We generated mice expressing a full-length Mpl transgene under the control of a 2-kb Mpl promoter in an Mpl−/− background, effectively obtaining mice that express full-length Mpl in the absence of other Mpl isoforms. These mice developed thrombocytosis with platelet levels approximately 5-fold higher than wild-type controls and markedly increased megakaryocyte numbers. The reintroduction of one wild-type Mpl allele restored normal platelet counts. We excluded the deletion of Mpl-tr, a dominant-negative isoform, as the underlying molecular cause for thrombocytosis. Instead, we found that transgene expression driven by the 2-kb Mpl promoter fragment was decreased during late megakaryocyte maturation, resulting in strongly diminished Mpl protein expression in platelets. Because platelets exert a negative feedback on thrombopoiesis by binding and consuming Tpo in the circulation through Mpl, we propose that the severe reduction of Mpl protein in platelets in Mpl-transgenic Mpl−/− mice shifts the equilibrium of this feedback loop, resulting in markedly elevated levels of megakaryocytes and platelets at steady state. Although the mechanism causing decreased expression of Mpl protein in platelets from patients with myeloproliferative disorders differs from this transgenic model, our results suggest that lowering Mpl protein in platelets could contribute to raising the platelet count.

Thrombopoietin (Tpo) and its receptor Mpl are the principal regulators of megakaryopoiesis.1,2  Mice deficient in Tpo or Mpl continue to produce functional platelets, albeit at much lower levels,3,4  suggesting that Mpl mainly controls quantitative aspects of thrombopoiesis. Tpo serum levels are controlled by the platelet mass through Mpl-mediated Tpo uptake and degradation.5,6  Consequently, Mpl−/− mice show increased Tpo levels.3  Although an important function of Mpl is to regulate platelet numbers, it is also expressed on hematopoietic stem cells (HSCs) and early progenitors.7,8  Consistently, Mpl-deficient mice show markedly decreased numbers of hematopoietic progenitors, and competitive repopulation assays indicate that the numbers of murine HSCs is reduced by 7- to 8-fold.7,8  In humans, loss-of-function mutations in Mpl lead to congenital amegakaryocytic thrombocytopenia, a disorder that frequently leads to bone marrow failure.9-11  The reason for the more severe phenotype in humans remains unknown.

Mutant versions of Mpl can lead to uncontrolled proliferation and survival signals as exemplified by the retroviral fusion oncogene v-Mpl, which can immortalize hematopoietic progenitors.12  An autosomal-dominant point mutation in the transmembrane domain of Mpl (S505N) was identified as the cause of thrombocytosis in families with hereditary thrombocytosis.13,14  Recently, point mutations in the cytoplasmic domain of Mpl (W515L, W515K) were identified in patients with primary myelofibrosis and essential thrombocythemia, and W515L was shown in mouse models to elicit myeloproliferative disease (MPD) with marked thrombocytosis.15,16  These findings underline that Mpl can trigger potent growth signals, and stringent mechanisms of signal attenuation are essential for ensuring an appropriate, controlled cellular response.

To study the effects of Mpl variants carrying mutations in the cytoplasmic domain that are expected to alter signaling, we used a transgene rescue strategy. The cDNAs encoding Mpl variants were placed under the control of a previously characterized Mpl promoter,17  and transgenic mice were generated on the Mpl−/− background.3  Unexpectedly, transgenic mice expressing the wild-type Mpl cDNA that was designed as a control displayed marked thrombocytosis, with 5-fold elevated platelets when examined on the Mpl−/− background. Since the platelet numbers were normal when the transgene was examined on the heterozygous Mpl−/+ background, we investigated the possibility that thrombocytosis was due to loss of an alternatively spliced Mpl mRNA. Expression of alternative cytokine receptor variants, which act as dominant-negative isoforms, can serve as a mechanism of modulating the responses to cytokines.18-20  Mechanistically, dominant-negative receptor variants can form nonfunctional heterodimers with the full-length receptor or, when expressed as secreted “soluble” isoforms, compete with the receptor for ligand binding.21  We have described a third mechanism, in which overexpression of the Mpl isoform Mpl-tr triggers protein degradation of the full-length receptor.22  Mpl-tr is the only splice variant found both in human and mouse. This variant results from splicing of exon 8 directly to exon 11, eliminating the juxtamembrane extracellular part and the transmembrane domain.23,24  However, our genetic analysis excluded loss of Mpl-tr as the cause of thrombocytosis in our transgene rescue mice. Instead, decreased expression of Mpl protein on platelets appears to be the cause of thrombocytosis through a dysbalance in the autoregulatory mechanism controlling Tpo. This mouse model recapitulates disease manifestations frequently associated with human MPDs, including thrombocytosis and a reduction of Mpl protein expression in platelets.

Generation of transgenic mice

We used a 2-kb HindIII-Apa I mouse Mpl promoter fragment,17  which was cloned in front of a mouse Mpl cDNA and an SV40 polyadenylation signal. The resulting 5-kb fragment, which contains no vector sequences, was used for oocyte microinjection. We generated 5 transgenic founder mice in the B6D2Fn background. This study was approved by the institutional review boards of all participating institutions. All mice used in this study were kept under specific pathogen–free conditions and in accordance to Swiss and German federal regulations.

Blood and tissue analysis

Animals were killed with CO2, and blood was obtained by cardiac puncture and mixed with EDTA. Blood counts were determined on an Advia 120 Hematology Analyzer (Bayer, Leverkusen, Germany). ELISA was performed with the Quantikine Mouse Tpo Immunoassay kit (R&D Systems, Abingdon, United Kingdom). For histology, freshly dissected tissues were fixed in Optimal*Fix (AMTS, Lodi, CA). Fixed specimens were embedded in paraffin, sectioned, and stained in the Transgenic Pathology Laboratory (University of California, Davis, CA). Megakaryocyte frequency was determined by 2-color flow cytometry as described.25  In brief, bone marrow cells from femurs and tibias were isolated in CATCH (129 nM NaCl, 8.614 nM Na2HPO4, 1.6 mM KH2PO4, 13.6 mM sodium citrate, 11.1 mM glucose, 1 mM adenosine, 2 mM theophilline, and 1% bovine serum albumin [BSA]; pH 7.0). Cells were filtered through a 100-μm nylon mesh, and megakaryocytes were stained using the monoclonal 4A5 rat anti–mouse platelet antibody and a fluoroisothiocyanate (FITC)–goat anti–rat IgG F(ab′)2 antibody (Tago, Burlingame, CA). Samples for electron microscopy were processed essentially as described.26  In brief, spleen tissue was diced into pieces of roughly 1 mm2, fixed for 1 hour in 3% glutaraldehyde in 0.1 M phosphate buffer (pH 7.4), treated with osmium tetroxide, washed, and embedded in Epon (Fluka Chemie, Buchs, Switzerland). Ultrathin sections were collected on grids and examined on a Philips Morgani 268D electron microscope (FEI, Hillsboro, OR) and images were analyzed with Photoshop (Adobe, San Jose, CA).

Platelet and megakaryocyte isolation and Western blot

For platelet isolation, blood was drawn by cardiac puncture into a syringe containing 3.8% citrate. Platelet-rich plasma was obtained by centrifugation at 350g for 10 minutes, and pure platelets were sedimented by centrifugation at 1300g, washed once with phosphate-buffered saline (PBS), and analyzed for purity on an Advia 120 Hematology Analyzer. For megakaryocyte isolation, femurs were flushed with CATCH medium. A total of 4 mL of a single-cell suspension was then mixed with 3 mL Percoll/PBS (1.02 g/mL; GE Healthcare, Chalfont St Giles, United Kingdom), gently layered on top of 4 mL Percoll/PBS (1.05 g/mL) and centrifuged for 20 minutes at 400g. The interface was then collected and washed with CATCH buffer. Alternatively, megakaryocytes were isolated using anti-CD41 antibodies (BD Pharmingen, Franklin Lakes, NJ) and Dynabeads conjugated with anti–rat IgG (Invitrogen, Carlsbad, CA). The purity of the megakaryocyte preparation was assessed by acetylcholine esterase staining on cytospin slides.27,28  For protein extraction, isolated platelets and megakaryocytes were lyzed by boiling in 2× SDS-PAGE loading buffer for 5 minutes at a concentration of 2× 106 platelets/μL and 50 000 megakaryocytes/μL. Protein lysates were size-separated by SDS-PAGE and transferred to nitrocellulose. The membranes were probed with a biotinylated mouse monoclonal anti-Mpl antibody and reprobed with a biotinylated 4A5 rat anti–mouse glycoprotein V monoclonal antibody (clone 4A5).8,29  Signals were detected using horseradish peroxidase–coupled streptavidin and the enhanced chemiluminescence (ECL) system (Amersham Biosciences, Piscataway, NJ).

RNA isolation and quantitative real-time PCR

Total RNA was isolated with TriFast (PeqLab, Erlangen, Germany) and treated with Turbo DNAse (Ambion, Austin, TX). Reverse transcription was performed with Omniscript reverse transcriptase (QIAGEN, Hilden, Germany), and control reactions without reverse transcriptase were included. Quantitative real-time polymerase chain reaction (qPCR) was performed using SYBR Green PCR master mix on an ABI Prism 7000 (Applied Biosystems, Foster City, CA) with the following primers: CTGTATGCCTACCGAGGAGAGAAG and GTTCCAAAGGTGGGCACACT for total mouse Mpl; TATTGGCAGCAGCCCTGAA and TGGATGGTGTTGAGGATGGATA for endogenous mouse Mpl (reverse primer located in 3′ untranslated region [UTR]); CAGCCCTGACTCGAATCGAA and TGCAGGAATTGACGGTATCG for transgenic Mpl (partially located in the SV40 polyadenylation signal); GGGCCTTCCTTCGGGAT and AGCACAATTGGGCTCAGCTT for mouse Itga2b (CD41); and ATCCGCAAGCCTGTGACTGT and TCGGGCCAGGGTGTTTTT for mouse Rpl19.

Mapping of the transgene integration site

The chromosomal integration site of the Yall transgene was determined by thermal asymmetric interlaced PCR.30  The gene-specific primers were GGAGCCTGGTGTAATAGCTCAC, AATCCCAGAACTTGGGAGAGAG, and GACCAGCCTGTTCTACAAGCAA.

Hematopoietic progenitors, megakaryocyte culture, and ploidy sort

Clonal cultures of hematopoietic cells were performed as described.31  Colonies containing at least 3 acetylcholinesterase-positive cells were scored as colony-forming unit–megakaryocyte (CFU-MK).32  For megakaryocyte cultures, femurs and tibias were flushed with CATCH medium, and cells were subjected to lineage depletion (Mouse Hematopoietic Cell Lineage Depletion Kit; R&D Systems), leading to a relative enrichment of megakaryocytes. RNA was prepared from cells immediately or after 2 or 4 days of culture in RPMI with 10% fetal calf serum (FCS), Tpo-conditioned media,6  and 10 μg/mL recombinant murine stem cell factor (BioSource, Camarillo, CA). For ploidy sorts, lineage-depleted bone marrow cells were stained in PBS with 0.1% BSA and 1 mM EDTA with a FITC-conjugated anti-CD41 antibody (Becton Dickinson) for 30 minutes at 4°C. After washing, cells were incubated for 2 hours at 37°C with 6.25 μg/mL Hoechst 33342 (Invitrogen) in RPMI without phenol red containing 0.1% BSA and 5 mM EDTA. CD41+ cells were sorted into 2 fractions (2n/4n/8n and 16n/32n/64n) on a FACSVantage cell sorter (Becton Dickinson).

Generation of transgenic mice expressing Mpl cDNA under the control of the Mpl promoter

We generated transgenic mice expressing the full-length mouse Mpl cDNA under the control of a 2-kb Mpl promoter fragment (Figure 1A), which we previously found to direct expression specifically to the megakaryocytic lineage.17  We obtained 5 founder mice and detected Mpl transgene expression in 3 of them (data not shown). These mice were viable, fertile, and showed no overt abnormalities. To eliminate the expression of the endogenous Mpl gene, we bred the founders into the Mpl knockout (Mpl−/−) genetic background.3  All 3 Mpl transgenic lines that expressed the transgene showed marked thrombocytosis in an Mpl−/− background with platelet counts 3- to 6-fold higher than the normal range, whereas a nonexpressing line showed thrombocytopenia similar to Mpl−/− mice (Table 1).3  One line, designated Yall (for all tyrosines [Y] of the cytoplasmic domain), was chosen for detailed analysis.

Figure 1

Expression of an Mpl transgene (Yall) in megakaryocytes. (A) Yall transgene construct: a 2-kb fragment corresponding to the genomic sequence immediately 5′ of the Mpl ATG start codon was placed as a promoter in front of the mouse Mpl cDNA. An alignment of endogenous mouse Mpl genomic sequence (above) and of the transgene (below) in the region upstream of the ATG start codon is shown. An SV40-derived sequence element containing an intron was placed at the 3′ end of the construct to ensure polyadenylation of the transcript. (B) Specific expression of the Yall transgene in bone marrow megakaryocytes. Expression of transgenic (■) and endogenous Mpl mRNA () was measured by qPCR in mouse organs from Yall;Mpl−/+ mice (error bars represent SD). Bone marrow was further fractionated into CD41+ and CD41 cells. BM indicates whole bone marrow; SPL, spleen; THY, thymus; LIV, liver; KID, kidney; LUN, lung; HEA, heart; INT, intestine; BRA, brain; OVA, ovary; and TES, testis. Primers for qPCR were specific for transgene and endogenous Mpl mRNA, respectively, and control cDNA synthesis reactions without reverse transcriptase were analyzed to exclude amplification of genomic DNA. Expression of mouse Rpl19 was used for normalization and relative expression was calculated with the ΔΔCT method using one bone marrow sample as calibrator. The mean value of 3 mice is shown. (C) Western blot with protein extracts from magnetic-activated cell sorter (MACS)–isolated megakaryocytes probed with antibodies against Mpl protein and the megakaryocyte-specific glycoprotein V (GP V). Megakaryocytes from wild-type (wt) mice and nontransgenic (−) or transgenic (Yall) Mpl−/− mice were analyzed.

Figure 1

Expression of an Mpl transgene (Yall) in megakaryocytes. (A) Yall transgene construct: a 2-kb fragment corresponding to the genomic sequence immediately 5′ of the Mpl ATG start codon was placed as a promoter in front of the mouse Mpl cDNA. An alignment of endogenous mouse Mpl genomic sequence (above) and of the transgene (below) in the region upstream of the ATG start codon is shown. An SV40-derived sequence element containing an intron was placed at the 3′ end of the construct to ensure polyadenylation of the transcript. (B) Specific expression of the Yall transgene in bone marrow megakaryocytes. Expression of transgenic (■) and endogenous Mpl mRNA () was measured by qPCR in mouse organs from Yall;Mpl−/+ mice (error bars represent SD). Bone marrow was further fractionated into CD41+ and CD41 cells. BM indicates whole bone marrow; SPL, spleen; THY, thymus; LIV, liver; KID, kidney; LUN, lung; HEA, heart; INT, intestine; BRA, brain; OVA, ovary; and TES, testis. Primers for qPCR were specific for transgene and endogenous Mpl mRNA, respectively, and control cDNA synthesis reactions without reverse transcriptase were analyzed to exclude amplification of genomic DNA. Expression of mouse Rpl19 was used for normalization and relative expression was calculated with the ΔΔCT method using one bone marrow sample as calibrator. The mean value of 3 mice is shown. (C) Western blot with protein extracts from magnetic-activated cell sorter (MACS)–isolated megakaryocytes probed with antibodies against Mpl protein and the megakaryocyte-specific glycoprotein V (GP V). Megakaryocytes from wild-type (wt) mice and nontransgenic (−) or transgenic (Yall) Mpl−/− mice were analyzed.

Close modal
Table 1

Blood counts of Mpl transgenic lines on the Mpl−/− background

Transgenic lineTransgene expressionBlood counts
nPlatelets, ×109/LHemoglobin, g/LWBC, ×106/L
M15 (YallYes 4791 ± 922 130 ± 11 6.96 ± 4.26 
M16 Yes 6165 ± 1262 127 ± 7.2 12.04 ± 4.62 
M3 Yes 3367 ± 397 147 ± 6.5 8.27 ± 2.2 
M8 No 194 ± 14 135 ± 1.3 5.22 ± 1.02 
Transgenic lineTransgene expressionBlood counts
nPlatelets, ×109/LHemoglobin, g/LWBC, ×106/L
M15 (YallYes 4791 ± 922 130 ± 11 6.96 ± 4.26 
M16 Yes 6165 ± 1262 127 ± 7.2 12.04 ± 4.62 
M3 Yes 3367 ± 397 147 ± 6.5 8.27 ± 2.2 
M8 No 194 ± 14 135 ± 1.3 5.22 ± 1.02 

Four Mpl transgenic lines were generated from the same transgenic cDNA construct and bred into the Mpl−/− genetic background. The blood counts are shown plus or minus standard deviation. n indicates number of transgenic mice on Mpl−/− background. Note that marked thrombocytosis was found in all 3 strains expressing the transgene. The strain M15 was chosen for detailed analysis and subsequently referred to as “Yall.”

The expression of the Yall transgene and endogenous Mpl in mouse organs was assessed by qPCR in transgenic mice heterozygous for Mpl knockout (Yall;Mpl−/+, Figure 1B). The distinction between transgenic and endogenous Mpl mRNA was achieved by placing the reverse primer of each qPCR primer pair in the respective 3′ UTRs. The transgene mRNA was expressed in bone marrow and brain, whereas the endogenous Mpl mRNA was found in bone marrow and testes. Megakaryocytes purified by anti-CD41 magnetic beads showed the highest expression of transgenic and endogenous Mpl mRNA. Comparable levels of Mpl protein were detected by immunoblot using anti-Mpl antibodies in purified megakaryocytes from Yall;Mpl−/− mice and wild-type controls, and—as expected—Mpl protein was absent in megakaryocytes from Mpl−/− mice (Figure 1C).

Increased megakaryopoiesis in Yall;Mpl−/− mice

Similar to the increase in platelets (Figure 2A), the percentage of megakaryocytes in the bone marrow of Yall;Mpl−/− mice was elevated as determined by flow cytometry (Figure 2B). To assess whether enhanced polyploidization could also be a contributing factor for the thrombocytosis, we determined ploidy of bone marrow megakaryocytes by flow cytometry (Figure 2C). Yall;Mpl−/− mice showed a slight shift toward higher ploidy, whereas a shift toward lower ploidy was seen in Mpl−/− mice. We also observed a marked increase in the number of megakaryocytic precursors (CFU-MK) in Yall;Mpl−/− mice, as determined by colony assays (Figure 2D). Thus, Mpl transgene expression in Mpl−/− mice resulted in an expansion of the megakaryocytic lineage at all stages of differentiation. In addition, we found that the frequencies of other hematopoietic progenitor types were also elevated in Yall;Mpl−/− mice (Figure 2E). This finding is in agreement with the role of Mpl in the maintenance of hematopoietic progenitors.7,8,33 

Figure 2

Increased number of platelets, megakaryocytes, and hematopoietic progenitors in Yall;Mpl−/− mice. (A) Platelet counts in peripheral blood from wild-type (wt), and nontransgenic (−), or transgenic (Yall) Mpl−/−mice. (B) Frequency of megakaryocytes in the bone marrow of mice with the same genotypes as in panel A, determined by flow cytometry. (C) Ploidy of megakaryocytes with the indicated genotypes, determined by flow cytometry. (D) CFU-MK assays showing an increased number of megakaryocyte (MK) colonies in Yall;Mpl−/− mice. (E) Hematopoietic colony assays (CFUs/burst-forming units [BFUs]) in methylcellulose indicate a general expansion of progenitors in the bone marrow of Yall;Mpl−/− mice. Error bars in panels A through D represent SD.

Figure 2

Increased number of platelets, megakaryocytes, and hematopoietic progenitors in Yall;Mpl−/− mice. (A) Platelet counts in peripheral blood from wild-type (wt), and nontransgenic (−), or transgenic (Yall) Mpl−/−mice. (B) Frequency of megakaryocytes in the bone marrow of mice with the same genotypes as in panel A, determined by flow cytometry. (C) Ploidy of megakaryocytes with the indicated genotypes, determined by flow cytometry. (D) CFU-MK assays showing an increased number of megakaryocyte (MK) colonies in Yall;Mpl−/− mice. (E) Hematopoietic colony assays (CFUs/burst-forming units [BFUs]) in methylcellulose indicate a general expansion of progenitors in the bone marrow of Yall;Mpl−/− mice. Error bars in panels A through D represent SD.

Close modal

Histopathologic analysis revealed a marked increase in megakaryocyte numbers in the bone marrow and spleen of Yall;Mpl−/− mice (Figure 3A). Furthermore, spleen sections of Yall;Mpl−/− but not wild-type mice contained aggregates of small particles (Figure 3A brown color). Such aggregates were also visible at a lower frequency in bone marrow. To identify the nature of the particles, electron microscopic analysis of spleen sections was performed. We detected areas containing densely packed platelets in Yall;Mpl−/− mice, but not in wild-type controls (Figure 3B). Platelets were identified by size, morphology, and the presence of dense granules.34,35 

Figure 3

Abnormally high numbers of megakaryocytes and areas of packed platelets in bone marrow and spleen of Yall;Mpl−/− mice. (A) Sections of bone marrow and spleen show an increase in the number of megakaryocytes in Yall;Mpl−/− (right) compared with wild-type (left) mice. Aggregates of small platelet-like particles (brown color) are only found in Yall;Mpl−/− mice and are more prominent in the spleen. (B) Electron microscopy reveals densely packed platelets in spleen sections of Yall;Mpl−/− mice. Large arrows indicate the boundaries of platelets; the small arrow points at a dense granule within a platelet. No such platelet clusters were found in wild-type mice (data not shown). Images were taken using an Axioscope 2a microscope with a Planar Neofluar objective (40×/0.75 NA) and an AxioCam HR digital camera (all from Carl Zeiss, Jena, Germany). The software for image acquisition was AxioVision Rel. 4.6.

Figure 3

Abnormally high numbers of megakaryocytes and areas of packed platelets in bone marrow and spleen of Yall;Mpl−/− mice. (A) Sections of bone marrow and spleen show an increase in the number of megakaryocytes in Yall;Mpl−/− (right) compared with wild-type (left) mice. Aggregates of small platelet-like particles (brown color) are only found in Yall;Mpl−/− mice and are more prominent in the spleen. (B) Electron microscopy reveals densely packed platelets in spleen sections of Yall;Mpl−/− mice. Large arrows indicate the boundaries of platelets; the small arrow points at a dense granule within a platelet. No such platelet clusters were found in wild-type mice (data not shown). Images were taken using an Axioscope 2a microscope with a Planar Neofluar objective (40×/0.75 NA) and an AxioCam HR digital camera (all from Carl Zeiss, Jena, Germany). The software for image acquisition was AxioVision Rel. 4.6.

Close modal

One copy of the wild-type Mpl gene restores normal platelet counts in Yall transgenic mice

We have recently shown that a splice variant of Mpl, called Mpl-tr, acts as a dominant-negative regulator of Mpl function in vitro.22,36  Because the Mpl knockout allele carries a neomycin cassette inserted in Mpl exon 3, the Mpl−/− mice cannot express functional Mpl-tr mRNA, which contains exon 3. We therefore hypothesized that the increased megakaryopoiesis and thrombopoiesis could be caused by the absence of the Mpl-tr transcript in Yall;Mpl−/− mice. Consistently, the Yall transgene on the Mpl−/+ background displayed platelet numbers similar to their nontransgenic littermates, indicating that a single copy of the wild-type Mpl gene is dominant over the transgene in respect to platelet production (Figure 4A). Similarly, comparable numbers of CFU-MKs were found in the bone marrow from transgenic and nontransgenic Mpl−/+ mice (data not shown). The Yall transgene on the Mpl+/+ background showed normal platelet numbers, and the heterozygous Mpl−/+ mice showed slightly higher platelet counts than Mpl+/+ mice independent of the presence or absence of the transgene (Figure 4A). To more directly test the hypothesis that the absence of Mpl-tr is the cause of thrombocytosis in Yall;Mpl−/− mice, we took advantage of an Mpl knockin allele named MplΔ60.37  This allele was generated by homologous recombination replacing exons 11 and 12, which encode the cytoplasmic signaling domain of Mpl, with a cDNA fragment encoding a truncated cytoplasmic domain that lacks the last 60 amino acids (Figure 4B). MplΔ60/Δ60 mice have normal platelet counts, demonstrating that the last 60 C-terminal amino acids of Mpl are not essential for steady-state thrombopoiesis in vivo.37  Mpl-tr cannot be produced by the MplΔ60 allele, because the splice acceptor for the generation of Mpl-tr is absent (Figure 4B). We confirmed the absence of Mpl-tr in MplΔ60/Δ60 mice by 3′ rapid amplification of cDNA ends (RACE; data not shown). We then crossed Yall;Mpl−/− mice with MplΔ60/Δ60 mice to obtain Yall;Mpl−/Δ60 mice, expecting that these mice would also show thrombocytosis. However, platelet levels in these mice were normalized to levels comparable with Yall;Mpl−/+ or Yall;Mpl+/+ mice (Figure 4A). Thus, the presence of the MplΔ60 allele was dominant over the transgene in respect to platelet production despite the fact that Mpl-tr cannot be expressed. These results exclude the hypothesis that the absence of Mpl-tr is the primary cause of thrombocytosis in Yall;Mpl−/− mice.

Figure 4

Suppression of thrombocytosis by adding back Mpl wild-type alleles or the Δ60 knockin allele. (A) Platelet counts (error bars = SD) in peripheral blood from nontransgenic () or transgenic (■) mice that are homozygous (Mpl−/−), heterozygous (Mpl−/+), or wild-type (Mpl+/+) at the Mpl locus or containing one knockout and one Δ60 knockin allele (Mpl−/Δ60). Values from at least 5 mice were determined. Note that in mice containing at least one Mpl wild-type allele or the Δ60 allele, platelet levels are only marginally affected by the presence of the Yall transgene, and reduced by approximately 70% compared with Yall;Mpl−/− mice. (B) Schematic representation of the Mpl gene and the Δ60 knockin. Numbered boxes represent exons. A line connecting exon 8 with exon 11 marks the splicing event that generates the dominant-negative variant Mpl-tr. In the Δ60 allele, exons 10, 11, and a truncated version of exon 12 are inserted as a cDNA fragment, followed by an SV40-derived polyadenylation signal. Consequently, Mpl-tr cannot be formed from this allele.

Figure 4

Suppression of thrombocytosis by adding back Mpl wild-type alleles or the Δ60 knockin allele. (A) Platelet counts (error bars = SD) in peripheral blood from nontransgenic () or transgenic (■) mice that are homozygous (Mpl−/−), heterozygous (Mpl−/+), or wild-type (Mpl+/+) at the Mpl locus or containing one knockout and one Δ60 knockin allele (Mpl−/Δ60). Values from at least 5 mice were determined. Note that in mice containing at least one Mpl wild-type allele or the Δ60 allele, platelet levels are only marginally affected by the presence of the Yall transgene, and reduced by approximately 70% compared with Yall;Mpl−/− mice. (B) Schematic representation of the Mpl gene and the Δ60 knockin. Numbered boxes represent exons. A line connecting exon 8 with exon 11 marks the splicing event that generates the dominant-negative variant Mpl-tr. In the Δ60 allele, exons 10, 11, and a truncated version of exon 12 are inserted as a cDNA fragment, followed by an SV40-derived polyadenylation signal. Consequently, Mpl-tr cannot be formed from this allele.

Close modal

Expression of the Yall transgene is dramatically decreased in platelets

To determine whether the function of Mpl protein in platelets as a negative regulator of Tpo serum levels was disrupted in Yall;Mpl−/− mice, we compared the expression of Mpl in platelets and megakaryocytes from Yall;Mpl−/−, MplΔ60/Δ60, and wild-type mice (Figure 5A). Mpl mRNA expression in platelets was strongly reduced in Yall;Mpl−/− mice compared with wild-type and MplΔ60/Δ60 mice (Figure 5A top panel), which translated into marked differences in protein levels as determined by Western blot analysis (Figure 5A bottom panel). In contrast, no differences in Mpl mRNA or protein expression were observed in purified megakaryocytes from the different genotypes. Thus, Mpl mRNA and protein encoded by the Yall transgene was dramatically reduced in platelets, but not in megakaryocytes, whereas Mpl expression driven by the endogenous Mpl promoter in wild-type and MplΔ60 mice was comparable in both platelets and megakaryocytes. Based on these findings, a possible cause for the observed thrombocytosis in Yall;Mpl−/− mice could be the decreased expression of Mpl in platelets and thus a reduced capacity of platelets to absorb circulating Tpo protein.

Figure 5

Reduced transgene expression in platelets of Yall;Mpl−/− mice, but normal Tpo levels and platelet half-life. (A) Mpl mRNA (top) and protein (bottom) expression in purified platelets and megakaryocytes. qPCR was performed with primers amplifying both transgenic and endogenous Mpl; Itga2b (CD41) primers were used for normalization. n.d. indicates not determined. Note that Mpl mRNA in Yall;Mpl−/− platelets is approximately 100-fold lower than in platelets of the other 2 genotypes. Similarly, Mpl protein is strongly decreased in platelets from Yall;Mpl−/− mice. Membranes were reprobed with anti-CD41 antibodies to control for equal loading. (B) Tpo levels in mice with the indicated genotypes. Tpo concentration in both serum and plasma was determined by ELISA. Consistent with previous reports, Tpo is elevated in Mpl−/− mice. In contrast, Yall;Mpl−/− mice displayed slightly reduced Tpo compared with controls. Note that the plasma values are generally lower than the serum values, with the exception of Mpl−/− mice. (C) Half-life of platelets is normal in Yall;Mpl−/− mice. After injection of chemically activated biotin, blood samples were taken at indicated time points and the fraction of biotin-labeled platelets was determined by flow cytometry. Error bars in panels A through C represent SD.

Figure 5

Reduced transgene expression in platelets of Yall;Mpl−/− mice, but normal Tpo levels and platelet half-life. (A) Mpl mRNA (top) and protein (bottom) expression in purified platelets and megakaryocytes. qPCR was performed with primers amplifying both transgenic and endogenous Mpl; Itga2b (CD41) primers were used for normalization. n.d. indicates not determined. Note that Mpl mRNA in Yall;Mpl−/− platelets is approximately 100-fold lower than in platelets of the other 2 genotypes. Similarly, Mpl protein is strongly decreased in platelets from Yall;Mpl−/− mice. Membranes were reprobed with anti-CD41 antibodies to control for equal loading. (B) Tpo levels in mice with the indicated genotypes. Tpo concentration in both serum and plasma was determined by ELISA. Consistent with previous reports, Tpo is elevated in Mpl−/− mice. In contrast, Yall;Mpl−/− mice displayed slightly reduced Tpo compared with controls. Note that the plasma values are generally lower than the serum values, with the exception of Mpl−/− mice. (C) Half-life of platelets is normal in Yall;Mpl−/− mice. After injection of chemically activated biotin, blood samples were taken at indicated time points and the fraction of biotin-labeled platelets was determined by flow cytometry. Error bars in panels A through C represent SD.

Close modal

Normal Tpo levels and platelet half-life in Yall;Mpl−/− mice

In Mpl−/− mice, which lack the Tpo receptor and have low platelet counts, Tpo levels are substantially increased.3  Platelets in Yall;Mpl−/− mice still express Mpl protein, albeit at much lower levels than wild-type mice. We therefore asked whether the reduction in Mpl protein in platelets leads to increased Tpo levels and determined Tpo by enzyme-linked immunosorbent assay (ELISA) in both serum and plasma. Tpo in serum and plasma from Yall;Mpl−/− mice was not elevated, but rather slightly diminished compared with the Tpo levels detected in Yall;Mpl−/+ control mice (Figure 5B). As shown previously, Mpl−/− mice displayed elevated Tpo levels. Thus, reduction in Mpl protein expression in platelets did not result in elevated Tpo serum levels in Yall;Mpl−/− mice. Because megakaryocytes have been shown to also have an effect on the circulating Tpo levels,38,39  it seems plausible that the increase in megakaryocyte mass in bone marrow may compensate for the decrease in Mpl protein expression in platelets in Yall;Mpl−/− mice. Thrombocytosis could also be the consequence of increased half-life of platelets in Yall;Mpl−/− mice. To address this possibility, we marked blood cells in wild-type and Yall;Mpl−/− mice with biotin and determined the fraction of labeled platelets over time by flow cytometry.40  No significant difference in half-life was observed (Figure 5C).

Mpl transgene expression is progressively decreasing during megakaryocyte maturation

To explain the reduction of Yall transgene expression in platelets, we considered 2 possibilities. First, the transgenic mRNA could be less stable than the endogenous Mpl mRNA. Since platelets do not contain nuclei and thus no longer transcribe mRNA, a faster decay of the transgene message could result in strongly reduced Mpl expression levels in platelets. Second, the 2-kb Mpl promoter used in the Yall transgenic mice could be transcriptionally less active in late stages of megakaryocyte maturation than the endogenous Mpl promoter.

The stability of many mRNAs is regulated by sequence elements in the 3′ UTR. The Yall transgene was constructed with a polyadenylation signal derived from the SV40 virus that is different from the polyadenylation signal of endogenous Mpl mRNA. However, the MplΔ60 knockin allele, which results in normal Mpl expression level in platelets, also features an SV40 polyadenylation signal. Since different variants of the SV40 polyadenylation signal were used in the Yall and the MplΔ60 constructs, we investigated whether this difference affects the half-life of the respective mRNAs. We constructed 2 Mpl expression plasmids differing only in their polyadenylation signal, one corresponding to Yall and the other to MplΔ60. Stable transfectants were obtained in murine hematopoietic BaF3 cells, and the half-life of Mpl mRNA was measured after blocking transcription with actinomycin D. We observed no difference in the mRNA half-life of the 2 constructs (data not shown), suggesting that differences in mRNA stability are not responsible for the loss of transgene expression in platelets of Yall;Mpl−/− mice.

Alternatively, expression of Mpl mRNA in our Yall transgene could be reduced at late but not early stages of megakaryocytic differentiation (eg, due to a lack of regulatory elements in the 2-kb promoter construct or due to effects related to transgene insertion). The Yall transgene was mapped to mouse chromosome 9, band D, between the genes Grinl1a, encoding a glutamate receptor, and Lipc, encoding hepatic lipase c, neither of which have any known function in megakaryopoiesis. Since the MplΔ60 allele has been generated by homologous recombination into the endogenous Mpl locus, transcriptional regulation of MplΔ60 is expected to be the same as for endogenous Mpl. To compare the expression of transgenic and endogenous Mpl mRNA during megakaryocyte maturation, we cultured megakaryocyte-enriched bone marrow from Yall;Mpl−/+ mice in the presence of Tpo and determined the mRNAs of Yall and endogenous Mpl on days 0, 2, and 4 of culture by qPCR with primers that are specific for the respective mRNAs (Figure 6A).

Figure 6

Expression of the Yall transgene but not of endogenous Mpl drops during megakaryocyte differentiation. (A) Comparison of transgenic (■) and endogenous () Mpl mRNA expression in megakaryocytes cultured for 0, 2, or 4 days with Tpo (error bars = SD). Bone marrow of mice containing both the Yall transgene and one wild-type Mpl allele (Yall;Mpl−/+) was lineage-depleted to enrich megakaryocytes and cultured in the presence of Tpo. Specific qPCR primers were used to distinguish mRNA of endogenous and transgene Mpl. Itga2b (CD41) mRNA was used as internal control, and data were normalized by arbitrarily setting one sample of the day 0 group to 100%. Results are based on bone marrow from 6 mice cultured separately. (B) Comparison of transgenic (■) and endogenous () Mpl in FACS-sorted megakaryocytes of low (R1) or high (R2) ploidy. Lineage-depleted bone marrow from Yall;Mpl−/+ mice was stained with FITC-labeled anti-CD41 antibodies and the DNA-binding dye Hoechst 33342. Only CD41+ cells are shown in the histogram. Megakaryocytes were sorted into 2 ploidy fractions—R1 (2n, 4n, 8n) and R2 (16n and higher)—based on their DNA content. The bar graph below indicates relative expression values measured as in panel A with the R1 value set to 100%.

Figure 6

Expression of the Yall transgene but not of endogenous Mpl drops during megakaryocyte differentiation. (A) Comparison of transgenic (■) and endogenous () Mpl mRNA expression in megakaryocytes cultured for 0, 2, or 4 days with Tpo (error bars = SD). Bone marrow of mice containing both the Yall transgene and one wild-type Mpl allele (Yall;Mpl−/+) was lineage-depleted to enrich megakaryocytes and cultured in the presence of Tpo. Specific qPCR primers were used to distinguish mRNA of endogenous and transgene Mpl. Itga2b (CD41) mRNA was used as internal control, and data were normalized by arbitrarily setting one sample of the day 0 group to 100%. Results are based on bone marrow from 6 mice cultured separately. (B) Comparison of transgenic (■) and endogenous () Mpl in FACS-sorted megakaryocytes of low (R1) or high (R2) ploidy. Lineage-depleted bone marrow from Yall;Mpl−/+ mice was stained with FITC-labeled anti-CD41 antibodies and the DNA-binding dye Hoechst 33342. Only CD41+ cells are shown in the histogram. Megakaryocytes were sorted into 2 ploidy fractions—R1 (2n, 4n, 8n) and R2 (16n and higher)—based on their DNA content. The bar graph below indicates relative expression values measured as in panel A with the R1 value set to 100%.

Close modal

Values were normalized to the mRNA of the megakaryocyte-specific marker CD41. We found that expression of endogenous Mpl slightly decreased after 2 and 4 days in culture, whereas the levels of Yall transgene mRNA dropped dramatically during the same period of time (Figure 6A). To show that this decrease correlates with megakaryocyte maturation, we sorted bone marrow–derived megakaryocytes according to increasing ploidy using a fluorescence-activated cell sorter (FACS). Megakaryocytes with a DNA content of 2n, 4n, and 8n were pooled (R1) and compared with a pool of megakaryocytes with a DNA content of 16n or higher (R2). The expression levels of the Yall transgene were decreased in megakaryocytes with higher ploidy, whereas the mRNA of endogenous Mpl was essentially equal (Figure 6B). We propose that such dysregulated Mpl expression during late megakaryopoiesis is causing thrombocytosis in Yall;Mpl−/− mice through decreased Tpo clearance by platelets in peripheral blood.

We generated mice expressing a wild-type Mpl cDNA transgene in the Mpl−/− background and surprisingly observed a marked thrombocytosis with approximately 5-fold higher platelet counts than normal. Initially, these experiments were designed to study Mpl transgenes with tyrosine-to-phenylalanine mutations in the intracellular domain, and the transgenic mice with the wild-type Mpl cDNA served as the positive control. At the same time, this transgenic model also allowed us to assess the in vivo role of alternatively spliced Mpl isoforms that are disrupted in the knockout allele and missing from the transgene. Here, we described in detail 1 of 3 transgenic strains that express the wild-type Mpl cDNA. Another team independently generated very similar Mpl transgenic mice in which the same 2-kb Mpl promoter was used, but was crossed with a different Mpl knockout strain.7  Interestingly, these mice also displayed thrombocytosis, indicating that the phenotype is independent of the Mpl knockout strain used (B. Lannutti and N. Josephson, written personal communication, March 2008).

We initially pursued the hypothesis that the reason for expansion of the megakaryocytic lineage in Yall;Mpl−/− mice could be the absence of Mpl-tr, an Mpl splice form which we have previously characterized as a negative regulator of Mpl in cell lines.22  However, we found that the MplΔ60 knockin allele, which cannot generate Mpl-tr, was nevertheless able to suppress thrombocytosis in Yall;Mpl−/Δ60 mice (Figure 4), which proves that loss of Mpl-tr cannot be the cause of thrombocytosis in Yall;Mpl−/− mice. In humans, 2 alternate mRNA Mpl species are known in addition to Mpl-tr. The Mpl-K variant is due to a read-through beyond the exon 10 splice donor site.41  The resulting K-form of Mpl diverges from the native sequence after the ninth cytoplasmic amino acid and terminates after 57 codons within intron 10. A second isoform, Mpl-del, arises as a consequence of alternative splicing between exons 8 and 9 and encodes a protein of unknown function with an in-frame deletion of 24 amino acids.42  There is no evidence for a negative regulatory role of Mpl-K or Mpl-del proteins, and these human Mpl isoforms have not been found in the mouse.

Analysis of the expression pattern of transgenic and endogenous Mpl revealed a likely cause for the phenotype of Yall;Mpl−/− mice: terminally differentiated megakaryocytes and platelets showed strongly reduced transgene mRNA expression, while in unfractionated megakaryocytes, the expression of transgenic Mpl mRNA and protein were not grossly different from endogenous Mpl of wild-type mice (Figures 5,6). Importantly, Yall-transgenic mice on a heterozygous Mpl−/+ background displayed almost normal platelet counts (Figure 2), and at the same time showed Mpl protein expression in platelets at levels similar to nontransgenic Mpl−/+ control mice (Figure 5). These findings are compatible with a model in which decreased expression of Mpl protein in late megakaryocytes and platelets concurrent with persistent Mpl expression on early megakaryocytes and progenitors is the cause for thrombocytosis in Yall;Mpl−/− mice (Figure 7). Interestingly, heterozygous Mpl−/+ mice consistently showed a slightly higher platelet count than wild-type littermates3,7  (Figure 4). According to our model, the subtle phenotype of heterozygous Mpl−/+ mice may be explained by a moderate decrease in Mpl expression in platelets. A trend toward a higher production of platelets in vitro has been described in Mpl−/− embryonic stem (ES) cells that were rescued with a full-length Mpl cDNA driven by the hEF-1alpha promoter.43  Since these experiments were conducted in vitro, we suspect that the mechanism of slightly increased platelet production in the ES cell experiments is different from the mechanism in our transgenic mice.

Figure 7

Model illustrating a hypothetical shift of the Mpl-Tpo equilibrium in Yall;Mpl−/− mice. (A) In wild-type mice, both platelets in the periphery and megakaryocytes in the bone marrow act as negative regulators of Tpo through absorption via surface Mpl, restricting the expansion of the megakaryocytic lineage. (B) In Yall;Mpl−/− mice, platelets are almost devoid of surface Mpl, thus having a reduced capacity to absorb Tpo (dashed blunt arrow). Consequently, the megakaryocytic lineage expands until the combined amount of Mpl on megakaryocytes and platelets is sufficiently high to reduce Tpo concentration to normal levels. In this new equilibrium, the increased megakaryocyte mass has a more pronounced role in Tpo absorption than in the wild-type equilibrium (large blunt arrow).

Figure 7

Model illustrating a hypothetical shift of the Mpl-Tpo equilibrium in Yall;Mpl−/− mice. (A) In wild-type mice, both platelets in the periphery and megakaryocytes in the bone marrow act as negative regulators of Tpo through absorption via surface Mpl, restricting the expansion of the megakaryocytic lineage. (B) In Yall;Mpl−/− mice, platelets are almost devoid of surface Mpl, thus having a reduced capacity to absorb Tpo (dashed blunt arrow). Consequently, the megakaryocytic lineage expands until the combined amount of Mpl on megakaryocytes and platelets is sufficiently high to reduce Tpo concentration to normal levels. In this new equilibrium, the increased megakaryocyte mass has a more pronounced role in Tpo absorption than in the wild-type equilibrium (large blunt arrow).

Close modal

It has been shown that Tpo serum levels are controlled by the platelet mass through Mpl-mediated Tpo uptake and degradation.5,6  Consequently, Mpl−/− mice showed increased Tpo levels3  (Figure 5B). In Yall;Mpl−/− mice, we found that Tpo levels were normal or slightly reduced (Figure 5). This could be explained by 2 factors: first, platelets make up for decreased Mpl expression by their increased numbers; second, the mass of megakaryocytes is increased and contributes to decreasing the Tpo levels. Megakaryocyte mass was previously shown to be an additional factor in the regulation of Tpo levels, as knockout mice lacking the transcription factor NF-E2, which are profoundly thrombocytopenic but have an increased mass of immature megakaryocytes, display normal Tpo levels.38,39  Similarly, in patients with platelet destructive disorders where megakaryocyte mass is increased, Tpo levels were found to be low.44  To reconcile the normal Tpo levels with our model, we suggest that the loss of the negative regulatory feedback normally exerted by platelets leads to an increased production of megakaryocytes and platelets in Yall;Mpl−/− mice. As these platelets lack the capability to provide a negative feedback, this expansion is expected to continue until the total amount of Mpl protein, contributed predominantly by an increase in megakaryocyte numbers, is able to consume Tpo at a normal rate. A new equilibrium is reached at a higher total number of megakaryocytes and platelets, keeping serum Tpo at normal levels. The production of platelets from mature megakaryocytes has been shown to be Tpo independent.45 

To explain why the Yall transgene is expressed at much lower levels than endogenous Mpl in late megakaryocytes and platelets in Yall;Mpl−/− mice, we considered several possible explanations. First, the mRNA of the Yall transgene could be less stable than the mRNA of endogenous Mpl, possibly due to the difference in the 3′ UTR. However, expression of Mpl constructs bearing different 3′ UTRs in cell lines did not support this hypothesis. Second, transcription of the Yall transgene might decrease during terminal megakaryocytic differentiation. This hypothesis is supported by our findings that Yall expression was reduced in ploidy-sorted mature megakaryocytes and also decreased during maturation of megakaryocytes cultured in vitro (Figure 6). A possible explanation for decreased Mpl expression in late megakaryocytes could be that the 2-kb Mpl promoter fragment used in the Yall construct lacks some elements required for expression in late megakaryocytes. A putative chromatin-dependent enhancer was described in Mpl intron 6.46  Alternatively, the integration site of the transgene could exert a silencing effect at later stages of megakaryocyte differentiation. Since several independent transgenic strains with the wild-type Mpl cDNA showed thrombocytosis when examined on the Mpl−/− background, this possibility appears less likely. Gene silencing of multicopy transgenes is a well-known phenomenon,47  but there is currently no evidence that such an effect could be selective for late stages of megakaryopoiesis. Finally, a decreased efficiency of the transgenic Mpl mRNA translation in mature megakaryocytes could in addition also contribute to decreasing the Mpl protein levels. This possibility is supported by the observation that the total Mpl mRNA was slightly higher in unfractionated megakaryocytes from Yall;Mpl−/− mice than from wild-type mice, while the Mpl protein levels were approximately the same (Figure 5).

Reduced expression of Mpl protein in platelets was described as a feature in many patients with MPD,48,49  and could be detected in patients with all 3 disease entities (polycythemia vera, essential thrombocythemia [ET], and primary myelofibrosis).50-52  Loss of Mpl protein was also observed in megakaryocytes from patients with MPD,49,53,54  but it is currently unclear whether this specifically affects late megakaryocytes, as we observed in our mice. Recently, a correlation between decreased expression of Mpl and mutant JAK2-V617F was noted.55  In 2 studies, platelets from patients with ET with decreased Mpl protein showed a concomitant reduction of Mpl mRNA resembling our Yall mouse model,48,56  whereas in another study, the platelet Mpl mRNA was normal in patients with MPD with decreased platelet Mpl protein51  (our unpublished data). Furthermore, decreased expression of Mpl protein was also found in patients with hereditary thrombocythemia due to a mutation in the THPO gene,51  and in some cases of reactive thrombocytosis.50  These data suggest that decreased platelet Mpl expression can contribute to thrombocytosis in MPD, but different mechanisms may be effective that need to be further investigated.

A single nucleotide substitution (G1238T) in the Mpl gene that results in a change from lysine to asparagine at amino acid 39 of the Mpl protein (MPLK39N) was found in 7% of African Americans.57  Termed MPLBaltimore, the defect is associated with mild thrombocytosis in heterozygous individuals and more marked thrombocytosis (> 800 × 109/L) in the homozygous state.57,58  These patients also had reduced platelet protein Mpl expression,57  suggesting that a decrease of Mpl protein expression in platelets can contribute to the development of thromobocytosis in human patients. In contrast, mutations in the transmembrane domain (MplS505N) or cytoplasmic juxtamembrane domain (MplW515K/L) cause thrombocytosis through a different mechanism (ie, constitutive activation of Mpl signaling).15,16,59-62  It will be interesting to develop mouse models for these mutations. Our results indicate that such mouse models will need to be examined in the presence of the wild-type Mpl allele, as we demonstrate here that dysregulated expression of Mpl even in the absence of any mutations can be sufficient to cause marked thrombocytosis when examined on an Mpl−/− background.

In summary, our findings support the concept that expression of Mpl protein in platelets and megakaryocytes is an important negative feedback factor in the regulation of thrombopoiesis, and that decreased Mpl expression can cause a phenotype resembling essential thrombocythemia.

An Inside Blood analysis of this article appears at the front of this issue.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

We thank Verena Jäggin for FACS sorting, Dr Kenneth Kaushansky and Dr Norma Fox for providing serum and plasma from Tpo−/− mice, and Dr Francois Lanza and Dr Lucia Kubovcakova for helpful comments on the manuscript.

This work was supported in part by grants 31-53677.98, 3100-066949.01, and 310000-108006/1 from the Swiss National Science Foundation to R.C.S.

Contribution: R.T. and J.C. designed and performed research, analyzed data, and wrote the paper; S.Z., A.W., H.H.-S., C.B., J.S., S.K., F.J.d.S., and C.W.J. performed research and analyzed data; and R.C.S. designed research, analyzed data, and wrote the paper.

Conflict-of-interest disclosure: F.J.d.S. is an employee and stockholder of Genentech. The remaining authors declare no competing financial interests.

Correspondence: Radek C. Skoda, Department of Research, Experimental Hematology, University Hospital Basel, Hebelstrasse 20, 4031 Basel, Switzerland; e-mail: radek.skoda@unibas.ch.

1
Kaushansky
 
K
Thrombopoietin.
N Engl J Med
1998
, vol. 
339
 (pg. 
746
-
754
)
2
Kaushansky
 
K
The molecular mechanisms that control thrombopoiesis.
J Clin Invest
2005
, vol. 
115
 (pg. 
3339
-
3347
)
3
Gurney
 
AL
Carver-Moore
 
K
de Sauvage
 
FJ
Moore
 
MW
Thrombocytopenia in c-mpl-deficient mice.
Science
1994
, vol. 
265
 (pg. 
1445
-
1447
)
4
de Sauvage
 
FJ
Carver-Moore
 
K
Luoh
 
SM
, et al. 
Physiological regulation of early and late stages of megakaryocytopoiesis by thrombopoietin.
J Exp Med
1996
, vol. 
183
 (pg. 
651
-
656
)
5
Fielder
 
PJ
Gurney
 
AL
Stefanich
 
E
, et al. 
Regulation of thrombopoietin levels by c-mpl–mediated binding to platelets.
Blood
1996
, vol. 
87
 (pg. 
2154
-
2161
)
6
Stoffel
 
R
Wiestner
 
A
Skoda
 
RC
Thrombopoietin in thrombocytopenic mice: evidence against regulation at the mRNA level and for a direct regulatory role of platelets.
Blood
1996
, vol. 
87
 (pg. 
567
-
573
)
7
Alexander
 
WS
Roberts
 
AW
Nicola
 
NA
Li
 
R
Metcalf
 
D
Deficiencies in progenitor cells of multiple hematopoietic lineages and defective megakaryocytopoiesis in mice lacking the thrombopoietic receptor c-Mpl.
Blood
1996
, vol. 
87
 (pg. 
2162
-
2170
)
8
Solar
 
GP
Kerr
 
WG
Zeigler
 
FC
, et al. 
Role of c-mpl in early hematopoiesis.
Blood
1998
, vol. 
92
 (pg. 
4
-
10
)
9
Ihara
 
K
Ishii
 
E
Eguchi
 
M
, et al. 
Identification of mutations in the c-mpl gene in congenital amegakaryocytic thrombocytopenia.
Proc Natl Acad Sci U S A
1999
, vol. 
96
 (pg. 
3132
-
3136
)
10
van den Oudenrijn
 
S
Bruin
 
M
Folman
 
CC
, et al. 
Mutations in the thrombopoietin receptor, Mpl, in children with congenital amegakaryocytic thrombocytopenia.
Br J Haematol
2000
, vol. 
110
 (pg. 
441
-
448
)
11
Ballmaier
 
M
Germeshausen
 
M
Schulze
 
H
, et al. 
c-mpl mutations are the cause of congenital amegakaryocytic thrombocytopenia.
Blood
2001
, vol. 
97
 (pg. 
139
-
146
)
12
Souyri
 
M
Vigon
 
I
Penciolelli
 
JF
Heard
 
JM
Tambourin
 
P
Wendling
 
F
A putative truncated cytokine receptor gene transduced by the myeloproliferative leukemia virus immortalizes hematopoietic progenitors.
Cell
1990
, vol. 
63
 (pg. 
1137
-
1147
)
13
Ding
 
J
Komatsu
 
H
Wakita
 
A
, et al. 
Familial essential thrombocythemia associated with a dominant-positive activating mutation of the c-MPL gene, which encodes for the receptor for thrombopoietin.
Blood
2004
, vol. 
103
 (pg. 
4198
-
4200
)
14
Teofili
 
L
Giona
 
F
Martini
 
M
, et al. 
Markers of myeloproliferative diseases in childhood polycythemia vera and essential thrombocythemia.
J Clin Oncol
2007
, vol. 
25
 (pg. 
1048
-
1053
)
15
Pikman
 
Y
Lee
 
BH
Mercher
 
T
, et al. 
MPLW515L is a novel somatic activating mutation in myelofibrosis with myeloid metaplasia.
PLoS Med
2006
, vol. 
3
 pg. 
e270
 
16
Pardanani
 
AD
Levine
 
RL
Lasho
 
T
, et al. 
MPL515 mutations in myeloproliferative and other myeloid disorders: a study of 1182 patients.
Blood
2006
, vol. 
108
 (pg. 
3472
-
3476
)
17
Ziegler
 
S
Burki
 
K
Skoda
 
RC
A 2-kb c-mpl promoter fragment is sufficient to direct expression to the megakaryocytic lineage and sites of embryonic hematopoiesis in transgenic mice.
Blood
2002
, vol. 
100
 (pg. 
1072
-
1074
)
18
Nakamura
 
Y
Takano
 
H
Osawa
 
M
, et al. 
Impaired erythropoiesis in transgenic mice overexpressing a truncated erythropoietin receptor.
Exp Hematol
1998
, vol. 
26
 (pg. 
1105
-
1110
)
19
Wagner
 
K
Kafert-Kasting
 
S
Heil
 
G
Ganser
 
A
Eder
 
M
Inhibition of granulocyte-macrophage colony-stimulating factor receptor function by a splice variant of the common beta-receptor subunit.
Blood
2001
, vol. 
98
 (pg. 
2689
-
2696
)
20
Mitsui
 
T
Watanabe
 
S
Taniguchi
 
Y
, et al. 
Impaired neutrophil maturation in truncated murine G-CSF receptor-transgenic mice.
Blood
2003
, vol. 
101
 (pg. 
2990
-
2995
)
21
Heaney
 
ML
Golde
 
DW
Soluble cytokine receptors.
Blood
1996
, vol. 
87
 (pg. 
847
-
857
)
22
Coers
 
J
Ranft
 
C
Skoda
 
RC
A truncated isoform of c-Mpl with an essential C-terminal peptide targets the full-length receptor for degradation.
J Biol Chem
2004
, vol. 
279
 (pg. 
36397
-
36404
)
23
Skoda
 
RC
Seldin
 
DC
Chiang
 
MK
Peichel
 
CL
Vogt
 
TF
Leder
 
P
Murine c-mpl: a member of the hematopoietic growth factor receptor superfamily that transduces a proliferative signal.
EMBO J
1993
, vol. 
12
 (pg. 
2645
-
2653
)
24
Vigon
 
I
Florindo
 
C
Fichelson
 
S
, et al. 
Characterization of the murine Mpl proto-oncogene, a member of the hematopoietic cytokine receptor family: molecular cloning, chromosomal location and evidence for a function in cell growth.
Oncogene
1993
, vol. 
8
 (pg. 
2607
-
2615
)
25
Arnold
 
JT
Daw
 
NC
Stenberg
 
PE
Jayawardene
 
D
Srivastava
 
DK
Jackson
 
CW
A single injection of pegylated murine megakaryocyte growth and development factor (MGDF) into mice is sufficient to produce a profound stimulation of megakaryocyte frequency, size, and ploidization.
Blood
1997
, vol. 
89
 (pg. 
823
-
833
)
26
Akimoto
 
Y
Ishiyama
 
T
Sano
 
M
, et al. 
[Essential thrombocythemia that transformed to myelofibrosis after three years].
Rinsho Ketsueki
1992
, vol. 
33
 (pg. 
520
-
524
)
27
Jackson
 
CW
Cholinesterase as a possible marker for early cells of the megakaryocytic series.
Blood
1973
, vol. 
42
 (pg. 
413
-
421
)
28
Long
 
MW
Williams
 
N
Immature megakaryocytes in the mouse: morphology and quantitation by acetylcholinesterase staining.
Blood
1981
, vol. 
58
 (pg. 
1032
-
1039
)
29
Burstein
 
SA
Friese
 
P
Downs
 
T
Mei
 
RL
Characteristics of a novel rat anti-mouse platelet monoclonal antibody: application to studies of megakaryocytes.
Exp Hematol
1992
, vol. 
20
 (pg. 
1170
-
1177
)
30
Pillai
 
MM
Venkataraman
 
GM
Kosak
 
S
Torok-Storb
 
B
Integration site analysis in transgenic mice by thermal asymmetric interlaced (TAIL)-PCR: segregating multiple-integrant founder lines and determining zygosity.
Transgenic Res
2008
, vol. 
17
 (pg. 
749
-
754
)
31
Stoffel
 
R
Ziegler
 
S
Ghilardi
 
N
Ledermann
 
B
de Sauvage
 
FJ
Skoda
 
RC
Permissive role of thrombopoietin and granulocyte colony-stimulating factor receptors in hematopoietic cell fate decisions in vivo.
Proc Natl Acad Sci U S A
1999
, vol. 
96
 (pg. 
698
-
702
)
32
Zhang
 
JL
Stenberg
 
PE
Baker
 
G
Levin
 
J
Immunocytochemical identification of murine and human megakaryocyte colonies in soft-agar cultures.
Histochem J
1994
, vol. 
26
 (pg. 
170
-
178
)
33
Kimura
 
S
Roberts
 
AW
Metcalf
 
D
Alexander
 
WS
Hematopoietic stem cell deficiencies in mice lacking c-mpl, the receptor for thrombopoietin.
Proc Natl Acad Sci U S A
1998
, vol. 
95
 (pg. 
1195
-
1200
)
34
White
 
JG
The dense bodies of human platelets: inherent electron opacity of the serotonin storage particles.
Blood
1969
, vol. 
33
 (pg. 
598
-
606
)
35
White
 
JG
Use of the electron microscope for diagnosis of platelet disorders.
Semin Thromb Hemost
1998
, vol. 
24
 (pg. 
163
-
168
)
36
Skoda
 
RC
Seldin
 
DC
Chiang
 
MK
Peichel
 
CL
Vogt
 
TF
Leder
 
P
Murine c-mpl: a member of the hematopoietic growth factor receptor superfamily that transduces a proliferative signal.
EMBO J
1993
, vol. 
12
 (pg. 
2645
-
2653
)
37
Luoh
 
SM
Stefanich
 
E
Solar
 
G
, et al. 
Role of the distal half of the c-Mpl intracellular domain in control of platelet production by thrombopoietin in vivo.
Mol Cell Biol
2000
, vol. 
20
 (pg. 
507
-
515
)
38
Shivdasani
 
RA
Rosenblatt
 
MF
Zucker-Franklin
 
D
, et al. 
Transcription factor NF-E2 is required for platelet formation independent of the actions of thrombopoietin/MGDF in megakaryocyte development.
Cell
1995
, vol. 
81
 (pg. 
695
-
704
)
39
Shivdasani
 
RA
Fielder
 
P
Keller
 
GA
Orkin
 
SH
De Sauvage
 
FJ
Regulation of the serum concentration of thrombopoietin in thrombocytopenic NF-E2 knockout mice.
Blood
1997
, vol. 
90
 (pg. 
1821
-
1827
)
40
Pan
 
D
Schomber
 
T
Kalberer
 
CP
, et al. 
Normal erythropoiesis but severe polyposis and bleeding anemia in Smad4 deficient mice.
Blood
2007
, vol. 
110
 (pg. 
3049
-
3055
)
41
Vigon
 
I
Mornon
 
JP
Cocault
 
L
, et al. 
Molecular cloning and characterization of MPL, the human homolog of the v-mpl oncogene: identification of a member of the hematopoietic growth factor receptor superfamily.
Proc Natl Acad Sci U S A
1992
, vol. 
89
 (pg. 
5640
-
5644
)
42
Li
 
J
Sabath
 
DF
Kuter
 
DJ
Cloning and functional characterization of a novel c-mpl variant expressed in human CD34 cells and platelets.
Cytokine
2000
, vol. 
12
 (pg. 
835
-
844
)
43
Filippi
 
MD
Porteu
 
F
Le Pesteur
 
F
, et al. 
Embryonic stem cell differentiation to hematopoietic cells: a model to study the function of various regions of the intracytoplasmic domain of cytokine receptors in vitro.
Exp Hematol
2000
, vol. 
28
 (pg. 
1363
-
1372
)
44
Emmons
 
RVB
Reid
 
DM
Cohen
 
RL
, et al. 
Human thrombopoietin levels are high when thrombocytopenia is due to megakaryocyte deficiency and low when due to increased platelet destruction.
Blood
1996
, vol. 
87
 (pg. 
4068
-
4071
)
45
Choi
 
ES
Hokom
 
MM
Chen
 
JL
, et al. 
The role of megakaryocyte growth and development factor in terminal stages of thrombopoiesis.
Br J Haematol
1996
, vol. 
95
 (pg. 
227
-
233
)
46
Mignotte
 
V
Deveaux
 
S
Filipe
 
A
Transcriptional regulation in megakaryocytes: the thrombopoietin receptor gene as a model.
Stem Cells
1996
, vol. 
14
 (pg. 
232
-
239
)
47
Garrick
 
D
Fiering
 
S
Martin
 
DI
Whitelaw
 
E
Repeat-induced gene silencing in mammals.
Nat Genet
1998
, vol. 
18
 (pg. 
56
-
59
)
48
Horikawa
 
Y
Matsumura
 
I
Hashimoto
 
K
, et al. 
Markedly reduced expression of platelet c-mpl receptor in essential thrombocythemia.
Blood
1997
, vol. 
90
 (pg. 
4031
-
4038
)
49
Moliterno
 
AR
Hankins
 
WD
Spivak
 
JL
Impaired expression of the thrombopoietin receptor by platelets from patients with polycythemia vera.
N Engl J Med
1998
, vol. 
338
 (pg. 
572
-
580
)
50
Harrison
 
CN
Gale
 
RE
Pezella
 
F
Mire-Sluis
 
A
MacHin
 
SJ
Linch
 
DC
Platelet c-mpl expression is dysregulated in patients with essential thrombocythaemia but this is not of diagnostic value
Br J Haematol
1999
, vol. 
107
 (pg. 
139
-
147
)
51
Kralovics
 
R
Buser
 
AS
Teo
 
SS
, et al. 
Comparison of molecular markers in a cohort of patients with chronic myeloproliferative disorders.
Blood
2003
, vol. 
102
 (pg. 
1869
-
1871
)
52
Antonioli
 
E
Guglielmelli
 
P
Pancrazzi
 
A
, et al. 
Clinical implications of the JAK2 V617F mutation in essential thrombocythemia.
Leukemia
2005
, vol. 
19
 (pg. 
1847
-
1849
)
53
Moliterno
 
AR
Spivak
 
JL
Posttranslational processing of the thrombopoietin receptor is impaired in polycythemia vera
Blood
1999
, vol. 
94
 (pg. 
2555
-
2561
)
54
Tefferi
 
A
Yoon
 
SY
Li
 
CY
Immunohistochemical staining for megakaryocyte c-mpl may complement morphologic distinction between polycythemia vera and secondary erythrocytosis.
Blood
2000
, vol. 
96
 (pg. 
771
-
772
)
55
Moliterno
 
AR
Williams
 
DM
Rogers
 
O
Spivak
 
JL
Molecular mimicry in the chronic myeloproliferative disorders: reciprocity between quantitative JAK2 V617F and Mpl expression.
Blood
2006
, vol. 
108
 (pg. 
3913
-
3915
)
56
Li
 
J
Xia
 
Y
Kuter
 
DJ
The platelet thrombopoietin receptor number and function are markedly decreased in patients with essential thrombocythaemia.
Br J Haematol
2000
, vol. 
111
 (pg. 
943
-
953
)
57
Moliterno
 
AR
Williams
 
DM
Gutierrez-Alamillo
 
LI
Salvatori
 
R
Ingersoll
 
RG
Spivak
 
JL
Mpl Baltimore: a thrombopoietin receptor polymorphism associated with thrombocytosis.
Proc Natl Acad Sci U S A
2004
, vol. 
101
 (pg. 
11444
-
11447
)
58
Standen
 
G
Clench
 
T
Rapid detection of MPL Baltimore using LightCycler technology and melting curve analysis.
Br J Haematol
2008
, vol. 
140
 (pg. 
714
-
716
)
59
Kitamura
 
T
Onishi
 
M
Yahata
 
T
Kanakura
 
Y
Asano
 
S
Activating mutations of the transmembrane domain of MPL in vitro and in vivo: incorrect sequence of MPL-K, an alternative spliced form of MPL [letter].
Blood
1998
, vol. 
92
 (pg. 
2596
-
2597
)
60
Ding
 
J
Komatsu
 
H
Wakita
 
A
, et al. 
Familial essential thrombocythemia associated with a dominant-positive activating mutation of the c-MPL gene, which encodes for the receptor for thrombopoietin.
Blood
2004
, vol. 
103
 (pg. 
4198
-
4200
)
61
Beer
 
PA
Campbell
 
PJ
Scott
 
LM
, et al. 
MPL mutations in myeloproliferative disorders: analysis of the PT-1 cohort.
Blood
2008
, vol. 
112
 (pg. 
141
-
149
)
62
Vannucchi
 
AM
Antonioli
 
E
Guglielmelli
 
P
, et al. 
Characteristics and clinical correlates of MPL 515W>L/K mutation in essential thrombocythemia.
Blood
2008
, vol. 
112
 (pg. 
844
-
847
)

Author notes

*R.T. and J.C. contributed equally to this study.

Sign in via your Institution