Skip to main content
Advertisement
  • Loading metrics

Malnutrition disrupts adaptive immunity during visceral leishmaniasis by enhancing IL-10 production

  • Laís Amorim Sacramento,

    Roles Conceptualization, Formal analysis, Investigation, Methodology, Validation, Visualization, Writing – original draft, Writing – review & editing

    Affiliation Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Pennsylvania, United States of America

  • Claudia Gonzalez-Lombana,

    Roles Methodology

    Affiliation Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Pennsylvania, United States of America

  • Phillip Scott

    Roles Conceptualization, Data curation, Funding acquisition, Investigation, Methodology, Project administration, Resources, Supervision, Validation, Visualization, Writing – review & editing

    pscott@vet.upenn.edu

    Affiliation Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Pennsylvania, United States of America

Abstract

Protein-energy malnutrition (PEM) is a risk factor for developing visceral leishmaniasis (VL). While nutrient deficiency can impair immunity, its mechanistic impact on protective adaptive immune responses following Leishmania infection remains unknown. To determine the potential negative impacts of malnutrition on anti-parasitic responses in chronic VL, we provided mice with a polynutrient-deficient diet (deficient protein, energy, zinc, and iron) that mimics moderate human malnutrition. The polynutrient-deficient diet resulted in growth stunting and reduced mass of visceral organs and following infection with Leishmania infantum, malnourished-mice harbored more parasites in the spleen and liver. Malnourished and infected mice also had fewer T lymphocytes, with reduced T cell production of IFN-γ required for parasite clearance and enhanced production of the immunosuppressive cytokine, IL-10. To determine if IL-10 was causative in disease progression in the malnourished mice, we treated infected mice with monoclonal antibody α-IL-10R. α-IL-10R treatment reduced the parasite number in malnourished mice, restored the number of T cells producing IFN-γ, and enhanced hepatic granuloma formation. Our results indicate that malnutrition increases VL susceptibility due to defective IFN-γ-mediated immunity attributable to increased IL-10 production.

Author summary

Malnutrition contributes to the development of visceral leishmaniasis (VL) following Leishmania infection. Despite the clear association, the impact of malnutrition on the adaptive immune mechanisms required for parasite control is still unclear. We found that malnutrition disrupts the ability to control parasite replication in the spleen and liver due to defective IFN-γ-mediated immunity, reduced hepatic granuloma formation, and enhanced IL-10 production. Blocking IL-10R signaling restored the protective mechanisms to control parasite replication in the malnourished mice without interfering with the undernutrition state. Thus, we demonstrate that increased IL-10 production driven by malnutrition disrupts protective immunity against Leishmania, thereby enhancing the susceptibility of VL. Understanding the association between malnutrition and VL will provide insights into therapeutic approaches to treat this aggressive disease.

Introduction

Protein-energy malnutrition (PEM) is the most frequent type of malnutrition, affecting at least 800 million people worldwide, especially children and elderly individuals [1], and is the most common cause of immunodeficiency worldwide [2]. Deficiency in macronutrients and/or micronutrients induces impaired immunity and consequently increases susceptibility to various pathogens [35]. The risk of developing visceral leishmaniasis (VL) following infection with Leishmania parasites is increased by malnutrition [68]. Moreover, murine models of moderate PEM demonstrate that malnutrition causes early dissemination of Leishmania parasites to visceral organs due to defective innate immunity [912]. Despite substantial efforts to define the mechanisms between malnutrition and VL, the immunopathogenesis of this association is not completely understood. This study aimed to determine the impact of malnutrition on adaptive immunity during VL. We took advantage of a well-established model of polynutrient deficiency that mimics moderate human malnutrition [13] to fill this knowledge gap. We found that malnutrition increases the susceptibility to VL due to defective IFN-γ-mediated immunity and hepatic granuloma formation due to an aberrant IL-10 response.

Results

Malnutrition disrupts anti-parasitic immunity during visceral leishmaniasis

We first characterized the impact of the polynutrient-deficient diet (PND diet) on the nutritional status of our model by monitoring body weight, tail lengths, and the size of the spleen and liver, which are the target organs of VL. Mice fed the PND diet gained significantly less weight over time than mice fed the control diet, but there was no statistical difference between naïve and infected malnourished mice (Fig 1A and 1B). Consistent with previous reports, the growth curve observed in PND diet-fed mice was considered a moderate level of malnutrition in mice that mimics malnutrition in humans [13]. At the end of 6 weeks of feeding, mice on the PND diet had shorter tails (Fig 1C) and there was a significant reduction in the weight of the spleen (Fig 1D) and liver (Fig 1E). These data suggest that a PND diet led to growth stunting and reduced visceral organ size.

thumbnail
Fig 1. Malnutrition disrupts anti-parasitic immunity during visceral leishmaniasis.

(A and B) Body weight of naive or L. infantum-infected control diet or polynutrient deficient diet (PND diet) over time. (C) Final tail lengths at 6 weeks of diet and 4 weeks post-infection (wpi). (D) Spleen and (E) liver weight. Parasite number in the (F) spleen and (G) liver of control diet and PND diet-fed mice at 4 weeks post-infection. Absolute number of (H) CD3+ cells, (I) CD4+ T cells, (J) CD4+ T cells expressing T-bet, and (K) CD4+ T cells expressing IFNγ in the spleen of control and PND diet-fed mice. Cells were gated based on their characteristic size (FSC) and granularity (SSC), singlet cells, live+, and CD45+ cells. (L) IL-10 levels in the supernatants from splenocytes restimulated with L. infantum crude antigen or medium for 72 h at 4 wpi. (M) IL-10 levels in the liver homogenate supernatant at 4 wpi. The data are expressed as mean ± SEM and represent two experiments (n = 4 mice/group). The statistical significance was calculated by one-way ANOVA or Student’s t-test (*p < 0.05, **p < 0.01, ***p < 0.001 and ****p < 0.0001).

https://doi.org/10.1371/journal.ppat.1012716.g001

To test whether malnutrition impacts the control of parasite replication, we performed limiting dilution to quantify parasite number in the target organs of VL. Infected-malnourished mice harbored significantly more parasites at the spleen and liver at 4 weeks post-infection (wpi) (Fig 1F and 1G). An assessment of immune cells revealed a reduction in the number of CD3+ cells (Fig 1H) and CD4+ T cells (Fig 1I) in the spleen of infected-malnourished mice compared to infected-control. No significant differences were observed between non-infected malnourished versus control mice. Moreover, the development of Th1-type immunity critical for parasite control [14,15] appears to be compromised, as we found a reduced number of CD4+ T-bet+ T cells in the spleen of infected-malnourished mice compared with infected-control mice (Fig 1J). Consistently, the number of CD4+ T cells producing IFN-γ was significantly reduced in the spleen of infected-malnourished mice compared with infected-control mice (Fig 1K).

IL-10, a key immunosuppressive factor, contributes to the pathology of VL [1620]. Notably, IL-10 levels were significantly higher in the supernatant of cultured cells from infected-malnourished mice versus the infected-control mice (Fig 1L). Consistently, liver samples from infected-malnourished mice exhibited increased amounts of IL-10 (Fig 1M). The flow cytometry analysis demonstrates that IL-10 is produced by many cells during VL, especially by Th1 cells (Tr1), as previously reported [21,22]. Interestingly, malnutrition enhances the frequency of NK cells and monocytes producing IL-10 (S1 Fig). These data suggest that malnutrition results in increased IL-10 production, thereby compromising the development of a protective Th1 response and a consequent increase in parasite burden.

IL-10R blockade restores anti-parasitic immunity

IL-10 is a pleiotropic cytokine that can influence VL development through multiple mechanisms [1821]. To investigate the effect of IL-10 production in the infected malnourished mice, we treated mice with either α-IL-10R mAb to block IL-10 signaling or an α-IgG mAb as a control. Treatment was initiated at the time of infection and continued weekly. The treatment with α-IL-10R did not alter the body weight (Fig 2A and 2B) or tail length (Fig 2C) of malnourished mice. We observed a small increase in the spleen (Fig 2D) and liver (Fig 2E) weight of PND diet-fed mice treated with α-IL-10R compared with the control α-IgG mAb.

thumbnail
Fig 2. IL-10R blockade restores anti-parasitic immunity.

L. infantum-infected mice fed with control diet or PND diet were treated with anti-IgG control or anti-IL-10R antibody and euthanized at 4 wpi. Treatment was initiated at the time of infection and continued weekly until the termination of the experiment. (A and B) Body weight of infected mice treated with (A) α-IgG control or (B) α-IL-10R antibody. (C) Tail lengths at 6 weeks of diet and 4 weeks post-infection (wpi). (D) Spleen and (E) liver weight. Parasite number in the (F) spleen and (G) liver. Absolute number of (H) CD4+ T cells, (I) percentage and (J) number of CD4+ T cells expressing CD44, (K) number of CD4+ T cells expressing IFNγ at the spleen of control and PND diet-fed mice treated with anti-IgG control or anti-IL-10R antibody. (L-M) Percentage and number of CD11b+ cells expressing iNOS at the spleen of infected control and PND diet-fed mice treated with α-IgG control or α-IL-10R antibody. Cells were gated based on their characteristic size (FSC) and granularity (SSC), singlet cells, live+, and CD45+ cells. T cells were gated on CD3+, CD4+, and myeloid cells on CD11b+, CD11b+ iNOS+ cells. (N-O) Representative pictures of H&E-stained liver sections (20x magnification) and bar graph of the number of granulomas per field (counted in 10x magnification). The data are expressed as mean ± SEM and represent two experiments (n = 3–5 mice/group). The statistical significance was calculated by one-way ANOVA (*p < 0.05, **p < 0.01, ***p < 0.001 and ****p < 0.0001).

https://doi.org/10.1371/journal.ppat.1012716.g002

As previously reported, α-IL-10R treatment reduced the parasite number in the spleen and liver of well-nourished mice [21]. Consistently, IL-10 signaling blockade also reduced the parasite burden in the target organs of infected-malnourished mice (Fig 2F and 2G). However, while we did not observe a difference in CD4+ T cell numbers in the spleen of α-IL-10R mAb treated mice compared with α-IgG control mice (Fig 2H), we did find that α-IL-10R treatment increased the percentage and number of CD4+ CD44+ T cells (Fig 2I and 2J) and the number of CD4+ IFNγ+ T cells in the spleen of the infected-malnourished mice (Fig 2K). Thus, our data support a mechanistic model in which the enhanced IL-10 production in infected-malnourished mice prevents the generation of CD4+ IFNγ+ T cells required for protection.

In addition to a protective Th1 response, iNOS-mediated nitric oxide production is essential for controlling parasites within macrophages [1820]. Therefore, we next evaluated whether IL-10R blockade interferes with the expression of iNOS in malnourished infected mice. Notably, treatment with α-IL-10R mAb enhanced the percentage and number of CD11b+ cells expressing iNOS from the spleen of infected malnourished mice (Fig 2L and 2M), supporting a pleiotropic role for this immunosuppressive cytokine.

Finally, control of liver infection during experimental VL depends on granuloma formation [23]. We, therefore, evaluated the liver sections to determine whether the enhanced IL-10 production in the infected-malnourished mice interfered with hepatic granuloma generation. α-IL-10R mAb treatment increased the granuloma formation in the control mice, as previously reported [19]. Notably, infected-malnourished mice had a reduced number of hepatic granulomas compared to control mice. Importantly, the blockade of IL-10 signaling partially restored the number of granulomas in the liver of infected malnourished mice (Fig 2O). We measured cytokine levels in liver samples to investigate whether IL-10R blockade affects systemic inflammatory markers. Corroborating the previous report, malnourished mice have increased levels of IL-1β, IL-6, and TNF-α compared with control mice at baseline [24], and the α-IL-10R mAb treatment has no impact on the modulation of systemic cytokine levels (S2 Fig). Our results indicate that malnutrition increases VL susceptibility due to defective activation and expansion of CD4+ IFNγ+ T cells, decreased iNOS expression, and reduced hepatic granuloma formation, which is attributable to increased IL-10 production.

Discussion

Malnutrition disrupts innate immune mechanisms, causes early visceralization of parasites during VL [9,11,13], and can affect T cell-mediated responses and granuloma formation [2527]. While increased IL-10 expression is also associated with VL progression in humans [17], the potential connection between these observations has remained unclear. Here, we found that malnutrition increases VL susceptibility due to an elevation in IL-10 production, which limits IFNγ-mediated immunity, iNOS production, and hepatic granuloma formation. Moreover, we found that α-IL-10R treatment restored activation and expansion of T cells producing IFNγ in the malnourished-infected mice without altering the undernutrition observed. The restoration of iNOS expression following α-IL-10R treatment could be either a consequence of enhanced CD4+ IFNγ+ T cells or a direct effect on the myeloid cells [17,28].

In agreement with our data, high levels of systemic IL-10 have been reported in malnourished mice challenged intraperitoneally with bacterial LPS [2931]. The observed increase in circulating levels of inflammatory cytokines in malnourished individuals [32,33] and murine models of malnutrition [24] has been attributed to a dysfunctional intestinal barrier and bacterial translocation [24]. Based on these findings, we hypothesize that the increased IL-10 production observed in the malnourished model results from an excessive systemic inflammatory process. In sepsis, high levels of IL-10 are critical for controlling inflammatory responses that can cause tissue damage [34,35]. In support, VL patients have a systemic overproduction of inflammatory mediators [36,37], and the high levels of IL-10 limit tissue damage in this disease. However, the tissue-preserving capacity of IL-10 is counterbalanced by its ability to generate a permissive environment for parasite replication [3840][16]. Additionally, malnutrition disturbs circulating metabolite levels, enhancing tryptophan catabolites such as kynurenine [41]. Given the influence of tryptophan catabolism on immunosuppression, the high level of IL-10 might result from kynurenine axis stimulation [42,43]. Specifically, our findings indicate that the enhanced IL-10 levels in malnutrition further contribute to VL by disrupting Th1 immunity and hepatic granuloma formation, thereby limiting parasite clearance. Further mechanistic dissection of this pathway may reveal opportunities to enhance the protective immune response without exacerbating tissue pathology.

Material and methods

Ethics statement

This study followed the Guide for the Care and Use of Laboratory Animals (National Institutes of Health). The protocol was approved by the Institutional Animal Care and Use Committee, University of Pennsylvania Animal Welfare Assurance Number 806959.

Mice and diets

4-week-old female C57BL/6 mice were purchased from Charles River. Mice were grouped in cages of 4–5 and were maintained in a specific pathogen–free facility with free access to water, nesting material, and housed at a temperature of 21°C at the University of Pennsylvania Animal Care Facilities. Mice received a control standard chow (17% protein, 100 ppm Iron, 30 ppm zinc—Teklad diet, #99103) or an isocaloric but protein, iron, and zinc-deficient diet (3% protein, 10 ppm Iron, 1 ppm zinc—Teklad diet, #99075). After 2 weeks of diet, mice were divided into two subgroups, one of which was infected with L. infantum. Polynutrient deficient diet-fed (PND diet) mice received 90% of the consumed food of the control mice over weeks calculated based on the food ingestion (g/mouse/day) of the control diet group obtained every 72 hours. The body weight of mice was monitored once a week. All animals were used in accordance with the recommendations in the Guide for the Care and Use of Laboratory Animals of the National Institutes of Health and the guidelines of the University of Pennsylvania Institutional Animal Use and Care Committee. The protocol was approved by the Institutional Animal Care and Use Committee, University of Pennsylvania Animal Welfare Assurance.

Parasite culture and infection

L. infantum (LLM-320) was cultured in Schneider medium with 20% heat-inactivated fetal bovine serum, 5% penicillin, and streptomycin. The mice were intravenously infected via the retro-orbital plexus with 107 L. infantum metacyclic enriched promastigotes in 100 μL of PBS. A quantitative limiting dilution assay determined parasite burdens.

In vivo α-IL-10R antibody treatment

Mice were treated intraperitoneally twice a week with 500 μg of α-mouse IL-10R antibody or rat anti-IgG1 isotype control. Treatment was initiated at the time of infection and continued weekly until the termination of the experiment at 4 weeks post-infection.

Statistical analysis

Data are shown as means ± SEM. Statistical significance was determined using the two-tailed unpaired Student’s t-test or one-way ANOVA. All statistical analysis was calculated using GraphPad Prism Software. Differences were considered significant when *p < 0.05, **p ≤ 0.01, ***p ≤ 0.001, ****p < .0001.

Supporting information

S1 Fig. IL-10 production by lymphoid and myeloid populations in malnourished mice.

The percentage of IL-10 in cells from the spleen and liver of naive or L. infantum-infected control diet or polynutrient deficient diet (PND diet) at 6 weeks of diet and 4 weeks post-infection. Leukocytes from the spleen and liver were stimulated with PMA, ionomycin, and monensin in the presence of BFA for 2.5 hr and subsequently harvested and stained for IL-10. Cells were gated based on size (FSC) and granularity (SSC), singlet cells, live+, and CD45+ cells. For NK and NKT cell identification, the NK1.1+ subset was identified as CD3-NK1.1+ and CD3+NK1.1+, respectively. NK1.1- subset was gated on CD3+ subset for subsequent identification of CD4+IFNγ+, CD4+ Foxp3+, and CD8+ cells. CD3- subset was followed by gate on CD19+ subset for B cell. For myeloid population identification, the CD19- subset was followed by the gate on CD11c+Ly6C- (dendritic cells), CD11c-Ly6C+CD11b+ (monocytes), and CD11c-Ly6C-F4/80+ (macrophages). The statistical significance was calculated by one-way ANOVA (*p < 0.05, **p < 0.01, and ***p < 0.001).

https://doi.org/10.1371/journal.ppat.1012716.s001

(DOCX)

S2 Fig. IL-10R blockade has no impact on systemic inflammation.

Levels of IL-1β, IL-6, and TNFα in supernatants from a liver homogenate at 6 weeks of diet and 4 weeks post-infection. The data are expressed as mean ± SEM and represent the combination of two experiments. The statistical significance was calculated by one-way ANOVA (*p < 0.05, **p < 0.01, and ***p < 0.001).

https://doi.org/10.1371/journal.ppat.1012716.s002

(DOCX)

Acknowledgments

We thank the Comparative Pathology Core (CPC).

References

  1. 1. Zhang X, Zhang L, Pu Y, Sun M, Zhao Y, Zhang D, et al. Global, Regional, and National Burden of Protein-Energy Malnutrition: A Systematic Analysis for the Global Burden of Disease Study. Nutrients. 2022;14. pmid:35807772
  2. 2. Bhargava A. Undernutrition, nutritionally acquired immunodeficiency, and tuberculosis control. BMJ. 2016;355: i5407. pmid:27733343
  3. 3. Keusch GT. The history of nutrition: malnutrition, infection and immunity. J Nutr. 2003;133: 336S–340S. pmid:12514322
  4. 4. Sukhina A, Queriault C, Hall E, Rome K, Aggarwal M, Nunn E, et al. Malnutrition drives infection susceptibility and dysregulated myelopoiesis that persists after refeeding intervention. BioRxiv. 2024. pmid:39229137
  5. 5. Cunningham-Rundles S, McNeeley DF, Moon A. Mechanisms of nutrient modulation of the immune response. J Allergy Clin Immunol. 2005;115: 1119–28; quiz 1129. pmid:15940121
  6. 6. Badaró R, Jones TC, Lorenço R, Cerf BJ, Sampaio D, Carvalho EM, et al. A prospective study of visceral leishmaniasis in an endemic area of Brazil. J Infect Dis. 1986;154: 639–649. pmid:3745974
  7. 7. Cerf BJ, Jones TC, Badaro R, Sampaio D, Teixeira R, Johnson WD. Malnutrition as a risk factor for severe visceral leishmaniasis. J Infect Dis. 1987;156: 1030–1033. pmid:3680989
  8. 8. Cuervo-Escobar S, Losada-Barragán M, Umaña-Pérez A, Porrozzi R, Saboia-Vahia L, Miranda LHM, et al. T-cell populations and cytokine expression are impaired in thymus and spleen of protein malnourished BALB/c mice infected with Leishmania infantum. PLoS ONE. 2014;9: e114584. pmid:25535967
  9. 9. Ibrahim MK, Barnes JL, Anstead GM, Jimenez F, Travi BL, Peniche AG, et al. The malnutrition-related increase in early visceralization of Leishmania donovani is associated with a reduced number of lymph node phagocytes and altered conduit system flow. PLoS Negl Trop Dis. 2013;7: e2329. pmid:23967356
  10. 10. Ibrahim MK, Barnes JL, Osorio EY, Anstead GM, Jimenez F, Osterholzer JJ, et al. Deficiency of lymph node-resident dendritic cells (DCs) and dysregulation of DC chemoattractants in a malnourished mouse model of Leishmania donovani infection. Infect Immun. 2014;82: 3098–3112. pmid:24818662
  11. 11. Anstead GM, Chandrasekar B, Zhang Q, Melby PC. Multinutrient undernutrition dysregulates the resident macrophage proinflammatory cytokine network, nuclear factor-kappaB activation, and nitric oxide production. J Leukoc Biol. 2003;74: 982–991. pmid:12960263
  12. 12. Osorio EY, Uscanga-Palomeque A, Patterson GE, Cordova E, Travi BL, Soong L, et al. Malnutrition-related parasite dissemination from the skin in visceral leishmaniasis is driven by PGE2-mediated amplification of CCR7-related trafficking of infected inflammatory monocytes. PLoS Negl Trop Dis. 2023;17: e0011040. pmid:36630476
  13. 13. Anstead GM, Chandrasekar B, Zhao W, Yang J, Perez LE, Melby PC. Malnutrition alters the innate immune response and increases early visceralization following Leishmania donovani infection. Infect Immun. 2001;69: 4709–4718. pmid:11447142
  14. 14. Squires KE, Schreiber RD, McElrath MJ, Rubin BY, Anderson SL, Murray HW. Experimental visceral leishmaniasis: role of endogenous IFN-gamma in host defense and tissue granulomatous response. J Immunol. 1989;143: 4244–4249. pmid:2512353
  15. 15. Murray HW, Rubin BY, Rothermel CD. Killing of intracellular Leishmania donovani by lymphokine-stimulated human mononuclear phagocytes. Evidence that interferon-gamma is the activating lymphokine. J Clin Invest. 1983;72: 1506–1510. pmid:6415111
  16. 16. Nylén S, Sacks D. Interleukin-10 and the pathogenesis of human visceral leishmaniasis. Trends Immunol. 2007;28: 378–384. pmid:17689290
  17. 17. Nylén S, Maurya R, Eidsmo L, Manandhar KD, Sundar S, Sacks D. Splenic accumulation of IL-10 mRNA in T cells distinct from CD4+CD25+ (Foxp3) regulatory T cells in human visceral leishmaniasis. J Exp Med. 2007;204: 805–817. pmid:17389235
  18. 18. Gautam S, Kumar R, Maurya R, Nylén S, Ansari N, Rai M, et al. IL-10 neutralization promotes parasite clearance in splenic aspirate cells from patients with visceral leishmaniasis. J Infect Dis. 2011;204: 1134–1137. pmid:21881130
  19. 19. Murray HW, Lu CM, Mauze S, Freeman S, Moreira AL, Kaplan G, et al. Interleukin-10 (IL-10) in experimental visceral leishmaniasis and IL-10 receptor blockade as immunotherapy. Infect Immun. 2002;70: 6284–6293. pmid:12379707
  20. 20. Murray HW, Moreira AL, Lu CM, DeVecchio JL, Matsuhashi M, Ma X, et al. Determinants of response to interleukin-10 receptor blockade immunotherapy in experimental visceral leishmaniasis. J Infect Dis. 2003;188: 458–464. pmid:12870129
  21. 21. Bunn PT, Montes de Oca M, de Labastida Rivera F, Kumar R, Ng SS, Edwards CL, et al. Distinct Roles for CD4+ Foxp3+ Regulatory T Cells and IL-10-Mediated Immunoregulatory Mechanisms during Experimental Visceral Leishmaniasis Caused by Leishmania donovani. J Immunol. 2018;201: 3362–3372. pmid:30355785
  22. 22. Edwards CL, Engel JA, de Labastida Rivera F, Ng SS, Corvino D, Montes de Oca M, et al. A molecular signature for IL-10-producing Th1 cells in protozoan parasitic diseases. JCI Insight. 2023;8. pmid:37917177
  23. 23. Stanley AC, Engwerda CR. Balancing immunity and pathology in visceral leishmaniasis. Immunol Cell Biol. 2007;85: 138–147. pmid:17146466
  24. 24. Patterson GT, Osorio EY, Peniche A, Dann SM, Cordova E, Preidis GA, et al. Pathologic inflammation in malnutrition is driven by proinflammatory intestinal microbiota, large intestine barrier dysfunction, and translocation of bacterial lipopolysaccharide. Front Immunol. 2022;13: 846155. pmid:35720380
  25. 25. Maciel PS, Gonçalves R, Antonelli LR do V, Fonseca CT. Schistosoma mansoni Infection Is Impacted by Malnutrition. Front Microbiol. 2021;12: 635843. pmid:33815321
  26. 26. Daumas A, Coiffard B, Chartier C, Ben Amara A, Alingrin J, Villani P, et al. Defective granuloma formation in elderly infected patients. Front Cell Infect Microbiol. 2020;10: 189. pmid:32411623
  27. 27. Chan J, Tian Y, Tanaka KE, Tsang MS, Yu K, Salgame P, et al. Effects of protein calorie malnutrition on tuberculosis in mice. Proc Natl Acad Sci USA. 1996;93: 14857–14861. pmid:8962145
  28. 28. Carvalho EM, Bacellar O, Brownell C, Regis T, Coffman RL, Reed SG. Restoration of IFN-gamma production and lymphocyte proliferation in visceral leishmaniasis. J Immunol. 1994;152: 5949–5956. pmid:8207220
  29. 29. Santos ACA, Correia CA, de Oliveira DC, Nogueira-Pedro A, Borelli P, Fock RA. Intravenous Glutamine Administration Modulates TNF-α/IL-10 Ratio and Attenuates NFkB Phosphorylation in a Protein Malnutrition Model. Inflammation. 2016;39: 1883–1891. pmid:27565164
  30. 30. Mello AS, de Oliveira DC, Bizzarro B, Sá-Nunes A, Hastreiter AA, Beltran JS de O, et al. Protein malnutrition alters spleen cell proliferation and IL-2 and IL-10 production by affecting the STAT-1 and STAT-3 balance. Inflammation. 2014;37: 2125–2138. pmid:24986442
  31. 31. Fock RA, Vinolo MAR, Crisma AR, Nakajima K, Rogero MM, Borelli P. Protein-energy malnutrition modifies the production of interleukin-10 in response to lipopolysaccharide (LPS) in a murine model. J Nutr Sci Vitaminol. 2008;54: 371–377. pmid:19001768
  32. 32. Takele Y, Adem E, Getahun M, Tajebe F, Kiflie A, Hailu A, et al. Malnutrition in healthy individuals results in increased mixed cytokine profiles, altered neutrophil subsets and function. PLoS ONE. 2016;11: e0157919. pmid:27548305
  33. 33. Patterson GT, Manthi D, Osuna F, Muia A, Olack B, Mbuchi M, et al. Environmental, metabolic, and inflammatory factors converge in the pathogenesis of moderate acute malnutrition in children: an observational cohort study. Am J Trop Med Hyg. 2021;104: 1877–1888. pmid:33755580
  34. 34. Marchant A, Bruyns C, Vandenabeele P, Abramowicz D, Gérard C, Delvaux A, et al. The protective role of interleukin-10 in endotoxin shock. Prog Clin Biol Res. 1994;388: 417–423. pmid:7831374
  35. 35. Steinhauser ML, Hogaboam CM, Kunkel SL, Lukacs NW, Strieter RM, Standiford TJ. IL-10 is a major mediator of sepsis-induced impairment in lung antibacterial host defense. J Immunol. 1999;162: 392–399. pmid:9886412
  36. 36. Ansari NA, Saluja S, Salotra P. Elevated levels of interferon-gamma, interleukin-10, and interleukin-6 during active disease in Indian kala azar. Clin Immunol. 2006;119: 339–345. pmid:16540374
  37. 37. Kurkjian KM, Mahmutovic AJ, Kellar KL, Haque R, Bern C, Secor WE. Multiplex analysis of circulating cytokines in the sera of patients with different clinical forms of visceral leishmaniasis. Cytometry A. 2006;69: 353–358. pmid:16604536
  38. 38. Ghalib HW, Piuvezam MR, Skeiky YA, Siddig M, Hashim FA, el-Hassan AM, et al. Interleukin 10 production correlates with pathology in human Leishmania donovani infections. J Clin Invest. 1993;92: 324–329. pmid:8326000
  39. 39. Mege J-L, Meghari S, Honstettre A, Capo C, Raoult D. The two faces of interleukin 10 in human infectious diseases. Lancet Infect Dis. 2006;6: 557–569. pmid:16931407
  40. 40. Moore KW, de Waal Malefyt R, Coffman RL, O’Garra A. Interleukin-10 and the interleukin-10 receptor. Annu Rev Immunol. 2001;19: 683–765. pmid:11244051
  41. 41. Mayneris-Perxachs J, Lima AAM, Guerrant RL, Leite ÁM, Moura AF, Lima NL, et al. Urinary N-methylnicotinamide and β-aminoisobutyric acid predict catch-up growth in undernourished Brazilian children. Sci Rep. 2016;6: 19780. pmid:26816084
  42. 42. Seo S-K, Kwon B. Immune regulation through tryptophan metabolism. Exp Mol Med. 2023;55: 1371–1379. pmid:37394584
  43. 43. Lanis JM, Alexeev EE, Curtis VF, Kitzenberg DA, Kao DJ, Battista KD, et al. Tryptophan metabolite activation of the aryl hydrocarbon receptor regulates IL-10 receptor expression on intestinal epithelia. Mucosal Immunol. 2017;10: 1133–1144. pmid:28098246