Skip to main content

Vaccination accelerates hepatic erythroblastosis induced by blood-stage malaria

Abstract

Background

Vaccination induces survival of otherwise lethal blood-stage infections of the experimental malaria Plasmodium chabaudi. Blood-stage malaria induces extramedullary erythropoiesis in the liver. This study investigates how vaccination affects the course of malaria-induced expression of erythrocytic genes in the liver.

Methods

Female Balb/c mice were vaccinated at week 3 and week 1 before challenging with 106 P. chabaudi-parasitized erythrocytes. The non-infectious vaccine consisted of erythrocyte ghosts isolated from P. chabaudi-infected erythrocytes. Gene expression microarrays and quantitative real-time PCR were used to compare mRNA expression of different erythrocytic genes in the liver of vaccination-protected and non-protected mice during infections on days 0, 1, 4, 8, and 11 p.i.

Results

Global transcriptomics analyses reveal vaccination-induced modifications of malaria-induced increases in hepatic gene expression on days 4 and 11 p.i. On these days, vaccination also alters hepatic expression of the erythropoiesis-involved genes Ermap, Kel, Rhd, Rhag, Slc4a1, Gypa, Add2, Ank1, Epb4.1, Epb4.2, Epb4.9, Spta1, Sptb, Tmod1, Ahsp, Acyp1, Gata1, Gfi1b, Tal1, Klf1, Epor, and Cldn13. In vaccination-protected mice, expression of these genes, except Epb4.1, is significantly higher on day 4 p.i. than in un-protected non-vaccinated mice, reaches maximal expression at peak parasitaemia on day 8 p.i., and is slowed down or even decreased towards the end of crisis phase on day 11 p.i.. After day 1 p.i., Epor expression takes about the same course as that of the other erythroid genes. Hepatic expression of Epo, however, is delayed in both vaccinated and non-vaccinated mice for the first 4 days p.i. and is maximal at significantly higher levels in vaccinated mice on day 8 p.i., before declining towards the end of crisis phase on day 11 p.i.

Conclusion

The present data indicate that vaccination accelerates malaria-induced erythroblastosis in the liver for 1–2 days. This may contribute to earlier replenishment of peripheral red blood cells by liver-derived reticulocytes, which may favour final survival of otherwise lethal blood-stage malaria, since reticulocytes are not preferred as host cells by P. chabaudi.

Background

Malaria has caused about 219 million cases and about 435,000 deaths globally in 2017, with about 266,000 deaths in children aged under 5 years [1]. Currently, promising vaccine candidates are tested, but an effective anti-malarial vaccine is not yet commercially available [2,3,4].

The malaria-causing agents are parasitic protozoans of the genus Plasmodium, whose blood-stages, developing and multiplicating within host erythrocytes, exclusively cause morbidity and mortality of malaria [3]. The deadliest human malaria species is Plasmodium falciparum causing about 99% of global malaria-related deaths in humans [1]. Plasmodium falciparum shares several characteristics with Plasmodium chabaudi in mice which is, therefore, widely used as an appropriate experimental system to study host defense mechanisms against blood-stage malaria [5, 6]. The P. chabaudi model is also appropriate to study effects of vaccination on the outcome of blood-stage malaria. Using an experimental non-infectious vaccine, survival of mice can be raised from 0% to over 80% [7, 8]. This vaccine induces a healing course of otherwise lethal infections. For instance, primary infections with 106 P. chabaudi-parasitized erythrocytes take a similar course in terms of parasitaemia in vaccinated and non-vaccinated mice of the inbred mouse strain Balb/c [8]. The prepatent phase lasts for about 3 days, before the patent period begins on day 4 post infectionem (p.i.), characterized by the appearance of parasitized erythrocytes in the peripheral blood. The parasitaemias vary among infected mice between 1–5% without any difference between vaccinated and non-vaccinated mice. Maximum parasitaemia is reached on day 8 p.i. with about 60% parasitaemia in non-vaccinated mice, whereas it is reduced to about 40% in vaccinated mice. Peak parasitaemia is followed by the crisis phase characterized by dramatically declining parasitaemias to 5–1% within 3–4 days. All non-vaccinated mice succumb to infection during crisis, whereas over 80% of the vaccinated mice survive the infections [8].

The massive clearance of P. chabaudi-parasitized erythrocytes from circulation during crisis is ascribed to the spleen, which is widely considered as the exclusive effector organ against blood-stage malaria [9, 10]. Indeed, the spleen is thought of using the same mechanisms for eliminating malaria-parasitized erythrocytes in its red pulp, by which it normally removes senescent erythrocytes from circulation. Moreover, the liver is increasingly recognized as an effector organ against blood-stage malaria [11] due to its intrinsic immune system, its specific iron disposal system, and its capability to remove senescent and other aberrant erythrocytes including P. chabaudi-parasitized erythrocytes [11,12,13,14,15,16].

In P. chabaudi infections, the dramatic low levels of peripheral erythrocytes towards the end of crisis occur, though erythropoiesis is concomitantly active evidenced as increasing levels of peripheral reticulocytes [8]. Erythropoiesis is a highly complex process proceeding from the commitment of a haematopoietic stem cell to the megakaryocyte-erythroid lineage, which generates the primitive progenitors of the erythrocyte lineage, i.e., the slowly proliferating burst-forming unit-erythroid cells followed by the rapidly expanding colony-forming unit-erythroid (CFU-e) cells. These in turn are the precursors for the nucleated pro-erythroblasts and erythroblasts [17,18,19], which are arranged in erythroblastic islands organized around a central macrophage [20, 21]. Erythroblasts denucleate to reticulocytes which terminally differentiate to erythrocytes [20, 22]. Erythropoietin (EPO) and erythropoietin receptor (EPOR), respectively, play a critical role in erythropoiesis, in particular during erythroblastosis. EPOR is constitutively expressed on CFU-e and erythroblasts [23, 24]. EPO-sensitive erythropoiesis is obviously also critical for the outcome of P. chabaudi blood-stage malaria [25, 26]. In particular, EPO-induced proliferation of erythroid precursors has been described to be suppressed by parasite factors thus causing severe anaemia and exacerbating effects on the final outcome of P. chabaudi infections [27].

Erythropoiesis takes place in the bone marrow [20, 21]. Evidence, however, is increasing that, under specific stress situations, erythropoiesis can also take place outside the bone marrow. This stress or extramedullary erythropoiesis has been reported to occur in several organs, particularly in the spleen, of mice and even humans [20, 28,29,30,31]. Previously, the spleen was found to be a site of increased erythropoiesis in P. chabaudi-resistant C57BL/6 mice [32]. Thereafter, the liver was also found to be a site of malaria-induced erythropoiesis. Thus, blood-stage infections with the non-lethal Plasmodium yoelii 17XL in C57BL/6 mice were reported to induce large cell clusters containing TER119+ nucleated reticulocytes in the parenchymal space of the liver [33, 34]. This aspect has been neglected until recently when several erythrocytic genes were detected by transcriptomics screening analyses to be expressed in the liver of mice infected with P. chabaudi [35, 36]. At present, however, there is only poor information available on the dynamics of stress-induced extramedullary erythropoiesis in general and, in particular, on the dynamics of malaria-induced extramedullary erythropoiesis in the liver at the gene expression level. To get a better understanding of extramedullary erythropoiesis in the liver induced by blood-stage malaria and, particularly, its modulation by vaccination, this study comparatively analyses the time-course of expression of a series of erythrocytic genes in the liver during primary blood-stage infections of P. chabaudi taking a lethal outcome in non-vaccinated mice and, in parallel, during healing infections in vaccination-protected surviving mice. These data are discussed with respect to P. chabaudi-induced changes in peripheral blood previously analysed under identical experimental conditions as used here [8].

Methods

Mice

Experiments were performed with female Balb/c mice aged 10–12 weeks, which were delivered from the central animal facility of the University of Düsseldorf, where mice were bred under specified pathogen-free conditions. During infection experiments, mice were housed in plastic cages and received a standard diet (Woehrlin, Bad Salzuflen, Germany) and water ad libitum.

Protective vaccination

Vaccination was performed under identical experimental conditions as described previously [8]. The non-infectious vaccine, consisting of erythrocyte ghosts isolated from P. chabaudi-parasitized erythrocytes, was prepared as detailed previously [7, 8, 37]. These membrane ghosts were previously characterized to contain parasite-synthesized proteins [38, 39]. Approximately 106 ghosts suspended in 100 µl Freund’s complete adjuvant (FCA) were subcutaneously injected at weeks 3 and 1 before infection with P. chabaudi blood-stage malaria. In parallel, only FCA was injected in control mice.

Blood-stage malaria of P. chabaudi

Plasmodium chabaudi infections were maintained in outbred mice by weekly passages of infected blood under sterile conditions. The non-clonal line of P. chabaudi [40], resembles P. chabaudi AS clone in terms of restriction fragment length polymorphism as well as dihydrofolate reductase and cysteine protease sequence identities [41]. Moreover, the used line of P. chabaudi is self-healing as the AS clone, which is under control of genes of the H-2 complex and the non-H-2 background as well as sex and sex hormones of the infected mouse strain [42]. The Balb/c mice were infected with 106 P. chabaudi-infected erythrocytes. Parasitaemia was evaluated in Giemsa-stained smears from tail blood and erythrocytes were counted in a Neubauer chamber as described previously [8]. Both groups of vaccinated and non-vaccinated mice contained 4 ‘control’ mice, which were not sacrificed for liver sampling. All 4 mice in the non-vaccinated group succumbed to infection during crisis. In the vaccinated group, however, only 1 mouse succumbed to infection during crisis, whereas 3 mice survived the infection for at least 3 weeks, in accordance with previous results [8].

Liver sampling

To analyse hepatic gene expression during the course of P. chabaudi infections, both vaccinated (V) and non-vaccinated mice (N) were concomitantly infected, and groups of 3 mice were sacrificed at different time points of infections: upon infection on day 0 p.i. (groups Vd0 and Nd0), at early prepatency on day 1 p.i. (groups Vd1 and Nd1), at early patency on day 4 p.i. (groups Vd4 and Nd4) when parasitized erythrocytes begin to appear in peripheral blood with parasitaemias varying between 1–5%, at peak parasitaemia on day 8 p.i. (groups Vd8 and Nd8), and towards the end of the crisis phase on day 11 p.i. (groups Vd11 and Nd11) when parasitaemia declined to 5–1%. The course of parasitaemia was previously determined in mice sacrificed in the different groups [43], which corresponded to that in living infected mice under identical experimental conditions [8]. Livers were aseptically removed from sacrificed mice, rapidly frozen in liquid nitrogen and stored at − 80 °C until use.

RNA isolation

Frozen livers were individually ground in a mortar under liquid nitrogen. Total RNA was isolated from ‘pulverized’ aliquots of each individual liver by the standard Trizol protocol (Qiagen, Hilden, Germany), followed by an additional cleaning up with the miRNeasy Kit (Qiagen). The Agilent 2100 Bioanalyser platform (Agilent Technologies) was then used to check integrity and quality of RNA. The RIN values of all 30 liver RNA samples ranged between 8.7 and 9.1.

Cy3 labelling of RNA

Each RNA sample (equivalents of 100 ng) were used to produce Cy3-labeled cRNA using the Agilent Low Input Quick Amp Labeling Kit (Agilent Technologies) according to the manufacturer’s protocol. Yields of cRNA and dye-incorporation were determined with the ND-1000 Spectrophotometer (NanoDrop Technologies). The incorporations ranged between 18 and 23 fmol Cy3/ng cRNA.

Hybridization of Agilent mouse whole genome oligo microarrays

Agilent’s 8x60 K oligo microarrays (design number 028005) were used, which contained 8 arrays per slide. Each array displayed 39,430 Entrez Gene RNAs. Using the Agilent Gene Expression Hybridization Kit, hybridization was performed according to the Agilent processing protocol (Agilent technologies). Specifically, 600 ng Cy3-labelled fragmented cRNA was hybridized overnight at 65 °C using Agilent’s recommended hybridization chamber and oven. Finally, the microarrays were washed with the Agilent Gene Expression Wash Buffer 1 for 1 min at room temperature and then with the preheated Agilent Gene Expression Wash Buffer 2 at 37 °C for 1 min.

Scanning and analyses of microarrays

Microarrays were scanned with the Agilent’s Microarray Scanner System (Agilent Technologies). Microarray image files were processed with the Agilent Feature Extraction Software determining feature intensities (including background substraction), rejecting outliers and calculating statistical confidences of the array spots. All the 30 microarrays were normalized by the quantile method. When required, the expression variance was stabilized through the log2 transform. In-home functions developed in Matlab (MathWorks) were used to construct heat maps, the hierarchical clustering dendrograms (calculated using the unweighted pair group method with arithmetic mean and Euclidean distance measure), and the principal component analysis (PCA). The microarray data have been deposited at the NCBI’s Gene Expression Omnibus (GEO) database with accession number GSE129133.

Microarray-based analyses of genes involved in erythropoiesis

The used Agilent’s 8x60K microarrays contain probes for the following genes: Acyp1 (acylphosphatase 1 erythrocyte isozyme), Add2 (adducin2), Ahsp (α hemoglobin stabilizing protein), Ank1 (ankyrin1), Cldn13 (claudin13), Epb4.1 (Erythrocyte membrane protein band 4.1), Epb4.2 (Erythrocyte membrane protein 4.2), Epb4.9 (Erythrocyte membrane protein 4.9), Epo (erythropoietin), Epor (erythropoietin receptor), Ermap (erythroblast membrane associated protein), Gata1 (GATA-binding factor 1), Gfi1b (growth factor independent 1B transcriptional repressor), Gypa (glycophorin A), Kel (Kell blood group antigen), Klf1 (Krueppel-like factor 1), Rhag (Rh-associated glycoprotein), Rhd (Rh blood group D antigen), Slc4a1 (solute carrier family 4 member 1 = protein band 3), Spta1 (spectrin α erythrocytic 1), Sptb (Spectrin β erythrocytic), Tal1 (T-cell acute lymphocytic leukemia protein 1), and Tmod1 (tropomodulin 1). The expression profiles of these 23 genes were determined from the normalized microarrays prepared from the individual livers of both vaccinated and non-vaccinated mice during infections with P. chabaudi on days 0, 1, 4, 8, and 11 p.i. Gene expression levels, measured as light intensities above the normalization level, were given as mean ± SD as a dispersion metric in all figures. T-test was used to determine statistical significance of differences of gene expression levels between vaccinated and non-vaccinated mice both at a given day p.i. and during the intervals between the different sampling days p.i. The number of ‘*’ marks in the figures that appear over interval lines between sampling points or over the sampling points are the number of zeros after the decimal point of the p-values for the statistical significance of the difference in instant of the two series and of the difference between two intervals, respectively.

Quantitative real-time PCR of erythroid genes

High Capacity cDNA Reverse Transcription Kit (Life Technologies) and TaqMan mRNA assays (Life Technologies) were used to perform reverse transcription of mRNAs coding for the following proteins: RHD (assay ID: Mm00456910_m1), ERMAP (Mm_00469273_m1), GATA1 (Mm01352636_m1), SLC4A1 (Mm00441492_m1), CLDN13 (Mm00491038_s1), ADD2 (Mm00478923_m1), ACYP1 (Mm00481325_m1), EPB4.2 (Mm00469111_m1), EPB4.9 (Mm00469121_m1), EPOR (Mm01202755_m1), and EPO (Mm00833882_m1). The TaqMan® gene expression master mix (Life Technologies) was used for PCR reactions according to the instructions given by the manufacturer on a 7900HT real-time PCR System, as previously described [35]. Raw Ct values were calculated using the SDS software v.2.4 with GAPDH (Glycerinaldehyd-3-phosphat-Dehydrogenase) for normalization, and the comparative Ct method (2−ΔΔCt) was used to calculate fold change of expression [44]. Statistical significance of corresponding data sets between vaccinated and non-vaccinated mice were analysed with the two-tailed unpaired heteroskedastic Student´s T-test (* = p-value < 0.05).

Results

Global transcriptomics analyses reveal modifications by vaccination of malaria-induced hepatic gene expression on days 4 and 11 p.i

The effect of protective vaccination on the time course of erythroid gene expression in the liver induced by P. chabaudi blood-stage malaria was analysed using mouse whole genome 8x60K oligo microarrays. In toto, 30 microarrays were used for 30 livers prepared from the 10 different groups of vaccination-protected and un-protected non-vaccinated mice at the different phases of infections, i.e., upon infection on day 0 p.i., at early prepatency on day 1 p.i., at early patency on day 4 p.i., at peak parasitaemia on day 8 p.i., and towards the end of crisis on day 11 p.i.

Figure 1a shows the heatmap of gene expression profiles of those probes whose range or variation across all samples were at least 7 in the 10 different groups. In general, there is a good replicability among the three replicates of each analysed condition. The heatmap also shows a gradual change in gene expression across time both in non-vaccinated and vaccinated samples with an abrupt change in gene expression at peak parasitaemia on day 8 p.i. As the heatmap, the hierarchical clustering of the 30 different individual samples, shown as a dendogram in Fig. 1b, also reveals a good replicability among the three replicates of each analysed condition and the abrupt change in gene expression on day 8 p.i. There are two main branches splitting the samples taken on days 0, 1, and 4 p.i. (red branches in Fig. 1b) from the samples taken on days 8 and 11 p.i. (blue branches in Fig. 1b). The principal component analysis (PCA) of gene expression (Fig. 1c) shows that the 1st principal component (PC1) accounts for 40% of the gene expression variability and the 2nd PC2 captures 10% of the variability. The 1st principal axis, that is the most informative, separates between those liver samples taken before day 8 p.i. (positive coordinates) and those taken after day 8 p.i. samples (negative coordinates).

Fig. 1
figure 1

Global expression profile of mRNAs in the liver of non-vaccinated (N) and vaccination-protected mice (V). a RNA was isolated from individual livers prepared on different days (d0, d1, d4, d8, d11) during infection with P. chabaudi blood-stage malaria. Gene expression levels for the individual livers were determined by microarrays and hierarchically clustered. Log2 transformed expression levels range from 0 to 18 as indicated at the top with blue (low expression) and red (high expression), respectively. b Hierarchical clustering of liver samples prepared from vaccination-protected (V) and non-vaccinated mice (N) during different phases of infections with P. chabaudi malaria on the indicated days (d0, d1, d4, d8, d11). Suffices indicate the three different livers taken per time point. The correlation metric and the average linkage method were used to construct the dendogram. c Principal Component Analysis (PCA) of gene expression data. The 1st principal component (PC1) captures 40% of the gene expression variability and the 2nd PC2 captures 10% of the variability. The Nd0 populations are depicted by blue spheres, the Nd1 by cyan spheres, the Nd4 by green spheres, the Nd8 by magenta spheres, the Nd11 by red spheres, the Vd0 by blue icosahedra, the Vd1 by cyan icosahedra, the Vd4 by green icosahedra, the Vd8 by magenta icosahedra and the Vd11 by red icosahedra

Collectively, the global transcriptomics analysis results indicate (i) that constitutive expression of the vast majority of hepatic genes is not essentially changed by protective vaccination, (ii) that P. chabaudi infections induce changes in hepatic RNA expression, being usually highest at peak parasitaemia on day 8 p.i. and towards the end of crisis on day 11 p.i., and (iii) that the malaria-induced changes in hepatic gene expression are modified by protective vaccination, particularly at early patency on day 4 p.i. and towards the end of crisis on day 11 p.i.

Erythropoiesis-involved genes are expressed in the liver of non-infected mice

The heatmap in Fig. 2 shows the expression profiles of the selected 23 genes known to encode important non-haemoglobin constituents of erythrocytes and the EPO encoding gene known to stimulate erythropoiesis. The numbers in the heatmap indicate the probe-detected expression levels of genes which were log2 transformed for variance stabilization. In the liver of non-vaccinated mice on day 0 p.i., the expression of a given gene is similar among the 3 microarrays prepared from the 3 mice (Nd0 in Fig. 2) and also with the corresponding probes on the microarrays of the 3 vaccinated mice on day 0 p.i. (Vd0 in Fig. 2). However, the 23 genes reveal varying constitutive expression levels, which, however, are approximately the same at both Nd0 and Vd0. For instance, low constitutive expression levels reveal the genes Epb4.2, Epb4.9, Rhag, Ermap, Spta, and Sptb at both Nd0 and Vd0, whereas higher expression levels are found for the genes Epor, Slc4a1, Gata1, Klf1, and Tal1 in both vaccinated and non-vaccinated mice (Fig. 2). This supports the view that constitutive expression of erythroid genes in non-infected mice is not essentially affected by vaccination.

Fig. 2
figure 2

Heatmap of hepatic expression profiles of erythropoiesis-involved genes. RNA was isolated from the individual livers prepared from non-vaccinated (N) and vaccinated (V) mice infected with P. chabaudi on days 0, 1, 4, 8, and 11 p.i. Log2 transformed expression levels range from 0 to 18; the higher is the expression the more red is the colour

Erythroid genes respond to infections with increasing expressions in non-vaccinated mice

The heatmap in Fig. 2 also shows that blood-stage infections of P. chabaudi malaria induce increasing expressions of the 23 erythroid genes in the liver, but with differences among vaccinated and non-vaccinated mice. To make these differences clearer and better comparable, the expression profiles were plotted in a linear scale as time series with the corresponding trajectories presented in Figs. 3, 4, 5, 6.

Fig. 3
figure 3

Time courses of hepatic expression of erythroid genes encoding proteins associated with the membrane and extracellular surface of erythroid cells. RNA was isolated from individual livers prepared from vaccination-protected (V, red) and non-vaccinated mice (N, blue) infected with P. chabaudi blood-stage malaria on days 0, 1, 4, 8, and 11 p.i. Data are plotted in linear scale. Means of 3 microarrays are given ± SD. The number of ‘*’ marks that appear over interval lines between sampling points and over the sampling points are the number of zeros after the decimal point of the p-values for the statistical significance of the difference in instant of the two series and of the difference between two intervals, respectively

Fig. 4
figure 4

Expression trajectories of erythroid genes coding for proteins associated with the inner surface of the erythroid plasma membrane in the liver of vaccinated and non-vaccinated mice. RNA was isolated from individual livers prepared from vaccination-protected (V, red) and non-vaccinated mice (N, blue) infected with P. chabaudi blood-stage malaria on days 0, 1, 4, 8, and 11 p.i. Data are plotted in linear scale. Means of 3 microarrays are given + SD. The number of ‘*’ marks indicate significances as detailed in legend to Fig. 3

Fig. 5
figure 5

Expression trajectories of genes encoding erythropoiesis-involved transcription factors and other internal erythroid proteins. RNA was isolated from individual livers prepared from vaccination-protected (V, red) and non-vaccinated mice (N, blue) infected with P. chabaudi blood-stage malaria on days 0, 1, 4, 8, and 11 p.i. Data are plotted in linear scale. Means of 3 microarrays are given ± SD. The number of ‘*’ marks indicate significances as detailed in legend to Fig. 3

Fig. 6
figure 6

Time course of expression of Epor and Epo in the liver of vaccination-protected and non-protected mice. RNA was isolated from individual livers prepared from vaccination-protected (V, red) and non-vaccinated mice (N, blue) infected with P. chabaudi blood-stage malaria on days 0, 1, 4, 8, and 11 p.i. The upper curves show microarray data, the lower curves qPCR data. The number of ‘*’ marks in microarray data indicate significances as detailed in legend to Fig. 3. ‘*’ in qPCR data indicate significant differences between vaccinated and non-vaccinated mice (p < 0.05)

Blood-stage infections with P. chabaudi induce only a slight increase in the expression of all the 23 erythropoiesis-involved genes between days 0 p.i. and 4 p.i. Thereafter, an abrupt increase in expression is observed at peak parasitaemia on day 8 p.i., and the increase even continues reaching still higher mRNA expression levels towards the end of the crisis phase on day 11 p.i. Among genes encoding membrane-associated proteins [45, 46], the gene Slc4a1 coding for the major integral multi-pass membrane protein band 3 and the gene Gypa encoding the major one-pass membrane protein of the Gyp family reach maximal mRNA expression levels of approximately 180 and 50 above the normalization level, respectively (Fig. 3). The genes Kel and Rhd coding for blood groups and Rhag coding for the Rh-blood group associated transmembrane glycoprotein reveal maximal expressions of approximately 100, 120, and 90, respectively, on day 11 p.i. (Fig. 3). Ermap known to encode a protein associated with the outer surface of erythroblasts displays maximal expression of approximately 90 on day 11 p.i. A very similar time-course of expression with a maximum level of approximately 100 reveals Cldn13 (Fig. 3), whose encoded protein has been previously suggested to be associated with erythroblastic islands [35]. Lower maximal expressions on day 11 p.i. exhibit those genes, whose encoded proteins are known to be associated with the cytoskeletal meshwork on the inner surface of the erythrocyte plasma membrane (Fig. 4). Among them are Spta, Sptb, Epb4.1, Epb4.2, Epb4.9, and Tmod1 [45, 46], whose maximal expressed mRNA levels vary between 15 and 50 above the normalization level, respectively. Only the genes Add2 involved in the assembly of the spectrin–actin cytoskeletal network and Ank1 involved in the linkage of integral membrane proteins to the underlying cytoskeletal meshwork display higher expression levels between 80 and 70, respectively (Fig. 4).

The genes Gata1, Gfi1b, Klf1, and Tal1 encode transcription factors which are known to be critically involved in erythropoiesis [47,48,49]. These genes also reach high expressions between approximately 80 and 110 above the normalization level towards the end of the crisis phase (Fig. 5). Among all erythroid genes responding to malaria, Ahsp, encoding the α haemoglobin stabilizing protein, exhibit the highest mRNA expression of approximately 190 towards the end of the crisis phase (Fig. 5). However, a maximal expression level of only approximately 35 reveals the gene Acyp1 encoding the erythrocytic acylphosphatase.

Erythropoiesis, in particular erythroblastosis, is critically dependent on EPO signaling through its receptor EPOR [23, 24]. Epor and Epo display a time-course of mRNA expression, which differs from that determined for the other erythroid genes in two aspects (Fig. 6). Epor reveals a sharp decline in its relative expression level from about 75 on day 0 p.i. to about 25 on day 1 p.i.., before it continuously increases almost linearly reaching its maximal level of about 120 towards the end of crisis on day 11 p.i. By contrast, Epo expression is impaired during the first 4 days of infection, then it sharply increases reaching its maximum expression of approximately 8 at peak parasitaemia on day 8 p.i., before declining to a level of about 3 towards the end of the crisis phase on day 11 p.i.

Expression of erythroid genes is accelerated in the liver of vaccination-protected mice

In contrast to un-protected non-vaccinated mice, the expression of the vast majority of erythroid genes in the liver of vaccination-protected mice is accelerated during infections, as evidenced by three facts. First, the erythroid genes are significantly much higher expressed at early patency on day 4 p.i. than the corresponding genes in the liver of non-vaccinated mice (cf. Figs. 3, 4, 5, 6). This significant higher expression is confirmed when the corresponding genes are directly compared between vaccinated and non-vaccinated mice on day 4 p.i. or when the increases in expression levels between day 1 p.i. and day 4 p.i. is compared in vaccinated mice vs non-vaccinated mice. The only exception is Epb4.1 (Fig. 4): its expression is significantly higher on day 1 p.i. in vaccinated mice, then it decreases to approximately the same low level as that of non-vaccinated mice on day 4 p.i., before it increases again reaching maximal expression at peak parasitaemia on day 8 p.i., which is about the same as that towards the end of the crisis phase on day 11 p.i. Secondly, the majority of erythroid genes reach their maximal expression levels already at peak parasitaemia on day 8 p.i. and keep about the same level during crisis, in contrast to non-vaccinated mice in which the corresponding genes reach their maximal expression levels on day 11 p.i. Thirdly, the final expression levels on day 11 p.i. are mostly lower with lower standard deviations in vaccination-protected mice than those of the corresponding genes in the liver of non-vaccinated un-protected mice (cf. Figs. 3, 4, 5, 6). A few genes in the liver of vaccinated mice, which still show a minor increase of mRNA expressions after peak parasitaemia on day 8 p.i., slow down their expressions towards the end of the crisis phase on day 11 p.i. Some genes, as e.g. Rhag, Epb4.1, Sptb, Tmod1 and Acyp1, have even significantly declined their expression levels on day 11 p.i.

Remarkably, the time course of expression of Epor in the liver of vaccinated mice is very similar to that of other genes proceeding from a level of about 25 on day 1 p.i. and reaches maximal levels of almost 100 on days 8 and 11 p.i. (Fig. 6). Also, the increase in expression of Epor between day 1 p.i. and day 4 p.i. as well as the expression on day 4 p.i. is significantly higher in vaccinated mice than in non-vaccinated mice. Remarkably, the time course of Epor expression significantly differs to that of Epo. Indeed, Epo expression is delayed during the first 4 days of infections, i.e., Epo is expressed at approximately the same very low level between day 0 p.i. and 4 p.i. in vaccinated mice as in non-vaccinated mice. Moreover, the maximal levels of Epo expression at peak parasitaemia on day 8 p.i. are significantly higher in the liver of vaccinated mice than in non-vaccinated mice (Fig. 6), though vaccination decreases maximum parasitaemia by approximately 30% in comparison with non-vaccinated mice [8, 43].

Quantitative PCR validates the response of erythroid genes to infections

Quantitative PCR (qPCR) was used to re-examine, in both non-vaccinated and vaccinated mice, the time courses of expression of some arbitrarily selected erythroid genes, whose expression was identified by microarrays to respond to P. chabaudi blood-stage malaria. The data are summarized in Fig. 7. The trajectories of Ermap expression in both vaccinated and non-vaccinated mice during infection take a similar course as those detected by the microarray probes (Fig. 3). Moreover, the qPCR-determined trajectories for expression of Gata1, Add2, Slc4a1, Rhd, and Cldn13 take a similar course as those determined by microarrays (cf. Fig. 7 with Figs. 3, 4, 5). In particular, there a higher increase in expression was observed at early patency on day 4 p.i. and a lower expression towards the end of the crisis phase on day 11 p.i. in vaccinated mice than in non-vaccinated mice. Furthermore, correspondence largely exists between microarray and qPCR data with respect to the tendency of increasing expression for Epb4.2, Epb4.9, and Acyp1 (Fig. 7). Similarly to microarray data, the 9 genes examined by qPCR reveal maximal expression levels in the liver of vaccinated mice at peak parasitaemia on day 8 p.i., whereas, in the liver of non-vaccinated mice, maximal expression of the majority of these genes is reached towards the end of crisis phase on day 11 p.i. Furthermore, the expressions of Epor and Epo measured by qPCR take very similar courses in vaccinated and non-vaccinated mice as those determined by microarrays (Fig. 6).

Fig. 7
figure 7

Quantitative PCR of mRNAs in the liver of vaccinated and non-vaccinated Balb/c mice. RNA was isolated from individual livers prepared from mice infected with P. chabaudi on different days p.i. Duplicate determinations were performed with liver aliquots from 3 different mice, and means are given ± SD. ‘*’ indicate significant differences between vaccinated and non-vaccinated mice (p < 0.05)

Discussion

This study provides evidence that vaccination, using a non-infectious vaccine consisting of erythrocyte ghosts isolated from P. chabaudi-infected erythrocytes, accelerates expression of erythroid genes in the liver induced by blood-stage malaria of P. chabaudi. Indeed, these genes encompass Slc4a1 and Gypa encoding band 3 and Glycophorin A, major integral membrane proteins of red blood cells; Rhd, Kel and Rhag coding for blood group antigens and Rh-associated antigen glycoprotein exposed on the outer membrane surface of red blood cells; Ermap and Cldn1 encoding presumably proteins associated with the outer surface of erythroblasts [25]; Add2 and Ank1 coding for proteins linking integral membrane proteins with peripheral skeletal proteins located on the inner surface of erythroid cells, as e.g., Epb4.2, Epb4.9, Spta, and Sptb [45, 46]; Acyp1 and Ahsp encoding proteins within erythroid cells; and even Gata1, Gfi1b, Tal1 and Klf1 coding for transcription factors critically involved in erythropoiesis [47,48,49]. Expression of these genes takes an accelerated time-course in surviving vaccinated mice, evidenced as it is significantly higher at early patency on day 4 p.i. and as it is often more slowed down or even lower towards the end of the crisis phase on day 11 p.i. than those expression time-courses taken by the corresponding genes during lethal infections in non-vaccinated mice. Thus, the results indicate that the liver responds to blood-stage malaria with extramedullary erythropoiesis, especially erythroblastosis in the liver, and erythroblastosis is obviously accelerated by vaccination.

Erythroblastosis is critically dependent on the interplay of EPO and EPOR, respectively [23, 24]. In accordance, extramedullary erythroblasts in the liver apparently display EPO sensitivity, evidenced as Epor expression taking approximately the same time-course as that taken by the other erythroid genes in vaccination-protected mice. Moreover, it is remarkable that Epo expression also occurs in the liver during extramedullary erythroblastosis. Indeed, there is evidence that EPO is not only produced by kidneys, but also by other cells including hepatocytes [23, 50]. In contrast to Epor and the other examined erythroid genes, however, hepatic expression of Epo is not accelerated during extramedullary erythroblastosis, rather it is delayed at least for the first 4 days of infections. Maximal Epo expression is reached at peak parasitaemia on day 8 p.i. both in vaccinated and non-vaccinated mice, but is much higher in vaccinated mice than in non-vaccinated mice, though, incidentally, non-vaccinated mice exhibit a higher peak parasitaemia by about 30% than vaccination-protected mice. The data, therefore, suggest that EPOR-mediated EPO sensitivity by hepatic erythroblasts precedes EPO production in the liver, and that malaria-induced erythroblastosis in the liver may require hepatic EPO particularly at peak parasitaemia. However, it cannot be excluded that hepatic EPO may be also involved in the stimulation of other processes in the liver, which are important to survive blood-stage malaria. For instance, EPO has been recently described to recruit Ly6Chi monocytes in response to liver injury [51] and to stimulate proliferation and phagocytotic activity of Kupffer cells—the liver-resident macrophages known for its capacity of erythrophagocytosing Plasmodium-parasitized red blood cells [11, 12, 14,15,16]. Liver injury does also occur during blood-stage malaria in mice and even humans [11, 52]. Moreover, EPO has been reported to inhibit inflammation and gluconeogenesis in the liver [53]. Attenuated inflammation of the liver and decreased glucose blood levels after crisis was also found in vaccination-protected mice infected with P. chabaudi [8, 11].

Extramedullary erythroblastosis in the liver of vaccination-protected mice may be reasonably assumed to contribute to overcome malaria-induced anemia and replenishment of peripheral erythrocytes, whose number is increasingly declining with progressing P. chabaudi blood-stage infections. This view is supported by previous results obtained under identical experimental conditions as used here [8]. Indeed, P. chabaudi-parasitized erythrocytes begin to appear in peripheral blood of both vaccinated and non-vaccinated mice on day 4 p.i., and peripheral red blood cells in both vaccinated and non-vaccinated mice begin to decrease after day 5 p.i., i.e., from approximately 9 × 106/µl on day 5 p.i. to approximately 5.5 × 106/µl on day 6 p.i. The lowest levels of approximately 1.8 × 106/µl peripheral red blood cells were reached at peak parasitaemia on day 8 p.i. [8]. This very low level of peripheral erythrocytes remains in non-vaccinated mice until deceasing at latest at the end of the crisis phase on approximately day 11 p.i., though there is observable a slight increase of peripheral reticulocytes at that time. In vaccinated mice, however, the increase in peripheral reticulocytes is accelerated and already begins about 1–2 days earlier than in non-vaccinated mice reaching higher levels at the end of crisis on day 11 p.i. [8]. This earlier appearance of peripheral reticulocytes may be due to an accelerated terminal erythroid differentiation in vaccination-protected mice, i.e., final denucleation of erythroblasts and release of denucleated reticulocytes into liver sinusoids and blood vessels, respectively, begin 1–2 days earlier in vaccination-protected mice than in unprotected non-vaccinated mice.

The accelerated extramedullary erythropoiesis in the liver of vaccination-protected mice may be also viewed as an important contribution of the liver to the host defense system against blood-stage malaria. At the end of crisis on day 11 p.i., for example, when parasitaemia has dramatically declined to about 1–5% in both vaccinated and non-vaccinated mice, there is accumulated a large excess of peripheral reticulocytes, representing approximately 90% of all peripheral red blood cells, only in vaccination protected mice [8]. This suggests that, at least at that time-point, vaccinated mice are less vulnerable to blood-stage infections, since reticulocytes—in contrast to erythrocytes—are not preferred as host cells by P. chabaudi [54]. A major reason for the host cell specificity of P. chabaudi for erythrocytes may be that reticulocytes differ from erythrocytes with respect to composition, organization and properties of their plasma membranes [45, 46]. Even erythrocytes during their aging are subjected to modifications in membrane components. For instance, there is evidence for an increase in the ratio of band 4.1a:4.1b during erythrocyte aging [55]. These hub proteins are not only involved in the organizational cross-linking of the spectrin network to the inner surface of red cell plasma membranes [56], but also favour the formation of elliptotic red cells, which are known to hinder red cell invasion by Plasmodium parasites [57, 58]. In accordance, there has been previously described a decrease of band 4.1a in erythrocytes of C57BL/10 mice after survival of P. chabaudi blood-stage infections [59]. Remarkably, the present study shows that the time course of Epb4.1 expression differs from that of all the other examined erythroid genes insofar as there is dramatic decline on day 4 p.i., when all other erythroid genes reveal an increased expression. Furthermore, there is ample evidence for other abnormalities, deficiencies and/or mutations of erythrocytes in their membrane or internal constituents which restrict invasion and/or internal survival of malaria parasites, respectively [60, 61]. It is therefore attractive to speculate that the accelerated extramedullary erythroblastosis in the liver of vaccination-protected mice ultimately leads, by the end of crisis phase, to an earlier transient excess of peripheral reticulocytes, being unfavourable for parasite survival, thus resulting in a dramatic decline of potential erythrocyte host cells for P. chabaudi parasites. This temporary impairment of parasite multiplication may concomitantly allow the evolvement of immune mechanisms finally contributing to resolve blood-stage infections of P. chabaudi.

Conclusion

This study indicates that vaccination accelerates extramedullary erythroblastosis, induced in the liver by blood-stage malaria of P. chabaudi, for 1–2 days. This presumably contributes to earlier replenishment of peripheral red blood cells by liver-derived reticulocytes. This in turn is speculated to favour final survival of otherwise lethal blood-stage malaria, since reticulocytes—in contrast to mature erythrocytes—are known to be not preferred as host cells by P. chabaudi.

Availability of data and materials

The datasets used and/or analysed during the current study are available from the corresponding author on reasonable request. The microarray data have been deposited at the NCBI’s Gene Expression Omnibus (GEO) database with accession number GSE129133.

Abbreviations

ACYP1:

acylphosphatase 1 erythrocyte isozyme

ADD2:

adducin2

AHSP:

α haemoglobin stabilizing protein

ANK1:

ankyrin1

CLDN13:

claudin13

CFU-e:

colony-forming unit-erythroid

EPB4.1:

Erythrocyte membrane protein band 4.1

EPB4.2:

Erythrocyte membrane protein 4.2

EPB4.9:

Erythrocyte membrane protein 4.9

EPO:

erythropoietin

EPOR:

erythropoietin receptor

ERMAP:

erythroblast membrane associated protein

FCA:

Freund’s complete adjuvant

GAPDH:

glycerinaldehyd-3-phosphat-Dehydrogenase

GATA1:

GATA-binding factor 1

GFI1B:

growth factor independent 1B transcriptional repressor

GYPA:

glycophorin A

KEL:

Kell blood group antigen

KLF1:

Krueppel-like factor 1

PCA:

principal component analysis

RHAG:

Rh-associated glycoprotein

RHD:

Rh blood group D antigen

SLC4A1:

solute carrier family 4 member 1

SPTA1:

spectrin α erythrocytic 1

SPTB:

spectrin β erythrocytic

TAL1:

T cell acute lymphocytic leukemia protein 1

TMOD1:

tropomodulin 1

References

  1. WHO. World malaria report 2018. Geneva: World Health Organization; 2018.

    Google Scholar 

  2. Gosling R, von Seidlein L. The future of the RTS, S/AS01 malaria vaccine. an alternative development plan. PLoS Med. 2016;13:e1001994.

    Article  PubMed  PubMed Central  Google Scholar 

  3. Ashley EA, Pyae Phyo A, Woodrow CJ. Malaria. Lancet. 2018;391:1608–21.

    Article  PubMed  Google Scholar 

  4. Draper SJ, Sack BK, Richter King C, Nielsen CN, Rayner JC, Higgins MK, et al. Malaria vaccines: recent advances and new horizons. Cell Host Microbe. 2018;24:43–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Longley R, Smith C, Fortin A, Berghout J, McMorran B, Burgio G, et al. Host resistance to malaria: using mouse models to explore the host response. Mamm Genome. 2011;22:32–42.

    Article  CAS  PubMed  Google Scholar 

  6. Stephens R, Culleton RL, Lamb TJ. The contribution of Plasmodium chabaudi to our understanding of malaria. Trends Parasitol. 2012;28:73–82.

    Article  PubMed  Google Scholar 

  7. Wunderlich F, Brenner H, Helwig M. Plasmodium chabaudi malaria: protective immunization with surface membranes of erythrocytes infected with Plasmodium chabaudi. Infect Immun. 1988;56:3326–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Krücken J, Delic D, Pauen H, Wojtalla A, El-Khadragy M, Dkhil MA, et al. Augmented particle trapping and attenuated inflammation in the liver by protective vaccination against Plasmodium chabaudi malaria. Malar J. 2009;8:54.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  9. Del Portillo HA, Ferrer M, Brugat T, Martin-Jaular L, Langhorne J, Lacerda MVG. The role of the spleen in malaria. Cell Microbiol. 2012;14:343–55.

    Article  PubMed  CAS  Google Scholar 

  10. White NJ. Malaria parasite clearence. Malar J. 2017;16:88.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  11. Wunderlich F, Al-Quraishy S, Dkhil M. Liver-inherent immune system: its role in blood-stage malaria. Front Microbiol. 2014;5:559.

    Article  PubMed  PubMed Central  Google Scholar 

  12. Terpstra V, Berkel TJ. Scavenger receptors on liver Kupffer cells mediate the in vitro uptake of oxidatively damaged red blood cells in mice. Blood. 2000;15:2157–63.

    Article  Google Scholar 

  13. Krücken J, Mehnert LI, Pauen H, Woijtalla A, El-Khadragy M. Massive destruction of malaria-parasitized red blood cells despite spleen closure. Infect Immun. 2005;73:6390–8.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. Delic D, Warskulat U, Borsch E, Al-Qahtani S, Al-Guraishy S, Häussinger D, et al. Loss of ability to self-heal malaria upon taurine transporter deletion. Infect Immun. 2010;78:1642–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Lee SJ, Park SY, Jung MY, Bae SM, Kim IS. Mechanism for phosphatidylserine-dependent erythrophagocytosis in mouse liver. Blood. 2011;117:5215–23.

    Article  CAS  PubMed  Google Scholar 

  16. Theurl I, Hilgendorf I, Nairz N, Tymoszuk P, Haschka D, Asshoff M, et al. On-demand erythrocyte disposal and iron recycling requires transient macrophages in the liver. Nat Med. 2016;22:945–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Dzierzak E, Philipsen S. Erythropoiesis: development and differentiation. Cold Spring Harb Perspect Med. 2013;3:a011601.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  18. An X, Schulz VP, Mohandas N, Gallagher PG. Human and murine erythropoiesis. Curr Opin Hematol. 2015;22:206–11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Zivot A, Lipton JM, Naria A, Blanc L. Erythropoiesis: insights into pathophysiology and treatments in 2017. Mol Med. 2017;24:11.

    Article  CAS  Google Scholar 

  20. Klei TRL, Meinderts SM, van den Berg TK, van Bruggen A. From the cradle to the grave: the role of macrophages in erythropoiesis and erythrophagocytosis. Front Immunol. 2017;8:73.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  21. Seu KG, Papoin J, Fessler R, Horn J, Huang G, Mohandas N, et al. Unraveling macrophage heterogeneity in erythroblastic islands. Front Immunol. 2017;8:1140.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  22. Moras M, Lefevre SD, Ostuni MA. From Erythroblasts to mature red blood cells: organelle clearance in mammals. Front Physiol. 2017;8:1076.

    Article  PubMed  PubMed Central  Google Scholar 

  23. Elliott S, Sinclair AM. The effect of erythropoietin on normal and neoplastic cells. Biologics. 2012;6:163–89.

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Wang J, Hayashi Y, Yokota A, Xu Z, Zhang Y, Huang R, et al. Expansion of EPOR-negative macrohpages besides erythroblasts by elevated EPOR signaling in erythrocytosis mouse models. Haematologica. 2018;103:40–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Chang KH, Stevenson MM. Malarial anaemia: mechanisms and implications of insufficient erythropoiesis during blood-stage malaria. Int J Parasitol. 2004;34:1501–16.

    Article  CAS  PubMed  Google Scholar 

  26. Chang KH, Tam M, Stevenson MM. Modulation of the course and outcome of blood-stage malaria by erythropoietin-induced reticulocytosis. J Infect Dis. 2004;189:735–43.

    Article  PubMed  Google Scholar 

  27. Thawani N, Tam M, Bellemare MJ, Bohle DS, Olivier M, de Souza JB, et al. Plasmodium products contribute to severe malarial anemia by inhibiting erythropoietin-induced proliferation of erythroid precursors. J Infect Dis. 2014;209:140–9.

    Article  CAS  PubMed  Google Scholar 

  28. Li LX, Benoun JM, Weiskopf K, Garcia KC, McSorley SJ. Salmonella infection enhances erythropoietin production by the kidney and liver, which correlates with elevated bacterial burden. Infect Immun. 2016;84:2833–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Otsuka H, Takito J, Endo Y, Yagi H, Soeta H, Yanagisawa N, et al. The expression of embryonic globin mRNA in a severely anemic mouse model induced by treament with nitrogen-containing bisphosphonate. Hematology. 2016;16:4.

    Google Scholar 

  30. Alamo IG, Kannan KG, Loftus TJ, Ramos H, Efron PA, Mohr AM. Severe trauma and chronic stress activates extramedullary erythropoiesis. J Trauma Acute Care Surg. 2017;83:144–50.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Myllimäki MN, Määttä J, Dimova EY, Izzi V, Väisänen T, Myllyharju J, et al. Notch downregulation and extramedullary erythropoiesis in hypoxia inducible factor prolyl 4-hydroxylase 2-deficient mice. Mol Cell Biol. 2017;37:e00529.

    Google Scholar 

  32. Yap GS, Stevenson MM. Plasmodium chabaudi AS: erythropoietic responses during infection in resistant and susceptible mice. Exp Parasitol. 1992;75:340–52.

    Article  CAS  PubMed  Google Scholar 

  33. Abo T, Sekikawa H. Extrathymic T cells in malaria protection, including evidence for the onset of erythropoiesis in the liver during infection. Arch Histol Cytol. 2002;65:127–32.

    Article  PubMed  Google Scholar 

  34. Halder RC, Abe T, Mannoor MK, Morshed SR, Ariyasinghe A, Watanabe H, et al. Onset of hepatic erythropoiesis after malarial infection in mice. Parasitol Int. 2003;52:259–68.

    Article  PubMed  Google Scholar 

  35. Al-Quraishy SA, Dkhil MA, Abdel-Baki AA, Delic D, Wunderlich F. Protective vaccination against blood-stage malaria of Plasmodium chabaudi: differential gene expression in the liver of Balb/c mice toward the end of crisis phase. Front Microbiol. 2016;7:1087.

    Article  PubMed  PubMed Central  Google Scholar 

  36. Al-Quraishy S, Dkhil MA, Al-Shaebi EM, Abdel-Baki AS, Araúzo-Bravo MJ, Delic D, et al. Gene expression of the liver of vaccine-protected mice in response to early patent infections of Plasmodium chabaudi blood-stage malaria. Malar J. 2018;17:215.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Wunderlich F, Helwig M, Schillinger G, Vial H, Philippot J, Speth V. Isolation and characterization of parasites and host cell ghosts from erythrocytes infected with Plasmodium chabaudi. Mol Biochem Parasitol. 1987;23:103–15.

    Article  CAS  PubMed  Google Scholar 

  38. Wunderlich F, Helwig M, Schillinger G, Speth V. Cryptic disposition of antigenic parasite proteins in plasma membranes of erythrocytes infected with Plasmodium chabaudi. Mol Biochem Parasitol. 1988;30:55–65.

    Article  CAS  PubMed  Google Scholar 

  39. Wunderlich F, Helwig M, Schillinger G, Speth V, Wiser MF. Expression of the parasite protein Pc90 in plasma membranes of erythrocytes infected with Plasmodium chabaudi. Eur J Cell Biol. 1988;47:157–64.

    CAS  PubMed  Google Scholar 

  40. Wunderlich F, Stuebig H, Koenigk E. Development of Plasmodium chabaudi in mouse red blood cells: structural properties of the host and parasite membranes. J Protozool. 1982;29:60–6.

    Article  CAS  PubMed  Google Scholar 

  41. Wunderlich F, Dkhil M, Mehnert L, Braun J, El-Khadragy M, Borsch E, et al. Testosterone responsiveness of spleen and liver in female lymphotoxin beta receptor-deficient mice resistant to blood-stage malaria. Microbes Infect. 2005;7:399–409.

    Article  CAS  PubMed  Google Scholar 

  42. Wunderlich F, Mossmann H, Helwig M, Schillinger G. Resistance to Plasmodium chabaudi in B10 mice: influence of the H-2 complex and testosterone. Infect Immun. 1988;56:2400–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Dkhil MA, Al-Quraishy SA, Abdel-Baki AS, Delic D, Wunderlich F. Differential miRNA expression in the liver of Balb/c mice protected by vaccination during crisis of Plasmodium chabaudi blood-stage malaria. Front Microbiol. 2017;7:2155.

    Article  PubMed  PubMed Central  Google Scholar 

  44. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 2002;25:402–8.

    Article  CAS  Google Scholar 

  45. Liu J, Guo X, Mohandas N, Chasis JA, An X. Membrane remodeling during reticulocyte maturation. Blood. 2010;8:241182.

    Google Scholar 

  46. Lux SE 4th. Anatomy of the red cell membrane skeleton: unanswered questions. Blood. 2016;127:187–99.

    Article  CAS  PubMed  Google Scholar 

  47. Love PE, Warzecha C, Li L. Lbd1 complexes: the new master regulators of erythroid gene transcription. Trends Genet. 2014;30:1–9.

    Article  CAS  PubMed  Google Scholar 

  48. Vassen L, Beauchemin H, Lemsaddek W, Krongold J, Trudel M, Moroy T. Growth factor independence 1b (gfi1b) is important for the maturation of erythroid cells and the regulation of embryonic globin expression. PLoS ONE. 2014;9:e96636.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  49. Barbarani G, Fugazza C, Strouboulis J, Ronchi AE. The pleiotropic effects of GATA1 and KLF1 in physiological erythropoiesis and in dyserythropoietic disorders. Front Physiol. 2019;10:91.

    Article  PubMed  PubMed Central  Google Scholar 

  50. Pinto JP, Ribeiro S, Pontes H, Thowfeequ S, Tosh D, Caevalho F, et al. Erythropoietin mediates hepcidin expression in hepatocytes through EPOR signaling and regulation of C/EBPalpha. Blood. 2008;2008(111):5727–33.

    Article  CAS  Google Scholar 

  51. Gilboa D, Haim-Ohana Y, Deshet-Unger N, Ben-Califa N, Hiram-Bab S, Reuveni D, et al. Erythropoietin enhances Kupffer cell number and activity in the challenged liver. Sci Rep. 2017;7:10379.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  52. Rupani AB, Amarapurkar AD. Hepatic changes in fatal malaria: an emerging problem. Ann Trop Med Parasitol. 2009;103:119–27.

    Article  CAS  PubMed  Google Scholar 

  53. Meng R, Zhu D, Bi Y, Yang D, Wang Y. Erythropoietin inhibits gluconeogenesis and inflammation in the liver and improves glucose tolerance in high-fat diet-fed mice. PLoS ONE. 2013;8:e53557.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Ott KJ. Influence of reticulocytosis on the course of infection of Plasmodium chabaudi and P. berghei. J Eukaryot Microbiol. 1968;15:365–9.

    CAS  Google Scholar 

  55. Mueller TJ, Jackson CW, Dockter ME, Morrison M. Membrane skeletal alterations during in vivo mouse red cell aging—increase in the band4.1a:4.1b ratio. J Clin Invest. 1987;79:492–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Baines AJ, Lu HC, Bennet PM. The protein 4.1 family: hub proteins in animals for organizing membrane proteins. Biochim Biophys Acta. 2014;1838:605–19.

    Article  CAS  PubMed  Google Scholar 

  57. Conboy JG, Chasis JA, Winardi R, Tchernia G, Kan YW, Mohandas N. An isoform-specific mutation in the protein 4.1 gene results in hereditary elliptocytosis and complete deficiency of protein 4.1 in erythrocytes but not in nonerythroid cells. J Clin Invest. 1993;91:77–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Chishti AH, Palek J, Fisher D, Maalouf GJ, Liu SC. Reduced invasion and growth of Plasmodium falciparum into elliptocytic red blood cells with a combined deficiency of protein 4.1, glycophorin C, and p55. Blood. 1996;87:3462–9.

    Article  CAS  PubMed  Google Scholar 

  59. Wunderlich F, Helwig M. Plasmodium chabaudi malaria: red blood cells with altered membrane proteins in immune mice. Eur J Cell Biol. 1987;43:499–500.

    CAS  PubMed  Google Scholar 

  60. Lelliot PM, McMorran BJ, Foote SJ, Burgio G. The influence of host genetics on erythrocytes and malaria infection: is there therapeutic potential? Malar J. 2015;14:289.

    Article  Google Scholar 

  61. Andolfo I, Russo R, Gambale A, Iolascon A. New insights on hereditary erythrocyte membrane defects. Haematol. 2016;101:1284–94.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This project was funded by the National Plan for Science, Technology and Innovation (MAARIFAH), King Abdulaziz City of Science and Technology, and Kingdom of Saudi Arabia, Award Number (13-B101206-02).

Author information

Authors and Affiliations

Authors

Contributions

SA, MAD and FW designed the study; MAD, ASA, MJA, and DD carried out the experiments and analysed the data. All authors wrote and revised the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Denis Delic.

Ethics declarations

Ethics approval and consent to participate

This study was carried out in strict accordance with the German law on animal protection. The maintenance of mice as well as the experimental protocol of the study were officially approved by the State-controlled Committee on the Ethics of Animal Experiments of the State Northrhine-Westphalia, Germany, and were regularly monitored without prior notice by the local authorities. All efforts were undertaken to minimize the suffering of mice.

Consent for publication

Not applicable.

Competing interests

Author Denis Delić was employed by company Boehringer Pharma GmbH & Co. KG, Ingelheim, Biberach, Germany. All other authors declare no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Delic, D., Wunderlich, F., Al-Quraishy, S. et al. Vaccination accelerates hepatic erythroblastosis induced by blood-stage malaria. Malar J 19, 49 (2020). https://doi.org/10.1186/s12936-020-3130-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12936-020-3130-2

Keywords