Acessibilidade / Reportar erro

Myeloproliferative neoplasms and the JAK/STAT signaling pathway: an overview

ABSTRACT

Myeloproliferative neoplasms are caused by a clonal proliferation of a hematopoietic progenitor. First described in 1951 as 'Myeloproliferative Diseases' and reevaluated by the World Health Organization classification system in 2011, myeloproliferative neoplasms include polycythemia vera, essential thrombocythemia and primary myelofibrosis in a subgroup called breakpoint cluster region-Abelson fusion oncogene-negative neoplasms. According to World Health Organization regarding diagnosis criteria for myeloproliferative neoplasms, the presence of the JAK2 V617F mutation is considered the most important criterion in the diagnosis of breakpoint cluster region-Abelson fusion oncogene-negative neoplasms and is thus used as a clonal marker. The V617F mutation in the Janus kinase 2(JAK2) gene produces an altered protein that constitutively activates the Janus kinase/signal transducers and activators of transcription pathway and other pathways downstream as a result of signal transducers and activators of transcription which are subsequently phosphorylated. This affects the expression of genes involved in the regulation of apoptosis and regulatory proteins and modifies the proliferation rate of hematopoietic stem cells.

Hematologic neoplasms; Janus kinase 2; STAT transcription factors; JAK2V617F mutation

Introduction

Myeloproliferative neoplasms (MPNs) are clonal disorders of hematopoietic stem cells, in which there is an increased proliferation of the myeloid lineage with effective maturation resulting in peripheral blood leukocytosis, and increased red blood cell mass; these neoplasms can progress to fibrosis or leukemic transformation. The MPNs are a group of diseases that include polycythemia vera (PV), essential thrombocythemia (ET) and primary myelofibrosis (PMF), which are collectively known as breakpoint cluster region-Abelson fusion oncogene (BCR-ABL)-negative neoplasms. These three diseases share common clinical features, such as proliferation of hematopoietic stem cells independent of growth factors, bone marrow hypercellularity, increased risk of thrombotic events and hemorrhages.1 Dameshek W. Editorial: some speculations on the myeloproliferative syndromes. Blood. 1951;6(4):372-5. , 2 Spivak JL, Barosi G, Tognoni G, Barbui T, Finazzi G, Marchioli R, et al. Chronic myeloproliferative disorders. Hematol Am Soc Hematol Educ Progr. 2003;2003(1):200-24. , 3 Finazzi G, Barbui T. How I treat patients with polycythemia vera. Blood. 2007;109(12):5104-11. and 4 Haferlach T, Bacher U, Kern W, Schnittger S, Haferlach C. The diagnosis of BCR/ABL- negative chronic myeloproliferative diseases (CMPD): a comprehensive approach based on morphology, cytogenetics, and molecular markers. Ann Hematol. 2008;87(1):1-10.

Studies carried out in 2005 identified abnormalities in the functioning of the Janus kinase 2 (JAK2) protein which is expressed by the JAK2 gene. Here, a sense mutation at exon 14 of the JAK2 gene (g.1849 G>T) 5 den Dunnen JT, Antonarakis SE. Mutation nomenclature extensions and suggestions to describe complex mutations: a discussion. Hum Mutat. 2000;15(1):7-12. and 6 Vainchenker W, Delhommeau F, Constantinescu SN, Bernard OA. New mutations and pathogenesis of myeloproliferative neoplasms. Review article. New mutations and pathogenesis of myeloproliferative neoplasms. Blood. 2011;118(7):1723-35. results in the substitution of a valine for a phenylalanine at position 617 located in the pseudokinase JH2 domain. It is responsible for constitutively activating the JAK/signal transducers and activators of transcription (STAT) signaling pathway and other pathways downstream. The JAK2 V617F mutation occurs at a frequency of 95% in PV and of 55-60% in ET and PMF. 5 den Dunnen JT, Antonarakis SE. Mutation nomenclature extensions and suggestions to describe complex mutations: a discussion. Hum Mutat. 2000;15(1):7-12. , 6 Vainchenker W, Delhommeau F, Constantinescu SN, Bernard OA. New mutations and pathogenesis of myeloproliferative neoplasms. Review article. New mutations and pathogenesis of myeloproliferative neoplasms. Blood. 2011;118(7):1723-35. , 7 Vannucchi AM, Antonioli E, Guglielmelli P, Rambaldi A, Barosi G, Marchioli R, et al. Clinical profile of homozygous JAK2 617V>F mutation in patients with polycythemia vera or essential thrombocythemia. Blood. 2007;110(3):840-6. , 8 Tefferi A. The history of myeloproliferative disorders: before and after Dameshek. Leukemia. 2008;22(1):3-13. , 9 Jones AV, Chase A, Silver RT, Oscier D, Zoi K, Wang YL, et al. JAK2 haplotype is a major risk factor for the development of myeloproliferative neoplasms. Nat Genet. 2009;41(4):446-9. and 1010  Gäbler K, Behrmann I, Haan C. JAK2 mutants (e.g., JAK2V617F) and their importance as drug targets in myeloproliferative neoplasms. JAKSTAT. 2013;2(3):e25025.

Myeloproliferative neoplasms

Dameshek1 Dameshek W. Editorial: some speculations on the myeloproliferative syndromes. Blood. 1951;6(4):372-5. carried out the first classification of Myeloproliferative Diseases in 1951. However, the World Health Organization (WHO) classification system published in 2011 introduced a new classification of tumors of hematopoietic and lymphoid tissue and changed the term to 'myeloproliferative neoplasms'. The MPNs include chronic myeloid leukemia (CML), PV, PMF, ET, chronic neutrophilic leukemia (CNL), chronic eosinophilic leukemia (CEL), mast cell disease (MCD) and unclassified myeloproliferative neoplasms (MPN-u).1111  Vardiman JW, Thiele J, Arber DA, Brunning RD, Borowitz MJ, Porwit A, et al. The 2008 revision of the World Health Organization (WHO) classification of myeloid neoplasms and acute leukemia: rationale and important changes. Blood. 2009;114(5):937-51. , 1212  Kiladjian JJ. The spectrum of JAK2 -positive myeloproliferative neoplasms. Hematol Am Soc Hematol Educ Progr. 2012;2012:561-6. and 1313  Kim HR, Choi HJ, Kim YK, Kim HJ, Shin JH, Suh SP, et al. Allelic expression imbalance of JAK2 V617F mutation in BCR-ABL- negative myeloproliferative neoplasms. PLoS ONE. 2013;8(1):e52518. CML is characterized by the presence of the Philadelphia chromosome (Ph) resulting from a translocation between chromosomes 9 and 22. The resulting hybrid gene, BCR-ABL, encodes a protein of 210 kDa, displaying tyrosine kinase activity and affects growth and cell differentiation.1414  Chauffaille ML. Neoplasias mieloproliferativas: revisão dos critérios diagnósticos e dos aspectos clínicos. Rev Bras Hematol Hemoter. 2010;32(4):308-16.

In addition to their common features, PV is characterized primarily by an increase in red blood cell production, ET for excessive production of platelets, and PMF for morphological changes in megakaryocytes and the development of monocytes that lead to the secretion of angiogenic factors responsible for promoting fibrosis in the bone marrow.1515  Quintás-Cardama A. The role of Janus kinase 2 (JAK2) in myeloproliferative neoplasms: therapeutic implications. Leuk Res. 2013;37(4):465-72.

Table 1 presents the MPN types, the incidences and clinical aspects of each.

Table 1
- Myeloproliferative neoplasms.

In addition to the JAK2 gene, works carried out since 2005 have found other mutations in candidate genes, including the myeloproliferative leukemia (MPL), ten-eleven translocation 2(TET2) a member of the TET oncogene family, additional sex comb-like 1 (ASXL1), Casitas B-lineage lymphoma (CBL), isocitrate dehydrogenase(IDH) and IKAROS family zinc finger 1(IKZF1) genes, all identified within the group of BCR-ABL-negative neoplasms. Mutations in the JAK2 and MPL encoder genes seem to have greater effect and are more associated with PV, ET and PMF with frequencies around 95%, 55% and 60% for JAK2 and 0%, 0.3% and 10% for MPL. 1616  Freitas RM, Santos MO, Maranduba CM. The JAK2 gene as a protagonist in chronic myeloproliferative neoplasms. Rev Bras Hematol Hemoter. 2013;35(4):278-9.

The functional consequences of these mutations in MPNs include disruption of the JAK/STAT signaling pathway, epigenetic modulation of transcription as well as abnormal accumulation of oncoproteins. However, it is still unclear how all these abnormalities contribute toward disease onset and clonal evolution of cells.1616  Freitas RM, Santos MO, Maranduba CM. The JAK2 gene as a protagonist in chronic myeloproliferative neoplasms. Rev Bras Hematol Hemoter. 2013;35(4):278-9.

Pathogenesis of myeloproliferative neoplasms

In the literature, some evidence suggests that phenotypic characteristics observed in MPNs are caused due to disorders in the hematopoietic cell signaling process. It has been reported that hematopoietic progenitors are hypersensitive to several growth factors in PV, ET and PMF,1717  Axelrad AA, Eskinazi D, Correa PN, Amato D. Hypersensitivity of circulating progenitor cells to megakaryocyte growth and development factor (PEG-rHu MGDF) in essential thrombocythemia. Blood. 2000;96(10):3310-21. and that abnormal myeloproliferation in patients with MPNs originates from constitutive activation of signal transduction pathways. These are caused by genetic rearrangements or mutations that affect tyrosine kinases or associated molecules.1818  Röder S, Steimle C, Meinhardt G, Pahl HL. STAT3 is constitutively active in some patients with Polycythemia rubra vera. Exp Hematol. 2001;29(6):694-702. Based on this and other evidence, some research groups started to work with the hypothesis that alterations in the function of the JAK2 protein could be related to MPNs, since this protein is mainly responsible for the activation of molecules involved in cell signaling processes and red blood cell production.1919  Kaushansky K. The chronic myeloproliferative disorders and mutation of JAK2: Dameshek's 54 year old speculation comes of age. Best Pract Res Clin Haematol. 2007;20(1):5-12.

The Janus kinase 2 gene

The JAK2 gene, located on chromosome 9p24 in humans, includes 25 exons encoding a protein of about 1132 amino acids. This protein is named JAK2 and it is a member of the Janus kinase (JAK) family. Four members of this family have already been identified: JAK1, JAK2, JAK3 and Tyk2. 2020  Yamaoka K, Saharinen P, Pesu M, Holt VE III, Silvennoinen O, O'Shea JJ. The Janus kinases (Jaks). Genome Biol. 2004;5(12):253-6.

In 2005, the discovery of the JAK2 mutation (JAK2 V617F) expanded the knowledge about the pathogenesis of BCR-ABL-negative neoplasms. A missense mutation at position g.1849 with a guanine-thymine transversion (g.1849 G>T)5 den Dunnen JT, Antonarakis SE. Mutation nomenclature extensions and suggestions to describe complex mutations: a discussion. Hum Mutat. 2000;15(1):7-12. and 6 Vainchenker W, Delhommeau F, Constantinescu SN, Bernard OA. New mutations and pathogenesis of myeloproliferative neoplasms. Review article. New mutations and pathogenesis of myeloproliferative neoplasms. Blood. 2011;118(7):1723-35. was identified in exon 14 of the JAK2 gene. This mutation results in the substitution of valine for phenylalanine at amino acid position p.617, 5 den Dunnen JT, Antonarakis SE. Mutation nomenclature extensions and suggestions to describe complex mutations: a discussion. Hum Mutat. 2000;15(1):7-12. affecting the pseudokinase domain (JH2) of the protein. The JAK2 V617F mutation is present in most patients with PV and in a subgroup of patients with ET and PMF (~95% in PV, 50-60% in ET and 50-60% in PMF). 5 den Dunnen JT, Antonarakis SE. Mutation nomenclature extensions and suggestions to describe complex mutations: a discussion. Hum Mutat. 2000;15(1):7-12. , 6 Vainchenker W, Delhommeau F, Constantinescu SN, Bernard OA. New mutations and pathogenesis of myeloproliferative neoplasms. Review article. New mutations and pathogenesis of myeloproliferative neoplasms. Blood. 2011;118(7):1723-35. , 7 Vannucchi AM, Antonioli E, Guglielmelli P, Rambaldi A, Barosi G, Marchioli R, et al. Clinical profile of homozygous JAK2 617V>F mutation in patients with polycythemia vera or essential thrombocythemia. Blood. 2007;110(3):840-6. , 8 Tefferi A. The history of myeloproliferative disorders: before and after Dameshek. Leukemia. 2008;22(1):3-13. , 9 Jones AV, Chase A, Silver RT, Oscier D, Zoi K, Wang YL, et al. JAK2 haplotype is a major risk factor for the development of myeloproliferative neoplasms. Nat Genet. 2009;41(4):446-9. and 1919  Kaushansky K. The chronic myeloproliferative disorders and mutation of JAK2: Dameshek's 54 year old speculation comes of age. Best Pract Res Clin Haematol. 2007;20(1):5-12.

It has been shown that the JAK2 V617F mutation causes genetic instability in gene expression by inducing changes in the chromatin structure and by reducing the apoptotic response to DNA damage. These are mechanisms that can increase the accumulation of genetic lesions leading to malignant transformations.1212  Kiladjian JJ. The spectrum of JAK2 -positive myeloproliferative neoplasms. Hematol Am Soc Hematol Educ Progr. 2012;2012:561-6. Besides the g.1849 G > T (p.V617F) mutation,5 den Dunnen JT, Antonarakis SE. Mutation nomenclature extensions and suggestions to describe complex mutations: a discussion. Hum Mutat. 2000;15(1):7-12. and 6 Vainchenker W, Delhommeau F, Constantinescu SN, Bernard OA. New mutations and pathogenesis of myeloproliferative neoplasms. Review article. New mutations and pathogenesis of myeloproliferative neoplasms. Blood. 2011;118(7):1723-35. other mutations were found in exon 12 in 2007 namely: p.N542-543del, p.E543-D544del, p.K539L, p.F537-K539delinsL, p.H538-p.K539delinsL, p.H538QK539L, p.V536-1546dup11, p.F537-1546dup10+F547L, p.R541 E543delinsK-and-p.I540 E543delinsMK.2121  Cazzola M. Somatic mutations of JAK2 exon 12 as a molecular basis of erythrocytosis. Haematologica. 2007;92(12):1585-9. , 2222  Williams DM, Kim AH, Rogers O, Spivak JL, Moliterno AR. Phenotypic variations and new mutations in JAK2 V617F- negative polycythemia vera, erythrocytosis, and idiopathic myelofibrosis. Exp Hematol. 2007;35(11):1641-6. and 2323  Laughlin TS, Moliterno AR, Stein BL, Rothberg PG. Detection of exon 12 mutations in the JAK2 gene enhanced analytical sensitivity using clamped PCR and nucleotide sequencing. J Mol Diagn. 2010;12(3):278-82. These were found to show low frequencies of about 3-4% in PV patients. There are valid techniques that detect these mutations and their active search is justified not only due to the absence of the exon 14 mutation, but also in patients that have erythrocytosis and in those with low levels of erythropoietin.2424  Kjær L, Westman M, Hasselbalch Riley C, Høgdall E, Weis Bjerrum O, Hasselbalch H. A highly sensitive quantitative real-time PCR assay for determination of mutant JAK2 exon 12 allele burden. PLoS ONE. 2012;7(3):e33100.

The Janus kinase 2 protein

Members of the JAK2 protein family have seven homologous domains (JH) that are numbered from 1 to 7. The JH1 domain acts as a kinase domain, which contains the adenosine triphosphate (ATP)-binding and signaling pathway activation regions. The JH2 domain is a pseudokinase that is homologous to the JH1 domain. It lacks catalytic activity due to a substitution of an aspartate residue in the catalytic loop (His-Arg-Asp - HRD motif). JH2 has an inhibitory function that regulates both the basal activity of JAK and the cytokine-induced activation by interacting with the active site, JH1, blocking ATP and/or substrates binding to the catalytic site.2525  Schafer AI. Molecular basis of the diagnosis and treatment of polycythemia vera and essential thrombocythemia. Blood. 2006;107(11):4214-22. , 2626  Smith CA, Fan G. The saga of JAK2 mutations and translocations in hematologic disorders: pathogenesis, diagnostic and therapeutic prospects, and revised World Health Organization diagnostic criteria for myeloproliferative neoplasms. Hum Pathol. 2008;39(6):795-810. and 2727  Ungureanu D, Wu J, Pekkala T, Niranjan Y, Young C, Jensen ON, et al. The pseudokinase domain of JAK2 is a dual-specificity protein kinase that negatively regulates cytokine signaling. Nat Struct Mol Biol. 2011;18(9):971-6.

Mutations in exon 12 of JAK2 are found in the binding region of the Src homology 2 (SH2) and JH2 domains. Although not directly located in the JH2 domain, these mutations may modify the structure of the pseudokinase domain, inducing erythropoietin hypersensitivity similar to that caused by the JAK2 p.V617F mutation.5 den Dunnen JT, Antonarakis SE. Mutation nomenclature extensions and suggestions to describe complex mutations: a discussion. Hum Mutat. 2000;15(1):7-12. , 9 Jones AV, Chase A, Silver RT, Oscier D, Zoi K, Wang YL, et al. JAK2 haplotype is a major risk factor for the development of myeloproliferative neoplasms. Nat Genet. 2009;41(4):446-9. , 2828  Lakey MA, Pardanani A, Hoyer JD, Nguyen PL, Lasho TL, Tefferi A, et al. Bone marrow morphologic features in polycythemia vera with JAK2 exon 12 mutations. Am J Clin Pathol. 2010;133(6):942-8. and 2929  Ikeda K, Ogawa K, Takeishi Y. The role of HMGA2 in the proliferation and expansion of a hematopoietic cell in myeloproliferative neoplasms. Fukushima J Med Sci. 2012;58(2):91-100.

The Janus kinase 2/signal transducers and activators of transcription signaling pathway

SIGNALING RECEPTORS:

Cytokines from the hematopoietic system include interleukins (ILs), colony stimulating factors (CSFs), interferon (INF), erythropoietin (EPO) and thrombopoietin (TPO) all of which activate the signal transduction of the JAK/STAT signaling pathway.3030  Yoshimura A, Naka T, Kubo M. SOCS proteins, cytokine signalling and immune regulation. Nat Rev Immunol. 2007;7(6):454-65. and 3131  Tamiya T, Kashiwagi I, Takahashi R, Yasukawa H, Yoshimura A. Suppressors of cytokine signaling (SOCS) proteins and JAK/STAT pathways: regulation of T-cell inflammation by SOCS1 and SOCS3. Arterioscler Thromb Vasc Biol. 2011;31(5):980-5. Most of these receptors are part of a type I-homodimer receptor family, that includes the erythropoietin (EPO-R) and thrombopoietin (TPO-R) receptors.3232  Morgan KJ, Gilliland DG. A role for JAK2 mutations in myeloproliferative diseases. Annu Rev Med. 2008;59:213-22.

The type I receptor is specifically expressed during erythroid and myeloid differentiation and its expression may account for the preferential activity of JAK2 V617F over myeloid lineage cells rather than lymphoid lineage cells, which lack this type of receptor.3232  Morgan KJ, Gilliland DG. A role for JAK2 mutations in myeloproliferative diseases. Annu Rev Med. 2008;59:213-22.

Janus kinases

JAKs are associated with the intracellular domains of cytokine receptors; they show intrinsic kinase activity mediated through their band-4.1 protein, ezrin, radixin, and moesin (FERM) and SH2 domains and are kept in an inactive state in the absence of specific receptor-activating ligands.3333  Tan AY, Westerman DA, Dobrovic A. A simple, rapid, and sensitive method for the detection of the JAK2 V617F mutation. Am J Clin Pathol. 2007;127(6):977-81. and 3434  Gari M. The role of Janus Kinase 2 (JAK2) in the pathologenesis of myeloproliferative disorders. J King Abdulaziz Univ Med Sci. 2009;16(1):3-19.

Cytokine binding leads to a conformational change of the receptor, cytoplasmically affecting associated JAKs, causing activation and phosphorylation. Phosphorylated tyrosine residues in JAKs act as binding sites for the SH2 domains in signaling molecules. Mutations in JAK2 generate constitutive activation of the JAK/STAT pathway, mainly in STAT3 and STAT5. In addition to 'hot spot' mutations, constitutive activation of tyrosine kinases also occurs by chromosomal translocation, deletion and tandem duplication, which are common pathogenic events in hematopoietic malignancies. 1010  Gäbler K, Behrmann I, Haan C. JAK2 mutants (e.g., JAK2V617F) and their importance as drug targets in myeloproliferative neoplasms. JAKSTAT. 2013;2(3):e25025. , 2727  Ungureanu D, Wu J, Pekkala T, Niranjan Y, Young C, Jensen ON, et al. The pseudokinase domain of JAK2 is a dual-specificity protein kinase that negatively regulates cytokine signaling. Nat Struct Mol Biol. 2011;18(9):971-6. , 3131  Tamiya T, Kashiwagi I, Takahashi R, Yasukawa H, Yoshimura A. Suppressors of cytokine signaling (SOCS) proteins and JAK/STAT pathways: regulation of T-cell inflammation by SOCS1 and SOCS3. Arterioscler Thromb Vasc Biol. 2011;31(5):980-5. , 3232  Morgan KJ, Gilliland DG. A role for JAK2 mutations in myeloproliferative diseases. Annu Rev Med. 2008;59:213-22. , 3333  Tan AY, Westerman DA, Dobrovic A. A simple, rapid, and sensitive method for the detection of the JAK2 V617F mutation. Am J Clin Pathol. 2007;127(6):977-81. , 3434  Gari M. The role of Janus Kinase 2 (JAK2) in the pathologenesis of myeloproliferative disorders. J King Abdulaziz Univ Med Sci. 2009;16(1):3-19. and 3535  Walz C, Cross NC, Van Etten RA, Reiter A. Comparison of mutated ABL1 and JAK2 as oncogenes and drug targets in myeloproliferative disorders. Leukemia. 2008;22(7):1320-34.

The STATs are located in the cytosol and migrate to the nucleus, regulating gene transcription only upon activation. Once associated with the phosphorylated sites of JAK, these STATs also phosphorylate and dimerize.2626  Smith CA, Fan G. The saga of JAK2 mutations and translocations in hematologic disorders: pathogenesis, diagnostic and therapeutic prospects, and revised World Health Organization diagnostic criteria for myeloproliferative neoplasms. Hum Pathol. 2008;39(6):795-810.

Dimerized STATs migrate toward the nucleus, where they act as transcription factors, activating or repressing genes that are important in cell proliferation and survival. Some of these genes include those that express cyclins as well as anti-apoptotic proteins.3636  Fiskus W, Ganguly S, Kambhampati S, Bhalla KN. Role of additional novel therapies in myeloproliferative neoplasms. Hematol Oncol Clin N Am. 2012;26(5):959-80. STAT dimers also activate genes that encode inhibitory proteins that contribute toward the termination of cellular response. Some of these proteins bind to phosphorylated JAKs and inactivate them, as well as their associated receptors. Others bind to phosphorylated STAT dimers and stop them from binding to their target gene.3737  Haricharan S, Li Y. STAT signaling in mammary gland differentiation, cell survival and tumorigenesis. Mol Cell Endocrinol. 2014;382(1):560-9.

JAK/STAT-mediated signaling control mechanisms include dephosphorylation of JAK and STAT. This is caused by an inhibition triggered by suppressors of cytokine signaling (SOCS) proteins, the lymphocyte adaptor protein (LNK also known as SH2B3) and CBL.3030  Yoshimura A, Naka T, Kubo M. SOCS proteins, cytokine signalling and immune regulation. Nat Rev Immunol. 2007;7(6):454-65. and 3232  Morgan KJ, Gilliland DG. A role for JAK2 mutations in myeloproliferative diseases. Annu Rev Med. 2008;59:213-22.

Negative regulation of the Janus kinase 2/signal transducers and activators of transcription pathway

As our knowledge about the JAK/STAT pathway has increased, important mechanisms for down-regulation of this pathway have been elucidated. These include removal of phosphates from cytokine receptors and activated STATs by tyrosine phosphatases, as well as by protein inhibitors of activated STAT (PIAS). PIAS inhibit transcriptional activation by binding and blocking access to target DNA. The next section will discuss three PIAS: SOCS, CBL and Lyme neuroborreliosis (LNB) proteins.

SOCS proteins are a family of cytokine signaling suppressors of the JAK/STAT pathway, characterized by the presence of an amino-terminal domain that is variable in size. It includes an inhibitory domain, a SH2 domain and a carboxy-terminal domain called SOCS box. Currently, there are eight members in this family, namely: SOCS1, SOCS2, SOCS3, SOCS4, SOCS5, SOCS6, SOCS7 and CIS. Under normal conditions, they are expressed at low levels in unstimulated cells while their expression increases when STATs are activated.3030  Yoshimura A, Naka T, Kubo M. SOCS proteins, cytokine signalling and immune regulation. Nat Rev Immunol. 2007;7(6):454-65. , 3131  Tamiya T, Kashiwagi I, Takahashi R, Yasukawa H, Yoshimura A. Suppressors of cytokine signaling (SOCS) proteins and JAK/STAT pathways: regulation of T-cell inflammation by SOCS1 and SOCS3. Arterioscler Thromb Vasc Biol. 2011;31(5):980-5. and 3838  Khwaja A. The role of Janus kinases in haemopoiesis and haematological malignancy. Br J Haematol. 2006;134(4):366-84.

SOCS can inhibit the JAK/STAT signaling through two mechanisms: (1) inhibition of JAK2 kinase activity by competing with the STATs SH2 domains for binding sites in the cytoplasmic domain of the receptor or (2) proteasomal degradation of signaling proteins by binding to the JH1 domain of JAK2, inhibiting the activation of all other JAK/STAT-associated pathways.3939  Hookham MB, Elliott J, Suessmuth Y, Staerk J, Ward AC, Vainchenker W, et al. The myeloproliferative disorder- associated JAK2 V617F mutant escapes negative regulation by suppressor of cytokine signaling 3. Blood. 2007;109(11):4924-9.

Some mutations in different SOCS have been identified in MPNs, but are rarely found. Hypermethylation of CpG islands in SOCS1 and SOCS3 is associated with a decrease in the expression of these regulators in p.V617F-positive PV and ET patients.5 den Dunnen JT, Antonarakis SE. Mutation nomenclature extensions and suggestions to describe complex mutations: a discussion. Hum Mutat. 2000;15(1):7-12. and 1010  Gäbler K, Behrmann I, Haan C. JAK2 mutants (e.g., JAK2V617F) and their importance as drug targets in myeloproliferative neoplasms. JAKSTAT. 2013;2(3):e25025.

Other important components of this negative regulatory complex include multifunctional proteins with ubiquitin ligase activity, such as CBL. CBL is usually involved in the negative feedback of tyrosine kinase receptors by competitive blocking signaling. It induces proteasomal degradation of tyrosine kinases activated by ubiquitination. All CBL have single domain proteins that recognizes phosphorylated tyrosine residues that are present on activated tyrosine kinases.1010  Gäbler K, Behrmann I, Haan C. JAK2 mutants (e.g., JAK2V617F) and their importance as drug targets in myeloproliferative neoplasms. JAKSTAT. 2013;2(3):e25025. and 4040  Naramura M, Nadeau S, Mohapatra B, Ahmad G, Mukhopadhyay C, Sattler M, et al. Mutant Cbl proteins as oncogenic drivers in myeloproliferative disorders. Oncotarget. 2011;2(3):245-50.

LNK, included in the SH2B protein family, negatively regulates the activity of JAK2 by binding to SH2-phosphorylated tyrosine. It also regulates the signaling of TPO-R and EPO-R, and it is detected in both JAK2 V617F-positive and -negative patients.1010  Gäbler K, Behrmann I, Haan C. JAK2 mutants (e.g., JAK2V617F) and their importance as drug targets in myeloproliferative neoplasms. JAKSTAT. 2013;2(3):e25025. Increased LNK deficiency increases the oncogenic ability of JAK2 to expand myeloid progenitors in vitro and in vivo. 4141  Jiang Q, Song C, Nangreave J, Liu X, Lin L, Qiu D, et al. DNA origami as a carrier for circumvention of drug resistance. J Am Chem Soc. 2012;134(32):13396-403.

Mutation in the Janus kinase 2 gene and changes in protein function

In addition to activating the JAK/STAT pathway, the p.V617F mutation5 den Dunnen JT, Antonarakis SE. Mutation nomenclature extensions and suggestions to describe complex mutations: a discussion. Hum Mutat. 2000;15(1):7-12. also constitutively activates the rat sarcoma (RAS)/Mitogen-activated protein kinases (MAPK) and phosphoinositide 3-kinase (PI3K)/protein kinase B (PKB, aka Akt) pathways, which results in an increased expression of mitotic proteins, regulatory proteins from the cellular cycle [cyclins D1, CDC25A (cell division cycle)] and anti-apoptotic genes [B-cell lymphoma-extra-large (Bcl-XL) and Bcl2]. These abnormalities involving the JAK/STAT and related pathways result in proliferation of affected cells (Figure 1).1010  Gäbler K, Behrmann I, Haan C. JAK2 mutants (e.g., JAK2V617F) and their importance as drug targets in myeloproliferative neoplasms. JAKSTAT. 2013;2(3):e25025.

Figure 1
- Signaling pathways activated by cytokine receptor binding. PI3K, Akt and STAT are activated after the phosphorylation of JAK2. The active negative feedback regulatory proteins complete the activation pathways. Such control is ineffective in the presence of a mutation in the JH2 domain of JAK2. PI3K: phosphoinositide 3-kinase; Akt: protein kinase B; STAT: signal transducers and activators of transcription; JAK2: Janus kinase 2; EPO-R: erythropoietin receptor; SOCS: suppressors of cytokine signaling; Bcl-XL: B-cell lymphoma-extra-large; Myc: myelocytomatosis oncogene.

In mutated cells, down-regulation becomes inadequate and/or inefficient. SH2 domains, normally present in PIAS proteins, cannot dephosphorylate JAK2. In addition, the anti-apoptotic mechanisms are expressed at much higher levels than normal. An acquired mutation in one of these regulatory proteins can trigger an exacerbated activation of STAT, even in the absence of specific mutations in JAK2.4242  Rawlings JS, Rosler KM, Harrison DA. The JAK/STAT signaling pathway. J Cell Sci. 2004;117 Pt 8:1281-3.

Additional molecular events appear to interfere with the activity of JAK2, such as cytogenetics, which increase the kinase activity of the protein, the regulatory ability of phosphorylases and SOCS proteins, polymorphisms, and mutations in cytokine receptors.4343  Perrotta S, Cucciolla V, Ferraro M, Ronzoni L, Tramontano A, Rossi F, et al. EPO receptor gain-of-function causes hereditary polycythemia, alters CD34+ cell differentiation and increases circulating endothelial precursors. PLoS ONE. 2010;5(8):e12015.

Questions about the different phenotypes in BCR-ABL-negative myeloproliferative neoplasms and the V617F mutation

A further intriguing question for researchers regarding MPNs is how the same mutation can trigger three phenotypically different diseases. There is still no clear understanding of the molecular mechanisms involved in the process, just hypotheses. One of them is the dose-response gene hypothesis, which is associated with allele load. Another is that there may be epigenetic changes that predispose the patient's organism to develop certain MPNs as well as changes in genes specific in the development of malignancies, as well as mutations in other signaling pathways that might affect signaling in regulatory proteins.1616  Freitas RM, Santos MO, Maranduba CM. The JAK2 gene as a protagonist in chronic myeloproliferative neoplasms. Rev Bras Hematol Hemoter. 2013;35(4):278-9.

For Kralovics et al., Plo et al., Smith & Fan, Cross and Pagliarini,2626  Smith CA, Fan G. The saga of JAK2 mutations and translocations in hematologic disorders: pathogenesis, diagnostic and therapeutic prospects, and revised World Health Organization diagnostic criteria for myeloproliferative neoplasms. Hum Pathol. 2008;39(6):795-810. , 4444  Kralovics R. Genetic complexity of myeloproliferative neoplasms. Leukemia. 2008;22(10):1841-8. , 4545  Plo I, Nakatake M, Malivert L, de Villartay JP, Giraudier S, Villeval JL, et al. JAK2 stimulates homologous recombination and genetic instability: potential implication in the heterogeneity of myeloproliferative disorders. Blood. 2008;112(4):1402-12. , 4646  Cross NC. Genetic and epigenetic complexity in myeloproliferative neoplasms. Hematol Am Soc Hematol Educ Progr. 2011;2011:208-14. and 4747  Pagliarini-e-Silva S, Santos BC, Pereira EM, Ferreira ME, Baraldi EC, Sell AM, et al. Evaluation of the association between the JAK2 46/1 haplotype and chronic myeloproliferative neoplasms in a Brazilian population. Clinics (Sao Paulo). 2013;68(1):5-9. the phenotype of the disease is related to the concept of allelic load. Thus, low levels of mutant alleles would result in ET phenotype, while higher levels of mutated alleles would lead to a phenotype similar to PV. These higher levels of mutated alleles in hematopoietic stem cells could be associated with the patient's homozygous condition, caused by loss of heterozygosity, as explained by the theory of uniparental disomy.4848  Scott LM, Rebel VI. JAK2 and genomic instability in the myeloproliferative neoplasms: a case of the chicken or the egg? Am J Hematol. 2012;87(11):1028-36.

Presumably, homozygosity confers a proliferative advantage over other cells, which present only one mutated allele, increasing the activity level of the tyrosine kinase generated by the mutated JAK2. Patients with ET and PMF are mostly heterozygous and seldom show homozygosity.4848  Scott LM, Rebel VI. JAK2 and genomic instability in the myeloproliferative neoplasms: a case of the chicken or the egg? Am J Hematol. 2012;87(11):1028-36.

Besides the association between phenotype and homozygosity/heterozygosity, there are other hypotheses that have attempted to correlate the different phenotypes observed related to the JAK2 V617F mutation.

In their study, Plo et al.4545  Plo I, Nakatake M, Malivert L, de Villartay JP, Giraudier S, Villeval JL, et al. JAK2 stimulates homologous recombination and genetic instability: potential implication in the heterogeneity of myeloproliferative disorders. Blood. 2008;112(4):1402-12. discuss the gene-dose hypothesis, which postulates that the JAK2 mutation can be a starting point for the three pathologies. They also discuss the existence of other genetic events that can modify the JAK2 kinase activity, which may explain the heterogeneity present in classic MPNs. Several mechanisms have been proposed to aid the understanding of loss of heterozygosity and/or the presence of new genetic abnormalities, including the double-stranded break repair mechanisms in DNA, homologous recombination and non-homologous end joining. All these lead to genomic instability, which highlights how essential the precise regulation of these mechanisms is for the maintenance of genome stability and diversity.4949  Kilpivaara O, Levine RL. JAK2 and MPL mutations in myeloproliferative neoplasms: discovery and science. Leukemia. 2008;22(10):1813-7. , 5050  Passamonti F, Rumi E. Clinical relevance of JAK2 (V617F) mutant allele burden. Haematologica. 2009;94(1):7-10. and 5151  Shih AH, Abdel- Wahab O, Patel JP, Levine RL. The role of mutations in epigenetic regulators in myeloid malignancies. Nat Rev Cancer. 2012;12(9):599-612.

According to Mascarenhas et al.5252  Mascarenhas J, Roper N, Chaurasia P, Hoffman R. Epigenetic abnormalities in myeloproliferative neoplasms: a target for novel therapeutic strategies. Clin Epigenet. 2011;2(2):197-212. biological events that lead to the initiation and progression of MPNs are linked not only to acquisition of genetic mutations such as JAK2 V617F. They are also due to epigenetic changes that do not affect the primary DNA sequence, but the gene expression responsible for chromatin remodeling. Deregulation of modulation processes between nucleosomes may result in silencing of tumor suppressor as well as differentiation genes, promoting cell survival by blocking apoptosis and senescence, contributing to malignant transformation. The researchers also highlighted two categories of epigenetic alterations in MPNs: (1) changes in the genes that encode proteins involved in remodeling of the chromatin structure, such as the TET2, ASXL1, JAK2 genes, among others, and (2) changes in the promoter site of genes essential for cell survival, differentiation and proliferation. Besides its role in signaling, the JAK2 V617F mutation also affects chromatin structure by blocking the recruitment of repressor proteins. 9 Jones AV, Chase A, Silver RT, Oscier D, Zoi K, Wang YL, et al. JAK2 haplotype is a major risk factor for the development of myeloproliferative neoplasms. Nat Genet. 2009;41(4):446-9.

References

  • 1
    Dameshek W. Editorial: some speculations on the myeloproliferative syndromes. Blood. 1951;6(4):372-5.
  • 2
    Spivak JL, Barosi G, Tognoni G, Barbui T, Finazzi G, Marchioli R, et al. Chronic myeloproliferative disorders. Hematol Am Soc Hematol Educ Progr. 2003;2003(1):200-24.
  • 3
    Finazzi G, Barbui T. How I treat patients with polycythemia vera. Blood. 2007;109(12):5104-11.
  • 4
    Haferlach T, Bacher U, Kern W, Schnittger S, Haferlach C. The diagnosis of BCR/ABL- negative chronic myeloproliferative diseases (CMPD): a comprehensive approach based on morphology, cytogenetics, and molecular markers. Ann Hematol. 2008;87(1):1-10.
  • 5
    den Dunnen JT, Antonarakis SE. Mutation nomenclature extensions and suggestions to describe complex mutations: a discussion. Hum Mutat. 2000;15(1):7-12.
  • 6
    Vainchenker W, Delhommeau F, Constantinescu SN, Bernard OA. New mutations and pathogenesis of myeloproliferative neoplasms. Review article. New mutations and pathogenesis of myeloproliferative neoplasms. Blood. 2011;118(7):1723-35.
  • 7
    Vannucchi AM, Antonioli E, Guglielmelli P, Rambaldi A, Barosi G, Marchioli R, et al. Clinical profile of homozygous JAK2 617V>F mutation in patients with polycythemia vera or essential thrombocythemia. Blood. 2007;110(3):840-6.
  • 8
    Tefferi A. The history of myeloproliferative disorders: before and after Dameshek. Leukemia. 2008;22(1):3-13.
  • 9
    Jones AV, Chase A, Silver RT, Oscier D, Zoi K, Wang YL, et al. JAK2 haplotype is a major risk factor for the development of myeloproliferative neoplasms. Nat Genet. 2009;41(4):446-9.
  • 10
    Gäbler K, Behrmann I, Haan C. JAK2 mutants (e.g., JAK2V617F) and their importance as drug targets in myeloproliferative neoplasms. JAKSTAT. 2013;2(3):e25025.
  • 11
    Vardiman JW, Thiele J, Arber DA, Brunning RD, Borowitz MJ, Porwit A, et al. The 2008 revision of the World Health Organization (WHO) classification of myeloid neoplasms and acute leukemia: rationale and important changes. Blood. 2009;114(5):937-51.
  • 12
    Kiladjian JJ. The spectrum of JAK2 -positive myeloproliferative neoplasms. Hematol Am Soc Hematol Educ Progr. 2012;2012:561-6.
  • 13
    Kim HR, Choi HJ, Kim YK, Kim HJ, Shin JH, Suh SP, et al. Allelic expression imbalance of JAK2 V617F mutation in BCR-ABL- negative myeloproliferative neoplasms. PLoS ONE. 2013;8(1):e52518.
  • 14
    Chauffaille ML. Neoplasias mieloproliferativas: revisão dos critérios diagnósticos e dos aspectos clínicos. Rev Bras Hematol Hemoter. 2010;32(4):308-16.
  • 15
    Quintás-Cardama A. The role of Janus kinase 2 (JAK2) in myeloproliferative neoplasms: therapeutic implications. Leuk Res. 2013;37(4):465-72.
  • 16
    Freitas RM, Santos MO, Maranduba CM. The JAK2 gene as a protagonist in chronic myeloproliferative neoplasms. Rev Bras Hematol Hemoter. 2013;35(4):278-9.
  • 17
    Axelrad AA, Eskinazi D, Correa PN, Amato D. Hypersensitivity of circulating progenitor cells to megakaryocyte growth and development factor (PEG-rHu MGDF) in essential thrombocythemia. Blood. 2000;96(10):3310-21.
  • 18
    Röder S, Steimle C, Meinhardt G, Pahl HL. STAT3 is constitutively active in some patients with Polycythemia rubra vera. Exp Hematol. 2001;29(6):694-702.
  • 19
    Kaushansky K. The chronic myeloproliferative disorders and mutation of JAK2: Dameshek's 54 year old speculation comes of age. Best Pract Res Clin Haematol. 2007;20(1):5-12.
  • 20
    Yamaoka K, Saharinen P, Pesu M, Holt VE III, Silvennoinen O, O'Shea JJ. The Janus kinases (Jaks). Genome Biol. 2004;5(12):253-6.
  • 21
    Cazzola M. Somatic mutations of JAK2 exon 12 as a molecular basis of erythrocytosis. Haematologica. 2007;92(12):1585-9.
  • 22
    Williams DM, Kim AH, Rogers O, Spivak JL, Moliterno AR. Phenotypic variations and new mutations in JAK2 V617F- negative polycythemia vera, erythrocytosis, and idiopathic myelofibrosis. Exp Hematol. 2007;35(11):1641-6.
  • 23
    Laughlin TS, Moliterno AR, Stein BL, Rothberg PG. Detection of exon 12 mutations in the JAK2 gene enhanced analytical sensitivity using clamped PCR and nucleotide sequencing. J Mol Diagn. 2010;12(3):278-82.
  • 24
    Kjær L, Westman M, Hasselbalch Riley C, Høgdall E, Weis Bjerrum O, Hasselbalch H. A highly sensitive quantitative real-time PCR assay for determination of mutant JAK2 exon 12 allele burden. PLoS ONE. 2012;7(3):e33100.
  • 25
    Schafer AI. Molecular basis of the diagnosis and treatment of polycythemia vera and essential thrombocythemia. Blood. 2006;107(11):4214-22.
  • 26
    Smith CA, Fan G. The saga of JAK2 mutations and translocations in hematologic disorders: pathogenesis, diagnostic and therapeutic prospects, and revised World Health Organization diagnostic criteria for myeloproliferative neoplasms. Hum Pathol. 2008;39(6):795-810.
  • 27
    Ungureanu D, Wu J, Pekkala T, Niranjan Y, Young C, Jensen ON, et al. The pseudokinase domain of JAK2 is a dual-specificity protein kinase that negatively regulates cytokine signaling. Nat Struct Mol Biol. 2011;18(9):971-6.
  • 28
    Lakey MA, Pardanani A, Hoyer JD, Nguyen PL, Lasho TL, Tefferi A, et al. Bone marrow morphologic features in polycythemia vera with JAK2 exon 12 mutations. Am J Clin Pathol. 2010;133(6):942-8.
  • 29
    Ikeda K, Ogawa K, Takeishi Y. The role of HMGA2 in the proliferation and expansion of a hematopoietic cell in myeloproliferative neoplasms. Fukushima J Med Sci. 2012;58(2):91-100.
  • 30
    Yoshimura A, Naka T, Kubo M. SOCS proteins, cytokine signalling and immune regulation. Nat Rev Immunol. 2007;7(6):454-65.
  • 31
    Tamiya T, Kashiwagi I, Takahashi R, Yasukawa H, Yoshimura A. Suppressors of cytokine signaling (SOCS) proteins and JAK/STAT pathways: regulation of T-cell inflammation by SOCS1 and SOCS3. Arterioscler Thromb Vasc Biol. 2011;31(5):980-5.
  • 32
    Morgan KJ, Gilliland DG. A role for JAK2 mutations in myeloproliferative diseases. Annu Rev Med. 2008;59:213-22.
  • 33
    Tan AY, Westerman DA, Dobrovic A. A simple, rapid, and sensitive method for the detection of the JAK2 V617F mutation. Am J Clin Pathol. 2007;127(6):977-81.
  • 34
    Gari M. The role of Janus Kinase 2 (JAK2) in the pathologenesis of myeloproliferative disorders. J King Abdulaziz Univ Med Sci. 2009;16(1):3-19.
  • 35
    Walz C, Cross NC, Van Etten RA, Reiter A. Comparison of mutated ABL1 and JAK2 as oncogenes and drug targets in myeloproliferative disorders. Leukemia. 2008;22(7):1320-34.
  • 36
    Fiskus W, Ganguly S, Kambhampati S, Bhalla KN. Role of additional novel therapies in myeloproliferative neoplasms. Hematol Oncol Clin N Am. 2012;26(5):959-80.
  • 37
    Haricharan S, Li Y. STAT signaling in mammary gland differentiation, cell survival and tumorigenesis. Mol Cell Endocrinol. 2014;382(1):560-9.
  • 38
    Khwaja A. The role of Janus kinases in haemopoiesis and haematological malignancy. Br J Haematol. 2006;134(4):366-84.
  • 39
    Hookham MB, Elliott J, Suessmuth Y, Staerk J, Ward AC, Vainchenker W, et al. The myeloproliferative disorder- associated JAK2 V617F mutant escapes negative regulation by suppressor of cytokine signaling 3. Blood. 2007;109(11):4924-9.
  • 40
    Naramura M, Nadeau S, Mohapatra B, Ahmad G, Mukhopadhyay C, Sattler M, et al. Mutant Cbl proteins as oncogenic drivers in myeloproliferative disorders. Oncotarget. 2011;2(3):245-50.
  • 41
    Jiang Q, Song C, Nangreave J, Liu X, Lin L, Qiu D, et al. DNA origami as a carrier for circumvention of drug resistance. J Am Chem Soc. 2012;134(32):13396-403.
  • 42
    Rawlings JS, Rosler KM, Harrison DA. The JAK/STAT signaling pathway. J Cell Sci. 2004;117 Pt 8:1281-3.
  • 43
    Perrotta S, Cucciolla V, Ferraro M, Ronzoni L, Tramontano A, Rossi F, et al. EPO receptor gain-of-function causes hereditary polycythemia, alters CD34+ cell differentiation and increases circulating endothelial precursors. PLoS ONE. 2010;5(8):e12015.
  • 44
    Kralovics R. Genetic complexity of myeloproliferative neoplasms. Leukemia. 2008;22(10):1841-8.
  • 45
    Plo I, Nakatake M, Malivert L, de Villartay JP, Giraudier S, Villeval JL, et al. JAK2 stimulates homologous recombination and genetic instability: potential implication in the heterogeneity of myeloproliferative disorders. Blood. 2008;112(4):1402-12.
  • 46
    Cross NC. Genetic and epigenetic complexity in myeloproliferative neoplasms. Hematol Am Soc Hematol Educ Progr. 2011;2011:208-14.
  • 47
    Pagliarini-e-Silva S, Santos BC, Pereira EM, Ferreira ME, Baraldi EC, Sell AM, et al. Evaluation of the association between the JAK2 46/1 haplotype and chronic myeloproliferative neoplasms in a Brazilian population. Clinics (Sao Paulo). 2013;68(1):5-9.
  • 48
    Scott LM, Rebel VI. JAK2 and genomic instability in the myeloproliferative neoplasms: a case of the chicken or the egg? Am J Hematol. 2012;87(11):1028-36.
  • 49
    Kilpivaara O, Levine RL. JAK2 and MPL mutations in myeloproliferative neoplasms: discovery and science. Leukemia. 2008;22(10):1813-7.
  • 50
    Passamonti F, Rumi E. Clinical relevance of JAK2 (V617F) mutant allele burden. Haematologica. 2009;94(1):7-10.
  • 51
    Shih AH, Abdel- Wahab O, Patel JP, Levine RL. The role of mutations in epigenetic regulators in myeloid malignancies. Nat Rev Cancer. 2012;12(9):599-612.
  • 52
    Mascarenhas J, Roper N, Chaurasia P, Hoffman R. Epigenetic abnormalities in myeloproliferative neoplasms: a target for novel therapeutic strategies. Clin Epigenet. 2011;2(2):197-212.

Publication Dates

  • Publication in this collection
    Sep-Oct 2015

History

  • Received
    30 June 2014
  • Accepted
    19 Oct 2014
Associação Brasileira de Hematologia e Hemoterapia e Terapia Celular R. Dr. Diogo de Faria, 775 cj 114, 04037-002 São Paulo/SP/Brasil, Tel. (55 11) 2369-7767/2338-6764 - São Paulo - SP - Brazil
E-mail: secretaria@rbhh.org