scispace - formally typeset
Search or ask a question
Posted ContentDOI

Diverse outcomes of controlled human malaria infection originate from host-intrinsic immune variation and not var gene switching

TL;DR: It is found that the diverse outcomes of CHMI therefore depend upon human immune variation and there is no evidence for switching or selection of var genes in naive hosts.
Abstract: Falciparum malaria is clinically heterogeneous and the relative contribution of parasite and host in shaping disease severity remains unclear. We explored the interaction between inflammation and parasite variant surface antigen (VSA) expression, asking whether this relationship underpins the variation observed in controlled human malaria infection (CHMI). We uncovered marked heterogeneity in the response of naive hosts to blood challenge; some volunteers remained quiescent, others triggered interferon-stimulated inflammation and some showed transcriptional evidence of myeloid cell suppression. Significantly, only inflammatory volunteers experienced hallmark symptoms of malaria. When we tracked temporal changes in parasite VSA expression to ask whether variants associated with severe disease preferentially expand in naive hosts (as predicted by current theory) we found that var gene profiles were unchanged after 10-days of infection. The diverse outcomes of CHMI therefore depend upon human immune variation and there is no evidence for switching or selection of var genes in naive hosts.

Summary (2 min read)

Introduction

  • 2Milne et al 2020 Falciparum malaria is clinically heterogeneous and the relative contribution of parasite and host in shaping disease severity remains unclear.
  • Significantly, a pairwise comparison between their diagnosis and pre-infection samples revealed a core signature of 77 genes that were differentially expressed in response to blood challenge (adj p < 0.05) (Table S6).
  • The authors data therefore support previous observations that the parasite density threshold required to trigger inflammation is highly variable between individuals (41, 42).
  • In this study the authors set out to explore variation in the immune response to falciparum malaria and identified at least three possible early outcomes of blood-stage infection.
  • Existing data do not therefore support the idea of parasite genotype-specific differences in var gene expression in naive hosts, but more studies with diverse parasite genotypes are needed.

Monitoring volunteers

  • Blood samples were collected to measure parasitaemia by qPCR twice daily, starting 2-days postinfection for blood challenge and 6.5-days post-infection for mosquito challenge.
  • In both trials, thick blood smears were also evaluated by experienced microscopists at each time-point.
  • And on days -1, 6, 28 and 90 post-infection (plus diagnosis) full blood counts and blood biochemistry were performed at the Churchill and John Radcliffe Hospitals in Oxford, providing 5-part differential white cell counts and a quantitative assessment of electrolytes, urea, creatinine, bilirubin, alanine aminotransferase, alkaline phosphatase and albumin.
  • QPCR & parasite multiplication rate modeling Blood was collected and prepared for qPCR analysis as previously described (32, 79).

Uninfected control volunteers

  • Four healthy adult volunteers were recruited at the University of Edinburgh to serve as uninfected controls for the blood challenge study, also known as Blood challenge.
  • Host RNA extraction, quantification and quality control RNA extraction was performed with the Tempus™ Spin RNA Isolation Kit (Applied Biosystems) according to the manufacturer’s instructions.
  • Log2 transformed intensity values were then scaled with scale (scale=TRUE, center=TRUE), plotted with pheatmap and clustered using hclust and cutree in R, with k=8.
  • To be classified as responsive to blood challenge volunteers had to pass 2 sequential thresholds: first, for every volunteer the mean variance of their 50 most variable protein-coding genes was calculated.
  • Note that to visualise and cluster the 226 unique genes and the 117-gene superset, read counts across the entire dataset were normalised using rlogs (blind=TRUE) in DESeq2 (84).

Quantification of cytokines & chemokines in plasma

  • Interferon alpha (IFNα), Interferon gamma (IFNγ), CXCL9 (MIG) and CXCL10 (IP-10) were quantified in plasma as part of a custom-design multiplex assay from BioLegend (LegendPlex™).
  • Samples were run in duplicate and the assay was performed according to the manufacturer’s instructions using low protein-binding filter-bottom plates .
  • Beads were acquired on a BD LSRFortessa™ running FACSDiva™ software and data were analysed with LEGENDPlex™ analysis software.
  • For blood challenge, data are presented as fold-change relative to pre-infection samples; when analytes were undetectable samples were simply assigned the largest integer below the limit of detection to allow normalisation and graphing of all time-points.

Quantification of Angiopoietin-2 in plasma

  • Angiopoietin-2 was measured in plasma at pre-infection and diagnosis time-points using the Human Angiopoietin-2 Quantikine ELISA kit from R&D Systems.
  • Samples were run in duplicate and the assay performed as per the manufacturer’s instructions.
  • Optical density (OD) was determined on a Multiskan Ascent plate reader by subtracting measurements taken at 570nm wavelength from readings at 450nm.
  • A 7-point standard curve was then constructed by plotting the mean absorbance of each standard against known concentration and using regression analysis to draw a best fit line (R2 = 0.998).
  • Data are presented as mean concentration of the duplicate samples.

Detection of anti-Cytomegalovirus IgG in plasma

  • Cytomegalovirus (CMV) seropositivity was assessed in all volunteers before CHMI using an antiCMV IgG Human ELISA Kit .
  • Next, the differentially abundant metabolites were rangescaled according to van den Berg et al (45) and a heatmap of range-scaled intensity values was generated using the gplots R package.
  • The flow-through was spun at 1,000xg for 10 min. at room temperature with brake off and the supernatant carefully aspirated (being careful not to disturb the pelleted red cells).
  • Parasites were then isolated from these mock samples according to the mosquito challenge protocol outlined above.

Did you find this useful? Give us your feedback

Figures (4)

Content maybe subject to copyright    Report

Diverse outcomes of controlled human malaria infection originate from
host-intrinsic immune variation and not var gene switching
Kathryn Milne,
1
* Alasdair Ivens,
1,2
* Adam J. Reid,
3
* Magda E. Lotkowska,
3
Áine O’Toole,
2,4
Geetha
Sankaranarayanan,
3
Diana Muñoz Sandoval,
1
Wiebke Nahrendorf,
1
Clement Regnault,
5,6
Nick J.
Edwards,
7
Sarah E. Silk,
7
Ruth O. Payne,
7
Angela M. Minassian,
7
Navin Venkatraman,
7
Mandy
Sanders,
3
Adrian V.S. Hill,
7
Michael P. Barrett,
5,6
Matthew Berriman,
3
Simon J. Draper,
7
J. Alexandra
Rowe,
1,2
† Philip J. Spence
1,2
1
Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, UK.
2
Centre
for Immunity, Infection and Evolution, University of Edinburgh, Edinburgh, UK.
3
Wellcome Sanger
Institute, Cambridge, UK.
4
Institute of Evolutionary Biology, University of Edinburgh, Edinburgh,
UK.
5
Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK.
6
Glasgow
Polyomics, University of Glasgow, Glasgow, UK.
7
The Jenner Institute, University of Oxford, Oxford,
UK.
*these authors contributed equally
†joint senior authors
email alex.rowe@ed.ac.uk or philip.spence@ed.ac.uk
. CC-BY 4.0 International licenseIt is made available under a
perpetuity.
is the author/funder, who has granted medRxiv a license to display the preprint in(which was not certified by peer review)preprint
The copyright holder for thisthis version posted September 7, 2020. ; https://doi.org/10.1101/2020.09.04.20188144doi: medRxiv preprint
NOTE: This preprint reports new research that has not been certified by peer review and should not be used to guide clinical practice.

2Milne et al 2020
Abstract
Falciparum malaria is clinically heterogeneous and the relative contribution of parasite and host in
shaping disease severity remains unclear. We explored the interaction between inammation and
parasite variant surface antigen (VSA) expression, asking whether this relationship underpins the
variation observed in controlled human malaria infection (CHMI). We uncovered marked heterogeneity
in the response of naive hosts to blood challenge; some volunteers remained quiescent, others
triggered interferon-stimulated inammation and some showed transcriptional evidence of myeloid
cell suppression. Signicantly, only inammatory volunteers experienced hallmark symptoms of
malaria. When we tracked temporal changes in parasite VSA expression to ask whether variants
associated with severe disease preferentially expand in naive hosts (as predicted by current theory)
we found that var gene proles were unchanged after 10-days of infection. The diverse outcomes
of CHMI therefore depend upon human immune variation and there is no evidence for switching or
selection of var genes in naive hosts.
. CC-BY 4.0 International licenseIt is made available under a
perpetuity.
is the author/funder, who has granted medRxiv a license to display the preprint in(which was not certified by peer review)preprint
The copyright holder for thisthis version posted September 7, 2020. ; https://doi.org/10.1101/2020.09.04.20188144doi: medRxiv preprint

3Milne et al 2020
Introduction
There is enormous diversity in the human response to identical immune challenge. This variation
is currently being interrogated in large cohorts of healthy volunteers to pinpoint the genetic and
non-genetic factors that dictate human immune decision-making (1-6). Controlled human malaria
infection (CHMI) provides a well-established challenge model in which to examine immune variation
in vivo, and heterogeneity in the host response to Plasmodium falciparum is well-recognised (7,
8). Prior work suggests a dichotomy in immune decision-making with some individuals triggering
an inammatory response, which is characterised by high circulating levels of IFNγ, whilst others
mount an alternative response distinguished by the early release of TGFβ and suppression of IFNγ
signaling (9-12). The precise mechanisms underpinning these divergent responses remain unknown.
Genome-wide transcriptional proling has the potential to map immune decision-making in
unprecedented detail and previous CHMI studies have revealed an upregulation of genes associated
with pathogen detection, NFκB activation and IFNγ signaling consistent with a systemic inammatory
response (13-15). A transcriptional signature of an alternative immune response has not been
described in naive hosts. Nevertheless, these prior studies were primarily designed to measure
shared or common signatures of infection, potentially masking variation between volunteers. Only
one study (16) has specically investigated transcriptional variation in CHMI and it reported two
distinct outcomes based on host microRNA proles; notably, the authors were able to correlate the
induction of 3 specic miRNAs with a lower parasite burden suggesting that events early in infection
may have important downstream consequences. Identifying the sources of immune variation may
therefore reveal some of the factors that underpin the clinical heterogeneity of falciparum malaria. In
this context, direct intravenous challenge with blood-stage parasites provides a unique opportunity to
assess the contribution of host-intrinsic mechanisms of variation as it ensures all volunteers receive
an identical immune challenge (17).
The potential role of parasite factors in inuencing human immune decision-making has not yet
been explored in CHMI. Expression of a subset of parasite variant surface antigens (VSA) known as
group A and DC8 var genes is associated with severe malaria in both children and adults (18-23).
These virulence-associated genes encode PfEMP1 adhesion molecules that mediate sequestration
of infected red cells in the microvasculature, contributing to pathology. Group A/DC8-expressing
parasites are thought to be more pathogenic than those expressing other var types (group B and C)
and can bind the endothelium with high afnity in sensitive sites, such as brain (24). It is unknown
whether expression of these variants can inuence the immune response, for example by causing
dysregulated activation that leads to widespread collateral tissue damage. Or conversely, whether
inammation might preferentially support the survival and expansion of these variants by upregulating
. CC-BY 4.0 International licenseIt is made available under a
perpetuity.
is the author/funder, who has granted medRxiv a license to display the preprint in(which was not certified by peer review)preprint
The copyright holder for thisthis version posted September 7, 2020. ; https://doi.org/10.1101/2020.09.04.20188144doi: medRxiv preprint

4Milne et al 2020
their binding sites on the endothelium.
It has been observed in previous CHMI studies that parasites transcribe a broad array of var genes
in naive hosts, with predominant expression of group B variants and no marked differences between
volunteers (25, 26). Indeed, we have shown that mosquitoes reset malaria parasites to ensure
diverse expression of their VSA repertoire at the start of the blood cycle (27). It therefore remains
unclear how group A and DC8 var genes seemingly come to dominate the PfEMP1 landscape in
severe malaria. Antibody-mediated clearance of parasites expressing group B variants is a possible
explanation, but is unlikely because severe malaria develops in hosts with low or absent immunity
during their rst few infections of life (28). It has instead been suggested that parasites expressing
group A/DC8 var genes preferentially expand in naive hosts because they sequester and avoid
host clearance more effectively than parasites expressing group B or C var genes (29-31). This
explanation is widely accepted but is not based on substantial evidence.
In this study, we have used a blood challenge model to investigate host-intrinsic variation in the
immune response to falciparum malaria and examined the interplay between parasite and host
factors in shaping outcome of infection. This model has the advantage that an identical immune
challenge is given to all volunteers and furthermore the frequency of each parasite variant can easily
be measured at the start and end of infection to track changes through time. We set out to test the
specic hypotheses that a, expression of group A and DC8 var genes would preferentially increase
as the infection progressed and b, there would be a measurable relationship between inammation
and parasite VSA expression, which would shape the clinical outcome of infection.
. CC-BY 4.0 International licenseIt is made available under a
perpetuity.
is the author/funder, who has granted medRxiv a license to display the preprint in(which was not certified by peer review)preprint
The copyright holder for thisthis version posted September 7, 2020. ; https://doi.org/10.1101/2020.09.04.20188144doi: medRxiv preprint

5Milne et al 2020
Results
Immune decision-making in falciparum malaria
To explore host-intrinsic variation in the immune response to P. falciparum a cohort of 14 malaria-
naive volunteers were infected with an equal number of blood-stage parasites (clone 3D7) by direct
intravenous inoculation (Table S1). This enabled the pathogenic blood cycle to be initiated in every
volunteer within a 30-minute window (32). Systemic changes in the host response were then captured
through time by transcriptionally proling whole blood every 48-hours from the day before infection
until the day of diagnosis (the point of drug treatment). This time-point varied between volunteers,
occurring 7.5 to 10.5 days post-infection when two out of three diagnostic criteria were met (positive
thick blood smear and/or parasite density > 500 parasites ml
-1
by qPCR and/or symptoms consistent
with malaria). To reveal the diversity of responses within our cohort each volunteer’s time-course
was analysed independently by tracking their dynamic changes in gene expression as the infection
progressed. Importantly, this approach does not assume shared features between individuals, nor
does it bias against uncommon (or rare) responses.
As a rst step, we measured the variance of every gene through time and visualised the 100 protein-
coding genes with highest variance in each volunteer (Fig. S1). To control for baseline variation in
gene expression, uninfected control volunteers were also analysed. We found that variance in one
third of infected volunteers (4/14) was comparable to the uninfected control group, suggesting these
four volunteers did not respond to infection (Table S2). To explore this further, we merged the top 100
gene lists from all infected volunteers to produce a single non-redundant list of the most dynamically
expressed protein-coding genes across the cohort (n = 517 unique genes, henceforth called the 517-
gene superset); we could then directly compare the transcriptional response between individuals.
Accordingly, we performed a principal component analysis (PCA) of these genes through time, and
for each volunteer we determined the Euclidean distance travelled to quantify the magnitude of
their response (Fig. S2 and Table S2). Using the variance and distance travelled metrics we then
set two thresholds to identify volunteers that trigger a measurable response to blood challenge
(see methods); four volunteers failed to cross both thresholds (v018, v012, v208 & v020) and were
therefore grouped and labelled unresponsive. A pairwise comparison between their diagnosis and
pre-infection samples conrmed that there were zero differentially expressed genes in this group
(adj p < 0.05). Immune quiescence is therefore a common outcome of controlled human malaria
infection.
In contrast, variance (Fig. S1) and distance travelled (Fig. S2) both suggested that the remaining 10
volunteers made a robust response to infection (Table S2). And when we clustered and visualised
the 517-gene superset expression data we found that these volunteers could separate easily into
. CC-BY 4.0 International licenseIt is made available under a
perpetuity.
is the author/funder, who has granted medRxiv a license to display the preprint in(which was not certified by peer review)preprint
The copyright holder for thisthis version posted September 7, 2020. ; https://doi.org/10.1101/2020.09.04.20188144doi: medRxiv preprint

Citations
More filters
10 Aug 2016
TL;DR: The authors compared whole-blood RNA-seq profiles at pre-and post-infection time points from Malian adults who were either asymptomatic or febrile during their first seasonal PCR-positive P. falciparum infection with those from malaria-naive Dutch adults after a single controlled human malaria infection.
Abstract: Identifying molecular predictors and mechanisms of malaria disease is important for understanding how Plasmodium falciparum malaria is controlled. Transcriptomic studies in humans have so far been limited to retrospective analysis of blood samples from clinical cases. In this prospective, proof-of-principle study, we compared whole-blood RNA-seq profiles at pre-and post-infection time points from Malian adults who were either asymptomatic (n = 5) or febrile (n = 3) during their first seasonal PCR-positive P. falciparum infection with those from malaria-naive Dutch adults after a single controlled human malaria infection (n = 5). Our data show a graded activation of pathways downstream of pro-inflammatory cytokines, with the highest activation in malaria-naive Dutch individuals and significantly reduced activation in malaria-experienced Malians. Newly febrile and asymptomatic infections in Malians were statistically indistinguishable except for genes activated by pro-inflammatory cytokines. The combined data provide a molecular basis for the development of a pyrogenic threshold as individuals acquire immunity to clinical malaria.

58 citations

10 Jun 2019
TL;DR: Insight is provided into the molecular and cellular basis of CPS-induced immune protection from P. falciparum infection by profiling whole-blood transcriptomes by RNA-seq before and during CPS immunization and following CHMI challenge.
Abstract: A highly effective vaccine that confers sterile protection to malaria is urgently needed. Immunization under chemoprophylaxis with sporozoites (CPS) consistently confers high levels of protection in the Controlled Human Malaria infection (CHMI) model. To provide a broad, unbiased assessment of the composition and kinetics of direct ex vivo human immune responses to CPS, we profiled whole-blood transcriptomes by RNA-seq before and during CPS immunization and following CHMI challenge. Differential expression of genes enriched in modules related to T cells, NK cells, protein synthesis, and mitochondrial processes were detected in fully protected individuals four weeks after the first immunization. Non-protected individuals demonstrated transcriptomic changes after the third immunization and the day of treatment, with upregulation of interferon and innate inflammatory genes and downregulation of B-cell signatures. Protected individuals demonstrated more significant interactions between blood transcription modules compared to non-protected individuals several weeks after the second and third immunizations. These data provide insight into the molecular and cellular basis of CPS-induced immune protection from P. falciparum infection.

8 citations

References
More filters
Journal ArticleDOI
TL;DR: This work presents DESeq2, a method for differential analysis of count data, using shrinkage estimation for dispersions and fold changes to improve stability and interpretability of estimates, which enables a more quantitative analysis focused on the strength rather than the mere presence of differential expression.
Abstract: In comparative high-throughput sequencing assays, a fundamental task is the analysis of count data, such as read counts per gene in RNA-seq, for evidence of systematic changes across experimental conditions. Small replicate numbers, discreteness, large dynamic range and the presence of outliers require a suitable statistical approach. We present DESeq2, a method for differential analysis of count data, using shrinkage estimation for dispersions and fold changes to improve stability and interpretability of estimates. This enables a more quantitative analysis focused on the strength rather than the mere presence of differential expression. The DESeq2 package is available at http://www.bioconductor.org/packages/release/bioc/html/DESeq2.html .

47,038 citations


"Diverse outcomes of controlled huma..." refers methods in this paper

  • ...Once again, we used DESeq2 to identify differentially expressed parasite genes but this time between inflammatory volunteers and the rest of the cohort....

    [...]

  • ...Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNAseq data with DESeq2....

    [...]

  • ...Note that to visualise and cluster the 226 unique genes and the 117-gene superset, read counts across the entire dataset were normalised using rlogs (blind=TRUE) in DESeq2 (84)....

    [...]

  • ...Calling differentially expressed parasite genes To determine whether any parasite genes were differentially expressed between volunteers with different clinical outcomes after blood challenge, we used DESeq2 (84) to compare parasite gene expression profiles from inflammatory volunteers (n = 8) versus the rest (unresponsive and 29Milne et al 2020 suppressor, n = 4)....

    [...]

  • ...Calling differentially expressed parasite genes To determine whether any parasite genes were differentially expressed between volunteers with different clinical outcomes after blood challenge, we used DESeq2 (84) to compare parasite gene expression profiles from inflammatory volunteers (n = 8) versus the rest (unresponsive and ....

    [...]

Journal ArticleDOI
TL;DR: EdgeR as mentioned in this paper is a Bioconductor software package for examining differential expression of replicated count data, which uses an overdispersed Poisson model to account for both biological and technical variability and empirical Bayes methods are used to moderate the degree of overdispersion across transcripts, improving the reliability of inference.
Abstract: Summary: It is expected that emerging digital gene expression (DGE) technologies will overtake microarray technologies in the near future for many functional genomics applications. One of the fundamental data analysis tasks, especially for gene expression studies, involves determining whether there is evidence that counts for a transcript or exon are significantly different across experimental conditions. edgeR is a Bioconductor software package for examining differential expression of replicated count data. An overdispersed Poisson model is used to account for both biological and technical variability. Empirical Bayes methods are used to moderate the degree of overdispersion across transcripts, improving the reliability of inference. The methodology can be used even with the most minimal levels of replication, provided at least one phenotype or experimental condition is replicated. The software may have other applications beyond sequencing data, such as proteome peptide count data. Availability: The package is freely available under the LGPL licence from the Bioconductor web site (http://bioconductor.org).

29,413 citations


"Diverse outcomes of controlled huma..." refers methods in this paper

  • ...EdgeR (83) was used to identify highly dispersed genes (BCV > 0....

    [...]

Journal ArticleDOI
TL;DR: A new software suite for the comparison, manipulation and annotation of genomic features in Browser Extensible Data (BED) and General Feature Format (GFF) format, which allows the user to compare large datasets (e.g. next-generation sequencing data) with both public and custom genome annotation tracks.
Abstract: Motivation: Testing for correlations between different sets of genomic features is a fundamental task in genomics research. However, searching for overlaps between features with existing webbased methods is complicated by the massive datasets that are routinely produced with current sequencing technologies. Fast and flexible tools are therefore required to ask complex questions of these data in an efficient manner. Results: This article introduces a new software suite for the comparison, manipulation and annotation of genomic features in Browser Extensible Data (BED) and General Feature Format (GFF) format. BEDTools also supports the comparison of sequence alignments in BAM format to both BED and GFF features. The tools are extremely efficient and allow the user to compare large datasets (e.g. next-generation sequencing data) with both public and custom genome annotation tracks. BEDTools can be combined with one another as well as with standard UNIX commands, thus facilitating routine genomics tasks as well as pipelines that can quickly answer intricate questions of large genomic datasets. Availability and implementation: BEDTools was written in C++. Source code and a comprehensive user manual are freely available at http://code.google.com/p/bedtools

18,858 citations


"Diverse outcomes of controlled huma..." refers methods in this paper

  • ...We identified reads overlapping annotated var genes using bedtools intersect (95)....

    [...]

Journal ArticleDOI
TL;DR: Tests showed that HISAT is the fastest system currently available, with equal or better accuracy than any other method, and requires only 4.3 gigabytes of memory.
Abstract: HISAT (hierarchical indexing for spliced alignment of transcripts) is a highly efficient system for aligning reads from RNA sequencing experiments. HISAT uses an indexing scheme based on the Burrows-Wheeler transform and the Ferragina-Manzini (FM) index, employing two types of indexes for alignment: a whole-genome FM index to anchor each alignment and numerous local FM indexes for very rapid extensions of these alignments. HISAT's hierarchical index for the human genome contains 48,000 local FM indexes, each representing a genomic region of ∼64,000 bp. Tests on real and simulated data sets showed that HISAT is the fastest system currently available, with equal or better accuracy than any other method. Despite its large number of indexes, HISAT requires only 4.3 gigabytes of memory. HISAT supports genomes of any size, including those larger than 4 billion bases.

13,192 citations


"Diverse outcomes of controlled huma..." refers methods in this paper

  • ...falciparum v3 genome sequence using default parameters and mapped reads with parameters --max-intronlen 5000 -p 12 (94)....

    [...]

Journal ArticleDOI
TL;DR: Kallisto pseudoaligns reads to a reference, producing a list of transcripts that are compatible with each read while avoiding alignment of individual bases, which removes a major computational bottleneck in RNA-seq analysis.
Abstract: We present kallisto, an RNA-seq quantification program that is two orders of magnitude faster than previous approaches and achieves similar accuracy. Kallisto pseudoaligns reads to a reference, producing a list of transcripts that are compatible with each read while avoiding alignment of individual bases. We use kallisto to analyze 30 million unaligned paired-end RNA-seq reads in <10 min on a standard laptop computer. This removes a major computational bottleneck in RNA-seq analysis.

6,468 citations

Related Papers (5)
Frequently Asked Questions (1)
Q1. What are the contributions in "Diverse outcomes of controlled human malaria infection originate from host-intrinsic immune variation and not var gene switching" ?

Kathryn Milne this paper,1 * Alasdair Ivens,1,2 * Adam J. Reid,3 * Magda E. Lotkowska,3 Áine O ’ Toole,2,4 Geetha Sankaranarayanan,3 Diana Muñoz Sandoval,1 Wiebke Nahrendorf,1 Clement Regnault,5,6 Nick J. Edwards,7 Sarah E. Silk,7 Ruth O. Payne,7 Angela M.