骨髓增殖性疾病及肥大细胞病受体酪氨酸激酶基因突变检测与分析
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
目的:骨髓增殖性疾病(myeloproliferative disorders,MPD)是骨髓细胞克隆性或肿瘤性增殖的疾病,包括属于慢性骨髓增殖性疾病(CMPD)范畴的慢性粒细胞白血病(CML)、真性红细胞增多症(PV)、原发性血小板增多症(ET)、慢性特发性骨髓纤维化症(CIMF)、慢性中性粒细胞白血病(CNL)及慢性嗜酸性粒细胞白血病(CEL)和属于骨髓增生异常/骨髓增殖性疾病(MDS/MPD)范畴的不典型慢粒(aCML)、慢性粒-单核细胞白血病(CMML)及幼年型粒.单核细胞白血病(JMML)等。慢性粒细胞白血病是MPD中最常见的类型,其中90%~95%的患者以Ph染色体阳性为特征。Ph染色体是由于位于9号染色体的原癌基因ABL易位到22号染色体与BCR基因相融合形成BCR/ABL融合基因所致,该融合基因所编码的融合蛋白具有很强的ABL蛋白酪氨酸激酶(protein tyrosine kinase,PTK)活性,可导致粒细胞的转化和增殖,在慢粒发病中起关键作用。BCR/ABL所介导的主要信号转导通路是包括TK(酪氨酸激酶)、RAS、RAF、MEK、和ERK的RAS通路、PI3K通路及STAT通路等。除BCR/ABL阳性慢粒外,MPD的其它类型是一组异质性疾病。这一类疾病的发病机理尚不很明确,多数病例没有细胞遗传学异常,但约在10%的病例中已发现涉及到一些受体酪氨酸激酶的融合基因,主要涉及PDGFRB、PDGFRA及FGFRI基因。这些融合基因编码的融合蛋白的功能与BCR/ABL融合蛋白相似,可导致酪氨酸激酶的持续激活。随着肿瘤分子靶向治疗的重大进展尤其是
Objective: Myeloproliferative disorders (MPD) are clonal disorders characterized by excess proliferation of cells from one or more myeloid lineages. It is consist of a heterogenous spectrum conditions mainly including chronic myeloid leukemia (CML), polycythemia vera (PV), essential thrombocythemia (ET), chronic idiopathic myelofibrosis (IMF), chronic neutrophilic leukemia (CNL) , chronic eosinophilic leukemia (CEL) , chronic myelomonocytic leukemia (CMML) and juvenile myelomonocytic leukemia (JMML) . CML is the most common type in MPD and 90%-95% of it is characterized by the presence of the BCR/ABL fusion gene which results in the production of constitutively active tyrosine kinase fusion protein that is believed to be the primary and possibly the only driving force behind the disease. The chimeric protein lead to an increase in activating multiple signalling pathways including RAS/RAF/MAPK, PI3K and STAT and has been found to be response very well to signal transduction inhibitor (STI) imatinib mesylate. The BCR/ABL negative CML and other MPD are rare and heterogeneous diseases of which the molecular pathogenesis is poorly understood. While the most of these cases have a normal karyotype the
    minority of them present an acquired reciprocal chromosomal translocation that disrupts specific receptor tyrosine kinase genes including PDGFRA, PDGFRB and FGFRl and produce constitutively active fusion proteins which are functionally and structurally analogous to BCR/ABL fusion protein. Signal transduction pathways have a wide spectrum of functions including regulations of cell proliferation and differentiation and have been reported to be involved in tumourgenesis. Constitutive activation of the signal transduction pathway may be due to the activating mutations at the receptor level (RTK) or downstream mediators (RAS or RAF) or protein tyrosine phosphatase (PTP).Receptor tyrosine kinase (RTK) and their downstream effectors (RAS, RAF) have emerged as significant components in the pathogenesis of haematological malignancies. Class III RTKs which include KIT, FMS, FLT3, PDGFRA and PDGFRB comprise a similar structure: a five immunoglobin-like domains in the extracellular ligand-binding region, a single hydrophobic transmembrane domain (TM) and two intracellular catalytic (TK1, TK2) domain which is separated by a kinase insert region (KI).The PTPN11 gene encodes the non-receptor-type protein tyrosine phosphatase SHP-2. SHP-2 is a member of the PTP family which is known to be signaling molecules that regulate a variety of cellular processes including cell growth, differentiation, mitotic cycle and oncogenic transformation. Recently mutations in exon 3 and exon 13 which encode the N-SH2 domain and PTP domain respectively were described in patients with NS and JMML causing a gain of function of this gene.With the advent of targeted signal transduction therapy, an accurate clinical and molecular diagnosis of MPD has become increasingly important. Therefore in this study, we undertook extensively mutation analysis using PCR, DHPLC, TOPO TA cloning and direct sequencing method in 42 MPD
    patients for the KIT mutations in JM (exonll) and AL (exonl7) regions and the analogous region of PDGFRs and FMS, the latter also including exon 7, exon 9 and exon 21/22 for the previously reported mutations. For FLT3 gene we screened FLT3/ITD in exon 14 and exon 15 and D835 point mutation in exon 20 and we also screened exon 3 and exon 13 of PTPN11 gene.Methods Genomic DNA from 42 BCR/ABL negative MPD patients were abstracted from bone marrow or peripheral blood using phenol/chloroform method.Genomic DNA amplification The entire coding sequence with exon-intron boundary was amplified. PCR was performed with AmpliTaq Gold DNA polymerase (Applied Biosystem) and 25ng genomic DNA in standard conditions. PCR products were resolved by electrophoresis on 1.5% agarose gel and visualised after staining with ethidium bromide.DHPLC analysis and sequencing Denaturing high-performance liquid chromatography (DHPLC) analysis was performed on a WAVE? DNA Fragment Analysis System (Transgenomic). Each PCR product was mixed with an equal quantity of amplified human placental DNA (sigma) and was denatured and reannealed to allow heteroduplexes formation. All the conditions for the DHPLC analysis, including melting temperature and buffer gradients were determined using the Transgenomic software Navigator VI.0. Samples with extra peaks or with different peak appearance were direct sequenced using the BigDye? Terminator V3.1 Cycle Sequencing Ready Reaction kit (Applied Biosystem) and ABI 3100 Genetics Analyser.Detection of FLT3/ITD PCR products which yielded an extra band on agarose gel were cloned into plasmid vector pCR4-TOPO using TOPO TA Cloning Kit (Invitrogen) and transformed to TOP 10 chemically competent E.coli cells for amplification. Plasmid DNA was abstracted using QIAprep Spin Miniprep Kit (Qiagen) and then direct sequenced.
    ARMS assay was used to screen known nucleotide change in normal controls.Results DHPLC and direct sequencing detected all the previously reported SNPs in screened exons of KIT and FMS as well as some novel silent changes in PDGFRs. We found two patients had the G413S change in exon 9 of FMS however we also found this change in 3 of 43 normal controls using ARMS assay suggesting that this change is a SNP. Two patients were found to have both a normal band and an aberrant band in agarose gel when using primers for exonl4 and exonl5 of FLT3 to amplify genomic DNA. Sequence analysis confirmed a 39 bp duplication with 9 bp insertion and a 57 bp duplication with 9 bp insertion in exon 14 of FLT3 resulting in an addition of 16 and 22 amino acids in these two patients respectively. In both cases the duplications and insertions maintained the integrity of the reading frame. No mutations were detected in PTPN11 gene. The previously reported mutations in KIT exonU and exonl7, PDGFRA exonl2 and exonl8, L301 and Y969 in FMS and D835 in FLT3 were not found in this cohort of patients. In addition, five samples were found to harbour mutations at codon 12 and codon 13 in NRAS gene in our lab's previous work, no mutation was revealed in KRAS, HRAS and BRAF.Conclusions In our study, we detected all the previously reported SNPs as well as some novel silent changes suggesting that DHPLC is a reliable and sensitive method for mutation detection. The silent single base pair substitutions found in PDGFRB and FMS gene may cause structure alternations to mRNA which will require further study to confirm. Two patients were found to have FLT3/ITD in exon 14 of FLT3 and we believe this mutation is crucial to the pathogenesis of the disease. We concluded that the RAS signal transduction pathway may play a very important role in molecular
引文
1. Harris NL, Jaffe ES, Diebold J, et al. The World Health Organization classification of neoplasms of the hematopoietic and lymphoid tissues: report of the Clinical Advisory Committee meeting—Airlie House, Virginia, November, 1997. Hematol J. 2000; 1(1): 53-66.
    2. Goldman JM, Melo JV. Chronic myeloid leukemia—advances in biology and new approaches to treatment. N Engl J Med. 2003; 349(15): 1451-64.
    3. Puil L, Liu J, Gish G, et al. Bcr-Abl oncoproteins bind directly to activators of the Ras signalling pathway. EMBO J. 1994; 13(4): 764-73.
    4. Deininger MW, Goldman JM, Melo JV. The molecular biology of chronic myeloid leukemia. Blood. 2000; 96(10): 3343-56.6
    5. Skorski T, Kanakaraj P, Nieborowska-Skorska M, et al. Phosphatidylinositol-3 kinase activity is regulated by BCR/ABL and is required for the growth of Philadelphia chromosome-positive cells. Blood. 1995; 86(2): 726-36.
    6. Ilaria RL Jr, Van Etten RA. P210 and P190 (BCR/ABL) induce the tyrosine phosphorylation and DNA binding activity of multiple specific STAT family members. J Biol Chem. 1996; 271(49): 31704-10.
    7. Sawyers CL, Callahan W, Witte ON. Dominant negative MYC blocks transformation by ABL oncogenes. Cell. 1992; 70(6): 901-10.
    8. Dancey JE. Recent advances of molecular targeted agents: opportunities for imaging. Cancer Biol Ther. 2003; 2(6): 601-9.
    9. Chrobak L, Voglova J. Imatinib mesylate (STI 571)—a new oral target therapy for chronic myelogenous leukemia (CML). Acta Medica. 2003; 46(3): 85-9.
    10. Cools J, DeAngelo DJ, Gotlib J, et al. A tyrosine kinase created by fusion of the PDGFRA and FIP1L1 genes as a therapeutic target of imatinib in idiopathic hypereosinophilic syndrome. N Engl J Med. 2003; 348(13):1201-14.
    11. Apperley JF, Gardembas M, Melo JV, et al. Response to imatinib mesylate in patients with chronic myeloproliferative diseases with rearrangements of the platelet-derived growth factor receptor beta. N Engl J Med. 2002; 347(7):481-7.
    12. Maroun CR, Naujokas MA, Holgado-Madruga M, et al. The tyrosine phosphatase SHP-2 is required for sustained activation of extracellular signal-regulated kinase and epithelial morphogenesis downstream from the met receptor tyrosine kinase. Mol Cell Biol. 2000; 20(22):8513-25.
    13. Xiao W, Oefner PJ. Denaturing high-performance liquid chromatography: A review. Hum Mutat. 2001; 1 7(6):439-74.
    14. O'Donovan MC, Oefner PJ, Roberts SC, et al. Blind analysis of denaturing high-performance liquid chromatography as a tool for mutation detection. Genomics. 1998; 52(l):44-9.
    15. Ravnik-Glavac M, Atkinson A, Glavac D, Dean M. DHPLC screening of cystic fibrosis gene mutations. Hum Mutat. 2002; 19(4):374-83.
    16. Hoogendoorn B, Owen MJ, Oefner PJ, et al. Genotyping single nucleotide polymorphisms by primer extension and high performance liquid chromatography. Hum Genet. 1999; 104(l):89-93.
    17. Brightwell G, Wycherley R, Waghorn A. SNP genotyping using a simple and rapid single-tube modification of ARMS illustrated by analysis of 6 SNPs in a population of males with FRAXA repeat expansions. Mol Cell Probes. 2002; 16(4):297-305.
    18. Ye S, Dhillon S, Ke X, et al. An efficient procedure for genotyping single nucleotide polymorphisms.Nucleic Acids Res. 2001; 29(17):E88-8.
    19. Pulford D, Meyer H, Brightwell G, et al. Amplification refractory mutation system PCR assays for the detection of variola and Orthopoxvirus. J Virol
     Methods. 2004; 117(1):81-90.
    20. Byrne JL, Paterson HF, Marshall CJ. p21Ras activation by the guanine nucleotide exchange factor Sos, requires the Sos/Grb2 interaction and a second ligand-dependent signal involving the Sos N-terminus. Oncogene. 1996; 13(10):2055-65.
    21. Liang Q, Mohan RR, Chen L, Wilson SE. Signaling by HGF and KGF in corneal epithelial cells: Ras/MAP kinase and Jak-STAT pathways. Invest Ophthalmol Vis Sci. 1998; 3 9(8):1329-38.
    22. Peyssonnaux C, Eychene A. The Raf/MEK/ERK pathway: new concepts of activation. Biol Cell. 2001; 93(l-2):53-62.
    23. Steelman LS, Pohnert SC, Shelton JG, et al. JAK/STAT, Raf/MEK/ERK, PI3K/Akt and BCR-ABL in cell cycle progression and leukemogenesis. Leukemia. 2004; 18(2): 189-218.
    24. Akin C, Brockow K, D'Ambrosio C, et al. Effects of tyrosine kinase inhibitor STI571 on human mast cells bearing wild-type or mutated c-kit. Exp Hematol. 2003; 31(8):686-92.
    25. Manning G, Whyte DB, Martinez R, et al. The protein kinase complement of the human genome. Science. 2002; 298(5600):1912-34.
    26. Zhou M, Felder S, Rubinstein M, et al. Real-time measurements of kinetics of EGF binding to soluble EGF receptor monomers and dimers support the dimerization model for receptor activation. Biochemistry. 1993; 32(32):8193-8.
    27. Ullrich A, Schlessinger J. Signal transduction by receptors with tyrosine kinase activity. Cell. 1990; 61(2):203-12.
    28. Reilly JT. Receptor tyrosine kinases in normal and malignant haematopoiesis. Blood Rev. 2003; 17(4):241-8.
    29. Robertson SC, Tynan JA, Donoghue DJ. RTK mutations and human syndromeswhen good receptors turn bad. Trends Genet. 2000; 16(6):265-71.
    30. Cross NC, Reiter A. Tyrosine kinase fusion genes in chronic myeloproliferative diseases. Leukemia. 2002; 16(7): 1207-12.
    31. Ma Y, Zeng S, Metcalfe D, et al. The c-KIT mutation causing human mastocytosis is resistant to STI571 and other KIT kinase inhibitors; kinases with enzymatic site mutations show different inhibitor sensitivity profiles than wild-type kinases and those with regulatory-type mutations. Blood. 2002; 99(5):1741-4.
    32. Tsujimura T, Kanakura Y, Kitamura Y. Mechanisms of constitutive activation of c-kit receptor. Leukemia. 1997; 11 Suppl 3: 396-398.
    33. Longley BJ, Reguera MJ, Ma Y. Classes of c-KIT activating mutations: proposed mechanisms of action and implications for disease classification and therapy. Leuk Res. 2001; 25(7):571-6.
    34. Beghini A, Peterlongo P, Ripamonti CB, et al. C-kit mutations in core binding factor leukemias. Blood. 2000; 95(2):726-7.
    35. Beghini A, Larizza L, Cairoli R, et al. c-kit activating mutations and mast cell proliferation in human leukemia. Blood. 1998; 92(2):701-2.
    36. Ridge SA, Worwood M, Oscier D, et al. FMS mutations in myelodysplastic, leukemic, and normal subjects. Proc Natl Acad Sci USA. 1990; 87(4):1377-80.
    37. Roussel MF, Downing JR, Rettenmier CW, Sherr CJ. A point mutation in the extracellular domain of the human CSF-1 receptor (c-fms proto-oncogene product) activates its transforming potential. Cell 1988; 55: 979-988.
    38. Roussel MF, Dull TJ, Rettenmier CW, et al. Transforming potential of the c-fms proto-oncogene (CSF-1 receptor). Nature. 1987; 325: 549-552.
    39. Tobal K, Pagliuca A, Bhatt B, et al. Mutation of the human FMS gene (M-CSF receptor) in myelodysplastic syndromes and acute myeloid leukemia. Leukemia. 1990; 4: 486-9.
    40. Abu-Duhier FM, Goodeve AC, et al. Mutational analysis of class III
     receptor tyrosine kinases (C-KIT, C-FMS, FLT3) in idiopathic myelofibrosis. Br J Haematol.2003; 20(3):464-70.
    41. Agnes F, Shamoon B, Dina C, et al. Genomic structure of the downstream part of the human FLT3 gene: exon/intron structure conservation among genes encoding receptor tyrosine kinases (RTK) of subclass III. Gene. 1994; 145(2):283-8.
    42. Turner AM, Lin NL, Issarachai S, et al. FLT3 receptor expression on the surface of normal and malignant human hematopoietic cells. Blood. 1996; 88: 3 383-3390.
    43. Nakao M, Yokota S, Iwai T, et al. Internal tandem duplication of the flt3 gene found in acute myeloid leukemia. Leukemia. 1996; 10: 1911-1918.
    44. Yokota S, Kiyoi H, Nakao M, et al. Internal tandem duplication of the FLT3 gene is preferentially seen in acute myeloid leukemia and myelodysplastic syndrome among various hematological malignancies. A study on a large series of patients and cell lines. Leukemia. 1997; 11(10): 1605-93.
    45. Abu-Duhier FM, Goodeve AC, Wilson GA, et al. Identification of novel FLT-3 Asp835 mutations in adult acute myeloid leukaemia. Br J Haematol 2001; 113:983-988.
    46. Sheikhha MH, Awan A, Tobal K, et al. Prognostic significance of FLT3 ITD and D835 mutations in AML patients. Hematol J 2003; 4: 41-46.
    47. Heinrich MC, Corless CL, Duensing A, et al. PDGFRA activating mutations in gastrointestinal stromal tumors. Science. 2003; 299(5607):708-10.
    48. Chompret A, Kannengiesser C, Barrois M, et al. PDGFRA germline mutation in a family with multiple cases of gastrointestinal stromal tumor. Gastroenterology. 2004; 126(1 ):31 8-21.
    49. Baxter EJ, Hochhaus A, Bolufer P, et al. The t(4;22)(ql2;ql 1) in atypical chronic myeloid leukaemia fuses BCR to PDGFRA. Hum Mol Genet. 2002; 11(12): 1391 -7.
    50. Golub TR, Barker GF, Lovett M, Gillil DG. Fusion of PDGF receptor beta to a novel ets-;ike gene, tel, in chronic myelomonocytic leukemia with t(5;12) chromosomal translocation. Cell 1994; 77: 307-316.
    51. Magnusson MK, Meade KE, Brown KE, et al. Rabaptin-5 is a novel fusion partner to platelet-derived growth factor beta receptor in chronic myelomonocytic leukemia. Blood 2001; 98(8):2518-25.
    52. Ross TS, Bernard OA, Berger R, Gilliland DG. Fusion of Huntingtin interacting protein 1 to platelet-derived growth factor beta receptor (PDGFbetaR) in chronic myelomonocytic leukemia with t(5;7) (q33;q11.2). Blood 1998; 91 (12):4419-26.
    53. Kulkarni S, Heath C, Parker S, et al. Fusion of H4/D10S170 to the platelet-derived growth factor receptor beta in BCR-ABL-negative myeloproliferative disorders with a t(5;10)(q33;q21). Cancer Res. 2000; 60(13):3592-8.
    54. Abe A, Emi N, Tanimoto M, et al. Fusion of the platelet-derived growth factor receptor beta to a novel gene CEV14 in acute myelogenous leukemia after clonal evolution. Blood. 1997; 90(11):4271-7.
    55. Steer EJ and Cross NC. Myeloproliferative disorders with translocations of chromosome 5q31-35: role of the platelet-derived growth factor receptor Beta. Acta Haematol. 2002; 107(2): 113-22.
    56. Baxter EJ, Kulkarni S, Vizmanos JL, et al. Novel translocations that disrupt the platelet-derived growth factor receptor beta (PDGFRB) gene in BCR-ABL-negative chronic myeloproliferative disorders. Br J Haematol. 2003; 120(2):251-6.
    57. Maroun CR, Naujokas MA, Holgado-Madruga M, et al. The tyrosine phosphatase SHP-2 is required for sustained activation of extracellular signal-regulated kinase and epithelial morphogenesis downstream from the met receptor tyrosine kinase. Mol Cell Biol. 2000; 20(22):8513-25.
    58. Zhang SQ, Tsiaras WG, Araki T, et al. Receptor-specific regulation of phosphatidylinositol 3'-kinase activation by the protein tyrosine phosphatase Shp2. Mol Cell Biol. 2002; 22(12):4062-72.
    59. Stein-Gerlach M, Wallasch C, Ullrich A. SHP-2, SH2-containing protein tyrosine phosphatase-2. Int J Biochem Cell Biol. 1998; 30(5):559-66.
    60. Tartaglia M, Mehler EL, Goldberg R, et al. Mutations in PTPN11, encoding the protein tyrosine phosphatase SHP-2, cause Noonan syndrome. Nat Genet. 2001;29(4):465-8.
    61. Tartaglia M, Niemeyer CM, Fragale A, et al. Somatic mutations in PTPN11 in juvenile myelomonocytic leukemia, myelodysplastic syndromes and acute myeloid leukemia. Nat Genet. 2003; 34(2): 148-50.
    62. Le Gac G, Mura C, Ferec C. Complete scanning of the hereditary hemochromatosis gene (HFE) by use of denaturing HPLC. Clin Chem. 2001; 47(9):1633-40.
    63. Kiyoi H, Towatari M, Yokota S, et al. Internal tandem duplication of the FLT3 gene is a novel modality of elongation mutation which causes constitutive activation of the product. Leukemia. 1998; 12(9): 1333-7.
    64. Kelly LM, Liu Q, Kutok JL, et al. FLT3 internal tandem duplication mutations associated with human acute myeloid leukemias induce myeloproliferative disease in a murine bone marrow transplant model. Blood. 2002; 99(l):310-8.
    65. Mizuki M, Fenski R, Halfter H, et al. Flt3 mutations from patients with acute myeloid leukemia induce transformation of 32D cells mediated by the Ras and STAT5 pathways. Blood. 2000; 96(12):3907-14.
    66. Kiyoi H, Naoe T, Yokota S, et al. Internal tandem duplication of FLT3 associated with leukocytosis in acute promyelocytic leukemia. Leukemia. 1997; ll(9):1447-52.
    67. Stirewalt DL, Kopecky KJ, Meshinchi S, et al. FLT3, RAS, and TP53
     mutations in elderly patients with acute myeloid leukemia. Blood. 2001; 97(11):3589-95.
    68. Kottaridis PD, Gale RE, Linch DC. Prognostic implications of the presence of FLT3 mutations in patients with acute myeloid leukemia. Leuk Lymphoma. 2003; 44(6):905-13.
    69. Kottaridis PD, Gale RE, Frew ME, et al. The presence of a FLT3 internal tandem duplication in patients with acute myeloid leukemia (AML) adds important prognostic information to cytogenetic risk group and response to the first cycle of chemotherapy: analysis of 854 patients from the United Kingdom Medical Res76earch Council AML 10 and 12 trials. Blood 2001; 98: 1752-1759.
    70. Kottaridis PD, Gale RE, Linch DC. Prognostic implications of the presence of FLT3 m tations in patients with acute myeloid leukemia. Leuk Lymphoma. 2003;44(6):905-13.
    71. Abu-Duhier FM, Goodeve AC, Wilson GA, et al. FLT3 internal tandem duplication mutations in adult acute myeloid leukaemia define a high-risk group. Br J Haematol 2000; 111: 190-195.
    72. Sheng XM, Kawamura M, Ohnishi H, et al. Mutations of the RAS genes in childhood acute myeloid leukemia, myelodysplastic syndrome and juvenile chronic myelocytic leukemia. Leuk Res. 1997; 21(8):697-701
    73. Padua RA, Guinn BA, Al-Sabah Al, et al. RAS, FMS and p53 mutations and poor clinical outcome in myelodysplasias: a 10-year follow-up. Leukemia 1998; 12: 887-892.
    74. Neubauer A, Dodge RK, George SL, et al. Prognostic importance of mutations in the ras proto-oncogenes in de novo acute myeloid leukemia. Blood. 1994; 83(6): 1603-11.
    75. Hirsch-Ginsberg C, LeMaistre AC, Kantarjian H, et al. RAS mutations are rare events in Philadelphia chromosome-negative/bcr gene rearrangement

© 2004-2018 中国地质图书馆版权所有 京ICP备05064691号 京公网安备11010802017129号

地址:北京市海淀区学院路29号 邮编:100083

电话:办公室:(+86 10)66554848;文献借阅、咨询服务、科技查新:66554700