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

Pathogenetic mechanisms in primary myelofibrosis

Pathogenetic mechanisms in primary myelofibrosis
Author:
Ayalew Tefferi, MD
Section Editor:
Richard A Larson, MD
Deputy Editor:
Alan G Rosmarin, MD
Literature review current through: Jan 2024.
This topic last updated: Mar 21, 2022.

INTRODUCTION — Primary myelofibrosis [1-3] (PMF, chronic idiopathic myelofibrosis, agnogenic myeloid metaplasia) is one of the chronic myeloproliferative neoplasms, which are collectively characterized by clonal proliferation of myeloid cells with variable morphologic maturity and hematopoietic efficiency (table 1).

The primary disease process in PMF is a clonal hematopoietic stem cell disorder that results in chronic myeloproliferation and atypical megakaryocytic hyperplasia [4]. The secondary process of bone marrow fibrosis (BMF) is the result of nonclonal fibroblastic proliferation and hyperactivity induced by growth factors abnormally shed from clonally expanded megakaryocytes [5]. BMF is the hallmark of PMF and contributes to the impaired hematopoiesis that leads to severe anemia. In addition to BMF and anemia, patients with PMF suffer from marked splenomegaly, extramedullary hematopoiesis, and severe constitutional symptoms. (See "Clinical manifestations and diagnosis of primary myelofibrosis".)

The potential to arrest or reverse the BMF in PMF may offer an alternative approach to palliative therapy. With this goal in mind, the pathogenesis of PMF will be discussed here. The prognosis and treatment of PMF are discussed separately. (See "Myelofibrosis (MF): Management of primary MF and secondary MF".)

ETIOLOGY — The exact cause of primary myelofibrosis (PMF) is unknown. PMF, along with the other chronic myeloproliferative disorders, chronic myeloid leukemia, polycythemia vera, and essential thrombocythemia, is considered to arise from a somatic mutation of a pluripotent hematopoietic progenitor cell [6,7]. A defective stem cell "niche" within the bone marrow has been postulated for PMF [8]. (See "Overview of the myeloproliferative neoplasms".)

The occurrence of PMF has, in a minority of cases, been linked to exposure to thorium dioxide, petroleum manufacturing plants (especially toluene and benzene), and ionizing radiation [9-11]. A very high incidence of PMF has been noted in patients given thorium-based radiographic contrast material and in individuals exposed to atomic bombs at Hiroshima [12,13].

Mice with a mutation of the GATA-1 transcription factor gene develop a hematologic picture similar to myelofibrosis at 15 months of age, and may represent a suitable animal model for the human disease [5,14,15].

CELLULAR ABNORMALITIES

Chromosomal and other genetic abnormalities — Approximately 50 to 60 percent of patients with primary myelofibrosis (PMF) have clonal karyotypic abnormalities at diagnosis [5,16-21]. However, none of these abnormalities is specific to this disorder. The most common findings, accounting for 50 to 65 percent of the karyotypic changes, are deletion of a segment of the chromosome bearing the retinoblastoma gene (13q-), 20q-, and partial trisomy 1q (table 2). Patients with these cytogenetic abnormalities are reported to have a worse prognosis [16,17,22].

In our series of 884 patients with PMF, unfavorable karyotype has been one of the factors associated with reduced overall survival, and includes such changes as +8, -7/7q-, i(17q), -5/5q-, 12p-, inv(3), and 11q23 rearrangement [23]. The detrimental effect of unfavorable karyotype was mostly attributed to monosomal karyotype or inv(3)/i(17q).

Gains of chromosome 9 or 9p have also been noted in our studies (13 percent) and those of others (50 percent), suggesting that genes on 9p may play a crucial role in the pathogenesis of PMF [24]. Of interest, mutations of JAK2 (Janus kinase 2), a gene found on 9p, have been found in approximately 50 percent of patients with PMF. (See 'JAK2 mutations' below and "Overview of the myeloproliferative neoplasms", section on 'Mutations in PV, ET, and PMF'.)

A number of genetic mutations and single nucleotide polymorphisms have been identified in patients with PMF and other myeloproliferative neoplasms associated with leukemic transformation [25-29]. As an example, a A3669G polymorphism of the glucocorticoid receptor was found to contribute to the phenotype of excess myeloproliferation in PMF (eg, higher white blood cell count, larger spleen, higher frequency of CD34+ cells at diagnosis) and, in cooperation with the JAK2V617F mutation, increasing the risk of blastic transformation [30].

In summary, mutations in PMF involve primarily JAK2 but also to a smaller extent MPL, LNK, CBL, TET2, ASXL1, IDH1, IDH2, IKZF1, EZH2, DNMT3A, TP53, SF3B1 and SFSR2. None of these mutations has been consistently traced back to the ancestral clone, and their disease-initiating potential is uncertain. Some (eg, JAK2 and SF3B1 mutations) might contribute to specific phenotypes such as erythrocytosis and ring sideroblasts, respectively. Available data suggest inferior survival in PMF associated with nullizygosity for JAK2 46/1 haplotype, low JAK2V617F allele burden, and the presence of IDH, ASXL1, SRSF2 or EZH2 mutations. Some of these mutations (eg, IDH, SRSF2) have also been associated with inferior leukemia-free survival. In contrast, the presence or absence of JAK2V617F, MPL or TET2 mutations do not appear to affect survival.

Cytokine abnormalities — Fibroblasts do not share the chromosomal abnormalities found in hematopoietic cells [31]. As a result, it is thought that bone marrow fibrosis, as well as many of the other signs and symptoms of PMF, are secondary reactions to the clonal hemopathy mediated by cytokines released from the neoplastic megakaryocytes and other clonally expanded hematopoietic cells, such as T and B cells [32,33].

Spontaneous hematopoietic colony growth in vitro — One of the hallmarks of polycythemia vera (PV) is in vitro erythropoietin- independent growth of red blood cell (RBC) colonies. Spontaneous growth of both RBC colonies and megakaryocytes (in the absence of exogenous cytokines) has also been described in a minority of patients with PMF and ET.

A number of findings support the involvement of the trophic hormone thrombopoietin (TPO) and/or its receptor (c-MPL) in the pathogenesis of PMF, particularly in the pathogenesis of bone marrow fibrosis (see 'Bone marrow fibrosis' below). There is also evidence that the TPO system is involved in the spontaneous megakaryocyte growth in PMF. As examples, both preparations of the soluble murine TPO receptor and antisense oligonucleotides against the TPO receptor markedly inhibit spontaneous megakaryocyte growth in PMF and in ET and PV [34,35].

JAK/STAT pathway

JAK2 mutations — One suggested mechanism of intrinsic growth factor hypersensitivity in PMF involves megakaryocyte overexpression of FKBP51 associated with JAK2/STAT5 activation, consistent with the discovery of an activation mutation of JAK2 in patients with PV, ET, and PMF [5]. (See "Overview of the myeloproliferative neoplasms", section on 'Mutations in PV, ET, and PMF' and "Molecular pathogenesis of congenital erythrocytoses and polycythemia vera", section on 'JAK2 mutations'.)

Constitutive activation of the JAK/STAT pathway appears to be an important pathogenetic event in patients with the myeloproliferative disorders. However, only approximately 50 percent of patients with PMF have the JAK2 mutation, suggesting the presence of other modes of JAK/STAT activation, such as activating mutations in hematopoietic-specific cytokine receptors (see below) [36].

Calreticulin gene mutations — Mutations in the calreticulin gene (CALR) have been reported in approximately 35 percent of patients with ET [37,38]. Thus far CALR mutations have been seen only in patients with ET or PMF and have not been observed in patients with PV or in those with JAK2 or MPL mutations. (See "Overview of the myeloproliferative neoplasms", section on 'Mutations in PV, ET, and PMF'.)

MPL mutations — Mutations of the thrombopoietin receptor (MPL) are also capable of activation of JAK/STAT signaling in patients with PMF [36]. The finding of novel somatic activating mutations in the thrombopoietin receptor (MPL mutations W515L and W515K) in approximately 5 percent of patients with PMF, 1 percent of those with ET, and in none of the patients with PV suggests that these MPL mutations favor megakaryocyte fate while the JAK2 mutation favors erythroid fate [36,39,40].

BONE MARROW FIBROSIS — Bone marrow fibrosis (BMF) in primary myelofibrosis (PMF) results from the abnormal deposition of excess collagen derived from fibroblasts. Although several types of collagen provide the reticular matrix support in normal bone marrow, types III, IV, and I are the major components of BMF associated with PMF [41].

The collagen-producing fibroblasts in PMF are functionally and physically similar to normal fibroblasts and are polyclonal [42,43]. They are secondarily stimulated to proliferate and overproduce collagen by growth factors secreted by the neighboring megakaryocytes [44,45]. Thus, BMF in PMF is a reactive process accompanying the underlying clonal stem cell disorder.

The major megakaryocyte-derived cytokine implicated in the mediation of BMF in PMF is transforming growth factor (TGF)-beta [4,46], which may interact with thrombopoietin (figure 1). Other growth factors also may contribute to the fibrotic reaction, including platelet derived growth factor (PDGF), epidermal growth factor (EGF), basic fibroblast growth factor (bFGF, FGF-2), vascular endothelial growth factor (VEGF), calmodulin, matrix metalloproteinase-9, and lysyl oxidase [4,5,46-53]. (See 'Transforming growth factor-beta' below.)

This fibrous reaction with abnormal accumulation of extracellular matrix components is also dependent on matrix metalloproteinases (MMP) and tissue inhibitors of MMPs (TIMP) [54]. In a study of 25 patients with PMF, significantly decreased plasma levels of MMP-3 were found, which correlated inversely with the degree of bone marrow fibrosis [55]. Elevated levels of TIMP-1 were also found, suggesting that the balance between MMPs and TIMP may be essential in fibrosis formation.

It has been suggested, based upon a murine model and on bone marrow samples from patients with PMF, that there is a significant degree of entry of hematopoietic cells into megakaryocyte cytoplasm, a phenomenon called emperipolesis [56]. This is probably brought about by increased expression and abnormal localization of P-selectin by the megakaryocytes, leading to engulfment, activation, and damage to the cells, mainly neutrophils and eosinophils, passing through the megakaryocyte cytoplasm. The result is release of lytic granules from the engulfed cells, progressive destruction of megakaryocytes with degradation and lysis of their alpha granules, and release of growth factors, resulting in the marked fibroblast activation and infiltration characteristic of PMF.

Transforming growth factor-beta — The major megakaryocyte-derived cytokine implicated in the mediation of BMF in PMF is transforming growth factor (TGF)-beta, which may interact with thrombopoietin (figure 1) [47,57-62].

TGF-beta is a glycoprotein that is synthesized and secreted primarily by the monocyte-macrophage system and endothelial cells, and also by megakaryocytes [4,57,58]. It is capable of enhancing the production and secretion of extracellular matrix proteins, including collagen types III and I from fibroblasts [59,60], and may also interact with IL-1, fibronectin, and substance P [63]. The expression of TGF-beta has been demonstrated at the level of mRNA and secreted peptide in clonal megakaryocytes [57,61]. In addition, circulating concentrations of TGF-beta are increased in patients with PMF [64].

In vitro, anti-TGF-beta antibodies decrease the collagen synthesis mediated by megakaryoblast-conditioned media [57]. This type of inhibition can also be achieved by tumor necrosis factor-alpha and interferon-gamma, which suppress the activation of type I collagen gene expression by TGF-beta [62].

Thrombopoietin — A PMF-like syndrome can be induced in mice via exposure to high concentrations of thrombopoietin [65-70]. Thrombopoietin (TPO) is the major growth factor required for megakaryocyte growth and development. The TPO glycoprotein can be modified by deglycosylation and subsequent coupling to polyethylene glycol (PEG) to be less immunogenic and have an extended half-life.

Mice injected daily with high doses of PEG-TPO develop a reversible bone marrow fibrosis accompanied by thrombocytosis, megakaryocytic hyperplasia, splenomegaly, extramedullary hematopoiesis, and anemia [66,68]. A similar syndrome has been observed in TPO-transfected mice [67,70] and has been attributed to increased TGF-beta activity secondary to an increased megakaryocyte mass [67,68]. Although the process was reversible by bone marrow transplantation [67], chronic exposure led to a fatal myeloproliferative disorder [65].

The role of TPO has been further evaluated in immune-compromised mice. Overexpression of the TPO gene using adenovectors in SCID mice (severe combined immune deficient) resulted in thrombocytosis, increased marrow megakaryocytes, fibrosis, and extramedullary hematopoiesis that mimicked PMF [71]. However, similar overexpression of thrombopoietin in NOD-SCID mice (which have reduced monocyte and macrophage function in addition to the lymphocyte deficiency in SCID mice) produced thrombocytosis and megakaryocytosis but no fibrosis. These results imply that other monocyte/macrophage mediators are involved in causing the fibrosis.

Overview — The above discussion supports the pathogenetic role of TGF-beta in BMF. The pathogenetic interaction of TPO and TGF-beta in the human disease has not been elucidated. Serum TPO concentrations in patients with PMF are higher than those of normal volunteers or patients with reactive thrombocytosis and have been correlated with the degree of BMF [72,73]. However, there was no correlation with megakaryocyte mass, and the demonstration of low TPO-receptor (c-Mpl) expression by platelets and megakaryocytes from patients with PMF suggests that defective TPO clearance may contribute to the above normal TPO values in patients with PMF [74,75].

Finally, CD34+ hematopoietic progenitor cells from patients with PMF have increased expression of mRNA transcripts of bFGF and its receptors and decreased expression of TGF-beta type II receptors [76]. The pathogenetic and clinical relevance of these observations is currently unknown, although the myeloproliferation characteristic of this disease may result from abnormal proliferation of these precursors [77].

EXTRAMEDULLARY HEMATOPOIESIS — The mechanisms responsible for extramedullary hematopoiesis (EMH) in primary myelofibrosis (PMF) are not understood. The distribution of hematopoietic tissue approximates that in the fetus. In both a mouse model of marrow fibrosis and in marrow fibrosis associated with metastatic disease, there is abnormal release of marrow precursors into distorted bone marrow sinusoids and then into the circulation [78,79]. These marrow precursors may be responsible for EMH in PMF [79,80], although increased trafficking of CD34+ cells into the circulation may also play a role [81]. (See "Clinical manifestations and diagnosis of primary myelofibrosis", section on 'Circulating CD34+ cells'.)

Foci of EMH may also be found in soft tissues, body cavities, serosal surfaces, central nervous system, skin, and other locations. EMH may resemble tumors at these sites, often causing obstructive symptoms, especially in the central nervous system. Such foci may enlarge significantly postsplenectomy, perhaps due to the loss of filtering function of the spleen.

In some patients with PMF, hematopoiesis is only present in extramedullary sites [82]. EMH is rarely as effective as medullary hematopoiesis, thereby leading to the cytopenias so often found in this disease.

INCREASED BONE MARROW VASCULARITY — A number of studies have shown that bone marrow microvessel density (MVD) is increased in patients with primary myelofibrosis (PMF) and other BCR/ABL1-negative myeloproliferative neoplasms [83-85]. In one of the studies, MVD was significantly increased in 114 patients with PMF, when compared with bone marrows from 44 normal controls, 15 patients with polycythemia vera and 17 with essential thrombocythemia [83]. This phenomenon, as with bone marrow fibrosis, is thought to be secondary to release of several growth factors by megakaryocytes (eg, VEGF, PDGF, TGF-beta, bFGF). (See 'Bone marrow fibrosis' above.)

In this study, increased MVD significantly correlated with the presence of megakaryocyte clumping, increased splenic size, and, in a multivariate model, decreased overall survival. Based on these correlations, phase II studies employing thalidomide and lenalidomide, agents with antiangiogenic properties, are ongoing [5,83,86]. (See "Overview of angiogenesis inhibitors", section on 'Immunomodulatory drugs (IMiDs)'.)

SUMMARY

Primary myelofibrosis (PMF) – PMF is characterized by chronic myeloproliferation, atypical megakaryocytic hyperplasia, increased bone marrow vascularity, and bone marrow fibrosis (BMF). Increased bone marrow vascularity and BMF are related to proliferation and hyperactivity of non-clonal fibroblasts that are induced by growth factors shed from clonally expanded megakaryocytes. (See 'Bone marrow fibrosis' above and 'Increased bone marrow vascularity' above.)

Pathogenesis – The following features have been implicated in the pathogenesis of PMF/BMF (see 'Overview' above):

Chromosomal abnormalities – Half of patients with PMF have clonal chromosomal abnormalities, but none is specific to this disorder. (See 'Chromosomal and other genetic abnormalities' above.)

Abnormal megakaryocyte growth – Growth of red blood cell (RBC) colonies and/or megakaryocytes with little or no exogenous cytokines is seen in some patients with PMF. (See 'Spontaneous hematopoietic colony growth in vitro' above.)

JAK/STAT activation – Constitutive activation of the JAK/STAT pathway, in association with mutations of JAK2, MPL, or CALR, contributes to the pathogenesis of PMF. (See 'JAK/STAT pathway' above.)

Bone marrow fibrosis – BMF may be related to abnormal expression of transforming growth factor-beta (TGF-beta), thrombopoietin (TPO), and/or the TPO receptor, MPL. (See 'Transforming growth factor-beta' above and 'Thrombopoietin' above and 'Cytokine abnormalities' above.)

ACKNOWLEDGMENT — The editors of UpToDate acknowledge the contributions of Stanley L Schrier, MD as Section Editor on this topic, his tenure as the founding Editor-in-Chief for UpToDate in Hematology, and his dedicated and longstanding involvement with the UpToDate program.

  1. Mesa RA, Verstovsek S, Cervantes F, et al. Primary myelofibrosis (PMF), post polycythemia vera myelofibrosis (post-PV MF), post essential thrombocythemia myelofibrosis (post-ET MF), blast phase PMF (PMF-BP): Consensus on terminology by the international working group for myelofibrosis research and treatment (IWG-MRT). Leuk Res 2007; 31:737.
  2. Thiele J, Kvasnicka HM, Tefferi A, et al. Primary myelofibrosis. In: WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues, Swerdlow SH, Campo E, Harris NL (Eds), IARC Press, Lyon 2008. p.44.
  3. Thiele J, Pierre R, Imbert M, et al. Chronic idiopathic myelofibrosis. In: World Health Organization Classification of Tumours. Pathology and Genetics of Tumours of Haematopoietic and Lymphoid Tissues, Jaffe ES, Harris NL, Stein H, Vardiman JW (Eds), IARC Press, Lyon 2001. p.35.
  4. Ciurea SO, Merchant D, Mahmud N, et al. Pivotal contributions of megakaryocytes to the biology of idiopathic myelofibrosis. Blood 2007; 110:986.
  5. Tefferi A. Pathogenesis of myelofibrosis with myeloid metaplasia. J Clin Oncol 2005; 23:8520.
  6. Campbell PJ, Green AR. The myeloproliferative disorders. N Engl J Med 2006; 355:2452.
  7. Tefferi A, Gangat N, Pardanani A, Crispino JD. Myelofibrosis: Genetic Characteristics and the Emerging Therapeutic Landscape. Cancer Res 2022; 82:749.
  8. Lataillade JJ, Pierre-Louis O, Hasselbalch HC, et al. Does primary myelofibrosis involve a defective stem cell niche? From concept to evidence. Blood 2008; 112:3026.
  9. Smith RE, Chelmowski MK, Szabo EJ. Myelofibrosis: a review of clinical and pathologic features and treatment. Crit Rev Oncol Hematol 1990; 10:305.
  10. Bosch X, Campistol JM, Montoliu J, et al. Toluene-associated myelofibrosis. Blut 1989; 58:219.
  11. Bosch X, Campistol JM, Montoliu J, Revert L. Myelofibrosis and focal segmental glomerulosclerosis associated with toluene poisoning. Hum Toxicol 1988; 7:357.
  12. Visfeldt J, Andersson M. Pathoanatomical aspects of malignant haematological disorders among Danish patients exposed to thorium dioxide. APMIS 1995; 103:29.
  13. ANDERSON RE, HOSHINO T, YAMAMOTO T. MYELOFIBROSIS WITH MYELOID METAPLASIA IN SURVIVORS OF THE ATOMIC BOMB IN HIROSHIMA. Ann Intern Med 1964; 60:1.
  14. Vannucchi AM, Bianchi L, Cellai C, et al. Development of myelofibrosis in mice genetically impaired for GATA-1 expression (GATA-1(low) mice). Blood 2002; 100:1123.
  15. Vannucchi AM, Bianchi L, Paoletti F, et al. A pathobiologic pathway linking thrombopoietin, GATA-1, and TGF-beta1 in the development of myelofibrosis. Blood 2005; 105:3493.
  16. Dupriez B, Morel P, Demory JL, et al. Prognostic factors in agnogenic myeloid metaplasia: a report on 195 cases with a new scoring system. Blood 1996; 88:1013.
  17. Reilly JT, Snowden JA, Spearing RL, et al. Cytogenetic abnormalities and their prognostic significance in idiopathic myelofibrosis: a study of 106 cases. Br J Haematol 1997; 98:96.
  18. Sinclair EJ, Forrest EC, Reilly JT, et al. Fluorescence in situ hybridization analysis of 25 cases of idiopathic myelofibrosis and two cases of secondary myelofibrosis: monoallelic loss of RB1, D13S319 and D13S25 loci associated with cytogenetic deletion and translocation involving 13q14. Br J Haematol 2001; 113:365.
  19. Tefferi A, Mesa RA, Schroeder G, et al. Cytogenetic findings and their clinical relevance in myelofibrosis with myeloid metaplasia. Br J Haematol 2001; 113:763.
  20. Tefferi A, Dingli D, Li CY, Dewald GW. Prognostic diversity among cytogenetic abnormalities in myelofibrosis with myeloid metaplasia. Cancer 2005; 104:1656.
  21. Strasser-Weippl K, Steurer M, Kees M, et al. Prognostic relevance of cytogenetics determined by fluorescent in situ hybridization in patients having myelofibrosis with myeloid metaplasia. Cancer 2006; 107:2801.
  22. Demory JL, Dupriez B, Fenaux P, et al. Cytogenetic studies and their prognostic significance in agnogenic myeloid metaplasia: a report on 47 cases. Blood 1988; 72:855.
  23. Tefferi A, Pardanani A, Gangat N, et al. Leukemia risk models in primary myelofibrosis: an International Working Group study. Leukemia 2012; 26:1439.
  24. Al-Assar O, Ul-Hassan A, Brown R, et al. Gains on 9p are common genomic aberrations in idiopathic myelofibrosis: a comparative genomic hybridization study. Br J Haematol 2005; 129:66.
  25. Thoennissen NH, Krug UO, Lee DH, et al. Prevalence and prognostic impact of allelic imbalances associated with leukemic transformation of Philadelphia chromosome-negative myeloproliferative neoplasms. Blood 2010; 115:2882.
  26. Tefferi A, Jimma T, Sulai NH, et al. IDH mutations in primary myelofibrosis predict leukemic transformation and shortened survival: clinical evidence for leukemogenic collaboration with JAK2V617F. Leukemia 2012; 26:475.
  27. Beer PA, Delhommeau F, LeCouédic JP, et al. Two routes to leukemic transformation after a JAK2 mutation-positive myeloproliferative neoplasm. Blood 2010; 115:2891.
  28. Zhang SJ, Rampal R, Manshouri T, et al. Genetic analysis of patients with leukemic transformation of myeloproliferative neoplasms shows recurrent SRSF2 mutations that are associated with adverse outcome. Blood 2012; 119:4480.
  29. Lasho TL, Jimma T, Finke CM, et al. SRSF2 mutations in primary myelofibrosis: significant clustering with IDH mutations and independent association with inferior overall and leukemia-free survival. Blood 2012; 120:4168.
  30. Poletto V, Rosti V, Villani L, et al. A3669G polymorphism of glucocorticoid receptor is a susceptibility allele for primary myelofibrosis and contributes to phenotypic diversity and blast transformation. Blood 2012; 120:3112.
  31. Wang JC, Lang HD, Lichter S, et al. Cytogenetic studies of bone marrow fibroblasts cultured from patients with myelofibrosis and myeloid metaplasia. Br J Haematol 1992; 80:184.
  32. Reeder TL, Bailey RJ, Dewald GW, Tefferi A. Both B and T lymphocytes may be clonally involved in myelofibrosis with myeloid metaplasia. Blood 2003; 101:1981.
  33. Tefferi A, Vaidya R, Caramazza D, et al. 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; 29:1356.
  34. Taksin AL, Couedic JP, Dusanter-Fourt I, et al. Autonomous megakaryocyte growth in essential thrombocythemia and idiopathic myelofibrosis is not related to a c-mpl mutation or to an autocrine stimulation by Mpl-L. Blood 1999; 93:125.
  35. Li Y, Hetet G, Kiladjian JJ, et al. Proto-oncogene c-mpl is involved in spontaneous megakaryocytopoiesis in myeloproliferative disorders. Br J Haematol 1996; 92:60.
  36. Pikman Y, Lee BH, Mercher T, et al. MPLW515L is a novel somatic activating mutation in myelofibrosis with myeloid metaplasia. PLoS Med 2006; 3:e270.
  37. Nangalia J, Massie CE, Baxter EJ, et al. Somatic CALR mutations in myeloproliferative neoplasms with nonmutated JAK2. N Engl J Med 2013; 369:2391.
  38. Klampfl T, Gisslinger H, Harutyunyan AS, et al. Somatic mutations of calreticulin in myeloproliferative neoplasms. N Engl J Med 2013; 369:2379.
  39. Pardanani AD, Levine RL, Lasho T, et al. MPL515 mutations in myeloproliferative and other myeloid disorders: a study of 1182 patients. Blood 2006; 108:3472.
  40. 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; 121:4388.
  41. Castro-Malaspina H, Jhanwar SC. Properties of myelofibrosis-derived fibroblasts. Prog Clin Biol Res 1984; 154:307.
  42. Jacobson RJ, Salo A, Fialkow PJ. Agnogenic myeloid metaplasia: a clonal proliferation of hematopoietic stem cells with secondary myelofibrosis. Blood 1978; 51:189.
  43. Lisse I, Hasselbalch H, Junker P. Bone marrow stroma in idiopathic myelofibrosis and other haematological diseases. An immunohistochemical study. APMIS 1991; 99:171.
  44. Groopman JE. The pathogenesis of myelofibrosis in myeloproliferative disorders. Ann Intern Med 1980; 92:857.
  45. Castro-Malaspina H, Rabellino EM, Yen A, et al. Human megakaryocyte stimulation of proliferation of bone marrow fibroblasts. Blood 1981; 57:781.
  46. 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.
  47. Kimura A, Katoh O, Kuramoto A. Effects of platelet derived growth factor, epidermal growth factor and transforming growth factor-beta on the growth of human marrow fibroblasts. Br J Haematol 1988; 69:9.
  48. Martyré MC, Le Bousse-Kerdiles MC, Romquin N, et al. Elevated levels of basic fibroblast growth factor in megakaryocytes and platelets from patients with idiopathic myelofibrosis. Br J Haematol 1997; 97:441.
  49. Dalley A, Smith JM, Reilly JT, Neil SM. Investigation of calmodulin and basic fibroblast growth factor (bFGF) in idiopathic myelofibrosis: evidence for a role of extracellular calmodulin in fibroblast proliferation. Br J Haematol 1996; 93:856.
  50. Bock O, Muth M, Theophile K, et al. Identification of new target molecules PTK2, TGFBR2 and CD9 overexpressed during advanced bone marrow remodelling in primary myelofibrosis. Br J Haematol 2009; 146:510.
  51. Papadantonakis N, Matsuura S, Ravid K. Megakaryocyte pathology and bone marrow fibrosis: the lysyl oxidase connection. Blood 2012; 120:1774.
  52. Tadmor T, Bejar J, Attias D, et al. The expression of lysyl-oxidase gene family members in myeloproliferative neoplasms. Am J Hematol 2013; 88:355.
  53. Bedekovics J, Kiss A, Beke L, et al. Platelet derived growth factor receptor-beta (PDGFRβ) expression is limited to activated stromal cells in the bone marrow and shows a strong correlation with the grade of myelofibrosis. Virchows Arch 2013; 463:57.
  54. Murate T, Yamashita K, Isogai C, et al. The production of tissue inhibitors of metalloproteinases (TIMPs) in megakaryopoiesis: possible role of platelet- and megakaryocyte-derived TIMPs in bone marrow fibrosis. Br J Haematol 1997; 99:181.
  55. Wang JC, Novetsky A, Chen C, Novetsky AD. Plasma matrix metalloproteinase and tissue inhibitor of metalloproteinase in patients with agnogenic myeloid metaplasia or idiopathic primary myelofibrosis. Br J Haematol 2002; 119:709.
  56. Schmitt A, Jouault H, Guichard J, et al. Pathologic interaction between megakaryocytes and polymorphonuclear leukocytes in myelofibrosis. Blood 2000; 96:1342.
  57. Terui T, Niitsu Y, Mahara K, et al. The production of transforming growth factor-beta in acute megakaryoblastic leukemia and its possible implications in myelofibrosis. Blood 1990; 75:1540.
  58. Ignotz RA, Massagué J. Transforming growth factor-beta stimulates the expression of fibronectin and collagen and their incorporation into the extracellular matrix. J Biol Chem 1986; 261:4337.
  59. Kimura A, Katoh O, Hyodo H, Kuramoto A. Transforming growth factor-beta regulates growth as well as collagen and fibronectin synthesis of human marrow fibroblasts. Br J Haematol 1989; 72:486.
  60. Reilly JT. Idiopathic myelofibrosis: pathogenesis, natural history and management. Blood Rev 1997; 11:233.
  61. Martyré MC, Romquin N, Le Bousse-Kerdiles MC, et al. Transforming growth factor-beta and megakaryocytes in the pathogenesis of idiopathic myelofibrosis. Br J Haematol 1994; 88:9.
  62. Kähäri VM, Chen YQ, Su MW, et al. Tumor necrosis factor-alpha and interferon-gamma suppress the activation of human type I collagen gene expression by transforming growth factor-beta 1. Evidence for two distinct mechanisms of inhibition at the transcriptional and posttranscriptional levels. J Clin Invest 1990; 86:1489.
  63. Chang VT, Yook C, Rameshwar P. Synergism between fibronectin and transforming growth factor-β1 in the production of substance P in monocytes of patients with myelofibrosis. Leuk Lymphoma 2013; 54:631.
  64. Rameshwar P, Chang VT, Thacker UF, Gascón P. Systemic transforming growth factor-beta in patients with bone marrow fibrosis--pathophysiological implications. Am J Hematol 1998; 59:133.
  65. Villeval JL, Cohen-Solal K, Tulliez M, et al. High thrombopoietin production by hematopoietic cells induces a fatal myeloproliferative syndrome in mice. Blood 1997; 90:4369.
  66. Ulich TR, del Castillo J, Senaldi G, et al. Systemic hematologic effects of PEG-rHuMGDF-induced megakaryocyte hyperplasia in mice. Blood 1996; 87:5006.
  67. Yan XQ, Lacey D, Hill D, et al. A model of myelofibrosis and osteosclerosis in mice induced by overexpressing thrombopoietin (mpl ligand): reversal of disease by bone marrow transplantation. Blood 1996; 88:402.
  68. Yanagida M, Ide Y, Imai A, et al. The role of transforming growth factor-beta in PEG-rHuMGDF-induced reversible myelofibrosis in rats. Br J Haematol 1997; 99:739.
  69. Abina MA, Tulliez M, Lacout C, et al. Major effects of TPO delivered by a single injection of a recombinant adenovirus on prevention of septicemia and anemia associated with myelosuppression in mice: risk of sustained expression inducing myelofibrosis due to immunosuppression. Gene Ther 1998; 5:497.
  70. Yan XQ, Lacey D, Fletcher F, et al. Chronic exposure to retroviral vector encoded MGDF (mpl-ligand) induces lineage-specific growth and differentiation of megakaryocytes in mice. Blood 1995; 86:4025.
  71. 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.
  72. Wang JC, Chen C, Lou LH, Mora M. Blood thrombopoietin, IL-6 and IL-11 levels in patients with agnogenic myeloid metaplasia. Leukemia 1997; 11:1827.
  73. Wang JC, Chen C, Novetsky AD, et al. Blood thrombopoietin levels in clonal thrombocytosis and reactive thrombocytosis. Am J Med 1998; 104:451.
  74. Moliterno AR, Hankins WD, Spivak JL. Impaired expression of the thrombopoietin receptor by platelets from patients with polycythemia vera. N Engl J Med 1998; 338:572.
  75. Sasaki A, Katoh O, Kawaishi K, et al. Expression of c-Mpl and c-Mpl ligand gene in hematopoietic cells of individuals with and without myeloproliferative disorders and leukemia cell lines. Int J Hematol 1995; 62:217.
  76. Le Bousse-Kerdilès MC, Chevillard S, Charpentier A, et al. Differential expression of transforming growth factor-beta, basic fibroblast growth factor, and their receptors in CD34+ hematopoietic progenitor cells from patients with myelofibrosis and myeloid metaplasia. Blood 1996; 88:4534.
  77. Xu M, Bruno E, Chao J, et al. The constitutive mobilization of bone marrow-repopulating cells into the peripheral blood in idiopathic myelofibrosis. Blood 2005; 105:1699.
  78. Silverstein MN. Agnogenic myeloid metaplasia, Publishing Sciences Group, Acton, MA 1975.
  79. O'Keane JC, Wolf BC, Neiman RS. The pathogenesis of splenic extramedullary hematopoiesis in metastatic carcinoma. Cancer 1989; 63:1539.
  80. Wolf BC, Neiman RS. Myelofibrosis with myeloid metaplasia: pathophysiologic implications of the correlation between bone marrow changes and progression of splenomegaly. Blood 1985; 65:803.
  81. Xu M, Bruno E, Chao J, et al. Constitutive mobilization of CD34+ cells into the peripheral blood in idiopathic myelofibrosis may be due to the action of a number of proteases. Blood 2005; 105:4508.
  82. Ferrant A, Rodhain J, Cauwe F, et al. Assessment of bone marrow and splenic erythropoiesis in myelofibrosis. Scand J Haematol 1982; 29:373.
  83. Mesa RA, Hanson CA, Rajkumar SV, et al. Evaluation and clinical correlations of bone marrow angiogenesis in myelofibrosis with myeloid metaplasia. Blood 2000; 96:3374.
  84. Boveri E, Passamonti F, Rumi E, et al. Bone marrow microvessel density in chronic myeloproliferative disorders: a study of 115 patients with clinicopathological and molecular correlations. Br J Haematol 2008; 140:162.
  85. Medinger M, Skoda R, Gratwohl A, et al. Angiogenesis and vascular endothelial growth factor-/receptor expression in myeloproliferative neoplasms: correlation with clinical parameters and JAK2-V617F mutational status. Br J Haematol 2009; 146:150.
  86. Thomas DA, Giles FJ, Albitar M, et al. Thalidomide therapy for myelofibrosis with myeloid metaplasia. Cancer 2006; 106:1974.
Topic 4526 Version 23.0

References

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