ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Megakaryocyte biology and platelet production

Megakaryocyte biology and platelet production
Literature review current through: Jan 2024.
This topic last updated: Jan 25, 2024.

INTRODUCTION — This topic reviews the biology of megakaryocytes and the mechanisms of platelet production in the bone marrow.

Separate topics discuss the biology of hematopoietic stem cells (HSCs), platelets, thrombopoietin (TPO), and clinical implications:

Basic science

HSCs – (See "Overview of hematopoietic stem cells".)

Platelet biology – (See "Platelet biology and mechanism of anti-platelet drugs".)

TPO biology – (See "Biology and physiology of thrombopoietin".)

Clinical

Inherited platelet function disorders – (See "Inherited platelet function disorders (IPFDs)".)

Platelet function tests – (See "Platelet function testing".)

Transfusion – (See "Platelet transfusion: Indications, ordering, and associated risks".)

TPO receptor agonists (TPO-RAs) – (See "Clinical applications of thrombopoietic growth factors".)

HISTORICAL BACKGROUND — The megakaryocyte was discovered to be the hematopoietic cell that produces platelets over 100 years ago.

Evidence for this relationship was first provided in 1906 by James Homer Wright, who demonstrated that circulating platelets and a giant bone marrow cell now known to be the megakaryocyte shared common tinctorial properties when subjected to a modified Romanowsky stain (picture 1) [1].

Wright went on to show that megakaryocytes sent out pseudopodia into the bone marrow sinusoids from which platelets appeared to be shed [2]. This model of how megakaryocytes produce platelets remains to this day.

Wright also demonstrated that changes in platelet number were associated with changes only in megakaryocytes [2].

Since these seminal observations, much has become known about megakaryocytes and how they produce platelets [3,4].

CHARACTERISTICS

Size and ploidy

Size – Megakaryocytes are the largest cell in the bone marrow (picture 2). They have an average diameter of 20 to 25 microns and a volume of 4700 ± 100 fL [5]. By comparison, red blood cells (RBCs) have a diameter of 7 to 8 microns and a volume of 85 to 100 fL [5]. Large megakaryocytes have diameters of 50 to 60 microns and volumes of 65,000 to 100,000 fL.

Ploidy – Mature megakaryocytes are polyploid, contain two (4N) to 32 (64N) times the normal diploid DNA content; the mean value is 16N in humans [6,7]. The DNA is contained within one highly lobulated nuclear envelope, in which each lobule represents one diploid amount of DNA (2N). This is the result of a process referred to as endomitosis [8]. (See "Basic genetics concepts: Chromosomes and cell division", section on 'Mitosis'.)

Few other cells are normally polyploid. Few other cells are normally polyploid. Placental trophoblasts invading the endometrium and myometrium may undergo polyploidization to create cells of 4 to 64N; this is felt to prevent their replication [9,10]. Occasional hepatocytes and macrophages have 4N or 8N DNA content contained in multiple separate nuclei.

Relationship between size and ploidy – Megakaryocyte polyploidy appears to result in functional gene amplification, perhaps to increase protein synthesis in parallel with megakaryocyte enlargement [11,12].

In general, increased ploidy is associated with increased megakaryocyte size. However, given the time needed for megakaryocyte cytoplasm to mature, not all small megakaryocytes are of low ploidy.

Membrane — Megakaryocyte plasma membrane is similar to that of the platelet.

One of the first signs of differentiation along the megakaryocyte lineage is the appearance of membrane glycoprotein (GP) receptors including integrin alphaIIbbeta3 (previously called GPIIbIIIa), which becomes active on activated platelets and binds to fibrinogen, and later by GPIb and other collagen receptors [13,14]. (See "Platelet biology and mechanism of anti-platelet drugs", section on 'Collagen receptors (GPIa/IIa and GPVI)' and "Platelet biology and mechanism of anti-platelet drugs", section on 'Integrin alphaIIbbeta3 (GPIIb/IIIa) binding to fibrinogen, activation, and platelet aggregation'.)

The surface membrane of the mature megakaryocyte is deeply invaginated and highly redundant, creating a demarcation membrane system that divides the cytoplasm into platelet-sized subunits (picture 3). This enormous amplification of the surface membrane is thought to provide sufficient membrane to form platelets [15-18].

Granules — Since platelets undergo little protein synthesis, their cytoplasmic characteristics are mostly determined by the megakaryocytes from which they arise.

The membrane bodies of the alpha and dense platelet granules and some of their contents are made in megakaryocytes. However, some of the granule contents are taken up from the plasma by both megakaryocytes and platelets.

Alpha granules – The alpha granule body is made early in megakaryocyte development before the demarcation membrane system. Alpha granules contain numerous platelet proteins and growth factors.

Megakaryocytes synthesize these proteins and transport them to the alpha granules [19]:

-Platelet-derived growth factor (PDGF)

-Transforming growth factor-beta (TGF-beta)

-Platelet factor-4 (PF4)

-von Willebrand factor

Megakaryocytes and platelets take up these proteins from the circulation:

-Fibrinogen undergoes receptor-mediated endocytosis by the integrin alphaIIbbeta3 receptor [20-22].

-Albumin and immunoglobulin G (IgG) undergo pinocytosis [23].

Dense granules – Dense granules are physically dense and, because they contain serotonin and calcium, they are also electron dense in transmission electron microscopy.

The dense granule membrane bodies are made in megakaryocytes, but do not take up serotonin and calcium until platelets are released into the circulation [24,25]. Most of the body's circulating serotonin is taken up by platelets.

A possible connection between selective serotonin reuptake inhibitors and bleeding is discussed separately. (See "Selective serotonin reuptake inhibitors: Pharmacology, administration, and side effects", section on 'Bleeding'.)

Platelet granule disorders are also discussed separately. (See "Inherited platelet function disorders (IPFDs)", section on 'Mechanisms and gene variants'.)

Microscopic appearance — Megakaryocytes normally account for approximately 0.05 to 0.1 percent of nucleated bone marrow cells. Their number increases as the demand for platelets rises.

Three stages of megakaryocytes may be identified [26]:

Mature megakaryocytes are large and polyploid. These cells are readily identified by light microscopy (picture 1 and picture 2). Platelet shedding can occasionally be seen.

Intermediate megakaryocytes have large polyploid nuclei with a modest amount of cytoplasm; they can be distinguished from immature megakaryocytes, which have scant cytoplasm and large basophilic nuclei.

Also present, but identified only by special stains for von Willebrand factor or GP IIb/IIIa, are small, diploid, lymphocyte-like megakaryocyte progenitor cells that have not yet undergone endomitosis [27].

ORIGIN AND DIFFERENTIATION

Stem and progenitor cells

HSCs – Megakaryocytes are derived from multipotent hematopoietic stem cells (HSCs) (figure 1). (See "Overview of hematopoietic stem cells", section on 'Hematopoietic stem cells'.)

MEPs – HSCs give rise to early bilineage progenitor cells, referred to as megakaryocyte-erythroid progenitors (MEPs), that can subsequently differentiate along the erythroid or megakaryocyte lineage.

A specific assay for MEPs is lacking, but their existence is supported by several findings:

Shared genetic regulation Expression of thymidine kinase in specific cell types allows selective killing upon administration of ganciclovir, which is converted to a toxic metabolite. In transgenic mice expressing the gene for thymidine kinase under the control of a platelet-specific promoter (ITGA2B, which encodes platelet glycoprotein [GP] IIb), administration of ganciclovir eradicated production of both platelets and erythrocytes [28]. This observation suggests that the GPIIb promoter was transcriptionally active in the MEP and possibly in a more potent progenitor cell [29,30].

Shared maturation – Early erythroid and megakaryocyte differentiation share molecular, cellular, and pathological features.

-The figure illustrates shared transcription factors (figure 2) [31].

-Regulatory hematopoietic cytokines for megakaryocytes (thrombopoietin [TPO]) and erythrocytes (erythropoietin) share 50 percent sequence similarity [32]. (See 'TPO role in maintaining platelet count' below and "Regulation of erythropoiesis", section on 'Erythropoietin'.)

Lineage specification – The microRNA miR-150 determines lineage specification for this common MEP, driving differentiation towards megakaryocytes and away from erythroid cells [33]. The transcription factor Myb is a critical target of miR-150 [34].

TPO increases miR-150, which in turn binds to the four miR-150 binding sites in the MYB 3' untranslated region and decreases Myb mRNA and protein [35]. Reduction in Myb expression subsequently leads to increased megakaryocyte differentiation and reduced erythroid differentiation.

Absence of miR-150 promotes erythroid differentiation.

Meg-CFC – Differentiation ultimately produces a committed precursor cell only able to produce megakaryocytes, called the megakaryocyte colony-forming cell (Meg-CFC).

Meg-CFCs have the following properties:

Can be assayed by cell culture methods

Express integrin alphaIIbbeta3 (previously called GPIIbIIIa), the receptor for fibrinogen [36,37]

Can undergo mitosis [38]

Are stimulated by interleukin (IL)-3 and TPO [39,40].

Immature megakaryocytes

Endomitotic growth – Eventually Meg-CFCs stop mitosis and enter endomitosis, in which DNA replication and chromosome segregation occur without nuclear division or cytokinesis, producing large polyploid cells (figure 1). (See 'Size and ploidy' above.)

It was initially assumed that endomitosis was simply the absence of mitosis after each round of DNA replication. However, studies in mice showed that the cells enter mitosis and progress through normal prophase, prometaphase, metaphase, and up to anaphase A, but not to anaphase B, telophase, or cytokinesis [41]. After anaphase, the nuclear membrane is reassembled around the sister chromatids as a single nucleus; the cells then enter the next round of DNA replication. (See "Basic genetics concepts: Chromosomes and cell division", section on 'Somatic cell division'.)

Endomitosis is associated with increased cyclin D3 and reduced cyclin B1 and cyclin B1-dependent Cdc2 kinase [42,43]. These changes may facilitate aborted mitosis and reentry into S phase without cytokinesis [44].

Cytoplasmic maturation – On completion of several rounds of endomitosis, immature megakaryocytes develop a mature cytoplasm, become morphologically identifiable megakaryocytes, and eventually release platelets.

It takes approximately five to seven days to produce platelets from Meg-CFCs.

Factors promoting differentiation — The mechanisms by which MEPs become committed to the megakaryocyte lineage and Meg-CFCs mature to become megakaryocytes are beginning to be unraveled.

GATA1 and NF-E2 transcription factors — GATA1 and NF-E2 (nuclear factor erythroid derived 2) are intrinsic lineage-specific transcription factors that help establish cell-specific phenotypes. They interact with a large number of other factors including JAK and STAT.

GATA1 – GATA1 is zinc-finger transcription factor expressed in many hematopoietic cell types, including progenitor cells and precursor cells of erythroid, megakaryocyte, eosinophil, and mast cell lineages [45]. GATA transcription factors bind to the DNA sequence G-A-T-A. (See "Regulation of erythropoiesis", section on 'GATA1'.)

GATA1 gene knockout in mice is embryonically lethal due to severe anemia [45]. In a mouse model in which a portion of the GATA1 promoter was disrupted to selectively eliminate expression in megakaryocytes but not erythroid progenitor cells, the platelet count was reduced to 15 percent of normal and small, abnormal megakaryocytes were seen, with multilobulated nuclei, scant cytoplasm, few demarcation membranes, no platelet "territories," and few platelet granules [31,46,47].

Numerous genes are downregulated by the loss of GATA1 in mice, including JAK2, which mediates TPO signaling, and STAT1, which mediates interferon-gamma signaling. GATA-1 may promote megakaryopoiesis in part via activation of interferon-gamma/STAT1 signaling [48]. This may explain the mechanism by which inflammatory disorders can increase platelet counts. (See 'Cancer and inflammation' below.)

NF-E2 – NF-E2 is a heterodimeric basic leucine zipper transcription factor composed of a p18 subunit that is widely expressed and a p45 subunit that is only expressed in erythroid, megakaryocyte, and mast cells.

NEF2 gene knockout in mice is embryonically lethal due to profound thrombocytopenia associated with a high rate of early hemorrhagic death [49,50]. The NFE2 gene encodes the p45 subunit.

NEF2 null mice have adequate numbers of large, abnormal megakaryocytes with hyperlobulated nuclei, rare granules, adequate amounts of demarcation membranes, but no platelet "territories," and they never produce proplatelets [51]. Proplatelets are elongated strands of megakaryocyte cytoplasm that are larger than normal platelets and later fragment into a number of platelets [52,53]. Thus, NF-E2 appears to affect megakaryocyte cytoplasmic differentiation and platelet production at a somewhat later step than GATA-1 [54].

Thrombopoietin (role in megakaryocyte maturation) — Thrombopoietin (TPO) has a major effect on almost all steps of megakaryocyte differentiation and maturation.

It has the following effects on Meg-CFCs [55,56]:

Promotes growth

Dramatically increases the rate of endomitosis

Inhibits apoptosis

Stimulates maturation

An increase in megakaryocyte ploidy is seen at even the lowest amounts of TPO and is one of the most prominent TPO effects. (See 'Size and ploidy' above.)

However, the final stage of platelet release occurs independently of TPO and in one model system was actually inhibited by large amounts of TPO [57]. (See 'Steps in platelet formation' below.)

TPO also promotes expansion of multipotent hematopoietic stem cells and precursor cells from other hematopoietic cell lineages. (See "Biology and physiology of thrombopoietin".)

Other cytokines — Other cytokines such as IL-3 and IL-11 can also promote Meg-CFC growth and megakaryocyte maturation but have little effect on endomitosis. Their actions may not be important in normal physiology. IL-6 also stimulates megakaryocyte growth, but its major in vivo effect is to increase hepatocyte production of TPO and thereby elevate the platelet count [58].

PLATELET PRODUCTION

Steps in platelet formation — Megakaryocytes normally account for 0.05 to 0.1 percent of all nucleated bone marrow cells. Each megakaryocyte produces a total of 1000 to 3000 platelets.

Historical observations suggested that platelets are shed by megakaryocytes, but electron microscopy images show the megakaryocyte cytoplasm to be divided by the demarcation membrane system into future platelet "territories" that facilitate the budding off of platelets in a highly organized process (picture 3) [59].

A modern model of platelet formation outlines the following steps, which are illustrated in the figure (figure 3):

Extension of pseudopodia – Chemoattractants such as stromal-derived factor 1 (SDF1; also called CXCL12) induce metalloproteinase production by megakaryocytes that is necessary for transendothelial migration and platelet production [56,60]. Studies in animals have suggested that chemokine-mediated interaction of megakaryocyte progenitor cells with the bone marrow vascular niche allows them to relocate to a microenvironment that is permissive for maturation and thrombopoiesis [61]. SDF1 may induce megakaryocytes to migrate to the bone marrow sinusoid where proplatelet formation occurs. Platelet granules track into the elongating pseudopodia.

Megakaryocytes use the highly redundant demarcation membrane system to send out pseudopodia into the bone marrow sinusoids [15,16,52]. The bone marrow sinusoids are lined by very thin endothelial cells that are tightly bound to each other and may even overlap [52]. The megakaryocyte pseudopodia pass through, not between, the endothelial cells, which may in turn play some role in regulating the process [52].

Budding of platelets and proplatelets – Platelets and proplatelets then bud off, possibly as a result of localized caspase activation, as illustrated in the electron micrograph (picture 4) [62]. The numbers of proplatelet processes in the sinusoids can be increased by thrombopoietic stimuli [28]. Images from a time lapse video are presented in the figure (picture 5).

Formation of these processes depends on cytoskeletal reorganization that produces pseudopods by evagination of the demarcation membrane system [63]. Platelet granules then track into the elongating pseudopodia, and the proplatelet fragments are released.

Transcription factors such as NF-E2 and GATA1 appear to play an essential role in proplatelet process formation [47,51,64]. The transcription factor nuclear factor (NF)-kB is necessary for platelet shedding from megakaryocytes [65].

Migration to the lungs – Proplatelets (or megakaryocytes) are released from the bone marrow and may travel to the lungs, where they are transformed into platelets [66-68].

Megakaryocytes are commonly found in the lungs. Early studies based on the distribution of platelet sizes suggested that megakaryocytes or large fragments of megakaryocytes passing through the lungs might be cleaved into platelets, but the extent of pulmonary platelet production could not be measured directly [67,69]. A 2017 study in living mice suggested that approximately one-half of normal platelet production occurs in the lungs [70]. This was subsequently brought into question by a 2024 study that showed that lung megakaryocytes accounted for minimal platelet production [71].

Additional findings include [28,66,68]:

Demonstration that megakaryocytes can cross the bone marrow endothelial cell barrier.

Detection of megakaryocytes and megakaryocyte nuclei in the circulation and in the pulmonary vessels.

Observations in mice of green fluorescent protein (GFP)-labeled megakaryocytes migrating to the lungs, extending proplatelet processes, and shedding platelets [70].

Although initially thought to be errant megakaryocytes that escaped from the marrow and became trapped in the lungs, this appears to be a major route of cell trafficking.

The lung was also demonstrated to be a hematopoietic organ containing multipotent hematopoietic stem cells. The clinical implications of these findings are only starting to be realized.

Bone marrow megakaryocytes respond to changes in the demand for platelets by altering their number, size, and ploidy. In animals made thrombocytopenic by the injection of antibody to platelets, bone marrow megakaryocytes increase their number, size (picture 6), and ploidy (figure 4) [7,72,73]. In animals made thrombocytotic by platelet transfusion, the opposite changes occur [7,73].

Many questions regarding the nuances of platelet production remain. Incomplete data suggest that platelets are not shed from megakaryocytes with ploidy less than 8N and that larger megakaryocytes make more platelets than smaller ones [74]. (See 'Size and ploidy' above.)

Megakaryocytes can migrate from the lungs back to the bone marrow, but the role of lung diseases in affecting platelet production or the role of platelet cytokines in contributing to lung disease are not well understood [70]. Platelets contain many cytokines with inflammatory and fibrotic characteristics, such as transforming growth factor beta (TGF-beta) and platelet-derived growth factor (PDGF).

The benefits of having platelets that are anucleate are also unexplained [75]. Platelets in lower vertebrate species such as fish and birds all contain a cell nucleus.

Control of platelet mass

Stability of platelet mass – The platelet count is remarkably constant throughout the life of an individual but remarkably variable between individuals [76,77]. It is actually the total platelet mass, rather than the platelet count, that is being controlled, and platelet mass is very similar across individuals.

Approximately one-third of the total platelet mass is normally sequestered in an exchangeable splenic pool [78]. The platelet count represents the concentration of the remaining platelets in the circulation.

Thrombocytopenia with normal platelet mass – Conditions that reduce the platelet count but not the total platelet mass include [77]:

-Pregnancy, in which the platelet count decreases as the plasma volume increases. (See "Thrombocytopenia in pregnancy", section on 'Gestational thrombocytopenia (GT)'.)

-Splenomegaly, in which the platelet count decreases proportionally to the increase in spleen size [78].

Altered platelet mass – Conditions that alter the total platelet mass include:

-Bone marrow disorders in which platelet production is impaired, such as myelodysplastic syndromes (MDS), or increased, such as myeloproliferative neoplasms. (See 'Myelodysplastic syndromes' below and 'Myeloproliferative neoplasms' below.)

-Severe liver disease, in which thrombopoietin (TPO) production is reduced. (See "Hemostatic abnormalities in patients with liver disease", section on 'Thrombocytopenia and platelet dysfunction'.)

Between-individual variation – The reference range for platelet count is large (from 150,000 to 450,000/microL), an approximately three-fold range [79]. This contrasts with the white blood cell (WBC) and red blood cell (RBC) counts, which show much less variability. However, the platelet volume (or platelet mass) across individuals has a much narrower range.

Platelet count versus platelet volume – Despite the large difference in normal platelet count between individuals, there is an inverse relation between the platelet count and the mean platelet volume (MPV) [80]; this results in a roughly constant circulating platelet mass for all individuals [80-82].

This inverse relationship extends to other species and is an example of "phylogenic canalization" [83]. As an example, mice have a normal platelet count of 1,200,000/microL and an MPV of 2.1 fL, whereas porcupines have a normal platelet count of 30,000/microL and an MPV of 105 fL.

TPO role in maintaining platelet count — TPO is the major hematopoietic growth factor responsible for regulating the circulating platelet mass. It is produced at a relatively constant rate by the liver and enters the circulation where most is cleared by avid TPO receptors (c-mpl) on normal platelets and possibly some by bone marrow megakaryocytes.

The residual amount of TPO (50 to 150 pg/mL) provides basal stimulation of megakaryocytes and a basal rate of platelet production.

When platelet production decreases, as during thrombocytopenia following chemotherapy, the platelet mass and the amount of c-mpl receptors decrease; clearance of TPO falls; TPO concentrations rise; and megakaryocyte growth is stimulated [84]. (See 'Chemotherapy' below.)

With experimental transfusion of platelets to supranormal levels, the total platelet mass and amount of c-mpl receptors rise; TPO clearance is increased; TPO concentrations fall, and megakaryocyte growth decreases (figure 4) [72].

Since the total number of circulating platelet c-mpl receptors determines the clearance rate of TPO, the normal circulating platelet mass, not the platelet count, remains relatively constant. (See 'Control of platelet mass' above.)

Platelet lifespan — Normal platelets undergo apoptosis, limiting their lifespan to approximately seven days [85-88]. (See "Platelet biology and mechanism of anti-platelet drugs".)

MEGAKARYOCYTES IN DISEASE — Several disorders produce characteristic changes in megakaryocytes [5].

The figures illustrate the effects of these disorders on megakaryocyte number and size (figure 5) and platelet count and platelet survival (figure 6).

Conditions with thrombocytopenia

Chemotherapy — Chemotherapy can alter megakaryocyte function and platelet production in several ways [89]:

Megakaryocyte apoptosis – Thrombocytopenia following most cytotoxic chemotherapy is due to a reduced number of megakaryocytes, perhaps due to drug-induced apoptosis of stem cells and/or megakaryocyte progenitor cells [90]. Platelet kinetic studies have also demonstrated ineffective thrombopoiesis with cytotoxic chemotherapy.

Hepatic thrombopoietin (TPO) production is constant, and when the platelet count decreases, clearance of TPO by platelets is reduced, resulting in a net increase in TPO levels. This increases the average ploidy of the remaining megakaryocytes, increasing platelet production [91]. (See 'Platelet production' above.)

Inhibited platelet production – The proteasome inhibitor bortezomib causes thrombocytopenia by inhibiting nuclear factor (NF)-kB, which is necessary for platelet shedding from megakaryocytes [65].

Platelet apoptosisEtoposide causes platelets to undergo apoptosis. Several drugs in development (eg, ABT-737) inhibit the anti-apoptotic factors Bcl-2 and Bcl-xL and produce rapid and profound thrombocytopenia by causing platelets apoptosis [87,89].

Vitamin B12 deficiency — Vitamin B12 is a cofactor in DNA synthesis pathways. (See "Causes and pathophysiology of vitamin B12 and folate deficiencies", section on 'DNA synthesis, RNA synthesis, DNA methylation'.)

In vitamin B12 deficiency, there is a marked increase in megakaryocyte number but decreased ploidy, likely due to decreased DNA replication in megakaryocyte progenitor cells. (See 'Stem and progenitor cells' above.)

This leads to an expanded megakaryocyte mass (figure 7) but reduced platelet production per megakaryocyte (figure 8).

Similar effects occur in the white blood cell (WBC) and red blood cell (RBC) lineages. (See "Causes and pathophysiology of vitamin B12 and folate deficiencies".)

Congestive splenomegaly — Congestive splenomegaly can be associated with thrombocytopenia by different mechanisms, depending on whether liver function is also abnormal.

Normal liver function – With congestive splenomegaly and normal liver function, such as in mononucleosis or other postinfectious splenomegaly, thrombocytopenia is primarily due to redistribution of the normal circulating mass of platelets to the spleen, without major alterations in megakaryocyte production or platelet survival (figure 6) [78,92]. (See 'Control of platelet mass' above and "Splenomegaly and other splenic disorders in adults", section on 'Hypersplenism'.)

Concomitant liver disease – In patients with splenomegaly due to liver disease, fewer platelets are produced because hepatic production of TPO is decreased (figure 5 and figure 7) [93]. Platelet redistribution to the spleen may also occur. (See "Biology and physiology of thrombopoietin".)

Immune thrombocytopenia (ITP) — ITP is caused by autoantibody-mediated platelet destruction, primarily by macrophages in the liver and spleen.

Reduced platelet numbers leads to an increase in TPO, which in turn increases the number, size, and ploidy of bone marrow megakaryocytes (figure 5).

However, antiplatelet antibodies and cytotoxic T cells target megakaryocytes and promote megakaryocyte apoptosis, thereby preventing platelet shedding [94-100]. (See 'Steps in platelet formation' above.)

The electron micrographs show megakaryocytes undergoing apoptosis in the bone marrows of patients with ITP (picture 7) [97]. (See "Immune thrombocytopenia (ITP) in adults: Clinical manifestations and diagnosis", section on 'Pathogenesis'.)

Myelodysplastic syndromes — Both thrombocytopenia and thrombocytosis may be seen in myelodysplastic syndromes (MDS), findings that have been attributed to abnormal megakaryocytes. The morphologic picture consists of an increased number of small megakaryocytes of low ploidy, occasionally displaying a characteristic "pawn ball" nucleus with three lobes (picture 8) [101-103]. (See "Clinical manifestations, diagnosis, and classification of myelodysplastic syndromes (MDS)", section on 'Cytopenias'.)

Platelet kinetic studies have demonstrated a greatly expanded megakaryocyte mass (increased number of megakaryocytes of low ploidy) and ineffective platelet production from the megakaryocytes (figure 5 and figure 8) [5].

However, platelet production may be increased in the 5q- syndrome. (See "Cytogenetics, molecular genetics, and pathophysiology of myelodysplastic syndromes/neoplasms (MDS)", section on 'del(5q)'.)

Conditions with thrombocytosis — Evaluation of thrombocytosis is presented separately. (See "Approach to the patient with thrombocytosis".)

Reactive thrombocytosis — Reactive thrombocytosis occurs in association with iron deficiency, malignancy, and inflammatory states; the table summarizes these and other causes (table 1). It is associated with an increased number of megakaryocytes and megakaryocyte mass but reduced ploidy (figure 5).

Iron deficiency — The platelet count rises in approximately one-third of patients with severe iron deficiency [104]; this increase may be associated with an increased risk of thrombosis [104]. TPO levels are not increased, and no iron-responsive regulatory elements have been identified in TPO-dependent signaling pathways.

Studies in mice rendered iron deficient due to knockout of the TMPRSS6 gene suggest that megakaryocytic-erythroid progenitor (MEP) cells were more likely to be committed to megakaryocytic differentiation due to a switch in gene expression and reduced signalling via ERK (extracellular signal-related kinase) pathways [105]. It was suggested that this switch of the MEP away from erythroid differentiation was a defense against making more RBCs in an iron-deficient environment.

Cancer and inflammation — Increases in platelet count may be seen with cancer and during acute infection or inflammatory states. TPO may be increased or decreased depending on the setting.

Increases in platelet count that can occur in cancer and inflammatory states are likely secondary to the expansion of megakaryocyte number due to inflammatory cytokines such as interleukin-6 (IL-6) and IL-11 [106-108]. IL-11 stimulates megakaryocyte growth and platelet production independent of TPO signaling. Ploidy is reduced due to reduced TPO, which occurs because the expanded platelet mass increases TPO clearance. (See 'TPO role in maintaining platelet count' above.)

In ovarian cancer, there is often marked thrombocytosis associated with increased levels of IL-6 and TPO. Studies have shown that the ovarian tumors secrete IL-6, which in turn increases hepatic production of TPO [108].

Rebound thrombocytosis — The platelet count often increases above normal during recovery from an acute thrombocytopenic event caused by chemotherapy, infection, or alcohol-induced bone marrow suppression. The peak platelet count occurs approximately 14 days after the platelet nadir and is presumed to reflect the effect of increased circulating TPO during the thrombocytopenic period [109-111].

Redistributive thrombocytosis — The spleen sequesters an exchangeable pool of about one-third of the platelet mass [82]. After splenectomy there is a period of redistributive thrombocytosis with the platelet count ultimately returning to values above baseline [112].

Myeloproliferative neoplasms — In essential thrombocythemia (ET) and the related myeloproliferative neoplasms (MPNs), chronic myeloid leukemia (CML), polycythemia vera (PV), and primary myelofibrosis (PMF), clonal proliferation of megakaryocytes of high ploidy and active platelet production can lead to very high platelet counts [113]. (See "Overview of the myeloproliferative neoplasms".)

Acquired mutations in certain genes are responsible for increased megakaryopoiesis and platelet production:

JAK2 – Mutations in JAK2, which encodes a Janus kinase, occur in approximately 60 to 65 percent of patients with ET and virtually 100 percent of patients with PV. These mutations make the early hematopoietic cells especially sensitive to growth factors and cytokines that act via altered GATA1-JAK/STAT signaling. (See "Overview of the myeloproliferative neoplasms", section on 'JAK2 mutations'.)

MPL – Mutations in MPL, which encodes the TPO receptor, are seen in approximately 5 to 10 percent of patients with ET [114,115]. (See "Overview of the myeloproliferative neoplasms", section on 'MPL mutations'.)

CALR – Mutations in CALR, which encodes calreticulin, occur in approximately 70 percent of patients with ET or PMF who do not carry a mutation in either JAK2 or MPL [114,115]. (See "Overview of the myeloproliferative neoplasms", section on 'Calreticulin (CALR) mutations'.)

TPO levels are not appropriately downregulated by the high platelet mass in MPNs; in most cases TPO levels are normal or slightly elevated [116,117]. TPO receptors on platelets and possibly megakaryocyte are markedly reduced via an uncertain mechanism [117,118]. This results in a net overall normal clearance of TPO despite elevated platelet counts. Platelet kinetic data suggest the clonal proliferation is autonomous of TPO, since platelet mass is increased (figure 6) and megakaryocyte ploidy is high (figure 5).

Thrombocytosis in PV may be present for years before the RBC mass becomes elevated.

In PMF, bone marrow megakaryocytes are increased, with a large amount of fibrosis and occasional megakaryocyte dysplasia. The fibrotic response is due to a polyclonal proliferation of fibroblasts, which has been attributed to the release of mesenchymal growth factors such as transforming growth factor (TGF)-beta from the abnormal megakaryocytes [119]. However, studies using mouse models are able to uncouple increases in bone marrow megakaryocytes from fibrosis, suggesting that other mediators may be responsible for the bone marrow fibrosis [120].

SOCIETY GUIDELINE LINKS — Links to society and government-sponsored guidelines from selected countries and regions around the world are provided separately. (See "Society guideline links: Immune thrombocytopenia (ITP) and other platelet disorders" and "Society guideline links: Myeloproliferative neoplasms".)

SUMMARY

Megakaryocytes – Megakaryocytes are the largest cell in the bone marrow (picture 2), with average diameter 20 to 25 (up to 50 to 60) microns. They are polyploid, containing 2 to 32 times the normal diploid DNA content within a lobulated nucleus, generated by endomitosis. They produce alpha granules and dense granules; megakaryocytes and platelets, also endocytose granule constituents. (See 'Characteristics' above.)

Megakaryocyte origins – Multipotent hematopoietic stem cells (HSCs) (figure 1) give rise to megakaryocyte-erythroid progenitor cells (MEPs) that can differentiate into megakaryocytes. Thrombopoietin (TPO) stimulates HSCs and acts on megakaryocyte colony-forming cells to promote growth, increase endomitosis, inhibit apoptosis, and stimulate maturation. (See 'Origin and differentiation' above.)

Platelet production – Each megakaryocyte produces 1000 to 3000 platelets (figure 3). Megakaryocytes in bone marrow sinusoids extend pseudopods into endothelial cells using the demarcation membrane system (picture 4). Proplatelets bud off (picture 5) and migrate to the lungs, where they are transformed into platelets. (See 'Steps in platelet formation' above.)

Platelet count regulation – The total platelet mass is kept constant by TPO. Megakaryocytes and platelets express TPO receptors that take up TPO, leaving residual TPO to stimulate megakaryocyte and platelet production. When the platelet count increases, TPO uptake is greater and platelet production declines. When the platelet count decreases, less TPO is taken up by platelets; circulating TPO levels rise; and platelet production is stimulated. Platelets live approximately seven days. Approximately one-third of the platelet mass is sequestered in the spleen. (See 'Control of platelet mass' above and 'TPO role in maintaining platelet count' above and 'Platelet lifespan' above.)

Megakaryocytes in disease

Thrombocytopenic conditions – (See 'Conditions with thrombocytopenia' above and "Diagnostic approach to thrombocytopenia in adults".)

-Cytotoxic chemotherapy can cause apoptosis of HSCs and/or MEPs; bortezomib can prevent platelet shedding from megakaryocytes, and etoposide can cause platelet apoptosis.

-Vitamin B12 deficiency decreases platelet production by reducing megakaryocyte ploidy.

-Splenomegaly redistributes the circulating platelet mass; concomitant liver disease decreases TPO production.

-In immune thrombocytopenia (ITP), autoantibodies and cytotoxic T cells promote platelet destruction by macrophages and cause megakaryocyte apoptosis.

-In myelodysplastic syndromes (MDS), megakaryocyte mass is increased but platelet production is ineffective.

Thrombocytotic conditions – (See 'Conditions with thrombocytosis' above and "Approach to the patient with thrombocytosis".)

-Reactive thrombocytosis can be seen with iron deficiency, cancer, inflammation, and other conditions listed in the table (table 1).

-Rebound thrombocytosis can occur with recovery from bone marrow suppression by chemotherapy, infection, or alcohol. Redistributive thrombocytosis can occur following splenectomy.

-Clonal myeloproliferative neoplasms (MPNs), including essential thrombocythemia (ET), chronic myeloid leukemia (CML), polycythemia vera (PV), and primary myelofibrosis (PMF), can cause thrombocytosis by a TPO-independent mechanism driven by mutations in JAK2, CALR, and MPL. MPL encodes the TPO receptor.

  1. Wright JH. The origin and nature of blood plates. Boston Medical and Surgical Journal 1906; 154:643.
  2. Wright JH. The histogenesis of the blood platelets. J Morphol 1910; 21:263.
  3. Deutsch VR, Tomer A. Megakaryocyte development and platelet production. Br J Haematol 2006; 134:453.
  4. Kaushansky K. Historical review: megakaryopoiesis and thrombopoiesis. Blood 2008; 111:981.
  5. Harker LA, Finch CA. Thrombokinetics in man. J Clin Invest 1969; 48:963.
  6. Kuter DJ, Greenberg SM, Rosenberg RD. Analysis of megakaryocyte ploidy in rat bone marrow cultures. Blood 1989; 74:1952.
  7. Jackson CW, Brown LK, Somerville BC, et al. Two-color flow cytometric measurement of DNA distributions of rat megakaryocytes in unfixed, unfractionated marrow cell suspensions. Blood 1984; 63:768.
  8. Machlus KR, Thon JN, Italiano JE Jr. Interpreting the developmental dance of the megakaryocyte: a review of the cellular and molecular processes mediating platelet formation. Br J Haematol 2014; 165:227.
  9. Zybina TG, Zybina EV. Role of cell cycling and polyploidy in placental trophoblast of different mammalian species. Reprod Domest Anim 2020; 55:895.
  10. Zybina TG, Zybina EV. Cell reproduction and genome multiplication in the proliferative and invasive trophoblast cell populations of mammalian placenta. Cell Biol Int 2005; 29:1071.
  11. Raslova H, Roy L, Vourc'h C, et al. Megakaryocyte polyploidization is associated with a functional gene amplification. Blood 2003; 101:541.
  12. Raslova H, Kauffmann A, Sekkaï D, et al. Interrelation between polyploidization and megakaryocyte differentiation: a gene profiling approach. Blood 2007; 109:3225.
  13. Berridge MV, Ralph SJ, Tan AS. Cell-lineage antigens of the stem cell-megakaryocyte-platelet lineage are associated with the platelet IIb-IIIa glycoprotein complex. Blood 1985; 66:76.
  14. Tomer A. Human marrow megakaryocyte differentiation: multiparameter correlative analysis identifies von Willebrand factor as a sensitive and distinctive marker for early (2N and 4N) megakaryocytes. Blood 2004; 104:2722.
  15. Radley JM, Haller CJ. The demarcation membrane system of the megakaryocyte: a misnomer? Blood 1982; 60:213.
  16. Behnke O. An electron microscope study of the megacaryocyte of the rat bone marrow. I. The development of the demarcation membrane system and the platelet surface coat. J Ultrastruct Res 1968; 24:412.
  17. MacPherson GG. Changes in megakaryocyte development following thrombocytopenia. Br J Haematol 1974; 26:105.
  18. Schulze H, Korpal M, Hurov J, et al. Characterization of the megakaryocyte demarcation membrane system and its role in thrombopoiesis. Blood 2006; 107:3868.
  19. Greenberg SM, Kuter DJ, Rosenberg RD. In vitro stimulation of megakaryocyte maturation by megakaryocyte stimulatory factor. J Biol Chem 1987; 262:3269.
  20. Handagama P, Bainton DF, Jacques Y, et al. Kistrin, an integrin antagonist, blocks endocytosis of fibrinogen into guinea pig megakaryocyte and platelet alpha-granules. J Clin Invest 1993; 91:193.
  21. Handagama PJ, Amrani DL, Shuman MA. Endocytosis of fibrinogen into hamster megakaryocyte alpha granules is dependent on a dimeric gamma A configuration. Blood 1995; 85:1790.
  22. Harrison P, Wilbourn B, Debili N, et al. Uptake of plasma fibrinogen into the alpha granules of human megakaryocytes and platelets. J Clin Invest 1989; 84:1320.
  23. Handagama PJ, Shuman MA, Bainton DF. Incorporation of intravenously injected albumin, immunoglobulin G, and fibrinogen in guinea pig megakaryocyte granules. J Clin Invest 1989; 84:73.
  24. White JG. Serotonin storage organelles in human megakaryocytes. Am J Pathol 1971; 63:403.
  25. Schick PK, Weinstein M. A marker for megakaryocytes: serotonin accumulation in guinea pig megakaryocytes. J Lab Clin Med 1981; 98:607.
  26. Odell TT Jr, Jackson CW. Polyploidy and maturation of rat megakaryocytes. Blood 1968; 32:102.
  27. Mazur EM, Hoffman R, Chasis J, et al. Immunofluorescent identification of human megakaryocyte colonies using an antiplatelet glycoprotein antiserum. Blood 1981; 57:277.
  28. Poujol C, Tronik-Le Roux D, Tropel P, et al. Ultrastructural analysis of bone marrow hematopoiesis in mice transgenic for the thymidine kinase gene driven by the alpha IIb promoter. Blood 1998; 92:2012.
  29. Tronik-Le Roux D, Roullot V, Schweitzer A, et al. Suppression of erythro-megakaryocytopoiesis and the induction of reversible thrombocytopenia in mice transgenic for the thymidine kinase gene targeted by the platelet glycoprotein alpha IIb promoter. J Exp Med 1995; 181:2141.
  30. Tropel P, Roullot V, Vernet M, et al. A 2.7-kb portion of the 5' flanking region of the murine glycoprotein alphaIIb gene is transcriptionally active in primitive hematopoietic progenitor cells. Blood 1997; 90:2995.
  31. Orkin SH, Shivdasani RA, Fujiwara Y, McDevitt MA. Transcription factor GATA-1 in megakaryocyte development. Stem Cells 1998; 16 Suppl 2:79.
  32. de Sauvage FJ, Hass PE, Spencer SD, et al. Stimulation of megakaryocytopoiesis and thrombopoiesis by the c-Mpl ligand. Nature 1994; 369:533.
  33. Edelstein LC, Bray PF. MicroRNAs in platelet production and activation. Blood 2011; 117:5289.
  34. Lu J, Guo S, Ebert BL, et al. MicroRNA-mediated control of cell fate in megakaryocyte-erythrocyte progenitors. Dev Cell 2008; 14:843.
  35. Barroga CF, Pham H, Kaushansky K. Thrombopoietin regulates c-Myb expression by modulating micro RNA 150 expression. Exp Hematol 2008; 36:1585.
  36. Miyazaki H, Inoue H, Yanagida M, et al. Purification of rat megakaryocyte colony-forming cells using a monoclonal antibody against rat platelet glycoprotein IIb/IIIa. Exp Hematol 1992; 20:855.
  37. Levene RB, Lamaziere JM, Broxmeyer HE, et al. Human megakaryocytes. V. Changes in the phenotypic profile of differentiating megakaryocytes. J Exp Med 1985; 161:457.
  38. Nakeff A. Colony-forming unit, megakaryocyte (CFU-m): Its use in elucidating the kinetics and humoral control of the megakaryocytic committed progenitor cell compartment. In: Experimental Hematology Today, Baum SJ, Ledney DG (Eds), Springer, New York 1977. p.111.
  39. Broudy VC, Lin NL, Fox N, et al. Thrombopoietin stimulates colony-forming unit-megakaryocyte proliferation and megakaryocyte maturation independently of cytokines that signal through the gp130 receptor subunit. Blood 1996; 88:2026.
  40. Dolzhanskiy A, Basch RS, Karpatkin S. The development of human megakaryocytes: III. Development of mature megakaryocytes from highly purified committed progenitors in synthetic culture media and inhibition of thrombopoietin-induced polyploidization by interleukin-3. Blood 1997; 89:426.
  41. Nagata Y, Muro Y, Todokoro K. Thrombopoietin-induced polyploidization of bone marrow megakaryocytes is due to a unique regulatory mechanism in late mitosis. J Cell Biol 1997; 139:449.
  42. Zimmet JM, Ladd D, Jackson CW, et al. A role for cyclin D3 in the endomitotic cell cycle. Mol Cell Biol 1997; 17:7248.
  43. Zhang Y, Wang Z, Liu DX, et al. Ubiquitin-dependent degradation of cyclin B is accelerated in polyploid megakaryocytes. J Biol Chem 1998; 273:1387.
  44. Zimmet JM, Toselli P, Ravid K. Cyclin D3 and megakaryocyte development: exploration of a transgenic phenotype. Stem Cells 1998; 16 Suppl 2:97.
  45. Fujiwara Y, Browne CP, Cunniff K, et al. Arrested development of embryonic red cell precursors in mouse embryos lacking transcription factor GATA-1. Proc Natl Acad Sci U S A 1996; 93:12355.
  46. Muntean AG, Crispino JD. Differential requirements for the activation domain and FOG-interaction surface of GATA-1 in megakaryocyte gene expression and development. Blood 2005; 106:1223.
  47. Shivdasani RA, Fujiwara Y, McDevitt MA, Orkin SH. A lineage-selective knockout establishes the critical role of transcription factor GATA-1 in megakaryocyte growth and platelet development. EMBO J 1997; 16:3965.
  48. Huang Z, Richmond TD, Muntean AG, et al. STAT1 promotes megakaryopoiesis downstream of GATA-1 in mice. J Clin Invest 2007; 117:3890.
  49. Shivdasani RA, Orkin SH. Erythropoiesis and globin gene expression in mice lacking the transcription factor NF-E2. Proc Natl Acad Sci U S A 1995; 92:8690.
  50. 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; 81:695.
  51. Lecine P, Villeval JL, Vyas P, et al. Mice lacking transcription factor NF-E2 provide in vivo validation of the proplatelet model of thrombocytopoiesis and show a platelet production defect that is intrinsic to megakaryocytes. Blood 1998; 92:1608.
  52. Becker RP, De Bruyn PP. The transmural passage of blood cells into myeloid sinusoids and the entry of platelets into the sinusoidal circulation; a scanning electron microscopic investigation. Am J Anat 1976; 145:183.
  53. Tong M, Seth P, Penington DG. Proplatelets and stress platelets. Blood 1987; 69:522.
  54. Lecine P, Shivdasani RA. Cellular and molecular biology of megakaryocyte differentiation in the absence of lineage-restricted transcription factors. Stem Cells 1998; 16 Suppl 2:91.
  55. Zauli G, Vitale M, Falcieri E, et al. In vitro senescence and apoptotic cell death of human megakaryocytes. Blood 1997; 90:2234.
  56. Majka M, Janowska-Wieczorek A, Ratajczak J, et al. Stromal-derived factor 1 and thrombopoietin regulate distinct aspects of human megakaryopoiesis. Blood 2000; 96:4142.
  57. 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; 95:227.
  58. Senchenkova EY, Komoto S, Russell J, et al. Interleukin-6 mediates the platelet abnormalities and thrombogenesis associated with experimental colitis. Am J Pathol 2013; 183:173.
  59. YAMADA E. The fine structure of the megakaryocyte in the mouse spleen. Acta Anat (Basel) 1957; 29:267.
  60. Lane WJ, Dias S, Hattori K, et al. Stromal-derived factor 1-induced megakaryocyte migration and platelet production is dependent on matrix metalloproteinases. Blood 2000; 96:4152.
  61. Avecilla ST, Hattori K, Heissig B, et al. Chemokine-mediated interaction of hematopoietic progenitors with the bone marrow vascular niche is required for thrombopoiesis. Nat Med 2004; 10:64.
  62. De Botton S, Sabri S, Daugas E, et al. Platelet formation is the consequence of caspase activation within megakaryocytes. Blood 2002; 100:1310.
  63. Thon JN, Italiano JE. Platelet formation. Semin Hematol 2010; 47:220.
  64. Lecine P, Blank V, Shivdasani R. Characterization of the hematopoietic transcription factor NF-E2 in primary murine megakaryocytes. J Biol Chem 1998; 273:7572.
  65. Lonial S, Waller EK, Richardson PG, et al. Risk factors and kinetics of thrombocytopenia associated with bortezomib for relapsed, refractory multiple myeloma. Blood 2005; 106:3777.
  66. Kaufman RM, Airo R, Pollack S, Crosby WH. Circulating megakaryocytes and platelet release in the lung. Blood 1965; 26:720.
  67. Trowbridge EA, Martin JF, Slater DN. Evidence for a theory of physical fragmentation of megakaryocytes, implying that all platelets are produced in the pulmonary circulation. Thromb Res 1982; 28:461.
  68. Pedersen NT. Occurrence of megakaryocytes in various vessels and their retention in the pulmonary capillaries in man. Scand J Haematol 1978; 21:369.
  69. Levine RF, Eldor A, Shoff PK, et al. Circulating megakaryocytes: delivery of large numbers of intact, mature megakaryocytes to the lungs. Eur J Haematol 1993; 51:233.
  70. Lefrançais E, Ortiz-Muñoz G, Caudrillier A, et al. The lung is a site of platelet biogenesis and a reservoir for haematopoietic progenitors. Nature 2017; 544:105.
  71. Asquith NL, Carminita E, Camacho V, et al. The bone marrow is the primary site of thrombopoiesis. Blood 2024; 143:272.
  72. Penington DG, Olsen TE. Megakaryocytes in states of altered platelet production: cell numbers, size and DNA content. Br J Haematol 1970; 18:447.
  73. Kuter DJ, Rosenberg RD. Regulation of megakaryocyte ploidy in vivo in the rat. Blood 1990; 75:74.
  74. Mattia G, Vulcano F, Milazzo L, et al. Different ploidy levels of megakaryocytes generated from peripheral or cord blood CD34+ cells are correlated with different levels of platelet release. Blood 2002; 99:888.
  75. Levin J. The evolution of mammalian platelets. In: Thrombopoiesis and Thrombopoietins: Molecular, Cellular, Preclinical, and Clinical Biology, Kuter DJ, Hunt P, Sheridan W, et al (Eds), Humana Press, Totowa 1997.
  76. Kuter DJ. The physiology of platelet production. Stem Cells 1996; 14 Suppl 1:88.
  77. BRECHER G, CRONKITE EP. Morphology and enumeration of human blood platelets. J Appl Physiol 1950; 3:365.
  78. Aster RH. Pooling of platelets in the spleen: role in the pathogenesis of "hypersplenic" thrombocytopenia. J Clin Invest 1966; 45:645.
  79. Giles C. The platelet count and mean platelet volume. Br J Haematol 1981; 48:31.
  80. Bessman JD, Williams LJ, Gilmer PR Jr. Mean platelet volume. The inverse relation of platelet size and count in normal subjects, and an artifact of other particles. Am J Clin Pathol 1981; 76:289.
  81. Thompson CB. From precursor to product; how do megakaryocytes produce platelets. In: Megakaryocyte Development and Function, Levine RF, Williams N, Levin J, et al (Eds), Liss, New York 1986. p.361.
  82. De Gabriele G, Penington DG. Regulation of platelet production: "hypersplenism" in the experimental animal. Br J Haematol 1967; 13:384.
  83. von Behrens WE. Evidence of phylogenetic canalisation of the circulating platelet mass in man. Thromb Diath Haemorrh 1972; 27:159.
  84. Kuter DJ, Beeler DL, Rosenberg RD. The purification of megapoietin: a physiological regulator of megakaryocyte growth and platelet production. Proc Natl Acad Sci U S A 1994; 91:11104.
  85. Li J, Xia Y, Bertino AM, et al. The mechanism of apoptosis in human platelets during storage. Transfusion 2000; 40:1320.
  86. Bertino AM, Qi XQ, Li J, et al. Apoptotic markers are increased in platelets stored at 37 degrees C. Transfusion 2003; 43:857.
  87. Zhang H, Nimmer PM, Tahir SK, et al. Bcl-2 family proteins are essential for platelet survival. Cell Death Differ 2007; 14:943.
  88. Mason KD, Carpinelli MR, Fletcher JI, et al. Programmed anuclear cell death delimits platelet life span. Cell 2007; 128:1173.
  89. Kuter DJ. Treatment of chemotherapy-induced thrombocytopenia in patients with non-hematologic malignancies. Haematologica 2022; 107:1243.
  90. Zeuner A, Signore M, Martinetti D, et al. Chemotherapy-induced thrombocytopenia derives from the selective death of megakaryocyte progenitors and can be rescued by stem cell factor. Cancer Res 2007; 67:4767.
  91. Nichol JL, Hokom MM, Hornkohl A, et al. Megakaryocyte growth and development factor. Analyses of in vitro effects on human megakaryopoiesis and endogenous serum levels during chemotherapy-induced thrombocytopenia. J Clin Invest 1995; 95:2973.
  92. Penington DG. Studies of platelet production and sequestration in the experimental animal. Proc R Soc Med 1968; 61:601.
  93. Peck-Radosavljevic M, Zacherl J, Meng YG, et al. Is inadequate thrombopoietin production a major cause of thrombocytopenia in cirrhosis of the liver? J Hepatol 1997; 27:127.
  94. Ballem PJ, Segal GM, Stratton JR, et al. Mechanisms of thrombocytopenia in chronic autoimmune thrombocytopenic purpura. Evidence of both impaired platelet production and increased platelet clearance. J Clin Invest 1987; 80:33.
  95. McMillan R, Wang L, Tomer A, et al. Suppression of in vitro megakaryocyte production by antiplatelet autoantibodies from adult patients with chronic ITP. Blood 2004; 103:1364.
  96. Li S, Wang L, Zhao C, et al. CD8+ T cells suppress autologous megakaryocyte apoptosis in idiopathic thrombocytopenic purpura. Br J Haematol 2007; 139:605.
  97. Houwerzijl EJ, Blom NR, van der Want JJ, et al. Ultrastructural study shows morphologic features of apoptosis and para-apoptosis in megakaryocytes from patients with idiopathic thrombocytopenic purpura. Blood 2004; 103:500.
  98. Kuter DJ. New drugs for familiar therapeutic targets: thrombopoietin receptor agonists and immune thrombocytopenic purpura. Eur J Haematol 2008; 9:.
  99. Bussel JB, Kuter DJ, George JN, et al. AMG 531, a thrombopoiesis-stimulating protein, for chronic ITP. N Engl J Med 2006; 355:1672.
  100. Chow L, Aslam R, Speck ER, et al. A murine model of severe immune thrombocytopenia is induced by antibody- and CD8+ T cell-mediated responses that are differentially sensitive to therapy. Blood 2010; 115:1247.
  101. Rosenthal DS, Moloney WC. Refractory dysmyelopoietic anemia and acute leukemia. Blood 1984; 63:314.
  102. Kobayashi Y, Ozawa M, Maruo N, Kondo M. Megakaryocytic ploidy in myelodysplastic syndromes. Leuk Lymphoma 1993; 9:55.
  103. Steensma DP. "Pawn Ball Megakaryocytes": from the marvellous medici and dear Old Saint Nick to the unsanctified marrow of myelodysplasia. Hematology 2003; 8:11.
  104. Song AB, Kuter DJ, Al-Samkari H. Characterization of the rate, predictors, and thrombotic complications of thrombocytosis in iron deficiency anemia. Am J Hematol 2020; 95:1180.
  105. Xavier-Ferrucio J, Scanlon V, Li X, et al. Low iron promotes megakaryocytic commitment of megakaryocytic-erythroid progenitors in humans and mice. Blood 2019; 134:1547.
  106. Beck JT, Hsu SM, Wijdenes J, et al. Brief report: alleviation of systemic manifestations of Castleman's disease by monoclonal anti-interleukin-6 antibody. N Engl J Med 1994; 330:602.
  107. Kaser A, Brandacher G, Steurer W, et al. Interleukin-6 stimulates thrombopoiesis through thrombopoietin: role in inflammatory thrombocytosis. Blood 2001; 98:2720.
  108. Stone RL, Nick AM, McNeish IA, et al. Paraneoplastic thrombocytosis in ovarian cancer. N Engl J Med 2012; 366:610.
  109. Moffatt TL, Schwartz A. Rebound thrombocytosis after alcohol withdrawal. N Engl J Med 1976; 295:1322.
  110. Othman TA, Mei M, Zhang J, et al. Rebound thrombocytosis is associated with response in AML patients treated with venetoclax and hypomethylating agents. Am J Hematol 2021; 96:E140.
  111. A R, R Y, B B, Kc G. Rebound thrombocytosis causing MI following dengue fever? Indian Heart J 2007; 59:94.
  112. HAYES DM, SPURR CL, HUTAFF LW, SHEETS JA. Post-splenectomy thrombocytosis. Ann Intern Med 1963; 58:259.
  113. Jacobsson S, Carneskog J, Ridell B, et al. Flow cytometric analysis of megakaryocyte ploidy in chronic myeloproliferative disorders and reactive thrombocytosis. Eur J Haematol 1996; 56:287.
  114. Nangalia J, Massie CE, Baxter EJ, et al. Somatic CALR mutations in myeloproliferative neoplasms with nonmutated JAK2. N Engl J Med 2013; 369:2391.
  115. Klampfl T, Gisslinger H, Harutyunyan AS, et al. Somatic mutations of calreticulin in myeloproliferative neoplasms. N Engl J Med 2013; 369:2379.
  116. Pitcher L, Taylor K, Nichol J, et al. Thrombopoietin measurement in thrombocytosis: dysregulation and lack of feedback inhibition in essential thrombocythaemia. Br J Haematol 1997; 99:929.
  117. Horikawa Y, Matsumura I, Hashimoto K, et al. Markedly reduced expression of platelet c-mpl receptor in essential thrombocythemia. Blood 1997; 90:4031.
  118. Li J, Xia Y, Kuter DJ. Analysis of the thrombopoietin receptor (MPL) on platelets from normal and essential thrombocythemic (ET) patients (abstract). Blood 1996; 88:545a.
  119. Kuter DJ, Bain B, Mufti G, et al. Bone marrow fibrosis: pathophysiology and clinical significance of increased bone marrow stromal fibres. Br J Haematol 2007; 139:351.
  120. Frey BM, Rafii S, Teterson M, et al. Adenovector-mediated expression of human thrombopoietin cDNA in immune-compromised mice: insights into the pathophysiology of osteomyelofibrosis. J Immunol 1998; 160:691.
Topic 6672 Version 28.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟