Myeloproliferative neoplasms (MPNs) are a heterogeneous group of chronic hematological diseases that arise from the clonal expansion of abnormal hematopoietic stem cells, of which polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF) have been extensively reviewed in the context of control of clonal expansion, fibrosis, and other phenotypes. Herein, we review current knowledge on the influence of different forms of MPN on bone health. In studies, murine models and human data have implicated various degrees of effect of different forms of MPN on bone density and on osteoblast proliferation and differentiation. Most results have shown that bone volume is generally increased in patients with PMF, whereas it is slightly decreased or not altered in patients with ET or PV, although possible differences between male and female phenotypes were not fully explored in most MPN forms. Osteosclerosis in patients with PMF is a serious complication that can lead to bone marrow failure, and the loss of bone reported in some patients with ET or PV can lead to osteoporotic fractures. Some MPN forms are associated with an increased number of megakaryocytes (MKs), and several of the MK-associated factors in MPN are known to affect bone development. We review known mechanisms involved in these processes, with a focus on the role of MKs and secreted factors. Understanding MPN-associated changes in bone health could improve early intervention and treatment of this side effect of the pathology.

Myeloproliferative neoplasms (MPNs) are a heterogeneous group of rare and chronic hematological diseases that arise from the clonal expansion of abnormal hematopoietic stem cells. According to the World Health Organization classification of 2016, the Philadelphia-negative, or classic MPNs consist of polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF).1 PMF can be described as either prefibrotic (pre PMF) or overt PMF, and myelofibrosis (MF) can be either primary (ie, diagnosed without any preceding MPN) or secondary (developed after a diagnosis of PV or ET [post-MPN MF]).1 ET is the most common classic MPN, followed by PV and PMF with an annual incidence of 1.03, 0.84, and 0.47 per 100 000, respectively.2 The median survival is ∼18 years for ET, 15 years for PV, and only 4.4 years for PMF.3,4

Driver mutations have been identified in the Janus kinase 2 (JAK2) gene and in the myeloproliferative leukemia virus oncogene (MPL) and calreticulin (CALR) gene that cause dysregulation of JAK-STAT signaling. The single-point mutation JAK2V617F in exon 14 is present in ∼96%, ∼55%, and ∼65% of patients with PV, ET, and PMF, respectively,4-6 and mutations in exon 12 of JAK2 have been found in ∼3% of PV cases.4-7 Approximately 3% of ET cases and ∼10% of cases of PMF have mutations in the MPL gene,4,6,8,9 and the presence of CALR gene mutations has been reported in ∼20% of ET and ∼25% of PMF cases.4,6,10 Yet, further genomic analysis revealed that >50% of patients with MPN have additional genetic aberrations.11 Various genes can be affected, such as SH2B3, DNMT3A, TET2, IDH1, IDH2, ASXL1, EZH2, U2AF1, SF3B1, SRSF2, ZRSR2, and TP53.4,6,12

The genomic classification of patients has provided the opportunity for generation of promising prognostic models that could enhance treatment management.13 That MPN driver mutations can be detected very early in life before diagnosis, with JAK2V617F and DNMT3A mutations being present even in utero, could provide early intervention and a better disease outcome.14,15 Although the different forms of MPN have been extensively studied and reviewed in the context of clonal expansion, fibrosis, and other phenotypes, the influence of MPN subtypes on bone homeostasis has been debated. This review summarizes published knowledge on the effects of MPN on bone health and the possible mechanisms leading to these influences.

A brief survey of major regulators of bone development is given as the basis for probing the mechanisms of the effects of MPN on bone health.

Regulation of bone formation

Skeletal bone consists of cortical bone, which is the hard outer layer, and trabecular bone, which is found inside the tissue and has a large surface exposed to the bone marrow (BM) and blood flow. Bone homeostasis is based on a balance between formation by osteoblasts (OBs) and resorption by osteoclasts (OCs). Bone can be formed by either intramembranous or endochondral osteogenesis. In the former process, mesenchymal stem cells (MSCs) differentiate directly into OBs, generating the flat bones, whereas in the latter process, MSCs differentiate into chondrocytes that form a cartilage intermediate that must undergo ossification by OBs, thus giving rise to the long bones.16 In endochondral osteogenesis, the formed chondrocytes switch from a proliferating to a nonproliferating state, and they become hypertrophic and then apoptotic, enabling OBs to start the ossification process. In both intramembranous and endochondral osteogenesis, osteoblastic bone formation is the same.

The first step in skeletogenesis requires the MSCs to form condensations. This process is controlled by factors such as transforming growth factor-β (TGF-β) and members of the HOX family.17 In addition, interactions of cells with the extracellular matrix are involved in the initiation of MSC differentiation.17 As they start to differentiate, MSCs give rise to osteochondroprogenitors, which are multipotent cells capable of generating either chondrocytes or OBs. The decision of lineage fate depends on the expression of runt-related transcription factor 2 (RUNX2) and SRY-Box9 (SOX9) by the osteochondroprogenitors. The SOX9 transcription factor is dominant over RUNX2 and triggers the activation of chondrocyte-specific genes,18 whereas RUNX2 determines OB vs chondrocyte differentiation, and its expression is modified by regulators that can cause its stabilization and upregulation, surpassing SOX9 dominance.19 Nevertheless, RUNX2 requires osterix (OSX or SP7), another key osteogenic transcription factor, to activate OB-specific genes and ensure full differentiation to OBs.20 

During their differentiation, OBs deposit bone matrix (osteoid) mainly composed by collagen type 1, and when the appropriate thickness is reached, some OBs get trapped and differentiate into osteocytes.21 Mineralization of the bone matrix is initiated at the stage of the late mature OBs and continues through the osteocyte level.21 Noncollagenous proteins produced by these cells regulate the mineralization process, which includes rapid mineral accumulation, carbonate substitution, collagen condensation, and water reduction.21 The formation of new bone, if not associated with normal mineralization, could lead to increased fragility.

Several signaling pathways are involved in bone development. In the canonical Wnt signaling pathway, translocation of β-catenin into the nucleus results in the transcription of specific genes that drive the osteoblastic precursor cells to become more mature OBs.22 Bone morphogenetic protein (BMP) signaling has various roles in the development and regulation of bone formation.23 Several subtypes of BMP (BMP-2, -6. -7, and -9) have been demonstrated to induce differentiation of MSCs into OBs, and BMP2 has been shown to upregulate the expression of RUNX2 level. BMPs normalize cartilage development as well; therefore, this signaling pathway is crucial for both intramembranous and endochondral osteogenesis.23 Similarly, fibroblast growth factors (FGFs), such as FGF-2, -3, -4, -9, and -18 are significant in chondrocyte and OB differentiation, as demonstrated by loss- or gain-of-function approaches.24 

Bone homeostasis also highly depends on the resorption processes, for which OCs are key. The receptor activator of the NF-κΒ ligand (RANKL) secreted by stromal cells or OBs binds to its receptor RANK on OC precursors, leading to OC differentiation. In contrast, osteoprotegerin (OPG), secreted by OBs, acts as a soluble decoy receptor that binds to RANKL and blocks the differentiation of OCs. Several hormones and cytokines regulate this process and are responsible for the final bone phenotype (reviewed in Zupan et al25).

Key factors affecting sex-dependent differences in bone health

Considering potential differences in the bone phenotypes observed in male and female patients with MPN and in mouse models, attention should be given to the effect of estrogen or/and calcium homeostasis. Estrogen regulates bone metabolism on multiple levels by inhibiting OB apoptosis, OC formation, and activation of bone remodeling, as well as by increasing OB lifespan and OC apoptosis.26 A decrease in estrogen level leads to augmented expression of macrophage colony-stimulating factor (M-CSF) and RANKL in OBs, and the production of interleukin-1 (IL-1), IL-6, and tumor necrosis factor-α (TNF-a) by T cells that enhance OC formation and function with a subsequent bone loss.26 This finding is noteworthy, because women may have osteopenia at the age when they are affected by MPN.

Calcium is important for healthy bone, and its homeostasis is regulated by parathyroid hormone (PTH) and 1,25-dihydroxy-vitamin D (1,25(OH)2D), which in turn are regulated by serum calcium levels.27 Calcium depletion is more common in menopausal women, whereas a decrease in serum calcium leads to increased PTH secretion by the parathyroid glands, causing increased calcium reabsorption in the kidneys, and augmented bone resorption for calcium release.27 In parallel, PTH triggers the secretion of 1,25(OH)2D from the kidneys, causing increased calcium absorption in the gut, increased bone resorption, and a subsequent decrease in PTH secretion.27 Calcium metabolism is interesting in the context of MPNs, because some patients have CALR mutations.10 Calcium is important for MK function,28 and normal CALR is involved in its homeostasis. Normal CALR regulates store-operated calcium entry28 by forming a complex with ER protein 57 and the endoplasmic reticulum calcium sensor stromal interaction molecule 1; however, a defective interaction of CALR mutants with these proteins cause the constitutive activation of store-operated calcium entry and an abnormal regulation of calcium.29 These alterations may affect calcium availability and, subsequently, bone homeostasis.

Reports on bone phenotypes in MPNs vary, depending on the type of MPN, sex, and method and system of detection. Accordingly, such reports are surveyed here based on studies using experimental models and human data pertinent to PMF vs PV or ET.

Bone phenotypes in PMF

Several investigations point to increased bone volume in PMF. In mouse models, expression of murine JAK2V617F under the H2Kb promoter led to the generation of transgenic mice that recapitulate PV, ET, or PMF. The mice with a PMF-like phenotype showed a slight increase in bone cortical thickness, with formation of new bony trabeculae, which was more pronounced in older mice.30 Similarly, other MK-driven MF mouse models assessed skeletal changes. GATA-1 and NF-E2 are transcription factors necessary for normal MK development, and mice deficient in those factors have an increased number of abnormal, immature MKs, with a striking increase in bone volume.31,32 Although, MF almost always precedes osteosclerosis in patients, the increased bone volume occurs much earlier than MF in these mice. Moreover, the increased TGF-β1 expression in the MKs of Gatalow-mutated mice may be of great interest, because its inhibition restores MK maturation and reduces osteogenesis in the mice.33 Based on these findings, it was suggested that the combination of JAK2 and TGF-β1 inhibitors could reduce disease manifestations. In these studies,30-33 sex-matched mice were used for analyses, but possible differences between sexes were not probed. Female mice with experimentally induced TPO overexpression display myeloproliferative syndrome accompanied by MF and osteosclerosis.34 Mice deficient in G6b-B, a receptor expressed on the surface of platelets and MKs, develop macrothrombocytopenia, MF, and osteosclerosis,35 with females displaying a stronger bone phenotype than males. MK/platelet-specific G6b-B-knockout animals demonstrated bone phenotypes similar to the global G6b-B–knockout mice.36 The role of the G6b-B gene in PMF was also supported by a publication, reporting that G6b-B loss-of-function mutations were found in a group of patients with PMF.37 Together, these studies suggest that the bone phenotype is attributable to the development of PMF, regardless of the gene inducing the pathology.

Similar to conclusions derived from mouse studies, several human studies showed that patients with PMF have increased bone volume, but comparison according to sex was not fully performed.38-45 In these later studies, bone volume was measured by various methods, as outlined in Table 1. In contrast, Farmer et al reported no significant differences in bone geometry, volumetric bone mineral density, and microstructure between patients with MF and controls, when using dual-energy X-ray absorptiometry (DXA) and high-resolution peripheral quantitative computed tomography.46 That BM specimens from these patients showing osteosclerosis led the researchers to assume that peripheral scanning measurements may underrate the actual new bone formation and the biomechanical properties.46 This result suggests that modes of bone assessment in PMF are indeed important, warranting further consideration.

Table 1.

Effect of different forms of MPN on human bone

ReferenceStudy purposePathology typePatients, nFindingMethod
Burkhardt et al, 198238  Assess bone biopsy specimens in hematological disorders. PV 232 Cohorts pooled for analyses.
Attenuation of the cancellous trabeculae in PV and ET.
Formation of irregular spicules of woven bone and progressive thickening of the trabeculae in osteomyelofibrosis.
Lytic osseous lesions in association with focal accumulations of megakaryoblasts and MKs in CML. 
Biopsy of the anterior or posterior iliac crests.
Histology 
ET (mentioned as MM) 52 
MF (fibrosis secondary to PV, CML) 100 
Osteomyelofibrosis (marrow fibrosis with trabecular thickening) 84 
CML 143 
Controls 158 
Coindre et al, 198439  Analyze sclerotic bone from 9 patients with IMF with histomorphometry. IMF (PMF) 9 (6M; 3F) Trabecular bone volume was higher in patients with IMF, compared with controls, and generally exceeded twice normal values. Female patients had higher trabecular volume than male patients. Iliac bone biopsy.
Histodynamic analysis on undecalcified bone. 
Controls 18 
Thiele et al, 198940  Elucidate osseous remodeling by assessment of trabecular bone area and number of OCs. CML 65 Cohorts were pooled for analysis.
No significant differences in the total area of trabecular bone and OC counts in CML, ET, and PV and initial stages of PMF.
In advanced stages of PMF, significant increase in total bone area is associated with a high count for all osteoclastic elements and OBs. 
Biopsy of posterior iliac crest.
Histomorphometry 
ET 25 
PV 25 
AMM (PMF) 50 
Control 20 (9M; 11F) 
Poulsen et al, 199841  Investigate degree of osteosclerosis and BM fibrosis in hematopoietic malignancies.
No controls; only patients. Comparison was made between the different MPNs. 
IMF (PMF) 20 (10M; 10F) Cohorts were pooled for analyses.
IMF showed greater BM fibrosis and total bone volume compared with all other MPNs
Fibrosis and osteosclerosis decreased gradually through IMF, CML, ET and PV.
Both the amount of woven bone and the size of osteons positively correlated with the degree of fibrosis. 
Jamshidi biopsies for histomorphometry. 
CML 10 (3M; 7F) 
ET 10 (3M; 7F) 
PV 5 (2M; 3F) 
Non-IMF 25 (8M; 17F) 
CMPD-NOS 2 (1M; 1F) 
Diamond et al, 200242  Analyze 4 males with IMF IMF (PMF) 4M Significant increases in osteoid thickness, trabecular thickness and cancellous bone volume.
Evidence of high bone turnover. 
Iliac crest bone biopsy for histomorphometry
Serum biochemistry
Markers of bone turnover
Lumbar spine and femoral neck DXA
Lumbar spine QCT 
Controls (based on control values from a previous study119113M 
Mellibovsky et al, 200443  Determine bone remodeling alterations in IMF. IMF (PMF) 9 (5M; 4F) Cohorts were pooled for analyses of the bone histomorphometric parameters.
Patients with IMF showed significantly higher bone volume (BV/TV); OB surface (Ob.S./BS); OC surface (Os.S/BS); OC number (n, OC/TA); mineralizing surface (MS/BS); and eroded surface (ES/BS). 
Transiliac bone biopsies
Static and dynamic histomorphometry 
Controls 40 (22M; 18F) 
Schmidt et al, 200744  Characterize cellular and tissular changes in bone from patients with MMM. MMM (same as PMF). Controls were age-matched from the French population with data available.120,121 13 (8M; 5F) No differences between M and F
Increased thickness of bone
units in some patients.
Bone formation rate increased
in 7 patients.
Tendency for decrease of OC number
Eroded surfaces increased in 10 patients. 
Transiliac bone biopsies
Histomorphometry
Micro-CT 
Fitter et al, 2008118  Examine the effect of long-term imatinib or α-interferon therapy on bone of patients with CML. CML 29 (20M; 9F) No differences between M and F.
Before treatment: Trabecular bone volume was within the normal range in 15 patients, below normal in 10 patients, and exceeded the normal range in 4 patients (based on control values from a previous study122). 
Posterior superior iliac spine trephine biopsies
Histomorphometry 
Teman et al, 201045  Test the utility of digital imaging as an objective quantification method for both BM fibrosis and osteosclerosis. PMF 34 (19M; 15F) Cohorts were pooled for analyses.
All categories of MPN, except newly diagnosed CML, had significantly higher trabecular volume compared with controls.
Trabecular volume was strikingly higher in PMF than in ET and PV.
PMF had higher trabecular volume than MF that was secondary to other MPNs.
There was no statistically significant relationship between JAK2 status and trabecular volume. 
Posterior superior iliac spine trephine biopsies.
Histomorphologic assessments of slides stained with hematoxylin and eosin and digitally scanned with a ScanScope XT system. 
ET 20 (9M; 11F) 
Post-ET MF 14 (6M; 8F) 
PV 14 (6M; 8F) 
Post-PV MF 7 (3M; 4F) 
CML 10 (6M; 4F) 
MPN-U (unclassifiable) 22 (12M; 10F) 
Controls 69 (41M; 28F) 
Farmer et al, 201546  Assess bone geometry, bone mineral density, and microarchitecture in patients with MF. MF (either primary MF or secondary to other MPN) 18 (9M; 9F) Cohorts were pooled for analyses.
No significant differences between MF patients and controls regarding bone mineral density, geometry, and microarchitecture, despite manifestations of osteosclerosis observed in bone marrow biopsies of the patients.
Bone stiffness and bone strength were similar between patients with MF and controls. 
Lumbar spine and nondominant total hip region DXA.
Nondominant distal radius and distal tibia HR-QCT.
Measurements of biochemical markers for assessment of bone turnover. 
Controls 18 (9M; 9F) 
ReferenceStudy purposePathology typePatients, nFindingMethod
Burkhardt et al, 198238  Assess bone biopsy specimens in hematological disorders. PV 232 Cohorts pooled for analyses.
Attenuation of the cancellous trabeculae in PV and ET.
Formation of irregular spicules of woven bone and progressive thickening of the trabeculae in osteomyelofibrosis.
Lytic osseous lesions in association with focal accumulations of megakaryoblasts and MKs in CML. 
Biopsy of the anterior or posterior iliac crests.
Histology 
ET (mentioned as MM) 52 
MF (fibrosis secondary to PV, CML) 100 
Osteomyelofibrosis (marrow fibrosis with trabecular thickening) 84 
CML 143 
Controls 158 
Coindre et al, 198439  Analyze sclerotic bone from 9 patients with IMF with histomorphometry. IMF (PMF) 9 (6M; 3F) Trabecular bone volume was higher in patients with IMF, compared with controls, and generally exceeded twice normal values. Female patients had higher trabecular volume than male patients. Iliac bone biopsy.
Histodynamic analysis on undecalcified bone. 
Controls 18 
Thiele et al, 198940  Elucidate osseous remodeling by assessment of trabecular bone area and number of OCs. CML 65 Cohorts were pooled for analysis.
No significant differences in the total area of trabecular bone and OC counts in CML, ET, and PV and initial stages of PMF.
In advanced stages of PMF, significant increase in total bone area is associated with a high count for all osteoclastic elements and OBs. 
Biopsy of posterior iliac crest.
Histomorphometry 
ET 25 
PV 25 
AMM (PMF) 50 
Control 20 (9M; 11F) 
Poulsen et al, 199841  Investigate degree of osteosclerosis and BM fibrosis in hematopoietic malignancies.
No controls; only patients. Comparison was made between the different MPNs. 
IMF (PMF) 20 (10M; 10F) Cohorts were pooled for analyses.
IMF showed greater BM fibrosis and total bone volume compared with all other MPNs
Fibrosis and osteosclerosis decreased gradually through IMF, CML, ET and PV.
Both the amount of woven bone and the size of osteons positively correlated with the degree of fibrosis. 
Jamshidi biopsies for histomorphometry. 
CML 10 (3M; 7F) 
ET 10 (3M; 7F) 
PV 5 (2M; 3F) 
Non-IMF 25 (8M; 17F) 
CMPD-NOS 2 (1M; 1F) 
Diamond et al, 200242  Analyze 4 males with IMF IMF (PMF) 4M Significant increases in osteoid thickness, trabecular thickness and cancellous bone volume.
Evidence of high bone turnover. 
Iliac crest bone biopsy for histomorphometry
Serum biochemistry
Markers of bone turnover
Lumbar spine and femoral neck DXA
Lumbar spine QCT 
Controls (based on control values from a previous study119113M 
Mellibovsky et al, 200443  Determine bone remodeling alterations in IMF. IMF (PMF) 9 (5M; 4F) Cohorts were pooled for analyses of the bone histomorphometric parameters.
Patients with IMF showed significantly higher bone volume (BV/TV); OB surface (Ob.S./BS); OC surface (Os.S/BS); OC number (n, OC/TA); mineralizing surface (MS/BS); and eroded surface (ES/BS). 
Transiliac bone biopsies
Static and dynamic histomorphometry 
Controls 40 (22M; 18F) 
Schmidt et al, 200744  Characterize cellular and tissular changes in bone from patients with MMM. MMM (same as PMF). Controls were age-matched from the French population with data available.120,121 13 (8M; 5F) No differences between M and F
Increased thickness of bone
units in some patients.
Bone formation rate increased
in 7 patients.
Tendency for decrease of OC number
Eroded surfaces increased in 10 patients. 
Transiliac bone biopsies
Histomorphometry
Micro-CT 
Fitter et al, 2008118  Examine the effect of long-term imatinib or α-interferon therapy on bone of patients with CML. CML 29 (20M; 9F) No differences between M and F.
Before treatment: Trabecular bone volume was within the normal range in 15 patients, below normal in 10 patients, and exceeded the normal range in 4 patients (based on control values from a previous study122). 
Posterior superior iliac spine trephine biopsies
Histomorphometry 
Teman et al, 201045  Test the utility of digital imaging as an objective quantification method for both BM fibrosis and osteosclerosis. PMF 34 (19M; 15F) Cohorts were pooled for analyses.
All categories of MPN, except newly diagnosed CML, had significantly higher trabecular volume compared with controls.
Trabecular volume was strikingly higher in PMF than in ET and PV.
PMF had higher trabecular volume than MF that was secondary to other MPNs.
There was no statistically significant relationship between JAK2 status and trabecular volume. 
Posterior superior iliac spine trephine biopsies.
Histomorphologic assessments of slides stained with hematoxylin and eosin and digitally scanned with a ScanScope XT system. 
ET 20 (9M; 11F) 
Post-ET MF 14 (6M; 8F) 
PV 14 (6M; 8F) 
Post-PV MF 7 (3M; 4F) 
CML 10 (6M; 4F) 
MPN-U (unclassifiable) 22 (12M; 10F) 
Controls 69 (41M; 28F) 
Farmer et al, 201546  Assess bone geometry, bone mineral density, and microarchitecture in patients with MF. MF (either primary MF or secondary to other MPN) 18 (9M; 9F) Cohorts were pooled for analyses.
No significant differences between MF patients and controls regarding bone mineral density, geometry, and microarchitecture, despite manifestations of osteosclerosis observed in bone marrow biopsies of the patients.
Bone stiffness and bone strength were similar between patients with MF and controls. 
Lumbar spine and nondominant total hip region DXA.
Nondominant distal radius and distal tibia HR-QCT.
Measurements of biochemical markers for assessment of bone turnover. 
Controls 18 (9M; 9F) 

Studies without M/F indication did not report according to sex.

AMM, agnogenic myeloid metaplasia; CMPD, chronic myeloproliferative disease; F, female; HR-pQCT, high-resolution-peripheral quantitative computed tomography; IMF, idiopathic MF; M, male; micro-CT, micro-computed tomography; MM, megakaryocytic myelosis; ΜΜΜ, myelofibrosis with myeloid metaplasia.

Bone phenotypes in PV and ET

The effect of PV and ET on bone is more obscure, and the literature shows that bone volume can be decreased, increased, or unchanged compared with controls, depending on the system examined. Mice receiving transplants of murine hematopoietic cells transduced with JAK2V617F displayed a human-PV phenotype that progressed to secondary MF with time.47 Histological examination of female mice in the early stages of the disease showed no difference in bone, compared with the controls, but increased bone cortical thickness and formation of newly bony trabeculae were present when the mice developed fibrosis secondary to PV, suggesting that development of MF is essential for a bone phenotype.47 On the other hand, a significant loss of trabecular bone was observed in a JAK2V617F-knockin mouse model that recapitulates PV.48 

Moreover, no definitive conclusion has been made in human studies. Similar to mice, patients with PV or ET either had attenuated cancellous bone38 or showed no significant changes in the total area of trabecular bone.40 One report stated that all categories of MPN, except newly diagnosed chronic myeloid leukemia (CML), had significantly higher trabecular volume than did the controls.45 A meta-analysis concluded that the effect of PV, ET, and CML on bone density is negative, but is statistically significant only in CML, whereas MF (primary or secondary) is characterized by an increase in bone density.49 Yet, there is an augmented risk of osteoporotic fractures in patients with ET, PV, or CML compared with the general population, but the mechanism remains to be elucidated.50 

Modes of bone measurement in patients with MPN

Table 1 summarizes the bone phenotypes in human studies, including modes of measurement in each case. Indeed, different technologies used to assess bone have their limitations, and this could contribute to the above-described discrepancies in the literature concerning the effects of MPN on bone volume. Histomorphometry, usually applied to histological sections of transiliac bone biopsy specimens, measures structural and remodeling bone parameters, providing information on bone mass, structure, demineralized bone, extent of resorption cavities, and bone formation rate. Nevertheless, it is a complex and invasive technique with the possibility of large measurement errors related to observer variations.51 DXA is a noninvasive method, often performed on the lower spine and hips, that measures areal bone mineral density from which the risk of fracture can be estimated. However, it does not analyze cortical and trabecular bone separately, and it can under- or overestimate real bone density.52 Both histomorphometry and DXA are 2-dimensional representations of a 3-dimensional structure. Quantitative computed tomography (QCT) provides a tridimensional study of the bone and measures the true volumetric bone mineral density.52 Moreover, QCT assesses the structure and geometry of the bone and analyzes the cortical and trabecular areas separately.52 High-resolution QCT can also quantify the trabecular and cortical architecture.52 Importantly, bone changes may not be the same at different anatomical sites, and the fact that the methods used for bone assessment do not always examine the same sample site could also be responsible for variation in study results.

MPN-induced secreted factors

Several cytokines and growth factors have been consistently identified as upregulated in patients with MPN.53 There is considerable evidence supporting a strong correlation between MF and increased TGF-β levels in patients with all 3 classic MPN forms.54,55 The role of TNF-α in MPNs is also well established,56,57 and it is associated with inferior overall survival in PV and PMF.58 In addition, increased platelet-derived growth factor (PDGF) levels have been reported in urine of patients with PMF, ET, or PV,59 and in circulating platelets and serum of patients with MPN and MF.60 Interestingly, PDGF is higher in ET than PV, and it is also higher in patients with ET who are positive for the JAK2V617F gene mutation than in those who are negative.61 Elevated levels of vascular endothelial growth factor (VEGF), IL-1β, IL-6, IL-8, and oncostatin M (OSM) were found in different forms of MPN.56-58,61-64 Upregulation of lysyl oxidase (LOX), a regulator of the extracellular matrix, was also observed in MKs of patients with MPN, especially in PMF, compared with controls.65,66 Finally, a very recent study highlighted the role of increased secretion of CXCL4/platelet factor 4 by MPN cells as a booster of fibrosis and a facilitator of upregulated inflammatory cytokines, such as TGF-β and TNF-α.67 This study also showed that hematopoietic stem cells carrying MPN-associated mutations alter the gene expression profile in mesenchymal stromal cells.67 The impact of this reprograming on OB differentiation is yet to be explored.

Factors causing bone loss

Of the several MPN-associated factors, some have been reported to lead to bone loss. IL-1β is a strong stimulator of bone resorption that exerts its function by increasing the expression of RANKL, through stimulation of OC development and activity, in stromal cells,68,69 and/or by having a direct effect on OCs.70 IL-1β also works in cooperation with TNF toward this effect.69 Furthermore, migration of OBs is inhibited in the presence of IL-1β.71 IL-8 participates in the altered MK growth in MF,62 while having an osteoclastogenic function as demonstrated either directly on the number of OCs or indirectly through upregulation of RANKL.72,73

Factors causing bone gain

LOX is elevated in both human and mouse JAK2V617F mutated MKs, the inhibition of which reduced MK adhesion to collagen66 and the fibrotic phenotype in mouse models of PMF.74 In contrast, LOX was shown to be necessary for normal OB proliferation and/or differentiation in studies using osteoblastic cell lines, primary mouse calvaria cells, and LOX-deficient mice.75-78 Excess TNF-α, also an MPN-associated factor, downregulates LOX expression and affects OB fate.76,79 Murine models treated with a LOX-inhibitor presented with altered collagen cross-linking patterns, reduced trabecular bone, and decreased cortical bone fracture toughness and strength.80,81

Factors causing bone gain and loss

Several of the released factors associated with MPNs exert a dual effect on bone, depending on concentration and timing, as well as possible effects on both OBs and OCs (Figure 1). For example, at early stages of MSC differentiation, TGF-β cooperates with BMP signaling to promote the osteoblastic lineage.82 Furthermore, it increases OB proliferation.83 On the other hand, TGF-β promotes OC differentiation at a low dose but inhibits the process at a higher dose, by modifying the RANKL/OPG ratio.84 PDGF (5 isoforms: PDGF-AA, PDGF-BB, PDGF-CC, PDGF-DD, PDGF-AB) has been considered to be a potential therapy in bone formation and healing of fractures.85 It can promote OB proliferation, acting as a mitogen for BM stromal cells, with a chemotactic effect on OB-like cells.86,87 Past studies concluded that PDGF is not a factor for OB differentiation and mineralization, even though it can promote their proliferation,88,89 whereas a recent study demonstrated that continuous exposure to PDGF-BB increases osteogenic differentiation from BM stromal cells .90 On the other hand, OC formation, osteoclastic activity, and OC precursor chemotaxis are enhanced by the presence of PDGF.91 Another factor, TNF-α, is known as a potent stimulator of bone resorption that stimulates osteoclastogenesis in a RANKL-dependent68,92 or -independent manner.93 In addition, TNF-α can both inhibit and promote osteoblastogenesis, and this contradictory effect can be explained based on the differentiation stage of the treated cells.94 VEGF has a role in both intramembranous95 and endochondral bone formation96 by coupling angiogenesis with osteogenesis, and it stimulates OB differentiation in an autocrine or paracrine manner.97 Moreover, it can control differentiation of MSCs by favoring osteoblastogenesis vs adipogenesis through an intracrine mechanism.98 On the other hand, VEGF can also increase bone resorption by acting on OCs and enhancing their recruitment, differentiation, and survival.99 Finally, both IL-6 and OSM have been shown to stimulate the function of OCs indirectly, by increasing the expression of RANKL and OPG, a protein with multiple effects, including on osteoporosis.100 In contrast, other publications have described IL-6 as a suppressor of OC differentiation, and OSM can promote bone formation by inhibiting sclerostin.101,102

Figure 1.

MPN MK-released factors affecting bone homeostasis. MK-released factors that enhance OB and/or OC development are depicted. Although not shown here, as noted in the text, depending on concentration, some factors can both enhance or inhibit the development of the same lineage, as in the case of the effect of TNF-α on OBs or TGF-β and IL-6 on OCs. Other factors have only a negative effect on function, such as the influence of IL-1β on OBs. Professional illustration by Patrick Lane, ScEYEnce Studios.

Figure 1.

MPN MK-released factors affecting bone homeostasis. MK-released factors that enhance OB and/or OC development are depicted. Although not shown here, as noted in the text, depending on concentration, some factors can both enhance or inhibit the development of the same lineage, as in the case of the effect of TNF-α on OBs or TGF-β and IL-6 on OCs. Other factors have only a negative effect on function, such as the influence of IL-1β on OBs. Professional illustration by Patrick Lane, ScEYEnce Studios.

Close modal

MKs as direct effectors of bone development

The BM consists of at least 2 major parts: the vascular niche that contains most sinusoids and arterioles and the endosteal niche that includes OBs, OCs, and a specific osteoblastic subpopulation.103 MKs are mainly located in the vascular niche; however, they have been associated with the endosteum, as well.103,104 Thus, MK localization in the BM allows for these cells to potentially affect bone homeostasis through direct interaction with MSCs and/or through released factors (Figure 1). In addition, the atypical localization of MKs at the paratrabecular area and endosteal border seen in myelodysplastic and myeloproliferative bone marrow (Figure 2) could indicate that this altered distribution influences bone in these pathologies.105-107 Accordingly, coculturing wild-type (WT) OBs with MKs derived from mice deficient in GATA-1 or NFE-2 transcription factors significantly enhanced OB proliferation, in accordance with a dramatic increase in trabecular bone volume observed in these mice.31 Interestingly, the use of conditioned medium from cultures of these gene-deleted MKs, not only failed to enhance OB proliferation, but caused a dose-dependent inhibition of this process, indicating that direct contact is necessary for the enhancement.31 A different study explored the effect of mechanical stresses on these processes, finding that the osteoblastic differentiation of MC3T3-E1 cells was decreased in the presence of coculture with the megakaryocytic cell line Meg-01 when exposed to fluid shear, as compared with cultures in a static condition.108 

Figure 2.

Atypical localization of MPN MKs close to bone areas. (A) A general structure of a long bone. (B) An enlarged area showing the cortical and trabecular bone. (C) An enlarged area depicting the organization of the bone marrow with its zones and the distribution of cells in healthy steady state. (D) An enlarged area depicting the organization of the bone marrow with its zones and the distribution of cells in MPNs. MKs are shown in red. The paratrabecular zone shown in panels C and D is adjacent to trabecular bone and normally has mostly myeloid precursor cells. The intermediate zone usually includes erythroid colonies and maturing myeloid cells. The central zone contains erythroid cells, maturing myeloid cells, sinusoids, and MKs. However, as cited in the text, MPNs are characterized by an increased number of MKs that tend to atypically localize closer to the paratrabecular zone and the endosteal border.

Figure 2.

Atypical localization of MPN MKs close to bone areas. (A) A general structure of a long bone. (B) An enlarged area showing the cortical and trabecular bone. (C) An enlarged area depicting the organization of the bone marrow with its zones and the distribution of cells in healthy steady state. (D) An enlarged area depicting the organization of the bone marrow with its zones and the distribution of cells in MPNs. MKs are shown in red. The paratrabecular zone shown in panels C and D is adjacent to trabecular bone and normally has mostly myeloid precursor cells. The intermediate zone usually includes erythroid colonies and maturing myeloid cells. The central zone contains erythroid cells, maturing myeloid cells, sinusoids, and MKs. However, as cited in the text, MPNs are characterized by an increased number of MKs that tend to atypically localize closer to the paratrabecular zone and the endosteal border.

Close modal

Whether MKs exert any effect on OCs was also examined.109 To this end, OCs from spleen or BM of WT mice were cocultured with MKs that were generated from fetal liver of WT or GATA-1–deficient mice. Both types of MKs blocked OC development, and MK-conditioned medium completely inhibited OC biogenesis, suggesting a role for a yet unidentified soluble factor. In line with the mouse data, human MKs derived from normal donors were tested for an effect on the synthesis of type 1 collagen, OPG, and RANKL in human OBs, finding a decreased expression of RANKL and an increased expression of OPG and type 1 collagen, which is indicative of enhanced OB differentiation.110 Human MKs also inhibit the development and the activity of OCs, and this inhibition is stronger when the MKs act on OC precursors, rather than on more mature OCs.111 It has been suggested that both cell-to-cell contact and MK-soluble factors are responsible for this result. No studies are available on the effect of human MPN MKs, or mouse JAK2V617F, or other MPN-mutated MKs on OB or OC differentiation.

Mechanisms of effect of MKs on osteogenesis

As to mechanisms by which MKs affect OBs, several pathways were explored. Because gap junction intercellular communication (GJIC) is necessary for cell proliferation and differentiation, its role in promoting MK-OB interaction was tested and confirmed by using two gap junction uncouplers.112 However, an unexpected observation was that OB proliferation was enhanced after inhibition of GJIC in the presence of MKs. OB differentiation was decreased when MKs were cocultured with OBs for extended periods, and blocking GJIC did not alter this MK-mediated reduction. Subsequent studies pointed to integrins α3β1 and α5β1 and glycoprotein IIb as essential for MK-OB interaction.113 Signal transduction pathways that are implicated in MK-induced OB proliferation were identified as Pyk2 signaling cascades and the p38/MAPKAPK2/p90RSK kinase cascade.114,115 Interestingly, the capacity of MKs to enhance OB proliferation weakens as the MKs age.116 In contrast, the number of MKs, per se, affected the phenotype observed.117 An increasing number of MKs in coculture experiments further increased OB proliferation and decreased OC formation, regardless of whether WT or mutant MKs were used. However, different maturation stages of MKs did not alter the aforementioned effects on OBs and OCs.117 The sex of the mice used in the cited publications was not specified.

Conclusions and future directions

There could be several reasons for the variability in reporting the effects of MPNs on patients’ bone health, with a major reason being the evolving methods of bone assessment. In addition, continuous updates in the classification system may result in misclassification of patients, topped by difficulty in recruiting the most appropriate controls. Usually, bone examination was conducted once, after diagnosis, whereas serial evaluations of the patient would be more informative for a correlation between disease progression and effect on bone. In some cases, patients may have taken medication that affects the bone, and in most studies, there is no comparison between men and women that could explain possible sex hormone effects. Larger studies that compare men with women and use individuals as their own controls via progressive bone density measurements are needed to better understand the bone phenotype in human MPNs. Future investigations could also include probing genes related to MF development for their effect on bone, such as NBEAL2, the mutations of which are present in patients with gray platelet syndrome who present with MF. Additional mouse studies could shed light on mechanisms by which MKs in different forms of MPN affect bone homeostasis directly or via secreted factors. Finally studying these processes in the context of bone regeneration after fracture, together could lead to developing means to ameliorate this side effect of the disease.

The authors thank Andrew Piasecki for help in editing the manuscript and Shinobu Matsuura for insights on some aspects of the review.

K.R. is an established investigator with the American Heart Association and is funded by National Institutes of Health, National Heart, Lung, and Blood Institute grant R01HL136363.

Contribution: A.K. searched the literature and wrote the first draft of the review, and K.R. added to the search and the content and finalized the review article, together with A.K.

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

Correspondence: Katya Ravid, Boston University School of Medicine, 700 Albany St, W-601, Boston, MA 02118; e-mail: kravid@bu.edu.

1.
Arber
DA
,
Orazi
A
,
Hasserjian
R
, et al
.
The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia
.
Blood.
2016
;
127
(
20
):
2391
-
2405
.
2.
Titmarsh
GJ
,
Duncombe
AS
,
McMullin
MF
, et al
.
How common are myeloproliferative neoplasms? A systematic review and meta-analysis
.
Am J Hematol.
2014
;
89
(
6
):
581
-
587
.
3.
Szuber
N
,
Mudireddy
M
,
Nicolosi
M
, et al
.
3023 Mayo Clinic patients with myeloproliferative neoplasms:risk-stratified comparison of survival and outcomes data among disease subgroups
.
Mayo Clinic Proceed.
2019
;
94
(
4
):
599
-
610
.
4.
Tefferi
A
,
Barbui
T
.
Polycythemia vera and essential thrombocythemia:2021 update on diagnosis, risk-stratification and management
.
Am J Hematol.
2020
;
95
(
12
):
1599
-
1613
.
5.
Baxter
EJ
,
Scott
LM
,
Campbell
PJ
,
S
et al
.
Acquired mutation of the tyrosine kinase JAK2 in human myeloproliferative disorders
.
Lancet.
2005
;
365
(
9464
):
1054
-
1061
.
6.
Tefferi
A
.
Primary myelofibrosis:2021 update on diagnosis, risk-stratification and management
.
Am J Hematol.
2021
;
96
(
1
):
145
-
162
.
7.
Scott
LM
,
Tong
W
,
Levine
RL
, et al
.
JAK2 exon 12 mutations in polycythemia vera and idiopathic erythrocytosis
.
N Engl J Med.
2007
;
356
(
5
):
459
-
468
.
8.
Pikman
Y
,
Lee
BH
,
Mercher
T
, et al
.
MPLW515L is a novel somatic activating mutation in myelofibrosis with myeloid metaplasia
.
PLoS Med.
2006
;
3
(
7
):
e270
.
9.
Rumi
E
,
Pietra
D
,
Guglielmelli
P
, et al
.
Acquired copy-neutral loss of heterozygosity of chromosome 1p as a molecular event associated with marrow fibrosis in MPL-mutated myeloproliferative neoplasms
.
Blood.
2013
;
121
(
21
):
4388
-
4395
.
10.
Klampfl
T
,
Gisslinger
H
,
Harutyunyan
AS
, et al
.
Somatic mutations of calreticulin in myeloproliferative neoplasms
.
N Engl J Med.
2013
;
369
(
25
):
2379
-
2390
.
11.
Marneth
AE
,
Mullally
A
.
The molecular genetics of myeloproliferative neoplasms
.
Cold Spring Harb Prospect Med.
2020
;
10
(
2
):
a034876
.
12.
Delic
S
,
Rose
D
,
Kern
W
, et al
.
Application of an NGS-based 28-gene panel in myeloproliferative neoplasms reveals distinct mutation patterns in essential thrombocythaemia, primary myelofibrosis and polycythaemia vera
.
Br J Haematol.
2016
;
175
(
3
):
419
-
426
.
13.
Grinfeld
J
,
Nangalia
J
,
Baxter
EJ
, et al
.
Classification and personalized prognosis in myeloproliferative neoplasms
.
N Engl J Med.
2018
;
379
(
15
):
1416
-
1430
.
14.
Williams
N
,
Lee
J
,
Moore
L
, et al
.
Phylogenetic reconstruction of myeloproliferative neoplasm reveals very early origins and lifelong evolution
.
BioRxiv
2020
:doi:10.1101/2020.11.09.374710.
15.
Van Egeren
D
,
Escabi
J
,
Nguyen
M
, et al
.
Reconstructing the lineage histories and differentiation trajectories of individual cancer cells in myeloproliferative neoplasms
.
Cell Stem Cell.
2021
;
28
(
3
):
514
-
523.e519
.
16.
Carroll
SH
,
Ravid
K
.
Differentiation of mesenchymal stem cells to osteoblasts and chondrocytes: a focus on adenosine receptors [online publication]
.
Expert Rev Mol Med.
2013
;
15
:14 February 2013.
17.
Hall
BK
,
Miyake
T
.
All for one and one for all: condensations and the initiation of skeletal development
.
BioEssays.
2000
;
22
(
2
):
138
-
147
.
18.
Zhou
G
,
Zheng
Q
,
Engin
F
, et al
.
Dominance of SOX9 function over RUNX2 during skeletogenesis
.
Proc Natl Acad Sci USA.
2006
;
103
(
50
):
19004
-
19009
.
19.
Komori
T
,
Yagi
H
,
Nomura
S
, et al
.
Targeted disruption of Cbfa1 results in a complete lack of bone formation owing to maturational arrest of osteoblasts
.
Cell.
1997
;
89
(
5
):
755
-
764
.
20.
Nakashima
K
,
Zhou
X
,
Kunkel
G
, et al
.
The novel zinc finger-containing transcription factor osterix is required for osteoblast differentiation and bone formation
.
Cell.
2002
;
108
(
1
):
17
-
29
.
21.
Blank
M
,
Sims
NA
.
Cellular processes by which osteoblasts and osteocytes control bone mineral deposition and maturation revealed by stage-specific EphrinB2 knockdown
.
Curr Osteoporos Rep.
2019
;
17
(
5
):
270
-
280
.
22.
Kobayashi
Y
,
Uehara
S
,
Udagawa
N
,
Takahashi
N
.
Regulation of bone metabolism by Wnt signals
.
J Biochem.
2016
;
159
(
4
):
387
-
392
.
23.
Beederman
M
,
Lamplot
JD
,
Nan
G
, et al
.
BMP signaling in mesenchymal stem cell differentiation and bone formation
.
J Biomed Sci Eng.
2013
;
6
(
08 8A
):
32
-
52
.
24.
Takei
Y
,
Minamizaki
T
,
Yoshiko
Y
.
Functional diversity of fibroblast growth factors in bone formation
.
Int J Endocrinol.
2015
;
2015
:
729352
.
25.
Zupan
J
,
Jeras
M
,
Marc
J
.
Osteoimmunology and the influence of pro-inflammatory cytokines on osteoclasts
.
Biochemia Med.
2013
;
23
(
1
):
43
-
63
.
26.
Khosla
S
,
Oursler
MJ
,
Monroe
DG
.
Estrogen and the skeleton
.
Trends Endocrinol Metab.
2012
;
23
(
11
):
576
-
581
.
27.
Peacock
M
.
Calcium metabolism in health and disease
.
Clin J Am Soc Nephrol.
2010
;
5
(
5 suppl 1
):
S23
-
S30
.
28.
Di Buduo
CA
,
Moccia
F
,
Battiston
M
, et al
.
The importance of calcium in the regulation of megakaryocyte function
.
Haematologica.
2014
;
99
(
4
):
769
-
778
.
29.
Di Buduo
CA
,
Abbonante
V
,
Marty
C
, et al
.
Defective interaction of mutant calreticulin and SOCE in megakaryocytes from patients with myeloproliferative neoplasms
.
Blood..
2020
;
135
(
2
):
133
-
144
.
30.
Shide
K
,
Shimoda
HK
,
Kumano
T
, et al
.
Development of ET, primary myelofibrosis and PV in mice expressing JAK2 V617F
.
Leukemia.
2008
;
22
(
1
):
87
-
95
.
31.
Kacena
MA
,
Shivdasani
RA
,
Wilson
K
, et al
.
Megakaryocyte-osteoblast interaction revealed in mice deficient in transcription factors GATA-1 and NF-E2
.
J Bone Miner Res.
2004
;
19
(
4
):
652
-
660
.
32.
Kacena
MA
,
Gundberg
CM
,
Nelson
T
,
Horowitz
MC
.
Loss of the transcription factor p45 NF-E2 results in a developmental arrest of megakaryocyte differentiation and the onset of a high bone mass phenotype
.
Bone.
2005
;
36
(
2
):
215
-
223
.
33.
Zingariello
M
,
Martelli
F
,
Ciaffoni
F
, et al
.
Characterization of the TGF-β1 signaling abnormalities in the Gata1low mouse model of myelofibrosis
.
Blood.
2013
;
121
(
17
):
3345
-
3363
.
34.
Villeval
JL
,
Cohen-Solal
K
,
Tulliez
M
, et al
.
High thrombopoietin production by hematopoietic cells induces a fatal myeloproliferative syndrome in mice
.
Blood.
1997
;
90
(
11
):
4369
-
4383
.
35.
Stavnichuk
M
,
Nagy
Z
,
Senis
YA
,
Komarova
SV
.
Megakaryocyte-driven myelofibrosis leads to progressive osteosclerosis in G6b-B knockout mice
.
Blood.
2019
;
134
(
suppl_1
). Abstract
4199
.
36.
Stavnichuk
M
,
Tauer
JT
,
Nagy
Z
, et al
.
Severity of megakaryocyte-driven osteosclerosis in Mpig6b-deficient mice is sex-linked
.
J Bone Miner Res.
2021
;
36
(
4
):
803
-
813
.
37.
Hofmann
I
,
Geer
MJ
,
Vögtle
T
, et al
.
Congenital macrothrombocytopenia with focal myelofibrosis due to mutations in human G6b-B is rescued in humanized mice
.
Blood.
2018
;
132
(
13
):
1399
-
1412
.
38.
Burkhardt
R
,
Frisch
B
,
Bartl
R
.
Bone biopsy in haematological disorders
.
J Clin Pathol.
1982
;
35
(
3
):
257
-
284
.
39.
Coindre
JM
,
Reiffers
J
,
Goussot
JF
,
De Mascarel
A
,
Broustet
A
.
Histomorphometric analysis of sclerotic bone from idiopathic myeloid metaplasia (nine cases)
.
J Pathol.
1984
;
144
(
3
):
163
-
169
.
40.
Thiele
J
,
Hoeppner
B
,
Wienhold
S
,
Schneider
G
,
Fischer
R
,
Zankovich
R
.
Osteoclasts and bone remodeling in chronic myeloproliferative disorders. A histochemical and morphometric study on trephine biopsies in 165 patients
.
Pathol Res Pract.
1989
;
184
(
6
):
591
-
599
.
41.
Poulsen
LW
,
Melsen
F
,
Bendix
K
.
A histomorphometric study of haematological disorders with respect to marrow fibrosis and osteosclerosis
.
APMIS.
1998
;
106
(
4
):
495
-
499
.
42.
Diamond
T
,
Smith
A
,
Schnier
R
,
Manoharan
A
.
Syndrome of myelofibrosis and osteosclerosis:a series of case reports and review of the literature
.
Bone.
2002
;
30
(
3
):
498
-
501
.
43.
Mellibovsky
L
,
Marinoso
ML
,
Cervantes
F
, et al
.
Relationship among densitometry, bone histomorphometry, and histologic stage in idiopathic myelofibrosis
.
Bone.
2004
;
34
(
2
):
330
-
335
.
44.
Schmidt
A
,
Blanchet
O
,
Dib
M
,
Basle
MF
,
Ifrah
N
,
Chappard
D
.
Bone changes in myelofibrosis with myeloid metaplasia: a histomorphometric and microcomputed tomographic study
.
Eur J Haematol.
2007
;
78
(
6
):
500
-
509
.
45.
Teman
CJ
,
Wilson
AR
,
Perkins
SL
,
Hickman
K
,
Prchal
JT
,
Salama
ME
.
Quantification of fibrosis and osteosclerosis in myeloproliferative neoplasms: a computer-assisted image study
.
Leukemia Res.
2010
;
34
(
7
):
871
-
876
.
46.
Farmer
S
,
Vestergaard
H
,
Hansen
S
, et al
.
Bone geometry, bone mineral density, and micro-architecture in patients with myelofibrosis: a cross-sectional study using DXA, HR-pQCT, and bone turnover markers [published correction appears in Int J Hemtol. 102:388]
.
Int J Hematol.
2015
;
102
(
1
):
67
-
75
.
47.
Lacout
C
,
Pisani
DF
,
Tulliez
M
,
Gachelin
FM
,
Vainchenker
W
,
Villeval
JL
.
JAK2V617F expression in murine hematopoietic cells leads to MPD mimicking human PV with secondary myelofibrosis
.
Blood.
2006
;
108
(
5
):
1652
-
1660
.
48.
Oikonomidou
PR
,
Casu
C
,
Yang
Z
, et al
.
Polycythemia is associated with bone loss and reduced osteoblast activity in mice
.
Osteoporosis Int.
2016
;
27
(
4
):
1559
-
1568
.
49.
Steer
K
,
Stavnichuk
M
,
Morris
M
,
Komarova
SV
.
Bone health in patients with hematopoietic disorders of bone marrow origin: systematic review and meta-analysis
.
J Bone Miner Res.
2017
;
32
(
4
):
731
-
742
.
50.
Farmer
S
,
Horváth-Puhó
E
,
Vestergaard
H
,
Hermann
AP
,
Frederiksen
H
.
Chronic myeloproliferative neoplasms and risk of osteoporotic fractures: a nationwide population-based cohort study
.
Br J Haematol.
2013
;
163
(
5
):
603
-
610
.
51.
Kulak
CA
,
Dempster
DW
.
Bone histomorphometry: a concise review for endocrinologists and clinicians
.
Arq Bras Endocrinol Metab.
2010
;
54
(
2
):
87
-
98
.
52.
Choksi
P
,
Jepsen
KJ
,
Clines
GA
.
The challenges of diagnosing osteoporosis and the limitations of currently available tools
.
Clin Diabetes Endocrinol.
2018
;
4
:
12
.
53.
Wang
Y
,
Zuo
X
.
Cytokines frequently implicated in myeloproliferative neoplasms
.
Cytokine X.
2019
;
1
(
1
):
100005
.
54.
Ponce
CC
,
de Lourdes
FCM
,
Ihara
SS
,
Silva
MR
.
The relationship of the active and latent forms of TGF-β1 with marrow fibrosis in essential thrombocythemia and primary myelofibrosis
.
Med Oncol.
2012
;
29
(
4
):
2337
-
2344
.
55.
Nakayama
S
,
Yokote
T
,
Hiraoka
N
, et al
.
Transforming growth factor β- and interleukin 13-producing mast cells are associated with fibrosis in bone marrow
.
Hum Pathol.
2017
;
62
(
62
):
180
-
186
.
56.
Cacemiro
MDC
,
Cominal
JG
,
Tognon
R
, et al
.
Philadelphia-negative myeloproliferative neoplasms as disorders marked by cytokine modulation [published correction appears in Hematol Transfus Cell Ther. 2021;43(1):117]
.
Hematol Transfus Cell Ther.
2018
;
40
(
2
):
120
-
131
.
57.
Tefferi
A
,
Vaidya
R
,
Caramazza
D
,
Finke
C
,
Lasho
T
,
Pardanani
A
.
Circulating interleukin (IL)-8, IL-2R, IL-12, and IL-15 levels are independently prognostic in primary myelofibrosis:a comprehensive cytokine profiling study
.
J Clin Oncol.
2011
;
29
(
10
):
1356
-
1363
.
58.
Vaidya
R
,
Gangat
N
,
Jimma
T
, et al
.
Plasma cytokines in polycythemia vera:phenotypic correlates, prognostic relevance, and comparison with myelofibrosis
.
Am J Hematol.
2012
;
87
(
11
):
1003
-
1005
.
59.
Gersuk
GM
,
Carmel
R
,
Pattengale
PK
.
Platelet-derived growth factor concentrations in platelet-poor plasma and urine from patients with myeloproliferative disorders
.
Blood.
1989
;
74
(
7
):
2330
-
2334
.
60.
Martyré
MC
,
Magdelenat
H
,
Bryckaert
MC
,
Laine-Bidron
C
,
Calvo
F
.
Increased intraplatelet levels of platelet-derived growth factor and transforming growth factor-beta in patients with myelofibrosis with myeloid metaplasia
.
Br J Haematol.
1991
;
77
(
1
):
80
-
86
.
61.
Pourcelot
E
,
Trocme
C
,
Mondet
J
,
Bailly
S
,
Toussaint
B
,
Mossuz
P
.
Cytokine profiles in polycythemia vera and essential thrombocythemia patients:clinical implications
.
Exp Hematol.
2014
;
42
(
5
):
360
-
368
.
62.
Emadi
S
,
Clay
D
,
Desterke
C
, et al
.
IL-8 and its CXCR1 and CXCR2 receptors participate in the control of megakaryocytic proliferation, differentiation, and ploidy in myeloid metaplasia with myelofibrosis
.
Blood.
2005
;
105
(
2
):
464
-473.
63.
Panteli
K
,
Bai
M
,
Hatzimichael
E
,
Zagorianakou
N
,
Agnantis
NJ
,
Bourantas
K
.
Serum levels, and bone marrow immunohistochemical expression of, vascular endothelial growth factor in patients with chronic myeloproliferative diseases
.
Hematology.
2007
;
12
(
6
):
481
-
486
.
64.
Lekovic
D
,
Gotic
M
,
Skoda
R
, et al
.
Bone marrow microvessel density and plasma angiogenic factors in myeloproliferative neoplasms:clinicopathological and molecular correlations
.
Ann Hematol.
2017
;
96
(
3
):
393
-
404
.
65.
Tadmor
T
,
Bejar
J
,
Attias
D
, et al
.
The expression of lysyl-oxidase gene family members in myeloproliferative neoplasms
.
Am J Hematol.
2013
;
88
(
5
):
355
-
358
.
66.
Abbonante
V
,
Chitalia
V
,
Rosti
V
, et al
.
Upregulation of lysyl oxidase and adhesion to collagen of human megakaryocytes and platelets in primary myelofibrosis
.
Blood.
2017
;
130
(
6
):
829
-
831
.
67.
Gleitz
HFE
,
Dugourd
AJF
,
Leimkühler
NB
, et al
.
Increased CXCL4 expression in hematopoietic cells links inflammation and progression of bone marrow fibrosis in MPN
.
Blood.
2020
;
136
(
18
):
2051
-
2064
.
68.
Quinn
JM
,
Horwood
NJ
,
Elliott
J
,
Gillespie
MT
,
Martin
TJ
.
Fibroblastic stromal cells express receptor activator of NF-kappa B ligand and support osteoclast differentiation
.
J Bone Miner Res.
2000
;
15
(
8
):
1459
-
1466
.
69.
Wei
S
,
Kitaura
H
,
Zhou
P
,
Ross
FP
,
Teitelbaum
SL
.
IL-1 mediates TNF-induced osteoclastogenesis
.
J Clin Invest.
2005
;
115
(
2
):
282
-
290
.
70.
Jimi
E
,
Nakamura
I
,
Duong
LT
, et al
.
Interleukin 1 induces multinucleation and bone-resorbing activity of osteoclasts in the absence of osteoblasts/stromal cells
.
Exp Cell Res.
1999
;
247
(
1
):
84
-
93
.
71.
Hengartner
N-E
,
Fiedler
J
,
Ignatius
A
,
Brenner
RE
.
IL-1β inhibits human osteoblast migration
.
Mol Med.
2013
;
19
(
1
):
36
-
42
.
72.
Bendre
MS
,
Margulies
AG
,
Walser
B
, et al
.
Tumor-derived interleukin-8 stimulates osteolysis independent of the receptor activator of nuclear factor-kappaB ligand pathway
.
Cancer Res.
2005
;
65
(
23
):
11001
-
11009
.
73.
Bendre
MS
,
Montague
DC
,
Peery
T
,
Akel
NS
,
Gaddy
D
,
Suva
LJ
.
Interleukin-8 stimulation of osteoclastogenesis and bone resorption is a mechanism for the increased osteolysis of metastatic bone disease
.
Bone.
2003
;
33
(
1
):
28
-
37
.
74.
Leiva
O
,
Ng
SK
,
Matsuura
S
, et al
.
Novel lysyl oxidase inhibitors attenuate hallmarks of primary myelofibrosis in mice
.
Int J Hematol.
2019
;
110
(
6
):
699
-
708
.
75.
Turecek
C
,
Fratzl-Zelman
N
,
Rumpler
M
, et al
.
Collagen cross-linking influences osteoblastic differentiation
.
Calcif Tissue Int.
2008
;
82
(
5
):
392
-
400
.
76.
Khosravi
R
,
Sodek
KL
,
Xu
WP
, et al
.
A novel function for lysyl oxidase in pluripotent mesenchymal cell proliferation and relevance to inflammation-associated osteopenia
.
PloS One.
2014
;
9
(
6
):
e100669
.
77.
Sharma-Bhandari
A
,
Park
SH
,
Kim
JY
,
Oh
J
,
Kim
Y
.
Lysyl oxidase modulates the osteoblast differentiation of primary mouse calvaria cells
.
Int J Mol Med.
2015
;
36
(
6
):
1664
-
1670
.
78.
Pischon
N
,
Mäki
JM
,
Weisshaupt
P
, et al
.
Lysyl oxidase (lox) gene deficiency affects osteoblastic phenotype
.
Calcif Tissue Int.
2009
;
85
(
2
):
119
-
126
.
79.
Pischon
N
,
Darbois
LM
,
Palamakumbura
AH
,
Kessler
E
,
Trackman
PC
.
Regulation of collagen deposition and lysyl oxidase by tumor necrosis factor-alpha in osteoblasts
.
J Biol Chem.
2004
;
279
(
29
):
30060
-
30065
.
80.
Paschalis
EP
,
Tatakis
DN
,
Robins
S
, et al
.
Lathyrism-induced alterations in collagen cross-links influence the mechanical properties of bone material without affecting the mineral
.
Bone.
2011
;
49
(
6
):
1232
-
1241
.
81.
McNerny
EM
,
Gong
B
,
Morris
MD
,
Kohn
DH
.
Bone fracture toughness and strength correlate with collagen cross-link maturity in a dose-controlled lathyrism mouse model
.
J Bone Miner Res.
2015
;
30
(
3
):
455
-
464
.
82.
Matsunobu
T
,
Torigoe
K
,
Ishikawa
M
, et al
.
Critical roles of the TGF-beta type I receptor ALK5 in perichondrial formation and function, cartilage integrity, and osteoblast differentiation during growth plate development
.
Dev Biol.
2009
;
332
(
2
):
325
-
338
.
83.
Kassem
M
,
Kveiborg
M
,
Eriksen
EF
.
Production and action of transforming growth factor-beta in human osteoblast cultures:dependence on cell differentiation and modulation by calcitriol
.
Eur J Clin Invest.
2000
;
30
(
5
):
429
-
437
.
84.
Karst
M
,
Gorny
G
,
Galvin
RJ
,
Oursler
MJ
.
Roles of stromal cell RANKL, OPG, and M-CSF expression in biphasic TGF-beta regulation of osteoclast differentiation
.
J Cell Physiol.
2004
;
200
(
1
):
99
-
106
.
85.
Graham
S
,
Leonidou
A
,
Lester
M
,
Heliotis
M
,
Mantalaris
A
,
Tsiridis
E
.
Investigating the role of PDGF as a potential drug therapy in bone formation and fracture healing
.
Expert Opin Investig Drugs.
2009
;
18
(
11
):
1633
-
1654
.
86.
Tanaka
H
,
Liang
CT
.
Effect of platelet-derived growth factor on DNA synthesis and gene expression in bone marrow stromal cells derived from adult and old rats
.
J Cell Physiol.
1995
;
164
(
2
):
367
-
375
.
87.
Tsukamoto
T
,
Matsui
T
,
Fukase
M
,
Fujita
T
.
Platelet-derived growth factor B chain homodimer enhances chemotaxis and DNA synthesis in normal osteoblast-like cells (MC3T3-E1)
.
Biochem Biophys Res Commun.
1991
;
175
(
3
):
745
-751.
88.
Cassiede
P
,
Dennis
JE
,
Ma
F
,
Caplan
AI
.
Osteochondrogenic potential of marrow mesenchymal progenitor cells exposed to TGF-beta 1 or PDGF-BB as assayed in vivo and in vitro
.
J Bone Miner Res.
1996
;
11
(
7
):
1264
-
1273
.
89.
Gruber
R
,
Karreth
F
,
Kandler
B
, et al
.
Platelet-released supernatants increase migration and proliferation, and decrease osteogenic differentiation of bone marrow-derived mesenchymal progenitor cells under in vitro conditions
.
Platelets.
2004
;
15
(
1
):
29
-
35
.
90.
Zhang
M
,
Yu
W
,
Niibe
K
, et al
.
The effects of platelet-derived growth factor-BB on bone marrow stromal cell-mediated vascularized bone regeneration
.
Stem Cells Int.
2018
;
2018
:
3272098
.
91.
Li
DQ
,
Wan
QL
,
Pathak
JL
,
Li
ZB
.
Platelet-derived growth factor BB enhances osteoclast formation and osteoclast precursor cell chemotaxis
.
J Bone Miner Metab.
2017
;
35
(
4
):
355
-
365
.
92.
Lam
J
,
Takeshita
S
,
Barker
JE
,
Kanagawa
O
,
Ross
FP
,
Teitelbaum
SL
.
TNF-alpha induces osteoclastogenesis by direct stimulation of macrophages exposed to permissive levels of RANK ligand
.
J Clin Invest.
2000
;
106
(
12
):
1481
-
1488
.
93.
Kudo
O
,
Fujikawa
Y
,
Itonaga
I
,
Sabokbar
A
,
Torisu
T
,
Athanasou
NA
.
Proinflammatory cytokine (TNFalpha/IL-1alpha) induction of human osteoclast formation
.
J Pathol.
2002
;
198
(
2
):
220
-
227
.
94.
Hess
K
,
Ushmorov
A
,
Fiedler
J
,
Brenner
RE
,
Wirth
T
.
TNFalpha promotes osteogenic differentiation of human mesenchymal stem cells by triggering the NF-kappaB signaling pathway
.
Bone.
2009
;
45
(
2
):
367
-
376
.
95.
Percival
CJ
,
Richtsmeier
JT
.
Angiogenesis and intramembranous osteogenesis
.
Dev Dyn.
2013
;
242
(
8
):
909
-
922
.
96.
Carlevaro
MF
,
Cermelli
S
,
Cancedda
R
,
Descalzi Cancedda
F
.
Vascular endothelial growth factor (VEGF) in cartilage neovascularization and chondrocyte differentiation: auto-paracrine role during endochondral bone formation
.
J Cell Sci.
2000
;
113
(
Pt 1
):
59
-
69
.
97.
Mayer
H
,
Bertram
H
,
Lindenmaier
W
,
Korff
T
,
Weber
H
,
Weich
H
.
Vascular endothelial growth factor (VEGF-A) expression in human mesenchymal stem cells:autocrine and paracrine role on osteoblastic and endothelial differentiation
.
J Cell Biochem.
2005
;
95
(
4
):
827
-
839
.
98.
Liu
Y
,
Berendsen
AD
,
Jia
S
, et al
.
Intracellular VEGF regulates the balance between osteoblast and adipocyte differentiation
.
J Clin Invest.
2012
;
122
(
9
):
3101
-
3113
.
99.
Yang
Q
,
McHugh
KP
,
Patntirapong
S
,
Gu
X
,
Wunderlich
L
,
Hauschka
PV
.
VEGF enhancement of osteoclast survival and bone resorption involves VEGF receptor-2 signaling and beta3-integrin
.
Matrix Biol.
2008
;
27
(
7
):
589
-
599
.
100.
Palmqvist
P
,
Persson
E
,
Conaway
HH
,
Lerner
UH
.
IL-6, leukemia inhibitory factor, and oncostatin M stimulate bone resorption and regulate the expression of receptor activator of NF-kappa B ligand, osteoprotegerin, and receptor activator of NF-kappa B in mouse calvariae
.
J Immunol.
2002
;
169
(
6
):
3353
-
3362
.
101.
Walker
EC
,
McGregor
NE
,
Poulton
IJ
, et al
.
Oncostatin M promotes bone formation independently of resorption when signaling through leukemia inhibitory factor receptor in mice
.
J Clin Invest.
2010
;
120
(
2
):
582
-
592
.
102.
Yoshitake
F
,
Itoh
S
,
Narita
H
,
Ishihara
K
,
Ebisu
S
.
Interleukin-6 directly inhibits osteoclast differentiation by suppressing receptor activator of NF-kappaB signaling pathways
.
J Biol Chem.
2008
;
283
(
17
):
11535
-
11540
.
103.
Malara
A
,
Abbonante
V
,
Di Buduo
CA
,
Tozzi
L
,
Currao
M
,
Balduini
A
.
The secret life of a megakaryocyte:emerging roles in bone marrow homeostasis control
.
Cell Mol Life Sci.
2015
;
72
(
8
):
1517
-
1536
.
104.
Olson
TS
,
Caselli
A
,
Otsuru
S
, et al
.
Megakaryocytes promote murine osteoblastic HSC niche expansion and stem cell engraftment after radioablative conditioning
.
Blood.
2013
;
121
(
26
):
5238
-
5249
.
105.
Fox
SB
,
Lorenzen
J
,
Heryet
A
,
Jones
M
,
Gatter
KC
,
Mason
DY
.
Megakaryocytes in myelodysplasia: an immunohistochemical study on bone marrow trephines
.
Histopathology.
1990
;
17
(
1
):
69
-
74
.
106.
Thiele
J
,
Quitmann
H
,
Wagner
S
,
Fischer
R
.
Dysmegakaryopoiesis in myelodysplastic syndromes (MDS): an immunomorphometric study of bone marrow trephine biopsy specimens.
J Clin Pathol.
1991
;
44
(
4
):
300
-
305
.
107.
Ghai
S
,
Rai
S
.
Megakaryocytic morphology in Janus kinase 2 V617F positive myeloproliferative neoplasm
.
South Asian J Cancer.
2017
;
6
(
2
):
75
-
78
.
108.
Soves
CP
,
Miller
JD
,
Begun
DL
,
Taichman
RS
,
Hankenson
KD
,
Goldstein
SA
.
Megakaryocytes are mechanically responsive and influence osteoblast proliferation and differentiation
.
Bone.
2014
;
66
(
66
):
111
-
120
.
109.
Kacena
MA
,
Nelson
T
,
Clough
ME
, et al
.
Megakaryocyte-mediated inhibition of osteoclast development
.
Bone.
2006
;
39
(
5
):
991
-
999
.
110.
Bord
S
,
Frith
E
,
Ireland
DC
,
Scott
MA
,
Craig
JI
,
Compston
JE
.
Megakaryocytes modulate osteoblast synthesis of type-l collagen, osteoprotegerin, and RANKL
.
Bone.
2005
;
36
(
5
):
812
-
819
.
111.
Beeton
CA
,
Bord
S
,
Ireland
D
,
Compston
JE
.
Osteoclast formation and bone resorption are inhibited by megakaryocytes
.
Bone.
2006
;
39
(
5
):
985
-
990
.
112.
Ciovacco
WA
,
Goldberg
CG
,
Taylor
AF
, et al
.
The role of gap junctions in megakaryocyte-mediated osteoblast proliferation and differentiation
.
Bone.
2009
;
44
(
1
):
80
-
86
.
113.
Lemieux
JM
,
Horowitz
MC
,
Kacena
MA
.
Involvement of integrins alpha(3)beta(1) and alpha(5)beta(1) and glycoprotein IIb in megakaryocyte-induced osteoblast proliferation
.
J Cell Biochem.
2010
;
109
(
5
):
927
-
932
.
114.
Kacena
MA
,
Eleniste
PP
,
Cheng
YH
, et al
.
Megakaryocytes regulate expression of Pyk2 isoforms and caspase-mediated cleavage of actin in osteoblasts
.
J Biol Chem.
2012
;
287
(
21
):
17257
-
17268
.
115.
Cheng
YH
,
Streicher
DA
,
Waning
DL
,
Chitteti
BR
,
Gerard-O'Riley
R
,
Horowitz
MC
et al
.
Signaling pathways involved in megakaryocyte-mediated proliferation of osteoblast lineage cells
.
J Cell Physiol.
2015
;
230
(
3
):
578
-
586
.
116.
Maupin
KA
,
Himes
ER
,
Plett
AP
, et al
.
Aging negatively impacts the ability of megakaryocytes to stimulate osteoblast proliferation and bone mass
.
Bone.
2019
;
127
(
127
):
452
-
459
.
117.
Ciovacco
WA
,
Cheng
YH
,
Horowitz
MC
,
Kacena
MA
.
Immature and mature megakaryocytes enhance osteoblast proliferation and inhibit osteoclast formation
.
J Cell Biochem.
2010
;
109
(
4
):
774
-
781
.
118.
Fitter
S
,
Dewar
AL
,
Kostakis
P
,
To
LB
,
Hughes
TP
,
Roberts
MM
et al
.
Long-term imatinib therapy promotes bone formation in CML patients
.
Blood.
2008
;
111
(
5
):
2538
-
2547
.
119.
Smerdely
P
,
Seller
M
,
Smith
A
,
Day
P
,
Diamond
T
.
Predictors of bone mass in healthy older men in the community
.
Med J Aust.
2000
;
173
(
4
):
183
-
186
.
120.
Courpron
P
,
Meunier
P
,
Edouard
C
,
Bernard
J
,
Bringuier
JP
,
Vignon
G
.
Donnèes histologiques quantitatives sur le vieillissement osseux humain [Quantitative histological data on the aging of human bone]
.
Rev Rhum Mal Osteoartic.
1973
;
40
(
7
):
469
483
.
121.
Meunier
PJ
,
Courpron
P
. Iliac trabecular bone volume in 236 controls. Representativeness of iliac samples. In:
Jaworski
ZFG
. ed.
Proceedings of the First Workshop on Bone Morphometry.
Ottawa, Canada
:
University of Ottawa Press
,
1973
;
100
.
122.
Melsen
F
,
Melsen
B
,
Mosekilde
L
,
Bergmann
S
.
Histomorphometric analysis of normal bone from the iliac crest.
Acta Pathol Microbiol Scand A.
1978
;
86
(
1
):
70
81
.
Sign in via your Institution