. To assess the effects of long-term cryopreservation, fixed cells from the stimulated bloods were analysed one week post-cryopreservation and at 3-month intervals over a 3-year period. RESULTS The limit of quantification for the different cytokines was variable: 0.04% for frequencies of IFN-γ- and IL-2-expressing T cells and less than 0.01% for TNF-α- and IL-17-expressing T cells. When measurement of the mycobacteria-specific T cells was assessed at levels above the detection limit, the whole blood intracellular cytokine assay showed high precision that was operator-independent. The assay was also robust: variation in staining conditions including temperature (4 °C or 20-23 °C) and time (45, 60 or 90 min) did not markedly affect quantification of specific T cells. Finally, prolonged periods of cryopreservation also did not significantly influence quantification of mycobacteria-specific CD4 T cells. CONCLUSIONS The whole blood intracellular cytokine assay is robust and reliable in quantification of the mycobacteria-specific T cells and is not significantly affected by cryopreservation of fixed cells.


American Journal of Respiratory and Critical Care Medicine | 2017

Optimization and Interpretation of Serial QuantiFERON Testing to Measure Acquisition of Mycobacterium tuberculosis Infection

Elisa Nemes; Virginie Rozot; Hennie Geldenhuys; Nicole Bilek; Simbarashe Mabwe; Deborah Abrahams; Lebohang Makhethe; Mzwandile Erasmus; Alana Keyser; Asma Toefy; Yolundi Cloete; Frances Ratangee; Thomas Blauenfeldt; Morten Ruhwald; Gerhard Walzl; Bronwyn Smith; Andre G. Loxton; Willem A. Hanekom; Jason R. Andrews; Maria D. Lempicki; Ruth D. Ellis; Ann M. Ginsberg; Mark Hatherill; Thomas J. Scriba

Rationale: Conversion from a negative to positive QuantiFERON‐TB test is indicative of Mycobacterium tuberculosis (Mtb) infection, which predisposes individuals to tuberculosis disease. Interpretation of serial tests is confounded by immunological and technical variability. Objectives: To improve the consistency of serial QuantiFERON‐TB testing algorithms and provide a data‐driven definition of conversion. Methods: Sources of QuantiFERON‐TB variability were assessed, and optimal procedures were identified. Distributions of IFN‐&ggr; response levels were analyzed in healthy adolescents, Mtb‐unexposed control subjects, and patients with pulmonary tuberculosis. Measurements and Main Results: Individuals with no known Mtb exposure had IFN‐&ggr; values less than 0.2 IU/ml. Among individuals with IFN‐&ggr; values less than 0.2 IU/ml, 0.2‐0.34 IU/ml, 0.35‐0.7 IU/ml, and greater than 0.7 IU/ml, tuberculin skin test positivity results were 15%, 53%, 66%, and 91% (P < 0.005), respectively. Together, these findings suggest that values less than 0.2 IU/ml were true negatives. In short‐term serial testing, “uncertain” conversions, with at least one value within the uncertainty zone (0.2‐0.7 IU/ml), were partly explained by technical assay variability. Individuals who had a change in QuantiFERON‐TB IFN‐&ggr; values from less than 0.2 to greater than 0.7 IU/ml had 10‐fold higher tuberculosis incidence rates than those who maintained values less than 0.2 IU/ml over 2 years (P = 0.0003). By contrast, “uncertain” converters were not at higher risk than nonconverters (P = 0.229). Eighty‐seven percent of patients with active tuberculosis had IFN‐&ggr; values greater than 0.7 IU/ml, suggesting that these values are consistent with established Mtb infection. Conclusions: Implementation of optimized procedures and a more rigorous QuantiFERON‐TB conversion definition (an increase from IFN‐&ggr; <0.2 to >0.7 IU/ml) would allow more definitive detection of recent Mtb infection and potentially improve identification of those more likely to develop disease.


Vaccine | 2017

H1:IC31 vaccination is safe and induces long-lived TNF-α+IL-2+CD4 T cell responses in M. tuberculosis infected and uninfected adolescents: A randomized trial

Helen Mearns; Hennie Geldenhuys; Benjamin M. Kagina; Munyaradzi Musvosvi; Francesca Little; Frances Ratangee; Hassan Mahomed; Willem A. Hanekom; Søren T. Hoff; Morten Ruhwald; Ingrid Kromann; Peter Bang; Mark Hatherill; Peter Andersen; Thomas J. Scriba; Virginie Rozot; Deborah Abrahams; Katya Mauff; Erica Smit; Yolande Brown; E. Jane Hughes; Edward Makgotlho; Alana Keyser; Mzwandile Erasmus; Lebohang Makhethe; Hadn Africa; Charles Hopley; Marcia Steyn

BACKGROUND Control of the tuberculosis epidemic requires a novel vaccine that is effective in preventing tuberculosis in adolescents, a key target population for vaccination against TB. METHODS Healthy adolescents, stratified by M. tuberculosis-infection status, were enrolled into this observer-blinded phase II clinical trial of the protein-subunit vaccine candidate, H1:IC31, comprising a fusion protein (H1) of Ag85B and ESAT-6, formulated with the IC31 adjuvant. Local and systemic adverse events and induced T cell responses were measured after one or two administrations of either 15μg or 50μg of the H1 protein. RESULTS Two hundred and forty participants were recruited and followed up for 224days. No notable safety events were observed regardless of H1 dose or vaccination schedule. H1:IC31 vaccination induced antigen-specific CD4 T cells, co-expressing IFN-γ, TNF-α and/or IL-2. H1:IC31 vaccination of M.tb-uninfected individuals preferentially drove the emergence of Ag85B and ESAT-6 specific TNF-α+IL-2+CD4 T cells, while H1:IC31 vaccination of M.tb-infected individuals resulted in the expansion of Ag85B-specific but not ESAT-6-specific TNF-α+IL-2+CD4 T cells. CONCLUSIONS H1:IC31 was safe and immunogenic in uninfected and M.tb-infected adolescents. Two administrations of the 15μg H1:IC31 dose induced the greatest magnitude immune response, and was considered optimal (South African National Clinical Trials Register, DoH-27-0612-3947; Pan African Clinical Trial Registry, PACTR201403000464306).


Clinical Infectious Diseases | 2018

Safety and Immunogenicity of Newborn MVA85A Vaccination and Selective, Delayed Bacille Calmette-Guerin for Infants of Human Immunodeficiency Virus-Infected Mothers: A Phase 2 Randomized, Controlled Trial

Elisa Nemes; Anneke C. Hesseling; Michele Tameris; Katya Mauff; Katrina Downing; Humphrey Mulenga; Penelope Rose; Marieke van der Zalm; Sharon Mbaba; Danelle Van As; Willem A. Hanekom; Gerhard Walzl; Thomas J. Scriba; Helen McShane; Mark Hatherill; Charmaine Abrahams; Deborah Abrahams; Hadn Africa; Veronica Baartman; Beauty Bavuma; Nicole Bilek; Natasja Botes; Yolande Brown; Yolundi Cloete; Margareth Damons; Ronel De Vos; Portia Dlakavu; Karen Du Preez; Mzwandile Erasmus; Claudia Francis

Newborn MVA85A prime vaccination was safe and induced an early immune response that did not interfere with immunogenicity of subsequent bacille Calmette-Guérin vaccination. New tuberculosis vaccine candidates should be tested using this strategy, which appears safe regardless of infant human immunodeficiency virus exposure.


Immunity, inflammation and disease | 2017

B‐cells with a FasL expressing regulatory phenotype are induced following successful anti‐tuberculosis treatment

Ilana C. van Rensburg; Léanie Kleynhans; Alana Keyser; Gerhard Walzl; Andre G. Loxton

Studies show that B‐cells, in addition to producing antibodies and antigen‐presentation, are able to produce cytokines as well. These include regulatory cytokines such as IL‐10 by regulatory B‐cells. Furthermore, a rare regulatory subset of B‐cells have the potential to express FasL, which is a death‐inducing ligand. This subset of B‐cells have a positive role during autoimmune disease, but has not yet been studied during tuberculosis. These FasL‐expressing B‐cells are induced by bacterial LPS and CpG, thus we hypothesized that this phenotype might be induced during tuberculosis as well.


Journal of Inflammation | 2016

Phenotypic analysis of peripheral B cell populations during Mycobacterium tuberculosis infection and disease

Willem J. du Plessis; Alana Keyser; Gerhard Walzl; Andre G. Loxton

BackgroundMycobacterium tuberculosis (Mtb) remains an unresolved threat resulting in great annual loss of life. The role of B cells during the protective immunity to Mtb is still unclear. B cells have been described as effector cells in addition to their role as antibody producing cells during disease.Here we aim to identify and characterize the frequency of peripheral B-cell subpopulations during active Tuberculosis and over treatment response. Analysis were done for both class switched (CS) and non-class switched (NCS) phenotypes. Methods We recruited participants with active untreated pulmonary Tuberculosis, other lung diseases and healthy community controls. All groups were followed up for one week from recruitment and the TB cases till the end of treatment (month 6).ResultsPeripheral blood samples were collected, stained with monoclonal antibodies to CD19+ cells, Immunoglobulin (Ig) M, plasma cells (CD 138+), marker of memory (CD27+), immune activation (CD23+) and acquired on a flow cytometer. Circulating Marginal zone B cells (CD19+IgM+CD23−CD27+) and memory phenotypes are able to distinguish between TB diagnosis and end of treatment. The frequency of mature B cells from TB cases are lower than that of other-lung diseases at diagnosis. A subpopulation of activated memory B cells (CD19+IgM+CD23+CD27+) cells are present at the end of TB treatment.ConclusionsThis study identified distinctive B cell subpopulations present during active TB disease and other lung disease conditions. These cell populations warrants further examination in larger studies as it may be informative as cell markers or as effectors/regulators in TB disease or TB treatment response.

Collaboration


Dive into the Alana Keyser's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gilla Kaplan

Public Health Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Brian Abel

University of Cape Town

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge