Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Genet Mol Biol ; 44(3): e20200332, 2021.
Article in English | MEDLINE | ID: mdl-34358284

ABSTRACT

Two Italian patients with the initial clinical diagnosis of Rothmund-Thomson syndrome were negative for RECQL4 mutations but showed in peripheral blood cells a spontaneous chromosomal instability significantly higher than controls. Revisiting after time their clinical phenotype, the suggestive matching with the autosomal dominant syndrome Poikiloderma, Hereditary Fibrosing with Tendon Contracture, Myopathy and Pulmonary fibrosis (POIKTMP) was confirmed by identification of the c.1879A>G (p.Arg627Gly) alteration in FAM111B. We compare the overall clinical signs of our patients with those of reported carriers of the same mutation and present the up-to-date mutational repertoire of FAM111B and the related phenotypic spectrum. Our snapshot highlights the age-dependent clinical expressivity of POIKTMP and the need to follow-up patients to monitor the multi-tissue impairment caused by FAM111B alterations. We link our chromosomal instability data to the role of FAM111B in cancer predisposition, pointed out by its implication in DNA-repair pathways and the outcome of pancreatic cancer in 2 out of 17 adult POIKTMP patients. The chromosomal instability herein highlighted well connects POIKTMP to cancer-predisposing syndromes, such as Rothmund-Thomson which represents the first hereditary poikiloderma entering in differential diagnosis with POIKTMP.

2.
Eur J Cell Biol ; 93(8-9): 355-65, 2014.
Article in English | MEDLINE | ID: mdl-25123532

ABSTRACT

MARK4 is a serine-threonine kinase that phosphorylates MAP proteins, increasing microtubule dynamics. MARK4 differs from the other members of the MARK family for encoding two isoforms (MARK4L and MARK4S), differentially expressed in the nervous system, and for the peculiar localisation at the centrosome and the midbody. By cytofluorimetric analysis we showed that MARK4 is expressed throughout the cell cycle and preferentially activated during mitosis. Depletion of MARK4S affected the morphology and proliferation of fibroblasts and glioma cells, as the percentages of cells in S and G2/M phases were reduced and the percentage of cells in G1 was increased. In MARK4S silenced cells, centrosomes were duplicated and positioned apically to the nucleus, indicating that the centrosome cycle was altered and the cells arrested in G1 phase. Overexpression of MARK4L or MARK4S reduced the density of the microtubule network, confirming microtubules as the main target of MARK4, and revealed a novel co-localisation of MARK4 and vimentin. Taken together, our data confirm that MARK4 is a key component in the regulation of microtubule dynamics and highlight its major role in cell cycle progression, particularly at the G1/S transition. The co-localisation of vimentin and MARK4L suggests that MARK4 has a wide-ranging influence on cytoskeleton.


Subject(s)
Microtubules/metabolism , Protein Serine-Threonine Kinases/metabolism , Cell Cycle Checkpoints , Cell Division , Cell Line, Tumor , Centrosome/metabolism , Fibroblasts/cytology , Humans , Phosphorylation , Protein Serine-Threonine Kinases/genetics
3.
Anal Cell Pathol (Amst) ; 34(6): 319-38, 2011.
Article in English | MEDLINE | ID: mdl-22156016

ABSTRACT

BACKGROUND: MAP/microtubule affinity-regulating kinase 4 (MARK4) is a serine-threonine kinase expressed in two spliced isoforms, MARK4L and MARK4S, of which MARK4L is a candidate for a role in neoplastic transformation. METHODS: We performed mutation analysis to identify sequence alterations possibly affecting MARK4 expression. We then investigated the MARK4L and MARK4S expression profile in 21 glioma cell lines and 36 tissues of different malignancy grades, glioblastoma-derived cancer stem cells (GBM CSCs) and mouse neural stem cells (NSCs) by real-time PCR, immunoblotting and immunohistochemistry. We also analyzed the sub-cellular localisation of MARK4 isoforms in glioma and normal cell lines by immunofluorescence. RESULTS: Mutation analysis rules out sequence variations as the cause of the altered MARK4 expression in glioma. Expression profiling confirms that MARK4L is the predominant isoform, whereas MARK4S levels are significantly decreased in comparison and show an inverse correlation with tumour grade. A high MARK4L/MARK4S ratio also characterizes undifferentiated cells, such as GBM CSCs and NSCs. Accordingly, only MARK4L is expressed in brain neurogenic regions. Moreover, while both MARK4 isoforms are localised to the centrosome and midbody in glioma and normal cells, the L isoform exhibits an additional nucleolar localisation in tumour cells. CONCLUSIONS: The observed switch towards MARK4L suggests that the balance between the MARK4 isoforms is carefully guarded during neural differentiation but may be subverted in gliomagenesis. Moreover, the MARK4L nucleolar localisation in tumour cells features this MARK4 isoform as a nucleolus-associated tumour marker.


Subject(s)
Biomarkers, Tumor/analysis , Centrosome/enzymology , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Glioma/metabolism , Protein Serine-Threonine Kinases/metabolism , Adult , Aged , Animals , Base Sequence , Biopsy , Cell Line, Tumor , Cell Nucleolus/metabolism , Child , Child, Preschool , DNA Mutational Analysis , Female , Genome, Human/genetics , Glioma/genetics , Glioma/pathology , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Male , Mice , Middle Aged , Neoplastic Stem Cells/enzymology , Neoplastic Stem Cells/pathology , Neural Stem Cells/enzymology , Neural Stem Cells/pathology , Phenotype , Protein Serine-Threonine Kinases/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism
4.
Leuk Res ; 34(10): 1287-95, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20227111

ABSTRACT

The t(8;21) Acute Myeloid Leukaemia (AML) Kasumi-1 cell line with N822K KIT mutation, is a model system for leukemogenesis. As AML initiating cells reside in the CD34(+)CD38(-) fraction, we addressed the refined cytogenomic characterization and miRNA expression of Kasumi-1 cell line and its FACS-sorted subpopulations focussing on this compartment. By conventional cytogenetics, Spectral-Karyotyping and array-CGH the cytogenomic profile of Kasumi-1 cells evidenced only subtle regions differentially represented in CD34(+)CD38(-) cells. Expression profiling by a miRNA platform showed a set of miRNA differentially expressed in paired subpopulations and the signature of miR-584 and miR-182 upregulation in the CD34(+)CD38(-) fraction.


Subject(s)
ADP-ribosyl Cyclase 1/analysis , Antigens, CD34/analysis , Leukemia, Myeloid, Acute/genetics , MicroRNAs/analysis , Cell Line, Tumor , Chromosomes, Human, Pair 4 , Comparative Genomic Hybridization , Gene Expression Profiling , Humans , Immunophenotyping , Leukemia, Myeloid, Acute/immunology , Proto-Oncogene Proteins c-kit/genetics
5.
Cell Oncol ; 31(5): 357-70, 2009.
Article in English | MEDLINE | ID: mdl-19759416

ABSTRACT

BACKGROUND: We have previously shown that the sustained expression of MARK4L transcripts in glioma and neural progenitors (NHNPs) declines after exposure to antisense MARK4L oligonucleotides in glioblastoma cell lines. Array-CGH confirmed the genomic duplication of MARK4L identified by FISH in a glioblastoma cell line. This background together with literature data on the exogenous association of MARK4 with interphase centrosome prompted us to investigate the sub-cellular localization of the endogenous MARK4L protein aiming at achieving insights on its possible role in the pathomechanisms of glioma. METHODS: Immunodetection was carried out to validate the specificity of MARK4L antibody in gliomas and NHNPs. Mass spectrometry was applied for MARK4L protein identification in a representative glioblastoma cell line. Combined biochemical fractionation and immunodetection analyses were performed to confirm the sub-cellular localization of MARK4L achieved by immunofluorescence in glioma cell lines. RESULTS: By assigning MARK4L protein within the band immunoprecipitated by the specific antibody we validated our anti-MARK4L antibody. We demonstrated that the endogenous MARK4L: (i) colocalizes with centrosomes at all mitotic stages and resides in centrosome-enriched fractions; (ii) associates with the nucleolus and the midbody and respective fractions, and (iii) co-stains the aberrant centrosome configurations observed in glioma cell lines. CONCLUSION: The overall data merge on the multiplex entry of MARK4L into the cell cycle and link it to the aberrant centrosomes in glioma cell lines suggesting a possible role of this kinase in the abnormal mitotic processes of human glioma.


Subject(s)
Glioma/metabolism , Protein Serine-Threonine Kinases/metabolism , Antibodies/immunology , Cell Line, Tumor , Cell Nucleus/metabolism , Centrosome/metabolism , Glioma/pathology , Humans , Immunoprecipitation , Interphase , Mass Spectrometry , Mitosis , Molecular Weight , Neurons/metabolism , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/immunology , Stem Cells/metabolism , Subcellular Fractions/metabolism
6.
Anticancer Drugs ; 18(6): 669-76, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17762395

ABSTRACT

Glioblastoma is a highly malignant tumor, characterized by an unfavorable prognosis even in response to multidisciplinary treatment strategies, owing to its high-invasive phenotype. Ukrain, a semisynthetic thiophosphoric acid derivative of the purified alkaloid chelidonine, has been used in the therapy of several solid tumors, but little is known about its effect on glioblastoma and, in general, about the molecular mechanisms responsible for its effects. In particular, we previously demonstrated that Ukrain modulates the expression of genes and proteins involved in tumor invasion, and here we investigate some unreported effects of Ukrain on human cultured glioblastoma cells. We used morphological and molecular biology methods to analyze the expression and the intracellular distribution pattern of glial fibrillary acidic protein, the expression of the gap junction protein connexin 43 and the apoptotic effect in human glioblastoma cells treated with 0.1, 1 and 10 micromol/l Ukrain for 72 h. After treatment with 10 micromol/l Ukrain, glial fibrillary acidic protein fluorescence increased and a higher number of cells displayed glial fibrillary acidic protein organized into a filamentous state. Western blot analysis of glial fibrillary acidic protein confirmed that Ukrain tended to upregulate the protein. Connexin 43 was not modulated by Ukrain both at the mRNA and at the protein level. Ukrain-induced apoptotic rate was 4.63, 10.9 and 28.9% after 0.1, 1 and 10 micromol/l Ukrain, respectively, likely mediated by cytochrome c release in the cytoplasm. Considered as a whole, these findings provide new information to complete the understanding of the mechanisms of Ukrain antitumor and chemopreventive effect, and support the possible potential of Ukrain for the therapy of brain tumors.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Berberine Alkaloids/pharmacology , Connexin 43/biosynthesis , Glial Fibrillary Acidic Protein/biosynthesis , Phenanthridines/pharmacology , Blotting, Western , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/ultrastructure , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Gap Junctions/drug effects , Gap Junctions/metabolism , Glioblastoma/pathology , Humans , Immunohistochemistry , Microscopy, Electron, Transmission , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation
7.
Am J Surg Pathol ; 31(5): 760-9, 2007 May.
Article in English | MEDLINE | ID: mdl-17460461

ABSTRACT

Caveolins are basic constituents of flask-shaped cell membrane microdomains (caveolae), which are involved in many cell functions, including signalling, trafficking, and cellular growth control. The distribution of caveolae within the normal brain and in brain tumors is controversial. In the present study, we describe the expression of caveolin-1 (cav-1) in 64 brain tumors of different grade, of either astroglial or oligodendroglial origin. All studied astrocitomas of any grade (from II to IV) were cav-1 positive, displaying staining patterns and intensity specifically associated to the different tumor grades. In all glioblastomas and gliosarcomas, cav-1 staining was extremely intense, typically localized at the cell membrane and recognized a variable percentage of cells, including the majority of spindle cells and palisade-oriented perinecrotic cells. In anaplastic astrocytomas, a less intense membrane staining or a cytoplasmic dotlike immunoreactivity were present, the latter being almost the exclusive pattern observed in diffuse astrocitomas grade II. In contrast to astroglial tumors, the striking totality of grade II oligodendrogliomas and the large majority of grade III were lacking cav-1 expression. Interestingly, a cav-1 distribution overlapping the pattern described in tissues was observed also in primary cell cultures of human glioblastomas and astrocytomas, and also in one established glioblastoma cell line (U251 MG), analyzed by means of confocal microscopy and flow cytometry. In conclusion, among astroglial tumors cav-1 expression varies in distribution, pattern, and intensity specifically according to tumor types and grades. The association between tumor progression and a more structured membranous pattern of cav-1 expression could suggest the hypothesis of a neoplastic shift towards a mesenchymal phenotype, whose behavioral and biologic significance worth further studies. Finally, the lack of cav-1 immunoreactivity in oligodendrogliomas suggests its concrete application as a useful diagnostic marker.


Subject(s)
Astrocytes/metabolism , Biomarkers, Tumor/metabolism , Brain Neoplasms/metabolism , Caveolin 1/metabolism , Glioblastoma/metabolism , Oligodendroglioma/metabolism , Adult , Aged , Aged, 80 and over , Astrocytes/pathology , Biomarkers, Tumor/genetics , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Count , Cell Line, Tumor , Cell Separation , Chromosome Deletion , Chromosomes, Human, Pair 1/genetics , Chromosomes, Human, Pair 19/genetics , Disease Progression , Flow Cytometry , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Immunohistochemistry , Middle Aged , Neoplasm Staging , Oligodendroglioma/genetics , Oligodendroglioma/pathology
8.
Anticancer Drugs ; 17(2): 189-94, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16428937

ABSTRACT

Glioblastoma is a highly malignant brain tumor with a highly invasive phenotype and hence an unfavorable prognosis even in response to multidisciplinary treatment strategies. Ukrain, a semi-synthetic thiophosphoric acid derivative of the purified alkaloid chelidonine, has been used in the therapy of several solid tumors, but little is known about its effect on glioblastoma and, in general, about the molecular mechanisms responsible for its effects. We used RT-PCR, Western blot and SDS-zymography to investigate the effects of three doses of Ukrain (0.1, 1 and 10 micromol/l) on the expression of genes and proteins involved in the extracellular matrix remodeling associated with tumor invasion in human cultured glioblastoma cells treated for 24, 48 and 72 h. We analyzed the expression of matrix metalloproteinase-2 and -9, the main mediators of glioblastoma invasiveness, and secreted protein acidic and rich in cysteine (SPARC), involved in the regulation of cell-matrix interactions. There was a significant, dose-related decrease of glioblastoma cell proliferation and a tendency to downregulation of SPARC at the protein level 72 h after 10 micromol/l Ukrain, suggesting the drug may be a useful therapeutic tool for brain tumors.


Subject(s)
Antineoplastic Agents/pharmacology , Berberine Alkaloids/pharmacology , Cell Proliferation/drug effects , Glioblastoma/drug therapy , Glioblastoma/metabolism , Matrix Metalloproteinase 2/metabolism , Osteonectin/metabolism , Phenanthridines/pharmacology , Blotting, Western , Brain Neoplasms , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Electrophoresis, Polyacrylamide Gel , Gene Expression Regulation, Neoplastic , Glioblastoma/pathology , Humans , Neoplasm Invasiveness/pathology , RNA, Messenger , Reverse Transcriptase Polymerase Chain Reaction
9.
Cancer Lett ; 232(1): 107-20, 2006 Jan 28.
Article in English | MEDLINE | ID: mdl-16271439

ABSTRACT

Rothmund-Thomson Syndrome (RTS) is a rare autosomal recessive genodermatosis with a heterogeneous clinical profile. Mutations in RECQL4, encoding a RecQ DNA helicase, are present in a large fraction, but not all clinically diagnosed patients, allowing to classify RTS among the RecQ helicase chromosomal instability defects including Bloom's and Werner's syndromes. Results of RECQL4 test coupled to the variable clinical presentation favored the splitting of RTS clinical phenotype into nosological entities under distinct genetic control. In parallel, lumping of the RECQL4 gene to two other diseases, RAPADILINO and Baller-Gerold has paved the way to unravel through allelic heterogeneity complex genotype-phenotype correlations. Recql4 knockout mice provided crucial insights into the comprehension of the functional role of RECQL4 helicase, which have been corroborated by the initial biochemical characterization of RECQL4 protein and its acting pathway and by studies on RECQL4 homologs in yeast and Xenopus. A role for RECQL4 in initiation of DNA replication and in sister chromatid cohesion has been proposed, which currently fits the pieces of evidence achieved by different approaches. Further work is needed to define the specific and shared functions of RECQL4 in relation to other RecQ helicases and to connect RECQL4 diseases to other genomic instability syndromes with birth defects and cancer predisposition.


Subject(s)
Adenosine Triphosphatases/deficiency , Chromosomal Instability , Chromosome Segregation , DNA Helicases/deficiency , Rothmund-Thomson Syndrome/metabolism , Adenosine Triphosphatases/genetics , Animals , DNA Helicases/genetics , Genetic Predisposition to Disease , Humans , Mutation , RecQ Helicases , Rothmund-Thomson Syndrome/genetics
10.
Cancer Genet Cytogenet ; 161(2): 140-5, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16102584

ABSTRACT

Glioblastomas, the most frequent and malignant glial tumors, are known to be phenotypically heterogeneous. A low fraction of glioblastomas is associated with specific chromosomal losses at 1p and 19q, which are commonly found in oligodendrogliomas and are generally considered to be a primary event in the development of these tumors. Subsequent progression of oligodendroglial tumors appears to be triggered by additional molecular features underlying the transition to anaplastic oligodendroglioma and glioblastoma multiforme (GBM) such as deletions of 9p and 10q, and alterations of CDKN2A (p16), which is located at 9p21. These findings strengthen the view that GBM on rare occasions may develop from oligodendroglial differentiated cells. In the present study, we evaluated the newly established MI-4 glioblastoma cell line, which displays 1p and 19q specific alterations targeting preferential regions of allelic loss in glial neoplasms, by array-CGH and fluorescence in situ hybridization (FISH) analyses that were combined to obtain a high resolution map of targeted chromosome rearrangements and copy number changes throughout the genome. Genome-wide and chromosome 19 full coverage array-CGH analysis of the MI-4 cell line revealed that in this particular cell line, 1p-specific loss, including the CDKN2 (p18) gene, is not accompanied by loss of the previously described 19q13.3 tumor suppressor candidate region. Interestingly, the array-CGH (CGHa) profile showed an increase in copy number along most of 19q including the AKT2 oncogene and the KLKs gene family, which have previously been shown to be amplified in pancreatic carcinomas and upregulated in several tumors, respectively. The concomitant 1p partial loss and chromosome 19 alterations, with the +7 and -10-specific GBM markers associated with homozygous deletion of 9p21.3 including CDKN2A (p16), are distinct features of the glioblastoma MI-4 cell line, illustrating its origin from an olidodendroglial tumor. Based on these results, we conclude that the MI-4 glioblastoma cell line might function as a model system for investigations into the behavior of a defined oligodendroglioma subtype.


Subject(s)
Brain Neoplasms/genetics , Cell Line, Tumor , Chromosome Aberrations , Chromosomes, Human, Pair 1 , Glioblastoma/genetics , Oligodendroglioma/genetics , Chromosomes, Human, Pair 19 , Gene Dosage , Humans , In Situ Hybridization, Fluorescence , Microarray Analysis
11.
Biomed Pharmacother ; 59(7): 359-64, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16084059

ABSTRACT

INTRODUCTION: Glioblastoma is a highly malignant brain tumor with a high-invasive phenotype, so the prognosis is unfavorable, even in response to multidisciplinary treatment strategies. Obviously, therefore, a better therapeutic strategy is needed. Resveratrol has been reported to be one of the most potent chemopreventive agents inhibiting the cellular processes associated with tumor development, including initiation, promotion, and progression. MATERIALS AND METHODS: In this study we used RT-PCR, western blot and SDS-zymography to investigate the effect of resveratrol on the expression of genes and proteins involved in the extracellular matrix remodeling associated with tumor invasion in human cultured glioblastoma cells treated for 24, 48 and 72 h. We analyzed the expression of matrix metalloproteinase-2 (MMP-2), the main mediator of glioblastoma invasiveness, and the Secreted Protein Acidic and Rich in Cysteine (SPARC), involved in the regulation of cell-matrix interactions. RESULTS: Our results show a dose-related decrease of MMP-2 mRNA and protein levels 72 h after resveratrol treatment, and lower SPARC gene and protein expression 72 h after resveratrol treatment. This indicates that resveratrol may influence the two major factors in the ECM remodeling occurring with tumor invasion, suggesting it may have uses as a therapeutic agent for brain tumors.


Subject(s)
Antineoplastic Agents/pharmacology , Glioblastoma/metabolism , Matrix Metalloproteinase 2/genetics , Osteonectin/genetics , Stilbenes/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Glioblastoma/drug therapy , Glioblastoma/pathology , Humans , RNA, Messenger/analysis , Resveratrol
12.
Exp Hematol ; 33(6): 682-8, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15911092

ABSTRACT

OBJECTIVE: Alterations in growth factor signaling pathways may be a frequent collaborating event in AML1-ETO-mediated leukemogenesis. Gain-of-function KIT receptor mutations have been reported in adult AML patients, especially those with core binding factor leukemia (CBFL). We have previously reported a new gain-of-function KIT(Asn822Lys) mutation that is constitutively expressed in the Kasumi-1 CBFL cell line, and has recently been described in two childhood AML patients. To explore the molecular basis of the effects of this mutation in the appropriate context of hemopoietic dysregulation, we investigated KIT downstream signaling in the Kasumi-1 cell line by means of STI 571 (Imatinib, Gleevec) pharmacological inhibition. MATERIALS AND METHODS: We investigated KIT(Asn822Lys) mutant-initiated signaling in Kasumi-1 cell line, and characterized the inhibitory effect of the STI 571 protein tyrosine kinase inhibitor on downstream signaling. RESULTS: The use of STI 571-mediated inhibition impaired the tyrosine phosphorylation of KIT(Asn822Lys) and its association with the p85 subunit of phosphatidylinositol 3'-kinase (p85PI3K). The downstream constitutive phosphorylation of JNK1/2 and STAT3 was also significantly inhibited, but STI 571 had no effect on the constitutive activation of Akt, thus suggesting that it is due to other signaling in Kasumi-1 cells. STI 571 inhibited the KIT-mediated proliferation of Kasumi-1 cells in a dose-dependent manner. CONCLUSIONS: These findings show the role of PI3K in KIT(Asn822Lys)-mediated constitutive activation through the Akt-independent downstream signaling pathway of JNK, and also demonstrate the mutant's susceptibility to STI 571, which may therefore have therapeutic potential in CBFL patients with susceptible KIT mutations.


Subject(s)
Antineoplastic Agents/pharmacology , Piperazines/pharmacology , Proto-Oncogene Proteins c-kit/physiology , Pyrimidines/pharmacology , Signal Transduction/drug effects , Asparagine/genetics , Benzamides , Cell Line, Tumor , Humans , Imatinib Mesylate , Lysine/genetics , Mutation , Phosphorylation , Proto-Oncogene Proteins c-kit/chemistry , Proto-Oncogene Proteins c-kit/genetics
13.
Leuk Lymphoma ; 46(2): 247-55, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15621809

ABSTRACT

The Kasumi-1 cell line is an intensively investigated model system of Acute Myeloid Leukemia with t(8;21) translocation, that represents 1 of the 2 main subtypes of Core Binding Factor Leukemia (CBFL). Since establishment in 1991 the Kasumi-1 cell line has provided the tool to study the peculiar molecular, morphologic, immunophenotypic findings of AML with t(8;21) and the functional consequences of the AML1-ETO fusion oncogene on myeloid differentiation. Leukemogenesis involves multiple genetic changes and, as suggested by murine experiments and other findings in humans, AML1-ETO expression may not be sufficient for full blown leukemia. In agreement with the "two hits" model of leukemogenesis, based on the cooperation between 1 class of mutations that impair hematopoietic differentiation and a second class of mutations that confer a proliferative and/or survival advantage to hematopoietic progenitors an activating mutation in the tyrosine kinase domain of the c-kit gene was identified in the AML1/ETO expressing Kasumi-1 cell line. The dosage of the Asn822Lys mutated allele was shown to be about 5-fold compared to the normal allele and c-kit amplification was found to map to minute 4cen-q11 marker chromosomes, likely derived from the extra chromosome 4 recorded in the newly established cell line. The combination of t(8;21) and trisomy 4 leading to enhanced dosage of a mutated kit allele is a feature of a few CBFL patients reproduced by the Kasumi-1 cell model. The Kasumi-1 cell line, paralleling the commitment stage of CBF leukemia also provides a valuable resource to investigate the effect of tyrosine kinase kit mutant on the main KIT-regulated signal transduction pathways, i.e. MAPK, PI3K/AKT and STAT3 and the diverse inhibitory effect exerted by STI 571 on these KIT mutant activated pathways. PI3K-dependent activation of AKT and STAT activation was observed in Kasumi-1 cells. Contrary to the expectations for an amplified tyrosine kinase kit mutant, we found that STI 571 inhibited KIT Asn822Lys tyrosine phosphorylation and downstream JNK and STAT3 effectors in Kasumi-1 cells, but had no effect on constitutive activation of AKT, suggesting that signaling by tyrosine kinases other than KIT may be responsible for its activation in Kasumi-1 cells. Independent findings on the same model system provide complementary insights into designing strategies for treatment of CBF leukemia associated with mutations in the KIT catalytic domain.


Subject(s)
Cell Line, Tumor , Leukemia, Myeloid/genetics , Mutation , Proto-Oncogene Proteins c-kit/genetics , Acute Disease , Chromosomes, Human, Pair 21 , Chromosomes, Human, Pair 8 , Humans , Leukemia, Myeloid/etiology , Leukemia, Myeloid/pathology , Signal Transduction/genetics , Translocation, Genetic
14.
Oncogene ; 22(17): 2581-91, 2003 May 01.
Article in English | MEDLINE | ID: mdl-12735302

ABSTRACT

Alterations of 19q13 are frequently observed in glial neoplasms, suggesting that this region harbors at least one gene involved in gliomagenesis. Following our previous studies on structural 19q chromosome rearrangements in gliomas, we have undertaken a detailed FISH analysis of the breakpoints and identified a 19q13.2 intrachromosomal amplification of the MAP/microtubule affinity-regulating kinase 4 (MARK4) gene in three primary glioblastoma cell lines. Recent data suggest that this gene is involved in the Wnt-signaling pathway. We observed that the expression of the alternatively spliced MARK4L isoform is upregulated in both fresh and cultured gliomas and overexpressed in all of the above three glioblastoma cell lines. Interestingly, we also found that MARK4L expression is restricted to undifferentiated neural progenitor cells or proliferating glial precursor cells, whereas its expression is downregulated during glial differentiation. Perturbation of expression using antisense oligonucleotides against MARK4 in glioblastoma cell lines, consistently induced a decreased proliferation of tumor cells. Taken together, these data show that MARK4, which is normally expressed in neural progenitors, is re-expressed in gliomas and may become a key target of intrachromosomal amplification upon 19q rearrangements.


Subject(s)
Glioblastoma/metabolism , Protein Isoforms/metabolism , Protein Serine-Threonine Kinases/metabolism , Cell Differentiation/physiology , Cell Division/physiology , Chromosomes, Human, Pair 19 , Humans , In Situ Hybridization, Fluorescence , Molecular Sequence Data , Neuroglia/physiology , Protein Isoforms/genetics , Protein Serine-Threonine Kinases/genetics , Sequence Deletion , Tumor Cells, Cultured , Up-Regulation
15.
Hematol J ; 3(3): 157-63, 2002.
Article in English | MEDLINE | ID: mdl-12111653

ABSTRACT

INTRODUCTION: A subset of AML-M2/M4Eo patients has been shown to carry c-kit mutations suggesting that myelomonoblastic leukemia cells, disrupting core binding factor through t(8;21) or inv(16) chromosomal rearrangements, have a common differentiation stage suitable to c-kit mutation. In rare core binding factor leukemia patients an increased dosage of a mutated Asp816(Tyr/Val) kit allele is achieved through nonrandom duplication of chromosome 4 where the c-kit gene is located. MATERIALS AND METHODS: The c-kit gene was studied in the core binding factor leukemia cell line Kasumi-1 with t(8;21) by fluorescence in situ hybridization and mutation analysis. The dosage of Asn822(Lys) mutated allele was evaluated by fluorescence semiquantitative PCR. The correct membrane homing of KIT receptor and its activating status was analysed by immunofluorescence and Western blotting respectively. RESULTS: We identified in the Kasumi-1 cell line a novel Asn822(Lys) ligand-independent c-kit activating mutation and demonstrated by semiquantitative PCR that the mutated allele is about fivefold amplified compared to the normal allele. Fluorescence In Situ Hybridization analysis revealed that c-kit amplification maps to minute 4cen-q11 derived marker chromosome, often carrying duplicated signals, which are unequally distributed in the cell population. The Asn822(Lys) mutation affects a highly conserved codon within the tyrosine kinase activation loop leading, likewise the Asp(816) mutants, to constitutive ligand-independent activation of the KIT receptor. DISCUSSION: Results obtained point to the Kasumi-1 cell line as powerful in-vitro model for further investigation of altered KIT signal transduction pathways in acute myeloid leukemia with core binding factor rearrangements and a useful tool for pharmacological therapeutic targeting.


Subject(s)
Enzyme Activation/genetics , Leukemia, Myeloid/genetics , Mutation, Missense , Proto-Oncogene Proteins c-kit/genetics , Acute Disease , Amino Acid Sequence , Chromosomes, Human, Pair 21 , Chromosomes, Human, Pair 8 , DNA Mutational Analysis , Humans , In Situ Hybridization, Fluorescence , Leukemia, Myeloid/pathology , Molecular Sequence Data , Phosphorylation , Tumor Cells, Cultured
16.
Cancer ; 94(4): 1037-48, 2002 Feb 15.
Article in English | MEDLINE | ID: mdl-11920474

ABSTRACT

BACKGROUND: Many studies found only a small fragment of the large T-antigen coding sequences in human tumors, raising doubts on authenticity of SV40 sequences detected in these samples. METHODS: Five different regions of SV40 DNA were investigated in 106 fresh human tumor biopsies (25 brain, 69 bone, 12 Wilms' tumors), 71 tumor-derived cell cultures (38 from brain and 33 from bone tumors) and normal tissues (5 fresh bone biopsies and 38 buffy coats) by polymerase chain reaction (PCR) techniques and filter hybridization with specific oligoprobes. Expression of SV40 Tag sequences was analyzed in human tumor specimens by RT-PCR. RESULTS: SV40 large T-antigen sequences were detected at high prevalence, in human biopsies of primary brain (37-44%) and bone (21-37%) tumors, in cell cultures derived from brain (30-54%) and bone (53-80%) tumors. SV40 Tag sequences were detected in 29% of buffy coats of blood donors. However, only four brain tumor cell lines showed all the five regions of the SV40 genome investigated. Expression of SV40 Tag sequences was found in 11 of 27 (41%) human tumor samples. DNA sequence analysis indicated that the PCR-amplified products belong to the SV40 wild type. Polymerase chain reaction products of Tag middle portion from 20 of 78 (26%) samples showed a 97% homology with telomeric sequences of human chromosomes 10 and 11. CONCLUSIONS: Authentic SV40 sequences were detected in human samples. The expression of SV40 Tag sequences indicates that SV40 could play a role, as a cofactor, in the onset/progression of specific human cancers. The inability to detect some regions of the virus genome may suggest that those regions are not required for tumor persistence or growth and have been lost or, alternatively, may be the result of assay conditions that were unable to PCR-amplify those regions in the tumors.


Subject(s)
Antigens, Polyomavirus Transforming/genetics , Bone Neoplasms/genetics , Bone Neoplasms/virology , Brain Neoplasms/genetics , Brain Neoplasms/virology , Sequence Alignment , Simian virus 40/genetics , Antigens, Polyomavirus Transforming/analysis , Blood Donors , DNA, Viral/analysis , Humans , Leukocytes/virology , Oligonucleotides , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...