Abstract
Trypanosomiasis is a parasitic disease, affecting both humans and animals, in the form of Human African Trypanosomiasis and Nagana disease, respectively. Anemia is one of the most common symptoms of trypanosomiasis, and if left unchecked, can cause severe complications and even death. Several factors have been associated with the development of this anemia, including dysregulation of iron homeostasis, but little is known about the molecular mechanisms involved. Here, using murine models, we study the involvement of hepcidin, the key regulator of iron metabolism and an important player in the development of anemia of inflammation. Our data shows two stages for the progression of anemia, to which hepcidin contributes: a first stage, when anemia develops, with a likely cytokine-mediated stimulation of hepcidin and subsequent limitation in iron availability and erythropoiesis, and a second stage, of recovery, where hepcidin elevation declines due to the decreased inflammatory signal and increased production of erythroid regulators by the kidney, spleen and bone marrow, thus leading to an increase in iron release and availability and enhanced erythropoiesis. In agreement, in hepcidin knockout mice, anaemia is much milder and its recovery is complete, in contrast to wild-type animals which do not fully recover from anaemia after 21 days. Aside from all other factors known to be involved in the development of anemia during trypanosomiasis, there clearly is an important contribution of hepcidin for both its development and recovery.Introduction
African trypanosomes are extracellular protozoan parasites transmitted by the hematophagous tsetse fly (Glossina spp), and are responsible for debilitating medical and veterinary diseases in sub-Saharan Africa.1 Trypanosoma brucei rhodesiense and Trypanosoma brucei gambiense infect humans and are responsible for the fatal Human African Trypanosomiasis, also known as sleeping sickness.2 Trypanosoma brucei brucei is responsible for animal trypanosomiasis, which mostly affects cattle.2
One of the most common complications of trypanosomiasis is anemia, both in humans and animals,3-5 which in conjunction with other symptoms can be a major cause of death if left untreated, particularly in livestock.6 Over the years, several causes for this anemia have been described, and these include both parasite and host-associated factors.7-9 Erythrophagocytosis by activated liver and spleen myeloid cells has been identified as a major contributor to erythrocyte clearance. In addition, the lipid composition of erythrocytes is altered during trypanosome infection and these are preferentially phagocytosed.8
Among the parasite factors that contribute to anemia are the expression of extracellular products, such as hemolysins,10,11 direct mechanical erythrocyte injury,12 lipid peroxidation,13-15 and extracellular vesicles that can fuse with erythrocytes resulting in rapid clearance and anemia.16 Furthermore, although there are clear indications that iron metabolism has a significant role in the establishment of anemia during trypanosomal infections,17,18 the overall molecular mechanisms that lead to it are still poorly understood, and this includes, in particular, the involvement of hepcidin.
Hepcidin is a small antimicrobial peptide and a key regulator of iron metabolism.19-21 During infectious/inflammatory processes, hepcidin leads to a systemic decrease in iron mobilization by blocking iron release from hepatocytes, enterocytes and macrophages. This impacts the proliferation of the pathogens but also affects the host by impairing erythropoiesis. The scarcity of iron and the subsequent impairment of erythropoiesis are thought to lead to a condition known as anemia of inflammation. This mechanism of response has been established for several bacterial infections22-26 and some intracellular parasites.27,28 However, studies of hepcidin involvement in the development of anemia in infections with extracellular parasites are extremely limited.
The present study was undertaken to determine the possible role of hepcidin in the regulation of iron metabolism during trypanosomal infections and its contribution to the onset, development and recovery from anemia.
Methods
Mice, parasites and infections
Five-week-old C57BL/6 and BALB/c female mice were purchased from Charles River Laboratories (Saint-Germain-Nuelles, France). Female hepcidin knockout (Hamp-/-) mice29 were bred at the institute facilities. The Trypanosoma brucei brucei, GVR35 strain, was used to infect the mice. All experiments were carried out in accordance with the IBMC.INEB Animal Ethics Committees and the Portuguese National Authorities for Animal Health guidelines according to the statements on the directive 2010/63/EU of the European Parliament and Council.
Hematologic and serum parameters, tissue iron content
Hematologic and serum parameters were blindly determined by a certified laboratory (CoreLab, Centro Hospitalar do Porto, Portugal). Liver and spleen iron content was evaluated by the bathophenanthroline method30 and Perls staining.
Cytokine profile analysis
Cytokine levels were measured in the serum using the BD CBA Mouse Inflammation Kit (BD Biosciences, San Jose, CA, USA).
Flow cytometry
Bone marrow (BM) cells were stained with anti-CD3e (17A2), anti-CD19 (6D5), anti-TER119, anti-CD71 (RI7217), anti-CD11c (N418), and anti-CD11b (M1/70) antibodies, and run in a BD FACSCanto II Flow Cytometer (BD Biosciences). Data were analyzed with FlowJo software (FlowJo LCC, Ashland, OR, USA).
RNA isolation and cDNA synthesis
Total RNA was isolated from tissues and cells with the PureLink RNA Mini Kit (Thermo Fisher Scientific) and converted to cDNA using the NZY First-Strand cDNA Synthesis Kit (NZYTech, Lisbon, Portugal).
Analysis of gene expression by quantitative-polymerase chain reaction
Relative levels of several genes mRNAs were quantified in relevant organs of control and infected animals, by quantitative-polymerase chain reaction (qPCR). The comparative CT method (2-ΔΔCT method) was used to analyze gene expression levels.
Analysis of ferroportin levels by Western blot
Levels of FPN1 protein were evaluated in the liver, spleen and duodenum of C57BL/6 and Hamp-/- mice by Western blot, with GAPDH being used as housekeeping protein. Primary antibodies used were rabbit anti-Ferroportin/SLC40A1 (Novus, Littletown, CO, USA; catalog #NPB1-21502), (1:1000) 1 hour (h) RT, rabbit anti-GAPDH (Abcam, Cambridge, UK, catalog #EPR16891), (1:1000) 1 h RT.
Statistical analysis
Statistical analysis was carried out using GraphPad Prism 8 (GraphPad Software Inc., La Jolla, CA, USA). Multiple comparisons were performed with one-way ANOVA and post hoc Student Newman-Keuls test. P<0.05 was considered statistically significant.
Further details of the study methods are available in the Online Supplementary Appendix.
Results
T.b. brucei infection in mice leads to macrocytic anemia, decreased erythropoietic activity and iron redistribution
The course of the infection with T.b. brucei GVR35 strain expressing luciferase was followed in BALB/c mice by bioluminescence imaging and counting the parasites in the blood (Online Supplementary Figure S1). A significant reduction in the number of red blood cells (RBC), reticulocytes, hematocrit and hemoglobin levels was observed up to day 7, with a gradual return to normal levels (Figure 1A-D), indicating an early onset of acute anemia, followed by a later recovery. A significant increase in the mean corpuscular volume (MCV) was also observed, indicative of macrocytic anemia (Figure 1E). Alterations in the development of erythroid lineage in the BM were evaluated by flow cytometry. Overall, a decreased total number of mature and developing erythrocytes was observed (Figure 1F), as well as decreased numbers of pro-erythroblasts (Figure 1G), basophilic erythroblasts (Figure 1H), and polychromatic erythroblasts (Figure 1, panel I).
Trypanosomal infection also caused a significant decrease in circulating serum iron levels (Figure 1J), transferrin saturation (Figure 1K), and increased total iron binding capacity (TIBC) (Figure 1L) and serum ferritin (Figure 1M), highlighting the inflammatory status of the animals and thus, indirectly, a condition of anemia of inflammation. The lower systemic iron concentration was accompanied by the accumulation of iron in the liver (Figure 1N) and spleen (Figure 1, panel O).
Circulating cytokine levels indicate the development of an acute infection
We evaluated the impact of T.b. brucei infection in the expression of several inflammatory cytokines. IL-6 levels were elevated as early as 1 day post infection and remained high up to day 7, with a gradual recovery to control levels (Figure 2A). IL-6 not only acts as a pro-inflammatory cytokine, inducing the expression of other cytokines and several acute phase proteins, but is also an important hepcidin inducer, thus potentially impacting iron metabolism during infection. Circulating levels of IFNg, TNF-a and IL-10 (Figure 2B-D) followed a similar pattern, with an increase up to day 4 followed by a decrease throughout the course of the experiment, with both IFN-g******and IL-10 returning to normal levels at 21 days post infection. Additionally, high levels of MCP-1 were observed as early as day 1 post infection, remaining high at day 4, and rapidly decreasing to almost normal levels (Figure 2E).
Evaluation of gene expression indicates an early onset of acute anemia and later recovery
Expression of hepcidin and other iron-related and hematopoietic genes was evaluated in the liver, spleen, kidney and BM. In the liver, a gradual increase in Hamp1 expression was observed up to day 7 post infection, followed by a decrease, reaching lower than control levels at day 21 (Figure 3A). Ferroportin (Fpn1) expression was significantly down-regulated as early as day 1, with a gradual recovery to control levels followed by an incremental upregulation up to day 21 post infection (Figure 3B). Transferrin (Tf) (Figure 3C) and ferritin H (Fth1) (Figure 3D) were also significantly up-regulated at the early stages of infection, returning to control levels before day 14, coinciding with the increases in TIBC and circulating ferritin levels, respectively (Figure 1L and M).
In the spleen, Hamp1 was observed to be up-regulated in the earlier days of infection, returning to control levels at day 7 and with a slight decrease at day 21 (Figure 3E). Expression levels of Fth1 started to increase at day 4, peaked at day 7, and then gradually decreased towards the end of the experiment (Figure 3G). Similar patterns of upregulation were observed for Fpn1, Hbb, Epor, Erfe and Twsg1 (Figure 3F, H-K), with significant increases in expression starting as early as day 4, and maintained up to day 21. In the kidney, an increase in Hamp1 expression was observed starting day 1, peaking at day 4, and returning to control levels at day 7 (Figure 3L). Levels of Epo were up-regulated throughout the duration of the experiment (Figure 3M).
Finally, in the BM, Hamp1 expression was found to be increased as early as day 1, followed by a gradual downregulation up to day 7, and a recovery to normal values towards day 21 (Figure 3N). For Hbb, Epor and Erfe, a similar pattern of expression was observed, with decreased expression at day 7, followed by gradual increases, and reaching maximum levels at day 21 (Figure 3, panels O-Q). Gdf15 and Twsg1 expressions gradually increased throughout the course of the infection, reaching maximum levels at day 21 (Figure 3R and S).
Hepcidin contributes to the development of anemia in trypanosomal infections
Gene expression profiles indicate that hepcidin might be involved in the development of anemia during trypanosomal infection. In order to investigate this, we performed experimental infections in hepcidin deficient (Hamp-/-) mice. No significant differences were found in the parasitemias or total parasite burdens between Hamp-/- and C57BL/6 mice (Online Supplementary Figure S2). Hematologic parameters show development of anemia, with a steady decline of RBC number, hematocrit and hemoglobin levels up to day 7 post infection, followed by a gradual recovery (Figure 4AD). However, whereas in C57BL/6 mice parameters never fully recover to normal values, in Hamp-/-- mice there is a complete reversal of anemia and a return to normal conditions, indicating a more severe anemia was established in the wild-type animals (Figure 4A, C and D). A significant increase in reticulocyte numbers and MCV was also observed at later stages of infection in both wild-type and knockout mice (Figure 4B and E). Variations in serological parameters and tissue iron content in C57BL/6 mice were mostly comparable to BALB/c, with decreases in serum iron levels and transferrin saturation (Figure 4F and G), increases in TIBC (Figure 4H), as well as in liver and spleen iron (Figure 4J and K, and Online Supplementary Figure S3) with no changes in ferritin levels (Figure 4, panel I). In Hamp-/- mice, only a similar increase in TIBC (Figure 4H) was observed, as well as increases in liver and spleen iron (Figure 4J and K) with no changes in transferrin saturation (Figure 4G). Furthermore, despite the higher levels of serum iron and circulating ferritin, when compared with C57BL/6, increases in both parameters were also observed in the early days of infection, with recoveries to near normal towards the end (Figure 4F and I).
Circulating cytokine levels in both C57BL/6 and Hamp-/- mice were mostly comparable to BALB/c. IL-6 levels increased as early as day 1 post infection and remained high up to day 7 or day 14 (in C57BL/6 and Hamp-/-) with a subsequent gradual recovery to control levels (Figure 5A). Circulating levels of IFN-g, TNF-a and IL-10 (Figure 5B-D) also followed similar patterns, with an increase up to day 4 followed by a decrease. Additionally, high levels of MCP1 were observed as early as day 1 post infection, reaching maximum levels at day 4 and rapidly decreasing to normal levels at day 7 post infection (Figure 5E).
Gene expression was also evaluated in the liver, spleen and kidney. Hepcidin and ferroportin liver expression profiles in C57BL/6 mice were similar to what was observed for BALB/c, with an increase of hepcidin up to day 7 followed by a decrease to lower than normal levels, and with ferroportin mirroring hepcidin (Figure 6A and B), coinciding with the early onset of anemia and the subsequent recovery. No discernible hepcidin expression was observed in the liver of Hamp-/- animals, and the increase in ferroportin expression was maintained throughout the duration of the experiment (Figure 6A and B). In the spleen, an increase in Hbb (Figure 6C) and Twsg1 (Figure 6E) was observed in both wild-type and knockout mice. Increases in the expression of Epo were also observed in the kidney of both C57BL/6 and Hamp- /- infected mice, remaining elevated throughout the infection in C57BL/6 mice, but decreasing at day 21 in Hamp- /- mice (Figure 6F). Despite the drive for enhanced erythropoiesis being present in both wild-type and knockout mice, Erfe was only up-regulated in C57BL/6 mice, similar to BALB/c, but no changes in expression were observed in Hamp-/- mice (Figure 6D). Ferroportin protein levels were also measured in the liver, spleen and duodenum of C57BL/6 and Hamp-/-. In the liver, variations in protein levels closely matched the variations in mRNA expression, with significant decreases in both C57BL/6 and Hamp-/- mice at days 1 and 4 followed by gradual recoveries towards day 21, although earlier and stronger in Hamp-/- (Figure 7A). In the spleen, ferroportin levels gradually decreased until they were no longer detected up to day 4, followed by a recovery up to day 21, but starting earlier and reaching higher levels in Hamp-/-, where ferroportin could already be observed at day 7, with much higher levels at day 21 (Figure 7B). No significant changes in ferroportin protein levels were observed in the duodenum (Figure 7C).
Discussion
Using a mouse model of trypanosomiasis (which has been shown to replicate the two stages of the disease31 occurring in humans) we performed an integrated analysis on the establishment/progression of anemia and the molecular mechanisms involved in iron homeostasis. Furthermore, this study also evaluates the contribution of hepcidin (the key regulator of iron homeostasis19,32) to the anemia established during T.b. brucei infection. Different degrees of anemia severity in trypanosomal infections have been reported and these have been described as being dependent on the host and sub-species causing the infection.3,7 In this study, an early decrease in several hematologic parameters such as the number of RBC and reticulocytes, hematocrit and hemoglobin levels, and an increase in MCV were seen in wild-type (BALB/c and C57BL/6) and in Hamp-/- (gene deficient in C57BL/6 background) mice. This was followed later by an increase in most parameters, never reaching normal levels in wildtype (BALB/c and C57BL/6) animals but fully recovering in Hamp-/- mice.
An in-depth investigation of several erythrocyte populations in the BM of BALB/c mice shows similar patterns in all of them, from the early immature pro-erythroblasts to the late mature polychromatic erythroblasts, most reaching the lowest numbers after 7 days of infection, and again followed by gradual recoveries. This was also accompanied by late increases in the expression of hemoglobin, in both the spleen and BM.
Iron is essential not only for the host but also for pathogen proliferation, and trypanosome infections are no exception.17,18 In the case of extracellular, blood circulating pathogens, iron is rapidly removed from circulation to prevent pathogens from accessing it. This is seen in BALB/c and C57BL/6 mice by the expression profiles of transferrin and ferritin, particularly in the liver, where early increases in both would facilitate iron retention and removal from circulation, as well as by decreases in circulating serum iron and transferrin saturation, and an increase in circulating ferritin.33 This response might be particularly important in the earliest days of infection, since T. brucei bloodstream forms can acquire iron through pathogen-specific receptors for transferrin and haptoglobin-hemoglobin complexes.34 On the other hand, the higher levels of serum iron and circulating ferritin levels observed in Hamp-/- mice do not appear to influence pathogen proliferation, when compared with the wild-type animals, with parasite levels remaining similar. Although limited data are available regarding extracellular and intracellular parasites, they can both benefit or be suppressed by high iron levels.27,28 This is in contrast with many bacterial infections, where hepcidin is known to have a crucial role in creating a hypoferrimic state, to limit iron availability, and protect the host both against infections caused by siderophilic bacteria23,35 and by others.36 Additionally, there is an increase in iron storage both in the liver, the major organ for iron accumulation, and the spleen, where iron recycling from senescent erythrocytes occurs. However, this redistribution of iron with the goal of limiting its mobilization and availability to pathogens is actually a double-edged sword; at the same time potentially limiting iron availability for erythropoiesis in the BM and leading to the condition known as anemia of inflammation,37,38 thus contributing to the overall trypanosome-related anemia.
On the inflammatory side, it has been well documented that the response to infectious/inflammatory stimuli involves the expression of numerous pro- and anti-inflammatory cytokines that have various effects on different leukocyte populations, from lymphocytes to macrophages, with the latter also being involved in the modulation of iron homeostasis.We evaluated the levels of some relevant circulating inflammatory cytokines, where we observed a strong type I cytokine response in all models, with increases in the levels of IL-6, IFN-g and TNF-a. IL-6, which is mostly produced by macrophages but also by Th2 T cells in response to the extracellular parasites, is a major inducer of hepcidin expression by the liver during inflammatory processes.37,40,41 In trypanosomiasis, increased levels of IFN-g can inhibit BM proliferation and suppress erythropoiesis,42 whereas TNF-a is known to be a key mediator involved in parasitemia control but can also contribute to enhanced erythrophagocytosis.43,44 Furthermore, these cytokines favor the maturation of naïve T cells into Th1 T cells, which are involved in cellmediated immunity. We also observed extremely high levels of MCP1, a chemokine that plays an important role in monocyte recruitment.45 Contrary to other protozoan infections, such as those from Leishmania major,46 Toxoplasma gondii47 or Plasmodium chabaudi,48 where this recruitment is essential for the effective control of the infection, in T. brucei infections expression of MCP1 and other chemokines seem to have deleterious effects, especially during early infection, contributing to enhanced pathogenesis.49,50 However, such effects might be mitigated by the production of the type II cytokine IL-10, which potentially limits MCP1 expression and reduces monocyte recruitment from the BM.51 IL-10 is also known to downregulate IFN-g and TNF-a, and, depending on the balance between these cytokines, it may contribute to attenuate the severity of the anemia.52
During the development of the immune response to various pathogens, hepcidin is known to be key in the regulation of iron metabolism, leading to reduced mobilization and redistribution of iron in order to limit its access by pathogens, and in turn, to the so-called anemia of inflammation. 37,38 However, there are cases where iron redistribution and anemia occur but by mechanisms that are hepcidin- independent.22 As such, we investigated the possible role of hepcidin in the development of trypanosome-related anemia and further looked into the molecular mechanisms subjacent to the transition from a status of acute anemia to a status of recovery/chronic anemia.
Increases in hepcidin expression were observed in BALB/c and C57BL/6 mice, with no discernible expression in Hamp-/- mice. The liver is long known to be the major contributor for systemic hepcidin levels, and thus the master regulator of iron homeostasis. In response to an infectious/ inflammatory stimulus, an increased expression of hepcidin is triggered in the liver, mostly mediated by IL-6. Hepcidin then binds to ferroportin, leading to its internalization and degradation, effectively blocking iron release from hepatocytes, intestinal enterocytes and macrophages.19-21,37,53 In prolonged infections, this limits iron availability for the pathogens, but also for the host itself, thus leading to the aforementioned anemia of inflammation. However, since there is no hepcidin in Hamp-/- mice, there is no limitation in iron availability, so the milder anemia observed in these animals is likely mediated by hepcidin-independent mechanisms, which is not always required for the onset of early inflammatory hypoferremia.54,55
The increased hepcidin expression in the spleen, kidney and BM is expected to have a low impact on systemic iron homeostasis, but may have an important role in the control of local iron fluxes. As with the hematologic parameters, there is a turning point at around day 7 when the infectious stimulus that leads to increased hepcidin expression seems to be replaced by an inhibitory signal that suppresses hepcidin. This could partially be explained by a decrease in IL-6 levels, but there are likely other signaling pathways contributing to this suppression. As such, we also evaluated the expression of genes that are influenced by hepcidin or, in turn, influence hepcidin expression.
Ferroportin is the major target for hepcidin, being removed from the cell surface and also inhibited at the expression level.21,56 As the sole known iron exporter, this interaction will severely limit iron release and mobilization, especially by the intestinal enterocytes, recycling macrophages and hepatocytes, leading to hypoferremia. In both BALB/c and C57BL/6 mice, ferroportin expression correlates both negatively with hepcidin expression and positively with the development of anemia, being downregulated at the earlier days of infection. This limits iron release for the production of new erythrocytes and leads to anemia. It is subsequently up-regulated over the following days when iron is again being released and enhanced erythropoiesis occurs, allowing a recovery from anemia. However, in Hamp-/- mice there is no such control of ferroportin due to the lack of hepcidin, so iron is readily available to allow for the faster recovery from anemia observed in these animals. A similar regulation of ferroportin was observed at the protein level. Levels in the liver of C57BL/6 mice closely matched variations in mRNA expression, and also mirrored hepcidin expression, with a decrease up to day 7. This is then followed by a recovery and an increase over subsequent days, indicating an early iron retention and a later release from the liver. In Hamp-/- mice, liver FPN1 levels also closely matched mRNA expression, and were kept elevated throughout the experiment, with the zenith at day 7. These results show that there was no limitation in iron release from the liver during infection, thus supporting the hypothesis of a faster erythrocyte recovery when compared with C57BL/6 mice. In the spleen, a similar response was observed for both C57BL/6 and Hamp-/-, with a very significant decrease in ferroportin levels, followed by a later increase in the second stage of infection, albeit faster and higher in the Hamp-/- mice. During anemia of inflammation, after erythrophagocytosis, iron is not properly released from macrophages due to ferroportin internalization mediated by hepcidin (hence the development of anemia). But at a later stage, during recovery, ferroportin levels are normalized, iron mobilized and erythropoiesis also normalizes, leading to a recovery from anemia. No significant changes were observed in duodenal levels of ferroportin. It is likely that body iron levels were already sufficient to cope with the erythropoietic demands, so there was no need for additional dietary iron absorption. Additionally, we must also consider that analysis of ferroportin by Western blot does not distinguish between functional ferroportin on the cell membrane and possibly non-functional ferroportin in intracellular compartments, which could hide the smaller differences between WT and KO mice. Nevertheless, it is clear that the lack of hepcidin allows for a faster recovery and normalization of ferroportin levels, and thus, for an earlier availability of iron required for erythropoiesis.
The later suppression of hepcidin also negatively correlates with increases in the expression of several erythroid regulators. Erythropoietin (EPO) is one of the signaling molecules driving erythropoiesis, being produced mostly by the kidney. It is essential for EPO receptor (R)-mediated erythropoiesis that occurs in the BM and the spleen. Although EPO can influence hepcidin expression, it does not seem to act directly on it, but rather indirectly through erythroferrone (ERFE) produced by erythroid progenitors. 57 Interestingly, there is no major role for ERFE in baseline erythropoiesis, but it rather functions during erythropoiesis- related stress58 and during recovery from anemia of inflammation,59 by suppressing hepcidin and increasing iron availability. Our data show that in Hamp-/- mice ERFE does not seem to be involved in the recovery from anemia, despite the increase in EPO, since no variations in expression were observed, which opens up the possibility that ERFE is not only involved in hepcidin suppression, but also acts as a sensor for hepcidin levels. Other erythroid regulators that can influence hepcidin, such as the predominantly erythroblast-produced GDF15 and TWSG1,60,61 were also found to be over-expressed at the later stage of infection and could contribute to hepcidin suppression. These findings are very similar to previous observations in bacterial infections. During injection with heat-inactivated Brucella abortus,25,26 C57BL/6 mice have similar patterns of hepcidin expression, with a significant increase in the early days and a decrease in later days of infection. Mice also develop anemia of inflammation and iron restriction, and can only partially recover from it. However, when hepcidin is suppressed (in Hamp-/- mice), anemia is ameliorated and there is a faster recovery. Furthermore, a role for IL-6 in the onset and resolution of anemia is also shown,26 both by triggering increased hepcidin expression and by interfering with erythropoiesis. However, recovery from anemia in IL-6-/- mice is not as fast as in Hamp-/- mice, showing that although IL-6 is a strong inducer of hepcidin during inflammatory conditions, it is not the only one.
In summary, T.b. brucei infection leads to the rapid development of anemia followed by a partial recovery (Figure 8). In the acute phase, a strong inflammatory signature is associated with hepcidin expression causing iron redistribution and limited availability. During the recovery phase, the decrease in hepcidin expression might be due to the decrease in the inflammatory response and the increased production of erythroid regulators. Importantly, the lack of hepcidin clearly reduces the severity of trypanosomederived anemia. This knowledge could contribute to the development of novel strategies for the treatment and control of trypanosomiasis-derived anemia, limiting its impact on human and non-human health.
Footnotes
- Received May 30, 2019
- Accepted January 2, 2020
Correspondence
Disclosures
This work was funded by National Funds through FCT—Fundação para a Ciência e a Tecnologia, I.P., under the project UIDB/04293/2020, and by individual funding from the Portuguese Foundation for Science and Technology (FCT) through CEECIND/00048/2017 (ACG), SFRH/BD/114899/2016 (CB), SFRH/BD/CEECIND/02362/2017 (JT), SFRH/BD/123734/2016 (DMC).
Contributions
JVN, ACG, DMC and CB performed research; JVN, ACG and DMC performed data analysis; JVN, ACG and DMC wrote the manuscript; JVN, JT, ACS and PNSR supervised the study; SV provided the Hamp KO animals; PNSR, JT, ACS, SV contributed data and edited the manuscript. All authors revised and approved the manuscript.
References
- Aksoy S, Buscher P, Lehane M, Solano P, Van Den Abbeele J. Human African trypanosomiasis control: Achievements and challenges. PLoS Negl Tro Dis. 2017; 11(4):e0005454. https://doi.org/10.1371/journal.pntd.0005454PubMedPubMed CentralGoogle Scholar
- Courtin D, Berthier D, Thevenon S, Dayo GK, Garcia A, Bucheton B. Host genetics in African trypanosomiasis. Infecti Genet Evol. 2008; 8(3):229-238. https://doi.org/10.1016/j.meegid.2008.02.007PubMedGoogle Scholar
- Amole BO, Clarkson AB, Shear HL. Pathogenesis of anemia in Trypanosoma brucei-infected mice. Infect Immun. 1982; 36(3):1060-1068. https://doi.org/10.1128/IAI.36.3.1060-1068.1982PubMedGoogle Scholar
- Cox FE. Pathogenesis of animal trypanosomiasis. Nature. 1979; 277(5698):603-604. https://doi.org/10.1038/277603a0PubMedGoogle Scholar
- Murray M, Dexter TM. Anaemia in bovine African trypanosomiasis. A review. Acta Trop. 1988; 45(4):389-432. https://doi.org/10.1002/j.1834-4453.1988.tb00198.xPubMedGoogle Scholar
- d'Ieteren GD, Authie E, Wissocq N, Murray M. Trypanotolerance, an option for sustainable livestock production in areas at risk from trypanosomosis. Rev Sci Tech. 1998; 17(1):154-175. https://doi.org/10.20506/rst.17.1.1088PubMedGoogle Scholar
- Mbaya A, Kumshe H, Nwosu CO. The mechanisms of anaemia in Trypanosomiasis: a review. 2010;269-82. Google Scholar
- Stijlemans B, Cnops J, Naniima P. Development of a pHrodo-based assay for the assessment of in vitro and in vivo erythrophagocytosis during experimental trypanosomosis. PLoS Negl Trop Dis. 2015; 9(3):e0003561. https://doi.org/10.1371/journal.pntd.0003561PubMedPubMed CentralGoogle Scholar
- Stijlemans B, De Baetselier P, Magez S, Van Ginderachter JA, De Trez C. African Trypanosomiasis-associated anemia: the contribution of the interplay between parasites and the mononuclear phagocyte system. Front Immunol. 2018; 9:218. https://doi.org/10.3389/fimmu.2018.00218PubMedPubMed CentralGoogle Scholar
- Huan CN, Webb L, Lambert PH, Miescher PA. Pathogenesis of the anemia in African Trypanosomiasis: characterization and purification of a hemolytic factor. Schweiz Med Wochenschr. 1975; 105(47):1582-1583. Google Scholar
- Tizard IR, Sheppard J, Neilsen K. The characterization of a second class of haemolysins from Trypanosoma brucei. Trans R Soc Trop Med Hyg. 1978; 72(2):198-200. https://doi.org/10.1016/0035-9203(78)90063-9PubMedGoogle Scholar
- Vickerman K, Tetley L. Biology and ultastructure of trypanosomes in relation to pathogenesis. 1978;23-31. Google Scholar
- Igbokwe IO, Esievo KA, Saror DI, Obagaiye OK. Increased susceptibility of erythrocytes to in vitro peroxidation in acute Trypanosoma brucei infection of mice. Vet Parasitol. 1994; 55(4):279-286. https://doi.org/10.1016/0304-4017(94)90070-1PubMedGoogle Scholar
- Umar IA, Ogenyi E, Okodaso D. Amelioration of anaemia and organ damage by combined intraperitoneal administration of vitamins A and C to Trypanosoma brucei brucei-infected rats. Afr J Biotechnol. 2007; 6(18):2083-2086. https://doi.org/10.5897/AJB2007.000-2322PubMedGoogle Scholar
- Clemens MR, Waller HD. Lipid peroxidation in erythrocytes. Chem Phys Lipids. 1987; 45(2-4):251-268. https://doi.org/10.1016/0009-3084(87)90068-5PubMedGoogle Scholar
- Szempruch AJ, Sykes SE, Kieft R. Extracellular vesicles from Trypanosoma brucei mediate virulence factor transfer and cause host anemia. Cell. 2016; 164(1-2):246-257. https://doi.org/10.1016/j.cell.2015.11.051PubMedPubMed CentralGoogle Scholar
- Stijlemans B, Beschin A, Magez S, Van Ginderachter JA, De Baetselier P. Iron homeostasis and Trypanosoma brucei associated immunopathogenicity development: a battle/ quest for iron. Biomed Res Int. 2015; 2015:819389. https://doi.org/10.1155/2015/819389PubMedPubMed CentralGoogle Scholar
- Stijlemans B, Vankrunkelsven A, Brys L, Magez S, De Baetselier P. Role of iron homeostasis in trypanosomiasis-associated anemia. Immunobiology. 2008; 213(9-10):823-835. https://doi.org/10.1016/j.imbio.2008.07.023PubMedGoogle Scholar
- Nemeth E, Ganz T. Regulation of iron metabolism by hepcidin. Annu Rev Nutr. 2006; 26:323-342. https://doi.org/10.1146/annurev.nutr.26.061505.111303PubMedGoogle Scholar
- Ganz T. Hepcidin-a regulator of intestinal iron absorption and iron recycling by macrophages. Best Pract Res Clin Haematol. 2005; 18(2):171-182. https://doi.org/10.1016/j.beha.2004.08.020PubMedGoogle Scholar
- Ramey G, Deschemin JC, Durel B, Canonne-Hergaux F, Nicolas G, Vaulont S. Hepcidin targets ferroportin for degradation in hepatocytes. Haematologica. 2010; 95(3):501-504. https://doi.org/10.3324/haematol.2009.014399PubMedPubMed CentralGoogle Scholar
- Moreira AC, Neves JV, Silva T, Oliveira P, Gomes MS, Rodrigues PN. Hepcidin- (in)dependent mechanisms of iron metabolism regulation during infection by Listeria and Salmonella. Infect Immun. 2017; 85(9)https://doi.org/10.1128/IAI.00353-17PubMedPubMed CentralGoogle Scholar
- Arezes J, Jung G, Gabayan V. Hepcidininduced hypoferremia is a critical host defense mechanism against the siderophilic bacterium Vibrio vulnificus. Cell Host Microbe. 2015; 17(1):47-57. https://doi.org/10.1016/j.chom.2014.12.001PubMedPubMed CentralGoogle Scholar
- Burns M, Muthupalani S, Ge Z. Helicobacter pylori infection induces anemia, depletes serum iron storage, and alters local iron-related and adult brain gene expression in male INS-GAS mice. PLoS One. 2015; 10(11):e0142630. https://doi.org/10.1371/journal.pone.0142630PubMedPubMed CentralGoogle Scholar
- Kim A, Fung E, Parikh SG. A mouse model of anemia of inflammation: complex pathogenesis with partial dependence on hepcidin. Blood. 2014; 123(8):1129-1136. https://doi.org/10.1182/blood-2013-08-521419PubMedGoogle Scholar
- Gardenghi S, Renaud TM, Meloni A. Distinct roles for hepcidin and interleukin-6 in the recovery from anemia in mice injected with heat-killed Brucella abortus. Blood. 2014; 123(8):1137-1145. https://doi.org/10.1182/blood-2013-08-521625PubMedPubMed CentralGoogle Scholar
- Ben-Othman R, Flannery AR, Miguel DC, Ward DM, Kaplan J, Andrews NW. Leishmania-mediated inhibition of iron export promotes parasite replication in macrophages. PLoS Pathog. 2014; 10(1):e1003901. https://doi.org/10.1371/journal.ppat.1003901PubMedPubMed CentralGoogle Scholar
- Ferrer P, Castillo-Neyra R, Roy CN, Sullivan DJ. Dynamic control of hepatic Plasmodium numbers by hepcidin despite elevated liver iron during iron supplementation. Microbes Infect. 2016; 18(1):48-56. https://doi.org/10.1016/j.micinf.2015.08.018PubMedPubMed CentralGoogle Scholar
- Lesbordes-Brion JC, Viatte L, Bennoun M. Targeted disruption of the hepcidin 1 gene results in severe hemochromatosis. Blood. 2006; 108(4):1402-1405. https://doi.org/10.1182/blood-2006-02-003376PubMedGoogle Scholar
- Torrence JD, Bothwell TH. Tissue Iron Stores. 1980;104-9. Google Scholar
- McLatchie AP, Burrell-Saward H, Myburgh E. Highly sensitive in vivo imaging of Trypanosoma brucei expressing "red-shifted" luciferase. PLoS Negl Trop Dis. 2013; 7(11):e2571. https://doi.org/10.1371/journal.pntd.0002571PubMedPubMed CentralGoogle Scholar
- Viatte L, Vaulont S. Hepcidin, the iron watcher. Biochimie. 2009; 91(10):1223-1228. https://doi.org/10.1016/j.biochi.2009.06.012PubMedGoogle Scholar
- Wang W, Knovich MA, Coffman LG, Torti FM, Torti SV. Serum ferritin: past, present and future. Biochim Biophys Acta. 2010; 1800(8):760-769. https://doi.org/10.1016/j.bbagen.2010.03.011PubMedPubMed CentralGoogle Scholar
- Basu S, Horakova E, Lukes J. Iron-associated biology of Trypanosoma brucei. Biochim Biophys Acta. 2016; 1860(2):363-370. https://doi.org/10.1016/j.bbagen.2015.10.027PubMedGoogle Scholar
- Stefanova D, Raychev A, Arezes J. Endogenous hepcidin and its agonist mediate resistance to selected infections by clearing non-transferrin-bound iron. Blood. 2017; 130(3):245-257. https://doi.org/10.1182/blood-2017-03-772715PubMedPubMed CentralGoogle Scholar
- Stefanova D, Raychev A, Deville J. Hepcidin protects against lethal Escherichia coli sepsis in mice inoculated with isolates from septic patients. Infect Immun. 2018; 86(7)https://doi.org/10.1128/IAI.00253-18PubMedPubMed CentralGoogle Scholar
- Andrews NC. Anemia of inflammation: the cytokine-hepcidin link. J Clin Invest. 2004; 113(9):1251-1253. https://doi.org/10.1172/JCI21441PubMedPubMed CentralGoogle Scholar
- Roy CN, Andrews NC. Anemia of inflammation: the hepcidin link. Curr Opin Hematol. 2005; 12(2):107-111. https://doi.org/10.1097/00062752-200503000-00001PubMedGoogle Scholar
- Soares MP, Hamza I. Macrophages and iron metabolism. Immunity. 2016; 44(3):492-504. https://doi.org/10.1016/j.immuni.2016.02.016PubMedPubMed CentralGoogle Scholar
- Nemeth E, Rivera S, Gabayan V. IL-6 mediates hypoferremia of inflammation by inducing the synthesis of the iron regulatory hormone hepcidin. J Clin Invest. 2004; 113(9):1271-1276. https://doi.org/10.1172/JCI200420945Google Scholar
- Wrighting DM, Andrews NC. Interleukin-6 induces hepcidin expression through STAT3. Blood. 2006; 108(9):3204-3209. https://doi.org/10.1182/blood-2006-06-027631PubMedPubMed CentralGoogle Scholar
- Felli N, Pedini F, Zeuner A. Multiple members of the TNF superfamily contribute to IFN-gamma-mediated inhibition of erythropoiesis. J Immunol. 2005; 175(3):1464-1472. https://doi.org/10.4049/jimmunol.175.3.1464PubMedGoogle Scholar
- Magez S, Radwanska M, Beschin A, Sekikawa K, De Baetselier P. Tumor necrosis factor alpha is a key mediator in the regulation of experimental Trypanosoma brucei infections. Infect Immun. 1999; 67(6):3128-3132. https://doi.org/10.1128/IAI.67.6.3128-3132.1999PubMedGoogle Scholar
- Magez S, Schwegmann A, Atkinson R. The role of B-cells and IgM antibodies in parasitemia, anemia, and VSG switching in Trypanosoma brucei-infected mice. PLoS Pathog. 2008; 4(8):e1000122. https://doi.org/10.1371/journal.ppat.1000122PubMedPubMed CentralGoogle Scholar
- Deshmane SL, Kremlev S, Amini S, Sawaya BE. Monocyte chemoattractant protein-1 (MCP-1): an overview. J Interferon Cytokine Res. 2009; 29(6):313-326. https://doi.org/10.1089/jir.2008.0027PubMedPubMed CentralGoogle Scholar
- Leon B, Lopez-Bravo M, Ardavin C. Monocyte-derived dendritic cells formed at the infection site control the induction of protective T helper 1 responses against Leishmania. Immunity. 2007; 26(4):519-531. https://doi.org/10.1016/j.immuni.2007.01.017PubMedGoogle Scholar
- Dunay IR, Damatta RA, Fux B. Gr1(+) inflammatory monocytes are required for mucosal resistance to the pathogen Toxoplasma gondii. Immunity. 2008; 29(2):306-317. https://doi.org/10.1016/j.immuni.2008.05.019PubMedPubMed CentralGoogle Scholar
- Sponaas AM, Freitas do Rosario AP, Voisine C. Migrating monocytes recruited to the spleen play an important role in control of blood stage malaria. Blood. 2009; 114(27):5522-5531. https://doi.org/10.1182/blood-2009-04-217489PubMedGoogle Scholar
- Bosschaerts T, Guilliams M, Stijlemans B. Tip-DC development during parasitic infection is regulated by IL-10 and requires CCL2/CCR2, IFN-gamma and MyD88 signaling. PLoS Pathog. 2010; 6(8):e1001045. https://doi.org/10.1371/journal.ppat.1001045PubMedPubMed CentralGoogle Scholar
- Liu Y, Li Z, Bakhiet M. Upregulation of the chemokines Rantes, MCP-1, MIP-1a and MIP-2 in early infection with Trypanosoma brucei brucei and inhibition by sympathetic denervation of the spleen. Trop Med Int Health. 1999; 4(2):85-92. https://doi.org/10.1046/j.1365-3156.1999.00364.xPubMedGoogle Scholar
- Guilliams M, Movahedi K, Bosschaerts T. IL-10 dampens TNF/inducible nitric oxide synthase-producing dendritic cell-mediated pathogenicity during parasitic infection. J Immunol. 2009; 182(2):1107-1118. https://doi.org/10.4049/jimmunol.182.2.1107PubMedGoogle Scholar
- Musaya J, Matovu E, Nyirenda M, Chisi J. Role of cytokines in Trypanosoma bruceiinduced anaemia: a review of the literature. Malawi Med J. 2015; 27(2):45-50. https://doi.org/10.4314/mmj.v27i2.3PubMedGoogle Scholar
- Rishi G, Wallace DF, Subramaniam VN. Hepcidin: regulation of the master iron regulator. Biosci Rep. 2015; 35(3)https://doi.org/10.1042/BSR20150014PubMedPubMed CentralGoogle Scholar
- Deschemin JC, Vaulont S. Role of hepcidin in the setting of hypoferremia during acute inflammation. PLoS One. 2013; 8(4):e61050. https://doi.org/10.1371/journal.pone.0061050PubMedPubMed CentralGoogle Scholar
- Willemetz A, Beatty S, Richer E. Ironand hepcidin-independent downregulation of the iron exporter ferroportin in macrophages during Salmonella infection. Front Immunol. 2017; 8:498. https://doi.org/10.3389/fimmu.2017.00498PubMedPubMed CentralGoogle Scholar
- Nemeth E, Tuttle MS, Powelson J. Hepcidin regulates cellular iron efflux by binding to ferroportin and inducing its internalization. Science. 2004; 306(5704):2090-2093. https://doi.org/10.1126/science.1104742PubMedGoogle Scholar
- Kautz L, Jung G, Valore EV, Rivella S, Nemeth E, Ganz T. Identification of erythroferrone as an erythroid regulator of iron metabolism. Nat Genet. 2014; 46(7):678-684. https://doi.org/10.1038/ng.2996PubMedPubMed CentralGoogle Scholar
- Arezes J, Foy N, McHugh K. Erythroferrone inhibits the induction of hepcidin by BMP6. Blood. 2018; 132(14):1473-1477. https://doi.org/10.1182/blood-2018-06-857995PubMedPubMed CentralGoogle Scholar
- Kautz L, Jung G, Nemeth E, Ganz T. Erythroferrone contributes to recovery from anemia of inflammation. Blood. 2014; 124(16):2569-2574. https://doi.org/10.1182/blood-2014-06-584607PubMedPubMed CentralGoogle Scholar
- Tanno T, Bhanu NV, Oneal PA. High levels of GDF15 in thalassemia suppress expression of the iron regulatory protein hepcidin. Nat Med. 2007; 13(9):1096-1101. https://doi.org/10.1038/nm1629PubMedGoogle Scholar
- Tanno T, Porayette P, Sripichai O. Identification of TWSG1 as a second novel erythroid regulator of hepcidin expression in murine and human cells. Blood. 2009; 114(1):181-186. https://doi.org/10.1182/blood-2008-12-195503PubMedPubMed CentralGoogle Scholar
Data Supplements
Figures & Tables
Article Information
This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.