1
|
Sankar S, Patterson E, Lewis EM, Waller LE, Tong C, Dearborn J, Wozniak D, Rubin JB, Kroll KL. Geminin deficiency enhances survival in a murine medulloblastoma model by inducing apoptosis of preneoplastic granule neuron precursors. Genes Cancer 2017; 8:725-744. [PMID: 29234490 PMCID: PMC5724806 DOI: 10.18632/genesandcancer.157] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Medulloblastoma is the most common malignant brain cancer of childhood. Further understanding of tumorigenic mechanisms may define new therapeutic targets. Geminin maintains genome fidelity by controlling re-initiation of DNA replication within a cell cycle. In some contexts, Geminin inhibition induces cancer-selective cell cycle arrest and apoptosis and/or sensitizes cancer cells to Topoisomerase IIα inhibitors such as etoposide, which is used in combination chemotherapies for medulloblastoma. However, Geminin's potential role in medulloblastoma tumorigenesis remained undefined. Here, we found that Geminin is highly expressed in human and mouse medulloblastomas and in murine granule neuron precursor (GNP) cells during cerebellar development. Conditional Geminin loss significantly enhanced survival in the SmoA1 mouse medulloblastoma model. Geminin loss in this model also reduced numbers of preneoplastic GNPs persisting at one postnatal month, while at two postnatal weeks these cells exhibited an elevated DNA damage response and apoptosis. Geminin knockdown likewise impaired human medulloblastoma cell growth, activating G2 checkpoint and DNA damage response pathways, triggering spontaneous apoptosis, and enhancing G2 accumulation of cells in response to etoposide treatment. Together, these data suggest preneoplastic and cancer cell-selective roles for Geminin in medulloblastoma, and suggest that targeting Geminin may impair tumor growth and enhance responsiveness to Topoisomerase IIα-directed chemotherapies.
Collapse
Affiliation(s)
- Savita Sankar
- Department of Developmental Biology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Ethan Patterson
- Department of Developmental Biology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Emily M Lewis
- Department of Developmental Biology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Laura E Waller
- Department of Developmental Biology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Caili Tong
- Department of Developmental Biology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Joshua Dearborn
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA
| | - David Wozniak
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA
| | - Joshua B Rubin
- Department of Pediatrics, Washington University School of Medicine, Saint Louis, MO, USA
| | - Kristen L Kroll
- Department of Developmental Biology, Washington University School of Medicine, Saint Louis, MO, USA
| |
Collapse
|
2
|
Salm F, Dimitrova V, von Bueren AO, Ćwiek P, Rehrauer H, Djonov V, Anderle P, Arcaro A. The Phosphoinositide 3-Kinase p110α Isoform Regulates Leukemia Inhibitory Factor Receptor Expression via c-Myc and miR-125b to Promote Cell Proliferation in Medulloblastoma. PLoS One 2015; 10:e0123958. [PMID: 25915540 PMCID: PMC4411098 DOI: 10.1371/journal.pone.0123958] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 02/26/2015] [Indexed: 12/22/2022] Open
Abstract
Medulloblastoma (MB) is the most common malignant brain tumor in childhood and represents the main cause of cancer-related death in this age group. The phosphoinositide 3-kinase (PI3K) pathway has been shown to play an important role in the regulation of medulloblastoma cell survival and proliferation, but the molecular mechanisms and downstream effectors underlying PI3K signaling still remain elusive. The impact of RNA interference (RNAi)-mediated silencing of PI3K isoforms p110α and p110δ on global gene expression was investigated by DNA microarray analysis in medulloblastoma cell lines. A subset of genes with selectively altered expression upon p110α silencing in comparison to silencing of the closely related p110δ isoform was revealed. Among these genes, the leukemia inhibitory factor receptor α (LIFR α) was validated as a novel p110α target in medulloblastoma. A network involving c-Myc and miR-125b was shown to be involved in the control of LIFRα expression downstream of p110α. Targeting the LIFRα by RNAi, or by using neutralizing reagents impaired medulloblastoma cell proliferation in vitro and induced a tumor volume reduction in vivo. An analysis of primary tumors revealed that LIFRα and p110α expression were elevated in the sonic hedgehog (SHH) subgroup of medulloblastoma, indicating its clinical relevance. Together, these data reveal a novel molecular signaling network, in which PI3K isoform p110α controls the expression of LIFRα via c-Myc and miR-125b to promote MB cell proliferation.
Collapse
Affiliation(s)
- Fabiana Salm
- Department of Clinical Research, Division of Pediatric Hematology/Oncology, University of Bern, Bern, Switzerland
| | - Valeriya Dimitrova
- Department of Clinical Research, Division of Pediatric Hematology/Oncology, University of Bern, Bern, Switzerland
| | - André O. von Bueren
- Department of Pediatrics and Department of Pediatric Hematology and Oncology, Georg August University Goettingen, Goettingen, Germany
| | - Paulina Ćwiek
- Department of Clinical Research, Division of Pediatric Hematology/Oncology, University of Bern, Bern, Switzerland
| | - Hubert Rehrauer
- Functional Genomics Center Zurich, ETH and University of Zurich, Zurich, Switzerland
| | | | - Pascale Anderle
- Institute of Biochemistry and Molecular Medicine, University of Bern, Swiss National Centre of Competence in Research TransCure, University of Bern, Bern, Switzerland
- Swiss National Centre of Competence in Research Molecular Oncology, Swiss Institute for Experimental Cancer Research, Ecole Polytechnique Fédérale de Lausanne, School of Life Sciences, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Alexandre Arcaro
- Department of Clinical Research, Division of Pediatric Hematology/Oncology, University of Bern, Bern, Switzerland
- * E-mail:
| |
Collapse
|
3
|
Ćwiek P, Leni Z, Salm F, Dimitrova V, Styp-Rekowska B, Chiriano G, Carroll M, Höland K, Djonov V, Scapozza L, Guiry P, Arcaro A. RNA interference screening identifies a novel role for PCTK1/CDK16 in medulloblastoma with c-Myc amplification. Oncotarget 2015; 6:116-29. [PMID: 25402633 PMCID: PMC4381582 DOI: 10.18632/oncotarget.2699] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 11/06/2014] [Indexed: 12/13/2022] Open
Abstract
Medulloblastoma (MB) is the most common malignant brain tumor in children and is associated with a poor outcome. cMYC amplification characterizes a subgroup of MB with very poor prognosis. However, there exist so far no targeted therapies for the subgroup of MB with cMYC amplification. Here we used kinome-wide RNA interference screening to identify novel kinases that may be targeted to inhibit the proliferation of c-Myc-overexpressing MB. The RNAi screen identified a set of 5 genes that could be targeted to selectively impair the proliferation of c-Myc-overexpressing MB cell lines: AKAP12 (A-kinase anchor protein), CSNK1α1 (casein kinase 1, alpha 1), EPHA7 (EPH receptor A7) and PCTK1 (PCTAIRE protein kinase 1). When using RNAi and a pharmacological inhibitor selective for PCTK1, we could show that this kinase plays a crucial role in the proliferation of MB cell lines and the activation of the mammalian target of rapamycin (mTOR) pathway. In addition, pharmacological PCTK1 inhibition reduced the expression levels of c-Myc. Finally, targeting PCTK1 selectively impaired the tumor growth of c-Myc-overexpressing MB cells in vivo. Together our data uncover a novel and crucial role for PCTK1 in the proliferation and survival of MB characterized by cMYC amplification.
Collapse
Affiliation(s)
- Paulina Ćwiek
- Division of Pediatric Hematology/Oncology, Bern University Hospital, Bern, Switzerland
| | - Zaira Leni
- Division of Pediatric Hematology/Oncology, Bern University Hospital, Bern, Switzerland
| | - Fabiana Salm
- Division of Pediatric Hematology/Oncology, Bern University Hospital, Bern, Switzerland
| | - Valeriya Dimitrova
- Division of Pediatric Hematology/Oncology, Bern University Hospital, Bern, Switzerland
| | | | - Gianpaolo Chiriano
- Pharmaceutical Biochemistry, School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, Geneva, Switzerland
| | - Michael Carroll
- Centre for Synthesis and Chemical Biology, University College Dublin, Belfield, Dublin, Ireland
| | - Katrin Höland
- Division of Pediatric Hematology/Oncology, Bern University Hospital, Bern, Switzerland
| | | | - Leonardo Scapozza
- Pharmaceutical Biochemistry, School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, Geneva, Switzerland
| | - Patrick Guiry
- Centre for Synthesis and Chemical Biology, University College Dublin, Belfield, Dublin, Ireland
| | - Alexandre Arcaro
- Division of Pediatric Hematology/Oncology, Bern University Hospital, Bern, Switzerland
| |
Collapse
|
4
|
Glaucarubinone inhibits colorectal cancer growth by suppression of hypoxia-inducible factor 1α and β-catenin via a p-21 activated kinase 1-dependent pathway. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1853:157-65. [PMID: 25409929 DOI: 10.1016/j.bbamcr.2014.10.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Revised: 09/17/2014] [Accepted: 10/15/2014] [Indexed: 01/22/2023]
Abstract
p-21-Activated kinase 1 (PAK1) enhances colorectal cancer (CRC) progression by stimulating Wnt/β-catenin, ERK and AKT pathways. PAK1 also promotes CRC survival via up-regulation of hypoxia-inducible factor 1α (HIF-1α), a key player in cancer survival. Glaucarubinone, a quassinoid natural product, inhibits pancreatic cancer growth by down-regulation of PAK1. The aim of this study was to investigate the effect of glaucarubinone on CRC growth and metastasis, and the mechanism involved. Cell proliferation was measured in vitro by [(3)H]-thymidine incorporation and in vivo by volume of tumor xenografts. Protein concentrations were measured by Western blotting of cell extracts. We report here that glaucarubinone inhibited CRC growth both in vitro and in vivo. The potency of glaucarubinone as an inhibitor of cell proliferation was negatively correlated to PAK1 expression in CRC cells. Glaucarubinone suppressed the expression of HIF-1α and β-catenin. Knockdown of PAK1 by shRNA enhanced inhibition by glaucarubinone while constitutively active PAK1 blocked the inhibitory effect. Our findings indicate that glaucarubinone inhibited CRC growth by down-regulation of HIF-1α and β-catenin via a PAK1-dependent pathway.
Collapse
|
5
|
Al-Salahi OSA, Ji D, Majid AMSA, Kit-Lam C, Abdullah WZ, Zaki A, Jamal Din SKK, Yusoff NM, Majid ASA. Anti-tumor activity of Eurycoma longifolia root extracts against K-562 cell line: in vitro and in vivo study. PLoS One 2014; 9:e83818. [PMID: 24409284 PMCID: PMC3883656 DOI: 10.1371/journal.pone.0083818] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Accepted: 11/08/2013] [Indexed: 01/28/2023] Open
Abstract
Eurycoma longifolia Jack has been widely used in traditional medicine for its antimalarial, aphrodisiac, anti-diabetic, antimicrobial and anti-pyretic activities. Its anticancer activity has also been recently reported on different solid tumors, however no anti-leukemic activity of this plant has been reported. Thus the present study assesses the in vitro and in vivo anti-proliferative and apoptotic potentials of E. longifolia on K-562 leukemic cell line. The K-562 cells (purchased from ATCC) were isolated from patients with chronic myelocytic leukemia (CML) were treated with the various fractions (TAF273, F3 and F4) of E. longifolia root methanolic extract at various concentrations and time intervals and the anti-proliferative activity assessed by MTS assay. Flow cytometry was used to assess the apoptosis and cell cycle arrest. Nude mice injected subcutaneously with 107 K-562 cells were used to study the anti-leukemic activity of TAF273 in vivo. TAF273, F3 and F4 showed various degrees of growth inhibition with IC50 values of 19, 55 and 62 µg/ml, respectively. TAF273 induced apoptosis in a dose and time dependent manner. TAF273 arrested cell cycle at G1and S phases. Intraperitoneal administration of TAF273 (50 mg/kg) resulted in a significant growth inhibition of subcutaneous tumor in TAF273-treated mice compared with the control mice (P = 0.024). TAF273 shows potent anti-proliferative activity in vitro and in vivo models of CML and therefore, justifies further efforts to define more clearly the potential benefits of using TAF273 as a novel therapeutic strategy for CML management.
Collapse
Affiliation(s)
- Omar Saeed Ali Al-Salahi
- Advanced Medical and Dental Institute (AMDI), Universiti Sains Malaysia (USM), Kepala Batas, Pulau Pinang, Malaysia
| | - Dan Ji
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Southwest Eye Hospital, Southwest Hospital, The Third Military Medical University, Chongqing, P.R. China
| | | | - Chan Kit-Lam
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Penang, Malaysia
| | - Wan Zaidah Abdullah
- Haematology Department, School of Medical Sciences, USM, Kubang Kerian, Kelantan, Malaysia
| | - Abdelhamid Zaki
- Advanced Medical and Dental Institute (AMDI), Universiti Sains Malaysia (USM), Kepala Batas, Pulau Pinang, Malaysia
- Therapeutic Chemistry Department, National Research Centre, Cairo University, Dokki, Cairo, Egypt
| | | | - Narazah Mohd Yusoff
- Advanced Medical and Dental Institute (AMDI), Universiti Sains Malaysia (USM), Kepala Batas, Pulau Pinang, Malaysia
- * E-mail: (ASAM); (NMY)
| | - Aman Shah Abdul Majid
- Advanced Medical and Dental Institute (AMDI), Universiti Sains Malaysia (USM), Kepala Batas, Pulau Pinang, Malaysia
- * E-mail: (ASAM); (NMY)
| |
Collapse
|
6
|
Fiaschetti G, Abela L, Nonoguchi N, Dubuc AM, Remke M, Boro A, Grunder E, Siler U, Ohgaki H, Taylor MD, Baumgartner M, Shalaby T, Grotzer MA. Epigenetic silencing of miRNA-9 is associated with HES1 oncogenic activity and poor prognosis of medulloblastoma. Br J Cancer 2013; 110:636-47. [PMID: 24346283 PMCID: PMC3915127 DOI: 10.1038/bjc.2013.764] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 10/31/2013] [Accepted: 11/13/2013] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND microRNA-9 is a key regulator of neuronal development aberrantly expressed in brain malignancies, including medulloblastoma. The mechanisms by which microRNA-9 contributes to medulloblastoma pathogenesis remain unclear, and factors that regulate this process have not been delineated. METHODS Expression and methylation status of microRNA-9 in medulloblastoma cell lines and primary samples were analysed. The association of microRNA-9 expression with medulloblastoma patients' clinical outcome was assessed, and the impact of microRNA-9 restoration was functionally validated in medulloblastoma cells. RESULTS microRNA-9 expression is repressed in a large subset of MB samples compared with normal fetal cerebellum. Low microRNA-9 expression correlates significantly with the diagnosis of unfavourable histopathological variants and with poor clinical outcome. microRNA-9 silencing occurs via cancer-specific CpG island hypermethylation. HES1 was identified as a direct target of microRNA-9 in medulloblastoma, and restoration of microRNA-9 was shown to trigger cell cycle arrest, to inhibit clonal growth and to promote medulloblastoma cell differentiation. CONCLUSIONS microRNA-9 is a methylation-silenced tumour suppressor that could be a potential candidate predictive marker for poor prognosis of medulloblastoma. Loss of microRNA-9 may confer a proliferative advantage to tumour cells, and it could possibly contribute to disease pathogenesis. Thus, re-expression of microRNA-9 may constitute a novel epigenetic regulation strategy against medulloblastoma.
Collapse
Affiliation(s)
- G Fiaschetti
- Neuro-Oncology group, Experimental Infectious Diseases and Cancer Research, August-Forel Strasse 1, Zurich CH-8008, Switzerland
| | - L Abela
- Division of Oncology, University Children's Hospital of Zürich, Steinwiesstrasse 75, Zurich CH-8032, Switzerland
| | - N Nonoguchi
- International Agency for Research on Cancer, World Health Organization, Section of Molecular Pathology, 150 Cours Albert Thomas, 69372, Lyon Cedex 08, France
| | - A M Dubuc
- Arthur and Sonia Labatt Brain Tumour Research Centre, MaRS Centre - 11-401M, 101 College Street, Toronto, ON M5G1L7, Canada
| | - M Remke
- Brain Tumor Research Centre, 101 College Street, TMDT-11-401M, Toronto, ON M5G1L7, Canada
| | - A Boro
- Oncology group, Experimental Infectious Diseases and Cancer Research, August-Forel Strasse 1, Zurich CH-8008, Switzerland
| | - E Grunder
- Division of Oncology, University Children's Hospital of Zürich, Steinwiesstrasse 75, Zurich CH-8032, Switzerland
| | - U Siler
- Division of Immunology, University Children's Hospital of Zürich, Steinwiesstrasse 75, Zurich CH-8032, Switzerland
| | - H Ohgaki
- International Agency for Research on Cancer, World Health Organization, Section of Molecular Pathology, 150 Cours Albert Thomas, 69372, Lyon Cedex 08, France
| | - M D Taylor
- The Hospital for Sick Children, Division of Neurosurgery, Suite 1504, 555 University Avenue, Toronto, ON M5G1X8, Canada
| | - M Baumgartner
- Neuro-Oncology group, Experimental Infectious Diseases and Cancer Research, August-Forel Strasse 1, Zurich CH-8008, Switzerland
| | - T Shalaby
- Neuro-Oncology group, Experimental Infectious Diseases and Cancer Research, August-Forel Strasse 1, Zurich CH-8008, Switzerland
| | - M A Grotzer
- Division of Oncology, University Children's Hospital of Zürich, Steinwiesstrasse 75, Zurich CH-8032, Switzerland
| |
Collapse
|
7
|
Shalaby T, Fiaschetti G, Nagasawa K, Shin-ya K, Baumgartner M, Grotzer M. G-quadruplexes as potential therapeutic targets for embryonal tumors. Molecules 2013; 18:12500-37. [PMID: 24152672 PMCID: PMC6269990 DOI: 10.3390/molecules181012500] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Revised: 09/18/2013] [Accepted: 09/25/2013] [Indexed: 12/27/2022] Open
Abstract
Embryonal tumors include a heterogeneous group of highly malignant neoplasms that primarily affect infants and children and are characterized by a high rate of mortality and treatment-related morbidity, hence improved therapies are clearly needed. G-quadruplexes are special secondary structures adopted in guanine (G)-rich DNA sequences that are often present in biologically important regions, e.g. at the end of telomeres and in the regulatory regions of oncogenes such as MYC. Owing to the significant roles that both telomeres and MYC play in cancer cell biology, G-quadruplexes have been viewed as emerging therapeutic targets in oncology and as tools for novel anticancer drug design. Several compounds that target these structures have shown promising anticancer activity in tumor xenograft models and some of them have entered Phase II clinical trials. In this review we examine approaches to DNA targeted cancer therapy, summarize the recent developments of G-quadruplex ligands as anticancer drugs and speculate on the future direction of such structures as a potential novel therapeutic strategy for embryonal tumors of the nervous system.
Collapse
Affiliation(s)
- Tarek Shalaby
- Division of Oncology, University Children's Hospital of Zurich, Zurich 8032, Switzerland.
| | | | | | | | | | | |
Collapse
|
8
|
Al-Salahi OSA, Kit-Lam C, Majid AMSA, Al-Suede FSR, Mohammed Saghir SA, Abdullah WZ, Ahamed MBK, Yusoff NM. Anti-angiogenic quassinoid-rich fraction from Eurycoma longifolia modulates endothelial cell function. Microvasc Res 2013; 90:30-9. [PMID: 23899415 DOI: 10.1016/j.mvr.2013.07.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2013] [Revised: 07/22/2013] [Accepted: 07/22/2013] [Indexed: 01/12/2023]
Abstract
Targeting angiogenesis could be an excellent strategy to combat angiogenesis-dependent pathophysiological conditions such as cancer, rheumatoid arthritis, obesity, systemic lupus erythematosus, psoriasis, proliferative retinopathy and atherosclerosis. Recently a number of clinical investigations are being undertaken to assess the potential therapeutic application of various anti-angiogenic agents. Many of these angiogenesis inhibitors are directed against the functions of endothelial cells, which are considered as the building blocks of blood vessels. Similarly, roots of a traditional medicinal plant, Eurycoma longifolia, can be used as an alternative treatment to prevent and treat the angiogenesis-related diseases. In the present study, antiangiogenic potential of partially purified quassinoid-rich fraction (TAF273) of E. longifolia root extract was evaluated using ex vivo and in vivo angiogenesis models and the anti-angiogenic efficacy of TAF273 was investigated in human umbilical vein endothelial cells (HUVEC). TAF273 caused significant suppression in sprouting of microvessels in rat aorta with IC50 11.5μg/ml. TAF273 (50μg/ml) showed remarkable inhibition (63.13%) of neovascularization in chorioallantoic membrane of chick embryo. Tumor histology also revealed marked reduction in extent of vascularization. In vitro, TAF273 significantly inhibited the major angiogenesis steps such as proliferation, migration and differentiation of HUVECs. Phytochemical analysis revealed high content of quassinoids in TAF273. Specially, HPLC characterization showed that TAF273 is enriched with eurycomanone, 13α(21)-epoxyeurycomanone and eurycomanol. These results demonstrated that the antiangiogenic activity of TAF273 may be due to its inhibitory effect on endothelial cell proliferation, differentiation and migration which could be attributed to the high content of quassinoids in E. longifolia.
Collapse
Affiliation(s)
- Omar Saeed Ali Al-Salahi
- Advanced Medical and Dental Institute (AMDI), Universiti Sains Malaysia (USM), 13200 Kepala Batas, Pulau Pinang, Malaysia
| | | | | | | | | | | | | | | |
Collapse
|
9
|
Wojtalla A, Salm F, Christiansen DG, Cremona T, Cwiek P, Shalaby T, Gross N, Grotzer MA, Arcaro A. Novel agents targeting the IGF-1R/PI3K pathway impair cell proliferation and survival in subsets of medulloblastoma and neuroblastoma. PLoS One 2012; 7:e47109. [PMID: 23056595 PMCID: PMC3466180 DOI: 10.1371/journal.pone.0047109] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Accepted: 09/10/2012] [Indexed: 12/31/2022] Open
Abstract
The receptor tyrosine kinase (RTK)/phosphoinositide 3-kinase (PI3K) pathway is fundamental for cancer cell proliferation and is known to be frequently altered and activated in neoplasia, including embryonal tumors. Based on the high frequency of alterations, targeting components of the PI3K signaling pathway is considered to be a promising therapeutic approach for cancer treatment. Here, we have investigated the potential of targeting the axis of the insulin-like growth factor-1 receptor (IGF-1R) and PI3K signaling in two common cancers of childhood: neuroblastoma, the most common extracranial tumor in children and medulloblastoma, the most frequent malignant childhood brain tumor. By treating neuroblastoma and medulloblastoma cells with R1507, a specific humanized monoclonal antibody against the IGF-1R, we could observe cell line-specific responses and in some cases a strong decrease in cell proliferation. In contrast, targeting the PI3K p110α with the specific inhibitor PIK75 resulted in broad anti-proliferative effects in a panel of neuro- and medulloblastoma cell lines. Additionally, sensitization to commonly used chemotherapeutic agents occurred in neuroblastoma cells upon treatment with R1507 or PIK75. Furthermore, by studying the expression and phosphorylation state of IGF-1R/PI3K downstream signaling targets we found down-regulated signaling pathway activation. In addition, apoptosis occurred in embryonal tumor cells after treatment with PIK75 or R1507. Together, our studies demonstrate the potential of targeting the IGF-1R/PI3K signaling axis in embryonal tumors. Hopefully, this knowledge will contribute to the development of urgently required new targeted therapies for embryonal tumors.
Collapse
Affiliation(s)
- Anna Wojtalla
- Division of Pediatric Hematology/Oncology, Department of Clinical Research, University of Bern, Bern, Switzerland
| | - Fabiana Salm
- Division of Pediatric Hematology/Oncology, Department of Clinical Research, University of Bern, Bern, Switzerland
| | - Ditte G. Christiansen
- Division of Pediatric Hematology/Oncology, Department of Clinical Research, University of Bern, Bern, Switzerland
| | - Tiziana Cremona
- Department of Oncology, University Children's Hospital Zurich, Zurich, Switzerland
| | - Paulina Cwiek
- Division of Pediatric Hematology/Oncology, Department of Clinical Research, University of Bern, Bern, Switzerland
| | - Tarek Shalaby
- Department of Oncology, University Children's Hospital Zurich, Zurich, Switzerland
| | - Nicole Gross
- Department of Pediatrics, Pediatric Oncology Research, University Hospital CHUV, Lausanne, Switzerland
| | - Michael A. Grotzer
- Department of Oncology, University Children's Hospital Zurich, Zurich, Switzerland
| | - Alexandre Arcaro
- Division of Pediatric Hematology/Oncology, Department of Clinical Research, University of Bern, Bern, Switzerland
- * E-mail:
| |
Collapse
|
10
|
Cavalcanti BC, da Costa PM, Carvalho AA, Rodrigues FAR, Amorim RCN, Silva ECC, Pohlit AM, Costa-Lotufo LV, Moraes MO, Pessoa C. Involvement of intrinsic mitochondrial pathway in neosergeolide-induced apoptosis of human HL-60 leukemia cells: the role of mitochondrial permeability transition pore and DNA damage. PHARMACEUTICAL BIOLOGY 2012; 50:980-993. [PMID: 22775415 DOI: 10.3109/13880209.2012.654921] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
CONTEXT Quassinoids are biologically active secondary metabolites found exclusively in the Simaroubaceae family of plants. These compounds generally present important biological properties, including cytotoxic and antitumor properties. OBJECTIVE In the present study, the cytotoxic effects of neosergeolide, a quassinoid isolated from Picrolemma sprucei Hook. f., were evaluated in human promyelocytic leukemia cells (HL-60). MATERIALS AND METHODS Cytotoxicity and antiproliferative effects were evaluated by the MTT assay, May-Grünwald-Giemsa's staining, BrdU incorporation test, and flow cytometry procedures. The comet assay and micronuclei analysis were applied to determine the genotoxic and mutagenic potential of neosergeolide. RESULTS After 24 h exposure, neosergeolide strongly inhibited cancer cell proliferation (IC₅₀ 0.1 µM), and its activity seemed to be selective to tumor cells because it had no antiproliferative effect on human peripheral blood mononuclear cells (PBMC) at tested concentrations. Apoptosis was induced at submicromolar concentrations (0.05, 0.1, and 0.2 µM) as evidenced by morphological changes, mitochondrial depolarization, phosphatidylserine externalization, caspases activation, and internucleosomal DNA fragmentation. Additionally, neosergeolide effects were prevented by cyclosporine A (CsA), an inhibitor of the mitochondrial permeability transition (MPT) pore, which reinforced the participation of intrinsic pathways in the apoptotic process induced by this natural quassinoid. Direct DNA damage was further confirmed by comet assay and cytokinesis-block micronucleus test. DISCUSSION AND CONCLUSION The present study provided experimental evidence to support the underlying mechanism of action involved in the neosergeolide-mediated apoptosis. In addition, no antiproliferative effect or DNA damage effect of neosergeolide was evident in PBMC, highlighting its therapeutic potential.
Collapse
MESH Headings
- Antineoplastic Agents, Phytogenic/adverse effects
- Antineoplastic Agents, Phytogenic/antagonists & inhibitors
- Antineoplastic Agents, Phytogenic/pharmacology
- Apoptosis/drug effects
- Cell Proliferation/drug effects
- Cells, Cultured
- Comet Assay
- Cyclosporine/pharmacology
- Cytokinesis/drug effects
- DNA Fragmentation/drug effects
- HL-60 Cells
- Humans
- Inhibitory Concentration 50
- Leukemia, Promyelocytic, Acute/drug therapy
- Leukemia, Promyelocytic, Acute/metabolism
- Leukemia, Promyelocytic, Acute/pathology
- Leukocytes, Mononuclear/drug effects
- Membrane Potential, Mitochondrial/drug effects
- Micronucleus Tests
- Mitochondria/drug effects
- Mitochondria/metabolism
- Mitochondrial Membrane Transport Proteins/antagonists & inhibitors
- Mitochondrial Membrane Transport Proteins/metabolism
- Mitochondrial Permeability Transition Pore
- Neoplasm Proteins/antagonists & inhibitors
- Neoplasm Proteins/metabolism
- Quassins/adverse effects
- Quassins/antagonists & inhibitors
- Quassins/pharmacology
- Simaroubaceae/chemistry
Collapse
Affiliation(s)
- Bruno C Cavalcanti
- National Laboratory of Experimental Oncology, Federal University of Ceará, CEP 60430-270, Fortaleza, CE, Brazil.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Astelbauer F, Gruber M, Brem B, Greger H, Obwaller A, Wernsdorfer G, Congpuong K, Wernsdorfer WH, Walochnik J. Activity of selected phytochemicals against Plasmodium falciparum. Acta Trop 2012; 123:96-100. [PMID: 22537982 DOI: 10.1016/j.actatropica.2012.04.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2010] [Revised: 03/31/2012] [Accepted: 04/03/2012] [Indexed: 01/03/2023]
Abstract
According to the WHO, in 2008, there were 247 million reported cases of malaria and nearly one million deaths from the disease. Parasite resistance against first-line drugs, including artemisinin and mefloquine, is increasing. In this study the plant-derived compounds aglafolin, rocaglamid, kokusaginine, arborine, arborinine and tuberostemonine were investigated for their anti-plasmodial activity in vitro. Fresh Plasmodium falciparum isolates were taken from patients in the area of Mae Sot, north-western Thailand in 2008 and the inhibition of schizont maturation was determined for the respective compounds. With inhibitory concentrations effecting 50%, 90% and 99% inhibition (IC(50), IC(90) and IC(99)) of 60.95 nM, 854.41 nM and 7351.49 nM, respectively, rocaglamid was the most active of the substances, closely followed by aglafoline with 53.49 nM, 864.55 nM and 8354.20 nM. The activity was significantly below that of artemisinin, but moderately higher than that of quinine. Arborine, arborinine, tuberostemonine and kokusaginine showed only marginal activity against P. falciparum characterized by IC(50) and IC(99) values higher than 350 nM and 180 μM, respectively, and regressions with relatively shallow slopes S>14.38. Analogues of rocaglamid and aglafoline merit further exploration of their anti-plasmodial activity.
Collapse
Affiliation(s)
- Florian Astelbauer
- Institute of Specific Prophylaxis and Tropical Medicine, Medical University of Vienna, Austria
| | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Schulte JH, Lindner S, Bohrer A, Maurer J, De Preter K, Lefever S, Heukamp L, Schulte S, Molenaar J, Versteeg R, Thor T, Künkele A, Vandesompele J, Speleman F, Schorle H, Eggert A, Schramm A. MYCN and ALKF1174L are sufficient to drive neuroblastoma development from neural crest progenitor cells. Oncogene 2012; 32:1059-65. [PMID: 22484425 DOI: 10.1038/onc.2012.106] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Neuroblastoma is an embryonal tumor with a heterogeneous clinical course. The tumor is presumed to be derived from the neural crest, but the cells of origin remain to be determined. To date, few recurrent genetic changes contributing to neuroblastoma formation, such as amplification of the MYCN oncogene and activating mutations of the ALK oncogene, have been identified. The possibility to model neuroblastoma in mice allows investigation of the cell of origin hypothesis in further detail. Here we present the evidence that murine neural crest progenitor cells can give rise to neuroblastoma upon transformation with MYCN or ALK(F1174L). For this purpose we used JoMa1, a multipotent neural crest progenitor cell line, which is kept in a viable and undifferentiated state by a tamoxifen-activated c-Myc transgene (c-MycER(T)). Expression of MYCN or ALK(F1174L), one of the oncogenic ALK variants identified in primary neuroblastomas, enabled these cells to grow independently of c-MycER(T) activity in vitro and caused formation of neuroblastoma-like tumors in vivo in contrast to parental JoMa1 cells and JoMa1 cells-expressing TrkA or GFP. Tumorigenicity was enhanced upon serial transplantation of tumor-derived cells, and tumor cells remained susceptible to the MYC-inhibitor, NBT-272, indicating that cell growth depended on functional MYCN. Our findings support neural crest progenitor cells as the precursor cells of neuroblastoma, and indicate that neuroblastomas arise as their malignant progeny.
Collapse
Affiliation(s)
- J H Schulte
- Department of Pediatric Oncology and Haematology, University Children's Hospital Essen, Essen, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Buss MC, Read TA, Schniederjan MJ, Gandhi K, Castellino RC. HDM2 promotes WIP1-mediated medulloblastoma growth. Neuro Oncol 2012; 14:440-58. [PMID: 22379189 DOI: 10.1093/neuonc/nos001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Medulloblastoma is the most common malignant childhood brain tumor. The protein phosphatase and oncogene WIP1 is over-expressed or amplified in a significant number of primary human medulloblastomas and cell lines. In the present study, we examine an important mechanism by which WIP1 promotes medulloblastoma growth using in vitro and in vivo models. Human cell lines and intracerebellar xenografted animal models were used to study the role of WIP1 and the major TP53 regulator, HDM2, in medulloblastoma growth. Stable expression of WIP1 enhances growth of TP53 wild-type medulloblastoma cells, compared with cells with stable expression of an empty-vector or mutant WIP1. In an animal model, WIP1 enhances proliferation and reduces the survival of immunodeficient mice bearing intracerebellar xenografted human medulloblastoma cells. Cells with increased WIP1 expression also exhibit increased expression of HDM2. HDM2 knockdown or treatment with the HDM2 inhibitor Nutlin-3a, the active enantomer of Nutlin-3, specifically inhibits the growth of medulloblastoma cells with increased WIP1 expression. Nutlin-3a does not affect growth of medulloblastoma cells with stable expression of an empty vector or of mutant WIP1. Knockdown of WIP1 or treatment with the WIP1 inhibitor CCT007093 results in increased phosphorylation of known WIP1 targets, reduced HDM2 expression, and reduced growth specifically in WIP1 wild-type and high-expressing medulloblastoma cells. Combined WIP1 and HDM2 inhibition is more effective than WIP1 inhibition alone in blocking growth of WIP1 high-expressing medulloblastoma cells. Our preclinical study supports a role for therapies that target WIP1 and HDM2 in the treatment of medulloblastoma.
Collapse
Affiliation(s)
- Meghan C Buss
- Department of Pediatrics, Aflac Cancer Center and Blood Disorders Service, Atlanta, Georgia, USA
| | | | | | | | | |
Collapse
|
14
|
Grunder E, D'Ambrosio R, Fiaschetti G, Abela L, Arcaro A, Zuzak T, Ohgaki H, Lv SQ, Shalaby T, Grotzer M. MicroRNA-21 suppression impedes medulloblastoma cell migration. Eur J Cancer 2011; 47:2479-90. [PMID: 21775132 DOI: 10.1016/j.ejca.2011.06.041] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2011] [Revised: 06/07/2011] [Accepted: 06/17/2011] [Indexed: 12/19/2022]
Abstract
Medulloblastoma (MB), the most common malignant brain tumour in children, is characterised by a high risk of leptomeningeal dissemination. But little is known about the molecular mechanisms that promote cancer cell migration in MB. Aberrant expression of miR-21 is recognised to be causatively linked to metastasis in a variety of human neoplasms including brain tumours; however its function in MB is still unknown. In this study we investigated the expression level and the role of miR-21 in MB cell migration. miR-21 was found to be up-regulated, compared to normal cerebellum, in 29/29 MB primary samples and 6/6 MB-derived cell lines. Inverse correlation was observed between miR-21 expression and the metastasis suppressor PDCD4, while miR-21 repression increased the release of PDCD4 protein, suggesting negative regulation of PDCD4 by miR-21 in MB cells. Anti-miR-21 decreased protein expression of the tumour cell invasion mediators MAP4K1 and JNK, which are also known to be negatively regulated by PDCD4, and down-regulated integrin protein that is essential for MB leptomeningeal dissemination. Moreover miR-21 knockdown in MB cells increased the expression of two eminent negative modulators of cancer cell migration, E-Cadherin and TIMP2 proteins that are known to be positively regulated by PDCD4. Finally and importantly, suppression of miR-21 decreased the motility of MB cells and reduced their migration across basement membranes in vitro. Together, these compelling data propose miR-21 pathway as a novel mechanism impacting MB cell dissemination and raises the possibility that curability of selected MB may be improved by pharmaceutical strategies directed towards microRNA-21.
Collapse
Affiliation(s)
- Eveline Grunder
- Oncology Department, University Children's Hospital of Zurich, Switzerland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Oehler C, von Bueren AO, Furmanova P, Broggini-Tenzer A, Orlowski K, Rutkowski S, Frei K, Grotzer MA, Pruschy M. The microtubule stabilizer patupilone (epothilone B) is a potent radiosensitizer in medulloblastoma cells. Neuro Oncol 2011; 13:1000-10. [PMID: 21743064 DOI: 10.1093/neuonc/nor069] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Concurrent radiochemotherapy for medulloblastoma includes the microtubule disrupting agent vincristine; however, vincristine alone or as part of a combined treatment regimen is highly toxic. A major goal is therefore to replace vincristine with novel potent chemotherapeutic agents-in particular, with microtubule stabilizing and destabilizing compounds-with a larger therapeutic window. Here, we investigated the antiproliferative, cytotoxic and radiosensitizing effect of patupilone (epothilone B [EPO906]), a novel, non-taxane-related and nonneurotoxic microtubule-stabilizing agent in human medulloblastoma cell lines. The antiproliferative and cytotoxic effects of patupilone alone and in combination with ionizing radiation was determined in the 3 representative human medulloblastoma cell lines D341Med, D425Med, and DAOY. Patupilone alone effectively reduced the proliferative activity and clonogenicity of all medulloblastoma cell lines tested at picomolar concentrations (50-200 pM) and resulted in an at least additive anticlonogenic effect in combination with clinically relevant doses of ionizing radiation (2 or 5 Gy). Cell-cycle analysis revealed a sequential G2-M arrest and sub-G1 accumulation in a dose- and treatment-dependent manner after exposure to patupilone. In tumor xenografts derived from D425Med cells, a minimal treatment regimen with patupilone and fractionated irradiation (1 × 2 mg/kg plus 3 × 3 Gy) resulted in an extended tumor growth delay for the 2 single treatment modalities alone and a supra-additive treatment response for the combined treatment modality, with complete tumor regressions. These results demonstrate the potent efficacy of patupilone against medulloblastoma cell lines and indicate that patupilone represents a promising candidate to replace vincristine as part of a combined treatment strategy with ionizing radiation.
Collapse
Affiliation(s)
- Christoph Oehler
- Department of Radiation Oncology, University Hospital Zurich, CH-8091 Zürich, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Small Interfering RNA to c-myc Inhibits Vein Graft Restenosis in a Rat Vein Graft Model. J Surg Res 2011; 169:e85-91. [PMID: 21571310 DOI: 10.1016/j.jss.2011.03.060] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2010] [Revised: 02/02/2011] [Accepted: 03/22/2011] [Indexed: 01/23/2023]
|
17
|
Guerreiro AS, Fattet S, Kulesza DW, Atamer A, Elsing AN, Shalaby T, Jackson SP, Schoenwaelder SM, Grotzer MA, Delattre O, Arcaro A. A sensitized RNA interference screen identifies a novel role for the PI3K p110γ isoform in medulloblastoma cell proliferation and chemoresistance. Mol Cancer Res 2011; 9:925-35. [PMID: 21652733 DOI: 10.1158/1541-7786.mcr-10-0200] [Citation(s) in RCA: 40] [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
Medulloblastoma is the most common malignant brain tumor in children and is associated with a poor outcome. We were interested in gaining further insight into the potential of targeting the human kinome as a novel approach to sensitize medulloblastoma to chemotherapeutic agents. A library of small interfering RNA (siRNA) was used to downregulate the known human protein and lipid kinases in medulloblastoma cell lines. The analysis of cell proliferation, in the presence or absence of a low dose of cisplatin after siRNA transfection, identified new protein and lipid kinases involved in medulloblastoma chemoresistance. PLK1 (polo-like kinase 1) was identified as a kinase involved in proliferation in medulloblastoma cell lines. Moreover, a set of 6 genes comprising ATR, LYK5, MPP2, PIK3CG, PIK4CA, and WNK4 were identified as contributing to both cell proliferation and resistance to cisplatin treatment in medulloblastoma cells. An analysis of the expression of the 6 target genes in primary medulloblastoma tumor samples and cell lines revealed overexpression of LYK5 and PIK3CG. The results of the siRNA screen were validated by target inhibition with specific pharmacological inhibitors. A pharmacological inhibitor of p110γ (encoded by PIK3CG) impaired cell proliferation in medulloblastoma cell lines and sensitized the cells to cisplatin treatment. Together, our data show that the p110γ phosphoinositide 3-kinase isoform is a novel target for combinatorial therapies in medulloblastoma.
Collapse
Affiliation(s)
- Ana S Guerreiro
- Department of Oncology, University Children's Hospital, Zurich, Switzerland
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Azizi AA, Li L, Ströbel T, Chen WQ, Slavc I, Lubec G. Identification of c-myc-dependent proteins in the medulloblastoma cell line D425Med. Amino Acids 2011; 42:2149-63. [DOI: 10.1007/s00726-011-0953-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2011] [Accepted: 05/26/2011] [Indexed: 12/27/2022]
|
19
|
von Bueren AO, Oehler C, Shalaby T, von Hoff K, Pruschy M, Seifert B, Gerber NU, Warmuth-Metz M, Stearns D, Eberhart CG, Kortmann RD, Rutkowski S, Grotzer MA. c-MYC expression sensitizes medulloblastoma cells to radio- and chemotherapy and has no impact on response in medulloblastoma patients. BMC Cancer 2011; 11:74. [PMID: 21324178 PMCID: PMC3050852 DOI: 10.1186/1471-2407-11-74] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2010] [Accepted: 02/16/2011] [Indexed: 12/27/2022] Open
Abstract
Background To study whether and how c-MYC expression determines response to radio- and chemotherapy in childhood medulloblastoma (MB). Methods We used DAOY and UW228 human MB cells engineered to stably express different levels of c-MYC, and tested whether c-MYC expression has an effect on radio- and chemosensitivity using the colorimetric 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium inner salt (MTS) assay, clonogenic survival, apoptosis assays, cell cycle analysis, and western blot assessment. In an effort to validate our results, we analyzed c-MYC mRNA expression in formalin-fixed paraffin-embedded tumor samples from well-documented patients with postoperative residual tumor and compared c-MYC mRNA expression with response to radio- and chemotherapy as examined by neuroradiological imaging. Results In DAOY - and to a lesser extent in UW228 - cells expressing high levels of c-MYC, the cytotoxicity of cisplatin, and etoposide was significantly higher when compared with DAOY/UW228 cells expressing low levels of c-MYC. Irradiation- and chemotherapy-induced apoptotic cell death was enhanced in DAOY cells expressing high levels of c-MYC. The response of 62 of 66 residual tumors was evaluable and response to postoperative radio- (14 responders (CR, PR) vs. 5 non-responders (SD, PD)) or chemotherapy (23 CR/PR vs. 20 SD/PD) was assessed. c-MYC mRNA expression was similar in primary MB samples of responders and non-responders (Mann-Whitney U test, p = 0.50, ratio 0.49, 95% CI 0.008-30.0 and p = 0.67, ratio 1.8, 95% CI 0.14-23.5, respectively). Conclusions c-MYC sensitizes MB cells to some anti-cancer treatments in vitro. As we failed to show evidence for such an effect on postoperative residual tumors when analyzed by imaging, additional investigations in xenografts and larger MB cohorts may help to define the exact function of c-MYC in modulating response to treatment.
Collapse
Affiliation(s)
- André O von Bueren
- Neuro-Oncology Program, University Children's Hospital, Zurich, Switzerland.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Bone morphogenetic protein-7 is a MYC target with prosurvival functions in childhood medulloblastoma. Oncogene 2011; 30:2823-35. [PMID: 21317922 DOI: 10.1038/onc.2011.10] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Medulloblastoma (MB) is the most common malignant brain tumor in children. It is known that overexpression and/or amplification of the MYC oncogene is associated with poor clinical outcome, but the molecular mechanisms and the MYC downstream effectors in MB remain still elusive. Besides contributing to elucidate how progression of MB takes place, most importantly, the identification of novel MYC-target genes will suggest novel candidates for targeted therapy in MB. A group of 209 MYC-responsive genes was obtained from a complementary DNA microarray analysis of a MB-derived cell line, following MYC overexpression and silencing. Among the MYC-responsive genes, we identified the members of the bone morphogenetic protein (BMP) signaling pathway, which have a crucial role during the development of the cerebellum. In particular, the gene BMP7 was identified as a direct target of MYC. A positive correlation between MYC and BMP7 expression was documented by analyzing two distinct sets of primary MB samples. Functional studies in vitro using a small-molecule inhibitor of the BMP/SMAD signaling pathway reproduced the effect of the small interfering RNA-mediated silencing of BMP7. Both approaches led to a block of proliferation in a panel of MB cells and to inhibition of SMAD phosphorylation. Altogether, our findings indicate that high MYC levels drive BMP7 overexpression, promoting cell survival in MB cells. This observation suggests the potential relevance of targeting the BMP/SMAD pathway as a novel therapeutic approach for the treatment of childhood MB.
Collapse
|
21
|
The Quassinoid Derivative NBT-272 Targets Both the AKT and ERK Signaling Pathways in Embryonal Tumors. Mol Cancer Ther 2010; 9:3145-57. [DOI: 10.1158/1535-7163.mct-10-0539] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
22
|
Faoro D, von Bueren AO, Shalaby T, Sciuscio D, Hürlimann ML, Arnold L, Gerber NU, Haybaeck J, Mittelbronn M, Rutkowski S, Hegi M, Grotzer MA. Expression of O6-methylguanine-DNA methyltransferase in childhood medulloblastoma. J Neurooncol 2010; 103:59-69. [DOI: 10.1007/s11060-010-0366-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2010] [Accepted: 08/17/2010] [Indexed: 11/28/2022]
|
23
|
Shalaby T, von Bueren AO, Hürlimann ML, Fiaschetti G, Castelletti D, Masayuki T, Nagasawa K, Arcaro A, Jelesarov I, Shin-ya K, Grotzer M. Disabling c-Myc in childhood medulloblastoma and atypical teratoid/rhabdoid tumor cells by the potent G-quadruplex interactive agent S2T1-6OTD. Mol Cancer Ther 2010; 9:167-79. [PMID: 20053783 DOI: 10.1158/1535-7163.mct-09-0586] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We investigated here the effects of S2T1-6OTD, a novel telomestatin derivative that is synthesized to target G-quadruplex-forming DNA sequences, on a representative panel of human medulloblastoma (MB) and atypical teratoid/rhabdoid (AT/RT) childhood brain cancer cell lines. S2T1-6OTD proved to be a potent c-Myc inhibitor through its high-affinity physical interaction with the G-quadruplex structure in the c-Myc promoter. Treatment with S2T1-6OTD reduced the mRNA and protein expressions of c-Myc and hTERT, which is transcriptionally regulated by c-Myc, and decreased the activities of both genes. In remarkable contrast to control cells, short-term (72-hour) treatment with S2T1-6OTD resulted in a dose- and time-dependent antiproliferative effect in all MB and AT/RT brain tumor cell lines tested (IC(50), 0.25-0.39 micromol/L). Under conditions where inhibition of both proliferation and c-Myc activity was observed, S2T1-6OTD treatment decreased the protein expression of the cell cycle activator cyclin-dependent kinase 2 and induced cell cycle arrest. Long-term treatment (5 weeks) with nontoxic concentrations of S2T1-6OTD resulted in a time-dependent (mainly c-Myc-dependent) telomere shortening. This was accompanied by cell growth arrest starting on day 28 followed by cell senescence and induction of apoptosis on day 35 in all of the five cell lines investigated. On in vivo animal testing, S2T1-6OTD may well represent a novel therapeutic strategy for childhood brain tumors.
Collapse
Affiliation(s)
- Tarek Shalaby
- Department of Oncology, University Children's Hospital Zurich, Zurich, Switzerland
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
RNA interference-mediated c-MYC inhibition prevents cell growth and decreases sensitivity to radio- and chemotherapy in childhood medulloblastoma cells. BMC Cancer 2009; 9:10. [PMID: 19134217 PMCID: PMC2648994 DOI: 10.1186/1471-2407-9-10] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2008] [Accepted: 01/10/2009] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND With current treatment strategies, nearly half of all medulloblastoma (MB) patients die from progressive tumors. Accordingly, the identification of novel therapeutic strategies remains a major goal. Deregulation of c-MYC is evident in numerous human cancers. In MB, over-expression of c-MYC has been shown to cause anaplasia and correlate with unfavorable prognosis. METHODS To study the role of c-MYC in MB biology, we down-regulated c-MYC expression by using small interfering RNA (siRNA) and investigated changes in cellular proliferation, cell cycle analysis, apoptosis, telomere maintenance, and response to ionizing radiation (IR) and chemotherapeutics in a representative panel of human MB cell lines expressing different levels of c-MYC (DAOY wild-type, DAOY transfected with the empty vector, DAOY transfected with c-MYC, D341, and D425). RESULTS siRNA-mediated c-MYC down-regulation resulted in an inhibition of cellular proliferation and clonogenic growth, inhibition of G1-S phase cell cycle progression, and a decrease in human telomerase reverse transcriptase (hTERT) expression and telomerase activity. On the other hand, down-regulation of c-MYC reduced apoptosis and decreased the sensitivity of human MB cells to IR, cisplatin, and etoposide. This effect was more pronounced in DAOY cells expressing high levels of c-MYC when compared with DAOY wild-type or DAOY cells transfected with the empty vector. CONCLUSION In human MB cells, in addition to its roles in growth and proliferation, c-MYC is also a potent inducer of apoptosis. Therefore, targeting c-MYC might be of therapeutic benefit when used sequentially with chemo- and radiotherapy rather than concomitantly.
Collapse
|
25
|
Targeting the PI3K p110α Isoform Inhibits Medulloblastoma Proliferation, Chemoresistance, and Migration. Clin Cancer Res 2008; 14:6761-9. [DOI: 10.1158/1078-0432.ccr-08-0385] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
26
|
Abstract
Medulloblastoma and other embronal brain tumors are similar in appearance and differentiation potential to neural stem and progenitor cells. Expression studies performed using human tumor samples, as well as the analysis of murine transgenic models, suggest that both multipotent cerebellar stem cells and lineage-restricted progenitors of the external germinal layer can be transformed into medulloblastoma by genetic alterations. These molecular changes frequently involve constitutive activation of signaling pathways such as Wnt, Hedgehog, and Notch, which play a key role in non-neoplastic neural stem cells. Pharmacologic blockade of the Hedgehog and Notch pathways suppresses the growth of medulloblastoma in culture and in vivo and may prove effective in targeting the small cancer stem-cell subpopulation required for tumor initiation and long-term propagation.
Collapse
Affiliation(s)
- Xing Fan
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | | |
Collapse
|
27
|
Abstract
PURPOSE OF REVIEW Chemotherapy has gained a larger importance in the management of brain tumours, especially in children. RECENT FINDINGS Converging results were presented in 2005 by the German, French and North-American cooperative groups indicating that a subgroup of young children with medulloblastoma (i.e. those with desmoplastic histology) could be cured with chemotherapy only strategies. The usefulness of high-dose chemotherapy followed by stem-cell transplant was shown not only as salvage strategy but also upfront in high-risk patients with medulloblastoma. Diffuse pontine glioma remains a devastating disease despite numerous attempts to improve on the standard radiotherapy. Targeted therapies have entered the paediatric neuro-oncology field as well. SUMMARY In the most frequent paediatric brain tumors (medulloblastoma and low grade gliomas), the improvements have been impressive in recent years. These patients still await new targeted therapies to lower the burden of treatments and their related side-effects. Most of the brain tumours, however, are rare and the development of specific protocols too slow. Likely, they may have very specific biologic abnormalities that could be efficiently targeted in the near future.
Collapse
Affiliation(s)
- Jacques Grill
- Department of Paediatric and Adolescent Oncology, Gustave Roussy Cancerology Institute, Villejuif, France.
| | | |
Collapse
|