Megakaryopoiesis is a complex, stepwise process that takes place largely in the bone marrow. At the apex of the hierarchy, hematopoietic stem cells undergo a number of lineage commitment decisions that ultimately lead to the production of polyploid megakaryocytes. On average, megakaryocytes release 1011 platelets per day into the blood that repair vascular injuries and prevent excessive bleeding. This differentiation process is tightly controlled by exogenous and endogenous factors, which have been the topics of intense research in the hematopoietic field. Indeed, a skewing of megakaryocyte commitment and differentiation may entail the onset of myeloproliferative neoplasms and other preleukemic disorders together with acute megakaryoblastic leukemia, whereas quantitative or qualitative defects in platelet production can lead to inherited platelet disorders. The recent advent of next-generation sequencing has prompted mapping of the genomic landscape of these conditions to provide an accurate view of the underlying lesions. The aims of this review are to introduce the physiological pathways of megakaryopoiesis and to present landmark studies on acquired and inherited disorders that target them. These studies have not only introduced a new era in the fields of molecular medicine and targeted therapies but may also provide us with a better understanding of the mechanisms underlying normal megakaryopoiesis and thrombopoiesis that can inform efforts to create alternative sources of megakaryocytes and platelets.

This review emphasizes how unraveling genomic lesions that underpin malignant and nonmalignant disorders of megakaryopoiesis and thrombopoiesis provides clues on the physiological mechanisms that control normal megakaryocyte differentiation, maturation, and platelet function. This knowledge, moreover, informs and influences the field of therapeutic megakaryopoiesis and platelet production, as discussed in the reviews by Sim et al, Eto and Kunishima, and Woolthuis and Park in this review series.

Megakaryocyte progenitors arise from the lineage restriction of hematopoietic stem cell–derived bipotent megakaryocyte-erythroid progenitors. Proliferating megakaryocyte progenitors terminally differentiate to megakaryocytes, which in turn undergo endomitosis and cytoplasmic maturation. Overall, the process of megakaryocyte development and maturation is called megakaryopoiesis. Mature megakaryocytes in turn produce platelets. Platelet biogenesis (called thrombopoiesis) mainly, but not exclusively,1  occurs through the organization of cytoplasmic extensions (proplatelets) that fragment and are released as platelets into the bloodstream (Figure 1A).

Figure 1

Main molecular mechanisms affected in malignant megakaryopoiesis and platelet function defects. Schematic representation of (A) TPO/MPL signaling pathway together with (B) the most relevant transcriptional regulators of megakaryocyte commitment, differentiation, and maturation. GP2B (CD41) is a component of the GPIIb-IIIa glycoprotein complex. GP9 (CD42a), GP1BA (CD42b), and GP1BB (CD42c) are components of the GPIb-IX-V glycoprotein complex. HSC, hematopoietic stem cell; MEP, megakaryocyte-erythroid progenitor; MK, megakaryocyte.

Figure 1

Main molecular mechanisms affected in malignant megakaryopoiesis and platelet function defects. Schematic representation of (A) TPO/MPL signaling pathway together with (B) the most relevant transcriptional regulators of megakaryocyte commitment, differentiation, and maturation. GP2B (CD41) is a component of the GPIIb-IIIa glycoprotein complex. GP9 (CD42a), GP1BA (CD42b), and GP1BB (CD42c) are components of the GPIb-IX-V glycoprotein complex. HSC, hematopoietic stem cell; MEP, megakaryocyte-erythroid progenitor; MK, megakaryocyte.

Close modal

Megakaryocyte-erythroid progenitor commitment toward the megakaryocyte lineage and maturation are tightly regulated through transcriptional and epigenetic mechanisms, which act in concert with extrinsically induced signal transduction events. Thrombopoietin (TPO) and its receptor, MPL, play a primary role in the extrinsic control of megakaryopoiesis. MPL is a homodimeric type I receptor that requires the tyrosine kinase Janus kinase 2 (JAK2) for signaling. Importantly, the TPO-MPL axis governs hematopoietic stem cell proliferation and megakaryocyte commitment, but it is dispensable for megakaryocyte maturation and platelet production1,2  (Figure 1A).

Several transcription factors drive megakaryopoiesis and platelet production. Among them, GATA1 is essential for proper erythroid and megakaryocyte differentiation, as demonstrated by the impaired maturation of GATA1 embryonic stem cell–derived erythro-megakaryocytic progenitors3  (Figure 1A). Indeed, germ line missense mutations in GATA1 underlie X-linked thrombocytopenia and anemia.4  Other transcription factors act in a coordinated, balanced manner to drive megakaryocyte-erythroid progenitor fate decision. The proto-oncogene MYB is a major regulator of this process. MYB favors erythropoiesis by inducing the expression of the erythroid transcription factor KLF1.5  In this way, MYB affects the functional antagonism between KLF1 that supports erythropoiesis and the transcription factor FLI1 that supports megakaryopoiesis.6,7  In addition, MYB induces the microRNA miR-486-3p, which in turn lowers the expression of the transcription factor MAF,8  which promotes megakaryopoiesis. The overall result is that, although it is required for the erythropoiesis, MYB constrains the megakaryocyte-erythroid progenitor commitment toward the megakaryocyte lineage. This role is experimentally supported by MYB knockdown or loss-of-function experiments, which show an enhanced megakaryopoiesis and platelet production.5,9  Interestingly, TPO-MPL signaling mimics experimental MYB knockdown, because MPL activation induces the expression of miR-150-5p, which in turn lowers MYB expression,10  thus enhancing megakaryopoiesis11  (Figure 1A). Strikingly, the inactivation of MYB transcriptional activity induces platelet production at supraphysiological levels, even in MPL−/− mice (ie, in the absence of MPL signaling).9 

During megakaryocyte lineage commitment, MYB downregulation enables FLI1 expression. FLI1 acts in concert with GATA1-FOG1 and ETS112,13  to transactivate the expression of several megakaryocyte-specific and platelet-specific receptors such as MPL, ITGA2B (ie, GP2B or CD41), and GP9 (ie, CD42a) (Figure 1B).

A number of transcription factors are similarly responsible for the transcriptional control of megakaryocyte maturation and platelet production (Figure 1B). Among them, RUNX1 affects ploidization and proplatelet formation during megakaryocyte maturation by regulating MYH9, MYL9, and MYH10 expression14,15  (Figure 1B). Similarly, GATA1 regulates polyploidization by driving expression of the gene coding for cyclin D1 (CCND1).16  GATA1 also transactivates the expression of the transcription factor p45 NFE2.17  NFE2 in turn induces the expression of several genes critical for proplatelet formation and platelet shedding such as TUBB1 (Figure 1B), which encodes for the tubulin β-1 chain, a major component of microtubules.18  Accordingly, p45 NFE2−/− mice lack circulating platelets.19 

Increasing knowledge of both extrinsic cues and intrinsic mechanisms that underpin megakaryopoiesis and thrombopoiesis broadens the panel of targets that could be used to overcome some issues such as yield in therapeutic, large-scale platelet production. For example, interleukin (IL)-1α was recently discovered to drive platelet production by megakaryocyte rupture. This mechanism is an alternative to TPO-induced thrombopoiesis via proplatelet formation, is activated in response to acute platelet needs, and can provide a 20-fold higher yield of platelets.1  This recent insight is emblematic of how unraveling the mechanisms underlying physiological megakaryopoiesis and platelet production can point to new ways for developing and optimizing large-scale platelet production protocols.

This section is focused on the genomic lesions causing malignant megakaryopoiesis that most clearly inform us about normal megakaryopoiesis. Preleukemic disorders characterized by a skewed megakaryopoiesis—namely, Philadelphia chromosome (Ph)-negative myeloproliferative neoplasms (MPNs), 5q- syndrome, and refractory anemia with ring sideroblasts associated with thrombocytosis (RARS-T)—will be reviewed first. Next, acute megakaryoblastic leukemia (AMKL) will be discussed as representing a possible scenario for the progression of the aforementioned preleukemic disorders to acute myeloid leukemia (AML). The diseases presented here share a common feature: the abnormal expansion of megakaryopoiesis. The identification of the underlying molecular defects capitalizes on a few mechanisms that entail an enhancement of megakaryopoiesis and therefore represent a target for overcoming some limitations (eg, the yield) in therapeutic megakaryocyte and platelet production.

Ph-negative MPNs

Classical Ph-negative MPNs are a heterogeneous group of clonal hematopoietic disorders characterized by an excessive production of mature myeloid cells and a tendency to progress to AML. The 2008 World Health Organization classification identifies three main diseases—polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF)—based on an expanded lineage in MPN patients.20  PV is characterized by erythrocytosis, whereas ET and PMF have abnormal megakaryopoiesis with alterations in platelet count. In particular, ET patients exhibit thrombocytosis (platelet count >450 × 109/L) and megakaryocyte proliferation in the absence of relevant bone marrow fibrosis, whereas PMF patients display bone marrow with megakaryocyte hyperplasia and dysplasia along with reticulin and/or collagen fiber deposition (Figure 2).

Figure 2

PMF, ET, RARS-T, and 5q- syndrome. The World Health Organization classification, the mutational landscape, and the distinctive clinical and morphologic features for the 4 malignant diseases are described. The schematic representation of megakaryocyte morphology, as a hallmark distinctive of PMF, ET, RARS-T, and 5q- syndrome, is shown at the bottom of the figure.

Figure 2

PMF, ET, RARS-T, and 5q- syndrome. The World Health Organization classification, the mutational landscape, and the distinctive clinical and morphologic features for the 4 malignant diseases are described. The schematic representation of megakaryocyte morphology, as a hallmark distinctive of PMF, ET, RARS-T, and 5q- syndrome, is shown at the bottom of the figure.

Close modal

Given the role of TPO as the main driver of megakaryocyte commitment and differentiation, most efforts to depict the genomic landscape of MPNs have focused on identifying genetic lesions that alter TPO signaling. The identification of a somatic gain-of-function point mutation in the nonreceptor tyrosine kinase JAK221-24  is considered a major milestone for the MPN field, because the JAK2V617F mutation is present in ∼95% of PV patients and in ∼50% to 60% of ET and PMF patients (Figure 2).21-24  JAK2 associates with the receptors for TPO, erythropoietin, and granulocyte colony-stimulating factor. Importantly, this amino acid substitution abrogates the negative regulatory function of the JH2 domain of JAK2, leading to constitutive JAK2 activation and subsequent cytokine-independent signaling. In vivo studies have provided a deeper insight into the mechanism by which the same lesion can cause 3 different phenotypes and suggest that gene dosage affects the disease phenotype.25  Indeed, Li et al26  demonstrated that acquisition of the homozygous JAK2V617F mutation drives a switch from an ET-like to a PV-like phenotype in knockin mice.

Somatic MPL mutations exist in ∼5% to 10% of ET and PMF patients27-30  (Figure 2), whereas no THPO mutations have been detected in MPNs thus far.31 MPL genetic lesions are most often gain-of-function mutations (eg, MPLW515L and MPLW515K) that lead to cytokine-independent MPL activation and TPO hypersensitivity.27,28  Other somatic mutations in MPL have been reported at lower frequencies in MPN patients (MPLW515A, MPLW515R, and MPLS505N).28-30 

In 2013, whole-exome sequencing revealed that somatic mutations in the calreticulin (CALR) gene occurs in 14% to 35% of PMF patients and in 25% to 27% of ET patients, all of which are JAK2 and MPL unmutated32,33  (Figure 2). CALR controls the folding of newly synthesized glycoproteins and acts as a major Ca2+ binding and storage protein in the endoplasmic reticulum. CALR somatic lesions frequently consist of frameshift mutations in exon 9, resulting in the loss of the C-terminal KDEL endoplasmic reticulum retention signal and a partial dislocation of the CALR protein.32  Although more than 50 different CALR insertions/deletions (indels) have been identified, only 2 of them—del52 (type 1) and ins5 (type 2)—collectively account for ∼85% of CALR mutations. Both CALRdel52 and CALRins5 mutants drive the expansion of megakaryopoiesis and thrombopoiesis leading to a human ET-like disease in a retroviral mouse model.34  Indeed, CALR mutants drive the constitutive activation of JAK2-STAT signaling through MPL35  with consequent TPO hypersensitivity. These data are consistent with the observation that JAK-STAT signaling is also aberrantly activated in patients with CALR mutations.32,36  Important differences between CALRdel52- and CALRins5-driven phenotypes in mice are detected, with CALRdel52 additionally expanding the stem cell compartment and causing myelofibrosis and more severe thrombocytosis than CALRins5.34,35  Of note, these features suggest that other molecular mechanisms besides the MPL pathway, such as Ca2+-dependent signaling, could be differently perturbed by CALR mutants.

However, approximately 10% of PMF and ET patients do not carry mutations in any of the 3 mutations (JAK2, MPL, CALR) described above (Figure 2). Somatic lesions in other genes involved in JAK-STAT signaling have been found in MPN patients. For instance, loss-of-function mutations in LNK and CBL, which encode a JAK2 inhibitor and a JAK2/MPL/EPOR–directed E3 ubiquitin ligase, respectively, have been identified in 3% to 6% of MPN patients and may co-occur alongside the JAK2V617F mutation.25,37  Additional lesions have also been associated with MPN pathogenesis and progression25,37,38  (Figure 2).

Taken together, these findings identify constitutive activation of MPL signaling as a mechanism shared by JAK2, MPL, and CALR driver mutations that affects megakaryopoiesis and thrombopoiesis. In addition, these finding further elucidate the role of key modulators of MPL-driven megakaryopoiesis. These data therefore pinpoint novel targets for the manipulation and tuning of the MPL signaling pathway to improve the yield of large-scale platelet generation systems as discussed in other reviews in this series. It is also noteworthy that, even though PMF and ET patients carry the same driver mutations (ie, JAK2, MPL, and CALR), their megakaryocytes show considerable morphologic differences (Figure 2). Therefore, additional mechanisms could be engaged to explain the differences in the skewing of megakaryocyte maturation between PMF and ET. Interestingly, the investigation of these mechanisms could also provide novel insights into the intrinsic and extrinsic signals that control physiological megakaryocyte maturation.

Genome-wide association studies have identified germ line susceptibility genes predisposing to MPN development, such as the JAK2 haplotype called 46/1 (or GGCC) and its associated single nucleotide polymorphism (rs10974944), which represents the main predisposition factor for both sporadic and familial MPNs with acquired JAK2V617F mutations.25  Additional MPN-predisposing variants have recently been identified. Among them, it is worth mentioning a single nucleotide polymorphism (rs9376092) close to the MYB locus associated with reduced MYB expression and strongly related to an ET rather than a PV phenotype in JAK2V617F-positive MPNs.39  These findings strongly suggest that MYB expression levels can affect the ET vs PV phenotype determination in JAK2V617F-positive MPNs, consistent with the role of MYB in megakaryocyte vs erythroid lineage choice illustrated above.5,8 

Recent genome-wide transcriptome studies have further characterized the molecular mechanisms underlying MPNs by profiling CD34+ hematopoietic stem and progenitor cells40-44  and have highlighted the abnormal expression of coding and noncoding transcripts involved in megakaryopoiesis and platelet production in PMF and ET patients (eg, NFE2, CD9, CXCR4, WT1, and miR-34a-5p).36,40  For instance, miR-155-5p is overexpressed in PMF CD34+ hematopoietic stem and progenitor cells and targets the polycomb repressive complex 2 component JARID2 to support the abnormal megakaryocyte lineage expansion seen in these patients.45  Similarly, miR-28, which targets MPL messenger RNA and inhibits megakaryocyte terminal differentiation, is overexpressed in MPN megakaryocytes and platelets that frequently display MPL downregulation.46,47 

These results provide important insights into how miRNA-mediated posttranscriptional control of gene expression affects megakaryopoiesis and thrombopoiesis in physiological and pathological conditions. They also suggest that modulation of miRNA expression might represent an important step to improve platelet production for therapeutic purposes.

As in MPNs, the MPL signaling pathway is also deregulated in hereditary thrombocytosis, a familial polyclonal disorder that results from germ line mutations in the THPO and MPL genes48  (Table 1). Hereditary thrombocytosis-associated THPO mutations generally lead to increased messenger RNA translation, which eventually results in cytokine overproduction. Of note, germ line and sporadic mutations in hereditary thrombocytosis and MPNs, respectively, occur in the same key megakaryocytic genes (MPL and JAK2), even though different residues are hit (Table 1).

Table 1

Genes involved in hereditary thrombocytosis

GeneProteinChromosome locationMutationInheritanceMolecular mechanism
THPO Thrombopoietin (TPO) 3q27 G>C in the splice donor site of intron 3 Autosomal dominant The mutation disrupts the inhibitory uORF leading to increase in mRNA translation. 
   G>T substitution in 5′UTR Autosomal dominant The mutation disrupts the inhibitory uORF leading to increase in mRNA translation. 
   Deletion of single G in 5′UTR Autosomal dominant The mutation disrupts the inhibitory uORF leading to increase in mRNA translation. 
   A>G mutation in intron 3 Autosomal dominant The A>G substitution in the +1 position of the splice donor of intron 3 leads to increase in mRNA translation. 
   T>C at the splice donor site of intron 2 Autosomal dominant The splice donor mutation leads to exon 2 skipping and loss of inhibitory 5′UTR sequence, leading to increased TPO expression. 
MPL Thrombopoietin receptor, MPL 1p34 S505N Autosomal dominant TPO-independent constitutive activation of MPL. 
   K39N (also called “MPL-Baltimore”) Autosomal dominant with incomplete penetrance This mutation is associated with incomplete processing and a reduction in MPL protein. 
   P106L Autosomal recessive The MPL P106L shows constitutive activity that can be further stimulated by TPO. 
   W515R Autosomal dominant The mutation increases MPL activity but to a lesser extent than the somatic MPLW515K/L/A mutations observed in MPNs. 
JAK2 Janus kinase 2, JAK2 9p24 V617I Autosomal dominant JAK2V617I has weak constitutive signaling compared with JAK2V617F because of reduction in the threshold for cytokine-induced activation. 
   R867Q Autosomal dominant The mutation in JH1 domain abolishes a salt bridge, which is present in the inactive JH1 and is lost upon activation. 
   S755R/R938Q Autosomal dominant The S755R mutation interferes with an important salt bridge and inhibits the function of JH2 domain. The R938Q is located in a sensitive region of the JH1 near the ATP loop and substrate access site. 
   R564Q Autosomal dominant JAK2 R564Q exhibits similar levels of increased kinase activity compared with JAK2V617F but fewer growth-promoting effects. 
   H608N Autosomal dominant The mutation in this region may abrogate the function of the inhibitory JH2 domain, resulting in increased kinase activity. 
GeneProteinChromosome locationMutationInheritanceMolecular mechanism
THPO Thrombopoietin (TPO) 3q27 G>C in the splice donor site of intron 3 Autosomal dominant The mutation disrupts the inhibitory uORF leading to increase in mRNA translation. 
   G>T substitution in 5′UTR Autosomal dominant The mutation disrupts the inhibitory uORF leading to increase in mRNA translation. 
   Deletion of single G in 5′UTR Autosomal dominant The mutation disrupts the inhibitory uORF leading to increase in mRNA translation. 
   A>G mutation in intron 3 Autosomal dominant The A>G substitution in the +1 position of the splice donor of intron 3 leads to increase in mRNA translation. 
   T>C at the splice donor site of intron 2 Autosomal dominant The splice donor mutation leads to exon 2 skipping and loss of inhibitory 5′UTR sequence, leading to increased TPO expression. 
MPL Thrombopoietin receptor, MPL 1p34 S505N Autosomal dominant TPO-independent constitutive activation of MPL. 
   K39N (also called “MPL-Baltimore”) Autosomal dominant with incomplete penetrance This mutation is associated with incomplete processing and a reduction in MPL protein. 
   P106L Autosomal recessive The MPL P106L shows constitutive activity that can be further stimulated by TPO. 
   W515R Autosomal dominant The mutation increases MPL activity but to a lesser extent than the somatic MPLW515K/L/A mutations observed in MPNs. 
JAK2 Janus kinase 2, JAK2 9p24 V617I Autosomal dominant JAK2V617I has weak constitutive signaling compared with JAK2V617F because of reduction in the threshold for cytokine-induced activation. 
   R867Q Autosomal dominant The mutation in JH1 domain abolishes a salt bridge, which is present in the inactive JH1 and is lost upon activation. 
   S755R/R938Q Autosomal dominant The S755R mutation interferes with an important salt bridge and inhibits the function of JH2 domain. The R938Q is located in a sensitive region of the JH1 near the ATP loop and substrate access site. 
   R564Q Autosomal dominant JAK2 R564Q exhibits similar levels of increased kinase activity compared with JAK2V617F but fewer growth-promoting effects. 
   H608N Autosomal dominant The mutation in this region may abrogate the function of the inhibitory JH2 domain, resulting in increased kinase activity. 

ATP, adenosine triphosphate; mRNA, messenger RNA; uORF, upstream open reading frame.

5q- syndrome.

Myelodisplastic syndromes (MDSs) are clonal preleukemic conditions characterized by ineffective hematopoiesis that frequently progress to AML. The 5q- syndrome is an MDS associated with isolated deletion on the long arm of chromosome 5 (del(5q)). Although ∼40% to 65% of MDS patients show thrombocytopenia, patients with the 5q- syndrome are characterized by normal or high platelet count and hypolobulated megakaryocytes in the bone marrow in addition to macrocytic anemia.49 

Over the past few years, the pathogenic roles of specific genes located in the 5q- syndrome commonly deleted region, including the casein kinase 1A gene (CSNK1A1), the ribosomal gene RPS14, and the miRNA genes miR-145 and miR-146a (Figure 2) have been unveiled. In particular, the heterozygous inactivation of CSNK1A1 was recently demonstrated to drive hematopoietic stem cell expansion in mice, suggesting that CSNK1A1 haploinsufficiency may play a role in the initial clonal expansion in patients with the 5q- syndrome.50  In addition, loss of the ribosomal gene RPS14 is responsible for dyserythropoiesis and anemia, whereas deletions affecting miR-145 and miR-146a genes are associated with the thrombocytosis and megakaryocyte dysplasia observed in some patients with 5q- syndrome. Of note, miR-145 targets FLI1, whose expression levels are increased in patients with del(5q) MDS. However, the most striking results involving miR-145 and miR-146a in 5q- pathogenesis come from miR-145/miR-146a double-knockdown mice that show upregulation of the miR-145/miR-146a target TRAF6, and subsequent ectopic TRAF6-mediated induction of IL-6 expression. The increase in IL-6 levels in turn likely accounts for megakaryocyte dysplasia and thrombocytosis through a paracrine mechanism.51  These data collectively identify a cell-nonautonomous mechanism that underpins 5q- syndrome-related megakaryopoiesis and thrombopoiesis defects. They also underline the importance of extrinsic factors such as IL-6 in regulating megakaryocyte maturation and platelet production, with important implications for the optimization of ex vivo platelet production protocols.

RARS-T.

RARS-T is a rare condition with features of both MDSs and MPNs.20  It is characterized by anemia with dysplastic erythropoiesis, ring sideroblasts >15% of erythroid precursors, and thrombocytosis associated with the increased proliferation of large atypical MPN-like megakaryocytes52  (Figure 2). Even though associated with JAK2V617F at first,53  the recent identification of somatically acquired mutations in the RNA splicing protein SF3B1 observed in 70% to 90% of MDS or MDS-MPN patients with ring sideroblasts has been the major breakthrough in the field.54,55 SF3B1 encodes an essential U2snRNP spliceosomal complex component that participates in normal RNA splicing. Mutations in SF3B1 are considered the first hit in RARS-T patients and account for the myelodysplastic features, although a second hit to JAK2, MPL, or CALR is necessary to confer a myeloproliferative phenotype52  (Figure 2). Indeed, somatic CALR mutations have been reported in patients with RARS-T in addition to those with PMF and ET,32,56,57  further emphasizing the link between CALR mutations and abnormal megakaryopoiesis and thrombopoiesis. However, TET2, DNMT3A, and ASXL1, which encode for epigenetic modifiers, are the most frequently mutated genes outside of SF3B1 and JAK2 in RARS-T.

Overall these data further confirm the connection between the constitutive activation of MPL signaling through JAK2, MPL, or CALR mutations and the expansion of megakaryopoiesis and platelet production as a pathogenetic mechanism shared by MPNs and RARS-T.

AMKL.

AMKL is a subtype of AML characterized by the presence of numerous abnormal megakaryoblasts in the bone marrow as well as by considerable myelofibrosis. AMKL displays a bimodal age distribution and is more frequent in infants (3% to 14% of AML patients) than adults (1% of AML patients). Three main subtypes of AMKL can be distinguished: pediatric Down syndrome–related AMKL, pediatric non-Down syndrome AMKL, and adult AMKL. Pediatric patients often exhibit de novo AMKL, whereas adult AMKL frequently arises after chemotherapy or is secondary to MPNs or MDSs.58,59 

AMKLs are commonly characterized by a higher incidence of complex karyotypes compared with other AML subtypes.58  There are several cytogenetic aberrations that are common in adult AMKL (t(9;22)(q34;q11), 3q21q26, −5/del(5q), −7/del(7q), and i(12)(p10)), but some pediatric mutations are also observed as described below.58  Of note, some of these lesions, such as del(5q) or del(7q), may also be found in MDS patients.

Although there are numerous cytogenetic aberrations that commonly cause adult AMKL, the most insightful mutations that inform us about normal megakaryopoiesis are found in children with Down syndrome who develop AMKL. These children often have a precedent indolent transient myeloproliferative disorder characterized by the accumulation of megakaryoblasts in the peripheral blood and by a moderate-to-high leukocytosis. Transient myeloproliferative disorder myeloid cells show just a few somatic lesions, with trisomy 21 (t21) and GATA1 mutations being the only recurrent alterations. The overexpression of some genes or microRNAs located in chromosome 21, such as DYRK1A,60 ERG,61  and miR-125b,62  has been shown to induce AMKL in mice. Recently, Ng et al63  have further characterized the role of the transcription factor ERG in a Down syndrome mouse model, suggesting that ERG is a key regulator of the erythroid-megakaryocytic cell fate in early progenitors, and its trisomy is sufficient to confer myeloproliferation in mice. In 2002, Wechsler and colleagues demonstrated that most of these patients have multiple, independent GATA1 mutations, resulting in a shorter GATA1 protein (GATA1s) with an N-terminal truncation.64  Interestingly, neither a mouse model carrying a complete copy of chromosome 21 nor GATA1s knockin mice develop any form of leukemia but show only an increased megakaryopoiesis. Surprisingly, the combination of these 2 hits further enhances the proliferation of megakaryocyte progenitors without leading to any overt leukemia phenotype.65,66 

The pediatric non-Down syndrome AMKLs generally show copy number variations and numerical chromosomal abnormalities. Approximately 30% of these patients carry the OTT-MAL fusion gene.67,68  The conditional OTT-MAL knockin mouse model69  recapitulates an AMKL-like phenotype, although with a low penetrance. Functionally, the OTT-MAL fusion protein activates RBPJ-mediated transcriptional regulation, leading to increased proliferation and megakaryocyte lineage skewing. Because RBPJ is a key mediator of canonical Notch signaling, this work further highlights the importance of this pathway whose activation favors megakaryopoiesis in mice.70  However, Poirault-Chassac et al71  have reported a suppression effect of Notch signaling on human megakaryopoiesis. Although puzzling, these findings might pave the way to design better systems to obtain and/or culture megakaryocytes ex vivo. Of note, human CD34+ cells expanded in vitro in the presence of Notch ligands have already been used as transplantation units in clinical trials.72 

Overall, the study of neoplastic megakaryopoiesis throughout the spectrum of malignancies affecting this lineage has allowed researchers to improve their understanding of how development of megakaryocytes can be driven by intrinsic and external stimuli. In particular, as discussed elsewhere in this review series, knowledge of cytokines (ie, TPO, IL-6), miRNAs (ie, miR-155, miR-28), and pathways (ie, JAK-STAT, Ca2+-dependent signaling, Notch pathway) can be exploited and applied to the production of megakaryocytes and platelets for therapy.

The transition of megakaryocytes to platelets for therapeutic purposes ideally requires optimal platelet function. Inherited platelet function disorders frequently entail alterations in platelet number and size.73  The characterization of their genomic landscape has provided much insight into the molecular mechanisms crucial for proper platelet production and function. Here we discuss the genomic landscape of the inherited platelet function disorders (Table 2), because it could provide important clues into how to improve and preserve the production of functional platelets for therapeutic purposes. Defects primarily responsible for thrombocytopenia are covered in the review by Eto and Kunishima.

Table 2

Principal inherited platelet function disorders and underlying genomic defects

DisorderGeneChromosome locationMain mode of inheritance
Adhesion defects    
 Bernard-Soulier syndrome GP1BA 17p13.2 Autosomal recessive 
 GP1BB 22q11.21 Autosomal recessive 
 GP9 3q21.3 Autosomal recessive 
 Platelet-type von Willebrand disease GP1BA 17p13.2 Autosomal dominant 
 GP6 deficiency GP6 19q13.4 Autosomal recessive 
Receptors and signaling defects    
 ADP receptor deficiency P2RY12 3q25.1 Autosomal recessive 
 Thromboxane A2 receptor deficiency TBXA2R 19p13.3 Autosomal recessive 
 PAR-4 receptor defects F2RL3 19p13.11 Autosomal dominant 
Storage pool disease    
 Gray platelet syndrome NBEAL2 3p21.31 Autosomal recessive 
GFI1B 9q34.13 Autosomal dominant 
 Quebec platelet syndrome PLAU 10q22.2 Autosomal dominant 
 Hermansky-Pudlak syndrome HPS1 10q23.1-q23.3 Autosomal recessive 
 AP3B1 5q14.1 Autosomal recessive 
 HPS3 3q24 Autosomal recessive 
 HPS4 22cen-q12.3 Autosomal recessive 
 HPS5 11p14 Autosomal recessive 
 HPS6 10q24.32 Autosomal recessive 
 HPS7 6p22.3 Autosomal recessive 
 HPS8 19q13.32 Autosomal recessive 
 HPS9 15q21.1 Autosomal recessive 
 Chediak-Higashi syndrome LYST 1q41.3 Autosomal recessive 
Aggregation defects    
 Glanzmann thrombasthenia ITGA2B 17q21.31 Autosomal recessive 
 ITGB3 17q21.32 Autosomal recessive 
 Glanzmann thrombasthenia-like disorder RASGRP2 11q13 Autosomal recessive 
 Leukocyte adhesion deficiency-III syndrome FERMT3 11q13.1 Autosomal recessive 
Procoagulant activity defects    
 Scott syndrome TMEM16F 12q12 Autosomal dominant 
DisorderGeneChromosome locationMain mode of inheritance
Adhesion defects    
 Bernard-Soulier syndrome GP1BA 17p13.2 Autosomal recessive 
 GP1BB 22q11.21 Autosomal recessive 
 GP9 3q21.3 Autosomal recessive 
 Platelet-type von Willebrand disease GP1BA 17p13.2 Autosomal dominant 
 GP6 deficiency GP6 19q13.4 Autosomal recessive 
Receptors and signaling defects    
 ADP receptor deficiency P2RY12 3q25.1 Autosomal recessive 
 Thromboxane A2 receptor deficiency TBXA2R 19p13.3 Autosomal recessive 
 PAR-4 receptor defects F2RL3 19p13.11 Autosomal dominant 
Storage pool disease    
 Gray platelet syndrome NBEAL2 3p21.31 Autosomal recessive 
GFI1B 9q34.13 Autosomal dominant 
 Quebec platelet syndrome PLAU 10q22.2 Autosomal dominant 
 Hermansky-Pudlak syndrome HPS1 10q23.1-q23.3 Autosomal recessive 
 AP3B1 5q14.1 Autosomal recessive 
 HPS3 3q24 Autosomal recessive 
 HPS4 22cen-q12.3 Autosomal recessive 
 HPS5 11p14 Autosomal recessive 
 HPS6 10q24.32 Autosomal recessive 
 HPS7 6p22.3 Autosomal recessive 
 HPS8 19q13.32 Autosomal recessive 
 HPS9 15q21.1 Autosomal recessive 
 Chediak-Higashi syndrome LYST 1q41.3 Autosomal recessive 
Aggregation defects    
 Glanzmann thrombasthenia ITGA2B 17q21.31 Autosomal recessive 
 ITGB3 17q21.32 Autosomal recessive 
 Glanzmann thrombasthenia-like disorder RASGRP2 11q13 Autosomal recessive 
 Leukocyte adhesion deficiency-III syndrome FERMT3 11q13.1 Autosomal recessive 
Procoagulant activity defects    
 Scott syndrome TMEM16F 12q12 Autosomal dominant 

Adhesion defects

Bernard-Soulier syndrome (BSS) is a rare disorder characterized by thrombocytopenia with giant platelets and bleeding tendency owing to a platelet adhesion deficiency. BSS is caused by qualitative or quantitative defects in the platelet GPIb-IX-V complex that includes GPIbA, GPIbB, GPIX, and GPV proteins (Figure 3) and binds the von Willebrand factor (VWF). The most comprehensive characterization of the genomic defects underlying BSS led to the identification of 112 different mutations in the GP1BA, GP1BB, or GP9 genes.74,75  With a few exceptions of monoallelic BSS resulting from dominant mutations (eg, the Bolzano mutation affecting GP1BA), biallelic GP1BA, GP1BB, or GP9 lesions are usually found in BSS patients. Most of them impair GPIb-IX-V complex assembly and/or trafficking but more rarely affect VWF binding ability.75-77 

Figure 3

Schematic cartoon representing the proteins mutated in inherited platelet function disorders. Light blue ovoids represent α-granules, dark blue spheres represent δ-granules. LAD-III, leukocyte adhesion deficiency-III syndrome; VWD, von Willebrand disease.

Figure 3

Schematic cartoon representing the proteins mutated in inherited platelet function disorders. Light blue ovoids represent α-granules, dark blue spheres represent δ-granules. LAD-III, leukocyte adhesion deficiency-III syndrome; VWD, von Willebrand disease.

Close modal

Megakaryocytes from both BSS patients78  and GP1BB−/− BSS mouse models79  show that GPIb-IX-V deficiency underpins an impaired proplatelet formation that in turn accounts for macrothrombocytopenia. Notably, the proplatelet formation defects are independent from VWF binding. Indeed, megakaryocytes from BSS patients display an impaired proplatelet formation in vitro not only when megakaryocytes are cultured in adhesion on VWF or fibrinogen-coated surfaces but also when megakaryocytes are cultured in suspension. These observations suggest that the proplatelet formation defects cannot be exclusively ascribed to the deficient interaction of GPIb-IX-V with extracellular ligands (eg, P-selectin in addition to VWF) but rather are intrinsic to megakaryocytes. Additional GPIb-IX-V–mediated actomyosin-to-plasma membrane connections might be involved.77,79 

In contrast to the loss-of-function mutations in GPIb-IX-V components that cause BSS, a few gain-of-function GP1BA mutations underlie the platelet hyperresponsiveness observed in platelet-type von Willibrand disease, a rare autosomal dominant disorder. Platelet-type von Willebrand disease patients display an excessive platelet-VWF interaction that exhausts the ability of platelets to bind to fibrinogen upon stimulation and leads to mild to moderate bleeding.73,80,81 

Receptors and signaling defects

Defects in signaling pathways that control platelet secretion and aggregation are the most common cause of mild to moderate bleeding in inherited platelet function disorders. Among them, mutations in P2RY12 and TBXA2R genes coding for G-protein–coupled receptors (P2Y12 and TBXA2R, respectively) for soluble agonists adenosine 5′-diphosphate (ADP) and thromboxane A2 have provided important insights into the molecular mechanisms underlying platelet activation.

Multiple P2RY12 genomic lesions have been described as causing decreased and reversible ADP-induced platelet aggregation. The majority of lesions are nonsense and missense mutations, the latter affecting ADP binding to P2Y12 or P2Y12 receptor trafficking and function.73,82  Most of the qualitative defects identified (eg, the recently described P2RY12H187Q mutation that impairs ADP binding83 ) have shed new light on P2Y12 structure-function relationships.

Although extremely rare, genomic lesions in TBXA2R similarly account for defects in TXA2-mediated platelet aggregation and underpin a mild bleeding phenotype.76  Genomic lesions that lead to the loss of TBXA2R expression or disrupt the TBXA2R cell surface expression, ligand binding affinity, or G-protein coupling have not been reported till now.73,82 

Worthy of mention is a recently uncovered missense mutation in F2RL3 gene, coding for the PAR-4 receptor that, upon thrombin activation, triggers integrin αIIbβ3 activation. The identified F2RL3 defect leads to an impaired thrombin-induced platelet response84  (Figure 3).

Storage pool diseases

Storage pool diseases are inherited defects of secretion from storage organelles (α-granules and δ-granules; Figure 3). α-granules are the most abundant platelet organelles. α-granule protein release triggers platelet adhesion, hemostasis, and wound healing.73  Gray platelet syndrome is the main inherited platelet function defect resulting from defective α-granule biogenesis, protein packaging, and storage.76  Patients with gray platelet syndrome display mild to moderate bleeding, myelofibrosis, and splenomegaly. In 2011, exome and RNA sequencing studies from 3 independent groups led to the identification of NBEAL2 hypomorphic mutations as the genetic lesion underlying autosomal recessive gray platelet syndrome.85-87  By an as yet unclarified mechanism, NBEAL2 is involved in α-granule formation and trafficking. It is noteworthy that NBEAL2−/− gray platelet syndrome mouse models reveal the role of α-granule protein deficiency and megakaryocyte proinflammatory phenotypes that drive the development of bone marrow fibrosis.88  However, NBEAL2 mutations cannot explain all gray platelet syndrome cases. Interestingly, Monteferraio et al89  recently identified a nonsense mutation in the GFI1B gene that codes for a transcriptional regulator of erythroid and megakaryocyte development as being responsible for an autosomal dominant form of gray platelet syndrome coupled with red cells defects.77,84,89 

Storage pool diseases affecting δ-granule secretion lead to platelet aggregation defects and bleeding diatheses.73  When associated with defects in other lysosome-related organelles, they entail clear phenotypic features. Notably, melanosomal defects account for the lack of skin and hair pigmentation in patients with Hermansky-Pudlak syndrome and Chediak-Higashi syndrome.76,80  Defects in formation, shuttle, and release of platelet δ-granules account for the bleeding tendency in patients with Hermansky-Pudlak syndrome. Genomic lesions in 9 genes (HPS1, AP3B1, HPS3-9) that code for biogenesis of lysosome-related organelle complexes 1, 2, and 3 (BLOC-1, BLOC-2, and BLOC-3) and AP-3 complex subunits have been linked to distinct Hermansky-Pudlak syndrome subtypes,73  with relatively mild clinical features for those linked to defects in BLOC-2 components (HPS-3, HPS-5, and HPS-6 subtypes) and more severe forms associated with mutations in BLOC-3 components (HPS-1 and HPS-4 subtypes).90  In Chediak-Higashi syndrome, δ-granule deficiency and hypopigmentation are associated with recurrent infections, severe immunologic defects, and progressive neurologic dysfunction.73,76  Frameshift and nonsense mutations in the LYST gene (coding for LYST, lysosomal-trafficking regulator) underlie the defective exocytosis of secretory lysosomes and lysosome-related organelles.73 

Defects in aggregation

Integrin αIIbβ3 on activated platelets enables the binding of fibrinogen and other soluble adhesive proteins involved in platelet aggregation.

Glanzmann thrombasthenia is caused by biallelic mutations in ITGA2B or ITGB3 genes encoding αIIb (ie, GPIIb) and β3 (ie, GPIIIa), respectively, which are usually unique to each family. Despite their normal platelet count and morphology, Glanzmann thrombasthenia patients have a severe bleeding diathesis due to αIIbβ3 qualitative or quantitative defects that impair platelet binding to fibrinogen during platelet activation.91  Genomic lesions underlying Glanzmann thrombasthenia are mainly represented by nonsense and missense mutations in ITGA2B or ITGB3 genes that impair either the αIIb and β3 subunit biosynthesis or the pro-αIIbβ3 complex formation, maturation, and translocation to the plasma membrane in megakaryocytes (Figure 3).73,84,92  Nurden et al93  recently screened a large cohort (76 families) of Glanzmann thrombasthenia patients by Sanger sequencing analysis on exon and splice sites and identified 55 novel mutations in ITGA2B and ITGB3 genes.

During platelet activation, integrin αIIbβ3 acts as a high-affinity binding site for fibrinogen. Proper αIIbβ3-mediated platelet activation requires interaction between β3 and the cytoplasmic proteins talin and kindlin-3. Indeed, β3 mutations that disrupt binding to talin/kindlin-3 cause platelet aggregation defects. Similarly, mutations affecting the kindlin-3–coding FERMT3 gene underlie leukocyte adhesion deficiency-III syndrome, whose features are a Glanzmann thrombasthenia-like phenotype because of defective β1 and β3 function in platelets, coupled with recurrent infections owing to the impaired β2 function in leukocytes.73,80,84,94,95 

The small GTPase RAP1 is required for full αIIbβ3 activation. RAP1 activity, in turn, is tuned by the guanine nucleotide exchange factor CalDAG-GEFI. Indeed, mutations affecting the CalDAG-GEFI–coding RASGRP2 gene have recently been described as causative for a variant Glanzmann thrombasthenia-like phenotype with normal αIIbβ3 expression but defective platelet aggregation and severe bleeding.84,96 

Defects in procoagulant activity

Phosphatidylserine flipping to the outer plasma membrane is essential for both proper platelet-mediated procoagulant activity and shedding of microparticles from activated, apoptotic, or necrotic cells. In fact, phosphatidylserine exposure upon vascular injury triggers the binding of coagulation factors and assembly of the prothrombinase complex, ultimately leading to platelet-mediated thrombin generation and fibrin clot formation. Platelet microparticles further enhance the hemostatic response through the exposure of phosphatidylserine and coagulation factor binding sites.

Notably, phosphatidylserine externalization is a Ca2+-dependent process regulated by TMEM16F activity. TMEM16F is a Ca2+-activated cation channel that tunes Ca2+ levels, therefore affecting phosphatidylserine scrambling in platelets during coagulation. Indeed, both homozygous nonsense and compound heterozygous in-frame deletion or nonsense mutations in the TMEM16F gene97,98  were identified in Scott syndrome patients. Scott syndrome is an extremely rare bleeding disorder characterized by defects in activation-induced phosphatidylserine externalization on platelets and subsequent impairment of tissue factor–induced thrombin generation and procoagulant microparticle shedding by platelets (Figure 3).76,84  Indeed, platelet-specific TMEM16F-null mice recapitulate all of these major platelet defects99  further emphasizing the crucial role of TMEM16F in platelet function.

Over the past decade, approaches based on next-generation sequencing have prompted the description of the mutational landscape for megakaryopoiesis and platelet function disorders. For some disorders, such as MPNs, genomics has reached the bedside to help patients, but unraveling these disorders is also providing us with a better knowledge of the mechanisms that underpin normal megakaryocyte-to-platelet transition. From this point of view, some of the newly uncovered molecular targets might be exploitable for the development of megakaryocyte precursor–related therapeutic strategies based on gene therapy for inherited platelet disorders as illustrated in the review by Wilcox in this series, but also in the quest to improve the megakaryocyte-to-platelet transition for ex vivo production of functional platelets as described in the review by Sim et al. Indeed, one of the major challenges for therapeutic platelet production is the requirement to produce functional platelets (ie, platelets that fulfill their proper physiological role in hemostasis). The genomic landscape of inherited platelet function defects further emphasizes which signaling pathways and molecular mechanisms are essential for proper platelet function.

With regard to the genomic landscape of malignant megakaryopoiesis, the recent development of methods for genome-wide analysis at a single-cell level will enable us to further investigate how clonal architecture can influence the disease phenotype in MPNs and further dissect the role of every single mutational event in the skewing of megakaryopoiesis and thrombopoiesis. In addition, clinical studies already underway that evaluate how mutations impact the clinical outcome of MPNs should be implemented through the assessment of global accuracy, sensitivity, and specificity of next-generation–based platforms compared with conventional assays.

Despite the effort to characterize the genomic landscape of AMKL, there are still gaps in our understanding. Thus, future studies should be focused on the development of targeted therapies toward recently identified fusion genes or deregulated downstream pathways.

Next-generation sequencing–based techniques have had a significant impact on the characterization of the mutational landscape of platelet function disorders.86,100  The ThromboGenomics Consortium of the International Society on Thrombosis and Haemostasis (https://haemgen.haem.cam.ac.uk/thrombogenomics) developed a platform for diagnosis of rare inherited bleeding and platelet disorders based on next-generation sequencing that has been available for clinical use since July 2015. Major efforts for characterizing platelet function disorders should be directed toward the assessment of tools for genotype-based prediction of bleeding severity to improve patient care.

This work was supported by Associazione Italiana per la Ricerca sul Cancro (AIRC) project No. 10005 “Special Program Molecular Clinical Oncology 5 × 1000” (AIRC-Gruppo Italiano Malattie Mieloproliferative; http://www.progettoagimm.it); AIRC project No. 15337; Italian Ministry of University and Research (Fondo per gli Investimenti della Ricerca di Base Project 2011 [project No. RBAP11CZLK], and Progetti di Rilevante Interesse Nazionale 2010-11 [project No. 2010NYKNS7]).

Contribution: E.B., R.N., V.P., R.Z., and R.M. wrote the paper.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Rossella Manfredini, Centre for Regenerative Medicine Stefano Ferrari, University of Modena and Reggio Emilia, via Gottardi 100, 41125 Modena, Italy; e-mail: rossella.manfredini@unimore.it.

1
Nishimura
 
S
Nagasaki
 
M
Kunishima
 
S
, et al. 
IL-1α induces thrombopoiesis through megakaryocyte rupture in response to acute platelet needs.
J Cell Biol
2015
, vol. 
209
 
3
(pg. 
453
-
466
)
2
Ng
 
AP
Kauppi
 
M
Metcalf
 
D
, et al. 
Mpl expression on megakaryocytes and platelets is dispensable for thrombopoiesis but essential to prevent myeloproliferation.
Proc Natl Acad Sci USA
2014
, vol. 
111
 
16
(pg. 
5884
-
5889
)
3
Stachura
 
DL
Chou
 
ST
Weiss
 
MJ
Early block to erythromegakaryocytic development conferred by loss of transcription factor GATA-1.
Blood
2006
, vol. 
107
 
1
(pg. 
87
-
97
)
4
Nichols
 
KE
Crispino
 
JD
Poncz
 
M
, et al. 
Familial dyserythropoietic anaemia and thrombocytopenia due to an inherited mutation in GATA1.
Nat Genet
2000
, vol. 
24
 
3
(pg. 
266
-
270
)
5
Bianchi
 
E
Zini
 
R
Salati
 
S
, et al. 
c-myb supports erythropoiesis through the transactivation of KLF1 and LMO2 expression.
Blood
2010
, vol. 
116
 
22
(pg. 
e99
-
e110
)
6
Starck
 
J
Cohet
 
N
Gonnet
 
C
, et al. 
Functional cross-antagonism between transcription factors FLI-1 and EKLF.
Mol Cell Biol
2003
, vol. 
23
 
4
(pg. 
1390
-
1402
)
7
Siatecka
 
M
Xue
 
L
Bieker
 
JJ
Sumoylation of EKLF promotes transcriptional repression and is involved in inhibition of megakaryopoiesis.
Mol Cell Biol
2007
, vol. 
27
 
24
(pg. 
8547
-
8560
)
8
Bianchi
 
E
Bulgarelli
 
J
Ruberti
 
S
, et al. 
MYB controls erythroid versus megakaryocyte lineage fate decision through the miR-486-3p-mediated downregulation of MAF.
Cell Death Differ
2015
, vol. 
22
 
12
(pg. 
1906
-
1921
)
9
Carpinelli
 
MR
Hilton
 
DJ
Metcalf
 
D
, et al. 
Suppressor screen in Mpl-/- mice: c-Myb mutation causes supraphysiological production of platelets in the absence of thrombopoietin signaling.
Proc Natl Acad Sci USA
2004
, vol. 
101
 
17
(pg. 
6553
-
6558
)
10
Barroga
 
CF
Pham
 
H
Kaushansky
 
K
Thrombopoietin regulates c-Myb expression by modulating micro RNA 150 expression.
Exp Hematol
2008
, vol. 
36
 
12
(pg. 
1585
-
1592
)
11
Lu
 
J
Guo
 
S
Ebert
 
BL
, et al. 
MicroRNA-mediated control of cell fate in megakaryocyte-erythrocyte progenitors.
Dev Cell
2008
, vol. 
14
 
6
(pg. 
843
-
853
)
12
Wang
 
X
Crispino
 
JD
Letting
 
DL
Nakazawa
 
M
Poncz
 
M
Blobel
 
GA
Control of megakaryocyte-specific gene expression by GATA-1 and FOG-1: role of Ets transcription factors.
EMBO J
2002
, vol. 
21
 
19
(pg. 
5225
-
5234
)
13
Jackers
 
P
Szalai
 
G
Moussa
 
O
Watson
 
DK
Ets-dependent regulation of target gene expression during megakaryopoiesis.
J Biol Chem
2004
, vol. 
279
 
50
(pg. 
52183
-
52190
)
14
Bluteau
 
D
Glembotsky
 
AC
Raimbault
 
A
, et al. 
Dysmegakaryopoiesis of FPD/AML pedigrees with constitutional RUNX1 mutations is linked to myosin II deregulated expression.
Blood
2012
, vol. 
120
 
13
(pg. 
2708
-
2718
)
15
Lordier
 
L
Bluteau
 
D
Jalil
 
A
, et al. 
RUNX1-induced silencing of non-muscle myosin heavy chain IIB contributes to megakaryocyte polyploidization.
Nat Commun
2012
, vol. 
3
 pg. 
717
 
16
Muntean
 
AG
Pang
 
L
Poncz
 
M
Dowdy
 
SF
Blobel
 
GA
Crispino
 
JD
Cyclin D-Cdk4 is regulated by GATA-1 and required for megakaryocyte growth and polyploidization.
Blood
2007
, vol. 
109
 
12
(pg. 
5199
-
5207
)
17
Takayama
 
M
Fujita
 
R
Suzuki
 
M
, et al. 
Genetic analysis of hierarchical regulation for Gata1 and NF-E2 p45 gene expression in megakaryopoiesis.
Mol Cell Biol
2010
, vol. 
30
 
11
(pg. 
2668
-
2680
)
18
Lecine
 
P
Italiano
 
JE
Kim
 
SW
Villeval
 
JL
Shivdasani
 
RA
Hematopoietic-specific beta 1 tubulin participates in a pathway of platelet biogenesis dependent on the transcription factor NF-E2.
Blood
2000
, vol. 
96
 
4
(pg. 
1366
-
1373
)
19
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
 
5
(pg. 
695
-
704
)
20
Tefferi
 
A
Vardiman
 
JW
Classification and diagnosis of myeloproliferative neoplasms: the 2008 World Health Organization criteria and point-of-care diagnostic algorithms.
Leukemia
2008
, vol. 
22
 
1
(pg. 
14
-
22
)
21
Baxter
 
EJ
Scott
 
LM
Campbell
 
PJ
, et al. 
Cancer Genome Project
Acquired mutation of the tyrosine kinase JAK2 in human myeloproliferative disorders.
Lancet
2005
, vol. 
365
 
9464
(pg. 
1054
-
1061
)
22
James
 
C
Ugo
 
V
Le Couédic
 
JP
, et al. 
A unique clonal JAK2 mutation leading to constitutive signalling causes polycythaemia vera.
Nature
2005
, vol. 
434
 
7037
(pg. 
1144
-
1148
)
23
Kralovics
 
R
Passamonti
 
F
Buser
 
AS
, et al. 
A gain-of-function mutation of JAK2 in myeloproliferative disorders.
N Engl J Med
2005
, vol. 
352
 
17
(pg. 
1779
-
1790
)
24
Levine
 
RL
Wadleigh
 
M
Cools
 
J
, et al. 
Activating mutation in the tyrosine kinase JAK2 in polycythemia vera, essential thrombocythemia, and myeloid metaplasia with myelofibrosis.
Cancer Cell
2005
, vol. 
7
 
4
(pg. 
387
-
397
)
25
Milosevic
 
JD
Kralovics
 
R
Genetic and epigenetic alterations of myeloproliferative disorders.
Int J Hematol
2013
, vol. 
97
 
2
(pg. 
183
-
197
)
26
Li
 
J
Kent
 
DG
Godfrey
 
AL
, et al. 
JAK2V617F homozygosity drives a phenotypic switch in myeloproliferative neoplasms, but is insufficient to sustain disease.
Blood
2014
, vol. 
123
 
20
(pg. 
3139
-
3151
)
27
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
 
7
pg. 
e270
 
28
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
 
10
(pg. 
3472
-
3476
)
29
Beer
 
PA
Campbell
 
PJ
Scott
 
LM
, et al. 
MPL mutations in myeloproliferative disorders: analysis of the PT-1 cohort.
Blood
2008
, vol. 
112
 
1
(pg. 
141
-
149
)
30
Schnittger
 
S
Bacher
 
U
Haferlach
 
C
, et al. 
Characterization of 35 new cases with four different MPLW515 mutations and essential thrombocytosis or primary myelofibrosis.
Haematologica
2009
, vol. 
94
 
1
(pg. 
141
-
144
)
31
Harrison
 
CN
Gale
 
RE
Wiestner
 
AC
Skoda
 
RC
Linch
 
DC
The activating splice mutation in intron 3 of the thrombopoietin gene is not found in patients with non-familial essential thrombocythaemia.
Br J Haematol
1998
, vol. 
102
 
5
(pg. 
1341
-
1343
)
32
Klampfl
 
T
Gisslinger
 
H
Harutyunyan
 
AS
, et al. 
Somatic mutations of calreticulin in myeloproliferative neoplasms.
N Engl J Med
2013
, vol. 
369
 
25
(pg. 
2379
-
2390
)
33
Nangalia
 
J
Massie
 
CE
Baxter
 
EJ
, et al. 
Somatic CALR mutations in myeloproliferative neoplasms with nonmutated JAK2.
N Engl J Med
2013
, vol. 
369
 
25
(pg. 
2391
-
2405
)
34
Marty
 
C
Pecquet
 
C
Nivarthi
 
H
, et al. 
Calreticulin mutants in mice induce an MPL-dependent thrombocytosis with frequent progression to myelofibrosis [published online ahead of print November 25, 2015].
Blood
 
doi:10.1182/blood-2015-11-679571
35
Chachoua
 
I
Pecquet
 
C
El-Khoury
 
M
, et al. 
Thrombopoietin receptor activation by myeloproliferative neoplasm associated calreticulin mutants [published online ahead of print December 14, 2015].
Blood
 
doi:10.1182/blood-2015-11-681932
36
Rampal
 
R
Al-Shahrour
 
F
Abdel-Wahab
 
O
, et al. 
Integrated genomic analysis illustrates the central role of JAK-STAT pathway activation in myeloproliferative neoplasm pathogenesis.
Blood
2014
, vol. 
123
 
22
(pg. 
e123
-
e133
)
37
Rampal
 
R
Levine
 
RL
A primer on genomic and epigenomic alterations in the myeloproliferative neoplasms.
Best Pract Res Clin Haematol
2014
, vol. 
27
 
2
(pg. 
83
-
93
)
38
Tenedini
 
E
Bernardis
 
I
Artusi
 
V
, et al. 
AGIMM investigators
Targeted cancer exome sequencing reveals recurrent mutations in myeloproliferative neoplasms.
Leukemia
2014
, vol. 
28
 
5
(pg. 
1052
-
1059
)
39
Tapper
 
W
Jones
 
AV
Kralovics
 
R
, et al. 
Genetic variation at MECOM, TERT, JAK2 and HBS1L-MYB predisposes to myeloproliferative neoplasms.
Nat Commun
2015
, vol. 
6
 pg. 
6691
 
40
Guglielmelli
 
P
Zini
 
R
Bogani
 
C
, et al. 
Molecular profiling of CD34+ cells in idiopathic myelofibrosis identifies a set of disease-associated genes and reveals the clinical significance of Wilms’ tumor gene 1 (WT1).
Stem Cells
2007
, vol. 
25
 
1
(pg. 
165
-
173
)
41
Catani
 
L
Zini
 
R
Sollazzo
 
D
, et al. 
Molecular profile of CD34+ stem/progenitor cells according to JAK2V617F mutation status in essential thrombocythemia.
Leukemia
2009
, vol. 
23
 
5
(pg. 
997
-
1000
)
42
Lin
 
X
Rice
 
KL
Buzzai
 
M
, et al. 
miR-433 is aberrantly expressed in myeloproliferative neoplasms and suppresses hematopoietic cell growth and differentiation.
Leukemia
2013
, vol. 
27
 
2
(pg. 
344
-
352
)
43
Zhan
 
H
Cardozo
 
C
Yu
 
W
, et al. 
MicroRNA deregulation in polycythemia vera and essential thrombocythemia patients.
Blood Cells Mol Dis
2013
, vol. 
50
 
3
(pg. 
190
-
195
)
44
Pennucci
 
V
Zini
 
R
Norfo
 
R
, et al. 
Associazione Italiana per la Ricerca sul Cancro Gruppo Italiano Malattie Mieloproliferative (AGIMM) Investigators
Abnormal expression patterns of WT1-as, MEG3 and ANRIL long non-coding RNAs in CD34+ cells from patients with primary myelofibrosis and their clinical correlations.
Leuk Lymphoma
2015
, vol. 
56
 
2
(pg. 
492
-
496
)
45
Norfo
 
R
Zini
 
R
Pennucci
 
V
, et al. 
Associazione Italiana per la Ricerca sul Cancro Gruppo Italiano Malattie Mieloproliferative Investigators
miRNA-mRNA integrative analysis in primary myelofibrosis CD34+ cells: role of miR-155/JARID2 axis in abnormal megakaryopoiesis.
Blood
2014
, vol. 
124
 
13
(pg. 
e21
-
e32
)
46
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
 
12
(pg. 
3913
-
3915
)
47
Girardot
 
M
Pecquet
 
C
Boukour
 
S
, et al. 
miR-28 is a thrombopoietin receptor targeting microRNA detected in a fraction of myeloproliferative neoplasm patient platelets.
Blood
2010
, vol. 
116
 
3
(pg. 
437
-
445
)
48
Hong
 
WJ
Gotlib
 
J
Hereditary erythrocytosis, thrombocytosis and neutrophilia.
Best Pract Res Clin Haematol
2014
, vol. 
27
 
2
(pg. 
95
-
106
)
49
Pellagatti
 
A
Boultwood
 
J
The molecular pathogenesis of the myelodysplastic syndromes.
Eur J Haematol
2015
, vol. 
95
 
1
(pg. 
3
-
15
)
50
Schneider
 
RK
Ademà
 
V
Heckl
 
D
, et al. 
Role of casein kinase 1A1 in the biology and targeted therapy of del(5q) MDS.
Cancer Cell
2014
, vol. 
26
 
4
(pg. 
509
-
520
)
51
Starczynowski
 
DT
Kuchenbauer
 
F
Argiropoulos
 
B
, et al. 
Identification of miR-145 and miR-146a as mediators of the 5q- syndrome phenotype.
Nat Med
2010
, vol. 
16
 
1
(pg. 
49
-
58
)
52
Malcovati
 
L
Cazzola
 
M
Refractory anemia with ring sideroblasts.
Best Pract Res Clin Haematol
2013
, vol. 
26
 
4
(pg. 
377
-
385
)
53
Szpurka
 
H
Tiu
 
R
Murugesan
 
G
, et al. 
Refractory anemia with ringed sideroblasts associated with marked thrombocytosis (RARS-T), another myeloproliferative condition characterized by JAK2 V617F mutation.
Blood
2006
, vol. 
108
 
7
(pg. 
2173
-
2181
)
54
Malcovati
 
L
Papaemmanuil
 
E
Bowen
 
DT
, et al. 
Chronic Myeloid Disorders Working Group of the International Cancer Genome Consortium and of the Associazione Italiana per la Ricerca sul Cancro Gruppo Italiano Malattie Mieloproliferative
Clinical significance of SF3B1 mutations in myelodysplastic syndromes and myelodysplastic/myeloproliferative neoplasms.
Blood
2011
, vol. 
118
 
24
(pg. 
6239
-
6246
)
55
Visconte
 
V
Rogers
 
HJ
Singh
 
J
, et al. 
SF3B1 haploinsufficiency leads to formation of ring sideroblasts in myelodysplastic syndromes.
Blood
2012
, vol. 
120
 
16
(pg. 
3173
-
3186
)
56
Broséus
 
J
Lippert
 
E
Harutyunyan
 
AS
, et al. 
Low rate of calreticulin mutations in refractory anaemia with ring sideroblasts and marked thrombocytosis.
Leukemia
2014
, vol. 
28
 
6
(pg. 
1374
-
1376
)
57
Malcovati
 
L
Papaemmanuil
 
E
Ambaglio
 
I
, et al. 
Driver somatic mutations identify distinct disease entities within myeloid neoplasms with myelodysplasia.
Blood
2014
, vol. 
124
 
9
(pg. 
1513
-
1521
)
58
Dastugue
 
N
Lafage-Pochitaloff
 
M
Pagès
 
MP
, et al. 
Groupe Français d’Hematologie Cellulaire
Cytogenetic profile of childhood and adult megakaryoblastic leukemia (M7): a study of the Groupe Français de Cytogénétique Hématologique (GFCH).
Blood
2002
, vol. 
100
 
2
(pg. 
618
-
626
)
59
Hama
 
A
Muramatsu
 
H
Makishima
 
H
, et al. 
Molecular lesions in childhood and adult acute megakaryoblastic leukaemia.
Br J Haematol
2012
, vol. 
156
 
3
(pg. 
316
-
325
)
60
Malinge
 
S
Bliss-Moreau
 
M
Kirsammer
 
G
, et al. 
Increased dosage of the chromosome 21 ortholog Dyrk1a promotes megakaryoblastic leukemia in a murine model of Down syndrome.
J Clin Invest
2012
, vol. 
122
 
3
(pg. 
948
-
962
)
61
Carmichael
 
CL
Metcalf
 
D
Henley
 
KJ
, et al. 
Hematopoietic overexpression of the transcription factor Erg induces lymphoid and erythro-megakaryocytic leukemia.
Proc Natl Acad Sci USA
2012
, vol. 
109
 
38
(pg. 
15437
-
15442
)
62
Klusmann
 
JH
Li
 
Z
Böhmer
 
K
, et al. 
miR-125b-2 is a potential oncomiR on human chromosome 21 in megakaryoblastic leukemia.
Genes Dev
2010
, vol. 
24
 
5
(pg. 
478
-
490
)
63
Ng
 
AP
Hu
 
Y
Metcalf
 
D
, et al. 
Early lineage priming by trisomy of erg leads to myeloproliferation in a down syndrome model.
PLoS Genet
2015
, vol. 
11
 
5
pg. 
e1005211
 
64
Wechsler
 
J
Greene
 
M
McDevitt
 
MA
, et al. 
Acquired mutations in GATA1 in the megakaryoblastic leukemia of Down syndrome.
Nat Genet
2002
, vol. 
32
 
1
(pg. 
148
-
152
)
65
Li
 
Z
Godinho
 
FJ
Klusmann
 
JH
Garriga-Canut
 
M
Yu
 
C
Orkin
 
SH
Developmental stage-selective effect of somatically mutated leukemogenic transcription factor GATA1.
Nat Genet
2005
, vol. 
37
 
6
(pg. 
613
-
619
)
66
Alford
 
KA
Slender
 
A
Vanes
 
L
, et al. 
Perturbed hematopoiesis in the Tc1 mouse model of Down syndrome.
Blood
2010
, vol. 
115
 
14
(pg. 
2928
-
2937
)
67
Lion
 
T
Haas
 
OA
Harbott
 
J
, et al. 
The translocation t(1;22)(p13;q13) is a nonrandom marker specifically associated with acute megakaryocytic leukemia in young children.
Blood
1992
, vol. 
79
 
12
(pg. 
3325
-
3330
)
68
Mercher
 
T
Busson-Le Coniat
 
M
Nguyen Khac
 
F
, et al. 
Recurrence of OTT-MAL fusion in t(1;22) of infant AML-M7.
Genes Chromosomes Cancer
2002
, vol. 
33
 
1
(pg. 
22
-
28
)
69
Mercher
 
T
Raffel
 
GD
Moore
 
SA
, et al. 
The OTT-MAL fusion oncogene activates RBPJ-mediated transcription and induces acute megakaryoblastic leukemia in a knockin mouse model.
J Clin Invest
2009
, vol. 
119
 
4
(pg. 
852
-
864
)
70
Mercher
 
T
Cornejo
 
MG
Sears
 
C
, et al. 
Notch signaling specifies megakaryocyte development from hematopoietic stem cells.
Cell Stem Cell
2008
, vol. 
3
 
3
(pg. 
314
-
326
)
71
Poirault-Chassac
 
S
Six
 
E
Catelain
 
C
, et al. 
Notch/Delta4 signaling inhibits human megakaryocytic terminal differentiation.
Blood
2010
, vol. 
116
 
25
(pg. 
5670
-
5678
)
72
Delaney
 
C
Heimfeld
 
S
Brashem-Stein
 
C
Voorhies
 
H
Manger
 
RL
Bernstein
 
ID
Notch-mediated expansion of human cord blood progenitor cells capable of rapid myeloid reconstitution.
Nat Med
2010
, vol. 
16
 
2
(pg. 
232
-
236
)
73
Nurden
 
AT
Nurden
 
P
Congenital platelet disorders and understanding of platelet function.
Br J Haematol
2014
, vol. 
165
 
2
(pg. 
165
-
178
)
74
Savoia
 
A
Pastore
 
A
De Rocco
 
D
, et al. 
Clinical and genetic aspects of Bernard-Soulier syndrome: searching for genotype/phenotype correlations.
Haematologica
2011
, vol. 
96
 
3
(pg. 
417
-
423
)
75
Savoia
 
A
Kunishima
 
S
De Rocco
 
D
, et al. 
Spectrum of the mutations in Bernard-Soulier syndrome.
Hum Mutat
2014
, vol. 
35
 
9
(pg. 
1033
-
1045
)
76
Nurden
 
AT
Freson
 
K
Seligsohn
 
U
 
Inherited platelet disorders. Haemophilia. 2012;18(Suppl 4):154-160
77
Pecci
 
A
Balduini
 
CL
Lessons in platelet production from inherited thrombocytopenias.
Br J Haematol
2014
, vol. 
165
 
2
(pg. 
179
-
192
)
78
Balduini
 
A
Malara
 
A
Pecci
 
A
, et al. 
Proplatelet formation in heterozygous Bernard-Soulier syndrome type Bolzano.
J Thromb Haemost
2009
, vol. 
7
 
3
(pg. 
478
-
484
)
79
Strassel
 
C
Eckly
 
A
Léon
 
C
, et al. 
Intrinsic impaired proplatelet formation and microtubule coil assembly of megakaryocytes in a mouse model of Bernard-Soulier syndrome.
Haematologica
2009
, vol. 
94
 
6
(pg. 
800
-
810
)
80
Nurden
 
A
Nurden
 
P
Advances in our understanding of the molecular basis of disorders of platelet function.
J Thromb Haemost
2011
, vol. 
9
 
Suppl 1
(pg. 
76
-
91
)
81
Woods
 
AI
Sanchez-Luceros
 
A
Bermejo
 
E
, et al. 
Identification of p.W246L as a novel mutation in the GP1BA gene responsible for platelet-type von Willebrand disease.
Semin Thromb Hemost
2014
, vol. 
40
 
2
(pg. 
151
-
160
)
82
Nisar
 
SP
Jones
 
ML
Cunningham
 
MR
Mumford
 
AD
Mundell
 
SJ
UK GAPP Study Group
Rare platelet GPCR variants: what can we learn?
Br J Pharmacol
2015
, vol. 
172
 
13
(pg. 
3242
-
3253
)
83
Lecchi
 
A
Razzari
 
C
Paoletta
 
S
, et al. 
Identification of a new dysfunctional platelet P2Y12 receptor variant associated with bleeding diathesis.
Blood
2015
, vol. 
125
 
6
(pg. 
1006
-
1013
)
84
Nurden
 
AT
Nurden
 
P
Inherited disorders of platelet function: selected updates.
J Thromb Haemost
2015
, vol. 
13
 
Suppl 1
(pg. 
S2
-
S9
)
85
Albers
 
CA
Cvejic
 
A
Favier
 
R
, et al. 
Exome sequencing identifies NBEAL2 as the causative gene for gray platelet syndrome.
Nat Genet
2011
, vol. 
43
 
8
(pg. 
735
-
737
)
86
Gunay-Aygun
 
M
Falik-Zaccai
 
TC
Vilboux
 
T
, et al. 
NBEAL2 is mutated in gray platelet syndrome and is required for biogenesis of platelet α-granules.
Nat Genet
2011
, vol. 
43
 
8
(pg. 
732
-
734
)
87
Kahr
 
WH
Hinckley
 
J
Li
 
L
, et al. 
Mutations in NBEAL2, encoding a BEACH protein, cause gray platelet syndrome.
Nat Genet
2011
, vol. 
43
 
8
(pg. 
738
-
740
)
88
Guerrero
 
JA
Bennett
 
C
van der Weyden
 
L
, et al. 
Gray platelet syndrome: proinflammatory megakaryocytes and α-granule loss cause myelofibrosis and confer metastasis resistance in mice.
Blood
2014
, vol. 
124
 
24
(pg. 
3624
-
3635
)
89
Monteferrario
 
D
Bolar
 
NA
Marneth
 
AE
, et al. 
A dominant-negative GFI1B mutation in the gray platelet syndrome.
N Engl J Med
2014
, vol. 
370
 
3
(pg. 
245
-
253
)
90
Huizing
 
M
Helip-Wooley
 
A
Westbroek
 
W
Gunay-Aygun
 
M
Gahl
 
WA
Disorders of lysosome-related organelle biogenesis: clinical and molecular genetics.
Annu Rev Genomics Hum Genet
2008
, vol. 
9
 (pg. 
359
-
386
)
91
Nurden
 
AT
Pillois
 
X
Wilcox
 
DA
Glanzmann thrombasthenia: state of the art and future directions.
Semin Thromb Hemost
2013
, vol. 
39
 
6
(pg. 
642
-
655
)
92
Nurden
 
AT
Fiore
 
M
Nurden
 
P
Pillois
 
X
Glanzmann thrombasthenia: a review of ITGA2B and ITGB3 defects with emphasis on variants, phenotypic variability, and mouse models.
Blood
2011
, vol. 
118
 
23
(pg. 
5996
-
6005
)
93
Nurden
 
AT
Pillois
 
X
Fiore
 
M
, et al. 
Expanding the Mutation Spectrum Affecting αIIbβ3 Integrin in Glanzmann Thrombasthenia: Screening of the ITGA2B and ITGB3 Genes in a Large International Cohort.
Hum Mutat
2015
, vol. 
36
 
5
(pg. 
548
-
561
)
94
Svensson
 
L
Howarth
 
K
McDowall
 
A
, et al. 
Leukocyte adhesion deficiency-III is caused by mutations in KINDLIN3 affecting integrin activation.
Nat Med
2009
, vol. 
15
 
3
(pg. 
306
-
312
)
95
Malinin
 
NL
Zhang
 
L
Choi
 
J
, et al. 
A point mutation in KINDLIN3 ablates activation of three integrin subfamilies in humans.
Nat Med
2009
, vol. 
15
 
3
(pg. 
313
-
318
)
96
Canault
 
M
Ghalloussi
 
D
Grosdidier
 
C
, et al. 
Human CalDAG-GEFI gene (RASGRP2) mutation affects platelet function and causes severe bleeding.
J Exp Med
2014
, vol. 
211
 
7
(pg. 
1349
-
1362
)
97
Castoldi
 
E
Collins
 
PW
Williamson
 
PL
Bevers
 
EM
Compound heterozygosity for 2 novel TMEM16F mutations in a patient with Scott syndrome.
Blood
2011
, vol. 
117
 
16
(pg. 
4399
-
4400
)
98
Suzuki
 
J
Umeda
 
M
Sims
 
PJ
Nagata
 
S
Calcium-dependent phospholipid scrambling by TMEM16F.
Nature
2010
, vol. 
468
 
7325
(pg. 
834
-
838
)
99
Fujii
 
T
Sakata
 
A
Nishimura
 
S
Eto
 
K
Nagata
 
S
TMEM16F is required for phosphatidylserine exposure and microparticle release in activated mouse platelets.
Proc Natl Acad Sci USA
2015
, vol. 
112
 
41
(pg. 
12800
-
12805
)
100
Leo
 
VC
Morgan
 
NV
Bem
 
D
, et al. 
UK GAPP Study Group
Use of next-generation sequencing and candidate gene analysis to identify underlying defects in patients with inherited platelet function disorders.
J Thromb Haemost
2015
, vol. 
13
 
4
(pg. 
643
-
650
)

Author notes

*

E.B. and R.N. contributed equally to this work.

Sign in via your Institution