Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2025 Feb 15.
Published in final edited form as: J Immunol. 2024 Feb 15;212(4):607–616. doi: 10.4049/jimmunol.2300195

Schistosome infection impacts hematopoiesis

Tobias Wijshake *, Joseph Rose III *, Jipeng Wang †,, Jacob Zielke *, Madeleine Marlar-Pavey *,§, Weina Chen , James J Collins III , Michalis Agathocleous *,
PMCID: PMC10872488  NIHMSID: NIHMS1951192  PMID: 38169327

Abstract

Helminth infections are common in animals. However, the impact of a helminth infection on the function of hematopoietic stem cells (HSCs) and other hematopoietic cells has not been comprehensively defined. Here we describe the hematopoietic response to infection of mice with Schistosoma mansoni, a parasitic flatworm which causes schistosomiasis. We analyzed the frequency or number of hematopoietic cell types in the bone marrow, spleen, liver, thymus, and blood, and observed multiple hematopoietic changes caused by infection. Schistosome infection impaired bone marrow HSC function after serial transplantation. Functional HSCs were present in the infected liver. Infection blocked bone marrow erythropoiesis and augmented spleen erythropoiesis, observations consistent with the anemia and splenomegaly prevalent in schistosomiasis patients. This work defines the hematopoietic response to schistosomiasis, a debilitating disease afflicting more than 200 million people, and identifies impairments in HSC function and erythropoiesis.

Introduction

Schistosomiasis is a parasitic disease caused by infection with Schistosoma spp. flatworms. It afflicts more than 200 million people in Africa, the Middle East, South-East Asia, and South America (1, 2). Vaccines are not available. Treatment can clear adult parasites but is ineffective against immature parasites, does not prevent reinfection, nor does it reverse multi-organ immunopathology (3). As a result, the chronic symptoms of schistosomiasis contribute substantially to the global disability burden, creating a cycle of poverty and infection (47). Schistosomes are shed as larvae from freshwater snails, infect humans by penetrating the skin, and can live in the circulation for decades (8), continuously laying eggs which lodge in liver, bladder, and other organs. Egg antigens trigger a Th2 response which dominates the chronic phase of the disease and is the main cause of pathology (1, 3, 9, 10). Several schistosomiasis symptoms including anemia, splenomegaly, and inflammation suggest hematopoietic involvement. However, the effect of schistosomiasis on HSCs and restricted progenitors is mostly uncharacterized.

Until recently most humans were likely to be parasitized (11). The prevalence of infection with schistosomes or other helminths was as high as 50% in some populations before the modern era (12, 13), or in modern untreated populations living in endemic areas (14). Primates and other animals are parasitized in the wild (15) and in some areas as many as 90% of primates have a history of schistosomiasis (16). Thus, HSCs and the hematopoietic system have likely evolved under near-constant pressure from schistosomes and other helminths. Recent studies have examined the impact on HSC function of various infections (17), including Mycobacterium avium, (18, 19), E. coli (20), Streptococcus (21), Plasmodium (22), Ehrlichia muris (23), Leishmania (24), C. albicans (25), Salmonella (26, 27) and several viruses (2830). Most of these infections induce a strong proinflammatory Th1 response. The impact of Th2 response-dominated helminth infections on hematopoiesis has been much less characterized (3133) and the effects of schistosomes or other helminths on HSCs and hematopoiesis have not been comprehensively analyzed. To address this, we investigated the impact of S. mansoni infection on HSCs and hematopoiesis in mice.

Materials and Methods

Mice.

Mice were on a C57BL/Ka background. Both male and female mice were used in all studies. Young adult mice were infected at the ages of 10–21 weeks and were either analyzed or used as donors for transplantation at 7 weeks after schistosome infection. C57BL/Ka-Thy-1.1 (CD45.2) and C57BL/Ka-Thy-1.2 (CD45.1) mice were used for transplantation experiments. Mice were housed in the Animal Resource Center of UT Southwestern and all procedures were approved by the UT Southwestern Institutional Animal Care and Use committee.

Schistosome infection.

Each mouse was infected with around 200 Schistosoma mansoni (NMRI strain) cercariae released from infected Biomphalaria glabrata snails (Schistosome Resource Center) by percutaneous tail exposure (34). Mice were restrained, and their tail was dipped in water containing cercariae for 30 minutes.

Cell isolation and hematopoietic analysis.

Bone marrow cells were obtained by flushing femurs and tibia with 25G needle, or crushing femurs, tibias, vertebrae, and pelvic bones with a mortar and pestle in staining medium consisting of Ca2+/Mg2+-free Hank’s balanced salt solution (HBSS; Gibco), supplemented with 2% heat-inactivated bovine serum (Gibco). Spleens and thymuses were mechanically dissociated by trituration in staining medium. Livers were enzymatically digested for 30 minutes at 37°C in 1.5 ml RPMI-1640 (Sigma), containing 250 μg/ml liberase (Roche) and 100 μg/ml DNase I (Roche). Cell suspensions were filtered through a 40 μm strainer. Cell number was assessed with a Vi-CELL cell viability analyzer (Beckman Coulter). Blood was collected by cardiac puncture using a 25G needle and mixed in a tube containing 5 μl 0.5M EDTA. Complete blood cell counts were determined using a hemavet HV950 (Drew Scientific). For hematopoietic analysis, 40 μl blood was lysed in 1 ml of ammonium chloride buffer (155mM NH4Cl; 10 mM KHCO3; 0.1 mM EDTA) for 10 minutes at 4°C. Cells were incubated with fluorescently conjugated antibodies for 90 minutes on ice when using CD34 antibody or for 30 minutes at 4°C. Cells were washed with staining media and resuspended in staining media containing 1 μg/ml DAPI or 1 μg/ml propidium iodide for live/dead discrimination. Cell populations were defined with the following markers: CD150+CD48LineageSca-1+Kit+ hematopoietic stem cells (HSCs), CD150CD48LineageSca-1+Kit+ multipotent progenitor cells (MPPs) (35), CD150CD48+LineageSca-1+Kit+ hematopoietic progenitor cells (HPC-1), CD150+CD48+LineageSca-1+Kit+ hematopoietic progenitor cells (HPC-2) (36), CD34+CD16/32LineageSca-1Kit+ common myeloid progenitors (CMPs), CD34+CD16/32+LineageSca-1Kit+ granulocyte–monocyte progenitors (GMPs), CD34CD16/32LineageSca-1Kit+ megakaryocyte–erythroid progenitors (MEPs) (37), Mac-1+ myeloid cells, Mac1+CD115+Ly6C+Ly6G inflammatory monocytes, Mac1+CD115Ly6Cmid/highSiglecF+ eosinophils, Mac1+CD115Ly6CmidLy6G+ neutrophils, CD11c+ dendritic cells (DCs), CD11c+Mac1+Ly6C+ monocytic DCs (moDCs), CD11c+Mac1 Mac1DCs, Mac1B220+ B cells, Mac1CD3+ T cells, Mac1B220CD3CD71midTer119 immature erythroid progenitors, Mac1B220CD3CD71+Ter119 erythroid progenitors, and Mac1B220CD3CD71+Ter119+ erythroid progenitors. The Lineage cocktail for HSCs and progenitors consisted of CD2, CD3, CD5, CD8, Ter119, B220, and Gr-1 antibodies. T cell progenitor populations in the thymus were defined with the following markers, after excluding Mac-1+, B220+, and Ter119+ cells: CD4+CD8+ double-positive (DP), CD3+CD4+CD8 (CD4+ single positive, CD4+SP), CD3+CD4CD8+ (CD8+SP), CD4CD8 double-negative (DN), CD4CD8CD44+CD25 (DN1), CD4CD8CD44+CD25+ (DN2), CD4CD8CD44CD25+ (DN3), CD4CD8CD44CD25 (DN4), and CD3CD4CD8+ immature single positive (ISP). Mature T cell populations were defined by the following markers, after excluding Mac-1+, B220+, and Ter119+ cells: CD4+ (CD4+ cells), CD4+CD44CD62L+ (naïve CD4+ cells), CD4+CD44+CD62L+ (CD4+ central memory cells), CD4+CD44+CD62LCD69+ (CD4+ resident memory cells), CD4+CD44+CD62LCD69CD103 (CD4+ effector memory cells), CD8+ (CD8+ cells), CD8+CD44CD62L+ (naïve CD8+ cells), CD8+CD44+CD62L+ (CD8+ central memory cells), CD8+CD44+CD62LCD69+ (CD8+ resident memory cells), and CD4+CD44+CD62LCD69CD103 (CD8+ effector memory cells). For all flow cytometry experiments, combinations of antibodies were used against the following cell-surface markers, conjugated to Violet Fluor 450, Brilliant Violet 421, Brilliant Violet 510, FITC, PE, PE594, PerCP-Cy5.5, PE-Cy7, APC, eFluor 660, Alexa Fluor 700, RedFluor 710, APC/Fire 750, or APC-Cy7: CD2 (RM2–5, Tonbo Biosciences/BioLegend), CD3 (17A2, Tonbo Biosciences/BioLegend), CD4 (GK1.5, Tonbo Biosciences/BioLegend), CD5 (53–7.3, BioLegend), CD8a (53–6.7, Tonbo Biosciences/BioLegend), CD11b (Mac1) (M1/70, Tonbo Biosciences/BioLegend), CD11c (N488, BioLegend), CD16/32 (93, BioLegend), CD25 (PC61, BioLegend), CD34 (RAM34, Invitrogen), CD44 (M7, Tonbo Biosciences), CD45.1 (A20, Tonbo Biosciences), CD45.2 (104, Tonbo Biosciences/BioLegend), CD45R (B220) (RA.3–6B2, Tonbo Biosciences/BioLegend), CD48 (HM48–1, BioLegend), CD62L (MEL-14, Tonbo Biosciences), CD69 (H1.2F3, Tonbo Biosciences), CD71 (R17217, BioLegend), CD103 (2E7, BioLegend), CD115 (CSF-1R) (AFS98, BioLegend), CD117 (c-Kit) (2B8, BioLegend), CD150 (TC15–12F12.2, BioLegend), Ly-6A/E (Sca-1) (D7, BioLegend), Ly-6C (HK1.4, BioLegend), Ly-6G (Gr-1) (RB6–8C5, Tonbo Biosciences; 1A8, BioLegend), SiglecF (E50–2440, BD Biosciences) or Ter119 (TER119, Tonbo Biosciences/BioLegend). Analysis and sorting were performed using the FACSAria flow cytometer (BD Biosciences) or a FACSCanto (BD Biosciences). Data were analyzed using FlowJo (Flowjo LLC) or FACSDiva (BD Biosciences).

Bone marrow and liver reconstitution assays.

Recipient mice (CD45.1) were irradiated using an XRAD 320 X-ray irradiator (Precision X-Ray) with two doses of 540 rad (1080 rad in total) delivered at least 3 hours apart. Bone marrow cells were injected into the retro-orbital venous sinus of anesthetized recipients. Seven weeks prior to transplant, donor mice were either infected with 200 cercariae or left uninfected (controls). For competitive transplants, 5 × 105 CD45+-selected bone marrow cells from infected or from uninfected donor (CD45.2) mice and 5 × 105 competitor (CD45.1;CD45.2) cells were mixed and transplanted by injection into the retro-orbital venous sinus of anesthetized recipients. Recipient mice were maintained on antibiotic water (Baytril 0.08 mg/ml) for 1 week pre-transplantation, and for 4 weeks after transplantation. Blood was obtained from the tail veins of recipient mice every four weeks for at least 16 weeks after transplantation. Red blood cells were lysed in ammonium chloride lysis buffer. The remaining cells were stained with antibodies against CD45.2, CD45.1, C11b (Mac1), CD115, Ly6G, Ly6C, CD45R (B220), and CD3 and analyzed by flow cytometry. For the secondary bone marrow transplants, 1 × 107 bone marrow cells from primary recipients were harvested 5 months after primary transplant and transplanted into lethally irradiated CD45.1 secondary recipients. For the competitive liver reconstitution assays, 2 × 106 CD45+-selected liver cells from infected or from uninfected donor (CD45.2) mice and 4 × 105 bone marrow competitor (CD45.1;CD45.2) cells from uninfected mice were mixed and transplanted into lethally irradiated CD45.1 recipient mice. Bone marrow cells for analysis of transplant recipient mice or for secondary transplantations was obtained by crushing femurs, tibias, vertebrae, and pelvic bones.

Histological evaluation, immunohistochemistry and immunofluorescence.

Livers and spleens from control and Schistosoma mansoni-infected mice were collected and fixed in 10% formalin for at least 24 hours. After fixation, tissues were submitted to the UTSW Histo Pathology Core for processing, paraffin embedding, sectioning and hematoxylin & eosin staining. H&E sections of livers and spleens from uninfected control and infected mice were evaluated for pathology, hematopoiesis, erythropoiesis, and inflammation by a pathologist (W.C). Immunohistochemistry was performed on formalin-fixed, paraffin-embedded livers and spleens. Sections were gradually rehydrated and heat-induced Tris-EDTA buffer (10mM Tris base, 1 mM EDTA, 0.1% Tween, pH 9.0) was used for antigen retrieval. Sections were washed in PBS with 0.1% Tween and blocked in PBS with 10% BSA containing 0.1% Tween for 1h. Sections were stained overnight with the following antibodies: myeloperoxidase (MPO; EPR20257, Abcam; 1/500), or CD61 (SJ19–09, Thermo Fisher; 1/100). Secondary antibody was IgG-HRP (Cell Signaling, 7074; 1/1000) and the signal was visualized by using DAB peroxidase substrate kit according to the manufacturer’s instructions (Vector Laboratories, SK-4100). Sections were counterstained with Mayer’s hematoxylin (Sigma, MHS32) and mounted with Vectashield Express mounting medium (H-5700). Images were acquired on an Olympus BX53 microscope. For immunofluorescence, dissected livers were fixed overnight in 4% paraformaldehyde (Fisher Scientific, AAJ19943K2) on a rotator at 4°C. Livers were then washed with PBS, rotated overnight in 20% sucrose at 4°C before being frozen in OCT compound on dry ice. Livers were sectioned into 8 μm slices using a cryostat (Leica CM3050S) and dried at 57 °C for 1 hour. Antigens were unmasked in 10 mM citric acid, 0.05% Tween20 buffer (pH 6) using a pressure cooker at 7.5 psi for 17 min and then cooled for 30 min. Sections were washed in PBS with 0.1% Tween and blocked with 10% BSA, 0.1% Tween in PBS for 1 hour at room temperature. Sections were stained overnight in a moist chamber at 4°C with a combination of the following primary antibodies: c-Kit (R&D Biosciences, AF1356; 1/150), desmin (Epredia, RB9014P0; 1/200), CD31 (Abcam, ab28364; 1/50), and MPO (Abcam, ab208670; 1/100). Sections were stained for 1 hour in the dark with DAPI and a combination of the following secondary antibodies: rabbit Alexa Fluor 568 (Invitrogen, A10042; 1/200), goat Alexa Fluor 647 (Invitrogen, A32849; 1/200), rabbit Alexa Fluor 488 (Invitrogen, A32790; 1/200). Slides were mounted with Aqua-Poly/Mount (Polysciences, 18606–20). Images were acquired on a Zeiss LSM780 inverted confocal microscope using the tiling function and tiled images were then stitched together using Zen software (Zeiss) to create a composite image of the tissue.

Quantitative RT-PCR.

Livers were homogenized in 1 ml TRIzol LS reagent (Life Technologies, 10296010) using the FastPrep-24 5G bead beater (MP Biomedicals). Total RNA was isolated by chloroform extraction, precipitated with isopropanol, washed with 75% ethanol and RNA pellets were dissolved in RNase/DNase-free water. Reverse transcription of RNA was performed using the iScript cDNA synthesis kit (BioRad, 1708891) according to the manufacturer’s instructions and cDNA was diluted 10-fold prior to use. 20 ng cDNA was used for quantitative RT-PCR with iTaq Universal SYBR Green Supermix (BioRad, 1725122) and a CFX384 Real-Time System (BioRad). Data were analyzed using the BioRad CFX Maestro software. GAPDH expression levels were used for normalization. The following primers were used: Gapdh: 5’-GGAGAGTGTTTCCTCGTCCC-3’, 5’-ACTGTGCCGTTGAATTTGCC-3’; Gm-csf (Csf2): 5’-GATATTCGAGCAGGGTCTACG-3’, 5’-AGGCTGTCTATGAAATCCGC-3’; M-csf (Csf1): 5’- ACCCAGGATGAGGACAGAC-3’, 5’-AGGAAGATGGTAGGAGAGGG-3’; Scf (Kitl): 5’-TCAAGAGGTGTAATTGTGGACG-3’, 5’- GGGTAGCAAGAACAGGTAAGG-3’.

Statistical analysis.

Most figure panels show the pooled results from mice we analyzed from multiple independent experiments. Mice were allocated to experiments randomly. For most experiments the operator was not blinded to the treatment. Uninfected littermate controls, or uninfected controls from litters of the same parental strains born a few days apart were used for experiments. Prior to analyzing the statistical significance of differences among treatments, we tested whether data were normally distributed and whether variance was similar among treatments. To test for normal distribution, we performed the Shapiro-Wilk test when 3 ≤ n < 20 or the D’Agostino & Pearson test when n ≥ 20. To test if variability significantly differed among treatments, we performed F-tests. If the data did not significantly (p < 0.01 for at least one treatment) deviate from normality, we used a parametric test, otherwise data were log-transformed and tested for a significant deviation from normality. If the log-transformed data passed normality, a parametric test was used on the transformed data. If both the untransformed and log-transformed data did not pass the normality test, a non-parametric test was used on the untransformed data. To assess the statistical significance of a difference between two treatments, we used a t-test for data that was normally distributed and had equal variability, or a t-test with Welch’s correction for data that was normally distributed and had unequal variability, or a Mann-Whitney test for data that was not normally distributed. To assess the statistical significance of differences between treatments when multiple measurements were taken across time, we used a repeated measures mixed-effects model for data for which some values were missing. To determine the statistical significance between treatments for the presence of multi-lineage reconstitution, we used a Fisher’s exact test.

Results

The effects of schistosome infection on hematopoiesis.

To understand the impact of schistosomiasis on HSCs and hematopoietic progenitors, we infected mice with the human pathogen Schistosoma mansoni by tail exposure in water containing cercariae. In this model, eggs deposited in the liver trigger granuloma formation and schistosomiasis pathology starting from ~ 5 weeks post-infection (10, 38, 39). We analyzed the hematopoietic system in the bone marrow and spleen of mice 7 weeks post-infection (Fig. 1A). As compared to uninfected mice, bone marrow cellularity did not significantly change after infection (Fig. 1B). The frequencies of CD150+CD48LineageSca-1+Kit+ HSCs and CD150CD48LineageSca-1+Kit+ multipotent progenitors (MPPs) were unchanged (Fig. 1CD). Infection increased the frequency of CD150CD48+LineageSca-1+Kit+ hematopoietic progenitor cells (HPC-1) (Fig. 1E) and decreased the frequency of CD34+CD16/32LineageSca-1Kit+ common myeloid progenitors (CMPs) and CD34CD16/32LineageSca-1Kit+ megakaryocyte-erythroid progenitors (MEPs) (Fig. 1F, 1G). The observed decline in CMP and MEP frequency was similar to a previous study examining the effects of schistosome infection in Apoe-deficient mice on a high-fat diet (40). The frequencies of CD150+CD48+LineageSca-1+Kit+ (HPC-2) and CD34+CD16/32+LineageSca-1Kit+ granulocyte-monocyte progenitors (GMPs) did not change (Fig. 1H, 1I). The spleen size and cellularity significantly increased after infection (Supplemental Fig. 1A, 1B), however the frequency of HSCs and most progenitor cell types in the spleen did not change (Supplemental Fig. 1CP). This suggests that in contrast to other infectious or inflammatory challenges, the spleen is not a reservoir for multilineage hematopoiesis in schistosomiasis despite its increased size.

Figure 1. The effects of schistosome infection on the frequency of hematopoietic cells in the bone marrow and blood cells.

Figure 1.

(A) Schematic overview of analysis of the hematopoietic and blood system 7 weeks after Schistosoma mansoni infection. (B-N) The frequency of HSCs, progenitors and mature cell types in the bone marrow of S. mansoni infected mice or uninfected controls (n = 7–13 mice per treatment). (O-R) Blood cell counts of white blood cells, neutrophils, lymphocytes, and monocytes of infected or uninfected mice (n = 9–13 mice per treatment). (S-W) The frequency of immune cell types in the blood of infected or uninfected mice (n = 4–11 mice per treatment). For all graphs both male and female mice were used. All graphs show mean ± s.d. In all figures, *p < 0.05, **p < 0.01, ***p < 0.001. Statistical significance was assessed with a t-test with Welch’s correction (B-C, E, G-H, J-K, O-Q, S and U-W), a t-test (F, I, L-M, R and T), or a Mann-Whitney test (D and N).

In the myeloid lineage, infection preferentially increased the frequency of bone marrow monocytes and eosinophils but not neutrophils or dendritic cells (Fig. 1JM). Infection significantly decreased bone marrow B cell frequency (Fig. 1N) in agreement with a recent report (41). The infected spleen had an increased frequency and number of most myeloid lineage cell types (Supplemental Fig. 1QAB). Infected mice had a higher white blood cell count than uninfected mice, and increased numbers or frequencies of neutrophils, monocytes, eosinophils, and dendritic cells in the blood (Fig. 1OW). The development of major T cell progenitor cell types in the thymus was not significantly impacted by infection (Supplemental Fig. 2). This contrasts with the severe impact of many other infections on thymus cellularity (42). To determine the effects of infection on T cells, we analyzed T-cell subpopulations in the blood, bone marrow, spleen, and liver. In the marrow, the frequency of total CD4+ and effector memory CD4+ T cells increased and the frequency of CD8+ T cells did not change (Supplemental Fig. 3AF). Infection did not cause significant changes in frequencies of T cell subsets in the spleen (Supplemental Fig. 3GL). The frequency of naïve CD4+ T cells declined in the blood (Supplemental Fig. 3MR). The liver had an increased frequency of total CD4+ or CD8+ T cells, particularly of a resident memory immunophenotype (Supplemental Fig. 3SV). Therefore, schistosome infection caused several changes in the frequency of hematopoietic and immune cells in the marrow, spleen, blood, and liver.

Infection impairs bone marrow HSC function after serial transplantation.

To test if infection changes HSC activity, donor bone marrow cells from infected or uninfected mice were mixed with competitor bone marrow cells from uninfected mice and transplanted into lethally irradiated recipients (Fig. 2A). This classical competition assay determines changes in long-term HSC activity irrespectively of changes in phenotypic surface markers, which can change after infection (17). There was no significant difference in donor cell reconstitution capacity between infected and uninfected mice (Fig. 2BE, Supplemental Fig. 4A, 4B). At 16 weeks after transplant, there was no significant difference in reconstituted lineage composition between blood cells from infected as compared to uninfected mice (Supplemental Fig. 4CG) suggesting infection did not cause long-term cell-intrinsic changes in HSC differentiation potential. Despite the maintenance of hematopoietic reconstitution in the peripheral blood, the bone marrow of transplant recipients had proportionately fewer infected donor-derived HSCs and some restricted progenitors as compared to uninfected donor-derived HSCs or progenitors (Fig. 2F, 2G). This suggested an impairment in HSC self-renewal after transplantation. To test HSC self-renewal after the primary transplant and the ability of HSCs to respond to repeated challenge, bone marrow cells from primary recipient mice were transplanted into lethally irradiated secondary transplant recipients 5 months after primary transplant (Fig. 2H). The blood reconstitution capacity of bone marrow cells from infected mice significantly decreased as compared to uninfected mice (Fig. 2IL, Supplemental Fig. 4H, 4I). The proportion of infected donor-derived myeloid progenitors and total hematopoietic cells in the marrow of secondary transplant recipients decreased (Fig. 2M, 2N). Therefore, schistosome infection impaired HSC function after serial transplant.

Figure 2. Schistosome infection impairs bone marrow HSC function.

Figure 2.

(A) Schematic overview of experiments to assess bone marrow HSC function after infection. 5 × 105 CD45.2+ donor cells from bone marrow of mice infected with S. mansoni for seven weeks, or from uninfected mice, were mixed with 5 × 105 CD45.1+CD45.2+ competitor bone marrow cells from uninfected mice and transplanted to each lethally irradiated CD45.1+ recipient mouse (n = 5 donor and 21–25 recipient mice per treatment). (B-E) Donor cell reconstitution of CD45+, myeloid, B, and T cells in the blood at the indicated time points. (F) The percentage of donor-derived hematopoietic stem and progenitor cells in the bone marrow at 5 months after transplantation (n = 21–25 mice per treatment). (G) The percentage of donor-derived myeloid, B and T cells in the bone marrow at 5 months after transplantation (n = 15 mice per treatment). (H) Schematic overview of the secondary transplantation. 1 × 107 bone marrow cells from primary recipient mice were harvested 5 months after primary transplant and transplanted into each lethally irradiated secondary recipient mouse (n = 3 donor mice and 14–15 recipient mice per treatment). (I-L) Donor cell reconstitution of CD45+, myeloid, B and T cells in the blood at the indicated time points after secondary transplantation. (M) The percentage of donor-derived hematopoietic stem and progenitor cells in the bone marrow after secondary transplantation (n = 13–14 mice per treatment). (N) Representative flow plots of donor-derived Lineage cells (left), LinSca-1+Kit+ (LSK) cells (middle), and LinSca-1Kit+ (LK) cells (right). Arrows indicate the respective parent population. All graphs show mean ± s.d. Statistical significance was assessed with a repeated measures mixed model (B-E, and I-L), a t-test (F-G, and M, HSC and HPC-2 cells), a t-test with Welch’s correction (M, HPC-1; CMP; GMP; MEP; and CD45+ cells) or a Mann-Whitney test (F, HPC-1 cells; and M, MPP cells).

Hematopoietic activity in the liver of infected mice.

The liver of infected mice harbors schistosome eggs which trigger granuloma formation. To test if schistosomiasis elicits multilineage hematopoiesis in the liver, we analyzed the frequency of phenotypic HSCs and other hematopoietic progenitors 7 weeks after infection with S. mansoni. The liver of infected mice had an increased frequency of phenotypic HSCs, HPC-1, HPC-2, CMPs, GMPs and MEPs (Fig. 3AF). Among mature cells, the frequency of inflammatory monocytes, eosinophils, dendritic cells, and monocytic dendritic cells also increased after infection, the frequency of neutrophils was unchanged, and the frequency of B cells decreased (Fig. 3GL). The infected liver was enlarged (Fig. 3M). Its architecture was distorted by chronic inflammation in the portal triads and increased hematopoiesis. Lodged S. mansoni eggs surrounded by mixed histiocytes/granulocytes formed granulomas (Fig. 3N). Liver sinuses harbored hematopoietic clusters with erythroid and megakaryocyte cell production (Fig. 3N). Immunohistochemistry staining for MPO and CD61 confirmed a dramatic increase of granulocytes throughout the liver, including in the granulomas and sinuses, and a mild increase in megakaryocytes (Fig. 3O, 3P). Therefore, inflammation and hematopoiesis increase in infected liver. To test the localization of hematopoietic cells in infected livers, we assessed immunofluorescence staining for the hematopoietic stem and progenitor marker c-kit in combination with other markers. In uninfected liver, almost all c-kit+ cells were also positive for the endothelial cell marker CD31 and localized around blood vessels, suggesting they were endothelial cells. By contrast in infected liver some c-kit+ cells were negative for CD31, localized within the hepatic tissue, and had the round morphology of hematopoietic cells, consistent with a hematopoietic progenitor identity (Fig. 4A). In infected liver, CD31c-kit+ cells localized near cells which were positive for desmin, a marker of hepatic stellate cells, consistent with the ubiquitous presence of desmin+ cells (Fig. 4B). Infected livers harbored clusters of MPO-positive cells, including in the periphery of granulomas and in the liver parenchyma, some of which co-stained with c-kit suggesting granulopoietic foci (Fig 4CE). Infection increased the expression in the liver of hematopoietic stem and progenitor cell growth factors including SCF, M-CSF, and GM-CSF (Fig. 4FH), suggesting the presence of niches to support active hematopoiesis in the infected liver.

Figure 3. Schistosome infection promotes liver hematopoiesis.

Figure 3.

(A-L) Frequency of hematopoietic stem, progenitor, and mature immune cells in the liver of schistosome infected or uninfected control mice (n = 4–12 mice per treatment). (M) Liver weight after schistosome infection. (N) Representative images of H&E staining of livers from uninfected control (left) and infected mice (middle and right). Middle panel shows inflammation composed of mixed histiocytes and granulocytes associated with heavy egg lodging (red arrow) in infected mice. Right panel shows a microabscess (red arrow) and increased hematopoiesis including erythropoiesis (green arrow) and megakaryopoiesis (black arrow) in infected mice. (O) Representative images of immunohistochemical staining for MPO+ granulocytes in livers from uninfected (left) or infected mice (middle and right). (P) Representative images of immunohistochemical staining for CD61+ megakaryocytes (black arrows) in livers from uninfected (left) or infected mice (right). All graphs show mean ± s.d. Statistical significance was assessed with a Mann-Whitney test (A), a t-test with Welch’s correction (B-C, E, H, K, and M), and a t-test (D, F-G, I-J, and L).

Figure 4. Localization of c-Kit+ cells in the liver of schistosome-infected mice.

Figure 4.

(A-B) Representative images of immunofluorescence staining for (A) CD31+ endothelial and c-kit+CD31 hematopoietic progenitor cells, and (B) desmin+ hepatic stellate cells and c-kit+ hematopoietic progenitor cells. White arrows indicate c-kit+ hematopoietic progenitor cells in (A-B). (C-E) Representative images of immunofluorescence staining for MPO+ granulocytes and c-kit+ hematopoietic progenitor cells (magnified image in C, white arrows in D, E) in livers from schistosome infected mice, including in (D) granulomas and the (E) liver parenchyma. For (A, B, D, and E) a green pseudocolor was used for Alexa Fluor 568. (F-H) Quantitative RT-PCR analysis of HSC growth factors in livers from uninfected control and infected mice (n = 6–8 mice per treatment). All graphs show mean ± s.d. Statistical significance was assessed with a t-test (F, H) or a t-test with Welch’s correction (G).

To test if the increased frequency of phenotypic HSCs corresponded to increased HSC function, we transplanted 2 × 106 CD45+ cells from the liver of infected or uninfected donor mice in competition with 4 × 105 bone marrow cells from uninfected mice into lethally irradiated recipient mice (Fig. 5A). The peripheral blood of recipient mice contained significantly more donor-derived cells from infected livers than from uninfected livers (Fig. 5BE). Nine out of ten mice receiving donor cells from infected liver showed donor-derived multilineage reconstitution as compared to only one out of fourteen mice receiving donor cells from uninfected liver (Fig. 5F). The bone marrow of transplant recipients had a significantly higher proportion of HSCs and restricted progenitors derived from infected donor livers as compared to uninfected donor livers (Fig. 5G, 5H). Thus, functional HSCs were present in the liver of infected mice.

Figure 5. HSC activity in the liver.

Figure 5.

(A) Schematic overview of experiments to assess HSC function in the infected liver. 2 × 106 CD45+ donor cells from livers of infected or uninfected control mice were mixed with 4 × 105 CD45+ competitor bone marrow cells from uninfected mice and transplanted to each lethally irradiated recipient mouse (n = 3 donor and 10–14 recipient mice per treatment). (B-E) Donor cell reconstitution of CD45+, myeloid, B and T cells in the blood at the indicated time points after transplantation. (F) The proportion of recipient mice which showed multilineage reconstitution after competitive transplantation of donor CD45+ cells from the liver of infected mice or uninfected controls. Multilineage reconstitution was defined as > 2% donor cell chimerism in peripheral blood myeloid, B and T cells at 16 weeks after transplantation. (G) The percentage of donor-derived hematopoietic stem and progenitor cells in the bone marrow at 16 weeks after transplantation (n = 10–14 mice per treatment). (H) Representative flow plots of chimerism analysis of LSK cells. All graphs show mean ± s.d. Statistical significance was assessed with a repeated measures mixed model (B-E), a Fisher’s exact test (F), a Mann-Whitney test (G, HSC; MPP; HPC-1; and GMP), a t-test with Welch’s correction (G, HPC-2; CMP; and CD45+), and a t-test (G, MEP).

Schistosome infection blocks marrow erythropoiesis and increases spleen erythropoiesis.

Schistosome-infected mice were anemic and thrombocytopenic (Fig. 6AD). They also showed increased red blood cell distribution width, a marker of anemia and inflammation which in humans correlates with increased mortality (43) (Fig. 6E). Chronic anemia is one of the most prevalent and disabling symptoms of schistosomiasis (44). Several causes have been proposed including anemia of inflammation, blood loss, erythrocyte spleen sequestration, or autoimmunity (44). Schistosome-infected mice showed a sharp decrease in CD71+Ter119+ erythroid progenitors and an accumulation of CD71midTer119 immature progenitors in the marrow (Fig. 6FI) suggesting a block in marrow erythropoiesis. In contrast the spleen of infected mice had an increased frequency of CD71+Ter119 and CD71+Ter119+ erythroid progenitors (Fig. 6JM). Therefore, erythropoiesis in schistosomiasis infection shifted from the bone marrow to the spleen. Splenic architecture was disrupted by reactive follicular hyperplasia with germinal centers in the white pulp and erythropoiesis and megakaryopoiesis in the red pulp (Fig. 6N, 6O). Staining for CD61 and MPO showed megakaryopoiesis and the presence of small numbers of granulocytes in spleens of infected mice (Fig. 6P, 6Q). S. mansoni egg lodging in spleens was rare, and granuloma formation was observed in only one out of five assessed spleens from infected mice. Consequently, inflammation associated with histiocytes and granulocytes was relatively mild in the spleen as compared to the liver (Fig. 6N). Our results are consistent with the idea that splenomegaly after S. mansoni infection is largely driven by extramedullary erythropoiesis rather than inflammation.

Figure 6. Schistosome infection blocks bone marrow erythropoiesis and increases spleen erythropoiesis.

Figure 6.

(A-E) Analysis of red blood cells, hemoglobin, hematocrit, platelets, and red blood cell distribution width of S. mansoni infected or uninfected mice. Analysis was performed 7 weeks after infection. (F-H) The frequency of CD71midTer119 immature progenitors, CD71+Ter119 and CD71+Ter119+ erythroid progenitors in the bone marrow of infected mice or uninfected control mice (n = 7–14 mice per treatment). (I) Representative flow plots of progenitors quantified in (F-H). (J-L) The frequency of CD71midTer119 immature progenitors, CD71+Ter119 and CD71+Ter119+ erythroid progenitors in the spleen of infected mice or uninfected control mice (n = 5–11 mice per treatment). (M) Representative flow plots of progenitors quantified in (J-L). (N-O) Images of H&E staining of spleens from uninfected or infected mice showing follicular hyperplasia with germinal centers in the white pulp (black arrow in N), significant increased hematopoiesis including erythropoiesis in the red pulp (green arrow in N), rare inflammation composed of mixed histiocytes and granulocytes associated with Schistosoma egg lodging (red arrow in N), and (O) significant increased hematopoiesis including erythropoiesis in the red pulp within the splenic cords/sinuses. (P-Q) Representative images of immunohistochemical staining for CD61+ megakaryocytes (black arrows in P), and MPO+ granulocytes (red arrow in Q) in the spleen of infected mice or uninfected controls. (R) Graphical summary. All data show mean ± s.d. Statistical significance was assessed with a t-test (A-C, E, G, and J-K) and a t-test with Welch’s correction (D, F, H, and L).

Discussion

The impact of schistosomiasis on hematopoietic stem cells.

Our findings provide a framework to understand how S. mansoni infection affects hematopoiesis (Fig. 6R). The frequency of bone marrow HSCs is not reduced after infection, and HSCs from infected mice can reconstitute primary recipients but are compromised in their ability to reconstitute secondary recipients. The molecular mechanisms that regulate HSC function in response to schistosome infection remain unknown. Schistosomiasis elevates the levels of several cytokines. Some type 2 cytokines, including IL-4 and IL-25 promote the development of restricted hematopoietic progenitors without impacting HSCs (45, 46), and consistent with this a type 2 immune response did not impact HSC frequency in mice infected with Trichuris muris (33). Other schistosomiasis-associated cytokines can have either protective or detrimental effects on HSCs. For example, eosinophilia caused by IL-5 overexpression negatively regulates HSC frequency and function through CCL-6 production (47). Conversely, IL-10 protects HSCs in inflammatory contexts and promotes HSC function during regeneration (48). Thus, the effects on HSC function we observe may reflect a balance of positive and negative effects from multiple cytokines. Populations in areas endemic for schistosomiasis face several hematopoietic challenges, including a high burden of anemia and of other infectious diseases such as malaria. Our findings that HSC function in serial transplantation is reduced after schistosome infection suggests that schistosomiasis impairs the ability of the hematopoietic system to regenerate after repeated challenge.

Erythropoiesis is blocked in the marrow but not the spleen.

It is thought that inflammation is the most common cause of anemia in schistosomiasis (49). Our results suggest that anemia in schistosomiasis is partly caused by a sharp block in bone marrow erythropoiesis. This block is accompanied by a striking increase in spleen erythropoiesis. Inflammation is known to reduce marrow erythropoiesis (50) and consistent with this schistosomiasis increased inflammatory monocytes in the bone marrow and blood. The mechanisms for the arrest in erythropoiesis are not known but may involve cytokines known to be elevated in schistosomiasis, such as IL-10 which can cause anemia (51). It is interesting that schistosomiasis arrested erythropoiesis in the marrow but not in the spleen. The inflammatory response in the spleen after infection is mild, and the spleen of infected mice does not harbor proportionately more HSCs or myeloid progenitors than the spleen of uninfected mice. Therefore, splenomegaly in schistosomiasis is not a consequence of general extramedullary hematopoiesis or inflammation but specifically of splenic erythropoiesis. Our results suggest that an important function of the spleen is to make erythrocytes in schistosome infections.

Hematopoiesis in the liver.

We show that HSCs and restricted progenitors are present in the liver after infection, as assayed by immunophenotypic, immunofluorescence and transplantation experiments. CD45+ cells from liver of infected mice competed against bone marrow cells from uninfected mice at a 5:1 donor:competitor ratio produced an average reconstitution of 20%, or 1:4 donor:competitor cells after transplant. This suggests that functional HSC frequency is 20-fold lower in infected liver than bone marrow. Given that the bone marrow typically contains more CD45+ cells than the liver, systemic output of blood cells from liver HSCs in infection is likely minor as compared to bone marrow hematopoiesis. However, hematopoiesis in the infected liver, including in the periphery of granulomas, may be important for the local immune response. The production of monocyte-derived macrophages in the infected liver has been suggested to be protective (5254), and, consistent with our results, proliferating myeloid lineage cells are found in the periphery of granulomas (55). The fact that HSC or immature progenitor frequency is not elevated in spleen of infected mice suggests some specificity to liver hematopoiesis as opposed to general activation of extramedullary hematopoiesis. Are HSCs and hematopoietic progenitors in the infected liver supported by specialized niches? Our results show that in the infected liver, c-kit+ hematopoietic progenitors localize close to hepatic stellate cells and the expression of HSC and progenitor growth factors increases. Hepatic stellate cells serve as the major HSC niche cell type in fetal liver hematopoiesis by secreting the c-kit ligand SCF (56). They can also express known HSC niche factors in adult mice, including SCF after schistosome infection (57) or CXCL12 in other contexts (58). Thus, we hypothesize that during schistosome infection HSCs migrate to the liver and are supported by reactivation of a dormant liver hematopoietic niche.

Supplementary Material

1

Key Points.

  • Schistosome infection impairs bone marrow HSC function after serial transplantation

  • Infection blocks marrow erythropoiesis and increases spleen erythropoiesis

  • Hematopoietic activity in the liver of infected mice

Acknowledgments

We thank members of the Collins lab for help with infections; the Moody Foundation Flow Cytometry Facility for flow cytometry; John Shelton and the UTSW Histo Pathology Core for sectioning and H&E staining; and the UTSW Quantitative Light Microscopy Core for confocal microscopy.

M.A. is a Cancer Prevention and Research Institute of Texas scholar and an American Society of Hematology faculty scholar. This work was supported by grants from Cancer Prevention and Research Institute of Texas (RR180007), the American Society of Hematology Faculty Scholar award, the Moody Foundation, and the National Institutes of Health (R01DK125713) to M.A and from the Welch Foundation (I-1948-20180324) and the National Institutes of Health (R01AI121037, R01AI150776) to JJC.

Abbreviations used in this article:

BM

bone marrow

CMP

common myeloid progenitor

DN

double negative thymocyte

DP

double positive thymocyte

GMP

granulocyte–monocyte progenitor

HPC-1

hematopoietic progenitor cell subset 1

HPC-2

hematopoietic progenitor cell subset 2

HSC

hematopoietic stem cell

LK

LineageSca1Kit+

LSK

LineageSca1+Kit+

MEP

megakaryocyte–erythroid progenitor

MPP

multipotent progenitor cell

MPO

myeloperoxidase

SCF

stem cell factor

Footnotes

Disclosure

The authors declare no financial conflicts of interest.

REFERENCES

  • 1.Colley DG, Bustinduy AL, Secor WE, and King CH. 2014. Human schistosomiasis. Lancet 383: 2253–2264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.McManus DP, Dunne DW, Sacko M, Utzinger J, Vennervald BJ, and Zhou XN. 2018. Schistosomiasis. Nat Rev Dis Primers 4: 13. [DOI] [PubMed] [Google Scholar]
  • 3.Abdel Aziz N, Musaigwa F, Mosala P, Berkiks I, and Brombacher F. 2022. Type 2 immunity: a two-edged sword in schistosomiasis immunopathology. Trends Immunol 43: 657–673. [DOI] [PubMed] [Google Scholar]
  • 4.King CH 2010. Parasites and poverty: the case of schistosomiasis. Acta Trop 113: 95–104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.King CH, Dickman K, and Tisch DJ. 2005. Reassessment of the cost of chronic helmintic infection: a meta-analysis of disability-related outcomes in endemic schistosomiasis. Lancet 365: 1561–1569. [DOI] [PubMed] [Google Scholar]
  • 6.King CH, and Dangerfield-Cha M. 2008. The unacknowledged impact of chronic schistosomiasis. Chronic Illn 4: 65–79. [DOI] [PubMed] [Google Scholar]
  • 7.Lo NC, Addiss DG, Hotez PJ, King CH, Stothard JR, Evans DS, Colley DG, Lin W, Coulibaly JT, Bustinduy AL, Raso G, Bendavid E, Bogoch II, Fenwick A, Savioli L, Molyneux D, Utzinger J, and Andrews JR. 2017. A call to strengthen the global strategy against schistosomiasis and soil-transmitted helminthiasis: the time is now. Lancet Infect Dis 17: e64–e69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Harris AR, Russell RJ, and Charters AD. 1984. A review of schistosomiasis in immigrants in Western Australia, demonstrating the unusual longevity of Schistosoma mansoni. Trans R Soc Trop Med Hyg 78: 385–388. [DOI] [PubMed] [Google Scholar]
  • 9.Fairfax K, Nascimento M, Huang SC, Everts B, and Pearce EJ. 2012. Th2 responses in schistosomiasis. Semin Immunopathol 34: 863–871. [DOI] [PubMed] [Google Scholar]
  • 10.Pearce EJ, and MacDonald AS. 2002. The immunobiology of schistosomiasis. Nat Rev Immunol 2: 499–511. [DOI] [PubMed] [Google Scholar]
  • 11.Girgis NM, Gundra UM, and Loke P. 2013. Immune regulation during helminth infections. PLoS Pathog 9: e1003250. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Flammer PG, Ryan H, Preston SG, Warren S, Prichystalova R, Weiss R, Palmowski V, Boschert S, Fellgiebel K, Jasch-Boley I, Kairies MS, Rummele E, Rieger D, Schmid B, Reeves B, Nicholson R, Loe L, Guy C, Waldron T, Machacek J, Wahl J, Pollard M, Larson G, and Smith AL. 2020. Epidemiological insights from a large-scale investigation of intestinal helminths in Medieval Europe. PLoS Negl Trop Dis 14: e0008600. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Wang T, Cessford C, Dittmar JM, Inskip S, Jones PM, and Mitchell PD. 2022. Intestinal parasite infection in the Augustinian friars and general population of medieval Cambridge, UK. Int J Paleopathol 39: 115–121. [DOI] [PubMed] [Google Scholar]
  • 14.Shehata MA, Chama MF, and Funjika E. 2018. Prevalence and intensity of Schistosoma haematobium infection among schoolchildren in central Zambia before and after mass treatment with a single dose of praziquantel. Trop Parasitol 8: 12–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Toft JD II 1986. The Pathoparasitology of Nonhuman Primates: A Review. In Primates. Proceedings in Life Sciences.. Benirschke K, ed. Springer, New York, NY. [Google Scholar]
  • 16.Standley CJ, Mugisha L, Dobson AP, and Stothard JR. 2012. Zoonotic schistosomiasis in non-human primates: past, present and future activities at the human-wildlife interface in Africa. J Helminthol 86: 131–140. [DOI] [PubMed] [Google Scholar]
  • 17.Caiado F, Pietras EM, and Manz MG. 2021. Inflammation as a regulator of hematopoietic stem cell function in disease, aging, and clonal selection. J Exp Med 218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Matatall KA, Jeong M, Chen S, Sun D, Chen F, Mo Q, Kimmel M, and King KY. 2016. Chronic Infection Depletes Hematopoietic Stem Cells through Stress-Induced Terminal Differentiation. Cell Rep 17: 2584–2595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Baldridge MT, King KY, Boles NC, Weksberg DC, and Goodell MA. 2010. Quiescent haematopoietic stem cells are activated by IFN-gamma in response to chronic infection. Nature 465: 793–797. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Burberry A, Zeng MY, Ding L, Wicks I, Inohara N, Morrison SJ, and Nunez G. 2014. Infection mobilizes hematopoietic stem cells through cooperative NOD-like receptor and Toll-like receptor signaling. Cell Host Microbe 15: 779–791. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Morales-Mantilla DE, Kain B, Le D, Flores AR, Paust S, and King KY. 2022. Hematopoietic stem and progenitor cells improve survival from sepsis by boosting immunomodulatory cells. Elife 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Haltalli MLR, Watcham S, Wilson NK, Eilers K, Lipien A, Ang H, Birch F, Anton SG, Pirillo C, Ruivo N, Vainieri ML, Pospori C, Sinden RE, Luis TC, Langhorne J, Duffy KR, Gottgens B, Blagborough AM, and Lo Celso C. 2020. Manipulating niche composition limits damage to haematopoietic stem cells during Plasmodium infection. Nat Cell Biol 22: 1399–1410. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.MacNamara KC, Jones M, Martin O, and Winslow GM. 2011. Transient activation of hematopoietic stem and progenitor cells by IFNgamma during acute bacterial infection. PLoS One 6: e28669. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Abidin BM, Hammami A, Stager S, and Heinonen KM. 2017. Infection-adapted emergency hematopoiesis promotes visceral leishmaniasis. PLoS Pathog 13: e1006422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Yanez A, Murciano C, O’Connor JE, Gozalbo D, and Gil ML. 2009. Candida albicans triggers proliferation and differentiation of hematopoietic stem and progenitor cells by a MyD88-dependent signaling. Microbes Infect 11: 531–535. [DOI] [PubMed] [Google Scholar]
  • 26.Mistry JJ, Hellmich C, Moore JA, Jibril A, Macaulay I, Moreno-Gonzalez M, Di Palma F, Beraza N, Bowles KM, and Rushworth SA. 2021. Free fatty-acid transport via CD36 drives beta-oxidation-mediated hematopoietic stem cell response to infection. Nat Commun 12: 7130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Takizawa H, Fritsch K, Kovtonyuk LV, Saito Y, Yakkala C, Jacobs K, Ahuja AK, Lopes M, Hausmann A, Hardt WD, Gomariz A, Nombela-Arrieta C, and Manz MG. 2017. Pathogen-Induced TLR4-TRIF Innate Immune Signaling in Hematopoietic Stem Cells Promotes Proliferation but Reduces Competitive Fitness. Cell Stem Cell 21: 225–240 e225. [DOI] [PubMed] [Google Scholar]
  • 28.Isringhausen S, Mun Y, Kovtonyuk L, Krautler NJ, Suessbier U, Gomariz A, Spaltro G, Helbling PM, Wong HC, Nagasawa T, Manz MG, Oxenius A, and Nombela-Arrieta C. 2021. Chronic viral infections persistently alter marrow stroma and impair hematopoietic stem cell fitness. J Exp Med 218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Hirche C, Frenz T, Haas SF, Doring M, Borst K, Tegtmeyer PK, Brizic I, Jordan S, Keyser K, Chhatbar C, Pronk E, Lin S, Messerle M, Jonjic S, Falk CS, Trumpp A, Essers MAG, and Kalinke U. 2017. Systemic Virus Infections Differentially Modulate Cell Cycle State and Functionality of Long-Term Hematopoietic Stem Cells In Vivo. Cell Rep 19: 2345–2356. [DOI] [PubMed] [Google Scholar]
  • 30.de Bruin AM, Demirel O, Hooibrink B, Brandts CH, and Nolte MA. 2013. Interferon-gamma impairs proliferation of hematopoietic stem cells in mice. Blood 121: 3578–3585. [DOI] [PubMed] [Google Scholar]
  • 31.Inclan-Rico JM, Hernandez CM, Henry EK, Federman HG, Sy CB, Ponessa JJ, Lemenze AD, Joseph N, Soteropoulos P, Beaulieu AM, Yap GS, and Siracusa MC. 2020. Trichinella spiralis-induced mastocytosis and erythropoiesis are simultaneously supported by a bipotent mast cell/erythrocyte precursor cell. PLoS Pathog 16: e1008579. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Rashidi NM, Scott MK, Scherf N, Krinner A, Kalchschmidt JS, Gounaris K, Selkirk ME, Roeder I, and Lo Celso C. 2014. In vivo time-lapse imaging shows diverse niche engagement by quiescent and naturally activated hematopoietic stem cells. Blood 124: 79–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Chenery AL, Antignano F, Hughes MR, Burrows K, McNagny KM, and Zaph C. 2016. Chronic Trichuris muris infection alters hematopoiesis and causes IFN-gamma-expressing T-cell accumulation in the mouse bone marrow. Eur J Immunol 46: 2587–2596. [DOI] [PubMed] [Google Scholar]
  • 34.Tucker MS, Karunaratne LB, Lewis FA, Freitas TC, and Liang YS. 2013. Schistosomiasis. Curr Protoc Immunol 103: 19 11 11–19 11 58. [DOI] [PubMed] [Google Scholar]
  • 35.Kiel MJ, Yilmaz OH, Iwashita T, Yilmaz OH, Terhorst C, and Morrison SJ. 2005. SLAM family receptors distinguish hematopoietic stem and progenitor cells and reveal endothelial niches for stem cells. Cell 121: 1109–1121. [DOI] [PubMed] [Google Scholar]
  • 36.Oguro H, Ding L, and Morrison SJ. 2013. SLAM family markers resolve functionally distinct subpopulations of hematopoietic stem cells and multipotent progenitors. Cell Stem Cell 13: 102–116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Akashi K, Traver D, Miyamoto T, and Weissman IL. 2000. A clonogenic common myeloid progenitor that gives rise to all myeloid lineages. Nature 404: 193–197. [DOI] [PubMed] [Google Scholar]
  • 38.Brunet LR, Finkelman FD, Cheever AW, Kopf MA, and Pearce EJ. 1997. IL-4 protects against TNF-alpha-mediated cachexia and death during acute schistosomiasis. J Immunol 159: 777–785. [PubMed] [Google Scholar]
  • 39.Fallon PG, Richardson EJ, McKenzie GJ, and McKenzie AN. 2000. Schistosome infection of transgenic mice defines distinct and contrasting pathogenic roles for IL-4 and IL-13: IL-13 is a profibrotic agent. J Immunol 164: 2585–2591. [DOI] [PubMed] [Google Scholar]
  • 40.Cortes-Selva D, Gibbs L, Maschek JA, Nascimento M, Van Ry T, Cox JE, Amiel E, and Fairfax KC. 2021. Metabolic reprogramming of the myeloid lineage by Schistosoma mansoni infection persists independently of antigen exposure. PLoS Pathog 17: e1009198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Musaigwa F, Kamdem SD, Mpotje T, Mosala P, Abdel Aziz N, Herbert DR, Brombacher F, and Nono JK. 2022. Schistosoma mansoni infection induces plasmablast and plasma cell death in the bone marrow and accelerates the decline of host vaccine responses. PLoS Pathog 18: e1010327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Nunes-Alves C, Nobrega C, Behar SM, and Correia-Neves M. 2013. Tolerance has its limits: how the thymus copes with infection. Trends Immunol 34: 502–510. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Perlstein TS, Weuve J, Pfeffer MA, and Beckman JA. 2009. Red blood cell distribution width and mortality risk in a community-based prospective cohort. Arch Intern Med 169: 588–594. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Friedman JF, Kanzaria HK, and McGarvey ST. 2005. Human schistosomiasis and anemia: the relationship and potential mechanisms. Trends Parasitol 21: 386–392. [DOI] [PubMed] [Google Scholar]
  • 45.Gao A, Gong Y, Zhu C, Yang W, Li Q, Zhao M, Ma S, Li J, Hao S, Cheng H, and Cheng T. 2019. Bone marrow endothelial cell-derived interleukin-4 contributes to thrombocytopenia in acute myeloid leukemia. Haematologica 104: 1950–1961. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Saenz SA, Siracusa MC, Perrigoue JG, Spencer SP, Urban JF Jr., Tocker JE, Budelsky AL, Kleinschek MA, Kastelein RA, Kambayashi T, Bhandoola A, and Artis D. 2010. IL25 elicits a multipotent progenitor cell population that promotes T(H)2 cytokine responses. Nature 464: 1362–1366. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Zhang C, Yi W, Li F, Du X, Wang H, Wu P, Peng C, Luo M, Hua W, Wong CC, Lee JJ, Li W, Chen Z, Ying S, Ju Z, and Shen H. 2018. Eosinophil-derived CCL-6 impairs hematopoietic stem cell homeostasis. Cell Res 28: 323–335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Kang YJ, Yang SJ, Park G, Cho B, Min CK, Kim TY, Lee JS, and Oh IH. 2007. A novel function of interleukin-10 promoting self-renewal of hematopoietic stem cells. Stem Cells 25: 1814–1822. [DOI] [PubMed] [Google Scholar]
  • 49.Butler SE, Muok EM, Montgomery SP, Odhiambo K, Mwinzi PM, Secor WE, and Karanja DM. 2012. Mechanism of anemia in Schistosoma mansoni-infected school children in Western Kenya. Am J Trop Med Hyg 87: 862–867. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Ganz T 2019. Anemia of Inflammation. N Engl J Med 381: 1148–1157. [DOI] [PubMed] [Google Scholar]
  • 51.Cardoso A, Martins AC, Maceiras AR, Liu W, Castro I, Castro AG, Bandeira A, Di Santo JP, Cumano A, Li Y, Vieira P, and Saraiva M. 2021. Interleukin-10 induces interferon-gamma-dependent emergency myelopoiesis. Cell Rep 37: 109887. [DOI] [PubMed] [Google Scholar]
  • 52.Gundra UM, Girgis NM, Gonzalez MA, San Tang M, Van Der Zande HJP, Lin JD, Ouimet M, Ma LJ, Poles J, Vozhilla N, Fisher EA, Moore KJ, and Loke P. 2017. Vitamin A mediates conversion of monocyte-derived macrophages into tissue-resident macrophages during alternative activation. Nat Immunol 18: 642–653. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Nascimento M, Huang SC, Smith A, Everts B, Lam W, Bassity E, Gautier EL, Randolph GJ, and Pearce EJ. 2014. Ly6Chi monocyte recruitment is responsible for Th2 associated host-protective macrophage accumulation in liver inflammation due to schistosomiasis. PLoS Pathog 10: e1004282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Girgis NM, Gundra UM, Ward LN, Cabrera M, Frevert U, and Loke P. 2014. Ly6C(high) monocytes become alternatively activated macrophages in schistosome granulomas with help from CD4+ cells. PLoS Pathog 10: e1004080. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Francisco JS, Terra M, Klein GCT, Dias de Oliveira B, and Pelajo-Machado M. 2022. The hepatic extramedullary hematopoiesis during experimental murine Schistosomiasis mansoni. Front Immunol 13: 955034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Lee Y, Leslie J, Yang Y, and Ding L. 2021. Hepatic stellate and endothelial cells maintain hematopoietic stem cells in the developing liver. J Exp Med 218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Brito JM, and Borojevic R. 1997. Liver granulomas in schistosomiasis: mast cell-dependent induction of SCF expression in hepatic stellate cells is mediated by TNF-alpha. J Leukoc Biol 62: 389–396. [DOI] [PubMed] [Google Scholar]
  • 58.Correia AL, Guimaraes JC, Auf der Maur P, De Silva D, Trefny MP, Okamoto R, Bruno S, Schmidt A, Mertz K, Volkmann K, Terracciano L, Zippelius A, Vetter M, Kurzeder C, Weber WP, and Bentires-Alj M. 2021. Hepatic stellate cells suppress NK cell-sustained breast cancer dormancy. Nature 594: 566–571. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

1

RESOURCES