Summary
The complexity of the Plasmodium parasite and its life cycle poses a challenge to our understanding of the host immune response against malaria. Studying human immune responses during natural and experimental Plasmodium infections can enhance our understanding of malaria-protective immunity and inform the design of disease-modifying adjunctive therapies and next-generation malaria vaccines. Systems immunology can complement conventional approaches to facilitate our understanding of the complex immune response to the highly dynamic malaria parasite. In this review, recent studies that used systems-based approaches to evaluate human immune responses during natural and experimental P. falciparum and P. vivax infections as well as during immunization with candidate malaria vaccines are summarized and related to each other. The potential for next-generation technologies to address the current limitations of systems-based studies of human malaria are discussed.
Keywords: malaria immunity, malaria immunology, host response, systems biology, systems immunology, systems vaccinology, transcriptomics, gene expression profiling, microarray analysis, Plasmodium falciparum, Plasmodium vivax
“The processes of disease are so complex that it is excessively difficult to search out the laws which control them, and, although we have seen a complete revolution in our ideas, what has been accomplished by the new school of medicine is only an earnest of what the future has in store.”
Sir William Osler
INTRODUCTION
Clinical malaria encompasses a spectrum of syndromes that initially manifest as the intermittent fevers and rigors typical of “uncomplicated” malaria, which if not promptly treated, can rapidly progress to severe disease, most notably the devastating neurological complications of cerebral malaria. Although much progress has been made towards reducing malaria morbidity and mortality over the last century, over 200 million people still suffered from the disease in 2017, leading to ~435,000 deaths1. Among the human infective Plasmodium species, Plasmodium falciparum is the most prevalent malaria parasite in Africa and much of Asia and is responsible for cerebral malaria and the overwhelming majority of malaria deaths; however, Plasmodium vivax also causes significant morbidity, being widely distributed across the Asia Pacific and the Americas. Control of the female anopheline mosquito, the vector of human malaria, and prompt access to malaria diagnosis and effective anti-malaria treatment have played key roles in reducing the global malaria burden and eliminating malaria in several countries2. However, further progress towards malaria eradication will require additional tools, such as vaccines that induce durable protection3. It is also critical to develop effective adjunctive therapies to improve survival during severe malaria before eradication is achieved4. The development of these tools would be aided by a more complete understanding of the processes that govern the balance between protective immunity and immunopathogenesis during malaria infection. Thus, studying human immune responses during natural and experimental Plasmodium infections can enhance our understanding of malaria-protective immunity and inform the design of disease-modifying adjunctive therapies and next-generation malaria vaccines.
However, the complexity of the Plasmodium parasite and its life cycle poses a challenge to our understanding of the host immune response against this organism. Plasmodium is an intracellular pathogen that directly invades two cell types in humans: liver cells (hepatocytes) and red blood cells (erythrocytes). After entering the human host as sporozoites via the bite of an infected Anopheles mosquito, Plasmodium parasites initiate the liver stage of infection by invading hepatocytes where they develop and replicate for ~7 days without causing symptoms. The parasites then rupture from hepatocytes into the bloodstream as merozoites and rapidly invade erythrocytes. Within erythrocytes, the parasite develops and replicates over 48 hours before rupturing out as daughter merozoites, each capable of invading a new erythrocyte. Blood-stage infection that begins as low-level, asymptomatic parasitemia can rapidly progress to mild or severe malaria or persist as a chronic, asymptomatic infection depending on the balance between protective and pathologic host immune responses, and possibly parasite factors that remain poorly understood. Not only does the parasite exist in multiple forms within the human host, but in the case of P. falciparum, also expresses over 5,000 protein-coding genes5, many of which are highly polymorphic, presumably due to immune selective pressure6,7. Thus, a strategy to comprehensively understand the host immune response to natural malaria infection would have to account for the diverse clinical phenotypes in the context of multiple forms of the parasite, each expressing a potentially unique set of polymorphic antigens.
Systems biology can complement conventional approaches to broaden our understanding of the complex immune response to the highly dynamic malaria parasite. Although systems biology has been defined a myriad of ways, it is generally described as an interdisciplinary approach that aims to unravel complex biological phenomena both conceptually and quantitatively via the integrated analysis of large-scale experimental data using computational and statistical methodologies, which may permit the accurate simulation of biological interactions and prediction of relevant outcomes8. In contrast to reductionist approaches, which attempt to dissect individual components of a system to address defined hypotheses, systems biology evaluates components within the context of the whole system or organism to address more general hypotheses while potentially generating more specific hypotheses. As a sub-specialization within systems biology, systems immunology attempts to integrate the cellular and molecular components of the immune system by applying computational methods to high-dimension data acquired from well-designed observational or experimental studies to better comprehend their interactions and function, especially in the context of immune perturbations such as infection or vaccination. Although Plasmodium parasites can directly affect several organs and tissues, including the liver, spleen, bone marrow and vascular endothelium, ongoing infections replicate within erythrocytes that predominantly circulate in the peripheral blood where they interact with host immune cells and molecules9, making the interrogation of immunological responses possible even in pediatric studies conducted in malaria-endemic settings. Thus, computational approaches employed in malaria systems immunology studies to date (Table 1) have generally been applied to data generated by -omics analyses of peripheral blood components: ex vivo whole blood, cryopreserved peripheral blood mononuclear cells (PBMCs), PBMCs re-stimulated with parasite antigen in vitro, and cryopreserved serum or plasma.
Table 1.
Objective | Methodology or Tool | Applicable to | Reference(s) |
---|---|---|---|
Determine differences between phenotypes or groups | Cufflinks/Cuffdiff | RNA-seq | Trapnell et al. 201210 |
DESeq2 | RNA-seq | Love et al. 201411 | |
edgeR | RNA-seq | Robinson et al. 201012 | |
empirical Bayes moderated t test (limma) | microarrays | Ritchie et al. 201513 | |
Power Law Global Error Model (PLGEM) | microarrays | Pavelka 200414 | |
significance analysis of microarrays (SAM) | microarrays | Tusher 200115 | |
voom-limma | RNA-seq | Ritchie et al 201513 | |
Deconvolution of transcriptomic data from heterogeneous tissue | Immune Response in Silico (IRIS) | transcriptomic datasets | Abbas et al. 200916 |
immunoStates | transcriptomic datasets | Vallania et al. 201817 | |
leukocyte signature matrix (LM22; CIBERSORT) | transcriptomic datasets | Newman et al. 201518 | |
Functional and pathways analysis | Camera | molecular -omic datasets | Wu and Smyth 201219 |
DAVID functional annotation | molecular -omic datasets | Huang et al. 200720,21 | |
Gene set enrichment analysis | molecular -omic datasets | Subramanian et al. 200522 | |
Ingenuity (for pathways analysis; proprietary) | molecular -omic datasets | Kramer et al. 2014 23 | |
Coincident extreme ranks in numerical observations (CERNO; implemented via tmod) | molecular -omic datasets | Zyla et al. 201924 | |
Gene Correlation Network analysis | weighted gene co-expression network analysis (WGCNA) | molecular -omic datasets | Langfelder and Horvath 200825 |
Integrative analysis with Plasmodium genome or proteome; protein arrays for antibody profiling | PlasmoDB Plasmodium genomics resource | Plasmodium -omic datasets | Aurrecoechea et al. 200926 |
Gene sets and transcriptional modules | blood transcription modules | transcriptomic datasets | Li et al. 201427 |
ImmuneSigDB (C7 collection) | transcriptomic datasets | Godec et al. 201628 | |
Kyoto Encyclopedia of Genes and Genomes (KEGG) | molecular -omic datasets | Kanehisa and Goto 200029 | |
modular transcriptional repertoire | transcriptomic datasets | Chaussabel et al. 201430 | |
Molecular signatures database (MSigDB) | molecular -omic datasets | Liberzon et al. 201131 | |
Platforms with accessible graphic user interfaces | DAVID Bioinformatics Resources 6.8 (https://david.ncifcrf.gov/home.jsp) | molecular -omic datasets | Huang et al. 200932 |
Galaxy (https://galaxyproject.org) | molecular -omic datasets | Afgan et al. 201633 | |
Gene set enrichment analysis (http://software.broadinstitute.org/gsea/index.jsp) | molecular -omic datasets | Subramanian et al. 200522 | |
Ingenuity (for pathways analysis; proprietary) | molecular -omic datasets | Kramer et al. 2014 23 | |
tmod (http://tmod.online/) | transcriptomic datasets | Zyla et al. 201924 |
Here, we review recent studies that used systems immunology approaches to evaluate human immune responses during P. falciparum and P. vivax infections and during immunization with candidate malaria vaccines. We will first discuss controlled human malaria infections (CHMI) in which individuals are infected with sporozoites either by the bite of mosquitoes or by direct venous inoculation, resulting in blood-stage infections approximately one week later. We next highlight recent systems vaccinology studies that assessed molecular signatures of immunogenicity and protection before and after immunization and challenge. Lastly, we discuss systems immunology studies of naturally transmitted Plasmodium infections that cover the spectrum of disease severity, from asymptomatic parasitemia to uncomplicated and severe malaria syndromes, including cerebral malaria and severe malaria anemia. The majority of these studies applied transcriptomic analysis of peripheral blood cells to assess differences in malaria-related clinical phenotypes or associated outcomes. A few notable studies further captured the complexity of the immune response by integrating transcriptomic analysis with data from genomic, parasitological, cellular, serological and/or functional assessments to gain further insight into malaria pathogenesis and immunity. We examine and summarize these recent studies in relation to each other to identify unifying concepts that might aid in our understanding of malaria, while also illuminating gaps in knowledge deserving of further investigation. Studies highlighted in this review are tabulated along with additional studies that applied transcriptomics, metabolomics, proteomics, or Plasmodium-specific antibody profiling by protein microarray to different clinical phenotypes associated with malaria (Table 2). Systems-based studies focusing primarily on mouse and non-human primate models of malaria are beyond the scope of this review, but their importance as model systems have recently been discussed by others34,35.
Table 2.
Study | Year |
Plasmodium species |
Comparison groups |
Study design |
Immune status |
Age | Primary platform |
---|---|---|---|---|---|---|---|
Arévalo-Herrera et al.36 | 2016 | Plasmodium vivax | asymptomatic infection and uncomplicated malaria | CHMI | naïve or semi-immune | adults | protein microarrays |
Bediako et al.37 | 2019 | Plasmodium falciparum | high and low prior malaria exposure | prospective cohort | semi-immune | children | RNA-seq transcriptomics |
Boldt et al.38 | 2019 | Plasmodium falciparum | asymptomatic infection, uncomplicated malaria, severe malarial anemia, and cerebral malaria | cross-sectional | semi-immune | children | microarray transcriptomics |
Chakraborty et al.39 | 2018 | Plasmodium falciparum and Plasmodum vivax | severe and uncomplicated malaria | cross-sectional | semi-immune | adults | microarray transcriptomics |
Chakravorty et al.40 | 2007 | Plasmodium falciparum | in vitro co-culture of human umbilical vein endothelial cells with P. falciparum infected erythrocytes and uninfected erythrocytes | in vitro culture | naïve | not applicable | microarray transcriptomics |
Chattopadhyay et al.41 | 2011 | Plasmodium falciparum | in vitro P. falciparum sporozoite infected and non-infected HepG2-A16 liver cells | in vitro culture | not applicable | not applicable | microarray transcriptomics |
Chen et al.42 | 2010 | Plasmodium vivax | uncomplicated malaria | cross-sectional | semi-immune | adults | protein microarrays |
Chen et al.43 | 2015 | Plasmodium vivax | P. vivax exposed | cross-sectional | naïve or semi-immune | adults | protein microarrays |
Colborn et al.44 | 2015 | Plasmodium falciparum | uncomplicated malaria | cross-sectional | semi-immune | children | microarray transcriptomics |
Cordy et al.45 | 2019 | Plasmodium coatneyi and Plasmodium falciparum | uncomplicated malaria and chronic Plasmodium infection | cross-sectional | semi-immune | adults | plasma metabolomics in macaques with validation in humans |
Crompton et al.46 | 2010 | Plasmodium falciparum | malaria exposed | prospective cohort | semi-immune | children and adults | protein microarrays |
Dent et al.47 | 2015 | Plasmodium falciparum | malaria exposed | prospective cohort | semi-immune | children and adults | protein microarrays |
Dobbs et al.48 | 2017 | Plasmodium falciparum | uncomplicated malaria, convalescent malaria, and asymptomatic P. falciparum infection | longitudinal analysis | semi-immune | children | microarray transcriptomics |
Doolan et al.49 | 2008 | Plasmodium falciparum | volunteers immunized with radiation-attenuated P. falciparum sporozoites and naturally exposed donors | longitudinal analysis (vaccine study) and cross-sectional (natural malaria exposure) | previously malaria-naïve or semi-immune | children and adults | protein microarrays |
Dunache et al.50 | 2015 | Plasmodium falciparum | antigen-stimulated PBMCs from volunteers in either RTS,S/AS02A and thrombospondin-related adhesive protein (TRAP) malaria vaccine trials | vaccination and CHMI | previously malaria-naïve | adults | microarray transcriptomics |
Feintuch et al. 51 | 2016 | Plasmodium falciparum | retinopathy-positive and retinopathy-negative cerebral malaria | cross-sectional | semi-immune | children | microarray transcriptomics |
Felgner et al.52 | 2013 | Plasmodium falciparum | uncomplicated malaria | CHMI and natural infection | naïve or semi-immune | adults | protein microarrays |
Franklin et al.53 | 2009 | Plasmodium falciparum | uncomplicated malaria | cross-sectional | semi-immune | adults | microarray transcriptomics |
Gardinassi et al.54 | 2018 | Plasmodium vivax | pre-symptomatic and uncomplicated malaria | CHMI | naïve or semi-immune | adults | integrated metabolomics and transcriptomics |
Grangeiro de Carvalho et al.55 | 2011 | Plasmodium falciparum | in vitro co-culture of NK cells with P. falciparum infected erythrocytes and uninfected erythrocytes | in vitro co-culture | naïve | adults | microarray transcriptomics |
Grangeiro de Carvalho et al.56 | 2011 | Plasmodium falciparum | in vitro co-culture of NK92 cells cells with P. falciparum infected erythrocytes and uninfected erythrocytes | in vitro co-culture | naïve | adults | microarray transcriptomics |
Idaghdour et al.57 | 2012 | Plasmodium falciparum | uncomplicated malaria | case-control | semi-immune | children | Integrated transcriptomics and genomics |
Kamuyu et al.58 | 2018 | Plasmodium falciparum | malaria-exposed and malaria-naïve | cross-sectional | naïve or semi-immune | adults | protein microarrays |
Kassa et al.59 | 2011 | Plasmodium falciparum | uncomplicated malaria and severe malaria | cross-sectional | semi-immune | adults | metabolomics |
Kazmin et al.60 | 2017 | Plasmodium falciparum | volunteers immunized with RTS,S (RRR) and RTS,S/AS01 following a primary immunization with adenovirus 35 (Ad35) vector expressing circumsporozoite protein (ARR) | vaccination and CHMI | naïve | adults | microarray transcriptomics |
Keller et al.61 | 2009 | Plasmodium falciparum | in vitro culture of PBMCs from malaria-exposed and malaria-naïve donors with and without P. falciparum hemozoin | in vitro co-culture | naïve or semi-immune | children and adults | microarray transcriptomics |
Krupka et al.62 | 2012 | Plasmodium falciparum | uncomplicated malaria and severe malaria | longitudinal analysis | semi-immune | children | microarray transcriptomics |
Kumar et al.63 | 2018 | Plasmodium falciparum | cerebral malaria | case-control | semi-immune | children and adults | proteomics of frontal lobe samples |
Lee et al.64 | 2018 | Plasmodium falciparum | uncomplicated malaria and severe malaria | cross-sectional | semi-immune | children | whole-blood RNA-seq transcriptomics |
Leopold et al.65 | 2019 | Plasmodium falciparum | uncomplicated malaria and severe malaria | nested case-control | semi-immune | adults | plasma metabolomics with in vitro validation |
Lu et al.66 | 2014 | Plasmodium vivax | uncomplicated malaria | cross-sectional | naïve or semi-immune | adults | protein microarrays |
Nallandhighal et al.67 | 2019 | Plasmodium falciparum | cerebral malaria, severe malarial anemia, malaria exposed community controls | nested case-control | semi-immune | children | microarray transcriptomics with plasma biomarkers |
Nnedu et al.68 | 2011 | Plasmodium falciparum | malaria exposed with HIV co-infection | cross-sectional | semi-immune | adults | protein microarrays |
Ockenhouse et al.69 | 2006 | Plasmodium falciparum | pre-symptomatic infection and uncomplicated malaria | CHMI and natural infection | naïve or semi-immune | adults | microarray transcriptomics |
Orikiiriza et al.70 | 2017 | Plasmodium falciparum | uncomplicated malaria and severe malaria | cross-sectional | semi-immune | children | lipidomics |
Portugal et al.71 | 2014 | Plasmodium falciparum | in vitro co-culture of PBMCs with P. falciparum infected erythrocytes and uninfected erythrocytes | longitudinal analysis | semi-immune | children | microarray transcriptomics |
Quin et al.72 | 2017 | Plasmodium falciparum | malaria exposed | cross-sectional | semi-immune | adults | RNA-seq transcriptomics and genome-wide DNA methylation |
Ray et al.73 | 2012 | Plasmodium falciparum and Plasmodium vivax | uncomplicated malaria | cross-sectional | semi-immune | adults | proteomics |
Ray et al.74 | 2012 | Plasmodium vivax | uncomplicated malaria | cross-sectional | naïve or semi-immune | adults | proteomics |
Ray et al.75 | 2015 | Plasmodium falciparum | uncomplicated malaria and severe malaria | cross-sectional | semi-immune | adults | proteomics |
Reuterswärd et al.76 | 2018 | Plasmodium falciparum | uncomplicated malaria and severe malaria | case-control | semi-immune | children | proteomics |
Rojas-Peña et al.77 | 2015 | Plasmodium vivax | uncomplicated malaria | case-control | naïve or semi-immune | adults | quantitative RT-PCR array and RNA-seq transcriptomics |
Rojas-Peña et al.78 | 2018 | Plasmodium falciparum and Plasmodum vivax | malaria exposed | longitudinal analysis | semi-immune | adults | RNA-seq transcriptomics |
Rothen et al.79 | 2018 | Plasmodium falciparum | pre-symptomatic infection and uncomplicated malaria | CHMI | semi-immune | adults | RNA-seq transcriptomics |
Torres et al.80 | 2015 | Plasmodium falciparum | uncomplicated malaria and asymptomatic infection | cross-sectional | naïve or semi-immune | adults | protein microarrays |
Tran et al.81 | 2016 | Plasmodium falciparum | uncomplicated malaria and asymptomatic infection | CHMI and natural infection | naïve or semi-immune | adults | RNA-seq transcriptomics |
Tran et al.82 | 2019 | Plasmodium falciparum | volunteers immunized with sporozoites under chemoprophylaxis (CPS) | vaccination and CHMI | naïve | adults | RNA-seq transcriptomics |
Tran et al.83 | 2019 | Plasmodium falciparum | asymptomatic infection and uncomplicated malaria | nested case-control in a prospective study | semi-immune | children | RNA-seq transcriptomics |
Tripathi et al.84 | 2009 | Plasmodium falciparum | in vitro co-culture of human brain microvascular endothelial cells with P. falciparum infected erythrocytes and uninfected erythrocytes | in vitro co-culture | naïve | not applicable | microarray transcriptomics |
Uplekar et al.85 | 2017 | Plasmodium falciparum and Plasmodum vivax | asymptomatic infection, uncomplicated malaria | cross-sectional | semi-immune | adults | protein microarrays |
Vahey et al.86 | 2010 | Plasmodium falciparum | volunteers immunized with the RTS,S malaria vaccine | vaccination and CHMI | previously malaria-naïve | adults | microarray transcriptomics |
Vallejo et al.87 | 2018 | Plasmodium vivax | pre-symptomatic and uncomplicated malaria | CHMI | naïve or semi-immune | adults | RNA-seq transcriptomics |
van den Berg et al.88 | 2017 | Plasmodium falciparum | volunteers immunized with the RTS,S malaria vaccine | vaccination and CHMI | naïve | adults | microarray transcriptomics |
Yamagishi et al.89 | 2014 | Plasmodium falciparum | uncomplicated malaria | cross-sectional | semi-immune | children and adults | RNA-seq transcriptomics |
Blood transcriptomic assessments of the early host response to controlled human malaria infections
Experimental malaria infections began in the early 1900s when Austrian physician Julius Wagner-Jauregg inoculated patients with malaria-infected blood to induce fever as a treatment for neurosyphilis, which was at that time a terminal illness90. Although penicillin would eventually supplant “malariotherapy” as a more effective and safer therapy for syphilis91, experimental malaria infection of humans has remained an invaluable tool for evaluating the in vivo efficacy of anti-malarial drugs and vaccines92 and–with the adoption of standardized safety procedures–is now referred to as controlled human malaria infection (CHMI)93. CHMI allows for high resolution temporal assessments of parasite replication in blood, providing an optimal framework for systems immunology studies of the initial host immune response to blood-stage infection. As discussed later, CHMI after immunization with candidate malaria vaccines has become the standard approach for evaluating vaccine efficacy against homologous and heterologous parasite challenge prior to conducting field trials in endemic areas92.
In one of the earliest transcriptomic studies of human malaria, Ockenhouse et al. employed Affymetrix GeneChip microarrays to assess gene-expression profiles of peripheral blood mononuclear cells (PBMCs) collected from 22 malaria-naïve North American adults experimentally infected with P. falciparum. Samples were collected on the day of first smear-detectable parasitemia, when subjects were either pre-symptomatic (n=16) or exhibited low-grade fevers consistent with blood-stage malaria (n=6), and compared to pre-infection baseline samples in paired analyses.69 Irrespective of symptomatology, infection induced upregulation of genes encoding the pattern recognition receptors Toll-like receptor 1 (TLR1), TLR2, TLR4; the glycoprotein CD36; and the lipopolysaccharide co-receptor CD14, but downregulation of TLR3 and TLR5. During early infection when parasitemia was at the limit of microscopic detection, pre-symptomatic subjects demonstrated upregulation of genes encoding IFN-γ and members of the IFN-γ signaling pathway as well as the inflammatory cytokine IL-1β and Caspase-1, which cleaves and activates IL-1 β to initiate pro-inflammatory responses. Increases in IFN-γ gene expression correlated with expression of CCL2, CXCL9, CXCL10 at both the gene and cytokine levels, providing evidence that transcriptomics was sensitive for detecting immune signatures even during clinically silent infections. Network analysis of genes overexpressed during P. falciparum blood-stage infection regardless of symptomatology revealed, perhaps not surprisingly, that the inflammatory cytokines TNF, IFN-γ, and IL-1β were the central nodes suspected of coordinating the immune response during early malaria. Similar activation of pro-inflammatory cytokines was also observed in subsequent CHMI studies that employed whole-blood RNA sequencing (RNA-seq) in malaria-naïve Dutch adults (n=5) at first detectable P. falciparum parasitemia81 and in both malaria-naïve (n=7) and semi-immune (n=9) Colombian adults at first detectable P. vivax parasitemia36.
Rothen et al. investigated whole-blood transcriptome changes following CHMI with P. falciparum in 10 Tanzanian adult male volunteers with prior malaria exposure using RNA-Seq79. Rather than evaluate gene expression changes at first detectable parasitemia, comparisons were made between expression at days 5, 9, and 28 post-CHMI with each other and the pre-infection baseline. Unexpectedly, differential gene expression was observed at day 5 post-CHMI during what was presumably the sub-clinical liver-stage, but not at day 9, which was on average ~2 days before microscopy detectable blood-stage parasitemia. Enrichment analysis of differentially expressed genes (DEGs) on Day 5 using blood transcription modules (BTMs) revealed overexpression of genes within modules related to the CORO1A-DEF6 network, platelet activation, regulation of localization, signaling events as well as processes involving translation and transcription. Grouping subjects by duration of prepatent period identified genes differentially expressed between early and average/late appearance of parasitemia, with over-representation of genes within modules linked to regulation of transcription, cell cycle, phosphatidylinositol signaling and erythrocyte development. One of the caveats with conducting CHMI studies in individuals with prior malaria exposure is high variability in the pre-patent period owing to differing levels of pre-existing blood-stage immunity, which introduces greater variation in the kinetics of host gene expression and reduces statistical power in detecting differential gene expression. More frequent sampling to capture the time of maximal differential gene expression can overcome this to some extent and would allow for systems analyses to determine which features of pre-existing malaria immunity influence the host response to CHMI.
Systems analyses of malaria vaccine candidates
Systems vaccinology is a sub-discipline of systems biology that attempts to unravel the complex interactions between components of the immune system after vaccination in order to better understand and predict vaccine responses. From a practical standpoint, systems vaccinology aims to determine molecular and cellular signatures induced by vaccination and detectable in peripheral blood that are predictive of vaccine immunogenicity and vaccine-induced protection. Such an approach has been applied to approved formulations of influenza94–96, yellow fever97,98, and pneumococcal96 vaccines in order to identify early molecular or cellular signatures induced by immunization that are predictive of subsequent pathogen-specific adaptive immune responses. In the case of infections for which immunologic correlates of protection are known and well defined (e.g. hepatitis A, hepatitis B, tetanus), the identification of signatures predictive of immunogenicity can be used to more rapidly evaluate the efficacy of next-generation vaccines. On the other hand, infections for which reliable immunologic correlates of protection are not yet known, such as malaria, integrated assessments of vaccine-induced molecular signatures with immunogenicity and vaccine efficacy data may lead to the identification of novel correlates of protection. Analyses of baseline (pre-vaccination) signatures can also reveal predictors of post-vaccination immune responses95, which can be used to identify individuals who may not respond optimally to vaccination. This last application is highly relevant to malaria vaccines, which appear to induce less robust antibody and cellular responses in malaria-exposed volunteers from endemic regions relative to naïve volunteers from non-endemic regions99–101.
It is important to note that protection conferred by malaria vaccines can be defined in terms of protection from parasitemia (sterile protection) or from febrile malaria (clinical protection), with vaccine efficacy assessed by proportional analysis or time to event analysis (either parasitemia or clinical malaria). Thus, a systems analysis of malaria vaccine efficacy would assess molecular signatures predictive of sterile or clinical protection using classifiers dichotomized by protection status or by time to event, with the latter being most useful for partially effective vaccines that may not have provided complete protection during the study period but nevertheless delayed either parasitemia or clinical malaria. Partially effective vaccines provide an opportunity to determine differences between protected and non-protected vaccinees using systems-based approaches.
The first systems vaccinology studies of a malaria vaccine focused on RTS,S—the most advanced malaria vaccine to date, having been licensed for use outside the European Union by the European Medicines Agency in 2015102. The RTS,S vaccine consists of a recombinant fusion protein (RTS) containing the circumsporozoite (CSP) NANP repeat region (R), multiple CSP T-cell epitopes (T), and the hepatitis B surface antigen (HBsAg; S) assembled with native HbsAg (S) into virus-like particles with the CSP component exposed on the surface103. The licensed version of RTS,S is formulated with the adjuvant AS01102. RTS,S/AS01 provided 23–41% protective efficacy against all malaria episodes in sub-Saharan Africa children 5 to 17 months of age, with declining efficacy over the follow-up period 104.
Vahey and colleagues examined PBMC transcriptomes by Affymetrix GeneChip microarrays in a subset of 39 volunteers enrolled in a phase 2b trial comparing RTS,S adjuvanted with either AS01 or AS02 to determine the pre-CHMI challenge signatures predictive of vaccine immunogenicity and efficacy86,105. Among these vaccinees, 13 (33%) were protected from P. falciparum parasitemia. Gene expression profiles were evaluated at the pre-vaccination baseline; the day of the third vaccination (just prior to injection); at 24 hours, 72 hours, and two weeks after the third vaccination; and 5 days after challenge by CHMI via P. falciparum-infected mosquito bites. Significant changes in gene expression were observed 24 and 72 hours after the third vaccination relative to the day of the third vaccination, but no differences were observed between protected and non-protected groups. Gene set enrichment analysis (GSEA), a computational method that determines whether a pre-defined set of genes are significantly enriched in a concordant manner between two groups22, revealed that the immunoproteasome degradation pathway was upregulated in protected relative to non-protected volunteers 2 weeks after the third vaccination, suggesting differential antigen presentation and processing of major histocompatibility complex peptides in RTS,S-induced malaria protection.
Rinchai and colleagues re-analyzed the microarray dataset generated by Vahey et al.86, applying a modular repertoire approach in which transcriptomic differences are assessed for sets of genes (“modules”) rather than as individual genes. These modules have been identified to be coordinately expressed (“co-clustered”) across multiple human disease states, including HIV, tuberculosis, sepsis, systemic lupus erythematosus, systemic arthritis, and also following liver transplantation30,106. Module response was measured as the percentage of responsive genes (i.e. DEGs relative to just prior to the third vaccination) within a given module. Protected volunteers had an increased interferon module response on day 1 post-vaccination compared to non-protected volunteers, while, intriguingly, non-protected volunteers had a sustained decrease in the interferon module response at days 3 and 14 after the third vaccination. Based on the decrease in abundance of interferon signatures in response to the third RTS,S vaccination among the non-protected individuals, the authors hypothesized that protective interferon responses may be antagonized by antigen-specific IgE induced by vaccination via mechanisms similar to that observed in human plasmacytoid dendritic cells107. Follow-up studies using serum from malaria-exposed Mozambican children enrolled in a phase 2b trial of RTS,S showed that RTS,S vaccination specifically induced CSP-specific IgE. However, post-vaccination levels were significantly lower in the non-protected group when compared to the protected group86, a finding that did not support the authors’ original hypothesis.
More recent transcriptomic analyses of the same RTS,S vaccination cohort (n=39) were performed by van den Berg and colleagues who used a different approach that allows for the assessment of gene expression changes over time and the incorporation of data trends in models to predict protection status88. Again, subjects were defined as ‘protected’ if they remained parasite negative for 4 weeks after challenge (n=13). However, among infected subjects, those who developed parasitemia were classified as ‘delayed parasitemia’ if their pre-patent period was longer than the longest pre-patent period in unvaccinated infectivity control subjects (n=11), while the remaining infected subjects were classified as ‘not protected’ (n=15). Gene expression was measured by Affymetrix GeneChip microarray at the time of the first and third vaccination as well 1, 3, and 14 days after the third vaccination. Serum IFN-γ levels measured before and 1 day after each dose showed consistent but non-significant increases in IFN-γ among protected subjects relative to non-protected subjects post-vaccination. Based on this evidence, the role of IFNγ in RTS,S-mediated protection was investigated. Genes within the IFNγ-signaling pathway from the proprietary Ingenuity pathways database23 demonstrated increased expression 14 days after the third vaccination in protected subjects relative to not protected subjects. Multiway partial least squares discriminant analysis (N-PLS-DA) incorporated subject, gene, and time point variables to correlate variability in expression data with the binary outcome of protection vs. non-protection (excluding delayed parasitemia). Using the entire data set as features, N-PLS-DA identified 110 genes, 42 of which had immune-related functions, within predictive models that showed 78% prediction accuracy by double cross-validation. Limiting features to only genes within the IFNγ-signaling pathway still allowed for the identification of 44 genes within predictive models that showed 77% prediction accuracy. Taken together, the blood transcriptomic studies of this vaccination cohort suggest that the vaccine-induced interferon response plays an important role in RTS,S-mediated protection against CHMI challenge.
Kazmin and colleagues used systems biology approaches to evaluate immune responses in volunteers from two different RTS,S vaccine trials. In the first, 21 volunteers received three consecutive immunizations with RTS,S/AS01 (RRR regimen). In the second, 25 volunteers received two immunizations with RTS,S/AS01 followed by immunization with adenovirus 35 expressing CSP (ARR regimen)60. Both regimens resulted in ~50% protective efficacy after challenge by CHMI via P. falciparum-infected mosquito bite three weeks after the final immunization. Gene expression of PBMCs was determined by Affymetrix GeneChip microarray at baseline; at each vaccination dose; on multiple days after each vaccination; and immediately before and after challenge. Vaccination with either regimen induced strong transcriptional responses in PBMCs at Day 1 or Day 6 after vaccination relative to the pre-vaccination baseline. Enrichment analysis with BTMs as gene sets to determine functionally relevant pathways induced by each regimen showed enrichment of BTMs related to cell cycle and B cells 6 days after each dose with persistence of the cell cycle BTM 14 days after the first vaccine dose and repression of the NK-cell-related BTM following vaccination. To assess molecular signatures of immunogenicity, BTM expression levels were then correlated to CSP-specific IgG antibody levels measured in serum collected on the day of CHMI challenge (day 77). The RRR regimen, which has been shown to induce robust anti-CSP antibody responses, induced expression of plasmablast, innate, and type I interferon (IFN)-related signatures within 6 days post-vaccination that positively correlated with these late CSP-specific IgG levels. In contrast, BTMs related to NK cells showed strong negative correlations to antibody levels, which may be interpreted as either negative regulation of B-cell responses by NK cells108, or migration of NK cells from the periphery to the lymphoid organs where they may act on infected erythrocytes via mechanisms such as antibody-dependent cellular cytoxicity.109 The ARR regimen did not induce a robust antibody response but enhanced frequencies of polyfunctional CSP-specific CD4+ T cells at Day 14 that were associated with protection, suggesting a mechanism of protection distinct from the RRR regimen. In the ARR arm, multiple modules related to innate immune activation, including antigen presentation, activated dendritic cells and monocytes, and inflammatory responses, were strongly associated with polyfunctional CSP-specific CD4+ T-cell response at day 14.
With regard to protection, the RRR regimen induced BTMs related to plasma cells, B cells, and cell cycle at day 1 after each vaccination that were positively associated with protection. In contrast, the ARR regimen induced innate immunity BTMs that positively correlated with protection. Intriguingly, both the RRR and ARR vaccination regimens induced NK-cell modules shortly after each vaccine dose that showed strong negative associations with protection. Lastly, the authors used discriminant analysis via mixed integer programming (DAMIP)110 to generate predictive signatures from the RRR vaccination group and validated these signatures using the dataset from the aforementioned study by Vahey et al.86 The successful predictive signatures were dominated by transcripts found in Day 56 (day of third vaccine dose) signatures, with a majority of these signatures containing the NK cell marker KIR2DS1. The authors show in their study the ability of three representative successful gene signatures to segregate protected and non-protected subjects in both the RRR group and the study by Vahey et al.
We recently performed whole-blood transcriptome profiling using RNA-seq to assess the composition and kinetics of direct ex vivo immune responses in 10 Dutch volunteers enrolled in a P. falciparum CPS vaccination trial in which 50% were sterilely protected after homologous CHMI challenge during the 21-day monitoring period82. To determine the time points that might provide the highest signal for blood transcriptome profiling during CPS, we assessed 15 time points before and after each of three CPS immunizations given 28 days apart and before and after challenge performed 19 weeks after the final immunization. Although no differences were observed between protected and non-protected groups, genes within BTMs related to T cells, NK cells, and mitochondrial processes were overexpressed in protected volunteers 28 days after the first immunization (one day before second immunization) relative to study baseline. The upregulation of NK-cell genes in protected subjects here contrasts with the negative association between NK modules and protection seen in the RTS,S vaccine trial described above, perhaps owing to differences in the mechanisms of protection. Differences observed in the protected group immediately prior to the second immunization corresponded with predicted activation of pathways involved in EIF2 signaling, oxidative phosphorylation and enhancement of T-cell activation pathways. Notably, correlation of summarized expression of BTMs at post-vaccination time points revealed more interactions between BTMs in protected subjects than in non-protected subjects, with the greatest number of interactions involving adaptive immunity modules observed in protected subjects relative to non-protected subjects several weeks after the second and third vaccinations. This suggests greater complexity of interactions and cross talk between adaptive and innate cells in protected subjects after CPS immunization.
Systems biology studies of naturally transmitted Plasmodium infections
Systems immunology analyses conducted with malaria-naïve volunteers enrolled in vaccine trials and CHMI studies have the distinct advantage of knowing the precise timing of the immunological perturbation (either vaccination or infection) and, in most cases, biological specimens can be sampled with high resolution and under ideal conditions. As such, in-depth assessments of the complexity of the host response to vaccination and/or infection over time can be achieved. In contrast, systems analyses of naturally transmitted Plasmodium infections must take into account a multitude of additional variables that could affect the interpretation of the data. These variables, listed in Table 3, can be both immunological and non-immunological in nature and can profoundly shape the quality and magnitude of the host response to Plasmodium infection. We previously reviewed the earliest studies that employed blood transcriptomic approaches to assess host responses during natural malaria infection111. Here, we examine more recent studies, referencing earlier investigations in their context and highlighting studies that extended beyond blood transcriptomics to incorporate other immunological, genetic, parasitological, and environmental variables into the analysis. A more complete listing of these studies along with those that rely primarily on data generated from a single platform are found in Table 2.Idaghdour and colleagues examined whole-blood transcriptomes of Beninese children with uncomplicated P. falciparum malaria (n=94) and age-matched P. falciparum negative controls from the city of Cotonou (n=61)57. Gene expression profiling was performed using Illumina BeadChips. Parasite load was noted to exert a profound influence on the whole-blood transcriptome, with P. falciparum infection and parasitemia class accounting for the majority of the transcriptomic variation in the combined dataset when modeled as a function of other variables such as sickle hemoglobin (HbS) genotype, sex, total blood counts, and ancestry. Enrichment analysis using the C2, C3, and C5 collections of the MsigDB database showed that, relative to controls, acute malaria induced signatures of innate immunity (including upregulation of IFN-inducible genes, neutrophil-associated modules, and markers of Fcγ-receptor mediated phagocytosis) but repressed adaptive signatures (including down-regulation of T cells, B cells, and cytotoxic T cell signaling pathways). Correlations of parasite load and average transcript abundance within each of six modules derived from cell type-specific expression profiles94 revealed significant effects of parasitemia on cell-type specific transcription and/or cellular composition. The authors used a mouse model of malaria to confirm the effect of high parasitemia on host gene expression observed in humans, including the strong induction of modules related to IFN response, antigen processing and presentation, the proteasome; suppression of the B-cell module; and regulation of Fcγ-receptor mediated phagocytosis. Lastly, a genome wide association test compared malaria cases and controls to explore a possible genetic basis for differential gene expression. Using a model that accounted for single nucleotide polymorphisms (SNP), malaria status, SNP by malaria status interaction, gender, location, and blood cell counts, significant genotype-by-infection interactions were identified for PRUNE2, SLC39A8, C3AR1, PADI3, and UNC119B.
Table 3.
Variable | Comments | Suggested References |
---|---|---|
age | In long-term residents of endemic areas, age is inextricably linked to malaria exposure. Additionally, there are age-related differences in immune cell composition and immunity that are independent of malaria exposure. | 112,113 |
previous malaria exposure | Prior malaria episodes can generate immunity that is protective against clinical malaria but can also shape the quality of the immune response. | 113,114 |
erythrocyte polymorphisms | Erythrocyte variants such as sickle hemoglobin (HbS) and thalassemias can adversely affect parasite growth and protect against malaria. | 115,116 |
spatial factors | Microgeography including residence near mosquito-breeding sites, near health clinics, and within clusters of asymptomatic carriers can influence malaria exposure. | 117,118 |
helminth co-infections | Co-infection with helminths can affect malaria risk and the quality of the immune response. | 119,120 |
parasite biomass | Parasite biomass, which includes the amount of sequestered parasite, can influence the host response to P. falciparum malaria. This is typically measured using plasma histidine-rich protein 2 (HRP2) levels. | 9,51 |
nutrition status | Although evidence on the effect of malnutrition of malaria risk remains inconclusive, primary childhood malnutrition impairs host defense mechanisms, potentially increasing the risk and severity of infection. | 121,122 |
microbiota | Gut microbiota composition has been shown to affect risk of P. falciparum infection in humans and malaria severity in a mouse malaria model. | 123–125 |
timing of blood sampling | Duration of parasitemia is generally not known at time of blood sampling in naturally infected individuals. Thus, there can be significant variability in immunological responses between subjects based on the natural history of acute malaria. | 126–128 |
parasite genetics | The parasite employs antigenic diversity and variation as immune evasion mechanisms. For example, certain parasite variants, such as those expressing PfEMP-1 variants with EPCR-binding phenotype, have been associated with cerebral malaria. | 129–131 |
host genetics | In addition to erythrocyte variants, other unknown host genetic determinants can potentially affect malaria severity. | 132,133 |
To investigate differences between cerebral malaria phenotypes, Feintuch and colleagues performed whole-blood transcriptional profiling by Affymetrix GeneChip microarray with samples obtained from 97 Malawian children from the Blantyre Malaria Research Project with cerebral malaria grouped by the presence (Ret+CM, n=64) or absence (Ret-CM, n=33) of malarial retinopathy, which is microvasculature pathology in the ocular fundus indicative of sequestration of parasitized erythrocytes in the cerebral vasculature51. Compared to Ret-CM, the Ret+CM phenotype had increased parasite biomass as measured by PfHRP2 levels; similar parasitemia; decreased hematocrit and platelets; and increased mortality. Gene set enrichment analysis using genes differentially expressed between CM phenotypes, with adjustment for peripheral parasitemia (not parasite biomass) and Gene Ontology (GO) pathways, identified an association between Ret+CM and the GO slim categories “cell adhesion”, which included genes encoding multimerin 1 (MMRN1), P-selectin (SELP), CD9, α-integrins and β-integrins; “cytokines”, which included genes encoding monocyte chemotactic protein 1 (MCP-1) and macrophage inflammatory protein 1α (MIP-1α); and the GO pathways related to degranulation of platelets. Plasma cytokines associated with inflammation such as TNF, MCP-1, and IL-10 were increased in Ret+CM relative to Ret-CM. Notably, extracellular matrix (ECM) pathways were also upregulated in Ret+CM children, with enrichment in genes encoding neutrophil-related proteins that included neutrophil collagenase (matrix metalloproteinase 8 [MMP8]), human neutrophil elastase (HNE), and secretory leukocyte peptidase inhibitor (SLPI). Plasma levels of neutrophil primary granule proteins HNE, myeloperoxidase (MPO), and proteinase 3 (PRTN3) were also significantly increased in Ret+CM relative to Ret-CM. This evidence suggestive of enhanced neutrophil activation during Ret+CM was also associated with neutrophil dysfunction, as neutrophils from Ret+CM children exhibited decreased chemotactic responses relative to neutrophils from Ret-CM children. The Ret-CM phenotype was positively associated with the GO slim categories “Immune response” which included TLR signaling pathways; “apoptosis”, and “antigen presentation”, as well as stress-response, proteasome complex, and heme biosynthetic process pathways. Interestingly, increased expression of GATA1, which encodes for the transcription factor involved in the growth and differentiation of erythrocytes and megakaryocytes, and genes encoding γ globin, which along with α globin form the subunits of fetal hemoglobin, were observed in Ret-CM children despite none of the participants having hemoglobinopathies such as sickle cell trait or α-thalassemia, suggesting relatively decreased erythropoiesis in Ret+CM children. Taken together, the Feintuch study suggests that children with cerebral malaria and evidence of retinopathy on exam have activated and perhaps dysfunctional neutrophils.
In a study by Lee and colleagues, dual RNA-seq was used to examine whole-blood transcriptomes of 46 Gambian children with P. falciparum uncomplicated malaria (n=21) and severe malaria (n=25, severe malarial anemia excluded), which allowed for an integrated analysis of host and parasite gene expression64. Deconvolution of host and parasite gene signatures revealed heterogeneity in leukocyte composition and parasite intraerythrocytic development stage between clinical groups. Thus, differential gene expression analysis between clinical groups included adjustments for leukocyte composition and parasite stage to remove their potential confounding effects. Notably, among genes overexpressed in severe malaria relative to uncomplicated malaria were neutrophil granule-related genes including MMP8, OLMF4, DEFA3, and ELANE. Colony-stimulating factor 3 (CSF3/GCSF), Fas cell surface death receptor, and prostaglandin E receptor 2 signaling were predicted to be activated by Ingenuity upstream regulator analysis in severe malaria. Given that host gene expression has been shown to be strongly influenced by parasitemia51,57, which differed between the two groups, the authors examined the relationship between host gene expression and parasite load. Differential host gene expression between the uncomplicated and severe malaria groups was minimally changed after adjustment for parasite density but was significantly reduced by ~99% after adjustment for PfHRP2, which is a biomarker for total parasite biomass that accounts for both circulating and sequestered parasites. Thus, the authors concluded that the host response in severe malaria is an appropriate response to excessive pathogen load.
In the same study, weight-gene co-expression network analysis (WGCNA) was applied to paired host and parasite gene expression data to identify co-expressed group of genes which were termed “modules” named after the “hub” gene with the greatest number of connections within the module64. Eleven modules included both host and parasite genes, with a module connected by HSPH1 [heat shock protein family H (Hsp110) member 1] having the greatest balance of genes of both species, being highly enriched in genes related to human heat shock response and parasite RNA metabolism. Distinct modules associated with disease severity included an MMP8-connected module enriched in defense response genes and reflective of granulopoiesis, an OAS1-connected module enriched for Type 1 IFN response genes, and a LYSMD3-connected module centered around IFN-γ. Comparison of co-expression networks for uncomplicated and severe malaria identified the MMP8-connected module as a unique feature of severe malaria, with predicted regulation by CEPBA, CSF3/GCSF, and TNF. Plasma levels of the neutrophil-related proteins defensin A3, elastase, and MMP8 significantly correlated with gene expression, but with MMP8 correlating only in severe malaria subjects.
To gain insight into the molecular and cellular processes that differentiate cerebral malaria and severe malarial anemia, we (Tran and colleagues) used Illumina Expression Beadchips to compare genome-wide transcriptional profiles of whole blood obtained from Ugandan children with retinopathy-positive cerebral malaria (n = 17) or severe malarial anemia (n = 17) enrolled in a larger study67. Gene expression data was also compared to healthy, community children negative for P. falciparum infection (n = 12) and correlated with hematological indices and relevant plasma biomarkers. Unsupervised clustering analysis showed that global differences in gene expression were influenced primarily by clinical phenotype, parasite biomass as measured by PfHRP2, sickle cell homozygosity (HbSS), platelet count, and hemoglobin levels. Subsequent statistical analyses included adjustments for PfHRP2 levels and HbSS status but not platelet counts or hemoglobin given the role of the latter two variables in defining the respective cerebral malaria and severe malarial anemia phenotypes134,135. When each was compared to healthy controls, cerebral malaria appeared to induce greater transcriptional activity relative to severe malarial anemia. Differential enrichment of functional pathways between clinical phenotypes was assessed using the CERNO test (a variant of Fisher’s combined probability test) with BTMs or KEGG biological pathways as gene sets.
Relative to controls, both cerebral malaria and severe malarial anemia groups showed upregulation of modules related to dendritic cell activation, monocyte, and inflammatory/Toll-like receptor/chemokines modules, but downregulated of modules related to natural killer and T cells. A direct comparison of cerebral malaria and severe malarial anemia revealed significant downregulation of erythroid-related modules; inhibition of heme biosynthesis pathways; predicted inhibition of GATA1 and the erythropoiesis-stimulating hormone erythropoietin (EPO); and decreased expression of a reticulocyte-specific gene signature in cerebral malaria. These findings were consistent with the prior study by Feintuch et al. (reviewed above) which found decreased GATA1 expression in retinopathy-positive cerebral malaria relative to retinopathy-negative cerebral malaria51. Hematological indices of reticulocytosis were decreased during cerebral malaria relative to severe malarial anemia despite evidence of an appropriate increase in plasma levels of erythropoietin in children in the study and the entire parent cohort (n=196), strongly suggesting ineffective erythropoiesis during cerebral malaria. Reticulocyte-related gene expression negatively correlated with expression of the module related to viral sensing and interferon-regulatory factor 2 (IRF2) targets networks (M111.1) as well as plasma concentration of the chemokine interferon gamma-induced protein 10 (CXCL10), which is an inflammatory chemokine known to inhibit human hematopoietic progenitors136.
To investigate the impact of cumulative malaria exposure on the immune system, Bediako and colleagues examined whole-blood transcriptomes using RNA-seq along with cellular and cytokine profiles in Kenyan children aged 7 to 10.5 years who either had a high (≥8) or low (<5) number of prior malaria episodes (n = 21 in each group), as well as age-matched malaria-naïve children from a cohort in a nearby community (n = 27)37. Malaria episodes were defined as body temperature above 37.5 °C with ≥ 2500 parasites/μl of blood. All children were free of malaria for the previous 110 days and were negative for Plasmodium by PCR at the time of sampling. None of the children carried the HbS allele. Differential gene expression analysis could not distinguish between the low-episode and naïve groups but did reveal significant DEGs between the high and low- groups involving innate immunity T-cell differentiation, and regulation of adaptive immune cells. Modular repertoire analysis of the RNA-seq data revealed upregulation of interferon-related modules comprising genes involved in inflammatory responses and type I and type II interferon signaling. Analysis of plasma cytokines and chemokines revealed significantly higher plasma IL-10 levels and a trend toward increasing inflammatory cytokines (TNF, IL-6) in the high vs. naïve group. Flow cytometric analysis showed that prior malaria exposure was associated with increased numbers of γδ T cells, CD11c+ B cells, CD4-CD8- T cells, and dendritic cells in both of the malaria- experienced groups compared to the naïve group. In addition, relative to the low-episode group, the high-episode group demonstrated significant expansion of γδ T cells, with this being the only subset evaluated that differed between the two malaria-experienced groups. Cellular deconvolution of whole-blood transcriptomes to infer cell-specific gene expression profiles showed transcriptional alterations in CD8+ T cells, neutrophils, and B cells in high-episode children. Specifically, enrichment analysis of significantly altered genes in CD8+ T cells showed upregulated enrichment of genes related to lymphocyte activation such as LAT, LCK, and CD40 in the high-episode children. Within the B-cell profile, there was increased expression of IGHE, which encodes for IgE, in the high-episode children. The main conclusion of this study was that high prior malaria exposure is associated with immune modifications characterized by upregulation of interferon-inducible genes and enhanced activation of neutrophils, B cells, and CD8+ T cells.
In order to better understand the cellular mechanisms of naturally acquired protection to uncomplicated, febrile malaria, we recently performed a systems biology analysis of children enrolled in a prospective cohort study of malaria immunity conducted in Kalifabougou, Mali, where malaria transmission is intense and seasonal83. During the first year of the cohort study, participants were followed weekly by active clinical surveillance and by symptom-triggered passive surveillance through self-referral. Retrospective PCR analysis of dried blood spots collected during clinic visits allowed us to determine the first time point at which an individual became positive for P. falciparum parasitemia. Combining this PCR data with clinical visit data allowed us to determine the time to progression to febrile malaria after initial parasitemia. Among a subset of the cohort consisting of children aged 6 to 11 years who started the malaria season negative for Plasmodium by PCR, three clinical phenotypes were identified: children who became parasitemic with P. falciparum and never progressed to fever during the remainder of the season (Immune, n =20); children who became parasitemic with P. falciparum but later progressed to fever within 2–14 days (Delayed Fever, n = 34); and children who were already febrile at their first parasitemic visit (Early Fever, n = 26). Consistent with the protective role of malaria-specific antibodies in naturally acquired immunity, antibody profiling by protein array showed increased magnitude and breadth of P. falciparum-specific IgG in Immune children relative to the susceptible Early Fever group. Whole-blood transcriptional profiling by RNA-seq was performed at baseline and at the first PCR-detectable parasitemia of the malaria season to determine if pre-infection transcriptional signatures and the transcriptional response to incident infection differed among the clinical phenotypes. The Immune children demonstrated a distinct molecular signature of B-cell enrichment and interferon activation that resembled that found in children with high malaria exposure in the Bediako study37. Flow cytometric analysis of PBMCs collected at the pre-infection baseline revealed an increased proportion of B cells in the Immune children along with increased expression of the IL-4 inducible, low-affinity IgE receptor CD23 on both atypical and activated memory B cells. However, it was unclear which cells were the source of IL-4 for the increased CD23 as there were no differences in Th2 transcription factors GATA3 or STAT6 in CD4+ T cells between groups. Notably, relative to the Early Fever group, both the Immune and Delayed Fever children showed increased whole-blood expression of TP53 as well as targets of p53, suggesting that activation of p53 pathways in peripheral blood was associated with protection from initial fever during incident blood-stage infection. Flow cytometry showed that expression of p53 in monocytes was increased in children protected from initial fever relative to children in the Early Fever group. Prior or recent malaria exposure could possibly explain these differences, given that p53 in monocytes increased during acute malaria relative to baseline and was higher in malaria-exposed Malian adults relative to malaria-naïve North American adults. Pharmacological activation of p53 using the MDM2/HDM2 inhibitor nutlin-3 reduced the production of TNF in malaria-naïve monocytes stimulated with P. falciparum lysate in vitro relative to vehicle-treated controls. Using a mouse malaria model, we showed that “super p53” mice, which have one extra copy of the gene encoding p53, had a blunted pro-inflammatory response relative to wild-type mice soon after infection with P. yoelii 17XNL. However, the kinetics of parasitemia were similar in both super p53 and wild-type mice and the blunting of inflammation was transient, occurring only during early blood-stage infection. Taken together, the data suggests that p53 activation plays a role in the initial control of malaria-induced inflammation without impacting parasitemia.
Next-generation studies of systems immunology in human malaria
The majority of published studies that have investigated the immune response to human malaria using high-throughput or highly multiplexed platforms (e.g. RNA-seq, gene microarrays, protein arrays, multiplex bead arrays for cytokines and chemokines) rely on measurements of bulk blood cells or serum analytes that are in essence the summation of responses from a heterogenous collection of cell types. Such bulk measurements are most sensitive to changes in the most abundant cell subsets and may not detect changes in relatively rare cell types such antigen-specific B or T cells137. The studies reviewed above employed transcriptomics using RNA isolated from bulk cell samples such as whole blood or PBMCs to evaluate differences between malaria-related clinical phenotypes. The main dilemma in bulk transcriptomics is distinguishing whether differential transcript abundance between comparison groups is due to gene expression differences within cells or differences in cell composition in the blood sample. Resolving this dilemma may involve adjusting for cellular composition with contemporaneous complete blood counts or with estimates of cell subset composition derived from deconvolution techniques that take advantage of cell subset-specific signatures derived from sorted immune cells 17,18,138. However, adjustments for cellular composition are not ideal given that it may overcorrect for gene expression changes or unnecessarily remove variations in cell composition that are actually specific to one comparison group. Highly multiplexed polychromatic flow cytometry of contemporaneously collected blood cells is one approach to measure differences in protein expression in a cell subset-specific manner139. As such, standard immunotyping of cells directly ex vivo or after cryopreservation can be complemented with in vitro stimulation with Plasmodium parasites to assess malaria-specific function. However, conventional polychromatic flow cytometry is mainly used for testing specific hypotheses generated by analysis of bulk transcriptomic data given limitations in the number of potential detectable targets to assay (both in terms of parameter limit and antibody availability) and the natural bias towards evaluating cell subsets with known functions.
The next generation of systems immunology studies will employ newer technologies to address the limitations inherent with bulk measurements. Single-cell (sc) technologies provide high-resolution assessments of the variation in the genomes, transcriptomes, and protein expression between individual cells and have recently been reviewed elsewhere in the context of immunological studies in general137,140–142. Single-cell immunoglobulin and T-cell receptor gene sequencing has been used to profile the respective repertoires of memory B cell and T cells during the host response to malaria143–145. Here, we specifically discuss the potential of single-cell technologies for systems-based interrogations of the immune response to human malaria.
Single-cell RNA-seq platforms became possible with the development of low-input RNA-seq protocols and allow for unbiased transcriptional profiling of even rare immune cell types. There are several general approaches for scRNA-seq. Individual cells can be 1) captured in barcoded gel beads (10X146 and inDrop147) or brittle resin droplets (Drop-Seq148) or 2) physically separated in 96-well plates using fluorescence-activated cell sorting or in microfluidic chips. After cell lysis, RNA is reversed transcribed and amplified by PCR with subsequent RNA-seq libraries prepared as cellular barcoded pools or from individual cells. Sequencing can be performed from the 3’ end of the transcript to detect only gene expression or for full-length transcripts to determine splice variants, B-cell receptor and T-cell receptor repertoires in addition to gene expression.
In studies of acute malaria using cryopreserved PBMCs, confounding due to differences in immune cell composition among malaria syndromes64,67,149 can be readily resolved by scRNA-seq provided that samples demonstrates adequate viability (generally >90%). Applying scRNA-seq to acute malaria blood samples collected ex vivo would be more physiologically relevant and allows for the interrogation of neutrophils and other granulocytes but could be challenging given the logistic hurdles of implementing such technologies in malaria-endemic regions and the need to remove erythrocytes and non-viable cells that would otherwise contribute to unwanted sequencing reads. Dual-RNA-seq has been applied to bulk whole blood from malaria patients to study the host-parasite “interactome”,9,64 but is limited to cases with high parasite densities (>10,000 parasites/μl, unpublished data) and thus probably not reliable for investigating less complicated malaria or asymptomatic infections. Here, scRNA-seq can potentially allow transcriptional profiling of individual Plasmodium-infected erythrocytes and human leukocytes collected ex vivo from patients with low parasitemia, provided that these target components can be effectively separated from both uninfected erythrocytes and non-viable cells.
Single-cell technologies can also be used to evaluate the epigenetic landscape of immune cells in unprecedented detail. For instance, applying single-cell Assay for Transposase Accessible Chromatin (scATAC-seq)150,151 with scRNA-seq on the same immune cell preparations can provide insight on the impact of malaria or prior malaria exposure on the coordinated accessibility and expression of genes involved in control of the host immune response and the degree of regulatory variation within cell subsets. Combined with single-cell bisulfite sequencing, which provides a map of the DNA methylome152,153, a more complete understanding of cell-state dynamics and epigenetic heterogeneity within immune cells during different malaria syndromes or malaria-immune phenotypes can be ascertained. Protocols have been developed to generate transcriptomes and methylomes from the same single cells154,155 as well as simultaneous profiling of chromatin accessibility, DNA methylation and the transcriptome using single-cell nucleosome, methylation and transcription sequencing (scNMT-seq)156. Joint profiling of single-cell chromatin accessibility and transcriptomes can now be performed in each of thousands of cells using single-cell combinatorial indexing methods157, with the scalable throughput being particularly useful for the analysis of peripheral blood samples from malaria patients that are often comprised of highly heterogeneous immune cell subsets.
Mass cytometry (as known as cytometry by time of flight or CyTOF) combined with single-cell fluidics is a relatively new, commercially available technique that can extend the number of quantifiable parameters beyond those available with conventional flow cytometry. Rather than using fluorochromes that are inherently limited by spectral overlap, mass cytometry relies on monoclonal antibodies tagged with rare-earth-metal isotopes that have minimal signal overlap, which currently allows for the simultaneous interrogation of nearly 50 immune markers including cell surface proteins, intracellular cytokines, and phosphorylated proteins158,159. Such expansion of parameters would provide deeper insight into the functional and activation states of immune cells in response to Plasmodium during natural exposure or after in vitro stimulation.
Lastly, more complete understanding variable immune responses in individuals living in malaria-endemic areas may require detailed assessments of co-infections and commensal microorganisms, both which can shape the host response to malaria and vaccination160–162. Future studies could integrate metagenomic analyses of microbiota from the skin and mucosal surfaces (e.g. the gut and urinary tract) with immunological signatures in the peripheral blood to assess the potential impact that specific microorganisms may have on acute malaria or immunization to malaria vaccines in an unbiased manner.
Summary
For over a decade ‘systems’ tools have been used to investigate the human immune response to malaria infection and vaccine candidates. These unbiased approaches have begun to provide insights into the nature of protective and pathologic immune responses to malaria, as well as potential correlates of immunogenicity and protective efficacy of vaccine candidates. Such advances may not have been readily achieved through hypothesis-driven, reductionist approaches. As more datasets become available from studies that vary by study population, clinical phenotype, and vaccine regimen, it is likely that common themes will continue to emerge that lead to a more comprehensive understanding of the complex and multi-faceted nature of immune response across various malaria-endemic settings. Many of these datasets have generated novel hypotheses of immunity to malaria that will require further testing in model systems to better elucidate the mechanisms of protection and immunopathogenesis. The limitations of current studies, most notably the inability of bulk approaches to adequately attribute differences in molecular abundance to increased cellular activity or changes in cell composition, could potentially be addressed with single-cell technologies. When coupled with other recently developed technologies such as mass cytometry, the resolution and interpretability of systems approaches will continue to increase. Comprehensive systems studies conducted in malaria-endemic settings may need to account for co-infections and variations in commensal flora by linking metagenomic profiles to distinct molecular signatures associated with malaria response phenotypes to provide a more complete understanding of the human immune response during naturally transmitted malaria.
ACKNOWLEDGMENTS
This work was supported by the Intramural Research Program of the National Institutes of Health, National Institute of Allergy and Infectious Diseases (NIAID). T.M.T. is also supported by K08AI125682 (NIAID) and Grant 2018091 from the Doris Duke Charitable Foundation.
Footnotes
CONFLICT OF INTEREST
The authors do not have any conflict of interest to declare.
REFERENCES
- 1.World Health Organization. World Malaria Report 2018. World Health Organization; 2018. [Google Scholar]
- 2.World Health Organization. Countries and territories certified malaria-free by WHO. 2019; Countries that have achieved at least 3 consecutive years of zero indigenous cases are eligible to apply for a WHO certification of malaria-free status. Available at: https://www.who.int/malaria/areas/elimination/malaria-free-countries/en/. Accessed July 10, 2019, 2019.
- 3.malERA Refresh Consultative Panel on Tools for Malaria Elimination. malERA: An updated research agenda for diagnostics, drugs, vaccines, and vector control in malaria elimination and eradication. PLoS Med. 2017;14(11):e1002455. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Varo R, Crowley VM, Sitoe A, et al. Adjunctive therapy for severe malaria: a review and critical appraisal. Malar J. 2018;17(1):47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Gardner MJ, Hall N, Fung E, et al. Genome sequence of the human malaria parasite Plasmodium falciparum. Nature. 2002;419(6906):498–511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Flanagan KL, Wilson KL, Plebanski M. Polymorphism in liver-stage malaria vaccine candidate proteins: immune evasion and implications for vaccine design. Expert Rev Vaccines. 2016;15(3):389–399. [DOI] [PubMed] [Google Scholar]
- 7.Ntege EH, Takashima E, Morita M, Nagaoka H, Ishino T, Tsuboi T. Blood-stage malaria vaccines: post-genome strategies for the identification of novel vaccine candidates. Expert Rev Vaccines. 2017;16(8):769–779. [DOI] [PubMed] [Google Scholar]
- 8.Germain RN. Will Systems Biology Deliver Its Promise and Contribute to the Development of New or Improved Vaccines? What Really Constitutes the Study of “Systems Biology” and How Might Such an Approach Facilitate Vaccine Design. Cold Spring Harb Perspect Biol. 2018;10(8). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Lee HJ, Georgiadou A, Otto TD, et al. Transcriptomic Studies of Malaria: a Paradigm for Investigation of Systemic Host-Pathogen Interactions. Microbiol Mol Biol Rev. 2018;82(2). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Trapnell C, Roberts A, Goff L, et al. Differential gene and transcript expression analysis of RNA-seq experiments with TopHat and Cufflinks. Nat Protoc. 2012;7(3):562–578. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014;15(12):550. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Robinson MD, McCarthy DJ, Smyth GK. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics. 2010;26(1):139–140. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Ritchie ME, Phipson B, Wu D, et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 2015;43(7):e47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Pavelka N, Pelizzola M, Vizzardelli C, et al. A power law global error model for the identification of differentially expressed genes in microarray data. BMC Bioinformatics. 2004;5:203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Tusher VG, Tibshirani R, Chu G. Significance analysis of microarrays applied to the ionizing radiation response. Proc Natl Acad Sci U S A. 2001;98(9):5116–5121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Abbas AR, Baldwin D, Ma Y, et al. Immune response in silico (IRIS): immune-specific genes identified from a compendium of microarray expression data. Genes Immun. 2005;6(4):319–331. [DOI] [PubMed] [Google Scholar]
- 17.Vallania F, Tam A, Lofgren S, et al. Leveraging heterogeneity across multiple datasets increases cell-mixture deconvolution accuracy and reduces biological and technical biases. Nat Commun. 2018;9(1):4735. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Newman AM, Liu CL, Green MR, et al. Robust enumeration of cell subsets from tissue expression profiles. Nat Methods. 2015;12(5):453–457. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Wu D, Smyth GK. Camera: a competitive gene set test accounting for inter-gene correlation. Nucleic Acids Res. 2012;40(17):e133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Huang DW, Sherman BT, Tan Q, et al. The DAVID Gene Functional Classification Tool: a novel biological module-centric algorithm to functionally analyze large gene lists. Genome Biol. 2007;8(9):R183. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Huang DW, Sherman BT, Tan Q, et al. DAVID Bioinformatics Resources: expanded annotation database and novel algorithms to better extract biology from large gene lists. Nucleic Acids Res. 2007;35(Web Server issue):W169–175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Subramanian A, Tamayo P, Mootha VK, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A. 2005;102(43):15545–15550. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Kramer A, Green J, Pollard J Jr., Tugendreich S. Causal analysis approaches in Ingenuity Pathway Analysis. Bioinformatics. 2014;30(4):523–530. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Zyla J, Marczyk M, Domaszewska T, Kaufmann SHE, Polanska J, Weiner J. Gene set enrichment for reproducible science: comparison of CERNO and eight other algorithms. Bioinformatics. 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Langfelder P, Horvath S. WGCNA: an R package for weighted correlation network analysis. BMC Bioinformatics. 2008;9:559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Aurrecoechea C, Brestelli J, Brunk BP, et al. PlasmoDB: a functional genomic database for malaria parasites. Nucleic Acids Res. 2009;37(Database issue):D539–543. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Li S, Rouphael N, Duraisingham S, et al. Molecular signatures of antibody responses derived from a systems biology study of five human vaccines. Nat Immunol. 2014;15(2):195–204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Godec J, Tan Y, Liberzon A, et al. Compendium of Immune Signatures Identifies Conserved and Species-Specific Biology in Response to Inflammation. Immunity. 2016;44(1):194–206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Kanehisa M, Goto S. KEGG: kyoto encyclopedia of genes and genomes. Nucleic Acids Res. 2000;28(1):27–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Chaussabel D, Baldwin N. Democratizing systems immunology with modular transcriptional repertoire analyses. Nat Rev Immunol. 2014;14(4):271–280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Liberzon A, Subramanian A, Pinchback R, Thorvaldsdottir H, Tamayo P, Mesirov JP. Molecular signatures database (MSigDB) 3.0. Bioinformatics. 2011;27(12):1739–1740. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Huang da W, Sherman BT, Lempicki RA. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc. 2009;4(1):44–57. [DOI] [PubMed] [Google Scholar]
- 33.Afgan E, Baker D, van den Beek M, et al. The Galaxy platform for accessible, reproducible and collaborative biomedical analyses: 2016 update. Nucleic Acids Res. 2016;44(W1):W3–W10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Zuck M, Austin LS, Danziger SA, Aitchison JD, Kaushansky A. The Promise of Systems Biology Approaches for Revealing Host Pathogen Interactions in Malaria. Front Microbiol. 2017;8:2183. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Galinski MR, Lapp SA, Peterson MS, et al. Plasmodium knowlesi: a superb in vivo nonhuman primate model of antigenic variation in malaria. Parasitology. 2018;145(1):85–100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Arevalo-Herrera M, Lopez-Perez M, Dotsey E, et al. Antibody Profiling in Naive and Semi-immune Individuals Experimentally Challenged with Plasmodium vivax Sporozoites. PLoS Negl Trop Dis. 2016;10(3):e0004563. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Bediako Y, Adams R, Reid AJ, et al. Repeated clinical malaria episodes are associated with modification of the immune system in children. BMC Med. 2019;17(1):60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Boldt ABW, van Tong H, Grobusch MP, et al. The blood transcriptome of childhood malaria. EBioMedicine. 2019;40:614–625. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Chakraborty B, Mondal P, Gajendra P, Mitra M, Das C, Sengupta S. Deciphering genetic regulation of CD14 by SP1 through characterization of peripheral blood mononuclear transcriptome of P. faiciparum and P. vivax infected malaria patients. EBioMedicine. 2018;37:442–452. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Chakravorty SJ, Carret C, Nash GB, Ivens A, Szestak T, Craig AG. Altered phenotype and gene transcription in endothelial cells, induced by Plasmodium falciparum-infected red blood cells: pathogenic or protective? Int J Parasitol. 2007;37(8–9):975–987. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Chattopadhyay R, de la Vega P, Paik SH, et al. Early transcriptional responses of HepG2-A16 liver cells to infection by Plasmodium falciparum sporozoites. J Biol Chem. 2011;286(30):26396–26405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Chen JH, Jung JW, Wang Y, et al. Immunoproteomics profiling of blood stage Plasmodium vivax infection by high-throughput screening assays. J Proteome Res. 2010;9(12):6479–6489. [DOI] [PubMed] [Google Scholar]
- 43.Chen JH, Chen SB, Wang Y, et al. An immunomics approach for the analysis of natural antibody responses to Plasmodium vivax infection. Mol Biosyst. 2015;11(8):2354–2363. [DOI] [PubMed] [Google Scholar]
- 44.Colborn JM, Ylostalo JH, Koita OA, Cisse OH, Krogstad DJ. Human Gene Expression in Uncomplicated Plasmodium falciparum Malaria. J Immunol Res. 2015;2015:162639. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Cordy RJ, Patrapuvich R, Lili LN, et al. Distinct amino acid and lipid perturbations characterize acute versus chronic malaria. JCI Insight. 2019;4(9). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Crompton PD, Kayala MA, Traore B, et al. A prospective analysis of the Ab response to Plasmodium falciparum before and after a malaria season by protein microarray. Proc Natl Acad Sci U S A. 2010;107(15):6958–6963. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Dent AE, Nakajima R, Liang L, et al. Plasmodium falciparum Protein Microarray Antibody Profiles Correlate With Protection From Symptomatic Malaria in Kenya. J Infect Dis. 2015;212(9):1429–1438. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Dobbs KR, Embury P, Vulule J, et al. Monocyte dysregulation and systemic inflammation during pediatric falciparum malaria. JCI Insight. 2017;2(18). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Doolan DL, Mu Y, Unal B, et al. Profiling humoral immune responses to P. falciparum infection with protein microarrays. Proteomics. 2008;8(22):4680–4694. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Dunachie S, Berthoud T, Hill AV, Fletcher HA. Transcriptional changes induced by candidate malaria vaccines and correlation with protection against malaria in a human challenge model. Vaccine. 2015;33(40):5321–5331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Feintuch CM, Saidi A, Seydel K, et al. Activated Neutrophils Are Associated with Pediatric Cerebral Malaria Vasculopathy in Malawian Children. MBio. 2016;7(1):e01300–01315. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Felgner PL, Roestenberg M, Liang L, et al. Pre-erythrocytic antibody profiles induced by controlled human malaria infections in healthy volunteers under chloroquine prophylaxis. Sci Rep. 2013;3:3549. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Franklin BS, Parroche P, Ataide MA, 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 U S A. 2009;106(14):5789–5794. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Gardinassi LG, Arevalo-Herrera M, Herrera S, 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;17:158–170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Grangeiro de Carvalho E, Bonin M, Kremsner PG, Kun JF. Plasmodium falciparum-infected erythrocytes and IL-12/IL-18 induce diverse transcriptomes in human NK cells: IFN-alpha/beta pathway versus TREM signaling. PLoS One. 2011;6(9):e24963. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.de Carvalho EG, Bottger E, Hoang VT, Kremsner PG, Kun JF. Limited response of NK92 cells to Plasmodium falciparum-infected erythrocytes. Malar J. 2011;10:311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Idaghdour Y, Quinlan J, Goulet JP, et al. Evidence for additive and interaction effects of host genotype and infection in malaria. Proc Natl Acad Sci U S A. 2012;109(42):16786–16793. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Kamuyu G, Tuju J, Kimathi R, et al. KILchip v1.0: A Novel Plasmodium falciparum Merozoite Protein Microarray to Facilitate Malaria Vaccine Candidate Prioritization. Front Immunol. 2018;9:2866. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Kassa FA, Shio MT, Bellemare MJ, Faye B, Ndao M, Olivier M. New inflammation-related biomarkers during malaria infection. PLoS One. 2011;6(10):e26495. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Kazmin D, Nakaya HI, Lee EK, et al. Systems analysis of protective immune responses to RTS,S malaria vaccination in humans. Proc Natl Acad Sci U S A. 2017;114(9):2425–2430. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Keller CC, Ouma C, Ouma Y, et al. Suppression of a novel hematopoietic mediator in children with severe malarial anemia. Infect Immun. 2009;77(9):3864–3871. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Krupka M, Seydel K, Feintuch CM, 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(3):1150–1155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Kumar M, Varun CN, Dey G, et al. Identification of Host-Response in Cerebral Malaria Patients Using Quantitative Proteomic Analysis. Proteomics Clin Appl. 2018;12(4):e1600187. [DOI] [PubMed] [Google Scholar]
- 64.Lee HJ, Georgiadou A, Walther M, et al. Integrated pathogen load and dual transcriptome analysis of systemic host-pathogen interactions in severe malaria. Sci Transl Med. 2018;10(447). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Leopold SJ, Ghose A, Allman EL, et al. Identifying the Components of Acidosis in Patients With Severe Plasmodium falciparum Malaria Using Metabolomics. J Infect Dis. 2019;219(11):1766–1776. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Lu F, Li J, Wang B, et al. Profiling the humoral immune responses to Plasmodium vivax infection and identification of candidate immunogenic rhoptry-associated membrane antigen (RAMA). J Proteomics. 2014;102:66–82. [DOI] [PubMed] [Google Scholar]
- 67.Nallandhighal S, Park GS, Ho YY, Opoka RO, John CC, Tran TM. Whole-Blood Transcriptional Signatures Composed of Erythropoietic and NRF2-Regulated Genes Differ Between Cerebral Malaria and Severe Malarial Anemia. J Infect Dis. 2019;219(1):154–164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Nnedu ON, O’Leary MP, Mutua D, et al. Humoral immune responses to Plasmodium falciparum among HIV-1-infected Kenyan adults. Proteomics Clin Appl. 2011;5(11–12):613–623. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Ockenhouse CF, Hu WC, Kester KE, 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(10):5561–5573. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Orikiiriza J, Surowiec I, Lindquist E, et al. Lipid response patterns in acute phase paediatric Plasmodium falciparum malaria. Metabolomics. 2017;13(4):41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Portugal S, Moebius J, Skinner J, et al. Exposure-dependent control of malaria-induced inflammation in children. PLoS Pathog. 2014;10(4):e1004079. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Quin JE, Bujila I, Cherif M, et al. Major transcriptional changes observed in the Fulani, an ethnic group less susceptible to malaria. Elife. 2017;6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Ray S, Renu D, Srivastava R, et al. Proteomic investigation of falciparum and vivax malaria for identification of surrogate protein markers. PLoS One. 2012;7(8):e41751. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Ray S, Kamath KS, Srivastava R, et al. Serum proteome analysis of vivax malaria: An insight into the disease pathogenesis and host immune response. J Proteomics. 2012;75(10):3063–3080. [DOI] [PubMed] [Google Scholar]
- 75.Ray S, Kumar V, Bhave A, et al. Proteomic analysis of Plasmodium falciparum induced alterations in humans from different endemic regions of India to decipher malaria pathogenesis and identify surrogate markers of severity. J Proteomics. 2015;127(Pt A):103–113. [DOI] [PubMed] [Google Scholar]
- 76.Reutersward P, Bergstrom S, Orikiiriza J, et al. Levels of human proteins in plasma associated with acute paediatric malaria. Malar J. 2018;17(1):426. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.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(8):e0003978. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Rojas-Pena ML, Duan M, Arafat D, et al. Individualized Transcriptional Resolution of Complicated Malaria in a Colombian Study. J Pers Med. 2018;8(3). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Rothen J, Murie C, Carnes J, 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(6):e0199392. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Torres KJ, Castrillon CE, Moss EL, et al. Genome-level determination of Plasmodium falciparum blood-stage targets of malarial clinical immunity in the Peruvian Amazon. J Infect Dis. 2015;211(8):1342–1351. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Tran TM, Jones MB, Ongoiba A, et al. Transcriptomic evidence for modulation of host inflammatory responses during febrile Plasmodium falciparum malaria. Sci Rep. 2016;6:31291. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Tran TM, Bijker EM, Haks MC, et al. Whole-blood transcriptomic signatures induced during immunization by chloroquine prophylaxis and Plasmodium falciparum sporozoites. Sci Rep. 2019;9(1):8386. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Tran TM, Guha R, Portugal S, et al. A Molecular Signature in Blood Reveals a Role for p53 in Regulating Malaria-Induced Inflammation. Immunity. 2019;51(4):750–765.e10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Tripathi AK, Sha W, Shulaev V, Stins MF, Sullivan DJ Jr. Plasmodium falciparum-infected erythrocytes induce NF-kappaB regulated inflammatory pathways in human cerebral endothelium. Blood. 2009;114(19):4243–4252. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Uplekar S, Rao PN, Ramanathapuram L, et al. Characterizing Antibody Responses to Plasmodium vivax and Plasmodium falciparum Antigens in India Using Genome-Scale Protein Microarrays. PLoS Negl Trop Dis. 2017;11(1):e0005323. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Vahey MT, Wang Z, Kester KE, et al. Expression of genes associated with immunoproteasome processing of major histocompatibility complex peptides is indicative of protection with adjuvanted RTS,S malaria vaccine. J Infect Dis. 2010;201(4):580–589. [DOI] [PubMed] [Google Scholar]
- 87.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(5):440–447. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.van den Berg RA, Coccia M, Ballou WR, et al. Predicting RTS,S Vaccine-Mediated Protection from Transcriptomes in a Malaria-Challenge Clinical Trial. Front Immunol. 2017;8:557. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Yamagishi J, Natori A, Tolba ME, et al. Interactive transcriptome analysis of malaria patients and infecting Plasmodium falciparum. Genome Res. 2014;24(9):1433–1444. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Austin SC, Stolley PD, Lasky T. The history of malariotherapy for neurosyphilis. Modern parallels. JAMA. 1992;268(4):516–519. [PubMed] [Google Scholar]
- 91.Marshall J. Penicillin in the Treatment of Syphilis. Nature. 1945;156(3974):769–772. [Google Scholar]
- 92.Sauerwein RW, Roestenberg M, Moorthy VS. Experimental human challenge infections can accelerate clinical malaria vaccine development. Nat Rev Immunol. 2011;11(1):57–64. [DOI] [PubMed] [Google Scholar]
- 93.Stanisic DI, McCarthy JS, Good MF. Controlled Human Malaria Infection: Applications, Advances, and Challenges. Infect Immun. 2018;86(1). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Nakaya HI, Wrammert J, Lee EK, et al. Systems biology of vaccination for seasonal influenza in humans. Nat Immunol. 2011;12(8):786–795. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Tsang JS, Schwartzberg PL, Kotliarov Y, et al. Global analyses of human immune variation reveal baseline predictors of postvaccination responses. Cell. 2014;157(2):499–513. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Obermoser G, Presnell S, Domico K, et al. Systems scale interactive exploration reveals quantitative and qualitative differences in response to influenza and pneumococcal vaccines. Immunity. 2013;38(4):831–844. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Querec TD, Akondy RS, Lee EK, et al. Systems biology approach predicts immunogenicity of the yellow fever vaccine in humans. Nature immunology. 2009;10(1):116–125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Gaucher D, Therrien R, Kettaf N, et al. Yellow fever vaccine induces integrated multilineage and polyfunctional immune responses. J Exp Med. 2008;205(13):3119–3131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Jongo SA, Church LWP, Mtoro AT, et al. Safety and Differential Antibody and T-Cell Responses to the Plasmodium falciparum Sporozoite Malaria Vaccine, PfSPZ Vaccine, by Age in Tanzanian Adults, Adolescents, Children, and Infants. Am J Trop Med Hyg. 2019;100(6):1433–1444. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Jongo SA, Shekalaghe SA, Church LWP, et al. Safety, Immunogenicity, and Protective Efficacy against Controlled Human Malaria Infection of Plasmodium falciparum Sporozoite Vaccine in Tanzanian Adults. Am J Trop Med Hyg. 2018;99(2):338–349. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Sissoko MS, Healy SA, Katile A, et al. Safety and efficacy of PfSPZ Vaccine against Plasmodium falciparum via direct venous inoculation in healthy malaria-exposed adults in Mali: a randomised, double-blind phase 1 trial. Lancet Infect Dis. 2017;17(5):498–509. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.World Health Organization. Malaria vaccine: WHO position paper, January 2016 - Recommendations. Vaccine. 2018;36(25):3576–3577. [DOI] [PubMed] [Google Scholar]
- 103.Gordon DM, McGovern TW, Krzych U, et al. Safety, immunogenicity, and efficacy of a recombinantly produced Plasmodium falciparum circumsporozoite protein-hepatitis B surface antigen subunit vaccine. J Infect Dis. 1995;171(6):1576–1585. [DOI] [PubMed] [Google Scholar]
- 104.RTS,S Clinical Trials Partnership. Efficacy and safety of RTS,S/AS01 malaria vaccine with or without a booster dose in infants and children in Africa: final results of a phase 3, individually randomised, controlled trial. Lancet. 2015;386(9988):31–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Kester KE, Cummings JF, Ofori-Anyinam O, et al. Randomized, double-blind, phase 2a trial of falciparum malaria vaccines RTS,S/AS01B and RTS,S/AS02A in malaria-naive adults: safety, efficacy, and immunologic associates of protection. J Infect Dis. 2009;200(3):337–346. [DOI] [PubMed] [Google Scholar]
- 106.Chaussabel D, Quinn C, Shen J, et al. A modular analysis framework for blood genomics studies: application to systemic lupus erythematosus. Immunity. 2008;29(1):150–164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Gill MA, Bajwa G, George TA, et al. Counterregulation between the FcepsilonRI pathway and antiviral responses in human plasmacytoid dendritic cells. J Immunol. 2010;184(11):5999–6006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Rydyznski C, Daniels KA, Karmele EP, et al. Generation of cellular immune memory and B-cell immunity is impaired by natural killer cells. Nat Commun. 2015;6:6375. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Arora G, Hart GT, Manzella-Lapeira J, et al. NK cells inhibit Plasmodium falciparum growth in red blood cells via antibody-dependent cellular cytotoxicity. Elife. 2018;7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Lee EK. Large-scale optimization-based classification models in medicine and biology. Ann Biomed Eng. 2007;35(6):1095–1109. [DOI] [PubMed] [Google Scholar]
- 111.Tran TM, Samal B, Kirkness E, Crompton PD. Systems immunology of human malaria. Trends Parasitol. 2012;28(6):248–257. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Montes de Oca M, Good MF, McCarthy JS, Engwerda CR. The Impact of Established Immunoregulatory Networks on Vaccine Efficacy and the Development of Immunity to Malaria. J Immunol. 2016;197(12):4518–4526. [DOI] [PubMed] [Google Scholar]
- 113.Doolan DL, Dobano C, Baird JK. Acquired immunity to malaria. Clin Microbiol Rev. 2009;22(1):13–36, Table of Contents. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Crompton PD, Moebius J, Portugal S, et al. Malaria immunity in man and mosquito: insights into unsolved mysteries of a deadly infectious disease. Annu Rev Immunol. 2014;32:157–187. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Goheen MM, Campino S, Cerami C. The role of the red blood cell in host defence against falciparum malaria: an expanding repertoire of evolutionary alterations. Br J Haematol. 2017;179(4):543–556. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Taylor SM, Parobek CM, Fairhurst RM. Haemoglobinopathies and the clinical epidemiology of malaria: a systematic review and meta-analysis. Lancet Infect Dis. 2012;12(6):457–468. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Bejon P, Williams TN, Nyundo C, et al. A micro-epidemiological analysis of febrile malaria in Coastal Kenya showing hotspots within hotspots. Elife. 2014;3:e02130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Bejon P, Williams TN, Liljander A, et al. Stable and unstable malaria hotspots in longitudinal cohort studies in Kenya. PLoS Med. 2010;7(7):e1000304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Salgame P, Yap GS, Gause WC. Effect of helminth-induced immunity on infections with microbial pathogens. Nat Immunol. 2013;14(11):1118–1126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Degarege A, Degarege D, Veledar E, et al. Plasmodium falciparum Infection Status among Children with Schistosoma in Sub-Saharan Africa: A Systematic Review and Meta-analysis. PLoS Negl Trop Dis. 2016;10(12):e0005193. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Ibrahim MK, Zambruni M, Melby CL, Melby PC. Impact of Childhood Malnutrition on Host Defense and Infection. Clin Microbiol Rev. 2017;30(4):919–971. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Das D, Grais RF, Okiro EA, et al. Complex interactions between malaria and malnutrition: a systematic literature review. BMC Med. 2018;16(1):186. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Villarino NF, LeCleir GR, Denny JE, et al. Composition of the gut microbiota modulates the severity of malaria. Proc Natl Acad Sci U S A. 2016;113(8):2235–2240. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Yilmaz B, Portugal S, Tran TM, et al. Gut microbiota elicits a protective immune response against malaria transmission. Cell. 2014;159(6):1277–1289. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Yooseph S, Kirkness EF, Tran TM, et al. Stool microbiota composition is associated with the prospective risk of Plasmodium falciparum infection. BMC Genomics. 2015;16:631. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.McElroy PD, Beier JC, Oster CN, et al. Dose- and time-dependent relations between infective Anopheles inoculation and outcomes of Plasmodium falciparum parasitemia among children in western Kenya. Am J Epidemiol. 1997;145(10):945–956. [DOI] [PubMed] [Google Scholar]
- 127.Hodgson SH, Juma E, Salim A, 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Dejon-Agobe JC, Ateba-Ngoa U, Lalremruata A, et al. Controlled human malaria infection of healthy lifelong malaria-exposed adults to assess safety, immunogenicity and efficacy of the asexual blood stage malaria vaccine candidate GMZ2. Clin Infect Dis. 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Mosnier LO, Lavstsen T. The role of EPCR in the pathogenesis of severe malaria. Thromb Res. 2016;141 Suppl 2:S46–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Saito F, Hirayasu K, Satoh T, et al. Immune evasion of Plasmodium falciparum by RIFIN via inhibitory receptors. Nature. 2017;552(7683):101–105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Wahlgren M, Goel S, Akhouri RR. Variant surface antigens of Plasmodium falciparum and their roles in severe malaria. Nat Rev Microbiol. 2017;15(8):479–491. [DOI] [PubMed] [Google Scholar]
- 132.Damena D, Denis A, Golassa L, Chimusa ER. Genome-wide association studies of severe P. falciparum malaria susceptibility: progress, pitfalls and prospects. BMC Med Genomics. 2019;12(1):120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Steri M, Orru V, Idda ML, et al. Overexpression of the Cytokine BAFF and Autoimmunity Risk. N Engl J Med. 2017;376(17):1615–1626. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Chimalizeni Y, Kawaza K, Taylor T, Molyneux M. The platelet count in cerebral malaria, is it useful to the clinician? Am J Trop Med Hyg. 2010;83(1):48–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.White NJ. Anaemia and malaria. Malar J. 2018;17(1):371. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Sarris AH, Broxmeyer HE, Wirthmueller U, et al. Human interferon-inducible protein 10: expression and purification of recombinant protein demonstrate inhibition of early human hematopoietic progenitors. J Exp Med. 1993;178(3):1127–1132. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Chattopadhyay PK, Gierahn TM, Roederer M, Love JC. Single-cell technologies for monitoring immune systems. Nat Immunol. 2014;15(2):128–135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Chen B, Khodadoust MS, Liu CL, Newman AM, Alizadeh AA. Profiling Tumor Infiltrating Immune Cells with CIBERSORT. Methods Mol Biol. 2018;1711:243–259. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Perfetto SP, Chattopadhyay PK, Roederer M. Seventeen-colour flow cytometry: unravelling the immune system. Nat Rev Immunol. 2004;4(8):648–655. [DOI] [PubMed] [Google Scholar]
- 140.Stubbington MJT, Rozenblatt-Rosen O, Regev A, Teichmann SA. Single-cell transcriptomics to explore the immune system in health and disease. Science. 2017;358(6359):58–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Papalexi E, Satija R. Single-cell RNA sequencing to explore immune cell heterogeneity. Nat Rev Immunol. 2018;18(1):35–45. [DOI] [PubMed] [Google Scholar]
- 142.Palit S, Heuser C, de Almeida GP, Theis FJ, Zielinski CE. Meeting the Challenges of High-Dimensional Single-Cell Data Analysis in Immunology. Front Immunol. 2019;10:1515. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Mpina M, Maurice NJ, Yajima M, et al. Controlled Human Malaria Infection Leads to Long-Lasting Changes in Innate and Innate-like Lymphocyte Populations. J Immunol. 2017;199(1):107–118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Murugan R, Buchauer L, Triller G, et al. Clonal selection drives protective memory B cell responses in controlled human malaria infection. Sci Immunol. 2018;3(20). [DOI] [PubMed] [Google Scholar]
- 145.Mamedov MR, Scholzen A, Nair RV, et al. A Macrophage Colony-Stimulating-Factor-Producing gammadelta T Cell Subset Prevents Malarial Parasitemic Recurrence. Immunity. 2018;48(2):350–363 e357. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Zheng GX, Terry JM, Belgrader P, et al. Massively parallel digital transcriptional profiling of single cells. Nat Commun. 2017;8:14049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Zilionis R, Nainys J, Veres A, et al. Single-cell barcoding and sequencing using droplet microfluidics. Nat Protoc. 2017;12(1):44–73. [DOI] [PubMed] [Google Scholar]
- 148.Macosko EZ, Basu A, Satija R, et al. Highly Parallel Genome-wide Expression Profiling of Individual Cells Using Nanoliter Droplets. Cell. 2015;161(5):1202–1214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Mandala WL, Msefula CL, Gondwe EN, et al. Lymphocyte Perturbations in Malawian Children with Severe and Uncomplicated Malaria. Clin Vaccine Immunol. 2015;23(2):95–103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Buenrostro JD, Wu B, Litzenburger UM, et al. Single-cell chromatin accessibility reveals principles of regulatory variation. Nature. 2015;523(7561):486–490. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Cusanovich DA, Daza R, Adey A, et al. Multiplex single cell profiling of chromatin accessibility by combinatorial cellular indexing. Science. 2015;348(6237):910–914. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Farlik M, Sheffield NC, Nuzzo A, et al. Single-cell DNA methylome sequencing and bioinformatic inference of epigenomic cell-state dynamics. Cell Rep. 2015;10(8):1386–1397. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Smallwood SA, Lee HJ, Angermueller C, et al. Single-cell genome-wide bisulfite sequencing for assessing epigenetic heterogeneity. Nat Methods. 2014;11(8):817–820. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Angermueller C, Clark SJ, Lee HJ, et al. Parallel single-cell sequencing links transcriptional and epigenetic heterogeneity. Nat Methods. 2016;13(3):229–232. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Hou Y, Guo H, Cao C, et al. Single-cell triple omics sequencing reveals genetic, epigenetic, and transcriptomic heterogeneity in hepatocellular carcinomas. Cell Res. 2016;26(3):304–319. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Clark SJ, Argelaguet R, Kapourani CA, et al. scNMT-seq enables joint profiling of chromatin accessibility DNA methylation and transcription in single cells. Nat Commun. 2018;9(1):781. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Cao J, Cusanovich DA, Ramani V, et al. Joint profiling of chromatin accessibility and gene expression in thousands of single cells. Science. 2018;361(6409):1380–1385. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Spitzer MH, Nolan GP. Mass Cytometry: Single Cells, Many Features. Cell. 2016;165(4):780–791. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Han G, Spitzer MH, Bendall SC, Fantl WJ, Nolan GP. Metal-isotope-tagged monoclonal antibodies for high-dimensional mass cytometry. Nat Protoc. 2018;13(10):2121–2148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Maizels RM, McSorley HJ. Regulation of the host immune system by helminth parasites. J Allergy Clin Immunol. 2016;138(3):666–675. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Ippolito MM, Denny JE, Langelier C, Sears CL, Schmidt NW. Malaria and the Microbiome: A Systematic Review. Clin Infect Dis. 2018;67(12):1831–1839. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Hagan T, Cortese M, Rouphael N, et al. Antibiotics-Driven Gut Microbiome Perturbation Alters Immunity to Vaccines in Humans. Cell. 2019;178(6):1313–1328 e1313. [DOI] [PMC free article] [PubMed] [Google Scholar]