Skip to main content
Advertisement
  • Loading metrics

Mechanistic insights into the interaction between the host gut microbiome and malaria

Abstract

Malaria is a devastating infectious disease and significant global health burden caused by the bite of a Plasmodium-infected female Anopheles mosquito. Gut microbiota was recently discovered as a risk factor of severe malaria. This review entails the recent advances on the impact of gut microbiota composition on malaria severity and consequence of malaria infection on gut microbiota in mammalian hosts. Additionally, this review provides mechanistic insight into interactions that might occur between gut microbiota and host immunity which in turn can modulate malaria severity. Finally, approaches to modulate gut microbiota composition are discussed. We anticipate this review will facilitate novel hypotheses to move the malaria-gut microbiome field forward.

1. Introduction

Malaria is an infectious disease caused by the bite of a female Anopheles mosquito infected with the parasite Plasmodium. Malaria remains a significant burden on the global healthcare system, causing more than 627,000 deaths and 241 million cases in 2020 [1,2]. Greater than 90% of infections and severe malaria in humans is caused by P. falciparum, with additional infections caused by other Plasmodium species including P. vivax, P. malaria, P. ovale, and P. knowlesi [3]. Sequestration of infected RBCs in internal organs lead to widespread organ damage and is a major cause of death in patients with severe P. falciparum malaria [4,5]. However, additional factors that determine the severity of malaria in humans are still evolving. Recently, it was shown that the gut microbiome (i.e., microorganism including their genetic content, microbial products, and environment within the gut) is a risk factor of severe malaria [6,7].

The phrase “you are what you eat” which can be extended to “what you eat is your gut microbiome” is very true in the context of gut microbiome that play a critical role in health and disease of individuals [810]. Bacteria make up the major organic fraction of feces (approximately 25% to 54% of dry solids) in humans [11]. Approximately 70% to 80% of immune cells are located in the gut that are trained by the intestinal microbiome [12]. Innate and adaptive immunity are required to control Plasmodium infection. Thus, gut microbiota that influence local and systemic immune system have the potential to significantly impact antimalarial immunity [1317].

Here, we have provided mechanistic insight into the role of gut microbiome in shaping malaria severity, including severe malaria anemia (SMA) and cerebral malaria. The presence of specific gut bacteria like Ruminococcaceae, Lachnospiraceae, Bacteroides, and Blautia were associated with severe malaria while Bifidobacterium and Escherichia are correlated with better outcome following Plasmodium infection. In addition to preinfection bacteria composition correlating with malaria outcomes, bacteria populations have also been shown to change in abundance following infection. Bacterial genera including Bacteroides, Alistipes, and Clostridia are relatively increased while Ruminococcaceae, Prevotellaceae, and Ruminococcus are decreased in abundance during Plasmodium infection. Importantly, gut bacteria represent a druggable target to boost antimalarial immunity and decrease malaria severity.

2. Associations between human gut microbiome and malaria

In 2014, Yilmaz and colleagues showed that older individuals (4 to 25 years old) with higher levels of Galα1-3Galβ1-4GlcNAc-R (α-gal) glycan-specific IgM antibodies in plasma were significantly protected against P. falciparum infection [18]. However, younger children (3 months to 4 years old) had no association between protection from P. falciparum infection and α-gal IgM antibodies in the plasma. Evolutionarily, humans do not express α-gal, allowing them to generate anti-α-gal antibodies when colonized by α-gal expressing bacteria [19]. Other microorganisms, including Plasmodium, express α-gal, providing an opportunity for cross-reaction between bacterial-induced anti-α-gal antibodies and other microbes. The authors further confirmed the role of α-gal produced by Escherichia coli O86:B7, a member of gut microbiota, in the mouse model of malaria (Fig 1). Anti-α-gal IgM antibodies confer protection against Plasmodium sporozoite infection independent of complement induced polymorphonuclear leukocyte recruitment, but rather by complement-induced cytotoxicity. Anti-α-gal IgM antibodies also conferred protection, in part, via blocking sporozoite migration and infection of hepatocytes, thereby blocking development of exoerythrocytic forms within hepatocytes. In contrast, anti-α-gal antibodies had no effect on the erythrocytic stage of infection in mice [18]. Additionally, in an RTS,S/AS0 vaccination trial conducted in Mozambique and Ghana, vaccinated infants (1.5 to 3 months) who had higher level of anti-α-gal IgM plasma antibodies were protected against clinical malaria over a one-year follow up compared to infants with low levels of IgM plasma antibody [20]. Nonetheless, anti α-gal IgM levels were not associated with protection against malaria in vaccinated older children (5 to 17 months) [20].

thumbnail
Fig 1. Gut bacteria associated with human malaria.

To date 5 peer-reviewed studies have been published on the impact of the gut microbiota in Plasmodium infection in humans. A cohort of children in Kalifabougou, Mali was used for 2 prospective studies (Yilmaz and colleagues (2014) [18] and Yooseph and colleagues (2015) [28]). Additionally, 3 case-control studies investigated association of gut microbiota at the time of Plasmodium infection (Huwe and colleagues (2019) [32] and Easton and colleagues (2020) [33]) and SMA (Mandal and colleagues (2021) [6]). Prospective and case-control studies have their own advantages and disadvantages. Bacteria associated with the clinical outcome is shown. Pf: P. falciparum, neg: negative, pos: positive. Figure was created with BioRender.com.

https://doi.org/10.1371/journal.ppat.1011665.g001

Multiple gut bacteria can produce α-gal including specific members of Klebsiella spps., Serratia spp., and Escherichia coli spps. Likewise, lactic acid bacteria (LAB) like Limosilactobacillus fermentum, Levilactobacillus brevis, Agrilactobacillus composti, Lacticaseibacillus paracasei, Leuconostoc mesenteroides, and Weissella confuse can express α-gal [13]. Probiotic bacteria Aeromonas veronii and Pseudomonas entomophila have high α-gal content [21]. Additionally, pathogenic bacteria including Salmonella spp. Beyond bacteria, Trypanosoma spp., Aspergillus fumigatus, and Leishmania spps. can produce α-gal [2225]. Of note, Klebsiella spps. and Escherichia are reported to be associated with malaria severity [6]. Although anti-α-gal antibodies were associated with protection from pre-erythrocytic stages of infection, they provided no benefit against blood-stage infection [18]. Therefore, it is possible that while bacteria-induced anti-α-gal antibodies confer protection against pre-erythrocytic stages, these same bacteria (e.g., Klebsiella spps. and Escherichia) acting through different mechanisms could also contribute to severe blood-stage infections. On the other hand, Trypanosoma brucei infection protects mice against experimental cerebral malaria in a coinfection model [26]. However, it’s not clear if trypanosomiasis protects against malaria in human [27].

A study by Yooseph and colleagues in 2015 [28] (using the same prospective cohort of Malian children as used by Yilmaz and colleagues (2014) [18]) reported that younger children (average age of 1.4 years) had a delayed time to first P. falciparum infection with median 121 days (95% CI 101 to 150) compared to older children (average age of 9.1 years) with median 85 days (95% CI 73 to 99). By accounting for age, gender, anemia, HbAS, S. hematobium infection, splenomegaly, and distance to river, the authors concluded that younger children stool microbiota significantly protected against prospective risk of P. falciparum infection compared to stool microbiome in older children. However, stool microbiota did not correlate with protection against febrile malaria episodes between the young and old children. Older children with increased prospective risk and shorter time to P. falciparum infection had significantly higher abundance of Ruminococcaceae unclassified, Lachnospiraceae unclassified, Prevotellaceae unclassified at family level; and Faecalibacterium, Blautia, and Dorea at genus level compared to younger children. Young children with delayed P. falciparum infection had significantly higher relative abundance of Bifidobacterium, Streptococcus, Escherichia/Shigella compared to older children among others (Fig 1). It was also reported that the stool microbiota composition was different between children (average age 3.2 years) who had a persistent asymptomatic P. falciparum infection carried over from the previous malaria transmission season than children (average age 1.3 years) who had no P. falciparum infection at the end of six-month dry season before start of subsequent six-month malaria season in Mali. This result may imply that gut microbiota composition in dry season might be involved in persistence or asymptomatic P. falciparum infection.

A limitation of Yilmaz and colleagues (2014) [18] towards our understanding into how gut microbiota impact Plasmodium infections and severity of malaria is that the authors did not identify the source of anti-α-gal IgM antibodies in the participants or if the abundance of E. coli O86:B7 in stool is associated with protection against P. falciparum infection. Likewise, Yooseph and colleagues (2015) [28] did not account for baseline anti-α-gal IgM antibody levels or if they are associated with time to delayed P. falciparum infection in younger kids during gut microbiota analysis. Thus, it’s not clear if the delayed P. falciparum infection in younger kids (1.4 years) compared to older kids is due to cross-reactivity between anti-α-gal IgM antibodies and P. falciparum or with differential gut microbiota composition. Noteworthy, Yooseph and colleagues (2015) [28] reported higher abundance of Escherichia/Shigella in the stool of younger kids with delayed protection from P. falciparum infection. Additionally, in the same Malian cohort P. falciparum reticulocyte-binding protein homologue 5 (PfRH5) specific IgG antibody was associated with a longer time to blood-stage infection and first febrile malaria and enhanced p53 expression in monocytes was predicted to be protective against febrile malaria [29,30]. Finally, P. falciparum Schizont Egress Antigen-1 anti-(PfSEA-1) in plasma is associated with a decreased risk of severe malaria in 1.5 to 4 years old kids in a holoendemic area of Tanzania [31]. These observations highlight the complexity of human malaria outcomes and the challenge to account for these while assessing the contribution of gut microbiota as a risk factor.

A study by Huwe and colleagues (2019) [32] in India where soil-transmitted helminths and malaria are endemic, reported a significantly higher abundance of Lactobacillus genus in the stool of individuals aged 0 to 68 years old infected with P. falciparum and P. vivax compared to noninfected individuals (Fig 1). Within this cohort (n = 68), 46% were infected with Plasmodium [32]. In another study, Easton and colleagues (2020) [33] found that stool bacterial taxa of Colombian children aged 4 to 16 years were stronger predictors of P. vivax parasitemia levels compared to the host peripheral blood transcriptome response or complete blood count. Although principal component analysis showed that overall gut microbiota composition was not significantly different between P. vivax-infected and uninfected children, differential abundance analysis identified higher prevalence of stool Bacteroides and lower abundance of Prevotella and Clostridiaceae were associated with P. vivax infection compared to uninfected individuals (Fig 1). A critical limitation of these studies is the comparison of gut bacteria between Plasmodium-infected and uninfected children because it is not possible to know what the malaria outcomes would be in the uninfected children if they, too, were infected with Plasmodium. This limitation is addressed in a study that compared gut bacteria between Ugandan children (0.5 to 4 years old) with an asymptomatic P. falciparum infection to children with severe malarial anemia (SMA) [6]. Children with SMA had significantly different gut bacteria composition compared to children that had asymptomatic P. falciparum infection. Higher abundance of stool bacteria including Escherichia coli, Parabacteroides distasonis, Bacteroides caccae, and Klebsiella pneumoniae among others were predictive of SMA (Fig 1). The possible mechanism by which gut bacteria may mediate immunity to malaria is described in later section. Limitations of this study include the small sample size (n = 7) of the Ugandan children with asymptomatic P. falciparum infection and the use of case-controls as opposed to a longitudinal prospective study. The latter limitation is potentially important as P. falciparum may cause changes in gut bacteria (discussed below).

3. Host gut microbiome impacts pathogenesis of malaria—Lessons from nonhuman vertebrate models

Several publications have reported the association between gut microbiota composition and malaria outcomes in mouse malaria models. Since 2016, it has been shown in several studies that baseline gut bacteria structure, function, and composition dictate susceptibility to P. yoelii 17XNL hyperparasitemia in specific-pathogen-free (SPF) C57BL/6 and BALB/c mice from different vendors and isolated barrier units (IBUs) [6,3439]. These outcomes are not restricted to mice from different vendors and IBUs, as antibiotic-induced changes in gut microbiota, both before P. yoelii 17XNL and up to 7 days after P. yoelii 17XNL infection can impact severity of malaria [6]. Additionally, in outbred Swiss Webster mice, gut microbiota composition determined P. chabudi chabudi AS infection pregnancy malaria outcomes [40]. Gut microbiota is a consortium of bacteria, fungi, archaea, viruses, and helminths. Among these microbes, it was shown that gut bacteria modulate the severity of P. yoelii 17XNL hyperparasitemia [6]. However, the potential contribution of other members of gut microbiota (e.g., fungi, archaea, and helminth) cannot be overlooked. Indeed, intestinal helminths are known to modulate malaria outcomes mostly exacerbating P. falciparum and P. vivax infection, yet the effects of intestinal helminths on malaria outcomes remains controversial [33,4144].

Gut bacteria could exert an effect on malaria outcomes by providing protection from severe malaria or by causing susceptibility to severe malaria. Presently, results from the P. yoelii 17XNL hyperparasitemia model suggest bacteria cause susceptibility, rather than resistance, to P. yoelii 17XNL hyperparasitemia. First, P. yoelii 17XNL hyperparasitemia-susceptible C57BL/6N mice treated with antibiotics have low parasitemia, while antibiotic-treated hyperparasitemia-resistant C57BL/6N mice were largely unaffected [6]. Second, fecal microbiota transplant (FMT) from hyperparasitemia-susceptible C57BL/6N mice➔hyperparasitemia-resistant C57BL/6N mice confers susceptibility while hyperparasitemia-resistant FMT➔hyperparasitemia-susceptible mice did not confer resistance [6]. Although these results favor gut bacteria can cause susceptibility to P. yoelii 17XNL hyperparasitemia, additional research in this model, and others, may identify protective roles of bacteria against severe malaria. C57BL/6N mice that are susceptible to severe hyperparasitemia have decreased numbers of germinal center (GC) B cells and follicular helper T (Tfh) cells and decreased titers of P. yoelii 17XNL-specific antibodies that recognize a smaller repertoire of P. yoelii 17XNL antigens [6,34]. C57BL/6N mice that develop P. yoelii 17XNL hyperparasitemia have significantly higher abundance of gut bacteria including Bacteroidaceae, Prevotellaceae, and Sutterellaceae at family level and Clostridium papyrosolvens, Alistipes putredinis, and Alistipes timonensis species compared to C57BL/6N mice resistant to P. yoelii 17XNL hyperparasitemia (Fig 2) [6,34].

thumbnail
Fig 2. Gut bacteria associated with nonhuman model of malaria.

Top 2 panels show baseline gut bacteria differentially abundant in mice either susceptible or resistant to P. yoelii 17XNL hyperparasitemia and P. chabaudi AS pregnancy outcomes. Fecal pellets are collected at baseline prior to Plasmodium infection to determine the gut microbiota composition. Bottom 2 panels show the shift in gut microbiota composition during Plasmodium infection in mice and monkeys. In nonlethal infection models, fecal pellet microbiota composition at peak parasitemia is compared to before Plasmodium infection. Bacteria that are significantly increased (up arrow) or decreased (down arrow) are shown. P. yoelii 17XL causes lethal infection due to hyperparasitemia while P. berghei ANKA causes mortality due to experimental cerebral malaria. Gut fecal samples are collected before mortality and compared to baseline gut microbiota. Changes in bacteria population at phylum, family, and genus level are shown. Figure was created with BioRender.com.

https://doi.org/10.1371/journal.ppat.1011665.g002

Functionally, as revealed by ceca metatranscriptomics, mice susceptible to P. yoelii 17XNL hyperparasitemia had overexpression of filC, ureABC, and 6 members of nuo gene family related to gut microbes [36]. Overexpression of filC, which encodes bacterial flagellin, is associated with mucosal barrier breakdown and inflammation [45,46]. Short-chain fatty acids (SCFAs) are bacterial metabolites that have pleotropic effects on host health. SCFAs play a vital role in energy metabolism, immunologic homeostasis, and gut barrier integrity [47]. Only the level of propionic acid (PA) among 7 other SCFAs was significantly higher in mice susceptible to P. yoelii 17XNL hyperparasitemia [39]. Bacteroidetes, which are abundant in P. yoelii 17XNL hyperparasitemia susceptible mice [6], are able to ferment polysaccharides to PA [48]. Bacteroides acidifaciens in the mouse gut are able to significantly increase the level of SCFAs especially PA [49]. Higher level of PA in gut suppresses inflammation and ameliorates liver ischemia and reperfusion injury in mice. Increased levels of circulating PA is linked to higher cognitive decline in older persons [50] and associated with innate neuroinflammation, increased oxidative stress, glutathione depletion, and altered phospholipid/acylcarnitine profiles linked to autism spectrum disorder [48,51]. The degree to which SCFAs contribute to malaria outcomes, including cerebral malaria, are unknown.

Plasmodium species infect red blood cells (RBCs) and undergo asexual replication to produce merozoites or enter sexual differentiation to produce gametocytes. It is not known if gut microbiota composition impact RBC physiology. Intriguingly, functional profiling of whole ceca (ceca content + ceca tissue) metatranscriptomics of P. yoelii 17XNL hyperparasitemia-susceptible mice revealed increased expression of basigin, a cell surface receptor required for P. falciparum invasion of RBCs [36]. Recently, the gut bacteria Flavonifractor plautii was found to be involved in the conversion blood type A to universal O type blood [52]. Higher abundance of Flavonifractor platutii was seen in the Ugandan kids with severe malaria [6]. How gut bacteria impact the function of RBCs and Plasmodium biology within RBCs is an unexplored area.

4. Impact of Plasmodium infection on gut microbiota composition

4.1. Non-cerebral malaria models

While gut microbiota has been shown to impact malaria outcomes, research has also shown that Plasmodium infections can alter gut bacteria composition dependent on the murine model, parasite strain, malaria severity, and baseline gut microbiota. In C57BL/6 mice, P. yoelii nigeriensis infection increased gut Bacteroides and decreased Ruminococcus on day 10 post infection (Fig 2) [53]. Bacterial diversities were lowest (alpha index) on day 10 p.i. and recovered to baseline level by day 30 post infection [53]. A similar decrease in alpha diversity and increase in abundance of bacteria belonging to Bacteroidota (such as Alistipes and Bacteroides) were reported in BALB/c mice following lethal P. yoelii 17XL infection at day 5 post infection compared to baseline level (Fig 2) [54]. In a nonhuman primate model, rhesus macaques infected by P. cynomolgi have decreased gut microbial alpha diversity at the peak of infection with a dramatic increase in relative abundance of Proteobacteria (family Helicobacteraceae) while decrease in Firmicutes (family Lactobacillaceae and Ruminococcaceae), Bacteroidetes (family Prevotellaceae) (Fig 2) [55]. However, Denny and colleagues and Yawen and colleagues showed increase in alpha diversity in mice following P. yoelii 17XNL infection [38,56]. Mice that are resistant to P. yoelii 17XNL hyperparasitemia (peak parasitemia is below 20%) have increase in alpha diversity (observed OTUs). In contrast, alpha diversity was stable in P. yoelii 17XNL hyperparasitemia-susceptible mice (peak parasitemia reaches up to 60%) following infection. Beta diversity (Bray–Curtis distance) was impacted by P. yoelii 17XNL infection in both hyperparasitemia-susceptible and -resistant mice, implying changes in predicted functional capacity [38]. In contrast to a shift in gut microbiota composition during P. yoelii 17XNL infection, untargeted metabolomics showed modest alterations in metabolite profile of small intestine and ceca content and plasma during parasitemia except at peak parasitemia in both P. yoelii 17XNL hyperparasitemia-susceptible and -resistant mice [38].

4.2. Experimental cerebral malaria

Cerebral malaria is one of the most severe forms of malaria and a leading cause of malaria mortality in children (15% death rate) and adults (20% death rate) [57]. Nearly a quarter of cerebral malaria survivors suffer from life-long neurological sequelae and ongoing comorbidities [58]. Five studies have investigated the impact of experimental cerebral malaria (ECM) on gut microbiome composition in murine malaria [5962] and one on upper gastrointestinal pathophysiology [63] with variable results. Taniguchi and colleagues (2015) and Shimada and colleagues (2019) reported intestinal pathology in C57BL/6 mice infected by P. berghei ANKA that develop ECM [62,63]. ECM caused weight loss, multiple red gastric patches, detachment of epithelia, gastric gas retention, enlargement of goblet cells, small intestine shortening, increased intestinal permeability, and caused dysbiosis [63]. Knowler and colleagues (2023) observed lengthening of small intestine in contrast to Shimada and colleagues (2019) [60,63]. P. berghei ANKA infection in C57BL/6 mice caused changes in gut bacteria composition with increased abundance of class Clostridia; family Enterobacteriaceae, Verrucomicrobiaceae; and genus Akkermansia, Alistipes, and Alloprevotella and decrease Lactobacillaceae family (Fig 2) [59,60,62]. Although P. berghei ANKA causes lethal ECM within 7 to 10 days, these P. berghei ANKA-induced changes in gut bacterial composition were long lasting if mice were treated with artemether [61].

4.3. Humans

There is limited information regarding the effect of malaria on gut microbiota composition in humans. Presently, 1 study has assessed this in the context of longitudinal analysis of stool bacteria compositions in Kenyan infants [64]. In this study, stool samples from infants (n = 10) from birth to 10 months were collected roughly 2 weeks before and 2 weeks after a febrile malarial episode and artemether-lumefantrine treatment. In contrast to model organisms, measurements of gut microbiota composition using multiple metrics of alpha and beta diversity did not show significant difference in structure and composition of gut microbiota [64]. That artemether-lumefantrine treatment had no effect on gut bacteria populations in the Kenyan infants, is consistent with a separate study in mice showing that artemether-lumefantrine and artesunate-amodiaquine treatment had no effect on gut bacteria communities [65]. There are important limitations to this study that must be considered including the small sample size of 10 infants, narrow age range of participants (<10 months old), lack of severe malaria, and lack of higher-resolution sample collection whereby gut bacteria could have changed and reverted to baseline. Therefore, additional longitudinal studies in humans that address these limitations and expand the geographical representation of participants are warranted to gain deeper insight. As other inflammatory and severe diseases have been shown to change gut bacteria compositions [6668], it would not be surprising if severe malaria in children is indeed associated with changes in gut bacteria compositions.

4.4. Implications of malaria-induced gut microbiota changes

These observations raise an important question, what effect does Plasmodium-induced changes in gut microbiota composition have on current/subsequent malaria outcomes? The one study to assess this question performed FMTs from convalescent P. yoelii 17XNL (day 60 post infection) hyperparasitemia-resistant and -susceptible mice into germ-free mice. As noted above, P. yoelii 17XNL hyperparasitemia-resistant and -susceptible mice showed changes in gut bacteria communities following P. yoelii 17XNL infection, with decreasing differences in gut bacteria communities between these groups of mice at convalescence compared to differences observed pre-P. yoelii 17XNL infection [38]. Yet, the ex-germ-free mice colonized with gut microbiota from P. yoelii 17XNL hyperparasitemia-resistant and -susceptible mice were resistant and susceptible, respectively, to hyperparasitemia following P. yoelii 17XNL infection [38]. These results demonstrate that, at least within this model, Plasmodium-induced changes in gut bacteria composition do not change susceptibility to future Plasmodium infections. That P. yoelii 17XNL-induced changes in gut bacteria did not impact subsequent malaria outcomes is beneficial to human case-control studies, as it demonstrates the bacteria that are present prior to Plasmodium infection and cause susceptibility to severe malaria are not lost in the susceptible mice nor do these bacteria appear in the resistant mice. Therefore, even if P. falciparum causes changes in human gut bacteria populations, these data suggest that comparing children with asymptomatic P. falciparum infections compared to children with severe malaria has the potential to identify bacteria that contributed to the differential malaria outcomes.

5. Immunological mechanisms by which host microbiota can impact malaria outcomes

Host immunity is essential to control Plasmodium infection [69], but immune responses are also involved in the pathology of malaria [7073]. As such, it is crucial to understand the components of host immunity that both contribute to protection and pathogenesis in malaria and how these are regulated by gut microbiota. This knowledge has the potential to aid in the development of microbiome-based therapeutics to control Plasmodium infection and prevent life-threatening severe malaria. The contribution of innate and adaptive immunity during Plasmodium infection is reviewed elsewhere [74]. Contribution of specific gut microbiome in immunity and immune-mediated disorders and CD4+ T cell differentiation and function is published previously [9,7577]. Here, we have reviewed components of the immune system that contribute towards control and pathogenesis of malaria and discuss specific gut microbiota that might affect these components of host immunity. Whether and how gut-derived immune cells impact the gut-distal immune response to malaria and other infections is yet to be fully understood (Fig 3).

thumbnail
Fig 3. Gut microbiota intervention strategy and possible immunological mechanism of action against severity to malaria.

Agents that can modulate gut microbiota composition or deplete gut bacteria that can influence gut or systemic immunity are shown. Dotted arrow indicates the potential interactions which need further validation. Color of arrows connect gut microbiota modifying agents and their impact on respective immune cells. Role of different immune populations to inhibit or exacerbate various stage and types of malaria are connected. Although, the exact mechanism on how gut microbiota impacts severe malaria is unknown, this figure provides a plausible connection between gut microbiota and malaria severity. Figure was created with BioRender.com.

https://doi.org/10.1371/journal.ppat.1011665.g003

5.1. GC B cells and TFh cells

GC reactions are required for production of high-affinity antibodies and long-term memory against malaria [69,7880]. We have shown that gut microbiota compositions dynamically regulate the quality and quantity of GC reactions during Plasmodium infection in C57BL/6 mice infected with P. yoelii 17XNL [6,34]. Relatively higher abundance of Clostridium papyrosolven, Alistipes putredinis, Alistipes timonensis, and Lactobacillus reuteri at genus level and Bacteroidiaceae and Lacnospiraceae at family level were associated with susceptibility to hyperparasitemia while Lactobacillus johnsonii, among others, with resistance to hyperparasitemia correlating to GC responses in mice [6]. Possibly, gut microbiota associated with resistance to hyperparasitemia might be linked to higher levels of SCFAs, flagellin, peptidoglycans, lipopolysaccharides, cross reactive epitopes, but not PA in the gut, resulting in B cells in Peyer’s patches and the spleen producing better antibody responses [39,81,82].

5.2. T helper 1 (TH1) cells

The contribution CD4+ TH1 cells to protection against blood-stage malaria is complicated and nuanced [79]. Both IFNγ producing Th1 cells and IL-10 producing Th1 cells (Tr1) are required to control Plasmodium infection [83]. Tissue damage done by inflammatory cytokines like IFNγ is regulated by anti-inflammatory cytokine IL-10 [83,84]. Gut microbiota-derived SCFAs promote IL-10 production by Th1 cells to maintain intestinal homeostasis. Gut microbiota depleted with antibiotics have enhanced intestinal Th1 cell response [85], and C57BL/6 mice become resistant to severe hyperparasitemia to P. yoelii 17XNL infection when treated with oral antibiotics [6]. Additionally, probiotic Lactobacillus strains can result in macrophage-mediated induction of Th1 response [75,86].

5.3. Regulatory T cells

Foxp3+ Tregs have ubiquitous roles in anti-malarial immunity in both mice and humans, yet owing to the pleiotropic effect of these cells and the complexity of anti-malaria immunity, the full contribution of these cells to protection and pathology during malaria is not fully understood [69,87]. Lower Treg cell numbers are associated with lower parasite burden and better outcomes in humans during blood-stage malaria [72,88,89]. However, depletion of Treg cells in FoxP3–diphtheria toxin receptor (DTR) transgenic (DEREG) C57BL/6 mice did not decrease ECM severity suggesting a limited role in ECM [90]. Still, depletion of Tregs or blocking Cytotoxic T-lymphocyte-Associated protein (CTLA-4) expressed on Tregs during a narrow window-of-time just prior to peak parasitemia enhanced immune responses and accelerated parasite clearance. This enhanced protection was attributed to increased Tfh:B cell interactions in GC reactions generating a more robust antibody response during blood-stage P. yoelii 17XNL infection in mice [72]. Germ-free mice colonized with SFB have increased expression of RORγt+ Tregs [91], and SCFAs (e.g., butyrate and propionate) produced by commensal gut bacteria promote peripheral Treg generation [92]. Consistent with gut microbiota stimulating expansion of these cells, Tregs are significantly reduced in germ-free or antibiotic-treated mice [91]. Although the mechanism is unknown, mice treated with antibiotics have significantly decreased P. yoelii 17XNL burden compared to untreated mice [6].

5.4. CD8+ T cells

Cytotoxic CD8+ T cells can protect against liver-stage malaria while providing little help in controlling blood-stage malaria [69,93]. However, CD8+ T cells have been shown in rodent ECM to cause breakdown of the blood–brain barrier [94] and have been found in the brain microvasculature of humans [70,95,96]. Several studies have demonstrated the ability of gut microbiota to regulate CD8+ T cell responses. Mice that develop more colitis-associated tumors have increased numbers of CD8+IFNγ+ T cells in lamina propria with higher relative abundance of Alistipes, Ruminococcus, Prevotellaceae, and lower abundance of Lachnospiraceae in the gut compared to mice that develop low tumor burden [97]. In contrast, circulating numbers of gut-derived CD8+IFNγ+ T cells induced by a mixture of 11 gut bacterial species belonging to 8 genera (Bacteroides, Parabacteroides, Alistipes, Paraprevotella, Eubacterim, Ruminococcaceae, Phascolarctobacterium, and Fusobacterium) had enhanced anti-tumor immunity against subcutaneous engraftment of MC38 adenocarcinoma cell and anti-microbial immunity against oral Listeria monocytogenes challenge [98]. The ability of gut microbiota to modulate CD8+ T cell responses during malaria, in particular during cerebral malaria, is an area ripe for exploration.

5.5. γδ T cells

γδ T cells recognize Plasmodium-infected erythrocytes and destroy infected RBCs either through cytotoxic molecules or antibody dependent phagocytosis [99,100]. Additionally, liver-stage dependent, low blood-stage Plasmodium parasite mass activates γδ T cells to produce IL-17, which protects mice from lethal cerebral malaria by promoting erythropoiesis [101]. Administration of lactic acid producing Lactobacillus plantarum probiotic promotes hematopoiesis and erythropoiesis [102], whether lactic acid-producing bacteria confer protection against severe malaria remains unclear. Interestingly, C57BL/6 mice fed a high fat diet (HFD) are resistant to ECM [103]. Numerous factors may contribute to HFD protection to ECM, but a HFD can modulate gut microbiota, generally leading to a decrease in Bacteroidetes and increase in Firmicutes and Proteobacteria [104]. Mice fed HFD for 3 weeks have increased number of IL-17+ γδ T cells, IFNγ+ Th1 cells, and CD8+ T cells while decreased numbers of Tregs in the colon and small intestine [105]. However, increased abundance of Proteobacteria and lower abundance of Clostridiaceae and S24-7 is also associated with induction of CD4+ Tregs cells [106], highlighting the complex interaction between gut microbiota and immune system. Whether HFD protection from ECM is attributed to diet-dependent effects on gut microbiota and their modulation of host immune cells or alternative microbiome-independent effects is not known.

5.6. NKT cells

NKT cells have been shown to reduce blood-stage parasitemia due to enhanced secretion of IFNγ [107]. NKT cells are usually found in thymus, spleen, liver, and bone marrow [108]. Depleting gram-positive gut bacteria with vancomycin that are involved in the conversion of primary bile acids to secondary bile acids was able to induce hepatic NKT cell accumulation and decreased liver tumor growth [109]. Vancomycin-treated mice had negligible presence of Bacteroidales and significantly reduced Clostridiales [109]. Interestingly, monocolonization with Clostridium scindens, which can transform bile acid, was able to reduce hepatic NKT cells and recover Bacteroidales [109,110]. Yet, the impact of gut microbiota modulating NKT cells other than in the liver is unknown; therefore, the ability of gut microbiota to modulate NKT cells and impact blood-stage parasite burden is unknown. Presently, there are no reported effects of gut microbiota on Plasmodium liver-stage burden, but as NKT cells are believed to have a protective role in liver-stage Plasmodium infections [74,111], it raises the possibility gut microbiota may impact liver-stage burden via modulation of NKT cells.

5.7. Natural killer (NK) cells

NK cells have beneficial roles during Plasmodium infection [112]. NK cells produce inflammatory cytokines, kill infected RBCs, and participate in initiation and development of adaptive immune response during malaria infection [113116]. Butyrate is one of the major SCFAs produced by gut microbiota that is reported to limit the effector function of human NK cells from blood in vitro by down-regulation of mTORC1 activity, c-Myc mRNA expression, and metabolism [117]. In contrast, dietary butyrate supplementation or treatment with Clostridium butyricum in mice treated with antibiotics early in life promoted the maturation and restored function of liver-resident NK cells [118]. Faecalibacterium, Roseburia, Fusobacteria, and Eubacterium are other bacteria that can produce butyrate [119]. Additionally, high salt diet can enhance NK cell functions in a gut microbiota dependent way by increasing the abundance of probiotic bacteria Bifidobacterium but increased gut permeability [120].

5.8. Monocytes

Monocytes have both protective and pathological role in malaria infection [121]. Enhanced p53 expression in monocytes are associated with attenuated Plasmodium-induced inflammation and protects from early fever during malaria infection in humans [30]. IFNγ leads to enhanced expression of p53 in monocytes to attenuate pro-inflammatory activation [30,122,123]. Gut microbial products have been associated with regulation and function of splenic monocytes [124]. Oral antibiotics treatment eliminated bacterial taxa from Bacteroides and Firmicutes along with other bacteria that reduced pattern recognition receptor ligands in the serum that led to immature phenotype of splenic Ly6Chigh monocytes exhibiting decreased level of pro-inflammatory cytokines and increased phagocytic abilities [124].

6. Targeting gut microbiota as a strategy to decrease severe malaria

There are numerous approaches by which gut microbiota can be modulated. In this section, we have reviewed some of the past efforts and recent advances and strategies to manipulate gut microbiota. These approaches may serve as useful approaches to interrogate gut microbiota–host–parasite interactions, with some approaches serving as potential gut microbiota-based approaches to mitigate severe malaria. The latter will require extensive investigation and clinical trials with positive malaria outcome before any recommendations can be made (Fig 3).

6.1. High fat, calorie restricted, and low protein diets

Studies have shown that high fat, calorie restricted, and low protein diets are associated with favorable parasitemia and mortality outcomes in rodent malaria models [103,125,126]. However, the role of gut microbiota, mechanisms of action, and identification of targetable pathways are required to advance dietary gut microbiota-based therapeutics.

6.2. Milk

Rodents on a milk diet suppress P. berghei infection, while having no effect on Nutallia rodhaini (Babesia) or Trypanosoma brucei infection [127129]. Similar protections were seen against 2 nonhuman primate (monkey) strains, P. knowlesi and P. cynomolgi [129]. Moreover, mice and monkeys fed a milk diet supplemented with p-aminobenzoate, a growth factor for many Plasmodium species, lost protection against malaria [129]. Milk consumption has been shown to decrease Bacteroidetes and Prevotella while increase Proteobacteria, Bifidobacterium, Lactobacillus, and Roseburia [130,131]. Connections between milk consumption and its effect on gut microbiota to modulate malaria severity warrants further investigation.

6.3. Folic acid (FA)

Controlled trials have found that FA supplementation compromised the efficacy of antimalaria drugs and should be avoided as supplement in children in malaria endemic regions [132,133]. FA induces Tregs [134] and production of folate is positively associated with higher relative abundance of Bacteroides, Sutterella, and Parasutterella [135] that are associated with high blood-stage parasite burden.

6.4. Probiotics

Probiotics are valued for the health benefits they confer upon the host, and these have been investigated in the context of Plasmodium infections. Heat killed Lactobacillus sakei HS-1 was able to minimize weight loss and mitigate intestinal pathology and limited small intestine shortening in C57BL/6 mice during P. berghei infection [136]. However, parasitemia load was not decreased in Lactobacillus sakei-treated mice [136]. Likewise, L. casei administration was shown to be protective against P. berghei as an adjunct therapy along with antimalarial treatment in BALB/C mice [137,138]. Additionally, C57BL/6 mice administered with Bifidobacterium longum alone had diminished P. berghei burden, colon inflammation, and significantly lower level of plasma TNFα and IFNγ compared to B. longum plus L. casei or L. casei alone [139]. Expression of intestinal CD103+ dendritic cells and intestinal Tregs were high in mice receiving B. longum [139]. CD103+ dendritic cells in nonlymphoid organs induces Tregs [140]. Dendritic cells have a critical role in initiating and regulating innate and adaptive immunity against malaria [141]. Finally, some probiotic bacteria strains express high levels of α-gal [142], which may facilitate protection against Plasmodium exoerythrocytic stages via induction of anti-α-gal antibodies, as discussed above.

One of the challenges with probiotics is poor engraftment. Understanding the dietary requirements of probiotic bacteria will be important as it can be targeted to improve engraftment. For example, some strains of Bacteroides ovatus can utilize the marine polysaccharide, porphyran, which is lacking in the vast majority of gut bacteria [143]. Transfer of the gene cluster for porphyran utilization from B. ovatus into another Bacteroides species lacking this cluster allowed fine tuning the engraftment of these bacteria into the competitive mouse gut microbiota niche when mice were provided porphyran in their diet. Similar approaches can be used to clone unique nutrient utilization gene clusters into probiotic strains with proven beneficial effect against malaria to overcome the probiotic wash-out effect.

6.5. Antimicrobial peptides

Non-immunogenic and non-toxic antimicrobial peptides from Lactobacillus plantarum strain LR/14 inhibited the growth of P. falciparum in vitro without any hemolysis [144]. L. plantarum can also inhibit many gram-positive and gram-negative bacteria [145], which can be exploited to manipulate gut microbiota composition.

6.6. Antibiotics

C57BL/6 mice treated with any of 4 antibiotics (ampicillin, gentamicin, metronidazole, and vancomycin) in drinking water prior to and during P. yoelii 17XNL infection significantly decrease the parasite burden. Of note, treatment of mice resistant to P. yoelii 17XNL hyperparasitemia showed no effect (i.e., the mice did not become susceptible). Moreover, treating with oral vancomycin 1 week prior to P. yoelii 17XNL infection, followed by cessation of treatment, significantly decreased P. yoelii 17XNL parasite burden. Intriguingly, preinfection vancomycin treatment afforded resistance to P. yoelii 17XNL hyperparasitemia for at least 3 months post-cessation of vancomycin treatment. Although antibiotic use poses threat to emergence of antibiotic resistance, clinical trials performed with antibiotics (amoxicillin, cefdinir, ceftriaxone, metronidazole) were associated with positive outcome in malnourished children [146148]. Thus, clinical trials to test antibiotics as an adjunct therapy to prevent and manage severe malaria may have merit.

6.7. Bacteriophages

Bacteriophages are viruses that can attack bacteria with specificity [149]. Bacteriophages, natural or engineered, can dynamically modulate gut microbiota and the metabolome [150]. Bacteriophages have been used to precisely modulate gut microbiota in diseases like IBD, colitis, type 2 diabetes, and colorectal cancer among others to improve prognosis with minimal damage to host gut microbiota [149,151,152]. Of course, this exciting technology comes with limitations and challenges [149]. For example, evolution of phage resistance, efficacy of phages against biofilms, and stability of phage preparations are a few important challenges [153]. Nevertheless, if specific gut bacteria are involved in susceptibility to severe malaria, then targeting these bacteria via bacteriophages might result in promising outcomes.

6.8. Short chain fatty acids (SCFAs)

Previously, we have shown that levels of SCFAs like propionic acid, butyric acid, and valeric acid were significantly different among mice susceptible and resistant to P. yoelii 17XNL hyperparasitemia [39]. Culminating evidence suggests important roles of SCFAs in the development and regulation of the immune system and gut barrier integrity that impact disease severity and enhanced health [154,155]. Consequently, SCFAs may impact malaria severity; however, the contribution of any specific SCFAs in malaria severity is yet to be studied.

6.9. Dietary tryptophan and indoles

Gut microbiota conversion of dietary tryptophan to indoles has important roles in enhancing gut barrier integrity and binds to AhR receptor on immune cells to induce anti-inflammatory and antimicrobial properties [156,157]. The indole moiety is one of the most promising chemotypes for the development of antiparasitic drugs [158]. Indole-3-acetic acid activated AhR pathway promotes anti-inflammatory cytokine IL-10 and up-regulated Foxp3 and increased Treg cells in a proteoglycan (PG)-induced ankylosis mouse model [159,160]. Therefore, the role of gut-derived indoles and the ability of indoles to induce Tregs in malaria will be an interesting avenue of research [161,162].

7. Conclusion

Within the last 2 decades, our understanding of gut microbiota function has increased exponentially due to advances in high-throughput omics technologies like nucleic acid sequencing, metabolomics, metatranscriptomics, proteomics, interbacterial and intrabacterial interaction within the host, interdisciplinary studies, and the rise of artificial intelligence and machine learning in biological sciences. With this new knowledge, the scientific community is positioned to identify specific gut microbiota and microbiota-derived products and their interactions with the host immune system in modulating the severity of malaria. The capacity to precisely identify these biomarkers will increase with continued development and innovation in the microbiome field. With these advancements, gut microbiome-based therapies may one day be used to mitigate the severity of malaria associated with Plasmodium infection.

Acknowledgments

We are thankful to members of the Schmidt Laboratory for helpful discussion of this article.

References

  1. 1. World Health Organization. World Malaria Report 2021 (World Health Organization). 2021.
  2. 2. Jagannathan P, Kakuru A. Malaria in 2022: Increasing challenges, cautious optimism. Nat Commun. 2022;13(1):2678. pmid:35562368
  3. 3. Zekar L, Sharman T. Plasmodium falciparum malaria. 2020.
  4. 4. Ashley EA, Pyae Phyo A, Woodrow CJ. Malaria Lancet. 2018;391(10130):1608–21. Epub 2018/04/11. pmid:29631781.
  5. 5. White NJ, Pukrittayakamee S, Hien TT, Faiz MA, Mokuolu OA, Dondorp AM. Malaria Lancet. 2014;383(9918):723–35. Epub 2013/08/21. pmid:23953767.
  6. 6. Mandal RK, Denny JE, Namazzi R, Opoka RO, Datta D, John CC, et al. Dynamic modulation of spleen germinal center reactions by gut bacteria during Plasmodium infection. Cell Rep. 2021;35(6):109094. pmid:33979614
  7. 7. Berg G, Rybakova D, Fischer D, Cernava T, Vergès M-CC, Charles T, et al. Microbiome definition re-visited: old concepts and new challenges. Microbiome. 2020;8:1–22.
  8. 8. York A. Your microbiome is what you eat. Nat Rev Microbiol. 2019;17(12):721. pmid:31624359
  9. 9. Zheng D, Liwinski T, Elinav E. Interaction between microbiota and immunity in health and disease. Cell Res. 2020;30(6):492–506. pmid:32433595
  10. 10. Fan Y, Pedersen O. Gut microbiota in human metabolic health and disease. Nat Rev Microbiol. 2021;19(1):55–71. pmid:32887946
  11. 11. Rose C, Parker A, Jefferson B, Cartmell E. The characterization of feces and urine: a review of the literature to inform advanced treatment technology. Crit Rev Environ Sci Technol. 2015;45(17):1827–1879. pmid:26246784
  12. 12. Wiertsema SP, van Bergenhenegouwen J, Garssen J, Knippels LM. The interplay between the gut microbiome and the immune system in the context of infectious diseases throughout life and the role of nutrition in optimizing treatment strategies. Nutrients. 2021;13(3):886. pmid:33803407
  13. 13. Bamgbose T, Alberdi P, Abdullahi IO, Inabo HI, Bello M, Sinha S, et al. Functional characterization of α-Gal producing lactic acid bacteria with potential probiotic properties. Sci Rep. 2022;12(1):1–12.
  14. 14. Mukherjee D, Chora ÂF, Mota MM. Microbiota, a third player in the host–Plasmodium affair. Trends Parasitol. 2020;36(1):11–18. pmid:31787522
  15. 15. Waide ML, Schmidt NW. The gut microbiome, immunity, and Plasmodium severity. Curr Opin Microbiol. 2020;58:56–61. pmid:33007644
  16. 16. Ippolito MM, Denny JE, Langelier C, Sears CL, Schmidt NW. Malaria and the microbiome: a systematic review. Clin Infect Dis. 2018;67(12):1831–1839. pmid:29701835
  17. 17. Winaris N, Pawestri AR, Azizah S, Alifia LI, Asiyah R, Ayuningtyas TR, et al. Plasmodium infection and dysbiosis: A new paradigm in the host–parasite interaction. Parasite Immunol. 2023:e12980. pmid:37092310
  18. 18. Yilmaz B, Portugal S, Tran TM, Gozzelino R, Ramos S, Gomes J, et al. Gut microbiota elicits a protective immune response against malaria transmission. Cell. 2014;159(6):1277–89. Epub 2014/12/07. pmid:25480293; PubMed Central PMCID: PMC4261137.
  19. 19. Steinke JW, Platts-Mills TA, Commins SP. The alpha-gal story: lessons learned from connecting the dots. J Allergy Clin Immunol. 2015;135(3):589–596. pmid:25747720
  20. 20. Aguilar R, Ubillos I, Vidal M, Balanza N, Crespo N, Jiménez A, et al. Antibody responses to α-Gal in African children vary with age and site and are associated with malaria protection. Sci Rep. 2018;8(1):9999.
  21. 21. Pacheco I, Díaz-Sánchez S, Contreras M, Villar M, Cabezas-Cruz A, Gortázar C, et al. Probiotic bacteria with high alpha-Gal content protect zebrafish against mycobacteriosis. Pharmaceuticals. 2021;14(7):635. pmid:34208966
  22. 22. Mateos-Hernández L, Risco-Castillo V, Torres-Maravilla E, Bermúdez-Humarán LG, Alberdi P, Hodžić A, et al. Gut Microbiota Abrogates Anti-α-Gal IgA Response in Lungs and Protects against Experimental Aspergillus Infection in Poultry. Vaccines (Basel). 2020;8(2). Epub 2020/06/11. pmid:32517302; PubMed Central PMCID: PMC7350254.
  23. 23. Portillo S, Zepeda BG, Iniguez E, Olivas JJ, Karimi NH, Moreira OC, et al. A prophylactic α-Gal-based glycovaccine effectively protects against murine acute Chagas disease. Npj Vaccines. 2019;4(1):1–15.
  24. 24. Iniguez E, Schocker NS, Subramaniam K, Portillo S, Montoya AL, Al-Salem WS, et al. An α-Gal-containing neoglycoprotein-based vaccine partially protects against murine cutaneous leishmaniasis caused by Leishmania major. PLoS Negl Trop Dis. 2017;11(10):e0006039.
  25. 25. Hodžić A, Mateos-Hernández L, de la Fuente J, Cabezas-Cruz A. α-Gal-based vaccines: advances, opportunities, and perspectives. Trends Parasitol. 2020;36(12):992–1001.
  26. 26. Sanches-Vaz M, Temporão A, Luis R, Nunes-Cabaço H, Mendes AM, Goellner S, et al. Trypanosoma brucei infection protects mice against malaria. PLoS Pathog. 2019;15(11):e1008145. pmid:31703103
  27. 27. Kotepui KU, Masangkay FR, De Jesus MG, Kotepui M. Prevalence and outcomes of malaria as co-infection among patients with human African trypanosomiasis: a systematic review and meta-analysis. Sci Rep. 2021;11(1):23777. pmid:34893680
  28. 28. Yooseph S, Kirkness EF, Tran TM, Harkins DM, Jones MB, Torralba MG, et al. Stool microbiota composition is associated with the prospective risk of Plasmodium falciparum infection. BMC Genomics. 2015;16(1):631. pmid:26296559
  29. 29. Tran TM, Ongoiba A, Coursen J, Crosnier C, Diouf A, Huang C-Y, et al. Naturally acquired antibodies specific for Plasmodium falciparum reticulocyte-binding protein homologue 5 inhibit parasite growth and predict protection from malaria. J Infect Dis. 2014;209(5):789–798. pmid:24133188
  30. 30. Tran TM, Guha R, Portugal S, Skinner J, Ongoiba A, Bhardwaj J, et al. A molecular signature in blood reveals a role for p53 in regulating malaria-induced inflammation. Immunity. 2019;51(4):750–65.e10. pmid:31492649
  31. 31. Raj DK, Nixon CP, Nixon CE, Dvorin JD, DiPetrillo CG, Pond-Tor S, et al. Antibodies to PfSEA-1 block parasite egress from RBCs and protect against malaria infection. Science. 2014;344(6186):871–877. pmid:24855263
  32. 32. Huwe T, Prusty BK, Ray A, Lee S, Ravindran B, Michael E. Interactions between parasitic infections and the human gut microbiome in Odisha, India. Am J Trop Med Hyg. 2019;100(6):1486. pmid:30963988
  33. 33. Easton AV, Raciny-Aleman M, Liu V, Ruan E, Marier C, Heguy A, et al. Immune response and microbiota profiles during coinfection with Plasmodium vivax and soil-transmitted helminths. MBio. 2020;11(5):e01705–e01720. pmid:33082257
  34. 34. Waide ML, Polidoro R, Powell WL, Denny JE, Kos J, Tieri DA, et al. Gut Microbiota Composition Modulates the Magnitude and Quality of Germinal Centers during Plasmodium Infections. Cell Rep. 2020;33(11):108503. pmid:33326773
  35. 35. Villarino NF, LeCleir GR, Denny JE, Dearth SP, Harding CL, Sloan SS, et al. Composition of the gut microbiota modulates the severity of malaria. Proc Natl Acad Sci U S A. 2016;113(8):2235–40. Epub 2016/02/10. pmid:26858424; PubMed Central PMCID: PMC4776451.
  36. 36. Stough J, Dearth SP, Denny JE, LeCleir GR, Schmidt NW, Campagna SR, et al. Functional characteristics of the gut microbiome in C57BL/6 mice differentially susceptible to Plasmodium yoelii. Front Microbiol. 2016;7:1520. pmid:27729904
  37. 37. Mandal RK, Denny JE, Waide ML, Li Q, Bhutiani N, Anderson CD, et al. Temporospatial shifts within commercial laboratory mouse gut microbiota impact experimental reproducibility. BMC Biol. 2020;18(1):1–12.
  38. 38. Denny JE, Powers JB, Castro HF, Zhang J, Joshi-Barve S, Campagna SR, et al. Differential sensitivity to plasmodium yoelii infection in C57BL/6 mice impacts gut-liver axis homeostasis. Sci Rep. 2019;9(1):1–15.
  39. 39. Chakravarty S, Mandal RK, Duff ML, Schmidt NW. Intestinal short-chain fatty acid composition does not explain gut microbiota-mediated effects on malaria severity. PLoS ONE. 2019;14(3):e0214449. pmid:30917184
  40. 40. Smith CDM, Gong M, Andrew AK, Russ BN, Ge Y, Zadeh M, et al. Composition of the gut microbiota transcends genetic determinants of malaria infection severity and influences pregnancy outcome. EBioMedicine. 2019;44:639–655. pmid:31160271
  41. 41. Mutoni JA, Coutelier J-P, Rujeni N, Mutesa L, Cani PD. Possible interactions between malaria, helminthiases and the gut microbiota: a short review. Microorganisms. 2022;10(4):721. pmid:35456772
  42. 42. Njua-Yafi C, Nkuo-Akenji T, Anchang-Kimbi J, Apinjoh T, Mugri R, Chi H, et al. The Effect of Helminth Co-infection on malaria-specific immunoglobulin g responses. BMJ Glob Health. 2017;2(Suppl 2).
  43. 43. Tuasha N, Hailemeskel E, Erko B, Petros B. Comorbidity of intestinal helminthiases among malaria outpatients of Wondo Genet health centers, southern Ethiopia: implications for integrated control. BMC Infect Dis. 2019;19(1):1–8.
  44. 44. Salazar-Castañon VH, Legorreta-Herrera M, Rodriguez-Sosa M. Helminth parasites alter protection against Plasmodium infection. Biomed Res Int. 2014;2014. pmid:25276830
  45. 45. Sanders D. Mucosal integrity and barrier function in the pathogenesis of early lesions in Crohn’s disease. J Clin Pathol. 2005;58(6):568–572. pmid:15917403
  46. 46. Gewirtz AT. Flag in the crossroads: flagellin modulates innate and adaptive immunity. Curr Opin Gastroenterol. 2006;22(1):8–12. pmid:16319670
  47. 47. Deleu S, Machiels K, Raes J, Verbeke K, Vermeire S. Short chain fatty acids and its producing organisms: An overlooked therapy for IBD? EBioMedicine. 2021;66:103293. pmid:33813134
  48. 48. MacFabe DF. Short-chain fatty acid fermentation products of the gut microbiome: implications in autism spectrum disorders. Microb Ecol Health Dis. 2012;23(1):19260. pmid:23990817
  49. 49. Kawasoe J, Uchida Y, Kawamoto H, Miyauchi T, Watanabe T, Saga K, et al. Propionic acid, induced in gut by an inulin diet, suppresses inflammation and ameliorates liver ischemia and reperfusion injury in mice. Front Immunol. 2022;13. pmid:35572528
  50. 50. Sa A-L, Roelofsen H, Rezaee F, Weening D, Hoek A, Vonk R, et al. Propionic acid affects immune status and metabolism in adipose tissue from overweight subjects. Eur J Clin Invest. 2012;42(4):357–364. pmid:21913915
  51. 51. Abdelli LS, Samsam A, Naser SA. Propionic acid induces gliosis and neuro-inflammation through modulation of PTEN/AKT pathway in autism spectrum disorder. Sci Rep. 2019;9(1):1–12.
  52. 52. Rahfeld P, Sim L, Moon H, Constantinescu I, Morgan-Lang C, Hallam SJ, et al. An enzymatic pathway in the human gut microbiome that converts A to universal O type blood. Nat Microbiol. 2019;4(9):1475–1485. pmid:31182795
  53. 53. Mooney JP, Lokken KL, Byndloss MX, George MD, Velazquez EM, Faber F, et al. Inflammation-associated alterations to the intestinal microbiota reduce colonization resistance against non-typhoidal Salmonella during concurrent malaria parasite infection. Sci Rep. 2015;5:14603. pmid:26434367
  54. 54. Guan W, Song X, Yang S, Zhu H, Li F, Li J. Observation of the gut microbiota profile in BALB/c mice induced by Plasmodium yoelii 17XL infection. Front Microbiol. 2022:13. pmid:35432291
  55. 55. Farinella DN, Kaur S, Tran V, Cabrera-Mora M, Joyner CJ, Lapp SA, et al. Malaria disrupts the rhesus macaque gut microbiome. Front Cell Infect Microbiol. 2023;12:1931. pmid:36710962
  56. 56. Yawen Z, Xiangyun C, Binyou L, Xingchen Y, Taiping L, Xuedong Z, et al. The dynamic landscape of parasitemia dependent intestinal microbiota shifting and the correlated gut transcriptome during Plasmodium yoelii infection. Microbiol Res. 2022;258:126994. pmid:35220138
  57. 57. Wang W, Qian H, Cao J. Stem cell therapy: a novel treatment option for cerebral malaria? Stem Cell Res Ther. 2015;6:1–3.
  58. 58. Birbeck GL, Molyneux ME, Kaplan PW, Seydel KB, Chimalizeni YF, Kawaza K, et al. Blantyre Malaria Project Epilepsy Study (BMPES) of neurological outcomes in retinopathy-positive paediatric cerebral malaria survivors: a prospective cohort study. Lancet Neurol. 2010;9(12):1173–1181. pmid:21056005
  59. 59. Xie Y, Guan W, Zhao Y, Yan S, Guo K, Chen S, et al. Deficiency of migration inhibitory factor influences the gut microbiota of C57BL/6 mice infected with Plasmodium berghei ANKA. Front Microbiol. 2022;13. pmid:36033889
  60. 60. Knowler SA, Shindler A, Wood JL, Lakkavaram A, Thomas CJ, de Koning-Ward TF, et al. Altered gastrointestinal tract structure and microbiome following cerebral malaria infection. Parasitol Res. 2023;1–11.
  61. 61. Fan Z-g, Li X, Fu H-y, Zhou L-m, Gong F-l, Fang M. Gut microbiota reconstruction following host infection with blood-stage Plasmodium berghei ANKA strain in a murine model. Curr Med Sci. 2019;39:883–889.
  62. 62. Taniguchi T, Miyauchi E, Nakamura S, Hirai M, Suzue K, Imai T, et al. Plasmodium berghei ANKA causes intestinal malaria associated with dysbiosis. Sci Rep. 2015;5(1):1–13. pmid:26503461
  63. 63. Shimada M, Hirose Y, Shimizu K, Yamamoto DS, Hayakawa EH, Matsuoka H. Upper gastrointestinal pathophysiology due to mouse malaria Plasmodium berghei ANKA infection. Trop Med Health. 2019;47(1):1–11. pmid:30872946
  64. 64. Mandal RK, Crane RJ, Berkley JA, Gumbi W, Wambua J, Ngoi JM, et al. Longitudinal analysis of infant stool bacteria communities before and after acute febrile malaria and artemether-lumefantrine treatment. J Infect Dis. 2018.
  65. 65. Denny JE, Schmidt NW. Oral Administration of Clinically Relevant Antimalarial Drugs Does Not Modify the Murine Gut Microbiota. Sci Rep. 2019;9(1):1–9.
  66. 66. Borsom EM, Conn K, Keefe CR, Herman C, Orsini GM, Hirsch AH, et al. Predicting Neurodegenerative Disease Using Prepathology Gut Microbiota Composition: a Longitudinal Study in Mice Modeling Alzheimer’s Disease Pathologies. Microbiol Spectr. 2023:e03458–e03422. pmid:36877047
  67. 67. Venzon M, Bernard-Raichon L, Klein J, Axelrad JE, Zhang C, Hussey GA, et al. Gut microbiome dysbiosis during COVID-19 is associated with increased risk for bacteremia and microbial translocation. Biorxiv. 2021. pmid:34341786
  68. 68. Zeng M, Inohara N, Nuñez G. Mechanisms of inflammation-driven bacterial dysbiosis in the gut. Mucosal Immunol. 2017;10(1):18–26. pmid:27554295
  69. 69. Kurup SP, Butler NS, Harty JT. T cell-mediated immunity to malaria. Nat Rev Immunol. 2019;19(7):457–471. pmid:30940932
  70. 70. Rénia L, Grau GE, Wassmer SC. CD8+ T cells and human cerebral malaria: a shifting episteme. J Clin Invest. 2020;130(3):1109–1111. pmid:32065593
  71. 71. Kumar R, Ng S, Engwerda C. The role of IL-10 in malaria: a double edged sword. Front Immunol. 2019;10:229. pmid:30809232
  72. 72. Kurup SP, Obeng-Adjei N, Anthony SM, Traore B, Doumbo OK, Butler NS, et al. Regulatory T cells impede acute and long-term immunity to blood-stage malaria through CTLA-4. Nat Med. 2017;23(10):1220–1225. pmid:28892065
  73. 73. Angulo I, Fresno M. Cytokines in the pathogenesis of and protection against malaria. Clin Vaccine Immunol. 2002;9(6):1145–1152. pmid:12414742
  74. 74. Belachew EB. Immune response and evasion mechanisms of Plasmodium falciparum parasites. J Immunol Res. 2018;2018. pmid:29765991
  75. 75. Shim JA, Ryu JH, Jo Y, Hong C. The role of gut microbiota in T cell immunity and immune mediated disorders. Int J Biol Sci. 2023;19(4):1178–1191. pmid:36923929
  76. 76. Soon MS, Haque A. Recent insights into CD4+ Th cell differentiation in malaria. J Immunol. 2018;200(6):1965–1975. pmid:29507121
  77. 77. Luckheeram RV, Zhou R, Verma AD, Xia B. CD4+ T cells: differentiation and functions. Clin Dev Immunol. 2012;2012.
  78. 78. Visweswaran GRR, Vijayan K, Chandrasekaran R, Trakhimets O, Brown SL, Vigdorovich V, et al. Germinal center activity and B cell maturation are associated with protective antibody responses against Plasmodium pre-erythrocytic infection. PLoS Pathog. 2022;18(7):e1010671. pmid:35793394
  79. 79. Perez-Mazliah D, Langhorne J. CD4 T-cell subsets in malaria: TH1/TH2 revisited. Front Immunol. 2015;5:671. pmid:25628621
  80. 80. Soon MS, Nalubega M, Boyle MJ. T-follicular helper cells in malaria infection and roles in antibody induction. Oxf Open Immunol. 2021;2(1):iqab008. pmid:36845571
  81. 81. Kim M, Qie Y, Park J, Kim CH. Gut microbial metabolites fuel host antibody responses. Cell Host Microbe. 2016;20(2):202–214. pmid:27476413
  82. 82. Lynn DJ, Benson SC, Lynn MA, Pulendran B. Modulation of immune responses to vaccination by the microbiota: implications and potential mechanisms. Nat Rev Immunol. 2022;22(1):33–46. pmid:34002068
  83. 83. Edwards CL, Ng SS, de Labastida RF, Corvino D, Engel JA, de Oca MM, et al. IL-10–producing Th1 cells possess a distinct molecular signature in malaria. J Clin Invest. 2023;133(4).
  84. 84. Ouyang W, Rutz S, Crellin NK, Valdez PA, Hymowitz SG. Regulation and functions of the IL-10 family of cytokines in inflammation and disease. Annu Rev Immunol. 2011;29:71–109. pmid:21166540
  85. 85. Kim M, Galan C, Hill AA, Wu W-J, Fehlner-Peach H, Song HW, et al. Critical role for the microbiota in CX3CR1+ intestinal mononuclear phagocyte regulation of intestinal T cell responses. Immunity. 2018;49(1):151–163.e5.
  86. 86. Won TJ, Kim B, Song DS, Lim YT, Oh ES, Lee DI, et al. Modulation of Th1/Th2 balance by Lactobacillus strains isolated from Kimchi via stimulation of macrophage cell line J774A. 1 in vitro. J Food Sci. 2011;76(2):H55–H61. pmid:21535768
  87. 87. Van Braeckel-Budimir N, Kurup SP, Harty JT. Regulatory issues in immunity to liver and blood-stage malaria. Curr Opin Immunol. 2016;42:91–97. pmid:27351448
  88. 88. Walther M, Tongren JE, Andrews L, Korbel D, King E, Fletcher H, et al. Upregulation of TGF-β, FOXP3, and CD4+ CD25+ regulatory T cells correlates with more rapid parasite growth in human malaria infection. Immunity. 2005;23(3):287–296.
  89. 89. Torcia MG, Santarlasci V, Cosmi L, Clemente A, Maggi L, Mangano VD, et al. Functional deficit of T regulatory cells in Fulani, an ethnic group with low susceptibility to Plasmodium falciparum malaria. Proc Natl Acad Sci U S A. 2008;105(2):646–651. pmid:18174328
  90. 90. Steeg C, Adler G, Sparwasser T, Fleischer B, Jacobs T. Limited role of CD4+ Foxp3+ regulatory T cells in the control of experimental cerebral malaria. J Immunol. 2009;183(11):7014–7022. pmid:19890049
  91. 91. Ohnmacht C, Park J-H, Cording S, Wing JB, Atarashi K, Obata Y, et al. The microbiota regulates type 2 immunity through RORγt+ T cells. Science. 2015;349(6251):989–993.
  92. 92. Arpaia N, Campbell C, Fan X, Dikiy S, Van Der Veeken J, Deroos P, et al. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature. 2013;504(7480):451–455. pmid:24226773
  93. 93. Vinetz JM, Kumar S, Good MF, Fowlkes B, Berzofsky J, Miller L. Adoptive transfer of CD8+ T cells from immune animals does not transfer immunity to blood stage Plasmodium yoelii malaria. J Immunol. 1990;144(3):1069–74. pmid:1967271
  94. 94. Swanson PA, Hart GT, Russo MV, Nayak D, Yazew T, Peña M, et al. CD8+ T cells induce fatal brainstem pathology during cerebral malaria via luminal antigen-specific engagement of brain vasculature. PLoS Pathog. 2016;12(12):e1006022. pmid:27907215
  95. 95. Barrera V, Haley MJ, Strangward P, Attree E, Kamiza S, Seydel KB, et al. Comparison of CD8+ T cell accumulation in the brain during human and murine cerebral malaria. Front Immunol. 2019;10:1747. pmid:31396236
  96. 96. Riggle BA, Manglani M, Maric D, Johnson KR, Lee M-H, Neto OLA, et al. CD8+ T cells target cerebrovasculature in children with cerebral malaria. J Clin Invest. 2020;130(3):1128–1138. pmid:31821175
  97. 97. Amy IY, Zhao L, Eaton KA, Ho S, Chen J, Poe S, et al. Gut microbiota modulate CD8 T cell responses to influence colitis-associated tumorigenesis. Cell Rep. 2020;31(1):107471.
  98. 98. Tanoue T, Morita S, Plichta DR, Skelly AN, Suda W, Sugiura Y, et al. A defined commensal consortium elicits CD8 T cells and anti-cancer immunity. Nature. 2019;565(7741):600–605. pmid:30675064
  99. 99. Junqueira C, Polidoro RB, Castro G, Absalon S, Liang Z, Sen Santara S, et al. γδ T cells suppress Plasmodium falciparum blood-stage infection by direct killing and phagocytosis. Nat Immunol. 2021;22(3):347–357.
  100. 100. Lefebvre MN, Harty JT. γδ T cells burst malaria’s bubble. Nat Immunol. 2021;22(3):270–272.
  101. 101. Chora ÂF, Marques S, Gonçalves JL, Lima P, da Costa DG, Fernandez-Ruiz D, et al. Interplay between liver and blood stages of Plasmodium infection dictates malaria severity via γδ T cells and IL-17-promoted stress erythropoiesis. Immunity. 2023;56(3):592–605.e8.
  102. 102. Lee Y-S, Kim T-Y, Kim Y, Kim S, Lee S-H, Seo S-U, et al. Microbiota-derived lactate promotes hematopoiesis and erythropoiesis by inducing stem cell factor production from leptin receptor+ niche cells. Exp Mol Med. 2021;53(9):1319–1331. pmid:34497346
  103. 103. Oliveira-Lima OC, Almeida NL, Almeida-Leite CM, Carvalho-Tavares J. Mice chronically fed a high-fat diet are resistant to malaria induced by Plasmodium berghei ANKA. Parasitol Res. 2019;118:2969–2977. pmid:31482465
  104. 104. Murphy EA, Velazquez KT, Herbert KM. Influence of high-fat-diet on gut microbiota: a driving force for chronic disease risk. Curr Opin Clin Nutr Metab Care. 2015;18(5):515. pmid:26154278
  105. 105. Luck H, Tsai S, Chung J, Clemente-Casares X, Ghazarian M, Revelo XS, et al. Regulation of obesity-related insulin resistance with gut anti-inflammatory agents. Cell Metab. 2015;21(4):527–542. pmid:25863246
  106. 106. Papotto PH, Yilmaz B, Silva-Santos B. Crosstalk between γδ T cells and the microbiota. Nat Microbiol. 2021;6(9):1110–1117.
  107. 107. Fu Y, Ding Y, Wang Q, Zhu F, Tan Y, Lu X, et al. Blood-stage malaria parasites manipulate host innate immune responses through the induction of sFGL2. Sci Adv. 2020;6(9):eaay9269. pmid:32133407
  108. 108. Wu L, Van Kaer L. Natural killer T cells in health and disease. Front Biosci (Schol Ed). 2011;3:236. pmid:21196373
  109. 109. Ma C, Han M, Heinrich B, Fu Q, Zhang Q, Sandhu M, et al. Gut microbiome–mediated bile acid metabolism regulates liver cancer via NKT cells. Science. 2018;360(6391):eaan5931. pmid:29798856
  110. 110. Marion S, Studer N, Desharnais L, Menin L, Escrig S, Meibom A, et al. In vitro and in vivo characterization of Clostridium scindens bile acid transformations. Gut Microbes. 2019;10(4):481–503. pmid:30589376
  111. 111. Taylor-Robinson A. NKT cells protect against pre-erythocytic malaria. Trends Microbiol. 2000;8(10):450. pmid:11044674
  112. 112. Burrack KS, Hart GT, Hamilton SE. Contributions of natural killer cells to the immune response against Plasmodium. Malar J. 2019;18(1):1–9.
  113. 113. Tukwasibwe S, Nakimuli A, Traherne J, Chazara O, Jayaraman J, Trowsdale J, et al. Variations in killer-cell immunoglobulin-like receptor and human leukocyte antigen genes and immunity to malaria. Cell Mol Immunol. 2020;17(8):799–806. pmid:32541835
  114. 114. Hart GT, Tran TM, Theorell J, Schlums H, Arora G, Rajagopalan S, et al. Adaptive NK cells in people exposed to Plasmodium falciparum correlate with protection from malaria. J Exp Med. 2019;216(6):1280–1290. pmid:30979790
  115. 115. Odera DO, Tuju J, Mwai K, Nkumama IN, Fürle K, Chege T, et al. Anti-merozoite antibodies induce natural killer cell effector function and are associated with immunity against malaria. Sci Transl Med. 2023;15(682):eabn5993. pmid:36753561
  116. 116. Ty M, Sun S, Callaway PC, Rek J, Press KD, van der Ploeg K, et al. Malaria-driven expansion of adaptive-like functional CD56-negative NK cells correlates with clinical immunity to malaria. Sci Transl Med. 2023;15(680):eadd9012. pmid:36696483
  117. 117. Zaiatz-Bittencourt V, Jones F, Tosetto M, Scaife C, Cagney G, Jones E, et al. Butyrate limits human natural killer cell effector function. Sci Rep. 2023;13(1):2715. pmid:36792800
  118. 118. Tian P, Yang W, Guo X, Wang T, Tan S, Sun R, et al. Early life gut microbiota sustains liver-resident natural killer cells maturation via the butyrate-IL-18 axis. Nat Commun. 2023;14(1):1710. pmid:36973277
  119. 119. Anand S, Kaur H, Mande SS. Comparative in silico analysis of butyrate production pathways in gut commensals and pathogens. Front Microbiol. 2016;7:1945. pmid:27994578
  120. 120. Rizvi ZA, Dalal R, Sadhu S, Kumar Y, Kumar S, Gupta SK, et al. High-salt diet mediates interplay between NK cells and gut microbiota to induce potent tumor immunity. Sci Adv. 2021;7(37):eabg5016. pmid:34516769
  121. 121. Dobbs KR, Crabtree JN, Dent AE. Innate immunity to malaria—the role of monocytes. Immunol Rev. 2020;293(1):8–24. pmid:31840836
  122. 122. Namgaladze D, Brüne B. Pharmacological Activation of p53 during Human Monocyte to Macrophage Differentiation Attenuates Their Pro-Inflammatory Activation by TLR4, TLR7 and TLR8 Agonists. Cancer. 2021;13(5):958. pmid:33668835
  123. 123. Liu G, Park Y-J, Tsuruta Y, Lorne E, Abraham E. p53 attenuates lipopolysaccharide-induced NF-κB activation and acute lung injury. J Immunol. 2009;182(8):5063–5071.
  124. 124. Kolypetri P, Liu S, Cox LM, Fujiwara M, Raheja R, Ghitza D, et al. Regulation of splenic monocyte homeostasis and function by gut microbial products. Iscience. 2021;24(4):102356. pmid:33898947
  125. 125. Mancio-Silva L, Slavic K, Grilo Ruivo MT, Grosso AR, Modrzynska KK, Vera IM, et al. Nutrient sensing modulates malaria parasite virulence. Nature. 2017;547(7662):213–216. pmid:28678779
  126. 126. Murr NJ, Olender TB, Smith MR, Smith AS, Pilotos J, Richard LB, et al. Plasmodium chabaudi infection alters intestinal morphology and mucosal innate immunity in moderately malnourished mice. Nutrients. 2021;13(3):913. pmid:33799736
  127. 127. Nowell F. The effect of a milk diet upon Plasmodium berghei, Nuttallia (= Babesia) rodhaini and Trypanosoma brucei infections in mice. Parasitology. 1970;61(3):425–433. pmid:4994294
  128. 128. Maegraith B, Deegan T, Jones ES. Suppression of malaria (P. berghei) by milk. Br Med J. 1952;2(4799):1382. pmid:12997791
  129. 129. Milk Hawking F., p-aminobenzoate, and malaria of rats and monkeys. Br Med J. 1954;1(4859):425.
  130. 130. Aslam H, Marx W, Rocks T, Loughman A, Chandrasekaran V, Ruusunen A, et al. The effects of dairy and dairy derivatives on the gut microbiota: A systematic literature review. Gut Microbes. 2020;12(1):1799533. pmid:32835617
  131. 131. Fernandez-Raudales D, Hoeflinger JL, Bringe NA, Cox SB, Dowd SE, Miller MJ, et al. Consumption of different soymilk formulations differentially affects the gut microbiomes of overweight and obese men. Gut Microbes. 2012;3(6):490–500. pmid:22895080
  132. 132. Ouma P, Parise ME, Hamel MJ, Kuile FOt, Otieno K, Ayisi JG et al. A randomized controlled trial of folate supplementation when treating malaria in pregnancy with sulfadoxine-pyrimethamine. PLoS Clinical Trials. 2006;1(6):e28. pmid:17053829
  133. 133. Kupka R. The role of folate in malaria–implications for home fortification programmes among children aged 6–59 months. Matern Child Nutr. 2015;11:1–15.
  134. 134. Kinoshita M, Kayama H, Kusu T, Yamaguchi T, Kunisawa J, Kiyono H, et al. Dietary folic acid promotes survival of Foxp3+ regulatory T cells in the colon. J Immunol. 2012;189(6):2869–2878. pmid:22869901
  135. 135. Malinowska AM, Schmidt M, Kok DE, Chmurzynska A. Ex vivo folate production by fecal bacteria does not predict human blood folate status: Associations between dietary patterns, gut microbiota, and folate metabolism. Food Res Int. 2022;156:111290. pmid:35651056
  136. 136. Shimada M, Hayakawa EH, Matsuoka H. Heat-killed Lactobacillus sakei HS-1 mitigates small intestinal pathophysiology on Plasmodium berghei ANKA infected C57BL/6 mice. 自治医科大学紀要 = Jichi Medical University Journal. 2021;43:13–19.
  137. 137. Mahajan E, Sinha S, Bhatia A, Sehgal R, Medhi B. Evaluation of the effect of probiotic as add-on therapy with conventional therapy and alone in malaria induced mice. BMC Res Notes. 2021;14(1):1–5.
  138. 138. Toukam LL, Fossi BT, Taiwe GS, Bila RB, Sofeu DDF, Ivo EP, et al. In vivo antimalarial activity of a probiotic bacterium Lactobacillus sakei isolated from traditionally fermented milk in BALB/c mice infected with Plasmodium berghei ANKA. J Ethnopharmacol. 2021;280:114448. pmid:34303805
  139. 139. Fitri LE, Sardjono TW, Winaris N, Pawestri AR, Endharti AT, Norahmawati E, et al. Bifidobacterium longum Administration Diminishes Parasitemia and Inflammation During Plasmodium berghei Infection in Mice. J Inflamm Res. 2023;1393–404. pmid:37006809
  140. 140. Cao Q, Lu J, Li Q, Wang C, Wang XM, Lee VW, et al. CD103+ dendritic cells elicit CD8+ T cell responses to accelerate kidney injury in adriamycin nephropathy. J Am Soc Nephrol. 2016;27(5):1344–1360. pmid:26376858
  141. 141. Yap XZ, Lundie RJ, Beeson JG, O’Keeffe M. Dendritic cell responses and function in malaria. Front Immunol. 2019;10:357. pmid:30886619
  142. 142. Singh S, Bastos-Amador P, Thompson JA, Truglio M, Yilmaz B, Cardoso S, et al. Glycan-based shaping of the microbiota during primate evolution. Elife. 2021;10:e67450. pmid:34009123
  143. 143. Shepherd ES, DeLoache WC, Pruss KM, Whitaker WR, Sonnenburg JL. An exclusive metabolic niche enables strain engraftment in the gut microbiota. Nature. 2018;557(7705):434–438. pmid:29743671
  144. 144. Gupta R, Rajendran V, Ghosh PC, Srivastava S. Assessment of anti-plasmodial activity of non-hemolytic, non-immunogenic, non-toxic antimicrobial peptides (AMPs LR14) produced by Lactobacillus plantarum LR/14. Drugs R&D. 2014;14:95–103. pmid:24797399
  145. 145. Dinev T, Beev G, Tzanova M, Denev S, Dermendzhieva D, Stoyanova A. Antimicrobial activity of Lactobacillus plantarum against pathogenic and food spoilage microorganisms: a review. Bulg J Vet Med. 2018;21(3).
  146. 146. Trehan I, Goldbach HS, LaGrone LN, Meuli GJ, Wang RJ, Maleta KM, et al. Research Article (New England Journal of Medicine) Antibiotics as part of the management of severe acute malnutrition. Malawi Med J. 2016;28(3):123–130.
  147. 147. Heikens GT, Schofield WN, Christie C, Gernay J, Dawson S. The Kingston Project. III. The effects of high energy supplement and metronidazole on malnourished children rehabilitated in the community: morbidity and growth. Eur J Clin Nutr. 1993;47(3):174–191. pmid:8458315
  148. 148. Standing JF, Ongas MO, Ogwang C, Kagwanja N, Murunga S, Mwaringa S, et al. Dosing of ceftriaxone and metronidazole for children with severe acute malnutrition. Clin Pharmacol Ther. 2018;104(6):1165–1174. pmid:29574688
  149. 149. Mirzaei MK, Deng L. New technologies for developing phage-based tools to manipulate the human microbiome. Trends Microbiol. 2022;30(2):131–142. pmid:34016512
  150. 150. Hsu BB, Gibson TE, Yeliseyev V, Liu Q, Lyon L, Bry L, et al. Dynamic modulation of the gut microbiota and metabolome by bacteriophages in a mouse model. Cell Host Microbe. 2019;25(6):803–814.e5. pmid:31175044
  151. 151. Zheng D-W, Dong X, Pan P, Chen K-W, Fan J-X, Cheng S-X, et al. Phage-guided modulation of the gut microbiota of mouse models of colorectal cancer augments their responses to chemotherapy. Nat Biomed Eng. 2019;3(9):717–728. pmid:31332342
  152. 152. El Haddad L, Mendoza JF, Jobin C. Bacteriophage-mediated manipulations of microbiota in gastrointestinal diseases. Front Microbiol. 2022;13:4512. pmid:36466675
  153. 153. Pires DP, Costa AR, Pinto G, Meneses L, Azeredo J. Current challenges and future opportunities of phage therapy. FEMS Microbiol Rev. 2020;44(6):684–700. pmid:32472938
  154. 154. Oliveira RA, Pamer EG. Assembling symbiotic bacterial species into live therapeutic consortia that reconstitute microbiome functions. Cell Host Microbe. 2023;31(4):472–484. pmid:37054670
  155. 155. Pérez-Reytor D, Puebla C, Karahanian E, García K. Use of short-chain fatty acids for the recovery of the intestinal epithelial barrier affected by bacterial toxins. Front Physiol. 2021;12:650313. pmid:34108884
  156. 156. Ye X, Li H, Anjum K, Zhong X, Miao S, Zheng G, et al. Dual role of indoles derived from intestinal microbiota on human health. Front Immunol. 2022;13. pmid:35784338
  157. 157. Zhang LS, Davies SS. Microbial metabolism of dietary components to bioactive metabolites: opportunities for new therapeutic interventions. Genome Med. 2016;8(1):1–18.
  158. 158. Pacheco PA, Santos MM. Recent Progress in the development of indole-based compounds active against malaria, trypanosomiasis and Leishmaniasis. Molecules. 2022;27(1):319. pmid:35011552
  159. 159. Konopelski P, Mogilnicka I. Biological effects of indole-3-propionic acid, a gut microbiota-derived metabolite, and its precursor tryptophan in mammals’ health and disease. Int J Mol Sci. 2022;23(3):1222. pmid:35163143
  160. 160. Shen J, Yang L, You K, Chen T, Su Z, Cui Z, et al. Indole-3-acetic acid alters intestinal microbiota and alleviates ankylosing spondylitis in mice. Front Immunol. 2022;13:262. pmid:35185872
  161. 161. Santos RO, Cruz MGS, Lopes SCP, Oliveira LB, Nogueira PA, Lima ES, et al. A first Plasmodium vivax natural infection induces increased activity of the interferon gamma-driven tryptophan catabolism pathway. Front Microbiol. 2020;11:400. pmid:32256470
  162. 162. Wyatt M, Greathouse KL. Targeting dietary and microbial tryptophan-indole metabolism as therapeutic approaches to colon cancer. Nutrients. 2021;13(4):1189. pmid:33916690