Skip to main content
Erschienen in: Malaria Journal 1/2019

Open Access 01.12.2019 | Review

Use of gene expression studies to investigate the human immunological response to malaria infection

verfasst von: Susanne H. Hodgson, Julius Muller, Helen E. Lockstone, Adrian V. S. Hill, Kevin Marsh, Simon J. Draper, Julian C. Knight

Erschienen in: Malaria Journal | Ausgabe 1/2019

Abstract

Background

Transcriptional profiling of the human immune response to malaria has been used to identify diagnostic markers, understand the pathogenicity of severe disease and dissect the mechanisms of naturally acquired immunity (NAI). However, interpreting this body of work is difficult given considerable variation in study design, definition of disease, patient selection and methodology employed. This work details a comprehensive review of gene expression profiling (GEP) of the human immune response to malaria to determine how this technology has been applied to date, instances where this has advanced understanding of NAI and the extent of variability in methodology between studies to allow informed comparison of data and interpretation of results.

Methods

Datasets from the gene expression omnibus (GEO) including the search terms; ‘plasmodium’ or ‘malaria’ or ‘sporozoite’ or ‘merozoite’ or ‘gametocyte’ and ‘Homo sapiens’ were identified and publications analysed. Datasets of gene expression changes in relation to malaria vaccines were excluded.

Results

Twenty-three GEO datasets and 25 related publications were included in the final review. All datasets related to Plasmodium falciparum infection, except two that related to Plasmodium vivax infection. The majority of datasets included samples from individuals infected with malaria ‘naturally’ in the field (n = 13, 57%), however some related to controlled human malaria infection (CHMI) studies (n = 6, 26%), or cells stimulated with Plasmodium in vitro (n = 6, 26%). The majority of studies examined gene expression changes relating to the blood stage of the parasite. Significant heterogeneity between datasets was identified in terms of study design, sample type, platform used and method of analysis. Seven datasets specifically investigated transcriptional changes associated with NAI to malaria, with evidence supporting suppression of the innate pro-inflammatory response as an important mechanism for this in the majority of these studies. However, further interpretation of this body of work was limited by heterogeneity between studies and small sample sizes.

Conclusions

GEP in malaria is a potentially powerful tool, but to date studies have been hypothesis generating with small sample sizes and widely varying methodology. As CHMI studies are increasingly performed in endemic settings, there will be growing opportunity to use GEP to understand detailed time-course changes in host response and understand in greater detail the mechanisms of NAI.
Hinweise

Supplementary information

Supplementary information accompanies this paper at https://​doi.​org/​10.​1186/​s12936-019-3035-0.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
CHMI
controlled human malaria infection
GEO
gene expression omnibus
GEP
gene expression profile
NAI
naturally acquired immunity
PBMC
peripheral blood mononuclear cells

Background

Malaria, caused by infection with parasites of the genus Plasmodium, remains a significant public health concern [1]. Despite a vaccine in pilot implementation trials [2] and widespread application of control measures [3], the disease is still responsible for a huge burden of mortality and morbidity worldwide and a concerning increase in incidence has been seen in previously well-controlled areas [3].
With repeated exposure to infection, individuals in malaria-endemic regions develop naturally acquired immunity (NAI), first to the most severe clinical forms, such as cerebral malaria and then more slowly to infection itself [1]. Although the role of antibodies in controlling parasite density, symptomatology and severity of disease is well established [4, 5], less is known about mechanism in terms of the role of the innate and cellular immune responses [6]. Increased understanding of the immune response to malaria, in particular those that mediate NAI, could aid identification of diagnostic and prognostic markers, inform vaccine development and assist with the identification of treatment strategies to modify the immunological mechanisms mediating severe pathology [1].
Transcriptomics, which allows the expression of thousands of genes to be assessed in parallel for a single RNA sample, is an exciting, expanding area of research with vast potential application in the field of infection [7]. Facilitating a systems biology approach, gene expression data from high-throughput technologies (such as microarrays [8] and next generation sequencing enabling RNA sequencing for bulk cell populations and at single-cell resolution [9, 10]) can allow greater understanding of individuals’ response to infection. To date, expression data have been used to dissect mechanisms of vaccine immunogenicity [11], inform the design of new vaccines [12, 13], predict response to infection and outcome [14, 15], characterize and improve understanding of sepsis [16], and offer a novel approach to the diagnosis of infectious pathogens [1719] together with RNA expression in the pathogen [20].
Given the limited understanding of the mechanisms of NAI to malaria from traditional immunological studies, a systems approach characterizing the gene expression patterns associated with infection could provide novel and valuable insights [21, 22]. Transcriptional profiling of the immune response to malaria in humans to date has sought to identify markers to aid diagnosis [23], to understand the pathogenicity of severe disease [24] and dissect the mechanisms of NAI [25, 26]. However, interpreting this body of work is difficult given considerable variation in study design, definition of disease, patient selection and methodology employed.
This review outlines a comprehensive analysis of all GEP studies of the human immune response to malaria with two aims: (i) to understand the application of this technology to date, in particular how these studies have informed understanding of NAI; and (ii) to determine the extent of variability in methodology between studies to allow informed comparison of data and interpretation of results.

Methods

A search of Gene Expression Omnibus (GEO) [27] for datasets including the search terms; ‘plasmodium’ or ‘malaria’ or ‘sporozoite’ or ‘merozoite’ or ‘gametocyte’ and ‘Homo sapiens’ was performed on 10th September 2019. Each of these datasets were examined and those not relating to the human immune response to malaria infection or using the Homo sapiens platform excluded. Of note, datasets of gene expression changes in relation to malaria vaccines were excluded.

Results

Studies identified

The search identified 30 GEO datasets. Seven of these datasets were excluded, as published analyses were unavailable. Twenty-three datasets and 25 related publications were therefore included in the final review (Table 1 and Additional file 1: Figure S1). All datasets related to Plasmodium falciparum infection except two that related to Plasmodium vivax infection (Table 1). The majority of datasets included samples from individuals infected with malaria ‘naturally’ in the field (n = 13, 57%), however some related to controlled human malaria infection (CHMI) studies (n = 6, 26%), or cells stimulated with Plasmodium in vitro (n = 6, 26%). Studies included samples from individuals with a wide range of ages (from 2 months—varying ages of adulthood) with differing degrees of prior exposure and, therefore, NAI to malaria. Samples were often collected as part of wider immuno-epidemiological studies or vaccine trials, leading to variation in study design and sampling intervals.
Table 1
Summary of gene expression datasets investigating the human immunological response to malaria infection
GEO series
Title of dataset
Publication
Design
Infection/antigenic Stimulation
Species
Tissue
Age
Participant origin
Expression profiling
Subjects (samples)a
Controls
Platform name
Platform technology
GSE2900
Host response malaria
Griffiths et al. (2005)
Comparison of GEP in febrile children with convalescent samples 2 weeks post discharge
Field
P. falciparum
Whole blood: PAX gene
Children 2–126 months
Kenya
Array
22 (28)
Subject paired samples: diagnosis and post treatment
LC-36
Spotted DNA/cDNA
GSE5418
Gene expression analysis in malaria infection
Ockenhouse et al. (2006)
Comparison of GEP in early, pre-symptomatic blood-stage infection post CHMI with symptomatic malaria-experienced adults with naturally acquired malaria
CHMI and Field
P. falciparum
PBMC
Adults; 19–49 years
USA and Cameroon
Array
37 (74)
22 un-infected malaria-naïve American adults
Affymetrix human genome U133A array
In situ oligonucleotide
GSE15221
Malaria primes the innate immune response due to IFNγ induced enhancement of Toll-like receptor expression and function
Franklin et al. (2009) and Sharma et al. (2011) and Hirako et al. (2018)
Comparison GEP at malaria diagnosis and 28 days post treatment
Field
P. falciparum
PBMC
Adults 30 ± 10 years
Brazil; Porto Velho
Array
21 (42)
Subject paired samples: diagnosis and post treatment
Illumina human-6 v2.0
Oligonucleotide beads
GSE26876
Time kinetics of gene expression in NK92 cells after P. falciparum-iRBC encounter
De Carvalho et al. (2011)
Comparison of GEP variation of NK92 cells after 6, 12, and 24 h of co-culture with either infected or uninfected RBC compared to time-point 0
In vitro—iRBC
P. falciparum
NK92 cell line
N/A
N/A
Array
N/A (12)
Paired samples: pre and post exposure
Affymetrix human gene 1.0 ST array
In situ oligonucleotide
GSE33811
Paired whole blood human transcription profiles from children with severe malaria and mild malaria
Krupka et al. (2012)
Comparison of GEP in severe malaria and subsequent mild malaria in same subjects 1 month later
Field
P. falciparum
Whole blood: tri-reagent BD
Children: 8–45 months
Malawi
Array
5 (10)
Subject paired samples: severe and mild malaria
Affymetrix Human Gene 1.0 ST Array
In situ oligonucleotide
GSE34404
The genomic architecture of host whole blood transcriptional response to malaria infection
Idaghdour et al. (2012)
Comparison of GEP in mild malaria with age matched un-infected controls
Field
P. falciparum
Whole blood: Tempus
Children; median age 3.7 years
Benin
Array
94 subjects (94) and 64 controls (64)
Uninfected age matched
Illumina HumanHT-12 V4.0 expression bead chip
Oligonucleotide beads
GSE55843
Loss and dysfunction of Vdelta2 + gamma delta-low T cells is associated with clinical tolerance to malaria
Jagannathan et al. (2014)
Comparison of GEP of Vδ2 + T cells from children with ‘high’ and ‘low’ episodes of malaria in the preceding year
In vitro—iRBC
P. falciparum
Vδ2 + T cells
Children: 4–5 years
Uganda
Array
78 (156)
N/A
Agilent-039494 SurePrint G3 Human GE v2 8 × 60K Microarray 03938
In situ oligonucleotide
GSE53292
Transcriptomic analysis of Plasmodium PBANKA, PBSLTRiP-KO, PB268-KO parasite infected and uninfected host cell
Jaijyan et al. (2015)
Comparison of GEP of uninfected HepG2 with those infected with wild-type and knock out sporozoites
In vitro—sporozoites
P. falciparum
HepG2 cells
N/A
N/A
High throughput sequencing
NK
NK
Illumina Genome Analyzer IIx (Homo sapiens)
High-throughput sequencing
GSE50957
Molecular hallmarks of experimentally acquired immunity to malaria [Pilot Study]
Tran et al. (2016) and Vallejo et al. (2018)
Comparison of GEP pre and post infection
CHMI
P. falciparum
Whole blood: PAX gene
Adults: 19–22 years
USA
High throughput sequencing
5 (10)
Subject paired samples: Pre and post infection
Illumina HiSeq 2000 (Homo sapiens)
High-throughput sequencing
GSE52166
Molecular hallmarks of naturally acquired immunity to malaria
Tran et al. (2016)
Comparison of GEP pre and post infection
Field
P. falciparum
Whole blood: Tempus
Adults and Children 13.5–23.3 years
Malawi
High throughput sequencing
8 (16)
Paired same subject pre infection
Illumina HiSeq 2000 (Homo sapiens)
High-throughput sequencing
GSE64338
Expression data from whole blood samples of Rwandan adults with mild malaria with matched sample 30 days later (convalescence)
Subramaniam et al. (2015)
Comparison of GEP in mild malaria and 30 days later
Field
P. falciparum
Whole blood: Tri-Reagent BD
Adults
Rwandan
Array
19 (38)
Subject paired samples: diagnosis and post treatment
[HuGene-1_0-st] Affymetrix Human Gene 1.0 ST Array
In situ oligonucleotide
GSE64493
FCRL5 delineates functionally impaired memory B cells associated with malaria exposure
Sullivan (2015)
Comparison of GEP between classical and atypical memory B cells in Uganda children
Field
P. falciparum
PBMC
Children 8–10 years
Uganda
Array
12
NK
Agilent-039494 SurePrint G3 Human GE v2 8 × 60K Microarray 039381
In situ oligonucleotide
GSE67184
Transcription profiling of malaria-naïve and semi-immune colombian volunteers in a Plasmodium vivax sporozoite challenge
Rojas-Penas (2015), Vallejo (2018) and Gardinassi (2018)
Comparison of GEP changes between malaria naïve and semi-immune adults pre-infection and at diagnosis
CHMI
P. vivax
Whole blood: Tempus
Adults
Columbia
High throughput sequencing
12 (24)
Subject paired samples: pre-infection and diagnosis
Illumina HiSeq 2500 (Homo sapiens)
High-throughput sequencing
GSE67469
Transcription profiling of malaria-naïve and semi-immune colombian volunteers in a Plasmodium vivax sporozoite challenge
Rojas-Penas (2015)
Comparison of GEP changes between malaria naïve and semi-immune adults over the time-course of malaria infection: pre-infection, day 5, day 7, day 9, diagnosis and month 4
CHMI
P. vivax
Whole blood: Tempus
Adults
Columbia
RT-qPCR
16 (85)
Subject paired samples: Pre infection and multiple time-points post infection
Fluidigm 96×96 nanofluidic arrays for 96 genes: blood informative transcripts
RT-PCR
GSE7586
Genome wide analysis of placental malaria
Muehlenbachs (2007)
Comparison of GEP in women with placental malaria and those without
Field
P. falciparum
Placenta
Adults
Tanzania
Array
20 (20)
NK
[HG-U133_Plus_2] Affymetrix Human Genome U133 Plus 2.0 Array
In situ oligonucleotide
GSE77122
Involvement of β-defensin 130 (DEFB130) in the macrophage microbicidal mechanisms for killing Plasmodium falciparum
Terkawi (2017)
Human monocyte-derived macrophages were co-cultured with P. falciparum iRBCs, saponin-treated iRBCs, or non-infected RBCs
In vitro—iRBC
P. falciparum
Macrophages
NK
NK
Array
NK (8)
NK
Agilent-028004 SurePrint G3 Human GE 8 × 60K Microarray
In situ oligonucleotide
GSE93664
Comparison of the transcriptomic profile of P. falciparum reactive polyfunctional and IFNγ monofunctional human CD4 T cells
Burel (2017)
Comparison of GEP in monofunctional and polyfunctional IFN producing T cells collected 21 days post CHMI infection
CHMI + in vitro—iRBC
P. falciparum
IFN producing T cells
18–42 years
Australia
Array
8 (2)
NK
[HuGene-2_0-st] Affymetrix Human Gene 2.0 ST Array
In situ oligonucleotide
GSE100562
RNA-sequencing analysis of response to P. falciparum infection in Fulani and Mossi ethnic groups, Burkina Faso
Quin (2017)
Comparison of GEP in onocytes and CD14− cells in P. falciparum infected and uninfected malaria-exposed Fulani and Mossi sympatric ethnic groups
Field
P. falciparum
Monocytes (CD14+) and lymphocytes (CD14−)
15–24 years
Burkino Faso
High throughput sequencing
23 (23)
NK
Illumina HiSeq 2500 (Homo sapiens)
High-throughput sequencing
GSE1124
Whole blood transcriptome of childhood malaria
Boldt (2019)
Comparison of GEP of children with asymptomatic parasitemia, uncomplicated malaria, malaria with severe anaemia and cerebral malaria
Field
P. falciparum
Whole blood: PAX gene
0.5–6 years
Gabon
Array
NK
Healthy control children
[HG-U133A] Affymetrix Human Genome U133A Array
In situ oligonucleotide
GSE114076
Differential gene expression profile of human neutrophils cultured with Plasmodium falciparum-parasitized erythrocytes
Terkawi (2018)
Comparison of GEP in neutrophils incubated with iRBC or non-infected RBC
In vitro—iRBC
P. falciparum
Neutrophils
NK
NK
Array
1 (8)
Culture with non-infected RBC
Agilent-072363 SurePrint G3 Human GE v3 8 × 60K Microarray
In situ oligonucleotide
GSE97158
Transcriptional responses induced by controlled human malaria infection (CHMI)
Rothan (2018)
Comparison of GEP in whole blood pre and post sporozoite CHMI in malaria exposed adults
CHMI
P. falciparum
Whole blood: PAX gene
Adults
Tanzania
High throughput sequencing
10 (40)
Subject paired samples: pre and post CHMI
Illumina HiSeq 2000 (Homo sapiens)
High-throughput sequencing
GSE65928
Malaria-associated atypical memory B cells exhibit markedly reduced B cell receptor signaling and effector function
Portugal (2015)
Comaprison of GEP of naïve B cells, classical and atypical memory B cells in immune adults
Field
P. falciparum
B cells
Adults: 18–37 years
Mali
Array
20 (20)
US healthy adults
[HuGene-2_0-st] Affymetrix Human Gene 2.0 ST Array [transcript (gene) version]
In situ oligonucleotide
GSE72058
Activated neutrophils are associated with pediatric cerebral malaria vasculopathy in Malawian children
Feintuch (2016)
Comparison of GEP in cerebral malaria between children with malaria retinopathy and those without
Field
P. falciparum
Whole blood: Tri-Reagent BD
Children 6 month–12 years
Mali
Array
98 (98)
NK
[HuGene-1_0-st] Affymetrix Human Gene 1.0 ST Array [transcript (gene) version]
In situ oligonucleotide
PBMC peripheral blood mononuclear cells, GEP gene expression profile, CHMI controlled human malaria infection, iRBCs infected red blood cells, N/A not applicable, NK not known
aSamples analysed for publication

Review of methodological approaches

Significant heterogeneity in the datasets was found in terms of study design, sample type, platform used and method of analysis (Tables 1, 2 and Fig. 1), making direct comparison of results between studies difficult. Most datasets were generated from whole blood samples (n = 11, 48%), however some used PBMCs (n = 3, 13%) or individual tissue or cells types (n = 8, 35%) (Table 1). For the majority of studies, expression profiling was performed by array (n = 16, 70%), with others using high throughput sequencing (n = 6, 26%) or RT-qPCR [28] (n = 1, 4%) (Table 1). There was heterogeneity in data generation between studies with variation in methods used for normalization of data and adjustment for co-variables (Table 2). Thresholds for significance varied considerably and not all studies applied corrections for multiple testing. Choice of database used for gene ontology analysis also varied and there was variable, often incomplete reporting of analysis methods used (Table 2).
Table 2
Comparison of methodological approaches for analysis of gene expression data
Dataset
Data generation
Gene ontology analysis
GEO series
Publication
RNA Quantification Platform
Normalization
Adjustment for covariates
Definition expression
Expressed genes
Threshold FC
Threshold P
Test
Multiple testing
GO analysis
Threshold GO enrichment p
Test
Multiple testing
GSE2900
Griffiths (2005)
Stanford University cDNA lymphochip two color microarray
Scaled to geometric mean of sample:reference signal ratio from all array features
NS
Signal threshold
9869
2.5 (from median in > 4 samples)
0.1
Permutation
FDR
NA
NA
NA
NA
GSE5418
Ockenhouse (2006)
Affymetrix U133A GeneChips
RMA
NS
NS
NS
No
0.01
SAM, t-test
FDR
Onto Express and Pathway Architect
0.05
NS
FDR
GSE15221
Franklin (2009) and Sharma (2011)
Illumina Human WG-6 v2.0
Cubic spline
NS
Signal threshold
NS
1.7
0.01
Paired t-test
FDR
Onto Express
Varying
NS
NS
GSE15221
Hirako (2018)
Illumina Human WG-6 v2.0
Cubic spline
NS
Signal threshold
NS
1.5
0.01
Permutation and t-test
FDR
DAVID, GSEA
0.05
Multiple
FDR
GSE26876
de Carvalho (2011)
Affymetrix Human Gene 1.0 ST Array
RMA
NS
NS
NS
1.5
0.05
Student t-test
No
Ingenuity pathway analysis
NS
NS
NS
GSE33811
Krupka (2012)
Affymetrix Human Gene 1.0 ST Array
RMA and Quantile
NS
Signal and variation threshold
3110
2
0.05
Paired t-test
No
Gene set enrichment analysis on selected GO terms
0.01
Paired t-test
FDR
GSE34404
Idaghdour (2012)
Illumina Human HT-12 BeadChips
Quantile
Location, Sex, Hb, total cell counts (RBCs and WBCs) and ancestry
Signal and normality threshold
NS
2 (for comparison)
0.01
ANOVA, ANCOVA
FDR
Gene set enrichment analysis on customized MsigDB database
0.05
NS
Bonferroni
GSE55843
Jagannathan (2014)
Agilent Sure Print G3 Human Gene Expression 8 × 60K v2 gene expression microarrays
Quantile
NS
Signal threshold
NS
2
0.05
SAM
FDR
NA
NA
NA
NA
GSE53292
Jaijyan (2015)
Illumina Genome Analyzer Iix 72SE
NS
NS
NS
NS
NS
0.05
t-test
No
GeneCodis3, Bingo 2.3 plugin (Cytoscape 2.8.3)
0.05
NS
NS
GSE50957 GSE52166
Tran (2016)
Illumina HiSeq 2000 2 × 100 PE
TAMM
Batch, Sex, Age, Pre-infection baseline
Signal and variation threshold, removal Y chromosomes
NS
1.5
0.05
Limma
FDR
Ingenuity pathway analysis
0.05
Fisher exact test
FDR
GSE50957 GSE67184
Vallejo (2018)
Illumina HiSeq 2000 2 × 100 PE
CPM, TPM
NS
Signal threshold
NS
NS
0.05
EdgeR
FDR
WGSEA, ToppGene, STRING
0.05
Multiple
FDR
GSE64338
Subramaniam (2015)
Affymetrix Human Gene 1.0 ST Array
Nonlinear normalization based on Li-Wong methods
NS
NS
NS
1.2
0.001
Paired t-test
FDR
Ingenuity Pathway Analysis
0.05
NS
FDR
GSE64493
Sullivan (2015)
Agilent Sure Print G3 Human Gene Expression 8 × 60K v2 gene expression microarrays
Quantile
NS
Signal threshold
NS
1.5
0.03
Limma
FDR
DAVID
0.05
NS
FDR
GSE67184
Rojas-Penas (2015)
Illumina HiSeq 2500 2 × 100 PE
SNM
Location/time-point, subject (random effect)
Signal threshold
6154
No
0.05
NS
FDR
NA
NA
NA
NA
GSE67184
Gardinassi (2018)
Illumina HiSeq 2500 2 × 100 PE
NS
NS
NS
NS
No
0.05
Limma, repeated measures ANOVA
FDR
GSEA on blood transcriptome modules (BTM, Li et al.)
0.05
permutation
FDR
GSE7586
Muehlenbachs (2007)
Affymetrix U133 Plus 2.0 GeneChip
GC RMA
NS
NS
NS
2.5
0.01
t-test
No
NA
NA
NA
NA
GSE77122
Tarawa (2017)
Agilent Sure Print G3 Human Gene Expression 8 × 60K gene expression microarrays
Each gene expression array dataset was normalized to the in silicon pool for the macrophages cultured with RBCs
NS
NS
NS
No
0.05
Paired t-test
No
DAVID
0.05
Fisher exact test
No
GSE93664
Burl (2017)
Affymetrix Human Gene ST 2.0 gene array
RMA
NS
NS
NS
2
0.05
NS
No
STRING
0.01
NS
Corrected unspecified
GSE100562
Quin (2017)
Illumina HiSeq 2500 2 × 50 PE
NS
NS
NS
NS
No
0.05
Limma
FDR
NA
NA
NA
NA
GSE1124
Boldt (2019)
Affymetrix U133A + B GeneChips
RMA
NS
Signal threshold
NS
1.9
0.004
SAM
FDR
DAVID and Ingenuity Pathway Analysis
0.05
NS
NS
GSE114076
Terkawi (2018)
Agilent Sure Print G3 Human Gene Expression 8 × 60K gene expression microarrays
Each gene expression array dataset was normalized to the in silicon pool for the neutrophils cultured with RBCs
NS
NS
NS
2
0.01
Limma
No
Genomatix GeneRanker, DAVID, NET-GE and Enricher
0.05
NS
Corrected unspecified
GSE97158
Rothan (2018)
Illumina HiSeq 2500 2 × 51 PE
TMM
Blocking by subject, in two separate models interaction with cell count and time of parasitemia was added
Signal threshold
16,473
1.5
0.05
Limma
FDR
GSEA (camera) on blood transcriptome modules (BTM, Li et al.)
0.05
Fisher exact test
FDR
GSE65928
Portugal (2015)
Affymetrix Human Gene ST 2.0 gene array
RMA
NS
NS
NS
NS
0.05
ANOVA
FDR
Ingenuity pathway analysis
NS
NS
NS
GSE72058
Feintuch (2016)
Affymetrix Human Gene 1.0 ST array
RMA and Quantile
Peripheral parasitemia
NS
NS
No
0.05
t-test
No
GSEA, CateGOrizer and ingenuity pathway analysis
0.2 and 0.06
NS
FDR
FDR false discovery rate, Hb haemoglobin, NA not available, NS not specified in publication, RBCs red blood cells, RMA Robust Multichip average, SNM supervised normalization of microarray, TMM trimmed mean of M-values, GEO Gene Expression Omnibus, GE gene ontology

Transcriptional insights into the immune response to malaria infection

Seven datasets provided insight into the transcriptional changes associated with NAI to malaria (Table 3) [2426, 2831]. However, given the difficulty in defining or quantifying NAI for an individual, studies varied in their approach, choosing to examine GEPs in settings of varying history of prior exposure to malaria [25, 26, 28, 29], symptomatology during infection [25] or severity of disease [24, 32]. All studies examining NAI included small numbers of subjects and all deployed different experimental designs (Table 3).
Table 3
Gene expression studies informing understanding of naturally acquired immunity to malaria infection
Measure of NAI
Publication
Design
Sample
Species
Subjects for comparison
Key finding
Comment
Prior exposure to malaria
Tran et al. (2016)
Comparison of GEP changes from paired infected and uninfected samples
Whole blood
P. falciparum
Malaria-naïve, symptomatic Dutch CHMI volunteers at diagnosis (n = 5)
Malaria experienced Malian children (> 13 years) and adults infected in the field (n = 8)
Graded activation of pathways of downstream proinflammatory cytokines with highest activation in malaria-naive subjects and significantly reduced activation in malaria experienced Malians
 
Ockenhouse et al. (2006)
Comparison of GEP changes in infection-controls samples US malaria naïve subjects
PBMC
P. falciparum
US malaria-naïve CHMI volunteers with early, blood-stage infection (n = 22)
Malaria-experienced Cameroonian adults presenting with naturally acquired febrile malaria (n = 15)
Similar induction of pro-inflammatory cytokines seen between pre-symptomatic and symptomatic individuals regardless of prior malaria exposure
 
Rojas-Pena et al. (2015) and Vallejo et al. (2018)
Comparison of GEP changes from paired infected and uninfected samples
Whole blood
P. vivax
Columbian malaria-naïve (MN) CHMI volunteers at diagnosis (n = 7)
Columbian malaria-exposed (ME) CHMI volunteers at diagnosis (n = 9)
Little differentiation seen between MN and ME populations by Rojas-Penas et al. However network co-expression analysis by Vallejo et al. showed the inflammatory response was attenuated in ME volunteers with decreased class II antigen presentation in dendritic cells
No significant difference between groups for pre-patent period or parasitaemia at diagnosis suggesting there may have been no difference in functional immunity between groups
Jagannathan et al. (2014)
Comparison of GEP between groups
Vδ2+ T cells
P. falciparum
Ugandan children with low prior malaria incidence (n = 4)
Ugandan children with low prior malaria incidence (n = 4)
Comparison of basal gene expression patterns of sorted, un-stimulated Vδ2+ T cells identified 48 differentially expressed genes, many with known roles in immunomodulation. For each of these genes, expression was higher among children with high prior exposure to malaria
Data suggest recurrent malaria infection causes up-regulation of immunoregulatory pathways that dampen the pro-inflammatory immune response to P. falciparum infection and help explain immunological tolerance to the parasite
Symptoms at diagnosis
Tran et al. (2016)
Comparison of GEP changes from paired infected and uninfected samples
Whole blood
P. falciparum
Malaria experienced Malian children (> 13 years) and adults infected in the field and asymptomatic at diagnosis (EA, n = 5)
Malaria experienced Malian children (> 13 years) and adults infected in the field and symptomatic with fever at the time of diagnosis (EF, n = 3)
Only 70 differentially expressed genes (DEGs) were identified between these groups despite the apparent clinical differences
2 of the 5 individuals in the EA group progressed to febrile malaria within 5 days of initial diagnosis by PCR
Disease severity
Krupka et al. (2012)
Comparison of GEP in same subjects at diagnosis with severe and subsequent mild malaria
Whole blood
P. falciparum
Malawian children who, after presenting with severe malaria (all had cerebral malaria), were found to have mild malaria one month later on screening by blood smear (n = 5)
Pathway analysis showed relative up regulation of Type I IFN signaling pathway, regulation of inflammation, regulation of leukocyte proliferation and T cell activation in episodes of mild malaria
 
Boldt et al. (2019)
Comparison of GEP between groups
Whole blood
P. falciparum
Healthy uninfected Gabonese children
Gabonese children with asymptomatic parasitaemia, mild malaria, malaria with severe anaemia and cerebral anaemia (0.5–6 years)
GEP of 22 genes significantly differed among groups. Immunoglobulin production, complement regulation and IFN beta signaling were most conspicuous
 
PBMC peripheral blood mononuclear cells, GEP gene expression profile, CHMI controlled human malaria infection
The findings from a number of studies supported a dampening of the innate pro-inflammatory immune response as a mechanism underpinning NAI [2426, 33] although this finding was not observed in all studies [28, 29, 31].
One study by Franklin et al. provided evidence of ‘pro-inflammatory priming’ of the innate immune system in acute malaria infection [34]. Comparison of GEP in Brazilian adults presenting with uncomplicated malaria with paired convalescent samples showed an increase in expression in genes involved in TLR signalling pathways supporting a role for TLR hyper responsiveness in the pathology of malaria infection [34, 35].
Quin et al. sought to use RNA sequencing to elucidate the mechanism driving lower infection rates, lower parasite densities and fewer symptomatic cases of P. falciparum in the population of Fulani compared to other sympatric ethnic groups [33]. Comparison of the GEP of monocytes from infected and uninfected Fulani and Mossi adults showed a marked difference, with a significantly greater number of differentially expressed (DE) genes in infected Fulani compared to infected Mossi participants (1239 versus 3 DE genes respectively). Pathway analysis showed that infected Fulani, but not infected Mossi, individuals demonstrated a marked reduction in expression of inflammasome pathway components, suggesting a blunting of the innate pro-inflammatory immune response post-infection could explain the differences in susceptibility.
Another study sought to examine the genetic basis of gene expression variation in malaria [36]. Idaghdour et al. compared GEP in children diagnosed with uncomplicated malaria (n = 94) in Benin with age matched controls (n = 64) [36] and performed a genome wide association test of transcript abundance. Testing for genotype-by-infection interactions demonstrated the existence of genome wide significant interactions and other genes subject to interaction effects beneath genome-wide significance but still likely to have important roles in modulating the course of infection. These interactions affected the complement system, antigen processing and presentation and T cell activation [36].
In work to identify a transcriptional signature to distinguish acute malaria from other febrile illnesses, Griffiths et al. compared the GEP of twenty-two Kenyan children admitted with febrile illnesses (fifteen of which had malaria infection alone) with six convalescent samples collected 2 weeks post discharge [23]. Two main GEPs relating to neutrophil and erythroid activity were shown to differentiate acutely ill and convalescent children, with significantly higher expression of genes in the neutrophil-related gene region in subjects with bacterial infections and significantly higher expression of genes related to lymphocyte and T cell activation in subjects with malaria. The authors also identified two gene profiles whose expression intensity correlated with host parasitaemia.
Only two datasets included gene expression changes following P. vivax infection [28, 30, 37]. Rojas-Penas et al. interrogated GEP changes in malaria naïve (MN) and malaria-exposed (ME) Columbian volunteers following infection with P. vivax in a CHMI setting [28]. Significant GEP changes were consistent with time-point rather than prior malaria exposure, with a decline in innate immune signalling and neutrophil number (in contrast to strong up regulation of the same genes reported by Igadour et al. [36]) and an increase in interferon induction seen at diagnosis. No significant GEP changes were noted at other time points, including those relating to the liver stage of infection. Further analysis of this dataset by Vallejo et al. using network co-expression analysis showed that while P. vivax infection induced strong inflammatory responses in all participants, the inflammatory response was attenuated with pathways associated with antigen processing and presentation less enriched in those with prior exposure to P. vivax, suggesting a more ‘tolerogenic’ immune response in these individuals [30].
In contrast to this work, Rothen et al. found that transcriptional changes post-CHMI via intradermal injection of cryopreserved P. falciparum sporozoites were most pronounced on day 5 after inoculation, during the clinically silent liver stage rather than during the blood-stage of infection [38].

Transcriptomic studies in specific cell types

Whilst the majority of studies examined the immune response from whole blood or PBMCs, some examined transcriptomic changes in other cell types or tissues [26, 33, 3945]. For example, the work of Muehlenbachs et al. with placental tissue highlighted a previously unappreciated role for B cells in chronic placental malaria [39]; whilst Sullivan et al. compared GEPs of classical and ‘atypical’ memory B cells obtained from Ugandan children showing the latter demonstrated down-regulation of B cell receptor signalling and apoptosis [43].

Discussion

GEP is a powerful tool to analyse the immune response to infection. As this review demonstrates, the application of these studies for malaria are wide-ranging, from attempts to dissect the mechanisms of NAI to improving understanding of the interaction between host genotype and infection outcome. However, as a field in its relative infancy, studies are often hypothesis generating with extremely small sample sizes. There is a lack of standardization ranging from methodological (such as sample type, RNA extraction, platform and analysis) to phenotype (including precision in disease context and immune status). This variation means interpreting published data and comparison between studies is challenging. Some of this is unavoidable, however, much could be addressed, for example by implementing standardization in blood sampling, methodological protocols for data generation and analysis with robust significance testing and approaches to confounders, use of ontologies (for example human phenotype and gene ontologies) and expert curation and annotation of data on deposition [4649].
GEP studies are well placed to examine the mechanisms of NAI and have already helped highlight the role of the innate and early adaptive immune responses [2426]. However, work has been limited by the lack of an in vitro correlate or universally accepted definition of NAI, meaning identifying the immune status of individuals or quantification of immunity is problematic [6, 50]. In field studies where the timing of infection and parasite burden and dynamics are unknown, and potentially hugely variable between individuals, only limited information can be reliably extrapolated from any GEP changes seen. Most studies assess gene expression from peripheral blood or its components, which does not provide reliable information regarding the transcriptional changes in key organs such as spleen, liver, and bone marrow. In addition, when subjects are recruited at presentation with disease, no baseline comparator data are available to use as a control. Even if a clear difference in GEP were to be reported between individuals with and without NAI, it would be near impossible to distinguish GEP changes associated with parasitaemia from those mediating immunity.
However, there is much potential for the future use of GEP studies, particularly in CHMI studies [51, 52] where the parasite burden can be pre-defined and dynamics of infection closely monitored using highly sensitive qPCR. As these studies are increasingly performed in endemic settings [5355], there will be growing opportunity to use GEP to understand detailed time-course changes in immune response, particularly at the skin, liver and pre-symptomatic blood-stage, which to date have been difficult to study in human subjects infected in the field.

Conclusion

GEP in malaria is a potentially powerful tool, but to date studies have been hypothesis generating with small sample sizes and widely varying methodology. As CHMI studies are increasingly performed in endemic settings, there will be growing opportunity to use GEP to understand detailed time-course changes in host response and understand in greater detail the mechanisms of NAI.

Supplementary information

Supplementary information accompanies this paper at https://​doi.​org/​10.​1186/​s12936-019-3035-0.

Acknowledgements

Not applicable.
Not applicable.
Not applicable.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat White NJ, Pukrittayakamee S, Hien TT, Faiz MA, Mokuolu OA, Dondorp AM. Malaria. Lancet. 2014;383:723–35.PubMedCrossRef White NJ, Pukrittayakamee S, Hien TT, Faiz MA, Mokuolu OA, Dondorp AM. Malaria. Lancet. 2014;383:723–35.PubMedCrossRef
2.
Zurück zum Zitat Cockburn IA, Seder RA. Malaria prevention: from immunological concepts to effective vaccines and protective antibodies. Nat Immunol. 2018;19:1199–211.PubMedCrossRef Cockburn IA, Seder RA. Malaria prevention: from immunological concepts to effective vaccines and protective antibodies. Nat Immunol. 2018;19:1199–211.PubMedCrossRef
3.
Zurück zum Zitat WHO. World malaria report. Geneva: World Health Organization; 2018. WHO. World malaria report. Geneva: World Health Organization; 2018.
4.
Zurück zum Zitat Cohen S, Mc GI, Carrington S. Gamma-globulin and acquired immunity to human malaria. Nature. 1961;192:733–7.PubMedCrossRef Cohen S, Mc GI, Carrington S. Gamma-globulin and acquired immunity to human malaria. Nature. 1961;192:733–7.PubMedCrossRef
5.
Zurück zum Zitat Sabchareon A, Burnouf T, Ouattara D, et al. Parasitologic and clinical human response to immunoglobulin administration in falciparum malaria. Am J Trop Med Hyg. 1991;45:297–308.PubMedCrossRef Sabchareon A, Burnouf T, Ouattara D, et al. Parasitologic and clinical human response to immunoglobulin administration in falciparum malaria. Am J Trop Med Hyg. 1991;45:297–308.PubMedCrossRef
8.
Zurück zum Zitat Schulze A, Downward J. Navigating gene expression using microarrays—a technology review. Nat Cell Biol. 2001;3:E190–5.PubMedCrossRef Schulze A, Downward J. Navigating gene expression using microarrays—a technology review. Nat Cell Biol. 2001;3:E190–5.PubMedCrossRef
10.
Zurück zum Zitat Papalexi E, Satija R. Single-cell RNA sequencing to explore immune cell heterogeneity. Nat Rev Immunol. 2018;18:35–45.PubMedCrossRef Papalexi E, Satija R. Single-cell RNA sequencing to explore immune cell heterogeneity. Nat Rev Immunol. 2018;18:35–45.PubMedCrossRef
11.
Zurück zum Zitat Querec TD, Akondy RS, Lee EK, Cao W, Nakaya HI, Teuwen D, et al. Systems biology approach predicts immunogenicity of the yellow fever vaccine in humans. Nat Immunol. 2009;10:116–25.PubMedCrossRef Querec TD, Akondy RS, Lee EK, Cao W, Nakaya HI, Teuwen D, et al. Systems biology approach predicts immunogenicity of the yellow fever vaccine in humans. Nat Immunol. 2009;10:116–25.PubMedCrossRef
13.
Zurück zum Zitat Rappuoli R, Aderem A. A 2020 vision for vaccines against HIV, tuberculosis and malaria. Nature. 2011;473:463–9.PubMedCrossRef Rappuoli R, Aderem A. A 2020 vision for vaccines against HIV, tuberculosis and malaria. Nature. 2011;473:463–9.PubMedCrossRef
14.
Zurück zum Zitat Hogan CM, Hammer SM. Host determinants in HIV infection and disease. Part 1: cellular and humoral immune responses. Ann Intern Med. 2001;134:761–76.PubMedCrossRef Hogan CM, Hammer SM. Host determinants in HIV infection and disease. Part 1: cellular and humoral immune responses. Ann Intern Med. 2001;134:761–76.PubMedCrossRef
15.
Zurück zum Zitat Blohmke CJ, Darton TC, Jones C, Suarez NM, Waddington CS, Angus B, et al. Interferon-driven alterations of the host’s amino acid metabolism in the pathogenesis of typhoid fever. J Exp Med. 2016;213:1061–77.PubMedPubMedCentralCrossRef Blohmke CJ, Darton TC, Jones C, Suarez NM, Waddington CS, Angus B, et al. Interferon-driven alterations of the host’s amino acid metabolism in the pathogenesis of typhoid fever. J Exp Med. 2016;213:1061–77.PubMedPubMedCentralCrossRef
17.
Zurück zum Zitat Anderson ST, Kaforou M, Brent AJ, Wright VJ, Banwell CM, Chagaluka G, et al. Diagnosis of childhood tuberculosis and host RNA expression in Africa. N Engl J Med. 2014;370:1712–23.PubMedPubMedCentralCrossRef Anderson ST, Kaforou M, Brent AJ, Wright VJ, Banwell CM, Chagaluka G, et al. Diagnosis of childhood tuberculosis and host RNA expression in Africa. N Engl J Med. 2014;370:1712–23.PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Mejias A, Ramilo O. Transcriptional profiling in infectious diseases: ready for prime time? J Infect. 2014;68(Suppl 1):S94–9.PubMedCrossRef Mejias A, Ramilo O. Transcriptional profiling in infectious diseases: ready for prime time? J Infect. 2014;68(Suppl 1):S94–9.PubMedCrossRef
19.
Zurück zum Zitat Berry MP, Graham CM, McNab FW, Xu Z, Bloch SA, Oni T, et al. An interferon-inducible neutrophil-driven blood transcriptional signature in human tuberculosis. Nature. 2010;466:973–7.PubMedPubMedCentralCrossRef Berry MP, Graham CM, McNab FW, Xu Z, Bloch SA, Oni T, et al. An interferon-inducible neutrophil-driven blood transcriptional signature in human tuberculosis. Nature. 2010;466:973–7.PubMedPubMedCentralCrossRef
20.
Zurück zum Zitat Westermann AJ, Forstner KU, Amman F, Barquist L, Chao Y, Schulte LN, et al. Dual RNA-seq unveils noncoding RNA functions in host–pathogen interactions. Nature. 2016;529:496–501.PubMedCrossRef Westermann AJ, Forstner KU, Amman F, Barquist L, Chao Y, Schulte LN, et al. Dual RNA-seq unveils noncoding RNA functions in host–pathogen interactions. Nature. 2016;529:496–501.PubMedCrossRef
22.
23.
Zurück zum Zitat Griffiths MJ, Shafi MJ, Popper SJ, Hemingway CA, Kortok MM, Wathen A, et al. Genomewide analysis of the host response to malaria in Kenyan children. J Infect Dis. 2005;191:1599–611.PubMedCrossRef Griffiths MJ, Shafi MJ, Popper SJ, Hemingway CA, Kortok MM, Wathen A, et al. Genomewide analysis of the host response to malaria in Kenyan children. J Infect Dis. 2005;191:1599–611.PubMedCrossRef
24.
Zurück zum Zitat Krupka M, Seydel K, Feintuch CM, Yee K, Kim R, Lin CY, et al. Mild Plasmodium falciparum malaria following an episode of severe malaria is associated with induction of the interferon pathway in Malawian children. Infect Immun. 2012;80:1150–5.PubMedPubMedCentralCrossRef Krupka M, Seydel K, Feintuch CM, Yee K, Kim R, Lin CY, et al. Mild Plasmodium falciparum malaria following an episode of severe malaria is associated with induction of the interferon pathway in Malawian children. Infect Immun. 2012;80:1150–5.PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Tran TM, Jones MB, Ongoiba A, Bijker EM, Schats R, Venepally P, et al. Transcriptomic evidence for modulation of host inflammatory responses during febrile Plasmodium falciparum malaria. Sci Rep. 2016;6:31291.PubMedPubMedCentralCrossRef Tran TM, Jones MB, Ongoiba A, Bijker EM, Schats R, Venepally P, et al. Transcriptomic evidence for modulation of host inflammatory responses during febrile Plasmodium falciparum malaria. Sci Rep. 2016;6:31291.PubMedPubMedCentralCrossRef
26.
Zurück zum Zitat Jagannathan P, Kim CC, Greenhouse B, Nankya F, Bowen K, Eccles-James I, et al. Loss and dysfunction of Vdelta2(+) gammadelta T cells are associated with clinical tolerance to malaria. Sci Transl Med. 2014;6:251ra117.PubMedPubMedCentralCrossRef Jagannathan P, Kim CC, Greenhouse B, Nankya F, Bowen K, Eccles-James I, et al. Loss and dysfunction of Vdelta2(+) gammadelta T cells are associated with clinical tolerance to malaria. Sci Transl Med. 2014;6:251ra117.PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Barrett T, Wilhite SE, Ledoux P, Evangelista C, Kim IF, Tomashevsky M, et al. NCBI GEO: archive for functional genomics data sets-update. Nucleic Acids Res. 2013;41:D991–5.PubMedCrossRef Barrett T, Wilhite SE, Ledoux P, Evangelista C, Kim IF, Tomashevsky M, et al. NCBI GEO: archive for functional genomics data sets-update. Nucleic Acids Res. 2013;41:D991–5.PubMedCrossRef
28.
Zurück zum Zitat Rojas-Pena ML, Vallejo A, Herrera S, Gibson G, Arevalo-Herrera M. Transcription profiling of malaria-naive and semi-immune Colombian volunteers in a Plasmodium vivax sporozoite challenge. PLoS Negl Trop Dis. 2015;9:e0003978.PubMedPubMedCentralCrossRef Rojas-Pena ML, Vallejo A, Herrera S, Gibson G, Arevalo-Herrera M. Transcription profiling of malaria-naive and semi-immune Colombian volunteers in a Plasmodium vivax sporozoite challenge. PLoS Negl Trop Dis. 2015;9:e0003978.PubMedPubMedCentralCrossRef
29.
Zurück zum Zitat Ockenhouse CF, Hu WC, Kester KE, Cummings JF, Stewart A, Heppner DG, et al. Common and divergent immune response signaling pathways discovered in peripheral blood mononuclear cell gene expression patterns in presymptomatic and clinically apparent malaria. Infect Immun. 2006;74:5561–73.PubMedPubMedCentralCrossRef Ockenhouse CF, Hu WC, Kester KE, Cummings JF, Stewart A, Heppner DG, et al. Common and divergent immune response signaling pathways discovered in peripheral blood mononuclear cell gene expression patterns in presymptomatic and clinically apparent malaria. Infect Immun. 2006;74:5561–73.PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Vallejo AF, Read RC, Arevalo-Herrera M, Herrera S, Elliott T, Polak ME. Malaria systems immunology: Plasmodium vivax induces tolerance during primary infection through dysregulation of neutrophils and dendritic cells. J Infect. 2018;77:440–7.PubMedPubMedCentralCrossRef Vallejo AF, Read RC, Arevalo-Herrera M, Herrera S, Elliott T, Polak ME. Malaria systems immunology: Plasmodium vivax induces tolerance during primary infection through dysregulation of neutrophils and dendritic cells. J Infect. 2018;77:440–7.PubMedPubMedCentralCrossRef
31.
Zurück zum Zitat Boldt ABW, van Tong H, Grobusch MP, Kalmbach Y, Dzeing Ella A, Kombila M, et al. The blood transcriptome of childhood malaria. EBioMedicine. 2019;40:614–25.PubMedPubMedCentralCrossRef Boldt ABW, van Tong H, Grobusch MP, Kalmbach Y, Dzeing Ella A, Kombila M, et al. The blood transcriptome of childhood malaria. EBioMedicine. 2019;40:614–25.PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Subramaniam KS, Spaulding E, Ivan E, Mutimura E, Kim RS, Liu X, et al. The T-cell inhibitory molecule Butyrophilin-Like 2 is up-regulated in mild Plasmodium falciparum infection and is protective during experimental cerebral malaria. J Infect Dis. 2015;212:1322–31.PubMedPubMedCentralCrossRef Subramaniam KS, Spaulding E, Ivan E, Mutimura E, Kim RS, Liu X, et al. The T-cell inhibitory molecule Butyrophilin-Like 2 is up-regulated in mild Plasmodium falciparum infection and is protective during experimental cerebral malaria. J Infect Dis. 2015;212:1322–31.PubMedPubMedCentralCrossRef
33.
Zurück zum Zitat Quin JE, Bujila I, Cherif M, Sanou GS, Qu Y, Vafa Homann M, et al. Major transcriptional changes observed in the Fulani, an ethnic group less susceptible to malaria. Elife. 2017;6:e29156.PubMedPubMedCentralCrossRef Quin JE, Bujila I, Cherif M, Sanou GS, Qu Y, Vafa Homann M, et al. Major transcriptional changes observed in the Fulani, an ethnic group less susceptible to malaria. Elife. 2017;6:e29156.PubMedPubMedCentralCrossRef
34.
Zurück zum Zitat Franklin BS, Parroche P, Ataide MA, Lauw F, Ropert C, de Oliveira RB, et al. Malaria primes the innate immune response due to interferon-gamma induced enhancement of toll-like receptor expression and function. Proc Natl Acad Sci USA. 2009;106:5789–94.PubMedCrossRefPubMedCentral Franklin BS, Parroche P, Ataide MA, Lauw F, Ropert C, de Oliveira RB, et al. Malaria primes the innate immune response due to interferon-gamma induced enhancement of toll-like receptor expression and function. Proc Natl Acad Sci USA. 2009;106:5789–94.PubMedCrossRefPubMedCentral
35.
Zurück zum Zitat Sharma S, DeOliveira RB, Kalantari P, Parroche P, Goutagny N, Jiang Z, et al. Innate immune recognition of an AT-rich stem-loop DNA motif in the Plasmodium falciparum genome. Immunity. 2011;35:194–207.PubMedPubMedCentralCrossRef Sharma S, DeOliveira RB, Kalantari P, Parroche P, Goutagny N, Jiang Z, et al. Innate immune recognition of an AT-rich stem-loop DNA motif in the Plasmodium falciparum genome. Immunity. 2011;35:194–207.PubMedPubMedCentralCrossRef
36.
Zurück zum Zitat Idaghdour Y, Quinlan J, Goulet JP, Berghout J, Gbeha E, Bruat V, et al. Evidence for additive and interaction effects of host genotype and infection in malaria. Proc Natl Acad Sci USA. 2012;109:16786–93.PubMedCrossRefPubMedCentral Idaghdour Y, Quinlan J, Goulet JP, Berghout J, Gbeha E, Bruat V, et al. Evidence for additive and interaction effects of host genotype and infection in malaria. Proc Natl Acad Sci USA. 2012;109:16786–93.PubMedCrossRefPubMedCentral
37.
Zurück zum Zitat Gardinassi LG, Arevalo-Herrera M, Herrera S, Cordy RJ, Tran V, Smith MR, et al. Integrative metabolomics and transcriptomics signatures of clinical tolerance to Plasmodium vivax reveal activation of innate cell immunity and T cell signaling. Redox Biol. 2018;17:158–70.PubMedPubMedCentralCrossRef Gardinassi LG, Arevalo-Herrera M, Herrera S, Cordy RJ, Tran V, Smith MR, et al. Integrative metabolomics and transcriptomics signatures of clinical tolerance to Plasmodium vivax reveal activation of innate cell immunity and T cell signaling. Redox Biol. 2018;17:158–70.PubMedPubMedCentralCrossRef
38.
Zurück zum Zitat Rothen J, Murie C, Carnes J, Anupama A, Abdulla S, Chemba M, et al. Whole blood transcriptome changes following controlled human malaria infection in malaria pre-exposed volunteers correlate with parasite prepatent period. PLoS ONE. 2018;13:e0199392.PubMedPubMedCentralCrossRef Rothen J, Murie C, Carnes J, Anupama A, Abdulla S, Chemba M, et al. Whole blood transcriptome changes following controlled human malaria infection in malaria pre-exposed volunteers correlate with parasite prepatent period. PLoS ONE. 2018;13:e0199392.PubMedPubMedCentralCrossRef
39.
Zurück zum Zitat Muehlenbachs A, Fried M, Lachowitzer J, Mutabingwa TK, Duffy PE. Genome-wide expression analysis of placental malaria reveals features of lymphoid neogenesis during chronic infection. J Immunol. 2007;179:557–65.PubMedCrossRef Muehlenbachs A, Fried M, Lachowitzer J, Mutabingwa TK, Duffy PE. Genome-wide expression analysis of placental malaria reveals features of lymphoid neogenesis during chronic infection. J Immunol. 2007;179:557–65.PubMedCrossRef
40.
Zurück zum Zitat Burel JG, Apte SH, Groves PL, McCarthy JS, Doolan DL. Polyfunctional and IFN-gamma monofunctional human CD4+ T cell populations are molecularly distinct. JCI Insight. 2017;2:e87499.PubMedPubMedCentralCrossRef Burel JG, Apte SH, Groves PL, McCarthy JS, Doolan DL. Polyfunctional and IFN-gamma monofunctional human CD4+ T cell populations are molecularly distinct. JCI Insight. 2017;2:e87499.PubMedPubMedCentralCrossRef
41.
Zurück zum Zitat Jaijyan DK, Singh H, Singh AP. A sporozoite- and liver stage-expressed tryptophan-rich protein plays an auxiliary role in Plasmodium liver stage development and is a potential vaccine candidate. J Biol Chem. 2015;290:19496–511.PubMedPubMedCentralCrossRef Jaijyan DK, Singh H, Singh AP. A sporozoite- and liver stage-expressed tryptophan-rich protein plays an auxiliary role in Plasmodium liver stage development and is a potential vaccine candidate. J Biol Chem. 2015;290:19496–511.PubMedPubMedCentralCrossRef
42.
Zurück zum Zitat Terkawi MA, Takano R, Furukawa A, Murakoshi F, Kato K. Involvement of beta-defensin 130 (DEFB130) in the macrophage microbicidal mechanisms for killing Plasmodium falciparum. Sci Rep. 2017;7:41772.PubMedPubMedCentralCrossRef Terkawi MA, Takano R, Furukawa A, Murakoshi F, Kato K. Involvement of beta-defensin 130 (DEFB130) in the macrophage microbicidal mechanisms for killing Plasmodium falciparum. Sci Rep. 2017;7:41772.PubMedPubMedCentralCrossRef
43.
Zurück zum Zitat Sullivan RT, Kim CC, Fontana MF, Feeney ME, Jagannathan P, Boyle MJ, et al. FCRL5 delineates functionally impaired memory B cells associated with Plasmodium falciparum exposure. PLoS Pathog. 2015;11:e1004894.PubMedPubMedCentralCrossRef Sullivan RT, Kim CC, Fontana MF, Feeney ME, Jagannathan P, Boyle MJ, et al. FCRL5 delineates functionally impaired memory B cells associated with Plasmodium falciparum exposure. PLoS Pathog. 2015;11:e1004894.PubMedPubMedCentralCrossRef
44.
Zurück zum Zitat Terkawi MA, Takano R, Kato K. Differential gene expression profile of human neutrophils cultured with Plasmodium falciparum-parasitized erythrocytes. J Immunol Res. 2018;2018:6709424.PubMedPubMedCentralCrossRef Terkawi MA, Takano R, Kato K. Differential gene expression profile of human neutrophils cultured with Plasmodium falciparum-parasitized erythrocytes. J Immunol Res. 2018;2018:6709424.PubMedPubMedCentralCrossRef
45.
Zurück zum Zitat Portugal S, Tipton CM, Sohn H, Kone Y, Wang J, Li S, et al. Malaria-associated atypical memory B cells exhibit markedly reduced B cell receptor signaling and effector function. Elife. 2015;4:e07218.PubMedCentralCrossRef Portugal S, Tipton CM, Sohn H, Kone Y, Wang J, Li S, et al. Malaria-associated atypical memory B cells exhibit markedly reduced B cell receptor signaling and effector function. Elife. 2015;4:e07218.PubMedCentralCrossRef
46.
Zurück zum Zitat Bammler T, Beyer RP, Bhattacharya S, Boorman GA, Boyles A, Bradford BU, et al. Standardizing global gene expression analysis between laboratories and across platforms. Nat Methods. 2005;2:351–6.PubMedCrossRef Bammler T, Beyer RP, Bhattacharya S, Boorman GA, Boyles A, Bradford BU, et al. Standardizing global gene expression analysis between laboratories and across platforms. Nat Methods. 2005;2:351–6.PubMedCrossRef
47.
Zurück zum Zitat Conesa A, Madrigal P, Tarazona S, Gomez-Cabrero D, Cervera A, McPherson A, et al. A survey of best practices for RNA-seq data analysis. Genome Biol. 2016;17:13.PubMedPubMedCentralCrossRef Conesa A, Madrigal P, Tarazona S, Gomez-Cabrero D, Cervera A, McPherson A, et al. A survey of best practices for RNA-seq data analysis. Genome Biol. 2016;17:13.PubMedPubMedCentralCrossRef
49.
Zurück zum Zitat Rung J, Brazma A. Reuse of public genome-wide gene expression data. Nat Rev Genet. 2013;14:89–99.PubMedCrossRef Rung J, Brazma A. Reuse of public genome-wide gene expression data. Nat Rev Genet. 2013;14:89–99.PubMedCrossRef
50.
Zurück zum Zitat Langhorne J, Ndungu FM, Sponaas AM, Marsh K. Immunity to malaria: more questions than answers. Nat Immunol. 2008;9:725–32.PubMedCrossRef Langhorne J, Ndungu FM, Sponaas AM, Marsh K. Immunity to malaria: more questions than answers. Nat Immunol. 2008;9:725–32.PubMedCrossRef
52.
Zurück zum Zitat Stanisic DI, McCarthy JS, Good MF. Controlled human malaria infection: applications, advances, and challenges. Infect Immun. 2018;86:e00479.PubMed Stanisic DI, McCarthy JS, Good MF. Controlled human malaria infection: applications, advances, and challenges. Infect Immun. 2018;86:e00479.PubMed
53.
Zurück zum Zitat Lell B, Mordmuller B, Dejon Agobe JC, Honkpehedji J, Zinsou J, Mengue JB, et al. Impact of sickle cell trait and naturally acquired immunity on uncomplicated malaria after controlled human malaria infection in adults in Gabon. Am J Trop Med Hyg. 2018;98:508–15.PubMedCrossRef Lell B, Mordmuller B, Dejon Agobe JC, Honkpehedji J, Zinsou J, Mengue JB, et al. Impact of sickle cell trait and naturally acquired immunity on uncomplicated malaria after controlled human malaria infection in adults in Gabon. Am J Trop Med Hyg. 2018;98:508–15.PubMedCrossRef
54.
Zurück zum Zitat Hodgson SH, Juma E, Salim A, Magiri C, Kimani D, Njenga D, et al. Evaluating controlled human malaria infection in Kenyan adults with varying degrees of prior exposure to Plasmodium falciparum using sporozoites administered by intramuscular injection. Front Microbiol. 2014;5:686.PubMedPubMedCentralCrossRef Hodgson SH, Juma E, Salim A, Magiri C, Kimani D, Njenga D, et al. Evaluating controlled human malaria infection in Kenyan adults with varying degrees of prior exposure to Plasmodium falciparum using sporozoites administered by intramuscular injection. Front Microbiol. 2014;5:686.PubMedPubMedCentralCrossRef
55.
Zurück zum Zitat Shekalaghe S, Rutaihwa M, Billingsley PF, Chemba M, Daubenberger CA, James ER, et al. Controlled human malaria infection of Tanzanians by intradermal injection of aseptic, purified, cryopreserved Plasmodium falciparum sporozoites. Am J Trop Med Hyg. 2014;91:471–80.PubMedPubMedCentralCrossRef Shekalaghe S, Rutaihwa M, Billingsley PF, Chemba M, Daubenberger CA, James ER, et al. Controlled human malaria infection of Tanzanians by intradermal injection of aseptic, purified, cryopreserved Plasmodium falciparum sporozoites. Am J Trop Med Hyg. 2014;91:471–80.PubMedPubMedCentralCrossRef
Metadaten
Titel
Use of gene expression studies to investigate the human immunological response to malaria infection
verfasst von
Susanne H. Hodgson
Julius Muller
Helen E. Lockstone
Adrian V. S. Hill
Kevin Marsh
Simon J. Draper
Julian C. Knight
Publikationsdatum
01.12.2019
Verlag
BioMed Central
Erschienen in
Malaria Journal / Ausgabe 1/2019
Elektronische ISSN: 1475-2875
DOI
https://doi.org/10.1186/s12936-019-3035-0

Weitere Artikel der Ausgabe 1/2019

Malaria Journal 1/2019 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Reizdarmsyndrom: Diäten wirksamer als Medikamente

29.04.2024 Reizdarmsyndrom Nachrichten

Bei Reizdarmsyndrom scheinen Diäten, wie etwa die FODMAP-arme oder die kohlenhydratreduzierte Ernährung, effektiver als eine medikamentöse Therapie zu sein. Das hat eine Studie aus Schweden ergeben, die die drei Therapieoptionen im direkten Vergleich analysierte.

Notfall-TEP der Hüfte ist auch bei 90-Jährigen machbar

26.04.2024 Hüft-TEP Nachrichten

Ob bei einer Notfalloperation nach Schenkelhalsfraktur eine Hemiarthroplastik oder eine totale Endoprothese (TEP) eingebaut wird, sollte nicht allein vom Alter der Patientinnen und Patienten abhängen. Auch über 90-Jährige können von der TEP profitieren.

Niedriger diastolischer Blutdruck erhöht Risiko für schwere kardiovaskuläre Komplikationen

25.04.2024 Hypotonie Nachrichten

Wenn unter einer medikamentösen Hochdrucktherapie der diastolische Blutdruck in den Keller geht, steigt das Risiko für schwere kardiovaskuläre Ereignisse: Darauf deutet eine Sekundäranalyse der SPRINT-Studie hin.

Bei schweren Reaktionen auf Insektenstiche empfiehlt sich eine spezifische Immuntherapie

Insektenstiche sind bei Erwachsenen die häufigsten Auslöser einer Anaphylaxie. Einen wirksamen Schutz vor schweren anaphylaktischen Reaktionen bietet die allergenspezifische Immuntherapie. Jedoch kommt sie noch viel zu selten zum Einsatz.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.