Skip to main content
Advertisement
  • Loading metrics

Intestinal injury and the gut microbiota in patients with Plasmodium falciparum malaria

  • Natthida Sriboonvorakul,

    Roles Conceptualization, Writing – original draft, Writing – review & editing

    Affiliation Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand

  • Kesinee Chotivanich,

    Roles Writing – review & editing

    Affiliations Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand

  • Udomsak Silachamroon,

    Roles Writing – review & editing

    Affiliation Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand

  • Weerapong Phumratanaprapin,

    Roles Writing – review & editing

    Affiliation Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand

  • John H. Adams,

    Roles Writing – original draft, Writing – review & editing

    Affiliation Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, Tampa, Florida, United States of America

  • Arjen M. Dondorp ,

    Roles Conceptualization, Writing – original draft, Writing – review & editing

    arjen@tropmedres.ac (AMD); s.j.leopold@amsterdamumc.nl (SJL)

    Affiliations Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand, Centre for Tropical Medicine, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom

  • Stije J. Leopold

    Roles Conceptualization, Writing – original draft, Writing – review & editing

    arjen@tropmedres.ac (AMD); s.j.leopold@amsterdamumc.nl (SJL)

    Affiliation Department of Internal Medicine, Division of Infectious Diseases, Amsterdam University Medical Center, location AMC, the Netherlands

Abstract

The pathophysiology of severe falciparum malaria involves a complex interaction between the host, parasite, and gut microbes. In this review, we focus on understanding parasite-induced intestinal injury and changes in the human intestinal microbiota composition in patients with Plasmodium falciparum malaria. During the blood stage of P. falciparum infection, infected red blood cells adhere to the vascular endothelium, leading to widespread microcirculatory obstruction in critical tissues, including the splanchnic vasculature. This process may cause intestinal injury and gut leakage. Epidemiological studies indicate higher rates of concurrent bacteraemia in severe malaria cases. Furthermore, severe malaria patients exhibit alterations in the composition and diversity of the intestinal microbiota, although the exact contribution to pathophysiology remains unclear. Mouse studies have demonstrated that the gut microbiota composition can impact susceptibility to Plasmodium infections. In patients with severe malaria, the microbiota shows an enrichment of pathobionts, including pathogens that are known to cause concomitant bloodstream infections. Microbial metabolites have also been detected in the plasma of severe malaria patients, potentially contributing to metabolic acidosis and other clinical complications. However, establishing causal relationships requires intervention studies targeting the gut microbiota.

Introduction

Malaria remains a significant global health burden, with an estimated 247 million cases and 620.000 deaths in 2021 [1]. The majority of severe and fatal malaria cases are caused by Plasmodium falciparum, one of the 5 malaria parasite species affecting humans. A crucial aspect of the pathophysiology of severe falciparum malaria is the extensive sequestration of parasitized red blood cells in the microcirculation, which impairs microcirculatory blood flow and leads to dysfunction of vital organs [2]. Additionally, endothelial activation and glycocalyx dysfunction are believed to further compromise tissue perfusion [3]. The degree of microvascular dysfunction can be assessed by directly observing the microcirculation and estimating the sequestered parasite biomass using plasma P. falciparum histidine-rich protein 2 (PfHRP2) [4,5]. Notably, microvascular sequestration of parasitized red blood cells is prominent in the gut [4,6], affecting gut perfusion and potentially disrupting tight and adherent junctions in the gut epithelium. This disruption compromises the gut’s barrier function and facilitates the translocation of enteric bacteria into the bloodstream, as previously reviewed [7].

The microbiome consists of a diverse group of bacteria, archaea, fungi, protozoa, and viruses. Among the human-associated microbial communities, the gut microbiota is the largest and most heterogeneous [8]. The gut microbiota has been implicated in numerous physiological processes, including energy homeostasis, metabolism, gut epithelial health, immunologic activity, and neurobehavioral development [9]. Furthermore, the microbiota’s composition may play a role in the pathogenesis of various infectious diseases, such as influenza [10], bacterial sepsis [11], Clostridium difficile enteritis [12], and in malaria as shown in prior reviews of mouse studies [13,14]. Previous mouse studies have demonstrated that different gut microbiota compositions result in varying disease severities [1518], in particular, in mouse models of malaria [15]. Several studies in both human and rodent malaria have shown changes in gut microbiota composition during Plasmodium infections, with an association observed with disease severity. The causal relationship between gut microbiota composition and disease severity in humans remains unclear, but it may offer new insights into the pathophysiology of severe malaria and potential targets for intervention.

In this review, we focus on the evidence from clinical studies in patients with malaria for intestinal injury, changes in microbiota composition, and their potential consequences in P. falciparum malaria. These consequences include the translocation of bacteria and their metabolites across the gut barrier and alterations in gut immunological functions. Furthermore, we briefly discuss potential interventions targeting the gut microbiota.

In a parallel review, Mandal and Schmidt discuss the evidence and insights from laboratory and experimental studies into the interaction between the host gut microbiome and malaria.

Parasite-induced intestinal injury

Patients with malaria commonly experience gastrointestinal symptoms such as nausea, vomiting, abdominal pain, and diarrhea. Intestinal damage occurs through a complex pathological cascade, primarily driven by the extensive sequestration of parasitized red blood cells in the splanchnic microcirculation of severe malaria patients. For a summary of the mechanisms involved, see Table 1.

thumbnail
Table 1. Evidence from clinical studies in patients with malaria for a pathological cascade of parasite-induced intestinal injury, impaired intestinal barrier function, translocation of bacteria and metabolites into the bloodstream, and an altered gut microbiota composition.

https://doi.org/10.1371/journal.ppat.1011661.t001

The splanchnic circulation supplies blood to abdominal organs, including the liver, spleen, stomach, pancreas, small intestine, and large intestine. It is perfused by 3 branches of the abdominal aorta: the coeliac artery, superior mesenteric artery, and inferior mesenteric artery.

The splanchnic blood flow is primarily influenced by systemic vascular resistance and cardiac output. Under normal conditions, splanchnic blood flow accounts for about 25% to 30% of cardiac output, but it can vary depending on factors such as recent feeding or physiological stress [19].

Hemodynamic shock is rare in severe malaria, as both systemic vascular resistance and cardiac output are typically maintained at adequate levels despite infections with a large parasite load. This is different from the macrovascular changes seen in bacterial sepsis. The key feature of septic shock is significant peripheral arteriolar vasodilation. This leads to low systemic vascular resistance, high cardiac output, severe hypotension, and shock, with subsequent inadequate tissue perfusion. Studies have shown that systemic vascular resistance remains at adequate levels in both uncomplicated and severe malaria, possibly due to the vasoconstrictive effects of cell-free hemoglobin released during red cell hemolysis [20]. Mildly elevated systemic vascular resistance has been observed in fatal cases of severe falciparum malaria [21]. It is unlikely that macrovascular changes contribute to splanchnic bed hypoperfusion and intestinal injury in severe malaria.

Sequestration of parasitized red blood cells, however, significantly affects the blood flow in the intestines’ microcirculation. Autopsy studies have shown that, given the total blood volume of the splanchnic circulation, the intestines represent a significant proportion of the total body sequestration [6,22]. Video-microscopy of the rectal circulation in living patients with severe malaria has confirmed widespread microcirculatory obstruction in the intestines [4]. Microvascular sequestration of parasitized red blood cells is believed to cause intestinal damage and contribute to the development of hyperlactatemia in severe malaria.

Additional mechanisms that may contribute to intestinal injury include ischemia-reperfusion injury, intestinal inflammation, and mast cell activation. Mast cell activation can damage the gut barrier, both physically and immunologically, through the release of Th2 cytokines that affect the defence against bacteria that may translocate from the intestine [23,24]. This may lead to disruption of the tight and adherent junctions between gut epithelial cells, further compromising gut barrier function, as previously reviewed [7].

Impaired intestinal barrier function

Intestinal injury in patients with malaria can progress to impaired intestinal barrier function, as evidenced by increased intestinal permeability and abnormal markers of intestinal integrity, Table 1. In adults with severe malaria, observational studies have shown that patients with a high parasite biomass exhibit reduced enterocyte integrity, indicated by decreased plasma L-citrulline, a marker produced by enterocytes in the small intestine [25]. In pediatric patients with malaria, elevated levels of trefoil factor 3 (TFF-3) and intestinal fatty acid binding protein (I-FABP), markers of intestinal injury, are associated with severe malaria and an increased risk of death [26].

Impairment of the gut barrier can increase the likelihood of translocation of enteric bacteria into the bloodstream, leading to concomitant bacteraemia and sepsis. In children with severe malaria in Africa, concurrent invasive bacterial infections are described, involving bacteria such as Streptococcus pneumoniae, nontyphoidal Salmonella, and Escherichia coli [2730]. Additionally, studies have observed elevated plasma concentrations of bacterial metabolites in patients with severe falciparum malaria [31,32].

Gut microbiota alterations in malaria

The composition of the gut microbiota can potentially influence the progression of malaria infection through various mechanisms. One important mechanism is colonization resistance, which refers to the ability of the gut microbiota to prevent the overgrowth of harmful bacteria by employing different mechanisms, including the reduction of gut pH [4143]. Perturbations in the gut microbiota can disrupt this balance and lead to the overgrowth and translocation of harmful bacteria, resulting in the dissemination of their metabolites into the bloodstream. This phenomenon has been observed in the development of C. difficile enteritis following broad-spectrum antibiotic treatment [44]. The gut microbiota also plays a role in modulating the immune response, both innate and adaptive, and changes in the microbiota due to antibiotic treatment have been shown to weaken the immune response to certain pathogens [45]. Additionally, the gut microbiota contributes to gut barrier function through the production of short-chain fatty acids [11].

In an observational study conducted in Bangladesh, the gut microbiota composition of adult patients with severe and uncomplicated falciparum malaria was compared to healthy volunteers [25]. The study utilized sequencing of the V4 region of the 16S rRNA gene amplified from fecal DNA [25]. Patients with severe malaria showed a significant enrichment of potentially pathogenic Enterococcus and Escherichia/Shigella species in their gut microbiota, pathogens that are known to be able to cause bloodstream infections [25]. Furthermore, an abundance of lactate-producing species, including Bacteroides, Streptococcus spp., and Lactobacillus spp., in the gut microbiota was associated with the severity of metabolic acidosis, which is a strong predictor of fatal outcomes in severe malaria [25,31]. However, the causal mechanisms underlying these associations remain unclear, and it is important to consider various environmental and patient-related factors that can influence the composition of the gut microbiota, such as age, diet, comorbidities, and prior treatment with antibiotics or antimalarials. To date, no intervention studies targeting the gut microbiota in patients with malaria have been conducted.

Furthermore, mouse models have shown changes in the gut microbiota composition following infection with different Plasmodium species. In Swiss Webster and C57BL/6 (B6) mice infected with Plasmodium yoelii, a reduction in the Firmicutes/Bacteroidetes ratio and a decrease in Proteobacteria were observed [46]. Another study in B6 and BALB/c mice infected with Plasmodium berghei revealed a decrease in Firmicutes and, specifically in one mouse strain, an increase in Proteobacteria and Verrucomicrobia [47]. In C57BL/6 mice, liver damage and bile acid depletion correlated with an increase in gut bacterial diversity during and after infection with P. yoelii, suggesting a potential role of bile acids in shaping the gut microbiota [48]. These findings highlight the alterations of the gut microbiota in malaria infection.

Impact of gut microbiota on Plasmodium infections

The role of the gut microbiota in influencing the immune response to malaria has been studied primarily in mouse models, with some evidence suggesting a similar effect in human malaria. Here, we summarize important findings from recent experimental studies, but for more insights into the interaction between the host gut microbiome and malaria, see the parallel review by Mandal and Schmidt, where the experimental evidence is further elaborated.

Mouse models of Plasmodium berghei or Plasmodium yoelii infection, including BALB/c and C57BL/6 mice, have been used to investigate the relationship between the gut microbiota and malaria pathogenesis. Studies have shown that mice colonized with the gut pathobiont Escherichia coli O86:B7, which expresses α-galactosyl, produce antibodies (against α-galactosyl) that cross-react with Plasmodium sporozoites, and could mediate clinical protection against malaria infection [49].

The composition of the gut microbiota in mouse models appears to impact parasite burden and fatality rates following infection with various Plasmodium species. In one study, fecal content from mice with different susceptibility to P. yoelii infection was transplanted into germ-free mice, demonstrating that resistance to infection could be transferred through fecal transplant [15]. In this study, relative protection against Plasmodium correlated with the abundance of Lactobacillus and Bifidobacterium bacteria [15].

Differences in parasite burden and bacterial community composition have been observed between different strains of mice. For example, Taconic mice showed lower peak parasite burden and faster recovery compared to Charles River mice, suggesting that the host–microbiota interaction plays a role in parasite burden rather than genetics or environmental factors [50]. The study also identified differences in gene expression, including the cell surface receptor basigin, which may link the gut microbiome and malaria resistance.

Intestinal helminth infections, such as hookworm (Ancylostoma duodenale, Necator americanus), ascaris (Ascaris lumbricoides), and whipworm (Trichuris trichiura), have also been associated with malaria susceptibility [51], although the data are not conclusive.

Studies on hookworm coinfections largely indicate increased susceptibility to malaria. A study from Ethiopia reported that the intensity of hookworm (and trichuriasis) coinfections was associated with increased densities of both P. falciparum and P. vivax [51]. Other studies from Uganda and Zimbabwe investigating hookworm coinfection reported early P. falciparum parasitemia [52,53], while another study from Uganda showed no association between hookworm infection and early or delayed parasitemia [54]. Most studies on Ascaris lumbricoides coinfections suggest a protective effect against malaria. A negative correlation was observed between the intensity of A. lumbricoides infection and P. falciparum and P. vivax parasitemia [51]. Another study from Thailand suggested protection from coinfection with Ascaris lumbricoides against the development of cerebral malaria or renal failure in patients with severe malaria [55,56]. However, a study from Cameroon reported no association between intestinal helminths (including ascaris and hookworm) and the clinical outcome of malaria [57].

Proposed mechanisms for the protective effect of ascaris infection include endothelial cell receptor down-regulation and the production of IgE-anti-IgE immune complexes that reduce the severity of falciparum malaria. Endothelial cell receptor down-regulation reduces parasite erythrocyte cytoadherence or selective splenic parasite clearance, thus reducing the proportion of virulent P. falciparum strains. It has also been suggested that IgE-anti-IgE immune complexes resulting from helminth infections reduce the severity of falciparum malaria and can mediate tolerance to the malaria parasite through the CD23/NO pathway [55]. Studies examining malaria in pregnancy have shown a negative correlation between A. lumbricoides infection and the risk of P. vivax malaria [58], while hookworm has been associated with an increased incidence of P. falciparum but not P. vivax parasitemia [56,59]. Overall, based on these epidemiological observations the relationship between intestinal helminth infections and malaria susceptibility or severity remains inconclusive.

Potential interventions targeting the gut microbiota

Therapies aimed at modifying the gut microbiota composition could be potential interventions to influence susceptibility to and severity of Plasmodium infections. These interventions include probiotics, selective digestive tract decontamination, fecal transplants, and antibiotics.

Probiotics

Probiotics are live microorganisms that can modify the gut microbiota. When combined with prebiotics, which support their growth, they are known as synbiotics [60,61]. Common probiotic bacteria include Lactobacilli spp., Bifidobacteria spp., Saccharomyces boulardii, and Bacillus coagulans. Probiotics and synbiotics have been suggested to reduce pathobionts through colonization resistance, prevent bacterial translocation, degrade toxins, and modulate the immune response [60].

While probiotics and synbiotics have been primarily studied for their potential in sepsis, their effects on Plasmodium infections remain largely unexplored in human studies. However, experimental mouse studies have shown promising results and have suggested a positive effect on time to death, reduction of bacteraemia, and improved gut wall integrity [62]. A large Indian study demonstrated a protective effect of a synbiotic containing Lactobacillus plantarum plus fructooligosaccharide on the prevalence of sepsis in neonates and infants. Although an earlier meta-analysis suggested a beneficial effect of probiotics on the prevention of ventilator-associated pneumonia (VAP) [63], a recent large multicenter, double-blinded, randomized controlled trial comparing the efficacy of the probiotic Lactobacillus rhamnosus GG (LGG) versus placebo in preventing VAP did not confirm these findings [64].

There are no human studies on the use of probiotics in patients with malaria.

Probiotics containing Lactobacillus and Bifidobacterium have demonstrated a beneficial effect in reducing P. yoelii parasitemia in another experimental mouse study [15]. The administration of Lactobacillus casei reduced the severity of Plasmodium chabaudi infection [65]. In a mouse model of Plasmodium berghei, the efficacy of Lactobacillus casei as adjuvant therapy to chloroquine, an antimalarial drug, was evaluated and showed a reduction in peripheral blood parasitemia with probiotic treatment [66].

Selective digestive tract decontamination

Selective digestive tract decontamination (SDD) involves the use of non-absorbable antimicrobials applied daily in the oropharynx and gastrointestinal tract [11]. SDD has been shown to reduce nosocomial infections and lower mortality in large trials involving critically ill patients in intensive care units in the Netherlands. It is now a standard infection prevention measure in Dutch ICUs [67]. SDD prevents colonization of potentially pathogenic microorganisms, including gram-negative aerobic microorganisms and Staphylococcus aureus, in the oropharynx and intestines [68]. SDD has not been studied as an adjunctive therapy in malaria and has also not been investigated in animal models of malaria.

Other interventions: Fecal transplants and antibiotics

Fecal microbiota transplantation (FMT) involves the administration of a solution of fecal material from a healthy donor into the intestinal tract of a recipient through a feeding tube to restore the gut microbiota [69]. FMT is currently used in the treatment of severe C. difficile infections (CDI) [70]. It has also been studied in patients with other causes of diarrhea or sepsis [71]. However, FMT has not been investigated in human or animal models of malaria.

Antibiotics represent a potential intervention for the treatment of bacterial coinfections in severe malaria. World Health Organization management guidelines recommend empirical broad-spectrum antibacterial therapy for all children diagnosed with severe falciparum malaria in malaria-endemic areas [72].

Conclusions

Intestinal injury and the gut microbiota appear to play a role in the severity and outcome of falciparum malaria. Mouse studies show that altering the microbiota affects susceptibility to Plasmodium infections. In humans, malaria leads to changes in the gut microbiota, including an increase in pathogens associated with severe disease and bacterial infections. Studies in patients with malaria show parasite-induced intestinal injury. Subsequently, impaired intestinal barrier function could allow translocation of gut microbiota and microbial metabolites into the bloodstream, potentially leading to concomitant sepsis. Observational studies in humans provide associations between the microbiota and disease severity or protection. Possible interventions include probiotic therapy, selective digestive tract decontamination, or fecal transplantation therapy. However, intervention studies are needed to establish causal relationships.

References

  1. 1. World Health Organization. World malaria report 2021. Geneva: World Health Organization; 2021.
  2. 2. White NJ, Pukrittayakamee S, Hien TT, Faiz MA, Mokuolu OA, Dondorp AM. Malaria Lancet. 2014;383(9918):723–35.
  3. 3. Bush MA, Anstey NM, Yeo TW, Florence SM, Granger DL, Mwaikambo ED, et al. Vascular Dysfunction in Malaria: Understanding the Role of the Endothelial Glycocalyx. Front Cell Dev Biol. 2021;9:757829. pmid:34858979
  4. 4. Dondorp AM, Ince C, Charunwatthana P, Hanson J, van Kuijen AM, Faiz MA, et al. Direct In Vivo Assessment of Microcirculatory Dysfunction in Severe Falciparum Malaria. J Infect Dis. 2008;197(1):79–84. pmid:18171289
  5. 5. Dondorp AM, Desakorn V, Pongtavornpinyo W, Sahassananda D, Silamut K, Chotivanich K, et al. Estimation of the Total Parasite Biomass in Acute Falciparum Malaria from Plasma PfHRP2. PLoS Med. 2005;2(8):e204. pmid:16104831
  6. 6. Pongponratn E, Riganti M, Punpoowong B, Aikawa M. Microvascular sequestration of parasitized erythrocytes in human falciparum malaria: a pathological study. Am J Trop Med Hyg. 1991;44(2):168–75. pmid:2012260
  7. 7. Donnelly E, de Water JV, Luckhart S. Malaria-induced bacteremia as a consequence of multiple parasite survival strategies. Curr Res Microb Sci. 2021;2:100036. pmid:34841327
  8. 8. Harris VC, Haak BW, B van Hensbroek M, Wiersinga WJ. The Intestinal Microbiome in Infectious Diseases: The Clinical Relevance of a Rapidly Emerging Field. Open Forum Infect Dis. 2017;4(3):ofx144. pmid:28852682
  9. 9. Barko PC, McMichael MA, Swanson KS, Williams DA. The Gastrointestinal Microbiome: A Review. J Vet Intern Med. 2018;32(1):9–25. pmid:29171095
  10. 10. Ichinohe T, Pang IK, Kumamoto Y, Peaper DR, Ho JH, Murray TS, et al. Microbiota regulates immune defense against respiratory tract influenza A virus infection. Proc Natl Acad Sci U S A. 2011;108(13):5354–9. pmid:21402903
  11. 11. Haak BW, Wiersinga WJ. The role of the gut microbiota in sepsis. Lancet Gastroenterol Hepatol. 2017;2(2):135–43. pmid:28403983
  12. 12. Seekatz AM, Young VB. Clostridium difficile and the microbiota. J Clin Invest. 2014;124(10):4182–9. pmid:25036699
  13. 13. Ippolito MM, Denny JE, Langelier C, Sears CL, Schmidt NW. Malaria and the Microbiome: A Systematic Review. Clin Infect Dis. 2018;67(12):1831–9. pmid:29701835
  14. 14. Ranallo RT, McDonald LC, Halpin AL, Hiltke T, Young VB. The State of Microbiome Science at the Intersection of Infectious Diseases and Antimicrobial Resistance. J Infect Dis. 2021;223(Supplement 3):S187–S93. pmid:33667294
  15. 15. 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. pmid:26858424
  16. 16. 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):83. pmid:32620114
  17. 17. 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
  18. 18. 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
  19. 19. Harper D, Chandler B. Splanchnic circulation. BJA Education. 2016;16(2):66–71.
  20. 20. Kingston HWF, Ghose A, Rungpradubvong V, Satitthummanid S, Herdman MT, Plewes K, et al. Cell-Free Hemoglobin Is Associated With Increased Vascular Resistance and Reduced Peripheral Perfusion in Severe Malaria. J Infect Dis. 2020;221(1):127–37. pmid:31693729
  21. 21. Hanson J, Lam SW, Mahanta KC, Pattnaik R, Alam S, Mohanty S, et al. Relative contributions of macrovascular and microvascular dysfunction to disease severity in falciparum malaria. J Infect Dis. 2012;206(4):571–9. pmid:22693227
  22. 22. Williams K, Milner J, Boudreau MD, Gokulan K, Cerniglia CE, Khare S. Effects of subchronic exposure of silver nanoparticles on intestinal microbiota and gut-associated immune responses in the ileum of Sprague-Dawley rats. Nanotoxicology. 2015;9(3):279–89. pmid:24877679
  23. 23. Bischoff SC, Krämer S. Human mast cells, bacteria, and intestinal immunity. Immunol Rev. 2007;217(1):329–37. pmid:17498069
  24. 24. Ribatti D. The Staining of Mast Cells: A Historical Overview. Int Arch Allergy Immunol. 2018;176(1):55–60. pmid:29597213
  25. 25. Leopold SJ, Ghose A. Gut microbiota composition in adult patients with severe and uncomplicated Plasmodium falciparum malaria compared to healthy volunteers. SSRN Electron J. 2021.
  26. 26. Sarangam ML, Namazzi R, Datta D, Bond C, Vanderpool CPB, Opoka RO, et al. Intestinal Injury Biomarkers Predict Mortality in Pediatric Severe Malaria. MBio. 2022;13(5):e01325–22. pmid:36069443
  27. 27. Phu NH, Day NPJ, Tuan PQ, Mai NTH, Chau TTH, Van Chuong L, et al. Concomitant Bacteremia in Adults With Severe Falciparum Malaria. Clin Infect Dis. 2020;71(9):e465–e70. pmid:32107527
  28. 28. Scott JA, Berkley JA, Mwangi I, Ochola L, Uyoga S, Macharia A, et al. Relation between falciparum malaria and bacteraemia in Kenyan children: a population-based, case-control study and a longitudinal study. Lancet. 2011;378(9799):1316–23. pmid:21903251
  29. 29. Nyein PP, Aung NM, Kyi TT, Htet ZW, Anstey NM, Kyi MM, et al. High Frequency of Clinically Significant Bacteremia in Adults Hospitalized With Falciparum Malaria. Open Forum Infect Dis. 2016;3(1):ofw028. pmid:26989752
  30. 30. Chau JY, Tiffany CM, Nimishakavi S, Lawrence JA, Pakpour N, Mooney JP, et al. Malaria-associated L-arginine deficiency induces mast cell-associated disruption to intestinal barrier defenses against nontyphoidal Salmonella bacteremia. Infect Immun. 2013;81(10):3515–26. pmid:23690397
  31. 31. Leopold SJ, Ghose A, Allman EL, Kingston HWF, Hossain A, Dutta AK, et al. Identifying the Components of Acidosis in Patients With Severe Plasmodium falciparum Malaria Using Metabolomics. J Infect Dis. 2019;219(11):1766–76. pmid:30566600
  32. 32. Olupot-Olupot P, Urban BC, Jemutai J, Nteziyaremye J, Fanjo HM, Karanja H, et al. Endotoxaemia is common in children with Plasmodium falciparum malaria. BMC Infect Dis. 2013;13:117. pmid:23497104
  33. 33. Milner DA Jr., Lee JJ, Frantzreb C, Whitten RO, Kamiza S, Carr RA, et al. Quantitative Assessment of Multiorgan Sequestration of Parasites in Fatal Pediatric Cerebral Malaria. J Infect Dis. 2015;212(8):1317–21. pmid:25852120
  34. 34. Hanson J, Lee SJ, Hossain MA, Anstey NM, Charunwatthana P, Maude RJ, et al. Microvascular obstruction and endothelial activation are independently associated with the clinical manifestations of severe falciparum malaria in adults: an observational study. BMC Med. 2015;13(1):122. pmid:26018532
  35. 35. Molyneux ME, Looareesuwan S, Menzies IS, Grainger SL, Phillips RE, Wattanagoon Y, et al. Reduced hepatic blood flow and intestinal malabsorption in severe falciparum malaria. Am J Trop Med Hyg. 1989;40(5):470–6. pmid:2729505
  36. 36. Wilairatana P, Meddings JB, Ho M, Vannaphan S, Looareesuwan S. Increased gastrointestinal permeability in patients with Plasmodium falciparum malaria. Clin Infect Dis. 1997;24(3):430–5. pmid:9114195
  37. 37. Olsson RA, Johnston EH. Histopathologic changes and small-bowel absorption in falciparum malaria. Am J Trop Med Hyg. 1969;18(3):355–9 pmid:4889829
  38. 38. Berkley JA, Bejon P, Mwangi T, Gwer S, Maitland K, Williams TN, et al. HIV infection, malnutrition, and invasive bacterial infection among children with severe malaria. Clin Infect Dis. 2009;49(3):336–43. pmid:19548833
  39. 39. Bassat Q, Guinovart C, Sigaúque B, Mandomando I, Aide P, Sacarlal J, et al. Severe malaria and concomitant bacteraemia in children admitted to a rural Mozambican hospital. Trop Med Int Health. 2009;14(9):1011–9. pmid:19552643
  40. 40. Aung NM, Nyein PP, Htut TY, Htet ZW, Kyi TT, Anstey NM, et al. Antibiotic Therapy in Adults with Malaria (ANTHEM): High Rate of Clinically Significant Bacteremia in Hospitalized Adults Diagnosed with Falciparum Malaria. Am J Trop Med Hyg. 2018;99(3):688–96. pmid:30014826
  41. 41. Lawley TD, Walker AW. Intestinal colonization resistance. Immunology. 2013;138(1):1–11. pmid:23240815
  42. 42. Kamada N, Chen GY, Inohara N, Núñez G. Control of pathogens and pathobionts by the gut microbiota. Nat Immunol. 2013;14(7):685–90. pmid:23778796
  43. 43. Liu P, Wang Y, Yang G, Zhang Q, Meng L, Xin Y, et al. The role of short-chain fatty acids in intestinal barrier function, inflammation, oxidative stress, and colonic carcinogenesis. Pharmacol Res. 2021;165:105420. pmid:33434620
  44. 44. Pike CM, Theriot CM. Mechanisms of colonization resistance against Clostridioides difficile. J Infect Dis. 2021;223(Supplement 3):S194–S200. pmid:33326565
  45. 45. Britton RA, Young VB. Role of the intestinal microbiota in resistance to colonization by Clostridium difficile. Gastroenterology. 2014;146(6):1547–53. pmid:24503131
  46. 46. 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
  47. 47. 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;5:15699. pmid:26503461
  48. 48. 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):15124.
  49. 49. 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. pmid:25480293
  50. 50. 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
  51. 51. Degarege A, Animut A, Legesse M, Erko B. Malaria severity status in patients with soil-transmitted helminth infections. Acta Trop. 2009;112(1):8–11. pmid:19497286
  52. 52. Hillier SD, Booth M, Muhangi L, Nkurunziza P, Khihembo M, Kakande M, et al. Plasmodium falciparum and helminth coinfection in a semi urban population of pregnant women in Uganda. J Infect Dis. 2008;198(6):920–7. pmid:18721060
  53. 53. Midzi N, Sangweme D, Zinyowera S, Mapingure MP, Brouwer KC, Munatsi A, et al. The burden of polyparasitism among primary schoolchildren in rural and farming areas in Zimbabwe. Trans R Soc Trop Med Hyg. 2008;102(10):1039–45. pmid:18656215
  54. 54. Shapiro AE, Tukahebwa EM, Kasten J, Clarke SE, Magnussen P, Olsen A, et al. Epidemiology of helminth infections and their relationship to clinical malaria in southwest Uganda. Trans R Soc Trop Med Hyg. 2005;99(1):18–24. pmid:15550257
  55. 55. Nacher M, Gay F, Singhasivanon P, Krudsood S, Treeprasertsuk S, Mazier D, et al. Ascaris lumbricoides infection is associated with protection from cerebral malaria. Parasite Immunol. 2000;22(3):107–13. pmid:10672191
  56. 56. Nacher M. Worms and malaria: blind men feeling the elephant? Parasitology. 2008;135(7):861–8. pmid:18377695
  57. 57. Kwenti TE, Nkume FA, Tanjeko AT, Kwenti TD. The Effect of Intestinal Parasitic Infection on the Clinical Outcome of Malaria in Coinfected Children in Cameroon. PLoS Negl Trop Dis. 2016;10(4):e0004673. pmid:27128975
  58. 58. Boel M, Carrara VI, Rijken M, Proux S, Nacher M, Pimanpanarak M, et al. Complex interactions between soil-transmitted helminths and malaria in pregnant women on the Thai-Burmese border. PLoS Negl Trop Dis. 2010;4(11):e887. pmid:21103367
  59. 59. Gebru T, Ajua A, Theisen M, Esen M, Ngoa UA, Issifou S, et al. Recognition of Plasmodium falciparum mature gametocyte-infected erythrocytes by antibodies of semi-immune adults and malaria-exposed children from Gabon. Malar J. 2017;16(1):111.
  60. 60. Pandey KR, Naik SR, Vakil BV. Probiotics, prebiotics and synbiotics- a review. J Food Sci Technol. 2015;52(12):7577–87. pmid:26604335
  61. 61. de Vrese M, Schrezenmeir J. Probiotics, prebiotics, and synbiotics. Adv Biochem Eng Biotechnol. 2008;111:1–66. pmid:18461293
  62. 62. Arumugam S, Lau CS, Chamberlain RS. Probiotics and Synbiotics Decrease Postoperative Sepsis in Elective Gastrointestinal Surgical Patients: a Meta-Analysis. J Gastrointest Surg. 2016;20(6):1123–31. pmid:27073082
  63. 63. Bo L, Li J, Tao T, Bai Y, Ye X, Hotchkiss RS, et al. Probiotics for preventing ventilator-associated pneumonia. Cochrane Database Syst Rev. 2014;10(10):CD009066. pmid:25344083
  64. 64. Lau VI, Cook DJ, Fowler R, Rochwerg B, Johnstone J, Lauzier F, et al. Economic evaluation alongside the Probiotics to Prevent Severe Pneumonia and Endotracheal Colonization Trial (E-PROSPECT): study protocol. BMJ Open. 2020;10(6):e036047. pmid:32595159
  65. 65. Martínez-Gómez F, Ixta-Rodríguez O, Aguilar-Figueroa B, Hernández-Cruz R, Monroy-Ostria A. Lactobacillus casei ssp. rhamnosus enhances non-specific protection against Plasmodium chabaudi AS in mice. Salud Publica Mex. 2006;48(6):498–503.
  66. 66. 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):246. pmid:34193269
  67. 67. Wittekamp BHJ, Oostdijk EAN, Cuthbertson BH, Brun-Buisson C, Bonten MJM. Selective decontamination of the digestive tract (SDD) in critically ill patients: a narrative review. Intensive Care Med. 2020;46(2):343–9. pmid:31820032
  68. 68. Zandstra DF, Van Saene HK. Selective decontamination of the digestive tract as infection prevention in the critically ill. A level 1 evidence-based strategy. Minerva Anestesiol. 2011;77(2):212–9. pmid:21102395
  69. 69. Smits LP, Bouter KE, de Vos WM, Borody TJ, Nieuwdorp M. Therapeutic potential of fecal microbiota transplantation. Gastroenterology. 2013;145(5):946–53. pmid:24018052
  70. 70. Han S, Shannahan S, Pellish R. Fecal Microbiota Transplant: Treatment Options for Clostridium difficile Infection in the Intensive Care Unit. J Intensive Care Med. 2016;31(9):577–86. pmid:26141116
  71. 71. Li Q, Wang C, Tang C, He Q, Zhao X, Li N, et al. Therapeutic modulation and reestablishment of the intestinal microbiota with fecal microbiota transplantation resolves sepsis and diarrhea in a patient. Am J Gastroenterol. 2014;109(11):1832–4. pmid:25373588
  72. 72. World Health Organisation. Severe malaria. Trop Med Int Health. 2014;19:7–131. pmid:25214480