Next Article in Journal
Genomic Characterization of Listeria innocua Isolates Recovered from Cattle Farms, Beef Abattoirs, and Retail Outlets in Gauteng Province, South Africa
Previous Article in Journal
The Program for the Control of Visceral Leishmaniasis in Brazil: The Effect of the Systematic Euthanasia of Seropositive Dogs as a Single Control Action in Porteirinha, a Brazilian City with an Intense Transmission of Visceral Leishmaniasis
Previous Article in Special Issue
Unravelling Antigenic Cross-Reactions toward the World of Coronaviruses: Extent of the Stability of Shared Epitopes and SARS-CoV-2 Anti-Spike Cross-Neutralizing Antibodies
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Malaria Genomics, Vaccine Development, and Microbiome

Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
*
Author to whom correspondence should be addressed.
Pathogens 2023, 12(8), 1061; https://doi.org/10.3390/pathogens12081061
Submission received: 21 July 2023 / Revised: 14 August 2023 / Accepted: 15 August 2023 / Published: 18 August 2023
(This article belongs to the Special Issue Models in Parasite and Pathogen Evolution)

Abstract

:
Recent advances in malaria genetics and genomics have transformed many aspects of malaria research in areas of molecular evolution, epidemiology, transmission, host–parasite interaction, drug resistance, pathogenicity, and vaccine development. Here, in addition to introducing some background information on malaria parasite biology, parasite genetics/genomics, and genotyping methods, we discuss some applications of genetic and genomic approaches in vaccine development and in studying interactions with microbiota. Genetic and genomic data can be used to search for novel vaccine targets, design an effective vaccine strategy, identify protective antigens in a whole-organism vaccine, and evaluate the efficacy of a vaccine. Microbiota has been shown to influence disease outcomes and vaccine efficacy; studying the effects of microbiota in pathogenicity and immunity may provide information for disease control. Malaria genetics and genomics will continue to contribute greatly to many fields of malaria research.

1. A Brief Introduction to Human Malaria Species and Life Cycle

Human malaria is a disease caused by Plasmodium parasites, including P. falciparum, P. vivax, P. ovale, P. malariae, and P. knowlesi. Whereas P. vivax is the most widespread species, P. falciparum causes the majority of the mortality, with an estimated 619,000 malaria deaths globally in 2021 [1]. Plasmodium parasites generally have a limited host range, e.g., human malaria parasites do not infect rodent hosts and vice versa, although some parasites can infect both humans and non-human primates (for example, P. knowlesi). Malaria parasites are single-cell eukaryotic organisms with complex life cycles involving the development of asexual and sexual stages (gametocytes) in vertebrate hosts (humans, non-human primates, rodents, birds, lizards, etc.) and sexual differentiation and fertilization in Anopheles mosquitoes [2] (Figure 1). Briefly, when a mosquito takes a blood meal that contains male and female gametocytes, fertilization and genetic exchange can occur in the mosquito midgut. Within the midgut, male and female gametocytes (1n) differentiate into male and female gametes that fertilize to produce zygotes (2n), which then turn into motile ookinetes (4n) after DNA replication. The ookinetes penetrate the mosquito midgut wall to develop into oocysts containing thousands of sporozoites (1n) after meiosis. Mature sporozoites move to the mosquito’s salivary glands and are injected into the skin when the mosquito bites a human host. The motile sporozoites travel from the skin to the liver, where they develop into schizonts containing thousands of merozoites. Mature schizonts release the merozoites into the bloodstream to invade erythrocytes. Within the erythrocytes, parasites develop into rings, trophozoites, and finally merozoites that can infect new erythrocytes when released. Some merozoites will convert into sexual stages, producing male and female gametocytes. Fertilization, DNA replication, and genetic recombination provide biological platforms for studying parasite population structure, transmission dynamics, genetic exchange, and evolution using genomic and genetic tools. The multiple developmental stages in the complex mosquito life cycles also allows for the identification of various potential targets for vaccine development (Figure 1).

2. Malaria Genetics and Genomics

Genomics is defined as “the study of the functions and interactions of all the genes in the genome” [3]. In infectious diseases, genomics includes interactions between the pathogen genomes and their hosts. Large amounts of genomic data are now available, such as genome sequences, proteomics, transcriptomics, and metabonomics from many species or strains of malaria parasites and their hosts. These data are being used to study gene function, gene–gene interaction, genotype–phenotype association, and genetic testing. Epidemiological principles and genomic methods can also be applied to discover genes, to characterize parasite populations, to track parasite origin and disease outbreak, to study parasite evolution history, and to improve vaccine efficacy. The major approaches and applications in malaria genetics and genomics research are summarized in Figure 2.

2.1. Genetic Typing and DNA Sequencing Methods Used in Malaria Genetic and Genomics

Based on the types of genetic variations, various genotyping methods have been developed to study malaria parasite populations, transmission, evolution, and gene function through linkage and association mapping. Electrophoretic variants of enzymes were first introduced to study polymorphisms in rodent malaria parasites [5]. Pulsed-field gel electrophoresis (PFGE) was then employed to show polymorphisms of tens to hundreds of kilobases in size in different P. falciparum isolates [6,7]. Based on restriction enzyme digestion, agarose gel electrophoresis, blotting, and the hybridization of radio-labeled probes, restriction fragment-length polymorphism (RFLP) analysis was used to study recombination rates and linkage groups in a genetic cross [8]. Later, amplified fragment-length polymorphism (AFLP) analysis was developed to genotype and map genes in Plasmodium c. chabaudi parasites [9,10,11]. The AFLP technique also involves digestion of parasite DNA, followed by the ligation of oligonucleotide adapters to the digested DNA, the amplification of the digested DNA fragments, the gel separation of the amplified fragments, and the visualization of the amplified products [9]. Based on the abundance of microsatellites (or simple sequence repeats) in the AT-rich P. falciparum genome, PCR amplification of DNA with a small-size polymorphism (microsatellite) was established to determine the type of P. falciparum isolates [12,13] and to map a determinant linked to chloroquine resistance [14]. With the advances in DNA sequencing technologies, large numbers of single-nucleotide polymorphisms (SNPs) were identified using DNA sequencing [15,16,17]. Based on the SNPs identified from sequencing, microarrays were developed to genotype P. falciparum and P. yoelii parasites [18,19,20,21]. With the reduced costs of DNA sequencing, genome sequencing of parasite DNA samples from patient blood is becoming a practical and popular method of performing genetic studies of malaria parasites [22,23].
DNA sequencing has been categorized into generations. DNA sequencing began using low levels of radioactive isotopes in DNA polymerization reactions [24,25]. The incorporation of radioactive ddNTP nucleotides into the DNA prevented further DNA extension. DNA fragments were visualized using electrophoresis gels. The sequencing reads were generally small—less than one kilobase (kb). Because of the AT-rich P. falciparum genome, sequencing using the isotope methods was very challenging initially. The second-generation sequencing used a luminescent method that measured pyrophosphate synthesis during amplification. This decreased the cost per base sequenced and allowed for higher throughput [26,27]. Light is released when a nucleotide is incorporated, and signals of four different colors representing four nucleotides are recorded. This method allowed for millions of fragments to be sequenced simultaneously per run. However, due to the amount of light produced per reaction, DNA fragments have to be amplified before sequencing reactions. The third-generation sequence allowed for the sequencing of single molecules and therefore eliminated the need for DNA amplification before sequencing [28]. The third-generation sequencing increased the read lengths and decreased the cost and time further. The rise of technologies has enabled the sequencing of the genomes of large numbers of malaria parasites and their hosts. The genotyping and sequencing methods are the key factors that have driven research in malaria genomics.

2.2. Genomes and Chromosomes of Plasmodium Parasites

The genomes of many Plasmodium species infecting humans, nonhuman primates, rodents, and birds have been sequenced. The first Plasmodium genome to be sequenced was the genome of the 3D7 clone of P. falciparum in 2002 [29]. The sequencing revealed an AT-rich genome of 22.8 Mb containing 14 chromosomes and 5300 predicted genes. It is now known that all species of malaria parasites have 14 chromosomes consisting of ~20–35 Mb of DNA, a linear 6 kb mitochondrial genome, and a 35 kb circular plastid genome [30,31,32,33,34,35,36,37,38,39,40,41]. There are also extensive gene syntenies among the malaria parasite species [42,43,44]. Similar genomic structure is an important common feature of the Plasmodium parasites. In contrast, the genome sequences of Plasmodium parasites can be very different in GC content, genome size, and the number of multigene families. For example, the P. falciparum genome is highly AT-rich (~80% AT), whereas the P. vivax genome is relatively more GC-balanced (~58% AT) [30,45], which suggests that Plasmodium parasites have a long and complex evolutionary history [40,46]. The genomes of many parasite strains or isolates of various Plasmodium species, particularly for P. falciparum and P. vivax, have been sequenced, revealing diverse genomes with large numbers of SNPs and indels within a single parasite species [40,47,48]. Like many other microorganisms, the chromosomes of the parasites can be divided into highly variable subtelomeric regions and conserved central regions [40,49]. The subtelomeric regions are known to include hypervariable repetitive sequences that often cause errors in genotyping and sequence alignment. It is not reliable to use the SNPs from the subtelomeric sequences to infer parasite ancestry or population structure due to frequent recombination.

2.3. Within Species Genetic Polymorphisms

Genetic polymorphisms within individual malaria parasite species include chromosome-size polymorphisms, variations in gene copy number, SNPs, indels (insertion or deletion of nucleotides), and nucleotide modifications. Chromosome-size polymorphisms were recognized in the 1980s after the separation of parasite chromosomes in PFGE [5,50,51]. Chromosome-size polymorphisms largely occur due to the deletion and translocation of chromosomal end segments [51]. Variations in the copy number of gene families also contribute to genetic differences in the parasite strains [44,52,53], and these gene families likely play a major role in parasite virulence and immune evasion [54,55,56,57,58,59]. Large numbers of SNPs and indels have been identified from many Plasmodium species, such as P. falciparum, P. vivax, and rodent malaria parasites, through the genome sequencing of parasite isolates or strains [18,40,48,60]. Recently, more than 4.5 million variable positions, including over 3 million SNPs and indels, were identified from 1895 P. vivax isolates collected from 88 worldwide locations [48]. For P. falciparum, a total of 3,125,721 SNPs and 2,742,938 indels were reported from genome sequences of ~20,000 parasite isolates after excluding variants in subtelomeric and internal hypervariable regions, as well as the mitochondrial and apicoplast genomes [47,61]. For an estimated 23 Mb P. falciparum genome, there is approximately one polymorphic site per four bp sequences. However, the polymorphisms are not evenly distributed throughout the genome. Additionally, most of the polymorphic minor alleles are low-frequency (<5%), occurring in one or several parasites and thus suggesting recent mutations. Genes encoding immune and drug targets are likely to have more SNPs due to immune and drug selections [60,62,63], and microsatellites are often found in AT-rich repeat regions [11,12]. Copy number variation can occur under drug pressures; the gene-encoding P. falciparum multidrug resistance 1 (PfMDR-1) can increase its copy number under mefloquine pressure [64,65]. Compared with SNPs, microsatellites generally have higher mutation rates and multiple alleles (in contrast to two alleles for most of the SNPs) and can be used to study recent evolutionary events in parasite populations. Because of maternal inheritance and limited recombination, SNPs and indels from the mitochondrial and plastid genomes are better markers for tracking parasite origin and studying parasite evolution [66,67,68,69,70]. Many genetic polymorphisms can influence gene functions and parasite development. On the other hand, they also provide tools for studying parasite evolution, population dynamics, gene function, and host–parasite interactions.

2.4. Identification of Drug Resistance Genes and Monitoring the Origin and Spread of Drug Resistance

Genetic and genomic tools have been employed to study malaria drug resistance extensively. The studies can be classified into three major directions: first, the identification of drug-resistant genes using linkage and association mappings, including genome-wide association study (GWAS); second, tracking and monitoring the origins and spread of drug-resistant parasites (drug-selective sweeps) using samples collected from patients; and third, drug selection in vitro or in vivo and genome sequencing or mapping of new mutations under drug pressure (Figure 2). These fields have been the most active in malaria research and will continue to receive great attention in the future. There have been many reviews on malaria drug resistance, the genetic mechanisms of resistance, and genetic/genomic approaches used to study drug resistance [71,72,73,74,75,76,77,78]. Readers who are interested in these subjects can consult the reviews listed above.

2.5. Host Genetic Polymorphisms and Protection against Malaria

Genetic variation in parasites leads to different disease outcomes, but host genetics can also impact the severity of the infection. For example, mutations in hemoglobin—leading to sickle cell amenia—or the absence of the Duffy antigen receptor give the host better survival outcomes. Sickle cell amenia is caused by a single amino acid mutation in hemoglobin, which leads to red blood cells having long crescent shapes [79,80]. Sickle red blood cells increase host survival via multiple means, including the increased clearance of infected cells by the spleen [81,82] and the trafficking of parasite proteins to the erythrocyte membrane [83]. While the sickle cell anemia trait led to the clearance of infected cells by the host, the absence of the Duffy antigen receptor prevented erythrocyte infection by P. vivax [84,85,86]. Infections by Plasmodium parasites placed selective pressure on the human host, resulting in mutations in the human genome.

3. Malaria Genomics and Vaccine Development

There are many malaria vaccines under various stages of development. Malaria vaccines can be divided into three major categories on the parasite developmental stages, including pre-erythrocytic vaccines (anti-liver stages), blood-stage vaccines, and transmission-blocking vaccines (anti-sexual stages) (Figure 1). Among the vaccines against each stage, there are single-molecule or multiantigen vaccines, multistage vaccines, and whole-organism-based vaccines [87,88,89,90,91,92,93,94,95]. The most successful malaria vaccine against P. falciparum parasites developed so far is the RTS,S/AS01 which consists of 18 copies of the central repeat and the C-terminal domain of PfCSP fused to the hepatitis B virus surface antigen (HBsAg) [93,94]. The RTS,S vaccine has been recommended for human use by the WHO [95]. However, a sterile malaria vaccine (or 100% protection) is not available currently; more research and development are required to achieve the goal of eradication of malaria through vaccination. Genetic and genomic tools have the potential to contribute significantly to the development of an effective malaria vaccine, providing information for the identification of vaccine targets, improving vaccine design, evaluating vaccine efficacy, and monitoring potential vaccine “resistance” or escape (Figure 3).

3.1. Identification of Vaccine Candidates

Genomic information and tools can be employed to identify candidate vaccine targets and predict T/B cell epitopes [96,97]. Computational methods can be used to search parasite genomes for putative surface-exposed or secreted proteins in an approach called “reverse vaccinology” [96]. A multivalent vaccine was developed against meningitis and sepsis caused by the bacteria Neisseria meningitidis using this approach [98]. Recombinant proteins from candidate genes identified through genome searches can be expressed, purified, and used to immunize mice to produce antibodies. Selected candidates can be checked for sequence conservation across a panel of different parasite strains to identify conserved epitopes or relatively conserved antigens. The ability of the antibodies to protect against a microorganism or parasite are tested in functional assays such as growth inhibition or transmission-blocking assays [99,100] and eventually evaluated in animal models and humans.
Most of the malaria antigen genes are highly polymorphic due to balancing selection, and searches for hotspots of genome diversity may lead to potential unknown immune targets. A survey of 3539 P. falciparum genes for polymorphisms identified some highly polymorphic loci and candidate genes. This included some genes of unknown function that were confirmed to encode antigens recognized by the human immune sera [60]. In another study, a genomic survey of 65 P. falciparum isolates from West Africa identified 2853 genes that contained three or more SNPs [63]. Genes with strong evidence of balancing selection and with peak expression at the stage of red blood cell (RBC) invasion were highlighted, including members of clag, PfMC-2TM, surfin, and msp3-like gene families. The genes under balancing selection can be prioritized for functional study and testing for use in potential vaccines. Unfortunately, highly polymorphic proteins may not be ideal vaccine candidates because of parasite-strain-specific immunity and immune evasion of parasites with variant alleles. However, it may be possible to search for protective antigens that are relatively conserved in parasite populations across geographic regions. Indeed, antigen candidates that had low naturally occurring sequence variation and high coverage across diverse bacterial populations were found after sequencing 2083 group A Streptococcus pyogenes bacterial isolates [101]. The most successful molecular malaria vaccine developed so far, RTS,S, is based on a highly-conserved tandem repeat tetrapeptide (NANP) in the central repeat region of the CSP and a C-terminal region that contains T- and B-cell epitopes, although the central repeat region itself is highly variable in size among parasite isolates [102]. The use of conserved tandem repeat sequences in a variable region in size as a vaccine may be good practice for malaria parasites because there are many proteins with repeated motifs in the parasite genomes, particularly in the P. falciparum genome. With genome sequences from over 20,000 P. falciparum isolates available, it is now possible to search for candidate vaccine targets that have relatively conserved motifs in most of the strains and are expressed in important parasite stages such as merozoite or sporozoite [47,61].
In addition to diversity-based search, experimental genetic crosses of parasites and genome-wide analyses have also been used to identify genes that encode targets of host immunity. In one study using progenies of Plasmodium chabaudi chabaudi genetic crosses, Cheeseman et al. developed a genetic approach called linkage group selection to identify target antigens of strain-specific protective immunity (SSPI) against malaria [103]. The analysis of the frequencies of genome-wide polymorphisms from 35 SSPI selection events on different populations of progeny pools identified a 79 kb genomic region on chromosome 8 as the region controlling SSPI. A gene encoding the merozoite surface protein 1 (MSP-1) within the locus was found to account for >60% of genetic polymorphism and was most frequently under the greatest selection by SSPI [103]. The results are consistent with the fact that MSP-1 is a known malaria antigen that is being used to develop a malaria vaccine [104]. In another study, a key molecule expressed on the P. falciparum merozoite, known as PfRh5, was identified through genetic mapping by the ability of the parasite to invade Aotus monkey erythrocytes as a phenotype [105]. PfRH5 plays an important role in erythrocyte invasion and has become a promising next-generation blood-stage vaccine candidate [106,107].
In addition to candidate antigens, large numbers of linear and discontinuous epitopes of T and B cells and MHCII have been detected in the genomes of Plasmodium species using genomic tools [108]. Many experimentally validated T and B cell and MHCII epitopes were found in well-known antigens such as RIFIN (repetitive interspersed families of polypeptides), STEVOR (subtelomeric variable open reading frame), PfEMP1 (P. falciparum erythrocyte membrane protein 1), MSPs, EBA (erythrocyte-binding antigen), and CSP proteins [108]. Additionally, approximately 50% of the epitopes from each species were predicted to induce IFN-γ, and approximately 26% of unique epitopes were predicted to have IL-10-inducing potential. The epitope analyses of vaccine candidates provide information that is valuable when designing a better vaccine.

3.2. Improving Vaccine Design

A difficulty in developing an effective malaria vaccine is the high level of polymorphism in candidate antigens, which may result in immune escape due to strain-specific immunity. Genomic information can be used to inform the design of malaria vaccines. A molecular vaccine can include multiple alleles of a target antigen to generate antibodies against different parasite strains circulating in a local endemic region as this will allow it overcome immune escape by parasite variants. For example, limited allelic variants (mostly three alleles) were found in genes encoding CSP, MSP-1, AMA-1, LAS-1 (liver stage antigen 1), and TRAP (thrombospondin-related anonymous protein) in 139 P. falciparum isolates collected from Amazon basin in Loreto, Peru [109]. A multivariant vaccine consisting of three major alleles for each specific antigen may be sufficient to cover most of the parasite variants in the regions. The P. falciparum populations in South America are relatively homogeneous due to drug-selective sweeps [110,111]. Parasite populations in Africa are generally more diverse, and more antigen variants are necessary to counter the diverse populations in Africa. However, for some antigens, genetic variation is relatively limited. A recent study showed a low level of genetic polymorphisms in the PfRH5 antigen in Lagos, Nigeria [112]. Sequence analysis revealed three haplotypes of PfRH5 with a negative Tajima’s D of − 1.717 and a dN/dS value of 0.011 (±0.020), suggesting a population expansion after a recent bottleneck and/or the selection on this gene. A vaccine with sequences covering the three PfRH5 haplotypes can be designed to vaccinate the human populations in the Lagos region. A region-specific vaccine based on the specific target antigen variants can be developed to protect the local human populations after a survey of parasite populations. This approach will be more practical for parasites in South America due to relatively homogenous parasite populations.

3.3. Evaluation of Vaccine Efficacy or Identification of Unknown Vaccine Targets

In vaccine trials, it is often necessary to know whether the parasites from patients who received a vaccine have the same genotype as the ones used in vaccine development. For a molecular vaccine based on a single-parasite antigen, it is relatively easy to sequence the antigen coding gene in parasite isolates collected from vaccinated individuals. Several studies investigated the RTS,S vaccine efficacy through sequencing or genotyping the vaccine targets from vaccine recipients and compared the frequencies of matched (3D7 strain) or mismatched sequences [113,114,115,116]. In particular, analysis of polymorphic sites and haplotypes within the CSP gene in samples from an RTS,S/AS01 vaccine trial found that the 1-year cumulative vaccine efficacy was 50.3% against clinical malaria with parasites matching the vaccine sequence in the entire CSP C-terminal, as compared with an efficacy of 33.4% against those with mismatching sequences [116].
For a whole-parasite vaccine such as attenuated sporozoite vaccines [90,117], determining the type of parasite genome using multiple genetic markers or even performing whole-genome sequencing may be necessary before a vaccine trial. In a recent study, whole-genome sequencing was performed to generate de novo genome assemblies for the vaccine strain (NF54) and other heterologous strains used in controlled human malaria infection (CHMI) trials in order to investigate genotypic and immunologic differences [118]. Based on the long genetic distance between the vaccine strain PfNF54 of West Africa and a Brazilian isolate Pf7G8 and drawing from the evaluation of host responses to vaccine trials, it was proposed that Pf7G8 could act as a stringent surrogate for P. falciparum parasites in Africa [119].
Comparison of the genome sequences of the vaccine strain and the strains before and after vaccination may help to identify some key protective epitopes or antigens in whole-organism vaccines such as P. falciparum sporozoite (PfSPZ) vaccines. For a whole-organism vaccine, the protective epitopes or antigens are generally unknown. An approach called ‘sieve analysis’ has been used to compare viral or parasite genetic sequences between vaccine and placebo recipients in order to study breakthrough populations, vaccine efficacy, and potentially identifying vaccine targets [116,120,121]. In the analysis, genetic and statistical approaches are used to measure the dissimilarity between the vaccine sequence and sequences isolated from trial participants, and the dissimilarities are then compared between vaccine and placebo recipients. The radiation-attenuated PfSPZ vaccine has been shown to protect > 90% of subjects against homologous CHMI, although it offers lower rates of protection against heterologous CHMI [122,123,124,125]. However, the critical protective targets or epitopes for homologous protection are currently unknown. Sequencing the genomes of parasite isolates before and after PfSPZ vaccination (if available) and/or between vaccine and placebo recipients may allow for the identification of a collection of protective epitopes via a concept similar to the linkage group selection method used in genetic crosses [126].

3.4. Monitoring Potential Vaccine Selection and Escape

Breakthrough infections in fully vaccinated individuals have been reported for vaccines against SARS-CoV-2 due to ineffective immune responses, waning immunity, or escape from immune recognition by viral evolution [127]. Malaria vaccines such as RTS,S do not induce sterile immunity, and infections of vaccinated individuals are likely to occur. Genome sequencing or genotyping using multiple polymorphic genetic markers can be employed to distinguish vaccine breakthrough parasites from new infections of parasites with similar genetic backgrounds, particularly for those patients vaccinated with a whole-organism vaccine. Partial protective vaccines like RTS,S put pressure on parasite populations and select parasites with CSP alleles that are different from those used in vaccine development. Genomics tools also help to monitor the potential emergence of “vaccine-escaping” parasite strains, which may become a challenge in malaria control. There are no solid data showing that the depletion of a parasite strain with a specific antigen allele will change parasite virulence or transmission capability. However, studies of rodent malaria species or strains clearly show that various degrees of virulence and disease severity (phenotype) can be caused by different strains of parasite species [128,129]. In human infections, disease severity is often the result of infections with a mixture of parasite strains. It is likely that strains of P. falciparum or P. vivax circulating in a human population have different levels of virulence and cause different disease pathologies. For example, the carriage of the msp1-MAD20 or msp2-FC27 allele family was associated with increased susceptibility to severe malaria [130]. Vaccine-targeted depletion of a parasite population or an allele in an individual may change the dynamics of parasite population structure and disease outcomes, particularly when a virulent gene is physically linked to the vaccine target gene, or when the vaccine antigen is a virulent factor itself. Indeed, an antigen used in a vaccine is the target of host immune responses, and different alleles of the antigen may stimulate different host responses. This effect of vaccine pressure may lead to decreased vaccine effectiveness. By surveying changes in the parasite’s genome over time, we can monitor potential parasite population dynamics in the vaccinated regions and predict the emergence of new parasite populations and possibly vaccine efficacy.

3.5. Screening for B Cells Producing Protective Monoclonal Antibodies

Monoclonal antibodies (mAbs) produced by a single B cell population can be used as prophylactic or therapeutic agents against infecting microbes, and exciting results have been reported when using monoclonal antibodies against malaria [131,132,133,134,135,136]. The identification and cloning of monoclonal antibodies that are protective against malaria relied on protocols that allow the efficient amplification, sequencing, and expression of many single B cells from malaria patients [137]. Again, tools of efficient DNA sequencing and genetic analyses are critical to identifying which clonal cells produce effective antibodies for prophylaxis and treatment of malaria.

4. Malaria, Microbiome, and Vaccine Efficacy

An emerging field taking advantage of high-throughput genomic technologies and biocomputational analyses is studying the association of and interaction between microbiomes and various diseases. Microbial communities that inhabit different body parts such as the intestine and skin have been studied intensively for their association with various diseases in recent years [138,139,140], including malaria [141,142,143]. The estimated number of microbial genes associated with the human body is estimated to be 2–20 million, or approximately 100X the ∼20,000 human genes, and the development of high-throughput genome sequencing technologies and computational methods makes it possible to analyze the genomic data from microbiota [144].
Although malaria parasite transmission typically requires and interacts with a vertebrate host and a mosquito vector, microbiota within the vertebrate host (human, mouse, or others) and mosquito can also influence host metabolism and immunity, thereby altering the outcomes of parasite growth and transmission (Figure 4). Elucidating host microbiota and their dynamics during malaria parasite infections may reveal molecular mechanisms that can be explored to develop better disease treatment and management. Studies of both rodent malaria models and human malaria have shown that gut microbiota composition is a factor in disease progression [141,142,143]. For example, the same strain of mice from different vendors infected with the same parasite had profound differences in disease severity, and differences in gut microbiota contributed to the differences in disease susceptibility [145]. Further, germ-free mice transplanted with cecal contents from ‘resistant’ or ‘susceptible’ mice had low and high parasite burdens, respectively. The ‘resistant’ mice had increased abundances of Lactobacillus and Bifidobacterium microbes and an elevated humoral immune response compared with susceptible mice. Additionally, the use of antibiotic treatment and fecal microbiota transplant to change the gut microbiota in outbred Swiss Webster mice also altered the susceptibility to P. c. chabaudi infection and pregnancy outcomes [146]. On the other hand, infection by Plasmodium parasites can induce alterations in the composition of the gut microbiota [143]. Plasmodium berghei (PbA) infection decreased the numbers of Bacteroidetes and Verrucomicrobia species in the mouse gut but increased the numbers of Firmicutes and Proteobacteria species [147].
Increasing evidence from clinical studies and animal models has also shown that the composition of the gut and skin microbiota plays a critical role in modulating immune responses, disease severity, and vaccine efficacy [148,149,150]. The composition of stool bacteria at the beginning of malaria transmission season was associated with susceptibility to P. falciparum infection [151]. Similarly, stool bacteria populations correlate with the severity of malaria in Ugandan children [152]. Children with malaria often have changes in the immune cell architecture of the spleen, which may affect the immunological function of the spleen against encapsulated bacteria [153]. Plasmodium infection can also lead to gut microbiota-dependent increases in splenic germinal center (GC)-associated immune cells in mice and in the titer of parasite-specific antibodies [154]. Malaria parasite sequestration in the lungs of PbA-infected mice results in sustained immune activation and production of the anti-inflammatory cytokine IL-10 by T cells, compromising microbial control and leading to severe lung disease [155]. Interestingly, modulation of mosquito microbiota by the anti-microbiota vaccination of birds against commensal Enterobacteriaceae disrupts P. relictum development within midguts and salivary glands of the vector Culex quinquefasciatus [156]. An increased ratio of Enterobacteriaceae to Bacteroides species was also observed in the responders of a vaccine against rotavirus in a study in Ghana [157]. The differences in the compositions of bacteria nucleic acid and/or lipopolysaccharides may play a role in immune responses during infection or after vaccination. Monitoring the dynamics of the host microbiota on the skin and in the gut using genomic tools may be necessary in order to better understand and improve vaccine efficacy.
Interestingly, the microbiota of the mosquito vector can also affect the transmission of the malaria parasite. One study found that the midguts of An. stephensi had Proteobacteria as the most dominant population among the three laboratory-reared strains of An. Stephensi, and that antibiotic treatment increased susceptibility to PbA infection [158]. Similarly, antibiotics in ingested blood enhanced the susceptibility of Anopheles gambiae mosquitoes to malaria infection by disturbing their gut microbiota [159]. In a follow-up study, the vectorial capacity of malaria mosquitoes was found to be dependent on antibiotics in the ingested blood [160]. Azithromycin could decrease P. falciparum infection load and mosquito lifespan, whereas doxycycline increased the parasite load at high concentrations. Additionally, gut bacteria may impair the effectiveness of mosquito control approaches using insecticide bacteria such as Bacillus thuringiensis subspecies israelensis (Bti) or Wolbachia to alter the fitness or inhibit insect reproductive organs of infected insects [161]. Therefore, studies of malaria genomics need to include the interactions of malaria parasites, their hosts, and the microbiome. There is increasing interest in studying the interaction of malaria and microbiota, and the continuous advance of genomic technologies and computational power will certainly advance the field of malaria and microbiota studies.

5. Conclusions and Perspectives

Malaria genetics and genomics are important approaches for studying disease mechanisms, host–parasite interaction, gene function, drug resistance, parasite population structure and transmission, and for developing better vaccines. Although great progress has been made in malaria genomics and the applications of genomics in studying various aspects of the disease, there are some areas where exciting discoveries may be obtained by applying genomic methods. In particular, analysis of parasite populations before and after vaccination (vaccine and placebo recipients) using a whole-organism vaccine may lead to the detection of immune targets if the vaccinated individuals are infected with multiple parasite strains. Multiple infections with two to eight P. falciparum clones were frequently detected in Africa [162], and diverse circulating parasite strains with frequent occurrence of multiple infections were observed in Nanoro, Burkina Faso [130]. If a vaccine removes some specific parasite strains, potential selection valleys can be detected. The presence of polymorphisms in vaccine molecules is another problem in developing an effective vaccine. It may be possible to design a vaccine that targets the local parasite populations after genotyping parasites in an endemic area. This approach may be practical for countries in South America where parasite populations are less diverse than those in Africa due to selective sweeps by chloroquine and other drugs [110,111]. After the selection of candidate antigens, parasite population surveys can be conducted to identify major alleles circulating in the local populations. A multivariant vaccine with limited variant sequences can be designed based on the major alleles in the endemic region. Finally, the genomes of microbiota have been shown to play a critical role in shaping malaria disease severity and vaccine efficacy, which brings a third dimension to the study of malaria genomics (in addition to parasite and host genomes). The study of microbiome genomics and interactions with malaria parasites and their hosts will enhance our understanding of malaria pathogenesis and the mechanism of disease severity. It can be expected that genomic and genetic tools will continue to contribute greatly to vaccine development and malaria elimination.

Author Contributions

X.S., conceived the manuscript and wrote the first draft. R.V.S. and F.X., writing, and J.W., drew Figure 1 and wrote the legend and improved other figures. All authors read and edited the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), USA.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

All data are included in the article.

Acknowledgments

We also thank Yolanda L. Jones, NIH Library Editing Services, for manuscript editing assistance.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. WHO. World Malaria Report 2022. Available online: https://www.who.int/teams/global-malaria-programme/reports/world-malaria-report-2022 (accessed on 15 July 2023).
  2. Su, X.Z.; Zhang, C.; Joy, D.A. Host-Malaria Parasite Interactions and Impacts on Mutual Evolution. Front. Cell. Infect. Microbiol. 2020, 10, 587933. [Google Scholar] [CrossRef] [PubMed]
  3. Khoury, M.J.; Millikan, R.; Little, J.; Gwinn, M. The emergence of epidemiology in the genomics age. Int. J. Epidemiol. 2004, 33, 936–944. [Google Scholar] [CrossRef] [PubMed]
  4. Carter, R. Enzyme variation in Plasmodium berghei. Trans. R. Soc. Trop. Med. Hyg. 1970, 64, 401–406. [Google Scholar] [CrossRef]
  5. Wu, J.; Cai, B.; Sun, W.; Huang, R.; Liu, X.; Lin, M.; Pattaradilokrat, S.; Martin, S.; Qi, Y.; Nair, S.C.; et al. Genome-wide Analysis of Host-Plasmodium yoelii Interactions Reveals Regulators of the Type I Interferon Response. Cell Rep. 2015, 12, 661–672. [Google Scholar] [CrossRef] [PubMed]
  6. Kemp, D.J.; Corcoran, L.M.; Coppel, R.L.; Stahl, H.D.; Bianco, A.E.; Brown, G.V.; Anders, R.F. Size variation in chromosomes from independent cultured isolates of Plasmodium falciparum. Nature 1985, 315, 347–350. [Google Scholar] [CrossRef] [PubMed]
  7. Corcoran, L.M.; Forsyth, K.P.; Bianco, A.E.; Brown, G.V.; Kemp, D.J. Chromosome size polymorphisms in Plasmodium falciparum can involve deletions and are frequent in natural parasite populations. Cell 1986, 44, 87–95. [Google Scholar] [CrossRef]
  8. Walker-Jonah, A.; Dolan, S.A.; Gwadz, R.W.; Panton, L.J.; Wellems, T.E. An RFLP map of the Plasmodium falciparum genome, recombination rates and favored linkage groups in a genetic cross. Mol. Biochem. Parasitol. 1992, 51, 313–320. [Google Scholar] [CrossRef]
  9. Vos, P.; Hogers, R.; Bleeker, M.; Reijans, M.; van de Lee, T.; Hornes, M.; Frijters, A.; Pot, J.; Peleman, J.; Kuiper, M.; et al. AFLP: A new technique for DNA fingerprinting. Nucleic Acids Res. 1995, 23, 4407–4414. [Google Scholar] [CrossRef]
  10. Grech, K.; Martinelli, A.; Pathirana, S.; Walliker, D.; Hunt, P.; Carter, R. Numerous, robust genetic markers for Plasmodium chabaudi by the method of amplified fragment length polymorphism. Mol. Biochem. Parasitol. 2002, 123, 95–104. [Google Scholar] [CrossRef]
  11. Martinelli, A.; Hunt, P.; Cheesman, S.J.; Carter, R. Amplified fragment length polymorphism measures proportions of malaria parasites carrying specific alleles in complex genetic mixtures. Mol. Biochem. Parasitol. 2004, 136, 117–122. [Google Scholar] [CrossRef]
  12. Su, X.; Wellems, T.E. Toward a high-resolution Plasmodium falciparum linkage map: Polymorphic markers from hundreds of simple sequence repeats. Genomics 1996, 33, 430–444. [Google Scholar] [CrossRef] [PubMed]
  13. Su, X.; Ferdig, M.T.; Huang, Y.; Huynh, C.Q.; Liu, A.; You, J.; Wootton, J.C.; Wellems, T.E. A genetic map and recombination parameters of the human malaria parasite Plasmodium falciparum. Science 1999, 286, 1351–1353. [Google Scholar] [CrossRef] [PubMed]
  14. Su, X.; Kirkman, L.A.; Fujioka, H.; Wellems, T.E. Complex polymorphisms in an approximately 330 kDa protein are linked to chloroquine-resistant P. falciparum in Southeast Asia and Africa. Cell 1997, 91, 593–603. [Google Scholar] [CrossRef] [PubMed]
  15. Mu, J.; Duan, J.; Makova, K.D.; Joy, D.A.; Huynh, C.Q.; Branch, O.H.; Li, W.H.; Su, X.Z. Chromosome-wide SNPs reveal an ancient origin for Plasmodium falciparum. Nature 2002, 418, 323–326. [Google Scholar] [CrossRef]
  16. Jeffares, D.C.; Pain, A.; Berry, A.; Cox, A.V.; Stalker, J.; Ingle, C.E.; Thomas, A.; Quail, M.A.; Siebenthall, K.; Uhlemann, A.C.; et al. Genome variation and evolution of the malaria parasite Plasmodium falciparum. Nat. Genet. 2007, 39, 120–125. [Google Scholar] [CrossRef]
  17. Volkman, S.K.; Sabeti, P.C.; DeCaprio, D.; Neafsey, D.E.; Schaffner, S.F.; Milner, D.A., Jr.; Daily, J.P.; Sarr, O.; Ndiaye, D.; Ndir, O.; et al. A genome-wide map of diversity in Plasmodium falciparum. Nat. Genet. 2007, 39, 113–119. [Google Scholar] [CrossRef]
  18. Jacob, C.G.; Tan, J.C.; Miller, B.A.; Tan, A.; Takala-Harrison, S.; Ferdig, M.T.; Plowe, C.V. A microarray platform and novel SNP calling algorithm to evaluate Plasmodium falciparum field samples of low DNA quantity. BMC Genom. 2014, 15, 719. [Google Scholar] [CrossRef]
  19. Nair, S.C.; Pattaradilokrat, S.; Zilversmit, M.M.; Dommer, J.; Nagarajan, V.; Stephens, M.T.; Xiao, W.; Tan, J.C.; Su, X.Z. Genome-wide polymorphisms and development of a microarray platform to detect genetic variations in Plasmodium yoelii. Mol. Biochem. Parasitol. 2014, 194, 9–15. [Google Scholar] [CrossRef]
  20. Mu, J.; Myers, R.A.; Jiang, H.; Liu, S.; Ricklefs, S.; Waisberg, M.; Chotivanich, K.; Wilairatana, P.; Krudsood, S.; White, N.J.; et al. Plasmodium falciparum genome-wide scans for positive selection, recombination hot spots and resistance to antimalarial drugs. Nat. Genet. 2010, 42, 268–271. [Google Scholar] [CrossRef]
  21. Takala-Harrison, S.; Clark, T.G.; Jacob, C.G.; Cummings, M.P.; Miotto, O.; Dondorp, A.M.; Fukuda, M.M.; Nosten, F.; Noedl, H.; Imwong, M.; et al. Genetic loci associated with delayed clearance of Plasmodium falciparum following artemisinin treatment in Southeast Asia. Proc. Natl. Acad. Sci. USA 2013, 110, 240–245. [Google Scholar] [CrossRef]
  22. Neafsey, D.E.; Galinsky, K.; Jiang, R.H.; Young, L.; Sykes, S.M.; Saif, S.; Gujja, S.; Goldberg, J.M.; Young, S.; Zeng, Q.; et al. The malaria parasite Plasmodium vivax exhibits greater genetic diversity than Plasmodium falciparum. Nat. Genet. 2012, 44, 1046–1050. [Google Scholar] [CrossRef] [PubMed]
  23. Miotto, O.; Amato, R.; Ashley, E.A.; MacInnis, B.; Almagro-Garcia, J.; Amaratunga, C.; Lim, P.; Mead, D.; Oyola, S.O.; Dhorda, M.; et al. Genetic architecture of artemisinin-resistant Plasmodium falciparum. Nat. Genet. 2015, 47, 226–234. [Google Scholar] [CrossRef] [PubMed]
  24. Jacobson, K.B.; Arlinghaus, H.F.; Schmitt, H.W.; Sachleben, R.A.; Brown, G.M.; Thonnard, N.; Sloop, F.V.; Foote, R.S.; Larimer, F.W.; Woychik, R.P.; et al. An approach to the use of stable isotopes for DNA sequencing. Genomics 1991, 9, 51–59. [Google Scholar] [CrossRef] [PubMed]
  25. Sanger, F.; Nicklen, S.; Coulson, A.R. DNA sequencing with chain-terminating inhibitors. Proc. Natl. Acad. Sci. USA 1977, 74, 5463–5467. [Google Scholar] [CrossRef] [PubMed]
  26. Ronaghi, M.; Uhlén, M.; Nyrén, P. A Sequencing Method Based on Real-Time Pyrophosphate. Science 1998, 281, 363–365. [Google Scholar] [CrossRef]
  27. Voelkerding, K.V.; Dames, S.A.; Durtschi, J.D. Next-Generation Sequencing: From Basic Research to Diagnostics. Clin. Chem. 2009, 55, 641–658. [Google Scholar] [CrossRef]
  28. Eid, J.; Fehr, A.; Gray, J.; Luong, K.; Lyle, J.; Otto, G.; Peluso, P.; Rank, D.; Baybayan, P.; Bettman, B.; et al. Real-Time DNA Sequencing from Single Polymerase Molecules. Science 2009, 323, 133–138. [Google Scholar] [CrossRef]
  29. Gardner, M.J.; Hall, N.; Fung, E.; White, O.; Berriman, M.; Hyman, R.W.; Carlton, J.M.; Pain, A.; Nelson, K.E.; Bowman, S.; et al. Genome sequence of the human malaria parasite Plasmodium falciparum. Nature 2002, 419, 498–511. [Google Scholar] [CrossRef]
  30. Carlton, J.M.; Angiuoli, S.V.; Suh, B.B.; Kooij, T.W.; Pertea, M.; Silva, J.C.; Ermolaeva, M.D.; Allen, J.E.; Selengut, J.D.; Koo, H.L.; et al. Genome sequence and comparative analysis of the model rodent malaria parasite Plasmodium yoelii yoelii. Nature 2002, 419, 512–519. [Google Scholar] [CrossRef]
  31. Pain, A.; Bohme, U.; Berry, A.E.; Mungall, K.; Finn, R.D.; Jackson, A.P.; Mourier, T.; Mistry, J.; Pasini, E.M.; Aslett, M.A.; et al. The genome of the simian and human malaria parasite Plasmodium knowlesi. Nature 2008, 455, 799–803. [Google Scholar] [CrossRef]
  32. Lapp, S.A.; Geraldo, J.A.; Chien, J.T.; Ay, F.; Pakala, S.B.; Batugedara, G.; Humphrey, J.; Ma, H.c.; De, B.J.; Le Roch, K.G.; et al. PacBio assembly of a Plasmodium knowlesi genome sequence with Hi-C correction and manual annotation of the SICAvar gene family. Parasitology 2018, 145, 71–84. [Google Scholar] [CrossRef] [PubMed]
  33. Otto, T.D.; Bohme, U.; Jackson, A.P.; Hunt, M.; Franke-Fayard, B.; Hoeijmakers, W.A.; Religa, A.A.; Robertson, L.; Sanders, M.; Ogun, S.A.; et al. A comprehensive evaluation of rodent malaria parasite genomes and gene expression. BMC Biol. 2014, 12, 86. [Google Scholar] [CrossRef] [PubMed]
  34. Otto, T.D.; Rayner, J.C.; Bohme, U.; Pain, A.; Spottiswoode, N.; Sanders, M.; Quail, M.; Ollomo, B.; Renaud, F.; Thomas, A.W.; et al. Genome sequencing of chimpanzee malaria parasites reveals possible pathways of adaptation to human hosts. Nat. Commun. 2014, 5, 4754. [Google Scholar] [CrossRef]
  35. Ansari, H.R.; Templeton, T.J.; Subudhi, A.K.; Ramaprasad, A.; Tang, J.; Lu, F.; Naeem, R.; Hashish, Y.; Oguike, M.C.; Benavente, E.D.; et al. Genome-scale comparison of expanded gene families in Plasmodium ovale wallikeri and Plasmodium ovale curtisi with Plasmodium malariae and with other Plasmodium species. Int. J. Parasitol. 2016, 46, 685–696. [Google Scholar] [CrossRef] [PubMed]
  36. Auburn, S.; Bohme, U.; Steinbiss, S.; Trimarsanto, H.; Hostetler, J.; Sanders, M.; Gao, Q.; Nosten, F.; Newbold, C.I.; Berriman, M.; et al. A new Plasmodium vivax reference sequence with improved assembly of the subtelomeres reveals an abundance of pir genes. Wellcome Open Res. 2016, 1, 4. [Google Scholar] [CrossRef]
  37. Rutledge, G.G.; Bohme, U.; Sanders, M.; Reid, A.J.; Cotton, J.A.; Maiga-Ascofare, O.; Djimde, A.A.; Apinjoh, T.O.; Amenga-Etego, L.; Manske, M.; et al. Plasmodium malariae and P. ovale genomes provide insights into malaria parasite evolution. Nature 2017, 542, 101–104. [Google Scholar] [CrossRef]
  38. Pasini, E.M.; Bohme, U.; Rutledge, G.G.; Voorberg-Van der Wel, A.; Sanders, M.; Berriman, M.; Kocken, C.H.; Otto, T.D. An improved Plasmodium cynomolgi genome assembly reveals an unexpected methyltransferase gene expansion. Wellcome Open Res. 2017, 2, 42. [Google Scholar] [CrossRef]
  39. Bohme, U.; Otto, T.D.; Sanders, M.; Newbold, C.I.; Berriman, M. Progression of the canonical reference malaria parasite genome from 2002–2019. Wellcome Open Res. 2019, 4, 58. [Google Scholar] [CrossRef]
  40. Daron, J.; Boissiere, A.; Boundenga, L.; Ngoubangoye, B.; Houze, S.; Arnathau, C.; Sidobre, C.; Trape, J.F.; Durand, P.; Renaud, F.; et al. Population genomic evidence of Plasmodium vivax Southeast Asian origin. Sci. Adv. 2021, 7, eabc3713. [Google Scholar] [CrossRef]
  41. Bajic, M.; Ravishankar, S.; Sheth, M.; Rowe, L.A.; Pacheco, M.A.; Patel, D.S.; Batra, D.; Loparev, V.; Olsen, C.; Escalante, A.A.; et al. The first complete genome of the simian malaria parasite Plasmodium brasilianum. Sci. Rep. 2022, 12, 19802. [Google Scholar] [CrossRef]
  42. Carlton, J.M.; Vinkenoog, R.; Waters, A.P.; Walliker, D. Gene synteny in species of Plasmodium. Mol. Biochem. Parasitol. 1998, 93, 285–294. [Google Scholar] [CrossRef]
  43. Kooij, T.W.; Carlton, J.M.; Bidwell, S.L.; Hall, N.; Ramesar, J.; Janse, C.J.; Waters, A.P. A Plasmodium whole-genome synteny map: Indels and synteny breakpoints as foci for species-specific genes. PLoS Pathog. 2005, 1, e44. [Google Scholar] [CrossRef]
  44. Otto, T.D.; Gilabert, A.; Crellen, T.; Bohme, U.; Arnathau, C.; Sanders, M.; Oyola, S.O.; Okouga, A.P.; Boundenga, L.; Willaume, E.; et al. Genomes of all known members of a Plasmodium subgenus reveal paths to virulent human malaria. Nat. Microbiol. 2018, 3, 687–697. [Google Scholar] [CrossRef] [PubMed]
  45. Carlton, J.M.; Adams, J.H.; Silva, J.C.; Bidwell, S.L.; Lorenzi, H.; Caler, E.; Crabtree, J.; Angiuoli, S.V.; Merino, E.F.; Amedeo, P.; et al. Comparative genomics of the neglected human malaria parasite Plasmodium vivax. Nature 2008, 455, 757–763. [Google Scholar] [CrossRef] [PubMed]
  46. Nikbakht, H.; Xia, X.; Hickey, D.A. The evolution of genomic GC content undergoes a rapid reversal within the genus Plasmodium. Genome 2014, 57, 507–511. [Google Scholar] [CrossRef]
  47. MalariaGEN; Ahouidi, A.; Ali, M.; Almagro-Garcia, J.; Amambua-Ngwa, A.; Amaratunga, C.; Amato, R.; Amenga-Etego, L.; Andagalu, B.; Anderson, T.J.C.; et al. An open dataset of Plasmodium falciparum genome variation in 7000 worldwide samples. Wellcome Open Res. 2021, 6, 42. [Google Scholar] [CrossRef]
  48. MalariaGEN; Adam, I.; Alam, M.S.; Alemu, S.; Amaratunga, C.; Amato, R.; Andrianaranjaka, V.; Anstey, N.M.; Aseffa, A.; Ashley, E.; et al. An open dataset of Plasmodium vivax genome variation in 1,895 worldwide samples. Wellcome Open Res. 2022, 7, 136. [Google Scholar] [CrossRef] [PubMed]
  49. Otto, T.D.; Bohme, U.; Sanders, M.; Reid, A.; Bruske, E.I.; Duffy, C.W.; Bull, P.C.; Pearson, R.D.; Abdi, A.; Dimonte, S.; et al. Long read assemblies of geographically dispersed Plasmodium falciparum isolates reveal highly structured subtelomeres. Wellcome Open Res. 2018, 3, 52. [Google Scholar] [CrossRef]
  50. Sharkey, A.; Langsley, G.; Patarapotikul, J.; Mercereau-Puijalon, O.; McLean, A.P.; Walliker, D. Chromosome size variation in the malaria parasite of rodents, Plasmodium chabaudi. Mol. Biochem. Parasitol. 1988, 28, 47–54. [Google Scholar] [CrossRef]
  51. Pologe, L.G.; Ravetch, J.V. Large deletions result from breakage and healing of P. falciparum chromosomes. Cell 1988, 55, 869–874. [Google Scholar] [CrossRef]
  52. Kyes, S.A.; Rowe, J.A.; Kriek, N.; Newbold, C.I. Rifins: A second family of clonally variant proteins expressed on the surface of red cells infected with Plasmodium falciparum. Proc. Natl. Acad. Sci. USA 1999, 96, 9333–9338. [Google Scholar] [CrossRef] [PubMed]
  53. Fischer, K.; Chavchich, M.; Huestis, R.; Wilson, D.W.; Kemp, D.J.; Saul, A. Ten families of variant genes encoded in subtelomeric regions of multiple chromosomes of Plasmodium chabaudi, a malaria species that undergoes antigenic variation in the laboratory mouse. Mol. Microbiol. 2003, 48, 1209–1223. [Google Scholar] [CrossRef] [PubMed]
  54. Smith, J.D.; Chitnis, C.E.; Craig, A.G.; Roberts, D.J.; Hudson-Taylor, D.E.; Peterson, D.S.; Pinches, R.; Newbold, C.I.; Miller, L.H. Switches in expression of Plasmodium falciparum var genes correlate with changes in antigenic and cytoadherent phenotypes of infected erythrocytes. Cell 1995, 82, 101–110. [Google Scholar] [CrossRef] [PubMed]
  55. Su, X.Z.; Heatwole, V.M.; Wertheimer, S.P.; Guinet, F.; Herrfeldt, J.A.; Peterson, D.S.; Ravetch, J.A.; Wellems, T.E. The large diverse gene family var encodes proteins involved in cytoadherence and antigenic variation of Plasmodium falciparum-infected erythrocytes. Cell 1995, 82, 89–100. [Google Scholar] [CrossRef]
  56. Baruch, D.I.; Pasloske, B.L.; Singh, H.B.; Bi, X.; Ma, X.C.; Feldman, M.; Taraschi, T.F.; Howard, R.J. Cloning the P. falciparum gene encoding PfEMP1, a malarial variant antigen and adherence receptor on the surface of parasitized human erythrocytes. Cell 1995, 82, 77–87. [Google Scholar] [CrossRef]
  57. Larremore, D.B.; Sundararaman, S.A.; Liu, W.; Proto, W.R.; Clauset, A.; Loy, D.E.; Speede, S.; Plenderleith, L.J.; Sharp, P.M.; Hahn, B.H.; et al. Ape parasite origins of human malaria virulence genes. Nat. Commun. 2015, 6, 8368. [Google Scholar] [CrossRef]
  58. Lavazec, C.; Sanyal, S.; Templeton, T.J. Expression switching in the stevor and Pfmc-2TM superfamilies in Plasmodium falciparum. Mol. Microbiol. 2007, 64, 1621–1634. [Google Scholar] [CrossRef]
  59. Ghorbal, M.; Scheidig-Benatar, C.; Bouizem, S.; Thomas, C.; Paisley, G.; Faltermeier, C.; Liu, M.; Scherf, A.; Lopez-Rubio, J.J.; Gopaul, D.N. Initial characterization of the Pf-Int recombinase from the malaria parasite Plasmodium falciparum. PLoS ONE 2012, 7, e46507. [Google Scholar] [CrossRef]
  60. Mu, J.; Awadalla, P.; Duan, J.; McGee, K.M.; Keebler, J.; Seydel, K.; McVean, G.A.; Su, X.Z. Genome-wide variation and identification of vaccine targets in the Plasmodium falciparum genome. Nat. Genet. 2007, 39, 126–130. [Google Scholar] [CrossRef]
  61. MalariaGEN; Abdel Hamid, M.M.; Abdelraheem, M.H.; Acheampong, D.O.; Ahouidi, A.; Ali, M.; Almagro-Garcia, J.; Amambua-Ngwa, A.; Amaratunga, C.; Amenga-Etego, L.; et al. Pf7: An open dataset of Plasmodium falciparum genome variation in 20,000 worldwide samples. Wellcome Open Res. 2023, 8, 22. [Google Scholar] [CrossRef]
  62. Amambua-Ngwa, A.; Park, D.J.; Volkman, S.K.; Barnes, K.G.; Bei, A.K.; Lukens, A.K.; Sene, P.; Van Tyne, D.; Ndiaye, D.; Wirth, D.F.; et al. SNP genotyping identifies new signatures of selection in a deep sample of West African Plasmodium falciparum malaria parasites. Mol. Biol. Evol. 2012, 29, 3249–3253. [Google Scholar] [CrossRef]
  63. Amambua-Ngwa, A.; Tetteh, K.K.; Manske, M.; Gomez-Escobar, N.; Stewart, L.B.; Deerhake, M.E.; Cheeseman, I.H.; Newbold, C.I.; Holder, A.A.; Knuepfer, E.; et al. Population genomic scan for candidate signatures of balancing selection to guide antigen characterization in malaria parasites. PLoS Genet. 2012, 8, e1002992. [Google Scholar] [CrossRef]
  64. Price, R.; Robinson, G.; Brockman, A.; Cowman, A.; Krishna, S. Assessment of pfmdr 1 gene copy number by tandem competitive polymerase chain reaction. Mol. Biochem. Parasitol. 1997, 85, 161–169. [Google Scholar] [CrossRef]
  65. Kiwuwa, M.S.; Byarugaba, J.; Wahlgren, M.; Kironde, F. Detection of copy number variation and single nucleotide polymorphisms in genes involved in drug resistance and other phenotypic traits in P. falciparum clinical isolates collected from Uganda. Acta Trop. 2013, 125, 269–275. [Google Scholar] [CrossRef] [PubMed]
  66. Escalante, A.A.; Freeland, D.E.; Collins, W.E.; Lal, A.A. The evolution of primate malaria parasites based on the gene encoding cytochrome b from the linear mitochondrial genome. Proc. Natl. Acad. Sci. USA 1998, 95, 8124–8129. [Google Scholar] [CrossRef] [PubMed]
  67. Conway, D.J.; Fanello, C.; Lloyd, J.M.; Al-Joubori, B.M.; Baloch, A.H.; Somanath, S.D.; Roper, C.; Oduola, A.M.; Mulder, B.; Povoa, M.M.; et al. Origin of Plasmodium falciparum malaria is traced by mitochondrial DNA. Mol. Biochem. Parasitol. 2000, 111, 163–171. [Google Scholar] [CrossRef] [PubMed]
  68. Joy, D.A.; Feng, X.; Mu, J.; Furuya, T.; Chotivanich, K.; Krettli, A.U.; Ho, M.; Wang, A.; White, N.J.; Suh, E.; et al. Early origin and recent expansion of Plasmodium falciparum. Science 2003, 300, 318–321. [Google Scholar] [CrossRef]
  69. Conway, D.J. Tracing the dawn of Plasmodium falciparum with mitochondrial genome sequences. Trends Genet. 2003, 19, 671–674. [Google Scholar] [CrossRef]
  70. Rodrigues, P.T.; Alves, J.M.; Santamaria, A.M.; Calzada, J.E.; Xayavong, M.; Parise, M.; da Silva, A.J.; Ferreira, M.U. Using mitochondrial genome sequences to track the origin of imported Plasmodium vivax infections diagnosed in the United States. Am. J. Trop. Med. Hyg. 2014, 90, 1102–1108. [Google Scholar] [CrossRef]
  71. Cui, L.; Mharakurwa, S.; Ndiaye, D.; Rathod, P.K.; Rosenthal, P.J. Antimalarial Drug Resistance: Literature Review and Activities and Findings of the ICEMR Network. Am. J. Trop. Med. Hyg. 2015, 93, 57–68. [Google Scholar] [CrossRef]
  72. Su, X.Z.; Wu, J.; Xu, F.; Pattaradilokrat, S. Genetic mapping of determinants in drug resistance, virulence, disease susceptibility, and interaction of host-rodent malaria parasites. Parasitol. Int. 2022, 91, 102637. [Google Scholar] [CrossRef] [PubMed]
  73. Su, X.Z.; Lane, K.D.; Xia, L.; Sa, J.M.; Wellems, T.E. Plasmodium Genomics and Genetics: New Insights into Malaria Pathogenesis, Drug Resistance, Epidemiology, and Evolution. Clin. Microbiol. Rev. 2019, 32, e00019-19. [Google Scholar] [CrossRef] [PubMed]
  74. Anderson, T.; Nkhoma, S.; Ecker, A.; Fidock, D. How can we identify parasite genes that underlie antimalarial drug resistance? Pharmacogenomics 2011, 12, 59–85. [Google Scholar] [CrossRef] [PubMed]
  75. Rocamora, F.; Winzeler, E.A. Genomic Approaches to Drug Resistance in Malaria. Annu. Rev. Microbiol. 2020, 74, 761–786. [Google Scholar] [CrossRef]
  76. Chaturvedi, R.; Chhibber-Goel, J.; Verma, I.; Gopinathan, S.; Parvez, S.; Sharma, A. Geographical spread and structural basis of sulfadoxine-pyrimethamine drug-resistant malaria parasites. Int. J. Parasitol. 2021, 51, 505–525. [Google Scholar] [CrossRef]
  77. Sen, S.; Ferdig, M. QTL analysis for discovery of genes involved in drug responses. Curr. Drug Targets Infect. Disord. 2004, 4, 53–63. [Google Scholar] [CrossRef]
  78. Hayton, K.; Su, X.Z. Drug resistance and genetic mapping in Plasmodium falciparum. Curr. Genet. 2008, 54, 223–239. [Google Scholar] [CrossRef]
  79. Modiano, D.; Luoni, G.; Sirima, B.S.; Simpore, J.; Verra, F.; Konate, A.; Rastrelli, E.; Olivieri, A.; Calissano, C.; Paganotti, G.M. Haemoglobin C protects against clinical Plasmodium falciparum malaria. Nature 2001, 414, 305–308. [Google Scholar] [CrossRef]
  80. Aidoo, M.; Terlouw, D.J.; Kolczak, M.S.; McElroy, P.D.; Ter Kuile, F.O.; Kariuki, S.; Nahlen, B.L.; Lal, A.A.; Udhayakumar, V. Protective effects of the sickle cell gene against malaria morbidity and mortality. Lancet 2002, 359, 1311–1312. [Google Scholar] [CrossRef]
  81. Luzzatto, L.; Nwachuku-Jarrett, E.S.; Reddy, S. Increased sickling of parasitised erythrocytes as mechanism of resistance against malaria in the sickle-cell trait. Lancet 1970, 1, 319–321. [Google Scholar] [CrossRef]
  82. Mackey, J.; Vivarelli, F. Sickle-cell anaemia. Br. Med. J. 1954, 1, 276. [Google Scholar] [CrossRef]
  83. Cyrklaff, M.; Sanchez, C.P.; Kilian, N.; Bisseye, C.; Simpore, J.; Frischknecht, F.; Lanzer, M. Hemoglobins S and C Interfere with Actin Remodeling in Plasmodium falciparum Infected Erythrocytes. Science 2011, 334, 1283–1286. [Google Scholar] [CrossRef]
  84. Miller, L.H.; Mason, S.J.; Dvorak, J.A.; McGinniss, M.H.; Rothman, I.K. Erythrocyte receptors for (Plasmodium knowlesi) malaria: Duffy blood group determinants. Science 1975, 189, 561–563. [Google Scholar] [CrossRef] [PubMed]
  85. Miller, L.H.; Mason, S.J.; Clyde, D.F.; McGinniss, M.H. The resistance factor to Plasmodium vivax in blacks: The Duffy-blood-group genotype, FyFy. N. Engl. J. Med. 1976, 295, 302–304. [Google Scholar] [CrossRef]
  86. Chitnis, C.E.; Miller, L.H. Identification of the erythrocyte binding domains of Plasmodium vivax and Plasmodium knowlesi proteins involved in erythrocyte invasion. J. Exp. Med. 1994, 180, 497–506. [Google Scholar] [CrossRef] [PubMed]
  87. Frimpong, A.; Kusi, K.A.; Ofori, M.F.; Ndifon, W. Novel Strategies for Malaria Vaccine Design. Front. Immunol. 2018, 9, 2769. [Google Scholar] [CrossRef] [PubMed]
  88. Laurens, M.B. The Promise of a Malaria Vaccine-Are We Closer? Annu. Rev. Microbiol. 2018, 72, 273–292. [Google Scholar] [CrossRef] [PubMed]
  89. Beeson, J.G.; Kurtovic, L.; Dobano, C.; Opi, D.H.; Chan, J.A.; Feng, G.; Good, M.F.; Reiling, L.; Boyle, M.J. Challenges and strategies for developing efficacious and long-lasting malaria vaccines. Sci. Transl. Med. 2019, 11, eaau1458. [Google Scholar] [CrossRef]
  90. Nunes-Cabaco, H.; Moita, D.; Prudencio, M. Five decades of clinical assessment of whole-sporozoite malaria vaccines. Front. Immunol. 2022, 13, 977472. [Google Scholar] [CrossRef]
  91. Duffy, P.E. Making a good malaria vaccine better. Trends Parasitol. 2022, 38, 9–10. [Google Scholar] [CrossRef]
  92. Tachibana, M.; Takashima, E.; Morita, M.; Sattabongkot, J.; Ishino, T.; Culleton, R.; Torii, M.; Tsuboi, T. Plasmodium vivax transmission-blocking vaccines: Progress, challenges and innovation. Parasitol. Int. 2022, 87, 102525. [Google Scholar] [CrossRef] [PubMed]
  93. Draper, S.J.; Sack, B.K.; King, C.R.; Nielsen, C.M.; Rayner, J.C.; Higgins, M.K.; Long, C.A.; Seder, R.A. Malaria Vaccines: Recent Advances and New Horizons. Cell Host Microbe 2018, 24, 43–56. [Google Scholar] [CrossRef]
  94. Beeson, J.G.; Kurtovic, L.; Valim, C.; Asante, K.P.; Boyle, M.J.; Mathanga, D.; Dobano, C.; Moncunill, G. The RTS,S malaria vaccine: Current impact and foundation for the future. Sci. Transl. Med. 2022, 14, eabo6646. [Google Scholar] [CrossRef] [PubMed]
  95. Zavala, F. RTS,S: The first malaria vaccine. J. Clin. Investig. 2022, 132, e156588. [Google Scholar] [CrossRef] [PubMed]
  96. Bambini, S.; Rappuoli, R. The use of genomics in microbial vaccine development. Drug Discov. Today 2009, 14, 252–260. [Google Scholar] [CrossRef]
  97. Conway, D.J. Paths to a malaria vaccine illuminated by parasite genomics. Trends Genet. 2015, 31, 97–107. [Google Scholar] [CrossRef]
  98. Giuliani, M.M.; Adu-Bobie, J.; Comanducci, M.; Arico, B.; Savino, S.; Santini, L.; Brunelli, B.; Bambini, S.; Biolchi, A.; Capecchi, B.; et al. A universal vaccine for serogroup B meningococcus. Proc. Natl. Acad. Sci. USA 2006, 103, 10834–10839. [Google Scholar] [CrossRef]
  99. Duncan, C.J.; Hill, A.V.; Ellis, R.D. Can growth inhibition assays (GIA) predict blood-stage malaria vaccine efficacy? Hum. Vaccin. Immunother. 2012, 8, 706–714. [Google Scholar] [CrossRef]
  100. Miura, K.; Deng, B.; Wu, Y.; Zhou, L.; Pham, T.P.; Diouf, A.; Wu, C.K.; Lee, S.M.; Plieskatt, J.L.; Morin, M.J.; et al. ELISA units, IgG subclass ratio and avidity determined functional activity of mouse anti-Pfs230 antibodies judged by a standard membrane-feeding assay with Plasmodium falciparum. Vaccine 2019, 37, 2073–2078. [Google Scholar] [CrossRef]
  101. Davies, M.R.; McIntyre, L.; Mutreja, A.; Lacey, J.A.; Lees, J.A.; Towers, R.J.; Duchene, S.; Smeesters, P.R.; Frost, H.R.; Price, D.J.; et al. Atlas of group A streptococcal vaccine candidates compiled using large-scale comparative genomics. Nat. Genet. 2019, 51, 1035–1043. [Google Scholar] [CrossRef]
  102. Laurens, M.B. RTS,S/AS01 vaccine (Mosquirix): An overview. Hum. Vaccin. Immunother. 2020, 16, 480–489. [Google Scholar] [CrossRef]
  103. Cheesman, S.; O’Mahony, E.; Pattaradilokrat, S.; Degnan, K.; Knott, S.; Carter, R. A single parasite gene determines strain-specific protective immunity against malaria: The role of the merozoite surface protein I. Int. J. Parasitol. 2010, 40, 951–961. [Google Scholar] [CrossRef]
  104. Holder, A.A.; Guevara Patino, J.A.; Uthaipibull, C.; Syed, S.E.; Ling, I.T.; Scott-Finnigan, T.; Blackman, M.J. Merozoite surface protein 1, immune evasion, and vaccines against asexual blood stage malaria. Parassitologia 1999, 41, 409–414. [Google Scholar]
  105. Hayton, K.; Gaur, D.; Liu, A.; Takahashi, J.; Henschen, B.; Singh, S.; Lambert, L.; Furuya, T.; Bouttenot, R.; Doll, M.; et al. Erythrocyte binding protein PfRH5 polymorphisms determine species-specific pathways of Plasmodium falciparum invasion. Cell Host Microbe 2008, 4, 40–51. [Google Scholar] [CrossRef] [PubMed]
  106. Ragotte, R.J.; Higgins, M.K.; Draper, S.J. The RH5-CyRPA-Ripr Complex as a Malaria Vaccine Target. Trends Parasitol. 2020, 36, 545–559. [Google Scholar] [CrossRef]
  107. Willcox, A.C.; Huber, A.S.; Diouf, A.; Barrett, J.R.; Silk, S.E.; Pulido, D.; King, L.D.W.; Alanine, D.G.W.; Minassian, A.M.; Diakite, M.; et al. Antibodies from malaria-exposed Malians generally interact additively or synergistically with human vaccine-induced RH5 antibodies. Cell Rep. Med. 2021, 2, 100326. [Google Scholar] [CrossRef] [PubMed]
  108. Kalkal, M.; Kalkal, A.; Dhanda, S.K.; Das, E.; Pande, V.; Das, J. A comprehensive study of epitopes and immune reactivity among Plasmodium species. BMC Microbiol. 2022, 22, 74. [Google Scholar] [CrossRef] [PubMed]
  109. Chenet, S.M.; Branch, O.H.; Escalante, A.A.; Lucas, C.M.; Bacon, D.J. Genetic diversity of vaccine candidate antigens in Plasmodium falciparum isolates from the Amazon basin of Peru. Malar. J. 2008, 7, 93. [Google Scholar] [CrossRef]
  110. Wootton, J.C.; Feng, X.; Ferdig, M.T.; Cooper, R.A.; Mu, J.; Baruch, D.I.; Magill, A.J.; Su, X.Z. Genetic diversity and chloroquine selective sweeps in Plasmodium falciparum. Nature 2002, 418, 320–323. [Google Scholar] [CrossRef]
  111. McCollum, A.M.; Basco, L.K.; Tahar, R.; Udhayakumar, V.; Escalante, A.A. Hitchhiking and selective sweeps of Plasmodium falciparum sulfadoxine and pyrimethamine resistance alleles in a population from central Africa. Antimicrob. Agents Chemother. 2008, 52, 4089–4097. [Google Scholar] [CrossRef]
  112. Ajibaye, O.; Osuntoki, A.A.; Balogun, E.O.; Olukosi, Y.A.; Iwalokun, B.A.; Oyebola, K.M.; Hikosaka, K.; Watanabe, Y.I.; Ebiloma, G.U.; Kita, K.; et al. Genetic polymorphisms in malaria vaccine candidate Plasmodium falciparum reticulocyte-binding protein homologue-5 among populations in Lagos, Nigeria. Malar. J. 2020, 19, 6. [Google Scholar] [CrossRef]
  113. Alloueche, A.; Milligan, P.; Conway, D.J.; Pinder, M.; Bojang, K.; Doherty, T.; Tornieporth, N.; Cohen, J.; Greenwood, B.M. Protective efficacy of the RTS,S/AS02 Plasmodium falciparum malaria vaccine is not strain specific. Am. J. Trop. Med. Hyg. 2003, 68, 97–101. [Google Scholar] [CrossRef]
  114. Enosse, S.; Dobano, C.; Quelhas, D.; Aponte, J.J.; Lievens, M.; Leach, A.; Sacarlal, J.; Greenwood, B.; Milman, J.; Dubovsky, F.; et al. RTS,S/AS02A malaria vaccine does not induce parasite CSP T cell epitope selection and reduces multiplicity of infection. PLoS Clin. Trials 2006, 1, e5. [Google Scholar] [CrossRef]
  115. Waitumbi, J.N.; Anyona, S.B.; Hunja, C.W.; Kifude, C.M.; Polhemus, M.E.; Walsh, D.S.; Ockenhouse, C.F.; Heppner, D.G.; Leach, A.; Lievens, M.; et al. Impact of RTS,S/AS02(A) and RTS,S/AS01(B) on genotypes of P. falciparum in adults participating in a malaria vaccine clinical trial. PLoS ONE 2009, 4, e7849. [Google Scholar] [CrossRef]
  116. Neafsey, D.E.; Juraska, M.; Bedford, T.; Benkeser, D.; Valim, C.; Griggs, A.; Lievens, M.; Abdulla, S.; Adjei, S.; Agbenyega, T.; et al. Genetic Diversity and Protective Efficacy of the RTS,S/AS01 Malaria Vaccine. N. Engl. J. Med. 2015, 373, 2025–2037. [Google Scholar] [CrossRef] [PubMed]
  117. Goh, Y.S.; McGuire, D.; Renia, L. Vaccination With Sporozoites: Models and Correlates of Protection. Front. Immunol. 2019, 10, 1227. [Google Scholar] [CrossRef] [PubMed]
  118. Moser, K.A.; Drabek, E.F.; Dwivedi, A.; Stucke, E.M.; Crabtree, J.; Dara, A.; Shah, Z.; Adams, M.; Li, T.; Rodrigues, P.T.; et al. Strains used in whole organism Plasmodium falciparum vaccine trials differ in genome structure, sequence, and immunogenic potential. Genome Med. 2020, 12, 6. [Google Scholar] [CrossRef]
  119. Silva, J.C.; Dwivedi, A.; Moser, K.A.; Sissoko, M.S.; Epstein, J.E.; Healy, S.A.; Lyke, K.E.; Mordmuller, B.; Kremsner, P.G.; Duffy, P.E.; et al. Plasmodium falciparum 7G8 challenge provides conservative prediction of efficacy of PfNF54-based PfSPZ Vaccine in Africa. Nat. Commun. 2022, 13, 3390. [Google Scholar] [CrossRef] [PubMed]
  120. Rolland, M.; Tovanabutra, S.; deCamp, A.C.; Frahm, N.; Gilbert, P.B.; Sanders-Buell, E.; Heath, L.; Magaret, C.A.; Bose, M.; Bradfield, A.; et al. Genetic impact of vaccination on breakthrough HIV-1 sequences from the STEP trial. Nat. Med. 2011, 17, 366–371. [Google Scholar] [CrossRef]
  121. Rolland, M.; Gilbert, P.B. Sieve analysis to understand how SARS-CoV-2 diversity can impact vaccine protection. PLoS Pathog. 2021, 17, e1009406. [Google Scholar] [CrossRef]
  122. Seder, R.A.; Chang, L.J.; Enama, M.E.; Zephir, K.L.; Sarwar, U.N.; Gordon, I.J.; Holman, L.A.; James, E.R.; Billingsley, P.F.; Gunasekera, A.; et al. Protection against malaria by intravenous immunization with a nonreplicating sporozoite vaccine. Science 2013, 341, 1359–1365. [Google Scholar] [CrossRef] [PubMed]
  123. Mordmuller, B.; Surat, G.; Lagler, H.; Chakravarty, S.; Ishizuka, A.S.; Lalremruata, A.; Gmeiner, M.; Campo, J.J.; Esen, M.; Ruben, A.J.; et al. Sterile protection against human malaria by chemoattenuated PfSPZ vaccine. Nature 2017, 542, 445–449. [Google Scholar] [CrossRef] [PubMed]
  124. Lyke, K.E.; Ishizuka, A.S.; Berry, A.A.; Chakravarty, S.; DeZure, A.; Enama, M.E.; James, E.R.; Billingsley, P.F.; Gunasekera, A.; Manoj, A.; et al. Attenuated PfSPZ Vaccine induces strain-transcending T cells and durable protection against heterologous controlled human malaria infection. Proc. Natl. Acad. Sci. USA 2017, 114, 2711–2716. [Google Scholar] [CrossRef] [PubMed]
  125. Sissoko, M.S.; Healy, S.A.; Katile, A.; Omaswa, F.; Zaidi, I.; Gabriel, E.E.; Kamate, B.; Samake, Y.; Guindo, M.A.; Dolo, 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, 498–509. [Google Scholar] [CrossRef]
  126. Culleton, R.; Martinelli, A.; Hunt, P.; Carter, R. Linkage group selection: Rapid gene discovery in malaria parasites. Genome Res. 2005, 15, 92–97. [Google Scholar] [CrossRef]
  127. Rumke, L.W.; Groenveld, F.C.; van Os, Y.M.G.; Praest, P.; Tanja, A.A.N.; de Jong, D.; Symons, J.; Schuurman, R.; Reinders, T.; Hofstra, L.M.; et al. In-depth Characterization of Vaccine Breakthrough Infections With SARS-CoV-2 Among Health Care Workers in a Dutch Academic Medical Center. Open Forum Infect. Dis. 2022, 9, ofab553. [Google Scholar] [CrossRef]
  128. Wu, J.; Tian, L.; Yu, X.; Pattaradilokrat, S.; Li, J.; Wang, M.; Yu, W.; Qi, Y.; Zeituni, A.E.; Nair, S.C.; et al. Strain-specific innate immune signaling pathways determine malaria parasitemia dynamics and host mortality. Proc. Natl. Acad. Sci. USA 2014, 111, E511–E520. [Google Scholar] [CrossRef]
  129. Xia, L.; Wu, J.; Pattaradilokrat, S.; Tumas, K.; He, X.; Peng, Y.C.; Huang, R.; Myers, T.G.; Long, C.A.; Wang, R.; et al. Detection of host pathways universally inhibited after Plasmodium yoelii infection for immune intervention. Sci. Rep. 2018, 8, 15280. [Google Scholar] [CrossRef]
  130. Sondo, P.; Derra, K.; Lefevre, T.; Diallo-Nakanabo, S.; Tarnagda, Z.; Zampa, O.; Kazienga, A.; Valea, I.; Sorgho, H.; Ouedraogo, J.B.; et al. Genetically diverse Plasmodium falciparum infections, within-host competition and symptomatic malaria in humans. Sci. Rep. 2019, 9, 127. [Google Scholar] [CrossRef]
  131. Kisalu, N.K.; Idris, A.H.; Weidle, C.; Flores-Garcia, Y.; Flynn, B.J.; Sack, B.K.; Murphy, S.; Schon, A.; Freire, E.; Francica, J.R.; et al. A human monoclonal antibody prevents malaria infection by targeting a new site of vulnerability on the parasite. Nat. Med. 2018, 24, 408–416. [Google Scholar] [CrossRef]
  132. Tan, J.; Sack, B.K.; Oyen, D.; Zenklusen, I.; Piccoli, L.; Barbieri, S.; Foglierini, M.; Fregni, C.S.; Marcandalli, J.; Jongo, S.; et al. A public antibody lineage that potently inhibits malaria infection through dual binding to the circumsporozoite protein. Nat. Med. 2018, 24, 401–407. [Google Scholar] [CrossRef] [PubMed]
  133. Foquet, L.; Hermsen, C.C.; van Gemert, G.J.; Van Braeckel, E.; Weening, K.E.; Sauerwein, R.; Meuleman, P.; Leroux-Roels, G. Vaccine-induced monoclonal antibodies targeting circumsporozoite protein prevent Plasmodium falciparum infection. J. Clin. Investig. 2014, 124, 140–144. [Google Scholar] [CrossRef] [PubMed]
  134. Wang, L.T.; Pereira, L.S.; Kiyuka, P.K.; Schon, A.; Kisalu, N.K.; Vistein, R.; Dillon, M.; Bonilla, B.G.; Molina-Cruz, A.; Barillas-Mury, C.; et al. Protective effects of combining monoclonal antibodies and vaccines against the Plasmodium falciparum circumsporozoite protein. PLoS Pathog. 2021, 17, e1010133. [Google Scholar] [CrossRef]
  135. Wu, R.L.; Idris, A.H.; Berkowitz, N.M.; Happe, M.; Gaudinski, M.R.; Buettner, C.; Strom, L.; Awan, S.F.; Holman, L.A.; Mendoza, F.; et al. Low-Dose Subcutaneous or Intravenous Monoclonal Antibody to Prevent Malaria. N. Engl. J. Med. 2022, 387, 397–407. [Google Scholar] [CrossRef]
  136. Gaudinski, M.R.; Berkowitz, N.M.; Idris, A.H.; Coates, E.E.; Holman, L.A.; Mendoza, F.; Gordon, I.J.; Plummer, S.H.; Trofymenko, O.; Hu, Z.; et al. A Monoclonal Antibody for Malaria Prevention. N. Engl. J. Med. 2021, 385, 803–814. [Google Scholar] [CrossRef] [PubMed]
  137. Tiller, T.; Meffre, E.; Yurasov, S.; Tsuiji, M.; Nussenzweig, M.C.; Wardemann, H. Efficient generation of monoclonal antibodies from single human B cells by single cell RT-PCR and expression vector cloning. J. Immunol. Methods 2008, 329, 112–124. [Google Scholar] [CrossRef]
  138. Durack, J.; Lynch, S.V. The gut microbiome: Relationships with disease and opportunities for therapy. J. Exp. Med. 2019, 216, 20–40. [Google Scholar] [CrossRef]
  139. Vijay, A.; Valdes, A.M. Role of the gut microbiome in chronic diseases: A narrative review. Eur. J. Clin. Nutr. 2022, 76, 489–501. [Google Scholar] [CrossRef]
  140. de Vos, W.M.; Tilg, H.; Van Hul, M.; Cani, P.D. Gut microbiome and health: Mechanistic insights. Gut 2022, 71, 1020–1032. [Google Scholar] [CrossRef]
  141. Ippolito, M.M.; Denny, J.E.; Langelier, C.; Sears, C.L.; Schmidt, N.W. Malaria and the Microbiome: A Systematic Review. Clin. Infect. Dis. 2018, 67, 1831–1839. [Google Scholar] [CrossRef]
  142. Waide, M.L.; Schmidt, N.W. The gut microbiome, immunity, and Plasmodium severity. Curr. Opin. Microbiol. 2020, 58, 56–61. [Google Scholar] [CrossRef] [PubMed]
  143. Mukherjee, D.; Chora, A.F.; Mota, M.M. Microbiota, a Third Player in the Host-Plasmodium Affair. Trends Parasitol. 2020, 36, 11–18. [Google Scholar] [CrossRef] [PubMed]
  144. Knight, R.; Callewaert, C.; Marotz, C.; Hyde, E.R.; Debelius, J.W.; McDonald, D.; Sogin, M.L. The Microbiome and Human Biology. Annu. Rev. Genom. Hum. Genet. 2017, 18, 65–86. [Google Scholar] [CrossRef]
  145. Villarino, N.F.; LeCleir, G.R.; Denny, J.E.; Dearth, S.P.; Harding, C.L.; Sloan, S.S.; Gribble, J.L.; Campagna, S.R.; Wilhelm, S.W.; Schmidt, N.W. Composition of the gut microbiota modulates the severity of malaria. Proc. Natl. Acad. Sci. USA 2016, 113, 2235–2240. [Google Scholar] [CrossRef]
  146. Morffy Smith, C.D.; Gong, M.; Andrew, A.K.; Russ, B.N.; Ge, Y.; Zadeh, M.; Cooper, C.A.; Mohamadzadeh, M.; Moore, J.M. Composition of the gut microbiota transcends genetic determinants of malaria infection severity and influences pregnancy outcome. EBioMedicine 2019, 44, 639–655. [Google Scholar] [CrossRef] [PubMed]
  147. Guan, W.; Yang, S.; Zhao, Y.; Cheng, W.; Song, X.; Yao, Y.; Xie, Y.; Du, W.; Li, J. Observation of the Gut Microbiota Profile in C57BL/6 Mice Induced by Plasmodium berghei ANKA Infection. Front. Cell. Infect. Microbiol. 2021, 11, 680383. [Google Scholar] [CrossRef]
  148. Lynn, D.J.; Benson, S.C.; Lynn, M.A.; Pulendran, B. Modulation of immune responses to vaccination by the microbiota: Implications and potential mechanisms. Nat. Rev. Immunol. 2022, 22, 33–46. [Google Scholar] [CrossRef]
  149. de Jong, S.E.; Olin, A.; Pulendran, B. The Impact of the Microbiome on Immunity to Vaccination in Humans. Cell Host Microbe 2020, 28, 169–179. [Google Scholar] [CrossRef]
  150. Stefanetti, G.; Kasper, D.L. Impact of the Host Microbiome on Vaccine Responsiveness: Lessons Learned and Future Perspective. Biochemistry 2022, 61, 2849–2855. [Google Scholar] [CrossRef]
  151. Yooseph, S.; Kirkness, E.F.; Tran, T.M.; Harkins, D.M.; Jones, M.B.; Torralba, M.G.; O’Connell, E.; Nutman, T.B.; Doumbo, S.; Doumbo, O.K.; et al. Stool microbiota composition is associated with the prospective risk of Plasmodium falciparum infection. BMC Genom. 2015, 16, 631. [Google Scholar] [CrossRef]
  152. Mandal, R.K.; Denny, J.E.; Namazzi, R.; Opoka, R.O.; Datta, D.; John, C.C.; Schmidt, N.W. Dynamic modulation of spleen germinal center reactions by gut bacteria during Plasmodium infection. Cell Rep. 2021, 35, 109094. [Google Scholar] [CrossRef]
  153. Gomez-Perez, G.P.; van Bruggen, R.; Grobusch, M.P.; Dobano, C. Plasmodium falciparum malaria and invasive bacterial co-infection in young African children: The dysfunctional spleen hypothesis. Malar. J. 2014, 13, 335. [Google Scholar] [CrossRef] [PubMed]
  154. Waide, M.L.; Polidoro, R.; Powell, W.L.; Denny, J.E.; Kos, J.; Tieri, D.A.; Watson, C.T.; Schmidt, N.W. Gut Microbiota Composition Modulates the Magnitude and Quality of Germinal Centers during Plasmodium Infections. Cell Rep. 2020, 33, 108503. [Google Scholar] [CrossRef] [PubMed]
  155. Mukherjee, D.; Chora, A.F.; Lone, J.C.; Ramiro, R.S.; Blankenhaus, B.; Serre, K.; Ramirez, M.; Gordo, I.; Veldhoen, M.; Varga-Weisz, P.; et al. Host lung microbiota promotes malaria-associated acute respiratory distress syndrome. Nat. Commun. 2022, 13, 3747. [Google Scholar] [CrossRef]
  156. Azelyte, J.; Wu-Chuang, A.; Ziegyte, R.; Platonova, E.; Mateos-Hernandez, L.; Maye, J.; Obregon, D.; Palinauskas, V.; Cabezas-Cruz, A. Anti-Microbiota Vaccine Reduces Avian Malaria Infection Within Mosquito Vectors. Front. Immunol. 2022, 13, 841835. [Google Scholar] [CrossRef]
  157. Harris, V.C.; Armah, G.; Fuentes, S.; Korpela, K.E.; Parashar, U.; Victor, J.C.; Tate, J.; de Weerth, C.; Giaquinto, C.; Wiersinga, W.J.; et al. Significant Correlation Between the Infant Gut Microbiome and Rotavirus Vaccine Response in Rural Ghana. J. Infect. Dis. 2017, 215, 34–41. [Google Scholar] [CrossRef] [PubMed]
  158. Kalappa, D.M.; Subramani, P.A.; Basavanna, S.K.; Ghosh, S.K.; Sundaramurthy, V.; Uragayala, S.; Tiwari, S.; Anvikar, A.R.; Valecha, N. Influence of midgut microbiota in Anopheles stephensi on Plasmodium berghei infections. Malar. J. 2018, 17, 385. [Google Scholar] [CrossRef]
  159. Gendrin, M.; Rodgers, F.H.; Yerbanga, R.S.; Ouedraogo, J.B.; Basanez, M.G.; Cohuet, A.; Christophides, G.K. Antibiotics in ingested human blood affect the mosquito microbiota and capacity to transmit malaria. Nat. Commun. 2015, 6, 5921. [Google Scholar] [CrossRef]
  160. Gendrin, M.; Yerbanga, R.S.; Ouedraogo, J.B.; Lefevre, T.; Cohuet, A.; Christophides, G.K. Differential Effects of Azithromycin, Doxycycline, and Cotrimoxazole in Ingested Blood on the Vectorial Capacity of Malaria Mosquitoes. Open Forum Infect. Dis. 2016, 3, ofw074. [Google Scholar] [CrossRef]
  161. Dacey, D.P.; Chain, F.J.J. The Challenges of Microbial Control of Mosquito-Borne Diseases Due to the Gut Microbiome. Front. Genet. 2020, 11, 504354. [Google Scholar] [CrossRef]
  162. Peyerl-Hoffmann, G.; Jelinek, T.; Kilian, A.; Kabagambe, G.; Metzger, W.G.; von Sonnenburg, F. Genetic diversity of Plasmodium falciparum and its relationship to parasite density in an area with different malaria endemicities in West Uganda. Trop. Med. Int. Health 2001, 6, 607–613. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The life cycle of malaria parasites, with major vaccine candidates targeting different developmental stages. The malaria life cycle starts with a mosquito bite injecting sporozoites into the human skin. The sporozoites travel to the bloodstream to invade hepatocytes, where the parasites replicate to produce thousands of merozoites. Some vaccines targeting the liver-stage parasites include RTS,S/AS01, R21, full-length circumsporozoite protein (CSP), radiation-attenuated sporozoites (RAS), and PfSPZ immunization–treatment–vaccination (PfSPZ ITV). Mature merozoites from the liver are then released into the bloodstream again to invade RBCs. Within an RBC, the parasite replicates to produce more merozoites that can invade new RBCs for additional cycles. Some vaccines targeting the P. falciparum blood stages include apical membrane antigen 1 (PfAMA1), merozoite surface protein (PfMSP1), PfMSP3, erythrocyte-binding antigen 175 (PfEBA-175), reticulocyte-binding protein homolog 5 (PfRh5), AMA1-rhoptry neck protein 2 (PfAMA1-PfRON2) complex, schizont egress antigen-1 (PfSEA1), glutamic-acid-rich protein (PfGARP), and erythrocyte membrane protein-binding chondroitin sulfate A (PfVAR2CSA). For unknown reasons, some merozoites may develop into male and female gametocytes that differentiate into male and female gametes in the mosquito midgut when another mosquito takes a blood meal. The male and female gametes fertilize to form zygotes that differentiate into motile ookinetes. The ookinetes penetrate the mosquito midgut and develop into oocysts outside the midgut wall. Each oocyst contains thousands of sporozoites that travel to mosquito salivary glands. Transmission-blocking vaccines are aimed at blocking the development of sexual stages within mosquitoes. Vaccines targeting these stages include Pfs48/45, Pfs230, Pfs25, Pfs28, Pfs47, and anopheline alanyl aminopeptidase N (AnAPN1) which is a mosquito midgut protein. When the mosquito bites another vertebrate host, the sporozoites are injected into the new host, starting another cycle.
Figure 1. The life cycle of malaria parasites, with major vaccine candidates targeting different developmental stages. The malaria life cycle starts with a mosquito bite injecting sporozoites into the human skin. The sporozoites travel to the bloodstream to invade hepatocytes, where the parasites replicate to produce thousands of merozoites. Some vaccines targeting the liver-stage parasites include RTS,S/AS01, R21, full-length circumsporozoite protein (CSP), radiation-attenuated sporozoites (RAS), and PfSPZ immunization–treatment–vaccination (PfSPZ ITV). Mature merozoites from the liver are then released into the bloodstream again to invade RBCs. Within an RBC, the parasite replicates to produce more merozoites that can invade new RBCs for additional cycles. Some vaccines targeting the P. falciparum blood stages include apical membrane antigen 1 (PfAMA1), merozoite surface protein (PfMSP1), PfMSP3, erythrocyte-binding antigen 175 (PfEBA-175), reticulocyte-binding protein homolog 5 (PfRh5), AMA1-rhoptry neck protein 2 (PfAMA1-PfRON2) complex, schizont egress antigen-1 (PfSEA1), glutamic-acid-rich protein (PfGARP), and erythrocyte membrane protein-binding chondroitin sulfate A (PfVAR2CSA). For unknown reasons, some merozoites may develop into male and female gametocytes that differentiate into male and female gametes in the mosquito midgut when another mosquito takes a blood meal. The male and female gametes fertilize to form zygotes that differentiate into motile ookinetes. The ookinetes penetrate the mosquito midgut and develop into oocysts outside the midgut wall. Each oocyst contains thousands of sporozoites that travel to mosquito salivary glands. Transmission-blocking vaccines are aimed at blocking the development of sexual stages within mosquitoes. Vaccines targeting these stages include Pfs48/45, Pfs230, Pfs25, Pfs28, Pfs47, and anopheline alanyl aminopeptidase N (AnAPN1) which is a mosquito midgut protein. When the mosquito bites another vertebrate host, the sporozoites are injected into the new host, starting another cycle.
Pathogens 12 01061 g001
Figure 2. Summary of major approaches and applications in genetic and genomic studies of malaria parasites. Many genetic and genomic approaches have been employed to study the malaria parasite populations, transmission, molecular evolution, drug resistances, host–parasite interaction, and vaccine targets. Genetic crosses can be performed using different strains of malaria parasites to map parasite genes conferring drug resistance or encoding vaccine targets. Similarly, genetic crosses of inbred mice will allow the mapping of host-protective genes. DNA collected from patient blood samples can be used in association studies, either using a single candidate gene or genome-wide association study (GWAS), to identify parasite and host genes of interest and/or to study parasite population structure, origin, molecular evolution, and drug-selective sweeps. Using animal models, it is also possible to study host–parasite genetic interactions, linking host gene response/expression to parasite genes using trans-species expression quantitative locus analysis (ts-eQTL) [4]. When identifying drug-resistant genes, parasites can be selected under drug pressure in vitro (P. falciparum) or in vivo (rodent Plasmodium species), and genome sequences from the parasites before and after selection can be compared to identify mutations selected using drug treatment. Similar genetic approaches have also been employed to map mosquito genes conferring resistance to malaria infections.
Figure 2. Summary of major approaches and applications in genetic and genomic studies of malaria parasites. Many genetic and genomic approaches have been employed to study the malaria parasite populations, transmission, molecular evolution, drug resistances, host–parasite interaction, and vaccine targets. Genetic crosses can be performed using different strains of malaria parasites to map parasite genes conferring drug resistance or encoding vaccine targets. Similarly, genetic crosses of inbred mice will allow the mapping of host-protective genes. DNA collected from patient blood samples can be used in association studies, either using a single candidate gene or genome-wide association study (GWAS), to identify parasite and host genes of interest and/or to study parasite population structure, origin, molecular evolution, and drug-selective sweeps. Using animal models, it is also possible to study host–parasite genetic interactions, linking host gene response/expression to parasite genes using trans-species expression quantitative locus analysis (ts-eQTL) [4]. When identifying drug-resistant genes, parasites can be selected under drug pressure in vitro (P. falciparum) or in vivo (rodent Plasmodium species), and genome sequences from the parasites before and after selection can be compared to identify mutations selected using drug treatment. Similar genetic approaches have also been employed to map mosquito genes conferring resistance to malaria infections.
Pathogens 12 01061 g002
Figure 3. Applications of malaria genomics in vaccine development. Genome sequences from parasite isolates can be studied for potential protective antigens. A candidate antigen can be tested for the efficacy of its protection in animal models before trials on humans. An antigen sequence in parasite populations from a specific endemic region can be surveyed for diversity, and a multivariant vaccine (with reduced diversity in a local setting) can be designed based on major alleles present in a specific location such as South American countries. Vaccine efficacy can be evaluated by surveying allele frequencies before and after vaccination. For whole-organism vaccines, a survey of genome-wide polymorphisms before and after vaccination (or vaccine vs placebo recipients) may help to identify the protective vaccine targets. Finally, there is a chance that a partially effective vaccine may change the parasite populations circulating in an endemic region, which may alter the virulence of parasite populations. New mutations may occur after vaccination, reducing the effectiveness of a vaccine.
Figure 3. Applications of malaria genomics in vaccine development. Genome sequences from parasite isolates can be studied for potential protective antigens. A candidate antigen can be tested for the efficacy of its protection in animal models before trials on humans. An antigen sequence in parasite populations from a specific endemic region can be surveyed for diversity, and a multivariant vaccine (with reduced diversity in a local setting) can be designed based on major alleles present in a specific location such as South American countries. Vaccine efficacy can be evaluated by surveying allele frequencies before and after vaccination. For whole-organism vaccines, a survey of genome-wide polymorphisms before and after vaccination (or vaccine vs placebo recipients) may help to identify the protective vaccine targets. Finally, there is a chance that a partially effective vaccine may change the parasite populations circulating in an endemic region, which may alter the virulence of parasite populations. New mutations may occur after vaccination, reducing the effectiveness of a vaccine.
Pathogens 12 01061 g003
Figure 4. Interactions of microbiota genomes with those of human and mosquito can influence the host responses to malaria parasite infections. Metabolites and components such as nucleic acids released from the microbes may trigger and/or alter the host (human and mosquito) immune responses, although whether there are microbes that can directly inhibit or enhance malaria parasite development is not clear. On the other hand, malaria parasite infections may also change host immunity and microbial populations in the hosts, allowing some commensal populations to become pathogenic.
Figure 4. Interactions of microbiota genomes with those of human and mosquito can influence the host responses to malaria parasite infections. Metabolites and components such as nucleic acids released from the microbes may trigger and/or alter the host (human and mosquito) immune responses, although whether there are microbes that can directly inhibit or enhance malaria parasite development is not clear. On the other hand, malaria parasite infections may also change host immunity and microbial populations in the hosts, allowing some commensal populations to become pathogenic.
Pathogens 12 01061 g004
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Su, X.; Stadler, R.V.; Xu, F.; Wu, J. Malaria Genomics, Vaccine Development, and Microbiome. Pathogens 2023, 12, 1061. https://doi.org/10.3390/pathogens12081061

AMA Style

Su X, Stadler RV, Xu F, Wu J. Malaria Genomics, Vaccine Development, and Microbiome. Pathogens. 2023; 12(8):1061. https://doi.org/10.3390/pathogens12081061

Chicago/Turabian Style

Su, Xinzhuan, Rachel V. Stadler, Fangzheng Xu, and Jian Wu. 2023. "Malaria Genomics, Vaccine Development, and Microbiome" Pathogens 12, no. 8: 1061. https://doi.org/10.3390/pathogens12081061

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop