1
|
Lomov NA, Viushkov VS, Rubtsov MA. Mechanisms of Secondary Leukemia Development Caused by Treatment with DNA Topoisomerase Inhibitors. BIOCHEMISTRY. BIOKHIMIIA 2023; 88:892-911. [PMID: 37751862 DOI: 10.1134/s0006297923070040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 04/14/2023] [Accepted: 04/20/2023] [Indexed: 09/28/2023]
Abstract
Leukemia is a blood cancer originating in the blood and bone marrow. Therapy-related leukemia is associated with prior chemotherapy. Although cancer therapy with DNA topoisomerase II inhibitors is one of the most effective cancer treatments, its side effects include development of secondary leukemia characterized by the chromosomal rearrangements affecting AML1 or MLL genes. Recurrent chromosomal translocations in the therapy-related leukemia differ from chromosomal rearrangements associated with other neoplasias. Here, we reviewed the factors that drive chromosomal translocations induced by cancer treatment with DNA topoisomerase II inhibitors, such as mobility of ends of double-strand DNA breaks formed before the translocation and gain of function of fusion proteins generated as a result of translocation.
Collapse
Affiliation(s)
- Nikolai A Lomov
- Department of Molecular Biology, Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia.
| | - Vladimir S Viushkov
- Department of Molecular Biology, Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia
| | - Mikhail A Rubtsov
- Department of Molecular Biology, Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia
- Department of Biochemistry, Center for Industrial Technologies and Entrepreneurship Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119435, Russia
| |
Collapse
|
2
|
ERK Phosphorylation Regulates the Aml1/Runx1 Splice Variants and the TRP Channels Expression during the Differentiation of Glioma Stem Cell Lines. Cells 2021; 10:cells10082052. [PMID: 34440820 PMCID: PMC8391729 DOI: 10.3390/cells10082052] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 08/06/2021] [Accepted: 08/07/2021] [Indexed: 12/15/2022] Open
Abstract
The identification of cancer stem cells in brain tumors paved the way for new therapeutic approaches. Recently, a role for the transcriptional factor Runx1/Aml1 and the downstream ion channel genes in brain cancer development and progression has been suggested. This study aimed to explore the expression and the role of Runx1/Aml1, its Aml1b and Aml1c splice variants and the downstream TRPA1 and TRPV1 ion channels in undifferentiated and day-14 differentiated neural stem cells (NSCs and D-NSCs) and glioblastoma stem cells (GSCs and D-GSCs) lines with different proneural (PN) or mesenchymal (MES) phenotype. Gene and protein expression were evaluated by qRT-PCR, cytofluorimetric, western blot and confocal microscopy analyses. Moreover, by western blot, we observed that ERK phosphorylation enhances the Aml1b and Aml1c protein expression during glioma differentiation. Furthermore, the agonists of TRPA1 and TRPV1 channels stimulated apoptosis/necrosis in GSCs and D-GSCs as evaluated by Annexin V and PI staining and cytofluorimetric analysis. Finally, by qRT-PCR, the modulation of Wnt/β catenin, FGF, and TGFβ/SMAD signaling pathways in PN- and MES-GSCs was reported. Overall, our results provide new evidence regarding Runx1/Aml1 isoform overexpression and modulation in TRP channel expression during gliomagenesis, thus offering new directions for glioblastoma therapy.
Collapse
|
3
|
Chen SF, Wang TZ, Jiang SH, Shen HJ, Xu Y, Zhou HF, Wu DP. [Clinical features and curative effect analysis of patients with myeloid neoplasms and RUNX1 mutations]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2018; 39:983-988. [PMID: 30612398 PMCID: PMC7348223 DOI: 10.3760/cma.j.issn.0253-2727.2018.12.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Indexed: 12/14/2022]
Abstract
Objective: To investigate the survival and prognostic factors of allogeneic hematopoietic stem-cell transplantation (allo-HSCT) for patients with myeloid neoplasms and RUNX1 mutations. Methods: From July 2014 to April 2018, the clinical data of forty-two AML/MDS patients with RUNX1 mutations in the First Affiliated Hospital of Soochow University were retrospectively analyzed. The clinical characteristic features and distribution of the mutations frequently observed with RUNX1 mutations were summarized, the prognosis of allo-HSCT for these patients was also analyzed. Results: Among 42 AML/MDS patients with RUNX1 mutations, 27 were male, 15 were female. The median age was 43.5 (16-68) years old. There are 31 patients in allo-HSCT group and 11 patients in chemotherapy group. RUNX1 mutations co-occurred with many other gene mutations, the most frequent mutations were FLT3 (26.2%, 11/42) . Interestingly, FLT3 mutations only occurred in AML patients compared with MDS patients (P=0.014) . ASXL1 (25%, 3/12) mutations were observed as the most frequent co-mutations in MDS patients. One-year overall survival (OS) , disease-free survival (DFS) of allo-HSCT and chemotherapy patients were (70.6±9.0) %, (61.0±9.4) % and (34.4±16.7) %, (22.4±15.3) %, respectively. When OS and DFS between allo-HSCT and chemotherapy patients were compared, significant differences (χ(2)=4.843, 4.320, P<0.05) were showed. In univariate analysis, transplant age >45 years was a negative effect for OS [HR=4.819 (95% CI 1.145-20.283) , P=0.032] and DFS [HR=5.945 (95% CI 1.715-20.604) , P=0.005]. Also, complex chromosome karyotype abnormality was a negative effect for OS [HR=5.572 (95%CI 1.104-28.113) , P=0.038]. Conclusion: Transplant age (>45 years) and complex chromosome karyotype abnormality were negative prognostic factors in allo-HSCT for myeloid neoplasms patients with RUNX1 mutations.
Collapse
Affiliation(s)
- S F Chen
- First Affiliated Hospital of Soochow University, Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis under Ministry of Health, Suzhou 215006, China
| | | | | | | | | | | | | |
Collapse
|
4
|
An AML1-ETO/miR-29b-1 regulatory circuit modulates phenotypic properties of acute myeloid leukemia cells. Oncotarget 2018; 8:39994-40005. [PMID: 28611288 PMCID: PMC5522207 DOI: 10.18632/oncotarget.18127] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 04/24/2017] [Indexed: 12/21/2022] Open
Abstract
Acute myeloid leukemia (AML) is characterized by an aggressive clinical course and frequent cytogenetic abnormalities that include specific chromosomal translocations. The 8;21 chromosomal rearrangement disrupts the key hematopoietic RUNX1 transcription factor, and contributes to leukemia through recruitment of co-repressor complexes to RUNX1 target genes, altered subnuclear localization, and deregulation of the myeloid gene regulatory program. However, a role of non-coding microRNAs (miRs) in t(8;21)-mediated leukemogenesis is minimally understood. We present evidence of an interplay between the tumor suppressor miR-29b-1 and the AML1-ETO (also designated RUNX1-RUNX1T1) oncogene that is encoded by the t(8;21). We find that AML1-ETO and corepressor NCoR co-occupy the miR-29a/b-1 locus and downregulate its expression in leukemia cells. Conversely, re-introduction of miR-29b-1 in leukemia cells expressing AML1-ETO causes significant downregulation at the protein level through direct targeting of the 3’ untranslated region of the chimeric transcript. Restoration of miR-29b-1 expression in leukemia cells results in decreased cell growth and increased apoptosis. The AML1-ETO-dependent differentiation block and transcriptional program are partially reversed by miR-29b-1. Our findings establish a novel regulatory circuit between the tumor-suppressive miR-29b-1 and the oncogenic AML1-ETO that controls the leukemic phenotype in t(8;21)-carrying acute myeloid leukemia.
Collapse
|
5
|
Fernando TR, Contreras JR, Zampini M, Rodriguez-Malave NI, Alberti MO, Anguiano J, Tran TM, Palanichamy JK, Gajeton J, Ung NM, Aros CJ, Waters EV, Casero D, Basso G, Pigazzi M, Rao DS. The lncRNA CASC15 regulates SOX4 expression in RUNX1-rearranged acute leukemia. Mol Cancer 2017; 16:126. [PMID: 28724437 PMCID: PMC5517805 DOI: 10.1186/s12943-017-0692-x] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 07/03/2017] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Long non-coding RNAs (lncRNAs) play a variety of cellular roles, including regulation of transcription and translation, leading to alterations in gene expression. Some lncRNAs modulate the expression of chromosomally adjacent genes. Here, we assess the roles of the lncRNA CASC15 in regulation of a chromosomally nearby gene, SOX4, and its function in RUNX1/AML translocated leukemia. RESULTS CASC15 is a conserved lncRNA that was upregulated in pediatric B-acute lymphoblastic leukemia (B-ALL) with t (12; 21) as well as pediatric acute myeloid leukemia (AML) with t (8; 21), both of which are associated with relatively better prognosis. Enforced expression of CASC15 led to a myeloid bias in development, and overall, decreased engraftment and colony formation. At the cellular level, CASC15 regulated cellular survival, proliferation, and the expression of its chromosomally adjacent gene, SOX4. Differentially regulated genes following CASC15 knockdown were enriched for predicted transcriptional targets of the Yin and Yang-1 (YY1) transcription factor. Interestingly, we found that CASC15 enhances YY1-mediated regulation of the SOX4 promoter. CONCLUSIONS Our findings represent the first characterization of this CASC15 in RUNX1-translocated leukemia, and point towards a mechanistic basis for its action.
Collapse
Affiliation(s)
- Thilini R Fernando
- Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, USA.,Present Address: Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA, 90048, USA
| | - Jorge R Contreras
- Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, USA.,Cellular and Molecular Pathology Ph.D. Program, UCLA, Los Angeles, USA
| | - Matteo Zampini
- Women and Child Health Department- Hematology-Oncology laboratory, University of Padova, Padova, Italy
| | - Norma I Rodriguez-Malave
- Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, USA.,Cellular and Molecular Pathology Ph.D. Program, UCLA, Los Angeles, USA.,Present Address: Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA, 90048, USA
| | - Michael O Alberti
- Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, USA.,Present Address: Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Jaime Anguiano
- Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, USA.,Present Address: University of San Francisco, 2130 Fulton St, San Francisco, CA, 94117, USA
| | - Tiffany M Tran
- Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, USA.,Molecular, Cellular and Integrative Physiology Ph.D. program, UCLA, Los Angeles, USA
| | - Jayanth K Palanichamy
- Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, USA.,Present Address: All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Jasmine Gajeton
- Microbiology, Immunology and Molecular Genetics Program, UCLA, Los Angeles, USA.,Present Address: Department of Molecular Cardiology Lerner Research Institute, 9500 Euclid Avenue. Cleveland, Cleveland, OH, 44195, USA
| | - Nolan M Ung
- Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, USA
| | - Cody J Aros
- Medical Scientist Training Program, David Geffen School of Medicine, UCLA, Los Angeles, USA
| | - Ella V Waters
- Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, USA.,Present Address: Department of Molecular and Cell Biology, UC Berkeley, Berkeley, USA
| | - David Casero
- Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, USA
| | - Giuseppe Basso
- Women and Child Health Department- Hematology-Oncology laboratory, University of Padova, Padova, Italy
| | - Martina Pigazzi
- Women and Child Health Department- Hematology-Oncology laboratory, University of Padova, Padova, Italy
| | - Dinesh S Rao
- Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, USA. .,Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, USA. .,Broad Stem Cell Research Center, UCLA, 650 Charles E. Young Drive, Factor 12-272, Los Angeles, CA, 90095, USA.
| |
Collapse
|
6
|
Perez Botero J, Chen D, Cousin MA, Majerus JA, Coon LM, Kruisselbrink TM, Klee EW, Lazaridis KN, Pruthi RK, Patnaik MM. Clinical characteristics and platelet phenotype in a family with RUNX1 mutated thrombocytopenia. Leuk Lymphoma 2016; 58:1963-1967. [PMID: 27931139 DOI: 10.1080/10428194.2016.1265118] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Juliana Perez Botero
- a Division of Hematology, Department of Medicine , Mayo Clinic , Rochester , MN , USA
| | - Dong Chen
- b Special Coagulation Laboratory, Department of Laboratory Medicine and Pathology , Mayo Clinic , Rochester , MN , USA.,c Division of Hematopathology, Department of Laboratory Medicine and Pathology , Mayo Clinic , Rochester , MN , USA
| | - Margot A Cousin
- d Center for Individualized Medicine, Department of Health Sciences Research , Mayo Clinic , Rochester , MN , USA
| | - Julie A Majerus
- b Special Coagulation Laboratory, Department of Laboratory Medicine and Pathology , Mayo Clinic , Rochester , MN , USA
| | - Lea M Coon
- b Special Coagulation Laboratory, Department of Laboratory Medicine and Pathology , Mayo Clinic , Rochester , MN , USA.,c Division of Hematopathology, Department of Laboratory Medicine and Pathology , Mayo Clinic , Rochester , MN , USA
| | - Teresa M Kruisselbrink
- d Center for Individualized Medicine, Department of Health Sciences Research , Mayo Clinic , Rochester , MN , USA
| | - Eric W Klee
- e Genetics and Bioinformatics, Department of Health Sciences Research , Mayo Clinic , Rochester , MN , USA
| | - Konstantinos N Lazaridis
- f Division of Gastroenterology and Hepatology, Department of Medicine , Mayo Clinic , Rochester , MN , USA
| | - Rajiv K Pruthi
- a Division of Hematology, Department of Medicine , Mayo Clinic , Rochester , MN , USA.,b Special Coagulation Laboratory, Department of Laboratory Medicine and Pathology , Mayo Clinic , Rochester , MN , USA
| | - Mrinal M Patnaik
- a Division of Hematology, Department of Medicine , Mayo Clinic , Rochester , MN , USA
| |
Collapse
|
7
|
Schmit JM, Turner DJ, Hromas RA, Wingard JR, Brown RA, Li Y, Li MM, Slayton WB, Cogle CR. Two novel RUNX1 mutations in a patient with congenital thrombocytopenia that evolved into a high grade myelodysplastic syndrome. Leuk Res Rep 2015; 4:24-7. [PMID: 25893166 PMCID: PMC4398854 DOI: 10.1016/j.lrr.2015.03.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Revised: 03/23/2015] [Accepted: 03/25/2015] [Indexed: 11/26/2022] Open
Abstract
Here we report two new RUNX1 mutations in one patient with congenital thrombocytopenia that transformed into a high grade myelodysplastic syndrome with myelomonocytic features. The first mutation was a nucleotide base substitution from guanine to adenine within exon 8, resulting in a nonsense mutation in the DNA-binding inhibitory domain of the Runx1 protein. This nonsense mutation is suspected a de novo germline mutation since both parents are negative for the mutation. The second mutation identified was an in-frame six nucleotide base pair insertion in exon 5 of the RUNX1 gene, which is predicted to result in an insertion in the DNA-binding runt homology domain (RHD). This mutation is believed to be a somatic mutation as it was mosaic before allogeneic hematopoietic cell transplantation and disappeared after transplant. As no other genetic mutation was found using genetic screening, it is speculated that the combined effect of these two RUNX1 mutations may have exerted a stronger dominant negative effect than either RUNX1 mutation alone, thus leading to a myeloid malignancy. We report two new RUNX1 mutations in a patient with thrombocytopenia and MDS. We demonstrate that a second hit to RUNX1 results in transformed MDS. Allogeneic transplant was successfully used to treat double RUNX1 mutant MDS.
Collapse
Affiliation(s)
- Jessica M Schmit
- Division of Hematology and Oncology, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Daniel J Turner
- Division of Hematology and Oncology, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Robert A Hromas
- Division of Hematology and Oncology, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL, USA
| | - John R Wingard
- Division of Hematology and Oncology, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Randy A Brown
- Division of Hematology and Oncology, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Ying Li
- Division of Hematopathology, Department of Pathology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Marilyn M Li
- Cancer Genetics Laboratory, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - William B Slayton
- Division of Hematology and Oncology, Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Christopher R Cogle
- Division of Hematology and Oncology, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL, USA
| |
Collapse
|
8
|
Esculetin Downregulates the Expression of AML1-ETO and C-Kit in Kasumi-1 Cell Line by Decreasing Half-Life of mRNA. JOURNAL OF ONCOLOGY 2015; 2015:781473. [PMID: 25861270 PMCID: PMC4377501 DOI: 10.1155/2015/781473] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Accepted: 02/23/2015] [Indexed: 01/05/2023]
Abstract
One of the most frequent genetic aberrations in acute myeloid leukemia (AML) is chromosomal translocation between AML1/RUNX1 on chromosome 21 and ETO gene on chromosome 8 resulting in the expression of chimeric oncogene AML1-ETO. Although patients with t(8;21) translocation have good prognosis, 5-year survival is observed only in 50% of the cases. AML1-ETO translocation is usually accompanied by overexpression of mutant C-Kit, a tyrosine kinase, which contributes to uncontrolled proliferation of premature blood cells leading to relapse and poor prognosis. We illustrate the potential use of esculetin on leukemic cell line, Kasumi-1, bearing t(8;21) translocation and mutated C-Kit gene. Esculetin decreases the expression of AML1-ETO at both protein and transcript level within 24 hours of treatment. Half-life of AML1-ETO mRNA was reduced from 7 hours to 1.5 hours. Similarly half-life of C-Kit mRNA was reduced to 2 hours from 5 hours in esculetin treated cells. Esculetin also perturbed the expression of ectopically expressed AML1-ETO in U937 cells. The decreased expression of AML1-ETO chimeric gene was associated with increased expression of LAT1 and RUNX3 genes, targets of AML1. We envisage that discovery of a drug candidate which could target both these mutated genes would be a considerable breakthrough for future application.
Collapse
|
9
|
Lukman S, Verma CS, Fuentes G. Exploiting Protein Intrinsic Flexibility in Drug Design. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 805:245-69. [DOI: 10.1007/978-3-319-02970-2_11] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
|
10
|
|
11
|
Rulina AV, Spirin PV, Prassolov VS. Activated leukemic oncogenes AML1-ETO and c-kit: role in development of acute myeloid leukemia and current approaches for their inhibition. BIOCHEMISTRY (MOSCOW) 2011; 75:1650-66. [PMID: 21417999 DOI: 10.1134/s0006297910130092] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Acute myeloid leukemia (AML) is a malignant blood disease caused by different mutations that enhance the proliferative activity and survival of blood cells and affect their differentiation and apoptosis. The most frequent disorders in AML are translocations between chromosomes 21 and 8 leading to production of a chimeric oncogene, AML1-ETO, and hyperexpression of the receptor tyrosine kinase KIT. Mutations in these genes often occur jointly. The presence in cells of two activated oncogenes is likely to trigger their malignization. The current approaches for treatment of oncologic diseases (bone marrow transplantation, radiotherapy, and chemotherapy) have significant shortcomings, and thus many laboratories are intensively developing new approaches against leukemias. Inhibiting expression of activated leukemic oncogenes based on the principle of RNA interference seems to be a promising approach in this field.
Collapse
Affiliation(s)
- A V Rulina
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.
| | | | | |
Collapse
|
12
|
Gu J, Ajani JA, Hawk ET, Ye Y, Lee JH, Bhutani MS, Hofstetter WL, Swisher SG, Wang KK, Wu X. Genome-wide catalogue of chromosomal aberrations in barrett's esophagus and esophageal adenocarcinoma: a high-density single nucleotide polymorphism array analysis. Cancer Prev Res (Phila) 2010; 3:1176-86. [PMID: 20651033 DOI: 10.1158/1940-6207.capr-09-0265] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
To better understand the molecular mechanisms behind esophageal adenocarcinoma (EAC) tumorigenesis, we used high-density single nucleotide polymorphism arrays to profile chromosomal aberrations at each of the four sequential progression stages, Barrett's metaplasia (BM), low-grade dysplasia (LGD), high-grade dysplasia (HGD), and EAC, in 101 patients. We observed a significant trend toward increasing loss of chromosomes with higher progression stage. For BM, LGD, HGD, and EAC, respectively, the average numbers of chromosome arms with loss per sample were 0.30, 3.21, 7.70, and 11.90 (P for trend = 4.82 x 10(-7)), and the mean percentages of single nucleotide polymorphisms with allele loss were 0.1%, 1.8%, 6.6%, and 17.2% (P for trend = 2.64 x 10(-6)). In LGD, loss of 3p14.2 (68.4%) and 16q23.1 (47.4%) was limited to narrow regions within the FHIT (3p14.2) and WWOX (16q23.1) genes, whereas loss of 9p21 (68.4%) occurred in larger regions. A significant increase in the loss of other chromosomal regions was seen in HGD and EAC. Loss of 17p (47.6%) was one of the most frequent events in EAC. Many recurrent small regions of chromosomal loss disrupted single genes, including FHIT, WWOX, RUNX1, KIF26B, MGC48628, PDE4D, C20orf133, GMDS, DMD, and PARK2, most of which are common fragile site regions in the human genome. However, RUNX1 at 21q22 seemed to be a potential tumor suppressor gene in EAC. Amplifications were less frequent than losses and mostly occurred in EAC. 8q24 (containing Myc) and 8p23.1 (containing CTSB) were the two most frequently amplified regions. In addition, a significant trend toward increasing amplification was associated with higher progression stage.
Collapse
Affiliation(s)
- Jian Gu
- Department of Epidemiology, The University of Texas M.D. Anderson Cancer Center, Unit 1340, 1155 Pressler Boulevard, Houston, TX 77030, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Baskaran D, Spirin PV, Prassolov VS. Activated leukemic oncogenes responsible for neoplastic transformation of hematopoietic cells. Mol Biol 2010. [DOI: 10.1134/s0026893310030039] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
14
|
Liakhovitskaia A, Gribi R, Stamateris E, Villain G, Jaffredo T, Wilkie R, Gilchrist D, Yang J, Ure J, Medvinsky A. Restoration of Runx1 expression in the Tie2 cell compartment rescues definitive hematopoietic stem cells and extends life of Runx1 knockout animals until birth. Stem Cells 2009; 27:1616-24. [PMID: 19544462 DOI: 10.1002/stem.71] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Mice deficient in the runt homology domain transcription factor Runx1/AML1 fail to generate functional clonogenic hematopoietic cells and die in utero by embryonic day 12.5. We previously generated Runx1 reversible knockout mice, in which the Runx1 locus can be restored by Cre-mediated recombination. We show here that selective restoration of the Runx1 locus in the Tie2 cell compartment rescues clonogenic hematopoietic progenitors in early Runx1-null embryos and rescues lymphoid and myeloid lineages during fetal development. Furthermore, fetal liver cells isolated from reactivated Runx1 embryos are capable of long-term multilineage lymphomyeloid reconstitution of adult irradiated recipients, demonstrating the rescue of definitive hematopoietic stem cells. However, this rescue of the definitive hematopoietic hierarchy is not sufficient to rescue the viability of animals beyond birth, pointing to an essential role for Runx1 in other vital developmental processes.
Collapse
Affiliation(s)
- Anna Liakhovitskaia
- Ontogeny of Haematopoietic Stem Cells Group, Medical Research Council Centre for Regenerative Medicine, Institute for Stem Cell Research, University of Edinburgh, Edinburgh, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Jaffredo T. [The aortic endothelium in the embryo: genesis and role in hematopoiesis]. JOURNAL DE LA SOCIETE DE BIOLOGIE 2009; 203:155-60. [PMID: 19527628 DOI: 10.1051/jbio/2009018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Intra-aortic haematopoiesis is a transient phenomenon, characterised by the emergence of Hematopoietic Stem Cells (HSC) from the ventral aortic endothelium through an endothelial cell (EC) to HSC lineage switch. HSC differentiation is followed by the colonization of definitive haematopoietic organs. Since intra-aortic haematopoiesis is born from EC of the aortic floor, we wondered how vascular integrity was maintained during hematopoietic production. We have used interspecific quail to chick grafts to study the aortic morphogenesis during hematopoiesis. We have demonstrated that: 1) before haematopoiesis, the aortic endothelium, originally entirely from splanchnic origin, was colonized by somitic EC, creating a new roof and sides derived from the somite, whereas the floor was contributed by splanchnopleural-derived EC. 2) As haematopoiesis proceeded, somite-derived EC colonized the aortic floor, where they settled underneath the HSC clusters. 3) After haematopoiesis, splanchnopleural ECs have disappeared from the aortic floor and have been replaced by somite-derived EC. At this stage, the whole aortic endothelium originated from somitic cells. 4) We have identified that the somite contributed to the vascular smooth muscle cells (VSMC). 5) Using grafts of either single quail dermomyotome or sclerotome in the chick, we showed that EC originated from the dermomyotome whereas the vascular smooth muscle cells originated from the sclerotome. Taken together, our results bring about new insights on aorta morphogenesis and the time-restricted production of HSCs.
Collapse
Affiliation(s)
- Thierry Jaffredo
- UPMC, CNRS UMR7622, Laboratoire de Biologie du Développement, 9 quai Saint-Bernard, 75252 Paris Cedex 05, France.
| |
Collapse
|
16
|
Karni S, Soreq H, Sharan R. A network-based method for predicting disease-causing genes. J Comput Biol 2009; 16:181-9. [PMID: 19193144 DOI: 10.1089/cmb.2008.05tt] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
A fundamental problem in human health is the inference of disease-causing genes, with important applications to diagnosis and treatment. Previous work in this direction relied on knowledge of multiple loci associated with the disease, or causal genes for similar diseases, which limited its applicability. Here we present a new approach to causal gene prediction that is based on integrating protein-protein interaction network data with gene expression data under a condition of interest. The latter are used to derive a set of disease-related genes which is assumed to be in close proximity in the network to the causal genes. Our method applies a set-cover-like heuristic to identify a small set of genes that best "cover" the disease-related genes. We perform comprehensive simulations to validate our method and test its robustness to noise. In addition, we validate our method on real gene expression data and on gene specific knockouts. Finally, we apply it to suggest possible genes that are involved in myasthenia gravis.
Collapse
Affiliation(s)
- Shaul Karni
- Blavatnik School of Computer Science, Tel-Aviv University, Tel-Aviv, Israel
| | | | | |
Collapse
|
17
|
Pabst T, Eyholzer M, Haefliger S, Schardt J, Mueller BU. Somatic CEBPA mutations are a frequent second event in families with germline CEBPA mutations and familial acute myeloid leukemia. J Clin Oncol 2008; 26:5088-93. [PMID: 18768433 DOI: 10.1200/jco.2008.16.5563] [Citation(s) in RCA: 138] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE The transcription factor CCAAT/enhancer binding protein-alpha (CEBPA) is crucial for normal myeloid differentiation. Mutations in the CEBPA gene are found in subsets of patients with acute myeloid leukemia (AML). Recently, three families were reported in whom several family members had germline CEBPA mutations and subsequently developed AML. Whereas familial AML is considered a rare event, the frequency of CEBPA germline mutations in AML is not known. PATIENTS AND METHODS In this study, we screened 187 consecutive AML patients for CEBPA mutations at diagnosis. We detected 18 patients (9.6%) with CEBPA mutations. We then analyzed remission samples and constitutive DNA from these patients. RESULTS We found that two (11.1%) of 18 AML patients with CEBPA mutations carried a germline N-terminal frameshift CEBPA mutation. Interestingly, additional members in the families of both of these patients have been affected by AML, and the germline CEBPA mutations were also observed in these patients. Additional somatic mutations in AML patients with germline CEBPA mutations in the two families comprised in-frame C-terminal CEBPA mutations in two patients, two nonsilent CEBPA point mutations in one patient, and monosomy 7 in one patient. CONCLUSION This study shows, for the first time to our knowledge, that germline CEBPA mutations are frequently observed among AML patients with CEBPA mutations. Including the families with germline CEBPA mutations reported previously, additional somatic CEBPA mutations represent a frequent second event in AML with germline CEBPA mutations. Our data strongly indicate that germline CEBPA mutations predispose to AML and that additional somatic CEBPA mutations contribute to the development of the disease.
Collapse
Affiliation(s)
- Thomas Pabst
- Department of Medical Oncology, University Hospital, Bern, Switzerland.
| | | | | | | | | |
Collapse
|
18
|
Five new pedigrees with inherited RUNX1 mutations causing familial platelet disorder with propensity to myeloid malignancy. Blood 2008; 112:4639-45. [PMID: 18723428 DOI: 10.1182/blood-2008-05-156745] [Citation(s) in RCA: 199] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Familial platelet disorder with propensity to myeloid malignancy (FPD/AML) is an autosomal dominant syndrome characterized by platelet abnormalities and a predisposition to myelodysplasia (MDS) and/or acute myeloid leukemia (AML). The disorder, caused by inherited mutations in RUNX1, is uncommon with only 14 pedigrees reported. We screened 10 families with a history of more than one first degree relative with MDS/AML for inherited mutations in RUNX1. Germ- line RUNX1 mutations were identified in 5 pedigrees with a 3:2 predominance of N-terminal mutations. Several affected members had normal platelet counts or platelet function, features not previously reported in FPD/AML. The median incidence of MDS/AML among carriers of RUNX1 mutation was 35%. Individual treatments varied but included hematopoietic stem cell transplantation from siblings before recognition of the inherited leukemogenic mutation. Transplantation was associated with a high incidence of complications including early relapse, failure of engraftment, and posttransplantation lymphoproliferative disorder. Given the small size of modern families and the clinical heterogeneity of this syndrome, the diagnosis of FPD/AML could be easily overlooked and may be more prevalent than previously recognized. Therefore, it would appear prudent to screen young patients with MDS/AML for RUNX1 mutation, before consideration of sibling hematopoietic stem cell transplantation.
Collapse
|
19
|
Owen C, Barnett M, Fitzgibbon J. Familial myelodysplasia and acute myeloid leukaemia--a review. Br J Haematol 2008; 140:123-32. [PMID: 18173751 DOI: 10.1111/j.1365-2141.2007.06909.x] [Citation(s) in RCA: 147] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Familial occurrence of myelodysplasia (MDS) and/or acute myeloid leukaemia (AML) is rare but can provide a useful resource for the investigation of predisposing mutations in these myeloid malignancies. To date, examination of families with MDS/AML has lead to the detection of two culprit genes, RUNX1 and CEBPA. Germline mutations in RUNX1 result in familial platelet disorder with propensity to myeloid malignancy and inherited mutations of CEBPA predispose to AML. Unfortunately, the genetic cause remains obscure in most other reported pedigrees. Further insight into the molecular mechanisms of familial MDS/AML will require awareness by clinicians of new patients with relevant family histories.
Collapse
Affiliation(s)
- Carolyn Owen
- Centre for Medical Oncology, Barts & the London School of Medicine & Dentistry, London, UK.
| | | | | |
Collapse
|
20
|
RUNX1 permits E4orf6-directed nuclear localization of the adenovirus E1B-55K protein and associates with centers of viral DNA and RNA synthesis. J Virol 2008; 82:6395-408. [PMID: 18417565 DOI: 10.1128/jvi.00043-08] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The localization of the adenovirus E1B-55K-E4orf6 protein complex is critical for its function. Prior studies demonstrated that E4orf6 directs the nuclear localization of E1B-55K in human cells and in rodent cells that contain part of human chromosome 21. We show here that the relevant activity on chromosome 21 maps to RUNX1. RUNX1 proteins are transcription factors that serve as scaffolds for the assembly of proteins that regulate transcription and RNA processing. After transfection, the RUNX1a, RUNX1b, and RUNX1-DeltaN variants allowed E4orf6-directed E1B-55K nuclear localization. The failure of RUNX1c to allow nuclear colocalization was relieved by the deletion of amino-terminal residues of this protein. In the adenovirus-infected mouse cell, RUNX1 proteins were localized to discrete structures about the periphery of viral replication centers. These sites are enriched in viral RNA and RNA-processing factors. RUNX1b and RUNX1a proteins displaced E4orf6 from these sites. The association of E1B-55K at viral replication centers was enhanced by the RUNX1a and RUNX1b proteins, but only in the absence of E4orf6. In the presence of E4orf6, E1B-55K occurred in a perinuclear cytoplasmic body resembling the aggresome and was excluded from the nucleus of the infected mouse cell. We interpret these findings to mean that a dynamic relationship exists between the E4orf6, E1B-55K, and RUNX1 proteins. In cooperation with E4orf6, RUNX1 proteins are able to modulate the localization of E1B-55K and even remodel virus-specific structures that form at late times of infection. Subsequent studies will need to determine a functional consequence of the interaction between E4orf6, E1B-55K, and RUNX1.
Collapse
|
21
|
Okumura AJ, Peterson LF, Lo MC, Zhang DE. Expression of AML/Runx and ETO/MTG family members during hematopoietic differentiation of embryonic stem cells. Exp Hematol 2007; 35:978-88. [PMID: 17533052 DOI: 10.1016/j.exphem.2007.03.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2006] [Revised: 02/28/2007] [Accepted: 03/01/2007] [Indexed: 11/20/2022]
Abstract
Runx1/AML1 plays important roles in hematopoiesis, including the commitment of cells to hematopoiesis during embryonic development, and in the maintenance of hematopoietic cell populations. It is also one of the most common genes involved in chromosomal translocations related to leukemia. One such translocation is t(8;21), which fuses the Runx1 gene to the MTG8/ETO gene and generates the Runx1-MTG8 (AML1-ETO) fusion gene. Both Runx1 and MTG8 have two additional family members that are much less studied in hematopoiesis. Here we report the expression of every member of the Runx and MTG families as well as the Runx heterodimerization partner CBFbeta during hematopoietic differentiation of murine embryonic stem cells. We observed substantially increased expression of Runx1, Runx2, and MTG16 during hematopoietic differentiation. Furthermore, the increase in Runx2 expression is delayed relative to Runx1 expression, suggesting their possible sequential contribution to hematopoiesis.
Collapse
Affiliation(s)
- Akiko Joo Okumura
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | | | | | |
Collapse
|
22
|
Abstract
Osteosarcoma is a devastating but rare disease, whose study has illuminated both the basic biology and clinical management of cancer over the past 30 years. These contributions have included insight into the roles of key cancer genes such as the retinoblastoma tumor suppressor gene and TP53, the identification of familial cancer syndromes implicating DNA helicases, and dramatic improvements in survival by the use of adjuvant chemotherapy. This review provides a synoptic overview of our current understanding of the molecular causes of osteosarcoma, and suggests future directions for study.
Collapse
Affiliation(s)
- Maya Kansara
- Ian Potter Foundation Centre for Cancer Genomics and Predictive Medicine and Research Division, Peter MacCallum Cancer Centre, Melbourne, Australia
| | | |
Collapse
|
23
|
Kurokawa M. AML1/Runx1 as a versatile regulator of hematopoiesis: regulation of its function and a role in adult hematopoiesis. Int J Hematol 2006; 84:136-42. [PMID: 16926135 DOI: 10.1532/ijh97.06070] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
AML1/Runx1, originally identified as a gene located at the breakpoint of the t(8;21) translocation, encodes a transcription factor that is widely expressed in multiple hematopoietic lineages and that regulates the expression of a variety of hematopoietic genes. Numerous studies have shown that AML1 is a critical regulator of hematopoietic development. In addition, AML1 is a frequent target for chromosomal translocation in human leukemia. The activity of AML1 can be modulated by various types of posttranslational modification, including phosphorylation and acetylation. Phosphorylation by extracellular signal-regulated kinase (ERK) is one of the mechanisms that dictate whether AML1 acts as either a transcriptional repressor or an activator of gene expression. Recently, a physiological role for AML1 in adult hematopoiesis was revealed by conditional gene targeting in mice. Remarkably, adult hematopoietic progenitors are maintained even in the absence of AML1, in stark contrast to the total disruption of definitive hematopoiesis during embryogenesis. AML1 is, however, critical for megakaryopoiesis and plays an important role in T-cell and B-cell development in adult mice. Recent analyses engineered to recreate hematopoiesis in vitro revealed that the transcriptional activity of AML1 is closely related with the potential of AML1 to generate hematopoietic cells and support thymocyte development.
Collapse
Affiliation(s)
- Mineo Kurokawa
- Department of Hematology and Oncology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan.
| |
Collapse
|
24
|
Thomas D, Kansara M. Epigenetic modifications in osteogenic differentiation and transformation. J Cell Biochem 2006; 98:757-69. [PMID: 16598744 DOI: 10.1002/jcb.20850] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Almost all tumors are characterized by both architectural and cellular abnormalities in differentiation. Osteoblast development is relatively well understood, making osteosarcoma a good model for understanding how tumorigenesis perturbs normal differentiation. We argue that there are two key transition points in normal cellular differentiation that are the focus of oncogenic events, in both of which epigenetic processes are critical. The first is the transition from an uncommitted pluripotent precursor (mesenchymal stem cell) to the 'transit-amplifying compartment' of the osteoblast lineage. This transition, normally exquisitely regulated in space and time, is abnormal in cancer. The second involves termination of lineage expansion, equally tightly regulated under normal circumstances. In cancer, the mechanisms that mandate eventual cessation of cell division are almost universally disrupted. This model predicts that key differentiation genes in bone, such as RUNX2, act in an oncogenic fashion to initiate entry into a proliferative phase of cell differentiation, and anti-oncogenically into the post-mitotic state, resulting in ambivalent roles in tumorigenesis. Polycomb genes exemplify epigenetic processes in the stem cell compartment and tumorigenesis, and are implicated in skeletal development in vivo. The epigenetic functions of the retinoblastoma protein, which plays a key role in tumorigenesis in bone, is discussed in the context of terminal cell cycle exit.
Collapse
Affiliation(s)
- David Thomas
- Ian Potter Foundation Centre for Cancer Genomics and Predictive Medicine, Peter MacCallum Cancer Centre, Victoria 3002, Melbourne, Australia.
| | | |
Collapse
|
25
|
Hamelin V, Letourneux C, Romeo PH, Porteu F, Gaudry M. Thrombopoietin regulates IEX-1 gene expression through ERK-induced AML1 phosphorylation. Blood 2006; 107:3106-13. [PMID: 16368886 DOI: 10.1182/blood-2005-07-2953] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Abstract
The extracellular signal-regulated kinases (ERKs) are required for thrombopoietin (TPO) functions on hematopoietic cells, but the ERKs targets involved remain unknown. Here we show that the regulation of the immediate early gene X-1 (IEX-1), identified as an ERK substrate in response to TPO, was mediated by an ERK-dependent phosphorylation of AML1. The addition of TPO to UT7-Mpl cells and primary megakaryocytes induced gene expression of IEX-1. Neither erythropoietin (EPO) nor granulocyte macrophage-colony stimulating factor (GM-CSF) was able to activate IEX-1 gene expression in UT7-Mpl cells. The induced expression was mediated by a transcriptional activation of the IEX-1 promoter and required an AML1-binding site located at –1068. The direct involvement of AML1 in the regulation of IEX-1 gene expression was shown by both the use of AML1 mutants and by shRNA experiments targeting endogenous AML1. Finally, the ability of TPO to induce the IEX-1 gene expression was inhibited by U0126, a specific inhibitor of the ERKs activator MEK and AML1 transcriptional activity was shown to be modulated by TPO through ERK-dependent phosphorylation. Taken together, these data suggest that AML1 plays a role in modulating the IEX-1 expression and that the ERK-dependent AML1 phosphorylation regulates the TPO-mediated activation of IEX-1.
Collapse
|
26
|
Abstract
Runx1/AML1 (also known as CBFA2 and PEBP23B) is a Runt family transcription factor critical for normal hematopoiesis. Runx1 forms a heterodimer with CBF3 and binds to the consensus PEBP2 sequence through the Runt domain. Runx1 enhances gene transcription by interacting with transcriptional coactivators such as p300 and CREB-binding protein. However, Runx1 can also suppress gene transcription by interacting with transcriptional corepressors, including mSin3A, TLE (mammalian homolog of Groucho), and histone deacetylases. Runx1 not only is critical for definitive hematopoiesis in the fetus but also is required for normal megakaryocytic maturation and T-lymphocyte and B-lymphocyte development in adult mice. Runx1 has been identified in leukemia-associated chromosomal translocations, including t(8;21) (Runx1-ETO/MTG8), t(16;21) (Runx1-MTG16), t(3;21) (Runx1-Evi1), t(12;21) (TEL-Runx1), and t(X;21) (Runx1-Fog2). The molecular mechanism of leukemogenesis by these fusion proteins is discussed. Various mutant mice expressing these fusion proteins have been created. However, expression of the fusion protein is not sufficient by itself to cause leukemia and likely requires additional events for leukemogenesis. Point mutations in a Runx1 allele cause haploinsufficiency and a biallelic null for Runx1, which are associated with familial platelet disorder with a propensity for acute myeloid leukemia (FPD/AML) and AML-M0, respectively. Thus, the correct protein structure and the precise dosage of Runx1 are essential for the maintenance of normal hematopoiesis.
Collapse
Affiliation(s)
- Tetsuya Yamagata
- Section on Immunology and Immunogenetics, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | | | | |
Collapse
|
27
|
Samokhvalov IM, Thomson AM, Lalancette C, Liakhovitskaia A, Ure J, Medvinsky A. Multifunctional reversible knockout/reporter system enabling fully functional reconstitution of the AML1/Runx1 locus and rescue of hematopoiesis. Genesis 2006; 44:115-21. [PMID: 16496309 DOI: 10.1002/gene.20190] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Mice deficient in the runt homology domain transcription factor Runx1 die of severe anemia in utero by embryonic day (E)12.5. A reactivatable Runx1 knockout embryonic stem cell (ESC) and mouse systems were generated by the targeted insertion of a loxP-flanked multipartite gene stop/trap cassette designed to simultaneously ablate the expression of Runx1 and report on the activity of its promoters. The cassette's in-frame LacZ reporter enabled activities of the proximal and the distal promoters to be differentially monitored. Although Runx1-null ESCs were capable of primitive erythroid differentiation in vitro, their capacity to generate granulocyte/macrophage or mixed myelo-erythroid embryoid bodies was lost. Cre-mediated reactivation restored Runx1 structural integrity and rescued the hematopoietic differentiation potential of ESCs. Mice with the reactivated allele survived, showed no hematopoietic deficit, and expressed all major splice isoforms of Runx1 appropriately. This multipurpose mouse model will be useful for the analysis of the critical Runx1-dependent check-point(s) in hematopoietic development.
Collapse
Affiliation(s)
- Igor M Samokhvalov
- Developmental Haematopoiesis Laboratory, Institute for Stem Cell Research, University of Edinburgh, Edinburgh EH9 3JQ, Scotland, United Kingdom
| | | | | | | | | | | |
Collapse
|
28
|
Senyuk V, Li D, Zakharov A, Mikhail FM, Nucifora G. The Distal Zinc Finger Domain of AML1/MDS1/EVI1 Is an Oligomerization Domain Involved in Induction of Hematopoietic Differentiation Defects in Primary Cells In vitro. Cancer Res 2005; 65:7603-11. [PMID: 16140925 DOI: 10.1158/0008-5472.can-05-0412] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
AML1/MDS1/EVI1 (AME) is a chimeric transcription factor produced by the (3;21)(q26;q22) translocation. This chromosomal translocation is associated with de novo and therapy-related acute myeloid leukemia and with the blast crisis of chronic myelogenous leukemia. AME is obtained by in-frame fusion of the AML1 and MDS1/EVI1 (ME) genes. The mechanisms by which AME induces a neoplastic transformation in bone marrow cells are unknown. AME interacts with the corepressors CtBP and HDAC1, and it was shown that AME is a repressor in contrast to the parent transcription factors AML1 and ME, which are transcription activators. Studies with murine bone marrow progenitors indicated that the introduction of a point mutation that destroys the CtBP-binding consensus impairs but does not abolish the disruption of cell differentiation and replication associated with AME expression, suggesting that additional events are required. Several chimeric proteins, such as AML1/ETO, BCR/ABL, and PML/RARa, are characterized by the presence of a self-interaction domain critical for transformation. We report that AME is also able to oligomerize and displays a complex pattern of self-interaction that involves at least three oligomerization regions, one of which is the distal zinc finger domain. Although the deletion of this short domain does not preclude the self-interaction of AME, it significantly reduces the differentiation defects caused in vitro by AME in primary murine bone marrow progenitors. The addition of a point mutation that inhibits CtBP binding completely abrogates the effects of AME on differentiation, suggesting that AME induces hematopoietic differentiation defects through at least two separate but cooperating pathways.
Collapse
Affiliation(s)
- Vitalyi Senyuk
- Department of Pathology and The Cancer Center, University of Illinois at Chicago, Chicago, Illinois 60607, USA.
| | | | | | | | | |
Collapse
|
29
|
Gutiérrez-Angulo M, González-García JR, Meza-Espinoza JP, Picos-Cárdenas VJ, Esparza-Flores MA, López-Guido B, Rivera H. Increased expression of AML1-a and acquired chromosomal abnormalities in childhood acute lymphoblastic leukemia. Hematol Oncol 2005; 22:85-90. [PMID: 15991223 DOI: 10.1002/hon.734] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
A semi-quantitative expression analysis of both AML1-a and AML1-total was performed by RT-PCR in 19 children with acute lymphoblastic leukemia (ALL) at diagnosis. AML1-a expression was assessed in 16 bone marrow (BM) and 13 peripheral blood (PB) samples whereas AML1-total was assessed in 17 BM and 16 PB samples. These analyses were also carried out in 15 PB samples of healthy controls. In addition, 18/19 patients were karyotyped: 11 had an unmodified constitutional karyotype (CK) and seven exhibited acquired chromosomal abnormalities (ACA). The expression of AML1-a was significantly increased in BM and PB when compared with the controls (p < 0.013 and p < 0.035, respectively). A significant increase was found in the expression of AML1-a in BM of the ACA group compared with the CK group (p < 0.0009). The expression of AML1-a in BM and PB showed a significant increase in the ACA group compared with controls (p < 0.00001 and p < 0.012, respectively); in contrast, the CK group did not differ from the controls. These observations may mean that the increase of AML1-a favours the progression of leukemia.
Collapse
Affiliation(s)
- M Gutiérrez-Angulo
- División de Genética, Centro de Investigación Biomédica de Occidente, IMSS, Tepatitlán, Jalisco, México
| | | | | | | | | | | | | |
Collapse
|
30
|
Thomas DM, Johnson SA, Sims NA, Trivett MK, Slavin JL, Rubin BP, Waring P, McArthur GA, Walkley CR, Holloway AJ, Diyagama D, Grim JE, Clurman BE, Bowtell DDL, Lee JS, Gutierrez GM, Piscopo DM, Carty SA, Hinds PW. Terminal osteoblast differentiation, mediated by runx2 and p27KIP1, is disrupted in osteosarcoma. ACTA ACUST UNITED AC 2005; 167:925-34. [PMID: 15583032 PMCID: PMC2172443 DOI: 10.1083/jcb.200409187] [Citation(s) in RCA: 171] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The molecular basis for the inverse relationship between differentiation and tumorigenesis is unknown. The function of runx2, a master regulator of osteoblast differentiation belonging to the runt family of tumor suppressor genes, is consistently disrupted in osteosarcoma cell lines. Ectopic expression of runx2 induces p27KIP1, thereby inhibiting the activity of S-phase cyclin complexes and leading to the dephosphorylation of the retinoblastoma tumor suppressor protein (pRb) and a G1 cell cycle arrest. Runx2 physically interacts with the hypophosphorylated form of pRb, a known coactivator of runx2, thereby completing a feed-forward loop in which progressive cell cycle exit promotes increased expression of the osteoblast phenotype. Loss of p27KIP1 perturbs transient and terminal cell cycle exit in osteoblasts. Consistent with the incompatibility of malignant transformation and permanent cell cycle exit, loss of p27KIP1 expression correlates with dedifferentiation in high-grade human osteosarcomas. Physiologic coupling of osteoblast differentiation to cell cycle withdrawal is mediated through runx2 and p27KIP1, and these processes are disrupted in osteosarcoma.
Collapse
Affiliation(s)
- David M Thomas
- Ian Potter Foundation Centre for Cancer Genomics and Predictive Medicine, and Sir Donald and Lady Trescowthick Laboratories, Peter MacCallum Cancer Center, Victoria, Melbourne, Australia.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Planagumà J, Díaz-Fuertes M, Gil-Moreno A, Abal M, Monge M, García A, Baró T, Thomson TM, Xercavins J, Alameda F, Reventós J. A Differential Gene Expression Profile Reveals Overexpression of RUNX1/AML1 in Invasive Endometrioid Carcinoma. Cancer Res 2004; 64:8846-53. [PMID: 15604243 DOI: 10.1158/0008-5472.can-04-2066] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Endometrial carcinoma is the most common gynecological malignant disease in industrialized countries. Two clinicopathological types of endometrial carcinoma have been described, based on estrogen relation and grade: endometrioid carcinoma (EEC) and non-EEC (NEEC). Some of the molecular events that occur during the development of endometrial carcinoma have been characterized, showing a dualistic genetic model for EEC and NEEC. However, the molecular bases for endometrial tumorigenesis are not clearly elucidated. In the present work, we attempted to identify new genes that could trigger cell transformation in EEC. We analyzed the differential gene expression profile between tumoral and nontumoral endometrial specimens with cDNA array hybridization. Among the 53 genes for which expression was found to be altered in EEC, the acute myeloid leukemia proto-oncogene, RUNX1/AML1, was one of the most highly up-regulated. The gene expression levels of RUNX1/AML1 were quantified by real-time quantitative PCR, and protein levels were characterized by tissue array immunohistochemistry. Real-time quantitative PCR validated RUNX1/AML1 up-regulation in EEC and demonstrated a specific and significantly stronger up-regulation in those tumor stages associated with myometrial invasion. Furthermore, tissue array immunohistochemistry showed that RUNX1/AML1 up-regulation correlates to the process of tumorigenesis, from normal atrophic endometrium to simple and complex hyperplasia and then, on to carcinoma. These results demonstrate for the first time the up-regulation of RUNX1/AML1 in EEC correlating with the initial steps of myometrial infiltration.
Collapse
Affiliation(s)
- Jesús Planagumà
- Unitat de Recerca Biomèdica, Institut de Recerca del Hospital Universitari Vall d'Hebron, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
North TE, Stacy T, Matheny CJ, Speck NA, de Bruijn MFTR. Runx1 is expressed in adult mouse hematopoietic stem cells and differentiating myeloid and lymphoid cells, but not in maturing erythroid cells. ACTA ACUST UNITED AC 2004; 22:158-68. [PMID: 14990855 DOI: 10.1634/stemcells.22-2-158] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The transcription factor Runx1 marks all functional hematopoietic stem cells (HSCs) in the embryo and is required for their generation. Mutations in Runx1 are found in approximately 25% of acute leukemias and in familial platelet disorder, suggesting a role for Runx1 in adult hematopoiesis as well. A comprehensive analysis of Runx1 expression in adult hematopoiesis is lacking. Here we show that Runx1 is expressed in functional HSCs in the adult mouse, as well as in cells with spleen colony-forming unit (CFU) and culture CFU capacities. Additionally, we document Runx1 expression in all hematopoietic lineages at the single cell level. Runx1 is expressed in the majority of myeloid cells and in a smaller proportion of lymphoid cells. Runx1 expression substantially decreases during erythroid differentiation. We also document effects of reduced Runx1 levels on adult hematopoiesis.
Collapse
Affiliation(s)
- Trista E North
- Department of Biochemistry, Dartmouth Medical School, Hanover, New Hampshire, USA. (Current address for Dr. de Bruijn) MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford, United Kingdom
| | | | | | | | | |
Collapse
|
33
|
Pick M, Flores-Flores C, Soreq H. From brain to blood: alternative splicing evidence for the cholinergic basis of Mammalian stress responses. Ann N Y Acad Sci 2004; 1018:85-98. [PMID: 15240356 DOI: 10.1196/annals.1296.010] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Three principal features of mammalian stress responses are that they span peripheral and CNS changes, modify blood cell composition and activities, and cover inter-related alterations in a large number of gene products. The finely tuned spatiotemporal regulation of these multiple events suggests the hierarchic involvement of modulatory neurotransmitters and modified process(es) in the pathway of gene expression that together would enable widely diverse stress responses. We report evidence supporting the notion that acetylcholine (ACh) acts as a stress-response-regulating transmitter and that altered ACh levels are variously associated with changes in the alternative splicing of pre-mRNA transcripts in brain neurons and peripheral blood cells. We used acetylcholinesterase (AChE) gene expression as a case study and developed distinct probes for its alternative splice variants at the mRNA and protein levels. In laboratory animals and human-derived cells, we found stress-induced changes in the alternative splicing patterns of AChE pre-mRNA, which attributes to this gene and its different protein products diverse stress responsive functions that are associated with the enzymatic and noncatalytic properties of AChE. Together, these approaches provide a conceptually unified view of the studied pathways for controlling stress responses in brain and blood.
Collapse
Affiliation(s)
- Marjorie Pick
- Department of Biological Chemistry, The Hebrew University of Jerusalem Safra Campus-Givat Ram, Jerusalem 91904, Israel.
| | | | | |
Collapse
|
34
|
|
35
|
Perry C, Sklan EH, Soreq H. CREB regulates AChE-R-induced proliferation of human glioblastoma cells. Neoplasia 2004; 6:279-86. [PMID: 15153340 PMCID: PMC1502102 DOI: 10.1593/neo.3424] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2003] [Revised: 12/09/2003] [Accepted: 12/11/2003] [Indexed: 11/18/2022]
Abstract
The cyclic adenosine monophosphate (AMP) response element-binding protein, CREB, often modulates stress responses. Here, we report that CREB suppresses the glioblastoma proliferative effect of the stress-induced acetylcholinesterase variant, AChE-R. In human U87MG glioblastoma cells, AChE-R formed a triple complex with protein kinase C (PKC) epsilon and the scaffold protein RACK1, enhanced PKCepsilon phosphorylation, and facilitated BrdU incorporation. Either overexpressed CREB, or antisense destruction of AChE-R mRNA, PKC, or protein kinase A (PKA) inhibitors-but not CREB combined with PKC inhibition suppressed-this proliferation, suggesting that CREB's repression of this process involves a PKC-mediated pathway, whereas impaired CREB regulation allows AChE-R-induced, PKA-mediated proliferation of glioblastoma tumors.
Collapse
Affiliation(s)
- Chava Perry
- Department of Biological Chemistry, The Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 91904, Israel
- Department of Hematology, The Tel-Aviv Sourasky Medical Center-Tel Aviv and Tel-Aviv University, Tel-Aviv 64239, Israel
| | - Ella H Sklan
- Department of Biological Chemistry, The Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 91904, Israel
| | - Hermona Soreq
- Department of Biological Chemistry, The Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 91904, Israel
| |
Collapse
|
36
|
Liu H, Holm M, Xie XQ, Wolf-Watz M, Grundström T. AML1/Runx1 recruits calcineurin to regulate granulocyte macrophage colony-stimulating factor by Ets1 activation. J Biol Chem 2004; 279:29398-408. [PMID: 15123671 DOI: 10.1074/jbc.m403173200] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Acute myeloid leukemia 1 (AML1), also denoted Runx1, is a transcription factor essential for hematopoiesis, and the AML1 gene is the most common target of chromosomal translocations in human leukemias. AML1 binds to sequences present in the regulatory regions of a number of hematopoiesis-specific genes, including certain cytokines such as granulocyte macrophage colony-stimulating factor (GM-CSF) up-regulated after T cell receptor stimulation. Here we show that both subunits of the Ca(2+)/calmodulin-dependent protein phosphatase calcineurin (CN), which is activated upon T cell receptor stimulation, interact directly with the N-terminal runt homology domain-containing part of AML1. The regulatory CN subunit binds AML1 with a higher affinity and in addition also interacts with the isolated runt homology domain. The related Runx2 transcription factor, which is essential for bone formation, also interacts with CN. A constitutively active derivative of CN is shown to activate synergistically the GM-CSF promoter/enhancer together with AML1 or Runx2. We also provide evidence that relief of the negative effect of the AML1 sites is important for Ca(2+) activation of the GM-CSF promoter/enhancer and that AML1 overexpression increases this Ca(2+) activation. Both subunits of CN interact with AML1 in coimmunoprecipitation analyses, and confocal microscopy analysis of cells expressing fluorescence-tagged protein derivatives shows that CN can be recruited to the nucleus by AML1 in vivo. Mutant analysis of the GM-CSF promoter shows that the Ets1 binding site of the promoter is essential for the synergy between AML1 and CN in Jurkat T cells. Analysis of the effects of inhibitors of the protein kinase glycogen synthase kinase-3beta and in vitro phosphorylation/dephosphorylation analysis of Ets1 suggest that glycogen synthase kinase-3beta-phosphorylated Ets1 is a target of AML1-recruited CN phosphatase at the GM-CSF promoter.
Collapse
Affiliation(s)
- Hebin Liu
- Department of Molecular Biology, Umeå University, Umeå S-901 87, Sweden
| | | | | | | | | |
Collapse
|
37
|
Hug BA, Ahmed N, Robbins JA, Lazar MA. A Chromatin Immunoprecipitation Screen Reveals Protein Kinase Cβ as a Direct RUNX1 Target Gene. J Biol Chem 2004; 279:825-30. [PMID: 14561740 DOI: 10.1074/jbc.m309524200] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
RUNX1 (also known as AML1) is a DNA-binding transcription factor that functions as a tumor suppressor and developmental determinant in hematopoietic cells. Target promoters have been identified primarily through the use of differential expression strategies and candidate gene approaches but not biochemical screens. Using a chromatin immunoprecipitation screen, we identified protein kinase Cbeta as a direct RUNX1 target gene and demonstrate that endogenous RUNX1 binds the chromatinized protein kinase Cbeta promoter of U937 cells. A phylogenetically conserved RUNX1-binding site within the PKCbeta promoter binds RUNX1 in electrophoretic mobility shift analyses and confers RUNX1 responsiveness on a heterologous promoter. Changes in RUNX1 activity affect endogenous protein kinase Cbeta expression, and a dominant-negative form of RUNX1 protects U937 cells from apoptotic stimuli previously shown to be dependent on protein kinase Cbeta. This protection can be reversed by the ectopic expression of protein kinase Cbeta. Together these findings demonstrate that protein kinase Cbeta is a direct, downstream target of RUNX1 and links RUNX1 to a myeloid apoptotic pathway.
Collapse
Affiliation(s)
- Bruce A Hug
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Pennsylvania Medical Center, Philadelphia, PA 19104, USA
| | | | | | | |
Collapse
|
38
|
Kurokawa M, Hirai H. Role of AML1/Runx1 in the pathogenesis of hematological malignancies. Cancer Sci 2003; 94:841-6. [PMID: 14556655 PMCID: PMC11160144 DOI: 10.1111/j.1349-7006.2003.tb01364.x] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2003] [Accepted: 08/20/2003] [Indexed: 11/26/2022] Open
Abstract
AML1/Runx1, originally identified as a gene located at the breakpoint of the t(8;21) translocation, encodes one of the two subunits forming a heterodimeric transcription factor. AML1 contains a highly evolutionally conserved domain called the Runt domain, responsible for both DNA binding and heterodimerization with the partner protein, CBFbeta. AML1 is widely expressed in all hematopoietic lineages, and regulates the expression of a variety of hematopoietic genes. Numerous studies have shown that AML is a critical regulator of hematopoietic development. In addition, AML1 and CBFbeta are frequent targets for chromosomal translocation in human leukemia. Translocations lead to the generation of fusion proteins, which play a causative role for the development of leukemia, primarily by inhibiting AML1 function. Point mutations that impair AML1 function are also associated with familial and sporadic leukemias. Loss of AML1 function is thus implicated in a number of leukemias through multiple pathogenic mechanisms. However, AML1-related translocations or haploinsufficiency of AML1 are not immediately leukemogenic in animal models, suggesting that additional genetic events are required for the development of full-blown leukemia.
Collapse
Affiliation(s)
- Mineo Kurokawa
- Department of Hematology and Oncology, Graduate School of Medicine, University of Tokyo, Bunkyo-ku, Tokyo113-8655, Japan
| | | |
Collapse
|
39
|
Tonks A, Pearn L, Tonks AJ, Pearce L, Hoy T, Phillips S, Fisher J, Downing JR, Burnett AK, Darley RL. The AML1-ETO fusion gene promotes extensive self-renewal of human primary erythroid cells. Blood 2003; 101:624-32. [PMID: 12393523 DOI: 10.1182/blood-2002-06-1732] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
The t(8;21) translocation, which encodes the AML1-ETO fusion protein (now known as RUNX1-CBF2T1), is one of the most frequent translocations in acute myeloid leukemia, although its role in leukemogenesis is unclear. Here, we report that exogenous expression of AML1-ETO in human CD34(+) cells severely disrupts normal erythropoiesis, resulting in virtual abrogation of erythroid colony formation. In contrast, in bulk liquid culture of purified erythroid cells, we found that while AML1-ETO initially inhibited proliferation during early (erythropoietin [EPO]-independent) erythropoiesis, growth inhibition gave way to a sustained EPO-independent expansion of early erythroid cells that continued for more than 60 days, whereas control cultures became growth arrested after 10 to 13 days (at the EPO-dependent stage of development). Phenotypic analysis showed that although these cells were CD13(-) and CD34(-), unlike control cultures, these cells failed to up-regulate CD36 or to down-regulate CD33, suggesting that expression of AML1-ETO suppressed the differentiation of these cells and allowed extensive self-renewal to occur. In the early stages of this expansion, addition of EPO was able to promote both phenotypic (CD36(+), CD33(-), glycophorin A(+)) and morphologic differentiation of these cells, almost as effectively as in control cultures. However, with extended culture, cells expressing AML1-ETO became refractory to addition of this cytokine, suggesting that a block in differentiation had been established. These data demonstrate the capacity of AML1-ETO to promote the self-renewal of human hematopoietic cells and therefore support a causal role for t(8;21) translocations in leukemogenesis.
Collapse
Affiliation(s)
- Alex Tonks
- Department of Haematology, University of Wales College of Medicine, Cardiff, United Kingdom.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Shim KS, Ferrando-Miguel R, Lubec G. Aberrant protein expression of transcription factors BACH1 and ERG, both encoded on chromosome 21, in brains of patients with Down syndrome and Alzheimer’s disease. ADVANCES IN DOWN SYNDROME RESEARCH 2003:39-49. [PMID: 15068237 DOI: 10.1007/978-3-7091-6721-2_3] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Down syndrome (DS; trisomy 21) is a genetic disorder associated with early mental retardation and patients inevitably develop Alzheimer's disease (AD)-like neuropathological changes. The molecular defects underlying the DS-phenotype may be due to overexpression of genes encoded on chromosome 21. This so-called gene dosage hypothesis is still controversial and demands systematic work on protein expression. A series of transcription factors (TF) are encoded on chromosome 21 and are considered to play a pathogenetic role in DS. We therefore decided to study brain expression of TF encoded on chromosome 21 in patients with DS and AD compared to controls: Frontal cortex of 6 male DS patients, 6 male patients with AD and 6 male controls were used for the experiments. Immunoblotting was used to determine protein levels of TF BACH1, ERG, SIM2 and RUNX1. SIM2 and RUNX1 were comparable between groups, while BACH1 was significantly reduced in DS, and ERG was increased in DS and AD as compared to controls. These findings may indicate that DS pathogenesis cannot be simply explained by the gene dosage effect hypothesis and that results of ERG expression in DS were paralleling those in AD probably reflecting a common pathogenetic mechanism possibly explaining why all DS patients develop AD like neuropathology from the fourth decade. We conclude that TF derangement is not only due to the process of neurodegeneration and propose that TFs BACH1 and ERG play a role for the development of AD-like neuropathology in DS and pathogenesis of AD per se and the manifold increase of ERG in both disorders may form a pivotal pathogenetic link.
Collapse
Affiliation(s)
- K S Shim
- Department of Pediatrics, University of Vienna, Vienna, Austria
| | | | | |
Collapse
|
41
|
Perry C, Sklan EH, Birikh K, Shapira M, Trejo L, Eldor A, Soreq H. Complex regulation of acetylcholinesterase gene expression in human brain tumors. Oncogene 2002; 21:8428-41. [PMID: 12466963 DOI: 10.1038/sj.onc.1205945] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2002] [Revised: 07/31/2002] [Accepted: 08/07/2002] [Indexed: 11/09/2022]
Abstract
To study the regulation of acetylcholinesterase (AChE) gene expression in human brain tumors, 3' splice variants of AChE mRNA and potentially relevant transcription factor mRNAs were labeled in primary astrocytomas and melanomas. AChE-S and AChE-R mRNA, as well as Runx1/AML1 mRNA accumulated in astrocytomas in correlation with tumor aggressiveness, but neither HNF3beta nor c-fos mRNA was observed in melanoma and astrocytomas. Immunohistochemistry demonstrated nuclear Runx1/AML1 and cellular AChE-S and AChE-R in melanomas, however, only AChE-S, and not the secreted AChE-R variant, was retained in astrocyte tumor cells. Runx1/AML1 revealed weak linkage with ACHE promoter sequences, yet enhanced ACHE gene expression in co-transfected COS1 cells. The p300 co-activator and the ACHE promoter's distal enhancer facilitated this effect, which was independent of much of the Runx1/AML1 trans-activation domain. Surprisingly, GASP, a fusion product of green fluorescence protein (GFP) and ASP(67), a peptide composed of the 67 C-terminal amino acid residues of AChE-S, localized to COS1 cell nuclei. However, GARP, the corresponding fusion product of GFP with a peptide having the 51 C-terminal residues of AChE-E or GFP alone, remained cytoplasmic. Runx1/AML1 exhibited improved nuclear retention in GASP-expressing COS1 cells, suggesting modulated nuclear localization processes. Together, these findings reveal brain tumor-specific regulation of both expression and cellular retention of variant ACHE gene products.
Collapse
Affiliation(s)
- Chava Perry
- Department of Biological Chemistry, The Institute of Life Sciences, The Hebrew University of Jerusalem, Israel 91904
| | | | | | | | | | | | | |
Collapse
|
42
|
Abstract
The RUNX family of transcription factors plays pivotal roles during normal development and in neoplasias. Recent data involve RUNX3 as an important tumor suppressor in gastric cancers and pose interesting questions about how perturbed levels and interspecific competition among RUNX family members may contribute to tumorigenesis.
Collapse
Affiliation(s)
- Anders H Lund
- Division of Molecular Genetics, The Netherlands Cancer Institute, Antoni van Leeuwenhoek ziekenhuis, Plesmanlaan 121, 1066CX, Amsterdam, The Netherlands
| | | |
Collapse
|