1
|
AlKhazal A, Chohan S, Ross DJ, Kim J, Brown EG. Emerging clinical and research approaches in targeted therapies for high-risk neuroblastoma. Front Oncol 2025; 15:1553511. [PMID: 40104501 PMCID: PMC11913827 DOI: 10.3389/fonc.2025.1553511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 02/06/2025] [Indexed: 03/20/2025] Open
Abstract
Neuroblastoma is a pediatric cancer that originates from neural crest cells and is the most common extracranial solid tumor in children under five years of age. While low-risk neuroblastoma often regresses spontaneously, high-risk neuroblastoma poses a significant clinical challenge. Recent advances in understanding neuroblastoma's molecular mechanisms have led to the development of targeted therapies that aim to selectively inhibit specific pathways involved in tumor growth and progression, improving patient outcomes while minimizing side effects. This review provides a comprehensive review of neuroblastoma biology and emerging therapeutic strategies. Key topics include (a) immunotherapies and immunotargets, (b) non-coding RNAs (long non-coding RNA, microRNA, and circular RNA), (c) molecular biomarkers and pathways, and (d) limitations and future directions.
Collapse
Affiliation(s)
- Albatool AlKhazal
- Department of Surgery, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Samiha Chohan
- Department of Surgery, School of Medicine, University of California, Davis, Davis, CA, United States
- Department of Biological Sciences, California State University, Sacramento, Sacramento, CA, United States
| | - Destani J Ross
- Department of Surgery, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Jinhwan Kim
- Department of Surgery, School of Medicine, University of California, Davis, Davis, CA, United States
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, United States
| | - Erin G Brown
- Department of Surgery, School of Medicine, University of California, Davis, Davis, CA, United States
| |
Collapse
|
2
|
Peggion S, Najem S, Kolman JP, Reinshagen K, Pagerols Raluy L. Revisiting Neuroblastoma: Nrf2, NF-κB and Phox2B as a Promising Network in Neuroblastoma. Curr Issues Mol Biol 2024; 46:3193-3208. [PMID: 38666930 PMCID: PMC11048850 DOI: 10.3390/cimb46040200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 03/29/2024] [Accepted: 04/02/2024] [Indexed: 04/28/2024] Open
Abstract
Neuroblastoma is the most common solid extracranial tumor during childhood; it displays extraordinary heterogeneous clinical courses, from spontaneous regression to poor outcome in high-risk patients due to aggressive growth, metastasizing, and treatment resistance. Therefore, the identification and detailed analysis of promising tumorigenic molecular mechanisms are inevitable. This review highlights the abnormal regulation of NF-κB, Nrf2, and Phox2B as well as their interactions among each other in neuroblastoma. NF-κB and Nrf2 play a key role in antioxidant responses, anti-inflammatory regulation and tumor chemoresistance. Recent studies revealed a regulation of NF-κB by means of the Nrf2/antioxidant response element (ARE) system. On the other hand, Phox2B contributes to the differentiation of immature sympathetic nervous system stem cells: this transcription factor regulates the expression of RET, thereby facilitating cell survival and proliferation. As observed in other tumors, we presume striking interactions between NF-κB, Nrf2, and Phox2B, which might constitute an important crosstalk triangle, whose decompensation may trigger a more aggressive phenotype. Consequently, these transcription factors could be a promising target for novel therapeutic approaches and hence, further investigation on their regulation in neuroblastoma shall be reinforced.
Collapse
Affiliation(s)
| | | | | | | | - Laia Pagerols Raluy
- Department of Pediatric Surgery, University Medical Centre Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| |
Collapse
|
3
|
Alhashem Z, Camargo-Sosa K, Kelsh RN, Linker C. Trunk Neural Crest Migratory Position and Asymmetric Division Predict Terminal Differentiation. Front Cell Dev Biol 2022; 10:887393. [PMID: 35756992 PMCID: PMC9214262 DOI: 10.3389/fcell.2022.887393] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 04/25/2022] [Indexed: 11/29/2022] Open
Abstract
The generation of complex structures during embryogenesis requires the controlled migration and differentiation of cells from distant origins. How these processes are coordinated and impact each other to form functional structures is not fully understood. Neural crest cells migrate extensively giving rise to many cell types. In the trunk, neural crest cells migrate collectively forming chains comprised of cells with distinct migratory identities: one leader cell at the front of the group directs migration, while followers track the leader forming the body of the chain. Herein we analysed the relationship between trunk neural crest migratory identity and terminal differentiation. We found that trunk neural crest migration and fate allocation is coherent. Leader cells that initiate movement give rise to the most distal derivativities. Interestingly, the asymmetric division of leaders separates migratory identity and fate. The distal daughter cell retains the leader identity and clonally forms the Sympathetic Ganglia. The proximal sibling migrates as a follower and gives rise to Schwann cells. The sympathetic neuron transcription factor phox2bb is strongly expressed by leaders from early stages of migration, suggesting that specification and migration occur concomitantly and in coordination. Followers divide symmetrically and their fate correlates with their position in the chain.
Collapse
Affiliation(s)
- Zain Alhashem
- Randall Centre for Cell and Molecular Biophysics, Guy's Campus, King's College London, London, United Kingdom
| | - Karen Camargo-Sosa
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
| | - Robert N Kelsh
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
| | - Claudia Linker
- Randall Centre for Cell and Molecular Biophysics, Guy's Campus, King's College London, London, United Kingdom
| |
Collapse
|
4
|
Kim HI, Schultz CR, Chandramouli GVR, Geerts D, Risinger JI, Bachmann AS. Pharmacological targeting of polyamine and hypusine biosynthesis reduces tumor activity of endometrial cancer. J Drug Target 2022; 30:623-633. [PMID: 35100927 DOI: 10.1080/1061186x.2022.2036164] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Endometrial cancer (EC) is a common and deadly cancer in women and novel therapeutic approaches are urgently needed. Polyamines (putrescine, spermidine, spermine) are critical for mammalian cell proliferation and MYC coordinately regulates polyamine metabolism through ornithine decarboxylase (ODC). ODC is a MYC target gene and rate-limiting enzyme of polyamine biosynthesis and the FDA-approved anti-protozoan drug α-difluoromethylornithine (DFMO) inhibits ODC activity and induces polyamine depletion that leads to tumor growth arrest. Spermidine is required for the hypusine-dependent activation of eukaryotic translation initiation factors 5A1 (eIF5A1) and 5A2 (eIF5A2) and connects the MYC/ODC-induced deregulation of spermidine to eIF5A1/2 protein translation, which is increased during cancer cell proliferation. We show that the eIF5A1 is significantly upregulated in EC cells compared to control cells (p = 0.000038) and that combined pharmacological targeting of ODC and eIF5A hypusination with cytostatic drugs DFMO and N1-guanyl-1,7-diaminoheptane (GC7), respectively, reduces eIF5A1 activation and synergistically induces apoptosis in EC cells. In vivo, DFMO/GC7 suppressed xenografted EC tumor growth in mice more potently than each drug alone compared to control (p = 0.002) and decreased putrescine (p = 0.045) and spermidine levels in tumor tissues. Our data suggest DFMO and GC7 combination therapy may be useful in the treatment or prevention of EC.
Collapse
Affiliation(s)
- Hong Im Kim
- Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids MI
| | - Chad R Schultz
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids MI
| | | | - Dirk Geerts
- Glycostem Therapeutics, Oss, The Netherlands
| | - John I Risinger
- Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids MI
| | - André S Bachmann
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids MI
| |
Collapse
|
5
|
The RNA-Binding Protein Musashi1 Regulates a Network of Cell Cycle Genes in Group 4 Medulloblastoma. Cells 2021; 11:cells11010056. [PMID: 35011618 PMCID: PMC8750343 DOI: 10.3390/cells11010056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 12/21/2021] [Accepted: 12/23/2021] [Indexed: 11/17/2022] Open
Abstract
Medulloblastoma is the most common malignant brain tumor in children. Treatment with surgery, irradiation, and chemotherapy has improved survival in recent years, but patients are frequently left with devastating neurocognitive and other sequelae. Patients in molecular subgroups 3 and 4 still experience a high mortality rate. To identify new pathways contributing to medulloblastoma development and create new routes for therapy, we have been studying oncogenic RNA-binding proteins. We defined Musashi1 (Msi1) as one of the main drivers of medulloblastoma development. The high expression of Msi1 is prevalent in Group 4 and correlates with poor prognosis while its knockdown disrupted cancer-relevant phenotypes. Genomic analyses (RNA-seq and RIP-seq) indicated that cell cycle and division are the main biological categories regulated by Msi1 in Group 4 medulloblastoma. The most prominent Msi1 targets include CDK2, CDK6, CCND1, CDKN2A, and CCNA1. The inhibition of Msi1 with luteolin affected the growth of CHLA-01 and CHLA-01R Group 4 medulloblastoma cells and a synergistic effect was observed when luteolin and the mitosis inhibitor, vincristine, were combined. These findings indicate that a combined therapeutic strategy (Msi1 + cell cycle/division inhibitors) could work as an alternative to treat Group 4 medulloblastoma.
Collapse
|
6
|
Nakkina SP, Gitto SB, Pandey V, Parikh JG, Geerts D, Maurer HC, Olive KP, Phanstiel O, Altomare DA. Differential Expression of Polyamine Pathways in Human Pancreatic Tumor Progression and Effects of Polyamine Blockade on Tumor Microenvironment. Cancers (Basel) 2021; 13:6391. [PMID: 34945011 PMCID: PMC8699198 DOI: 10.3390/cancers13246391] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 12/12/2021] [Accepted: 12/14/2021] [Indexed: 12/18/2022] Open
Abstract
Pancreatic cancer is the fourth leading cause of cancer death. Existing therapies only moderately improve pancreatic ductal adenocarcinoma (PDAC) patient prognosis. The present study investigates the importance of the polyamine metabolism in the pancreatic tumor microenvironment. Relative mRNA expression analysis identified differential expression of polyamine biosynthesis, homeostasis, and transport mediators in both pancreatic epithelial and stromal cells from low-grade pancreatic intraepithelial neoplasia (PanIN-1) or primary PDAC patient samples. We found dysregulated mRNA levels that encode for proteins associated with the polyamine pathway of PDAC tumors compared to early lesions. Next, bioinformatic databases were used to assess expression of select genes involved in polyamine metabolism and their impact on patient survival. Higher expression of pro-polyamine genes was associated with poor patient prognosis, supporting the use of a polyamine blockade therapy (PBT) strategy for inhibiting pancreatic tumor progression. Moreover, PBT treatment of syngeneic mice injected intra-pancreatic with PAN 02 tumor cells resulted in increased survival and decreased tumor weights of PDAC-bearing mice. Histological assessment of PBT-treated tumors revealed macrophage presence and significantly increased expression of CD86, a T cell co-stimulatory marker. Collectively, therapies which target polyamine metabolism can be used to disrupt tumor progression, modulate tumor microenvironment, and extend overall survival.
Collapse
Affiliation(s)
- Sai Preethi Nakkina
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA;
| | - Sarah B. Gitto
- Ovarian Cancer Research Center, Division of Gynecology Oncology, Department of Obstetrics and Gynecology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (S.B.G.); (V.P.)
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Veethika Pandey
- Ovarian Cancer Research Center, Division of Gynecology Oncology, Department of Obstetrics and Gynecology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (S.B.G.); (V.P.)
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jignesh G. Parikh
- Department of Pathology, Orlando VA Medical Center, 13800 Veterans Way, Orlando, FL 32827, USA;
| | - Dirk Geerts
- Department of Medical Biology, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands;
| | - Hans Carlo Maurer
- Internal Medicine II, School of Medicine, Technische Universität München, 81675 Munich, Germany;
| | - Kenneth P. Olive
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA;
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Otto Phanstiel
- Department of Medical Education, College of Medicine, University of Central Florida, Orlando, FL 32826, USA
| | - Deborah A. Altomare
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA;
| |
Collapse
|
7
|
Perri P, Ponzoni M, Corrias MV, Ceccherini I, Candiani S, Bachetti T. A Focus on Regulatory Networks Linking MicroRNAs, Transcription Factors and Target Genes in Neuroblastoma. Cancers (Basel) 2021; 13:5528. [PMID: 34771690 PMCID: PMC8582685 DOI: 10.3390/cancers13215528] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 10/26/2021] [Accepted: 10/28/2021] [Indexed: 12/17/2022] Open
Abstract
Neuroblastoma (NB) is a tumor of the peripheral sympathetic nervous system that substantially contributes to childhood cancer mortality. NB originates from neural crest cells (NCCs) undergoing a defective sympathetic neuronal differentiation and although the starting events leading to the development of NB remain to be fully elucidated, the master role of genetic alterations in key oncogenes has been ascertained: (1) amplification and/or over-expression of MYCN, which is strongly associated with tumor progression and invasion; (2) activating mutations, amplification and/or over-expression of ALK, which is involved in tumor initiation, angiogenesis and invasion; (3) amplification and/or over-expression of LIN28B, promoting proliferation and suppression of neuroblast differentiation; (4) mutations and/or over-expression of PHOX2B, which is involved in the regulation of NB differentiation, stemness maintenance, migration and metastasis. Moreover, altered microRNA (miRNA) expression takes part in generating pathogenetic networks, in which the regulatory loops among transcription factors, miRNAs and target genes lead to complex and aberrant oncogene expression that underlies the development of a tumor. In this review, we have focused on the circuitry linking the oncogenic transcription factors MYCN and PHOX2B with their transcriptional targets ALK and LIN28B and the tumor suppressor microRNAs let-7, miR-34 and miR-204, which should act as down-regulators of their expression. We have also looked at the physiologic role of these genetic and epigenetic determinants in NC development, as well as in terminal differentiation, with their pathogenic dysregulation leading to NB oncogenesis.
Collapse
Affiliation(s)
- Patrizia Perri
- Laboratory of Experimental Therapies in Oncology, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy; (M.P.); (M.V.C.)
| | - Mirco Ponzoni
- Laboratory of Experimental Therapies in Oncology, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy; (M.P.); (M.V.C.)
| | - Maria Valeria Corrias
- Laboratory of Experimental Therapies in Oncology, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy; (M.P.); (M.V.C.)
| | - Isabella Ceccherini
- Laboratory of Genetics and Genomics of Rare Diseases, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy;
| | - Simona Candiani
- Department of Earth, Environment and Life Sciences, University of Genoa, 16132 Genoa, Italy;
| | - Tiziana Bachetti
- Laboratory of Genetics and Genomics of Rare Diseases, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy;
- Department of Earth, Environment and Life Sciences, University of Genoa, 16132 Genoa, Italy;
| |
Collapse
|
8
|
Kempers L, Wakayama Y, van der Bijl I, Furumaya C, De Cuyper IM, Jongejan A, Kat M, van Stalborch AMD, van Boxtel AL, Hubert M, Geerts D, van Buul JD, de Korte D, Herzog W, Margadant C. The endosomal RIN2/Rab5C machinery prevents VEGFR2 degradation to control gene expression and tip cell identity during angiogenesis. Angiogenesis 2021; 24:695-714. [PMID: 33983539 PMCID: PMC8292304 DOI: 10.1007/s10456-021-09788-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 03/05/2021] [Indexed: 12/13/2022]
Abstract
Sprouting angiogenesis is key to many pathophysiological conditions, and is strongly regulated by vascular endothelial growth factor (VEGF) signaling through VEGF receptor 2 (VEGFR2). Here we report that the early endosomal GTPase Rab5C and its activator RIN2 prevent lysosomal routing and degradation of VEGF-bound, internalized VEGFR2 in human endothelial cells. Stabilization of endosomal VEGFR2 levels by RIN2/Rab5C is crucial for VEGF signaling through the ERK and PI3-K pathways, the expression of immediate VEGF target genes, as well as specification of angiogenic 'tip' and 'stalk' cell phenotypes and cell sprouting. Using overexpression of Rab mutants, knockdown and CRISPR/Cas9-mediated gene editing, and live-cell imaging in zebrafish, we further show that endosomal stabilization of VEGFR2 levels is required for developmental angiogenesis in vivo. In contrast, the premature degradation of internalized VEGFR2 disrupts VEGF signaling, gene expression, and tip cell formation and migration. Thus, an endosomal feedforward mechanism maintains receptor signaling by preventing lysosomal degradation, which is directly linked to the induction of target genes and cell fate in collectively migrating cells during morphogenesis.
Collapse
Affiliation(s)
- Lanette Kempers
- Sanquin Research, Plesmanlaan 125, 1066 CX, Amsterdam, The Netherlands
| | - Yuki Wakayama
- Max Planck Institute for Molecular Biomedicine, Roentgenstrasse 20, 48149, Muenster, Germany
| | - Ivo van der Bijl
- Sanquin Research, Plesmanlaan 125, 1066 CX, Amsterdam, The Netherlands
| | - Charita Furumaya
- Sanquin Research, Plesmanlaan 125, 1066 CX, Amsterdam, The Netherlands
| | - Iris M De Cuyper
- Sanquin Research, Plesmanlaan 125, 1066 CX, Amsterdam, The Netherlands
| | - Aldo Jongejan
- Department of Epidemiology and Data Science /Amsterdam Public Health Research Institute, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Marije Kat
- Sanquin Research, Plesmanlaan 125, 1066 CX, Amsterdam, The Netherlands
| | | | - Antonius L van Boxtel
- Cancer Biology and Genetics and Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.,Developmental, Stem Cell and Cancer Biology, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, The Netherlands
| | - Marvin Hubert
- University of Muenster, Schlossplatz 2, 48149, Muenster, Germany
| | - Dirk Geerts
- Department of Medical Biology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Jaap D van Buul
- Sanquin Research, Plesmanlaan 125, 1066 CX, Amsterdam, The Netherlands
| | - Dirk de Korte
- Sanquin Research, Plesmanlaan 125, 1066 CX, Amsterdam, The Netherlands.,Sanquin Blood Bank, Plesmanlaan 125, 1066 CX, Amsterdam, The Netherlands
| | - Wiebke Herzog
- Max Planck Institute for Molecular Biomedicine, Roentgenstrasse 20, 48149, Muenster, Germany.,University of Muenster, Schlossplatz 2, 48149, Muenster, Germany
| | - Coert Margadant
- Angiogenesis Laboratory, Department of Medical Oncology, Amsterdam University Medical Center, location VUmc, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.
| |
Collapse
|
9
|
Liu C, Huang M, Han C, Li H, Wang J, Huang Y, Chen Y, Zhu J, Fu G, Yu H, Lei Z, Chu X. A narrative review of the roles of muscle segment homeobox transcription factor family in cancer. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:810. [PMID: 34268423 PMCID: PMC8246185 DOI: 10.21037/atm-21-220] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 03/05/2021] [Indexed: 11/23/2022]
Abstract
Deregulation of many homeobox genes has been observed in various cancers and has caused functional implications in the tumor progression. In this review, we will focus on the roles of the human muscle segment homeobox (MSX) transcription factor family in the process of tumorigenesis. The MSX transcription factors, through complex downstream regulation mechanisms, are promoters or inhibitors of diverse cancers by participating in cell proliferation, cell invasion, cell metastasis, cell apoptosis, cell differentiation, drug resistance of tumors, maintenance of tumor stemness, and tumor angiogenesis. Moreover, their upstream regulatory mechanisms in cancers may include: gene mutation and chromosome aberration; DNA methylation and chromatin modification; regulation by non-coding RNAs; regulation by other transcription factors and post-translational modification. These mechanisms may provide a better understanding of why MSX transcription factors are abnormally expressed in tumors. Notably, intermolecular interactions and post-translational modification can regulate the transcriptional activity of MSX transcription factors. It is also crucial to know what affects the transcriptional activity of MSX transcription factors in tumors for possible interventions in them in the future. This systematic summary of the regulatory patterns of the MSX transcription factor family may help to further understand the mechanisms involved in transcriptional regulation and also provide new therapeutic approaches for tumor progression.
Collapse
Affiliation(s)
- Chao Liu
- Department of Medical Oncology, Jinling Hospital, Nanjing Medical University, Nanjing, China
| | - Mengxi Huang
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Chao Han
- Department of Medical Oncology, Jinling Hospital, Nanjing Medical University, Nanjing, China
| | - Huiyu Li
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Jing Wang
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Yadi Huang
- Department of Medical Oncology, Jinling Hospital, First School of Clinical Medicine, Southern Medical University, Nanjing, China
| | - Yanyan Chen
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Jialong Zhu
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Gongbo Fu
- Department of Medical Oncology, Jinling Hospital, Nanjing Medical University, Nanjing, China.,Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Hanqing Yu
- Department of Clinical Laboratory, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Zengjie Lei
- Department of Medical Oncology, Jinling Hospital, Nanjing Medical University, Nanjing, China.,Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Xiaoyuan Chu
- Department of Medical Oncology, Jinling Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
10
|
Zafar A, Wang W, Liu G, Wang X, Xian W, McKeon F, Foster J, Zhou J, Zhang R. Molecular targeting therapies for neuroblastoma: Progress and challenges. Med Res Rev 2020; 41:961-1021. [PMID: 33155698 PMCID: PMC7906923 DOI: 10.1002/med.21750] [Citation(s) in RCA: 213] [Impact Index Per Article: 42.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 09/25/2020] [Accepted: 10/28/2020] [Indexed: 01/09/2023]
Abstract
There is an urgent need to identify novel therapies for childhood cancers. Neuroblastoma is the most common pediatric solid tumor, and accounts for ~15% of childhood cancer‐related mortality. Neuroblastomas exhibit genetic, morphological and clinical heterogeneity, which limits the efficacy of existing treatment modalities. Gaining detailed knowledge of the molecular signatures and genetic variations involved in the pathogenesis of neuroblastoma is necessary to develop safer and more effective treatments for this devastating disease. Recent studies with advanced high‐throughput “omics” techniques have revealed numerous genetic/genomic alterations and dysfunctional pathways that drive the onset, growth, progression, and resistance of neuroblastoma to therapy. A variety of molecular signatures are being evaluated to better understand the disease, with many of them being used as targets to develop new treatments for neuroblastoma patients. In this review, we have summarized the contemporary understanding of the molecular pathways and genetic aberrations, such as those in MYCN, BIRC5, PHOX2B, and LIN28B, involved in the pathogenesis of neuroblastoma, and provide a comprehensive overview of the molecular targeted therapies under preclinical and clinical investigations, particularly those targeting ALK signaling, MDM2, PI3K/Akt/mTOR and RAS‐MAPK pathways, as well as epigenetic regulators. We also give insights on the use of combination therapies involving novel agents that target various pathways. Further, we discuss the future directions that would help identify novel targets and therapeutics and improve the currently available therapies, enhancing the treatment outcomes and survival of patients with neuroblastoma.
Collapse
Affiliation(s)
- Atif Zafar
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas, USA
| | - Wei Wang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas, USA.,Drug Discovery Institute, University of Houston, Houston, Texas, USA
| | - Gang Liu
- Department of Pharmacology and Toxicology, Chemical Biology Program, University of Texas Medical Branch, Galveston, Texas, USA
| | - Xinjie Wang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas, USA
| | - Wa Xian
- Department of Biology and Biochemistry, Stem Cell Center, University of Houston, Houston, Texas, USA
| | - Frank McKeon
- Department of Biology and Biochemistry, Stem Cell Center, University of Houston, Houston, Texas, USA
| | - Jennifer Foster
- Department of Pediatrics, Texas Children's Hospital, Section of Hematology-Oncology Baylor College of Medicine, Houston, Texas, USA
| | - Jia Zhou
- Department of Pharmacology and Toxicology, Chemical Biology Program, University of Texas Medical Branch, Galveston, Texas, USA
| | - Ruiwen Zhang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas, USA.,Drug Discovery Institute, University of Houston, Houston, Texas, USA
| |
Collapse
|
11
|
Abstract
Neuroblastoma (NB) is a pediatric tumor of embryonic origin. About 1-2% of all NBs are familial cases, and genetic predisposition is suspected for the remaining cases. During the last decade, genome-wide association studies (GWAS) and high-throughput sequencing approaches have been used to identify associations among common and rare genetic variants and NB risk. Substantial data has been produced by large patient cohorts that implicate various genes in NB tumorigenesis, such as CASC15, BARD1, CHEK2, LMO1, LIN28B, AXIN2, BRCA1, TP53, SMARCA4, and CDK1NB. NB, as well as other pediatric cancers, has few recurrent mutations but several copy number variations (CNVs). Almost all NBs show both numerical and structural CNVs. The proportion between numerical and structural CNVs differs between localized and metastatic tumors, with a greater prevalence of structural CNVs in metastatic NB. This genomic chaos frequently identified in NBs suggests that chromosome instability (CIN) could be one of the major actors in NB oncogenesis. Interestingly, many NB-predisposing variants occur in genes involved in the control of genome stability, mitosis, and normal chromosome separation. Here, we discuss the relationship between genetic predisposition and CIN in NB.
Collapse
Affiliation(s)
- Gian Paolo Tonini
- Neuroblastoma Laboratory, Pediatric Research Institute, Città della Speranza, Corso Stati Uniti 4, 35127, Padova, Italy.
| | - Mario Capasso
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Univeristà degli Studi di Napoli Federico II, Naples, Italy. .,CEINGE Biotecnologie Avanzate, Naples, Italy.
| |
Collapse
|
12
|
Eppich S, Kuhn C, Schmoeckel E, Mayr D, Mahner S, Jeschke U, Gallwas J, Heidegger HH. MSX1-A Potential Marker for Uterus-Preserving Therapy of Endometrial Carcinomas. Int J Mol Sci 2020; 21:ijms21124529. [PMID: 32630554 PMCID: PMC7350265 DOI: 10.3390/ijms21124529] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 06/17/2020] [Accepted: 06/22/2020] [Indexed: 12/28/2022] Open
Abstract
Prognostic factors are of great interest in patients with endometrial cancer. One potential factor could be the protein MSX1, a transcription repressor, that has an inhibitory effect on the cell cycle. For this study, endometrioid endometrial carcinomas (n = 53), clear cell endometrial carcinomas (n = 6), endometrioid ovarian carcinomas (n = 19), and clear cell ovarian carcinomas (n = 11) were immunochemically stained for the protein MSX1 and evaluated using the immunoreactive score (IRS). A significant stronger expression of MSX1 was found in endometrioid endometrial carcinomas (p < 0.001), in grading 2 (moderate differentiation) (p = 0.001), and in tumor material of patients with no involvement of lymph nodes (p = 0.031). Correlations were found between MSX1 expression and the expression of β-Catenin, p21, p53, and the steroid receptors ERα, ERβ, PRα, and PRβ. A significant (p = 0.023) better survival for patients with an MSX1 expression in more than 10% of the tumor cells was observed for endometrioid endometrial carcinomas (21.3 years median survival (MSX1-positive) versus 17.3 years (MSX1-negative)). Although there is evidence that MSX1 expression correlates with improved long-term survival, further studies are necessary to evaluate if MSX1 can be used as a prognostic marker.
Collapse
Affiliation(s)
- Simon Eppich
- Department of Obstetrics and Gynecology, University Hospital, Ludwig Maximilians University (LMU), Marchioninistraße 15, 81377 Munich, Germany; (S.E.); (C.K.); (S.M.); (J.G.); (H.H.H.)
| | - Christina Kuhn
- Department of Obstetrics and Gynecology, University Hospital, Ludwig Maximilians University (LMU), Marchioninistraße 15, 81377 Munich, Germany; (S.E.); (C.K.); (S.M.); (J.G.); (H.H.H.)
| | - Elisa Schmoeckel
- Department of Pathology, LMU Munich, Thalkirchner Str. 56, 80337 Munich, Germany; (E.S.); (D.M.)
| | - Doris Mayr
- Department of Pathology, LMU Munich, Thalkirchner Str. 56, 80337 Munich, Germany; (E.S.); (D.M.)
| | - Sven Mahner
- Department of Obstetrics and Gynecology, University Hospital, Ludwig Maximilians University (LMU), Marchioninistraße 15, 81377 Munich, Germany; (S.E.); (C.K.); (S.M.); (J.G.); (H.H.H.)
| | - Udo Jeschke
- Department of Obstetrics and Gynecology, University Hospital, Ludwig Maximilians University (LMU), Marchioninistraße 15, 81377 Munich, Germany; (S.E.); (C.K.); (S.M.); (J.G.); (H.H.H.)
- Department of Obstetrics and Gynecology, University Hospital Augsburg, Stenglinstr. 2, 86156 Augsburg, Germany
- Correspondence: ; Tel.: +49-89-4400-54240
| | - Julia Gallwas
- Department of Obstetrics and Gynecology, University Hospital, Ludwig Maximilians University (LMU), Marchioninistraße 15, 81377 Munich, Germany; (S.E.); (C.K.); (S.M.); (J.G.); (H.H.H.)
- Department of Gynecology and Obstetrics, Georg August University Goettingen, University Medicine, 37075 Goettingen, Germany
| | - Helene Hildegard Heidegger
- Department of Obstetrics and Gynecology, University Hospital, Ludwig Maximilians University (LMU), Marchioninistraße 15, 81377 Munich, Germany; (S.E.); (C.K.); (S.M.); (J.G.); (H.H.H.)
| |
Collapse
|
13
|
A Wnt-BMP4 Signaling Axis Induces MSX and NOTCH Proteins and Promotes Growth Suppression and Differentiation in Neuroblastoma. Cells 2020; 9:cells9030783. [PMID: 32210188 PMCID: PMC7140810 DOI: 10.3390/cells9030783] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 03/18/2020] [Accepted: 03/19/2020] [Indexed: 01/09/2023] Open
Abstract
The Wnt and bone morphogenetic protein (BMP) signaling pathways are known to be crucial in the development of neural crest lineages, including the sympathetic nervous system. Surprisingly, their role in paediatric neuroblastoma, the prototypic tumor arising from this lineage, remains relatively uncharacterised. We previously demonstrated that Wnt/b-catenin signaling can have cell-type-specific effects on neuroblastoma phenotypes, including growth inhibition and differentiation, and that BMP4 mRNA and protein were induced by Wnt3a/Rspo2. In this study, we characterised the phenotypic effects of BMP4 on neuroblastoma cells, demonstrating convergent induction of MSX homeobox transcription factors by Wnt and BMP4 signaling and BMP4-induced growth suppression and differentiation. An immunohistochemical analysis of BMP4 expression in primary neuroblastomas confirms a striking absence of BMP4 in poorly differentiated tumors, in contrast to a high expression in ganglion cells. These results are consistent with a tumor suppressive role for BMP4 in neuroblastoma. RNA sequencing following BMP4 treatment revealed induction of Notch signaling, verified by increases of Notch3 and Hes1 proteins. Together, our data demonstrate, for the first time, Wnt-BMP-Notch signaling crosstalk associated with growth suppression of neuroblastoma.
Collapse
|
14
|
Lin A, Fu W, Wang W, Zhu J, Liu J, Xia H, Liu G, He J. Association between PHOX2B gene rs28647582 T>C polymorphism and Wilms tumor susceptibility. Biosci Rep 2019; 39:BSR20192529. [PMID: 31652452 PMCID: PMC6822530 DOI: 10.1042/bsr20192529] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 09/08/2019] [Accepted: 10/03/2019] [Indexed: 02/07/2023] Open
Abstract
Wilms tumor is one of the most common pediatric solid tumors. The pair-like homeobox 2b (PHOX2B) gene is an important transcription factor that regulates cellular proliferation and differentiation in early life. The association between PHOX2B single nucleotide polymorphisms (SNPs) and Wilms tumor risk has not been investigated. Therefore, we conducted a case-control study involving 145 Wilms tumor patients and 531 controls to explore the association between the PHOX2B rs28647582 T>C polymorphism and Wilms tumor susceptibility. The association between the PHOX2B rs28647582 T>C polymorphism and Wilms tumor susceptibility was assessed by odds ratios (ORs) and 95% confidence intervals (CIs). Our results indicated that PHOX2B rs28647582 T>C polymorphism did not significantly alter Wilms tumor susceptibility. However, in the stratified analysis, we found that TC/CC genotypes significantly increased Wilms tumor risk among children older than 18 months (adjusted OR = 1.77, 95% CI = 1.07-2.95, P=0.027) and those with clinical stages III+IV (adjusted OR = 1.75, 95% CI = 1.09-2.82, P=0.022), when compared with those with TT genotype. Our study suggested that PHOX2B rs28647582 T>C was weakly associated with Wilms tumor susceptibility. Our conclusions need further validation with a larger sample size.
Collapse
Affiliation(s)
- Ao Lin
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Wen Fu
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Wenwen Wang
- Department of Oncology, The Affiliated Wuxi No.2 People’s Hospital of Nanjing Medical University, Wuxi 214000, Jiangsu, China
| | - Jinhong Zhu
- Department of Clinical Laboratory, Biobank, Harbin Medical University Cancer Hospital, Harbin 150040, Heilongjiang, China
| | - Jiabin Liu
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Huimin Xia
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Guochang Liu
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Jing He
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| |
Collapse
|
15
|
Bachetti T, Ceccherini I. Causative and commonPHOX2Bvariants define a broad phenotypic spectrum. Clin Genet 2019; 97:103-113. [DOI: 10.1111/cge.13633] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 07/31/2019] [Accepted: 08/15/2019] [Indexed: 11/25/2022]
Affiliation(s)
- Tiziana Bachetti
- Laboratorio Neurobiologia dello Sviluppo, Dipartimento di Scienze della Terra dell'Ambiente e della Vita (DISTAV)Università di Genova Genova Italy
| | | |
Collapse
|
16
|
Testori A, Lasorsa VA, Cimmino F, Cantalupo S, Cardinale A, Avitabile M, Limongelli G, Russo MG, Diskin S, Maris J, Devoto M, Keavney B, Cordell HJ, Iolascon A, Capasso M. Exploring Shared Susceptibility between Two Neural Crest Cells Originating Conditions: Neuroblastoma and Congenital Heart Disease. Genes (Basel) 2019; 10:genes10090663. [PMID: 31480262 PMCID: PMC6771154 DOI: 10.3390/genes10090663] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 08/12/2019] [Accepted: 08/26/2019] [Indexed: 12/13/2022] Open
Abstract
In the past years, genome wide association studies (GWAS) have provided evidence that inter-individual susceptibility to diverse pathological conditions can reveal a common genetic architecture. Through the analysis of congenital heart disease (CHD) and neuroblastoma (NB) GWAS data, we aimed to dissect the genetic susceptibility shared between these conditions, which are known to arise from neural crest cell (NCC) migration or development abnormalities, via identification and functional characterization of common regions of association. Two loci (2q35 and 3q25.32) harbor single nucleotide polymorphisms (SNPs) that are associated at a p-value < 10-3 with conotruncal malformations and ventricular septal defect respectively, as well as with NB. In addition, the lead SNP in 4p16.2 for atrial septal defect and the lead SNP in 3q25.32 for tetralogy of Fallot are less than 250 Kb distant from the lead SNPs for NB at the same genomic regions. Some of these shared susceptibility loci regulate the expression of relevant genes involved in NCC formation and developmental processes (such as BARD1, MSX1, and SHOX2) and are enriched in several epigenetic markers from NB and fetal heart cell lines. Although the clinical correlation between NB and CHD is unclear, our exploration of a possible common genetic basis between NB and a subset of cardiac malformations can help shed light on their shared embryological origin and pathogenetic mechanisms.
Collapse
Affiliation(s)
- Alessandro Testori
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, 80131 Naples, Italy
- CEINGE Biotecnologie Avanzate, 80145 Naples, Italy
| | - Vito A Lasorsa
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, 80131 Naples, Italy
- CEINGE Biotecnologie Avanzate, 80145 Naples, Italy
| | - Flora Cimmino
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, 80131 Naples, Italy
- CEINGE Biotecnologie Avanzate, 80145 Naples, Italy
| | - Sueva Cantalupo
- IRCCS SDN, Istituto di Ricerca Diagnostica e Nucleare, 80143 Naples, Italy
| | - Antonella Cardinale
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, 80131 Naples, Italy
- CEINGE Biotecnologie Avanzate, 80145 Naples, Italy
| | - Marianna Avitabile
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, 80131 Naples, Italy
- CEINGE Biotecnologie Avanzate, 80145 Naples, Italy
| | - Giuseppe Limongelli
- Division of Cardiology, Università degli Studi della Campania "Luigi Vanvitelli" - AO dei Colli, Presidio Monaldi, 80121 Naples, Italy
| | - Maria Giovanna Russo
- Division of Cardiology, Università degli Studi della Campania "Luigi Vanvitelli" - AO dei Colli, Presidio Monaldi, 80121 Naples, Italy
| | - Sharon Diskin
- Division of Oncology and Center for Childhood Cancer Research, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - John Maris
- Division of Oncology and Center for Childhood Cancer Research, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Marcella Devoto
- Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Division of Genetics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Translational and Precision Medicine, University of Rome "La Sapienza", 00185 Rome, Italy
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Bernard Keavney
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK
- Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester M20 4BX, UK
| | - Heather J Cordell
- Institute of Genetic Medicine, Newcastle University, Central Parkway, Newcastle upon Tyne NE1 3BZ, UK
| | - Achille Iolascon
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, 80131 Naples, Italy
- CEINGE Biotecnologie Avanzate, 80145 Naples, Italy
| | - Mario Capasso
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, 80131 Naples, Italy.
- CEINGE Biotecnologie Avanzate, 80145 Naples, Italy.
- IRCCS SDN, Istituto di Ricerca Diagnostica e Nucleare, 80143 Naples, Italy.
| |
Collapse
|
17
|
Mooney MR, Geerts D, Kort EJ, Bachmann AS. Anti-tumor effect of sulfasalazine in neuroblastoma. Biochem Pharmacol 2019; 162:237-249. [DOI: 10.1016/j.bcp.2019.01.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 01/08/2019] [Indexed: 01/18/2023]
|
18
|
Di Lascio S, Benfante R, Cardani S, Fornasari D. Advances in the molecular biology and pathogenesis of congenital central hypoventilation syndrome—implications for new therapeutic targets. Expert Opin Orphan Drugs 2018. [DOI: 10.1080/21678707.2018.1540978] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Affiliation(s)
- Simona Di Lascio
- Department of Medical Biotechnology and Translational Medicine (BIOMETRA), Università degli Studi di Milano, Milan, Italy
| | - Roberta Benfante
- Department of Medical Biotechnology and Translational Medicine (BIOMETRA), Università degli Studi di Milano, Milan, Italy
- CNR- Neuroscience Institute, Milan, Italy
| | - Silvia Cardani
- Department of Medical Biotechnology and Translational Medicine (BIOMETRA), Università degli Studi di Milano, Milan, Italy
| | - Diego Fornasari
- Department of Medical Biotechnology and Translational Medicine (BIOMETRA), Università degli Studi di Milano, Milan, Italy
- CNR- Neuroscience Institute, Milan, Italy
| |
Collapse
|
19
|
Yue Y, Zhou K, Li J, Jiang S, Li C, Men H. MSX1 induces G0/G1 arrest and apoptosis by suppressing Notch signaling and is frequently methylated in cervical cancer. Onco Targets Ther 2018; 11:4769-4780. [PMID: 30127625 PMCID: PMC6091477 DOI: 10.2147/ott.s165144] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
PURPOSE The objectives of this study were to investigate the expression of MSX1 in cervical cells and tissues, the methylation status of the MSX1 promoter, the influence of overexpression of gene MSX1 on the proliferation, migration, and invasion of HeLa and SiHa cells, and finally the possible molecular mechanisms responsible for the suppressive effects of MSX1 upon cervical cancer cells. PATIENTS AND METHODS Semi-quantitative and quantitative reverse transcription-polymerase chain reactions were used to investigate the expression levels of MSX1, and methylation-specific polymerase chain reaction (MSP) was performed to investigate promoter methylation status in cervical cancer cell lines, primary cervical tissues, and normal cervical tissues. Clone formation, Cell Counting Kit-8 (CCK-8), cell wound scratch, and transwell assays were performed to verify whether MSX1 could inhibit the proliferation and migration of cervical cancer cells. Western blot was used to analyze the effect of MSX1 upon Notch1, Jagged1, c-Myc, cleaved PARP, cleaved caspse-3, and cyclin D1 (CCND1). RESULTS MSX1 was frequently downregulated or silenced in 60.0% (3/5) of cervical cancer cell lines. The promoter methylation of MSX1 was detected in 42.0% (42/100) of primary tumor tissues, while no methylation was observed in normal cervical tissues. Pharmacological demethylation reduced MSX1 promoter methylation levels and restored the expression of MSX1. The overexpression of MSX1 in cervical cancer cells thus inhibited the proliferation and migration of cervical cancer cells. The overexpression of MSX1 in cervical cancer cells downregulated the expression levels of Notch1, Jagged1, and c-Myc but upregulated the expression levels of CCND1, cleaved PARP, and cleaved caspase-3. CONCLUSION MSX1 appears to be a functional tumor suppressor that regulates tumorigenesis in cervical cancer by antagonizing Notch signaling.
Collapse
Affiliation(s)
- Yujuan Yue
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Kun Zhou
- Clinical Center for Tumor Therapy, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China,
| | - Jiachu Li
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Shan Jiang
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Chunyan Li
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Haitao Men
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| |
Collapse
|
20
|
Uhl KL, Schultz CR, Geerts D, Bachmann AS. Harmine, a dual-specificity tyrosine phosphorylation-regulated kinase (DYRK) inhibitor induces caspase-mediated apoptosis in neuroblastoma. Cancer Cell Int 2018; 18:82. [PMID: 29977157 PMCID: PMC5992763 DOI: 10.1186/s12935-018-0574-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 05/26/2018] [Indexed: 11/12/2022] Open
Abstract
Background Neuroblastoma (NB) is an early childhood malignancy that arises from the developing sympathetic nervous system. Harmine is a tricyclic β-carboline alkaloid isolated from the harmal plant that exhibits both cytostatic and cytotoxic effects. Harmine is capable of blocking the activities of dual-specificity tyrosine phosphorylation-regulated kinase (DYRK) family proteins and mitogen-activated protein kinase. These kinases promote proliferation and inhibit apoptosis. Methods Four human NB cell lines were used to study the effects of harmine treatment: SKNBE and KELLY (MYCN-amplified) as well as SKNAS and SKNFI (MYCN non-amplified). The anti-cancer properties of harmine were examined by RealTime-Glo MT cell viability assays, caspase activity assays, PARP cleavage using Western blot analysis, and flow cytometry-based Annexin V detection. A molecular interaction model of harmine bound to the DYRK2 family kinase was generated by computational docking using X-ray structures. NB tumors from human patients were profiled for DYRK mRNA expression patterns and clinical correlations using the R2 platform. Results The IC50 values for harmine after 72 h treatment were 169.6, 170.8, and 791.7 μM for SKNBE, KELLY, and SKNFI, respectively. Exposure of these NB cell lines to 100 μM of harmine resulted in caspase-3/7 and caspase-9 activation as well as caspase-mediated PARP cleavage and Annexin V-positive stained cells, as early as 24 h after treatment, clearly suggesting apoptosis induction, especially in MYCN-amplified cell lines. Elevated DYRK2 mRNA levels correlated with poor prognosis in a large cohort of NB tumors. Conclusion Harmine is a known inhibitor of DYRK family kinases. It can induce apoptosis in NB cell lines, which led us to investigate the clinical correlations of DYRK family gene expression in NB tumors. The patient results support our hypothesis that DYRK inhibition by harmine and the subsequent triggering of caspase-mediated apoptosis might present a novel approach to NB therapy. Electronic supplementary material The online version of this article (10.1186/s12935-018-0574-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Katie L Uhl
- 1Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, 400 Monroe Avenue NW, Grand Rapids, MI 49503 USA
| | - Chad R Schultz
- 1Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, 400 Monroe Avenue NW, Grand Rapids, MI 49503 USA
| | - Dirk Geerts
- 2Department of Medical Biology, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - André S Bachmann
- 1Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, 400 Monroe Avenue NW, Grand Rapids, MI 49503 USA
| |
Collapse
|
21
|
Ménard M, Costechareyre C, Ichim G, Blachier J, Neves D, Jarrosson-Wuilleme L, Depping R, Koster J, Saintigny P, Mehlen P, Tauszig-Delamasure S. Hey1- and p53-dependent TrkC proapoptotic activity controls neuroblastoma growth. PLoS Biol 2018; 16:e2002912. [PMID: 29750782 PMCID: PMC5965893 DOI: 10.1371/journal.pbio.2002912] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 05/23/2018] [Accepted: 04/13/2018] [Indexed: 11/19/2022] Open
Abstract
The neurotrophin-3 (NT-3) receptor tropomyosin receptor kinase C (TrkC/NTRK3) has been described as a dependence receptor and, as such, triggers apoptosis in the absence of its ligand NT-3. This proapoptotic activity has been proposed to confer a tumor suppressor activity to this classic tyrosine kinase receptor (RTK). By investigating interacting partners that might facilitate TrkC-induced cell death, we have identified the basic helix-loop-helix (bHLH) transcription factor Hey1 and importin-α3 (karyopherin alpha 4 [KPNA4]) as direct interactors of TrkC intracellular domain, and we show that Hey1 is required for TrkC-induced apoptosis. We propose here that the cleaved proapoptotic portion of TrkC intracellular domain (called TrkC killer-fragment [TrkC-KF]) is translocated to the nucleus by importins and interacts there with Hey1. We also demonstrate that Hey1 and TrkC-KF transcriptionally silence mouse double minute 2 homolog (MDM2), thus contributing to p53 stabilization. p53 transcriptionally regulates the expression of TrkC-KF cytoplasmic and mitochondrial interactors cofactor of breast cancer 1 (COBRA1) and B cell lymphoma 2–associated X (BAX), which will subsequently trigger the intrinsic pathway of apoptosis. Of interest, TrkC was proposed to constrain tumor progression in neuroblastoma (NB), and we demonstrate in an avian model that TrkC tumor suppressor activity requires Hey1 and p53. Tropomyosin receptor kinase C (TrkC) is a transmembrane receptor at the cell surface and has been described to work paradoxically both as an oncogene and as a tumor suppressor. We partly solved this paradox in a previous study, demonstrating that TrkC is a double-facet receptor: Upon interaction with its ligand neurotrophin-3 (NT-3), TrkC has a tyrosine kinase activity and induces survival and proliferation of the cell; conversely, in the absence of the ligand, TrkC is cleaved and releases a "killer-fragment" that triggers apoptosis. In this study, we analyze the fate of this fragment and show that TrkC killer-fragment is translocated to the nucleus, where it stabilizes the apoptosis inducer p53. We further find that p53 activates the transcription of cytoplasmic molecular partners, which interact with TrkC killer-fragment and induce apoptosis. We also demonstrate that alteration of this mechanism favors tumor growth in neuroblastoma (NB), an avian tumor progression model for a pediatric cancer.
Collapse
Affiliation(s)
- Marie Ménard
- Apoptosis, Cancer and Development Laboratory—Equipe labellisée ‘La Ligue’, LabEx DEVweCAN, Centre de Recherche en Cancérologie de Lyon, INSERM U1052-CNRS UMR5286, Université de Lyon, Centre Léon Bérard, Lyon, France
| | - Clélia Costechareyre
- Apoptosis, Cancer and Development Laboratory—Equipe labellisée ‘La Ligue’, LabEx DEVweCAN, Centre de Recherche en Cancérologie de Lyon, INSERM U1052-CNRS UMR5286, Université de Lyon, Centre Léon Bérard, Lyon, France
| | - Gabriel Ichim
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052-CNRS UMR5286, Université de Lyon, Centre Léon Bérard, Lyon, France
| | - Jonathan Blachier
- Apoptosis, Cancer and Development Laboratory—Equipe labellisée ‘La Ligue’, LabEx DEVweCAN, Centre de Recherche en Cancérologie de Lyon, INSERM U1052-CNRS UMR5286, Université de Lyon, Centre Léon Bérard, Lyon, France
| | - David Neves
- Apoptosis, Cancer and Development Laboratory—Equipe labellisée ‘La Ligue’, LabEx DEVweCAN, Centre de Recherche en Cancérologie de Lyon, INSERM U1052-CNRS UMR5286, Université de Lyon, Centre Léon Bérard, Lyon, France
| | - Loraine Jarrosson-Wuilleme
- Apoptosis, Cancer and Development Laboratory—Equipe labellisée ‘La Ligue’, LabEx DEVweCAN, Centre de Recherche en Cancérologie de Lyon, INSERM U1052-CNRS UMR5286, Université de Lyon, Centre Léon Bérard, Lyon, France
| | - Reinhard Depping
- Universität zu Lübeck, Institut für Physiologie, Zentrum für Medizinische Struktur und Zellbiologie, Lübeck, Germany
| | - Jan Koster
- Department of Oncogenomics, Academic Medical Center, Amsterdam, the Netherlands
| | - Pierre Saintigny
- Department of translational Research and Innovation, Centre Léon Bérard, Lyon, France
| | - Patrick Mehlen
- Apoptosis, Cancer and Development Laboratory—Equipe labellisée ‘La Ligue’, LabEx DEVweCAN, Centre de Recherche en Cancérologie de Lyon, INSERM U1052-CNRS UMR5286, Université de Lyon, Centre Léon Bérard, Lyon, France
- Department of translational Research and Innovation, Centre Léon Bérard, Lyon, France
- * E-mail: (PM); (ST)
| | - Servane Tauszig-Delamasure
- Apoptosis, Cancer and Development Laboratory—Equipe labellisée ‘La Ligue’, LabEx DEVweCAN, Centre de Recherche en Cancérologie de Lyon, INSERM U1052-CNRS UMR5286, Université de Lyon, Centre Léon Bérard, Lyon, France
- * E-mail: (PM); (ST)
| |
Collapse
|
22
|
Inamura K. Clinicopathological Characteristics and Mutations Driving Development of Early Lung Adenocarcinoma: Tumor Initiation and Progression. Int J Mol Sci 2018; 19:ijms19041259. [PMID: 29690599 PMCID: PMC5979290 DOI: 10.3390/ijms19041259] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 04/19/2018] [Accepted: 04/20/2018] [Indexed: 01/01/2023] Open
Abstract
Lung cancer is the leading cause of cancer-related deaths worldwide, with lung adenocarcinoma representing the most common lung cancer subtype. Among all lung adenocarcinomas, the most prevalent subset develops via tumorigenesis and progression from atypical adenomatous hyperplasia (AAH) to adenocarcinoma in situ (AIS), to minimally invasive adenocarcinoma (MIA), to overt invasive adenocarcinoma with a lepidic pattern. This stepwise development is supported by the clinicopathological and molecular characteristics of these tumors. In the 2015 World Health Organization classification, AAH and AIS are both defined as preinvasive lesions, whereas MIA is identified as an early invasive adenocarcinoma that is not expected to recur if removed completely. Recent studies have examined the molecular features of lung adenocarcinoma tumorigenesis and progression. EGFR-mutated adenocarcinoma frequently develops via the multistep progression. Oncogene-induced senescence appears to decrease the frequency of the multistep progression in KRAS- or BRAF-mutated adenocarcinoma, whose tumor evolution may be associated with epigenetic alterations and kinase-inactive mutations. This review summarizes the current knowledge of tumorigenesis and tumor progression in early lung adenocarcinoma, with special focus on its clinicopathological characteristics and their associations with driver mutations (EGFR, KRAS, and BRAF) as well as on its molecular pathogenesis and progression.
Collapse
Affiliation(s)
- Kentaro Inamura
- Division of Pathology, The Cancer Institute, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo 135-8550, Japan.
| |
Collapse
|
23
|
RSK2 activity mediates glioblastoma invasiveness and is a potential target for new therapeutics. Oncotarget 2018; 7:79869-79884. [PMID: 27829215 PMCID: PMC5346757 DOI: 10.18632/oncotarget.13084] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Accepted: 10/21/2016] [Indexed: 01/06/2023] Open
Abstract
In glioblastoma (GBM), infiltration of primary tumor cells into the normal tissue and dispersal throughout the brain is a central challenge to successful treatment that remains unmet. Indeed, patients respond poorly to the current therapies of tumor resection followed by chemotherapy with radiotherapy and have only a 16-month median survival. It is therefore imperative to develop novel therapies. RSK2 is a kinase that regulates proliferation and adhesion and can promote metastasis. We demonstrate that active RSK2 regulates GBM cell adhesion and is essential for cell motility and invasion of patient-derived GBM neurospheres. RSK2 control of adhesion and migration is mediated in part by its effects on integrin-Filamin A complexes. Importantly, inhibition of RSK2 by either RSK inhibitors or shRNA silencing impairs invasion and combining RSK2 inhibitors with temozolomide improves efficacy in vitro. In agreement with the in vitro data, using public datasets, we find that RSK2 is significantly upregulated in vivo in human GBM patient tumors, and that high RSK2 expression significantly correlates with advanced tumor stage and poor patient survival. Together, our data provide strong evidence that RSK inhibitors could enhance the effectiveness of existing GBM treatment, and support RSK2 targeting as a promising approach for novel GBM therapy.
Collapse
|
24
|
Hasna J, Hague F, Rodat-Despoix L, Geerts D, Leroy C, Tulasne D, Ouadid-Ahidouch H, Kischel P. Orai3 calcium channel and resistance to chemotherapy in breast cancer cells: the p53 connection. Cell Death Differ 2018; 25:693-707. [PMID: 29323264 DOI: 10.1038/s41418-017-0007-1] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 09/23/2017] [Accepted: 09/25/2017] [Indexed: 02/06/2023] Open
Abstract
Orai proteins are highly selective calcium channels playing an important role in calcium entry. Orai3 channels are overexpressed in breast cancer (BC) tissues, and involved in their proliferation, cell cycle progression and survival. Herein, we sought to address the involvement of Orai3 in resistance to chemotherapeutic drugs. Using high-throughput approaches, we investigated major changes induced by Orai3 overexpression, including downstream signaling mechanisms involved in BC chemotherapy resistance. Resistance was dependent on external calcium presence and thus Orai3 functionality. This effect allowed a downregulation of the p53 tumor suppressor protein expression via the pro-survival PI3K/Sgk-1/Sek-1 pathway. We demonstrated that p53 degradation occurred not only via Mdm2, but also via another unexpected E3 ubiquitin ligase, Nedd4-2. We found supporting bioinformatic evidence linking Orai3 overexpression and chemoresistance in large human BC data sets. Altogether, our results shed light on the molecular mechanisms activated in BC cells commonly found to overexpress Orai3, allowing resistance to chemotherapeutic drugs.
Collapse
Affiliation(s)
- Jessy Hasna
- Université de Picardie Jules Verne, UFR des Sciences, Laboratoire de Physiologie Cellulaire et Moléculaire (EA 4667), SFR CAP-SANTE (FED 4231), Amiens, France
| | - Frédéric Hague
- Université de Picardie Jules Verne, UFR des Sciences, Laboratoire de Physiologie Cellulaire et Moléculaire (EA 4667), SFR CAP-SANTE (FED 4231), Amiens, France
| | - Lise Rodat-Despoix
- Université de Picardie Jules Verne, UFR des Sciences, Laboratoire de Physiologie Cellulaire et Moléculaire (EA 4667), SFR CAP-SANTE (FED 4231), Amiens, France
| | - Dirk Geerts
- Department of Medical Biology L2-109, Academic Medical Center-University of Amsterdam, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands
| | - Catherine Leroy
- Equipe Signalisation, Apoptose et Cancer CNRS UMR8161, Institut de Biologie de Lille - Institut Pasteur, 1 Rue Pr. Calmette, CS50447, 59021, Lille Cedex, France
| | - David Tulasne
- Equipe Signalisation, Apoptose et Cancer CNRS UMR8161, Institut de Biologie de Lille - Institut Pasteur, 1 Rue Pr. Calmette, CS50447, 59021, Lille Cedex, France
| | - Halima Ouadid-Ahidouch
- Université de Picardie Jules Verne, UFR des Sciences, Laboratoire de Physiologie Cellulaire et Moléculaire (EA 4667), SFR CAP-SANTE (FED 4231), Amiens, France.
| | - Philippe Kischel
- Université de Picardie Jules Verne, UFR des Sciences, Laboratoire de Physiologie Cellulaire et Moléculaire (EA 4667), SFR CAP-SANTE (FED 4231), Amiens, France.
| |
Collapse
|
25
|
Faouzi M, Hague F, Geerts D, Ay AS, Potier-Cartereau M, Ahidouch A, Ouadid-Ahidouch H. Functional cooperation between KCa3.1 and TRPC1 channels in human breast cancer: Role in cell proliferation and patient prognosis. Oncotarget 2017; 7:36419-36435. [PMID: 27183905 PMCID: PMC5095010 DOI: 10.18632/oncotarget.9261] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 04/08/2016] [Indexed: 12/24/2022] Open
Abstract
Intracellular Ca2+ levels are important regulators of cell cycle and proliferation. We, and others, have previously reported the role of KCa3.1 (KCNN4) channels in regulating the membrane potential and the Ca2+ entry in association with cell proliferation. However, the relevance of KC3.1 channels in cancer prognosis as well as the molecular mechanism of Ca2+ entry triggered by their activation remain undetermined. Here, we show that RNAi-mediated knockdown of KCa3.1 and/or TRPC1 leads to a significant decrease in cell proliferation due to cell cycle arrest in the G1 phase. These results are consistent with the observed upregulation of both channels in synchronized cells at the end of G1 phase. Additionally, knockdown of TRPC1 suppressed the Ca2+ entry induced by 1-EBIO-mediated KCa3.1 activation, suggesting a functional cooperation between TRPC1 and KCa3.1 in the regulation of Ca2+ entry, possibly within lipid raft microdomains where these two channels seem to co-localize. We also show significant correlations between KCa3.1 mRNA expression and poor patient prognosis and unfavorable clinical breast cancer parameters by mining large datasets in the public domain. Together, these results highlight the importance of KCa3.1 in regulating the proliferative mechanisms in breast cancer cells as well as in providing a promising novel target in prognosis and therapy.
Collapse
Affiliation(s)
- Malika Faouzi
- University of Picardie Jules Verne, UFR of Sciences, EA4667 Laboratory of Cell and Molecular Physiology, SFR CAP-SANTE (FED 4231), Amiens, France.,Queen's Center for Biomedical Research, The Queen's Medical Center, Honolulu, HI 96813, USA
| | - Frederic Hague
- University of Picardie Jules Verne, UFR of Sciences, EA4667 Laboratory of Cell and Molecular Physiology, SFR CAP-SANTE (FED 4231), Amiens, France
| | - Dirk Geerts
- Department of Pediatric Oncology/Hematology, Erasmus University Medical Center, 3015 GE Rotterdam, The Netherlands
| | - Anne-Sophie Ay
- University of Picardie Jules Verne, UFR of Sciences, EA4667 Laboratory of Cell and Molecular Physiology, SFR CAP-SANTE (FED 4231), Amiens, France
| | - Marie Potier-Cartereau
- University of Picardie Jules Verne, UFR of Sciences, EA4667 Laboratory of Cell and Molecular Physiology, SFR CAP-SANTE (FED 4231), Amiens, France.,Inserm, UMR1069, Nutrition, Growth and Cancer, University of François Rabelais, Tours F-37032, France
| | - Ahmed Ahidouch
- University of Picardie Jules Verne, UFR of Sciences, EA4667 Laboratory of Cell and Molecular Physiology, SFR CAP-SANTE (FED 4231), Amiens, France
| | - Halima Ouadid-Ahidouch
- University of Picardie Jules Verne, UFR of Sciences, EA4667 Laboratory of Cell and Molecular Physiology, SFR CAP-SANTE (FED 4231), Amiens, France
| |
Collapse
|
26
|
Tomolonis JA, Agarwal S, Shohet JM. Neuroblastoma pathogenesis: deregulation of embryonic neural crest development. Cell Tissue Res 2017; 372:245-262. [PMID: 29222693 DOI: 10.1007/s00441-017-2747-0] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 11/21/2017] [Indexed: 12/12/2022]
Abstract
Neuroblastoma (NB) is an aggressive pediatric cancer that originates from neural crest tissues of the sympathetic nervous system. NB is highly heterogeneous both from a clinical and a molecular perspective. Clinically, this cancer represents a wide range of phenotypes ranging from spontaneous regression of 4S disease to unremitting treatment-refractory progression and death of high-risk metastatic disease. At a cellular level, the heterogeneous behavior of NB likely arises from an arrest and deregulation of normal neural crest development. In the present review, we summarize our current knowledge of neural crest development as it relates to pathways promoting 'stemness' and how deregulation may contribute to the development of tumor-initiating CSCs. There is an emerging consensus that such tumor subpopulations contribute to the evolution of drug resistance, metastasis and relapse in other equally aggressive malignancies. As relapsed, refractory disease remains the primary cause of death for neuroblastoma, the identification and targeting of CSCs or other primary drivers of tumor progression remains a critical, clinically significant goal for neuroblastoma. We will critically review recent and past evidence in the literature supporting the concept of CSCs as drivers of neuroblastoma pathogenesis.
Collapse
Affiliation(s)
- Julie A Tomolonis
- Department of Pediatrics, Section of Hematology-Oncology, Texas Children's Cancer Center, Houston, TX, 77030, USA.,Medical Scientist Training Program (MSTP), Baylor College of Medicine, Houston, TX, 77030, USA.,Translational Biology & Molecular Medicine (TBMM) Graduate Program, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Saurabh Agarwal
- Department of Pediatrics, Section of Hematology-Oncology, Texas Children's Cancer Center, Houston, TX, 77030, USA.,Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jason M Shohet
- Department of Pediatrics, Section of Hematology-Oncology, Texas Children's Cancer Center, Houston, TX, 77030, USA. .,Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, 77030, USA. .,Neuroblastoma Research Program, Texas Children's Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
27
|
MSX1-Induced Neural Crest-Like Reprogramming Promotes Melanoma Progression. J Invest Dermatol 2017; 138:141-149. [PMID: 28927893 DOI: 10.1016/j.jid.2017.05.038] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 04/30/2017] [Accepted: 05/15/2017] [Indexed: 12/14/2022]
Abstract
Melanoma cells share many biological properties with neural crest stem cells. Here we show that the homeodomain transcription factor MSX1, which is significantly correlated with melanoma disease progression, reprograms melanocytes and melanoma cells toward a neural crest precursor-like state. MSX1-reprogrammed normal human melanocytes express the neural crest marker p75 and become multipotent. MSX1 induces a phenotypic switch in melanoma, which is characterized by an oncogenic transition from an E-cadherin-high nonmigratory state toward a ZEB1-high invasive state. ZEB1 up-regulation is responsible for the MSX1-induced migratory phenotype in melanoma cells. Depletion of MSX1 significantly inhibits melanoma metastasis in vivo. These results show that neural crest-like reprogramming achieved by a single factor is a critical process for melanoma progression.
Collapse
|
28
|
Vaz M, Hwang SY, Kagiampakis I, Phallen J, Patil A, O'Hagan HM, Murphy L, Zahnow CA, Gabrielson E, Velculescu VE, Easwaran HP, Baylin SB. Chronic Cigarette Smoke-Induced Epigenomic Changes Precede Sensitization of Bronchial Epithelial Cells to Single-Step Transformation by KRAS Mutations. Cancer Cell 2017; 32:360-376.e6. [PMID: 28898697 PMCID: PMC5596892 DOI: 10.1016/j.ccell.2017.08.006] [Citation(s) in RCA: 132] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 06/21/2017] [Accepted: 08/11/2017] [Indexed: 12/21/2022]
Abstract
We define how chronic cigarette smoke-induced time-dependent epigenetic alterations can sensitize human bronchial epithelial cells for transformation by a single oncogene. The smoke-induced chromatin changes include initial repressive polycomb marking of genes, later manifesting abnormal DNA methylation by 10 months. At this time, cells exhibit epithelial-to-mesenchymal changes, anchorage-independent growth, and upregulated RAS/MAPK signaling with silencing of hypermethylated genes, which normally inhibit these pathways and are associated with smoking-related non-small cell lung cancer. These cells, in the absence of any driver gene mutations, now transform by introducing a single KRAS mutation and form adenosquamous lung carcinomas in mice. Thus, epigenetic abnormalities may prime for changing oncogene senescence to addiction for a single key oncogene involved in lung cancer initiation.
Collapse
Affiliation(s)
- Michelle Vaz
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Stephen Y Hwang
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Ioannis Kagiampakis
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Jillian Phallen
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Ashwini Patil
- Krieger School of Arts and Sciences, Baltimore, MD 21218, USA
| | - Heather M O'Hagan
- Medical Sciences, Indiana University School of Medicine, Bloomington, IN 47405, USA; Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN 46202, USA
| | - Lauren Murphy
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Cynthia A Zahnow
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Edward Gabrielson
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Victor E Velculescu
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Hariharan P Easwaran
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| | - Stephen B Baylin
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| |
Collapse
|
29
|
Neto M, Naval-Sánchez M, Potier D, Pereira PS, Geerts D, Aerts S, Casares F. Nuclear receptors connect progenitor transcription factors to cell cycle control. Sci Rep 2017; 7:4845. [PMID: 28687780 PMCID: PMC5501803 DOI: 10.1038/s41598-017-04936-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 05/23/2017] [Indexed: 01/31/2023] Open
Abstract
The specification and growth of organs is controlled simultaneously by networks of transcription factors. While the connection between these transcription factors with fate determinants is increasingly clear, how they establish the link with the cell cycle is far less understood. Here we investigate this link in the developing Drosophila eye, where two transcription factors, the MEIS1 homologue hth and the Zn-finger tsh, synergize to stimulate the proliferation of naïve eye progenitors. Experiments combining transcriptomics, open-chromatin profiling, motif analysis and functional assays indicate that these progenitor transcription factors exert a global regulation of the proliferation program. Rather than directly regulating cell cycle genes, they control proliferation through an intermediary layer of nuclear receptors of the ecdysone/estrogen-signaling pathway. This regulatory subnetwork between hth, tsh and nuclear receptors might be conserved from Drosophila to mammals, as we find a significant co-overexpression of their human homologues in specific cancer types.
Collapse
Affiliation(s)
- Marta Neto
- CABD, Andalusian Centre for Developmental Biology, CSIC-UPO-JA, 41013, Seville, Spain.,i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal.,IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
| | | | - Delphine Potier
- School of Medicine, University of Leuven, box 602 3000, Leuven, Belgium
| | - Paulo S Pereira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal.,IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
| | - Dirk Geerts
- Department of Medical Biology L2-109, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Stein Aerts
- School of Medicine, University of Leuven, box 602 3000, Leuven, Belgium.
| | - Fernando Casares
- CABD, Andalusian Centre for Developmental Biology, CSIC-UPO-JA, 41013, Seville, Spain.
| |
Collapse
|
30
|
Abstract
Neuroblastoma is the most common extracranial solid tumor of childhood originating from sympathetic nervous system cells. Neuroblastoma has also been diagnosed in conjunction with other congenital conditions such as Hirschsprung's disease, congenital hypoventilation disorder, and neurofibromatosis type 1. Wolf-Hirschhorn syndrome is a congenital disorder caused by microdeletion of short arm of chromosome 4 encoding MSX1 gene with characteristic facial features. We describe a child with dysmorphic features, developmental delay, mental retardation who developed neuroblastoma at 2 years of age and cytogenetic analysis of blood lymphocytes revealed an interstitial deletion of 4p(15,2). To best our knowledge, this report is the first report of neuroblastoma in a child with Wolf-Hirschhorn syndrome; and the reported association may be an important clue for oncological follow-up of patients with Wolf-Hirschhorn syndrome.
Collapse
|
31
|
Ding K, Liu WY, Zeng Q, Hou F, Xu JZ, Yang Z. Msx1-modulated muscle satellite cells retain a primitive state and exhibit an enhanced capacity for osteogenic differentiation. Exp Cell Res 2017; 352:84-94. [DOI: 10.1016/j.yexcr.2017.01.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 01/03/2017] [Accepted: 01/29/2017] [Indexed: 12/14/2022]
|
32
|
Chung STM, Geerts D, Roseman K, Renaud A, Connelly L. Osteoprotegerin mediates tumor-promoting effects of Interleukin-1beta in breast cancer cells. Mol Cancer 2017; 16:27. [PMID: 28143606 PMCID: PMC5286681 DOI: 10.1186/s12943-017-0606-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 01/25/2017] [Indexed: 12/26/2022] Open
Abstract
Background It is widely recognized that inflammation promotes breast cancer invasion and metastasis. Given the complex nature of the breast tumor inflammatory microenvironment, much remains to be understood of the molecular mechanisms that govern these effects. We have previously shown that osteoprotegerin knockdown in breast cancer cells resulted in reduced invasion and metastasis. Here we present novel insight into the role of osteoprotegerin in inflammation-driven tumor progression in breast cancer by investigating the link between osteoprotegerin, macrophages and the potent pro-inflammatory cytokine Interleukin-1beta. Methods We used human breast cancer cell lines to investigate the effects of Interleukin-1beta treatment on osteoprotegerin secretion as measured by ELISA. We analyzed public datasets containing human breast cancer genome-wide mRNA expression data to reveal a significant and positive correlation between osteoprotegerin mRNA expression and the mRNA expression of Interleukin-1beta and of monocyte chemoattractant protein CC-chemokine ligand 2. Osteoprotegerin, Interleukin-1beta and CC-chemokine ligand 2 mRNA levels were also examined by qPCR on cDNA from normal and cancerous human breast tissue. We determined the effect of Interleukin-1beta–producing macrophages on osteoprotegerin expression by co-culturing breast cancer cells and differentiated THP-1 macrophages. Immunohistochemistry was performed on human breast tumor tissue microarrays to assess macrophage infiltration and osteoprotegerin expression. To demonstrate that osteoprotegerin mediated functional effects of Interleukin-1beta we performed cell invasion studies with control and OPG siRNA knockdown on Interleukin-1beta-treated breast cancer cells. Results We report that Interleukin-1beta induces osteoprotegerin secretion, independent of breast cancer subtype and basal osteoprotegerin levels. Co-culture of breast cancer cells with Interleukin-1beta-secreting macrophages resulted in a similar increase in osteoprotegerin secretion in breast cancer cells as Interleukin-1beta treatment. Macrophage infiltration correlates with osteoprotegerin secretion in human breast tumor tissue samples. We show that osteoprotegerin secretion is regulated by Interleukin-1beta in a p38- and p42/44-dependent manner. We also demonstrate that osteoprotegerin knockdown represses Interleukin-1beta expression, Interleukin-1beta-mediated breast cancer cell invasion and MMP3 expression. Conclusions These data indicate a novel role for osteoprotegerin as a mediator of inflammation- promoted breast cancer progression. Electronic supplementary material The online version of this article (doi:10.1186/s12943-017-0606-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Stephanie Tsang Mui Chung
- Department of Pharmaceutical Sciences, Daniel K. Inouye College of Pharmacy, University of Hawaii at Hilo, Hilo, Hawaii, USA
| | - Dirk Geerts
- Department of Pediatric Oncology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Kim Roseman
- Department of Pharmaceutical Sciences, Daniel K. Inouye College of Pharmacy, University of Hawaii at Hilo, Hilo, Hawaii, USA
| | - Ashleigh Renaud
- Department of Pharmaceutical Sciences, Daniel K. Inouye College of Pharmacy, University of Hawaii at Hilo, Hilo, Hawaii, USA
| | - Linda Connelly
- Department of Pharmaceutical Sciences, Daniel K. Inouye College of Pharmacy, University of Hawaii at Hilo, Hilo, Hawaii, USA.
| |
Collapse
|
33
|
Taminiau A, Draime A, Tys J, Lambert B, Vandeputte J, Nguyen N, Renard P, Geerts D, Rezsöhazy R. HOXA1 binds RBCK1/HOIL-1 and TRAF2 and modulates the TNF/NF-κB pathway in a transcription-independent manner. Nucleic Acids Res 2016; 44:7331-49. [PMID: 27382069 PMCID: PMC5009750 DOI: 10.1093/nar/gkw606] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 06/24/2016] [Indexed: 11/14/2022] Open
Abstract
HOX proteins define a family of key transcription factors regulating animal embryogenesis. HOX genes have also been linked to oncogenesis and HOXA1 has been described to be active in several cancers, including breast cancer. Through a proteome-wide interaction screening, we previously identified the TNFR-associated proteins RBCK1/HOIL-1 and TRAF2 as HOXA1 interactors suggesting that HOXA1 is functionally linked to the TNF/NF-κB signaling pathway. Here, we reveal a strong positive correlation between expression of HOXA1 and of members of the TNF/NF-κB pathway in breast tumor datasets. Functionally, we demonstrate that HOXA1 can activate NF-κB and operates upstream of the NF-κB inhibitor IκB. Consistently, we next demonstrate that the HOXA1-mediated activation of NF-κB is non-transcriptional and that RBCK1 and TRAF2 influences on NF-κB are epistatic to HOXA1. We also identify an 11 Histidine repeat and the homeodomain of HOXA1 to be required both for RBCK1 and TRAF2 interaction and NF-κB stimulation. Finally, we highlight that activation of NF-κB is crucial for HOXA1 oncogenic activity.
Collapse
Affiliation(s)
- Arnaud Taminiau
- Animal Molecular and Cellular Biology Group (AMCB), Life Sciences Institute (ISV), Université catholique de Louvain, Louvain-la-Neuve 1348, Belgium
| | - Amandine Draime
- Animal Molecular and Cellular Biology Group (AMCB), Life Sciences Institute (ISV), Université catholique de Louvain, Louvain-la-Neuve 1348, Belgium
| | - Janne Tys
- Animal Molecular and Cellular Biology Group (AMCB), Life Sciences Institute (ISV), Université catholique de Louvain, Louvain-la-Neuve 1348, Belgium
| | - Barbara Lambert
- Animal Molecular and Cellular Biology Group (AMCB), Life Sciences Institute (ISV), Université catholique de Louvain, Louvain-la-Neuve 1348, Belgium
| | - Julie Vandeputte
- Animal Molecular and Cellular Biology Group (AMCB), Life Sciences Institute (ISV), Université catholique de Louvain, Louvain-la-Neuve 1348, Belgium
| | - Nathan Nguyen
- Animal Molecular and Cellular Biology Group (AMCB), Life Sciences Institute (ISV), Université catholique de Louvain, Louvain-la-Neuve 1348, Belgium
| | - Patricia Renard
- Cellular Biology Research Unit, Université de Namur, Namur 5000, Belgium
| | - Dirk Geerts
- Department of Pediatric Oncology/Hematology, Erasmus University Medical Center, Rotterdam 3015, The Netherlands
| | - René Rezsöhazy
- Animal Molecular and Cellular Biology Group (AMCB), Life Sciences Institute (ISV), Université catholique de Louvain, Louvain-la-Neuve 1348, Belgium
| |
Collapse
|
34
|
Chen LT, Hu MM, Xu ZS, Liu Y, Shu HB. MSX1 Modulates RLR-Mediated Innate Antiviral Signaling by Facilitating Assembly of TBK1-Associated Complexes. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2016; 197:199-207. [PMID: 27194789 DOI: 10.4049/jimmunol.1600039] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 04/22/2016] [Indexed: 12/16/2023]
Abstract
Recognition of viral dsRNA by the retinoic acid-inducible gene-1-like receptors (RLRs) triggers signaling cascades that lead to activation of the TBK1 kinase and transcription factor IFN regulatory factor 3, induction of downstream antiviral genes, and innate antiviral responses. In this study, we identified muscle segment homeobox1 (MSX1) as an important modulator of RLR-mediated signaling pathways. Knockdown or knockout of MSX1 significantly impaired Sendai virus-triggered activation of TBK1 and IFN regulatory factor 3, induction of downstream antiviral genes, and cellular antiviral responses. Interestingly, MSX1 was translocated from the nucleus to cytoplasm, particularly mitochondria upon infection of Sendai virus. Biochemcially, MSX1 was important for assembly of TBK1/IKK-related kinase-associated protein 1/TNFR-associated factor-associated NF-κB activator complexes. Our results suggest that MSX1 is an important component of RLR-mediated signaling and reveal mechanisms on innate immune responses against RNA viruses.
Collapse
Affiliation(s)
- Liu-Ting Chen
- College of Life Sciences, Medical Research Institute, Collaborative Innovation Center for Viral Immunology, Wuhan University, Wuhan 430072, China
| | - Ming-Ming Hu
- College of Life Sciences, Medical Research Institute, Collaborative Innovation Center for Viral Immunology, Wuhan University, Wuhan 430072, China
| | - Zhi-Sheng Xu
- College of Life Sciences, Medical Research Institute, Collaborative Innovation Center for Viral Immunology, Wuhan University, Wuhan 430072, China
| | - Yu Liu
- College of Life Sciences, Medical Research Institute, Collaborative Innovation Center for Viral Immunology, Wuhan University, Wuhan 430072, China
| | - Hong-Bing Shu
- College of Life Sciences, Medical Research Institute, Collaborative Innovation Center for Viral Immunology, Wuhan University, Wuhan 430072, China
| |
Collapse
|
35
|
Pannella M, Caliceti C, Fortini F, Aquila G, Vieceli Dalla Sega F, Pannuti A, Fortini C, Morelli MB, Fucili A, Francolini G, Voltan R, Secchiero P, Dinelli G, Leoncini E, Ferracin M, Hrelia S, Miele L, Rizzo P. Serum From Advanced Heart Failure Patients Promotes Angiogenic Sprouting and Affects the Notch Pathway in Human Endothelial Cells. J Cell Physiol 2016; 231:2700-10. [PMID: 26987674 DOI: 10.1002/jcp.25373] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 03/10/2016] [Indexed: 12/21/2022]
Abstract
It is unknown whether components present in heart failure (HF) patients' serum provide an angiogenic stimulus. We sought to determine whether serum from HF patients affects angiogenesis and its major modulator, the Notch pathway, in human umbilical vein endothelial cells (HUVECs). In cells treated with serum from healthy subjects or from patients at different HF stage we determined: (1) Sprouting angiogenesis, by measuring cells network (closed tubes) in collagen gel. (2) Protein levels of Notch receptors 1, 2, 4, and ligands Jagged1, Delta-like4. We found a higher number of closed tubes in HUVECs treated with advanced HF patients serum in comparison with cells treated with serum from mild HF patients or controls. Furthermore, as indicated by the reduction of the active form of Notch4 (N4IC) and of Jagged1, advanced HF patients serum inhibited Notch signalling in HUVECs in comparison with mild HF patients' serum and controls. The circulating levels of NT-proBNP (N-terminal of the pro-hormone brain natriuretic peptide), a marker for the detection and evalutation of HF, were positively correlated with the number of closed tubes (r = 0.485) and negatively with Notch4IC and Jagged1 levels in sera-treated cells (r = -0.526 and r = -0.604, respectively). In conclusion, we found that sera from advanced HF patients promote sprouting angiogenesis and dysregulate Notch signaling in HUVECs. Our study provides in vitro evidence of an angiogenic stimulus arising during HF progression and suggests a role for the Notch pathway in it. J. Cell. Physiol. 231: 2700-2710, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Micaela Pannella
- Goldyne Savad Institute of Gene Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Cristiana Caliceti
- Department of Chemistry "G. Ciamician", University of Bologna, Bologna, Italy
| | - Francesca Fortini
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Giorgio Aquila
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | | | - Antonio Pannuti
- Stanley Scott Cancer Center, Louisiana State University Health Sciences Center and Louisiana Cancer Research Consortium, New Orleans, Louisiana
| | - Cinzia Fortini
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | | | - Alessandro Fucili
- University Hospital of Ferrara, Ferrara, Italy.,Maria Cecilia Hospital, GVM Care & Research, E.S. Health Science Foundation, Cotignola, Italy
| | - Gloria Francolini
- Cardiovascular Research Center, Salvatore Maugeri Foundation IRCCS, Lumezzane, Italy
| | - Rebecca Voltan
- Department of Morphology, Surgery, and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Paola Secchiero
- Department of Morphology, Surgery, and Experimental Medicine, University of Ferrara, Ferrara, Italy.,Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Giovanni Dinelli
- Department of Agricultural Sciences, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Emanuela Leoncini
- Department for Life Quality Studies, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Manuela Ferracin
- Department of Experimental, Diagnostic and Specialty Medicine-DIMES, University of Bologna, Bologna, Italy
| | - Silvana Hrelia
- Department for Life Quality Studies, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Lucio Miele
- Stanley Scott Cancer Center, Louisiana State University Health Sciences Center and Louisiana Cancer Research Consortium, New Orleans, Louisiana
| | - Paola Rizzo
- Department of Morphology, Surgery, and Experimental Medicine, University of Ferrara, Ferrara, Italy
| |
Collapse
|
36
|
Middelbeek J, Visser D, Henneman L, Kamermans A, Kuipers AJ, Hoogerbrugge PM, Jalink K, van Leeuwen FN. TRPM7 maintains progenitor-like features of neuroblastoma cells: implications for metastasis formation. Oncotarget 2016; 6:8760-76. [PMID: 25797249 PMCID: PMC4496182 DOI: 10.18632/oncotarget.3315] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 02/08/2015] [Indexed: 12/18/2022] Open
Abstract
Neuroblastoma is an embryonal tumor derived from poorly differentiated neural crest cells. Current research is aimed at identifying the molecular mechanisms that maintain the progenitor state of neuroblastoma cells and to develop novel therapeutic strategies that induce neuroblastoma cell differentiation. Mechanisms controlling neural crest development are typically dysregulated during neuroblastoma progression, and provide an appealing starting point for drug target discovery. Transcriptional programs involved in neural crest development act as a context dependent gene regulatory network. In addition to BMP, Wnt and Notch signaling, activation of developmental gene expression programs depends on the physical characteristics of the tissue microenvironment. TRPM7, a mechanically regulated TRP channel with kinase activity, was previously found essential for embryogenesis and the maintenance of undifferentiated neural crest progenitors. Hence, we hypothesized that TRPM7 may preserve progenitor-like, metastatic features of neuroblastoma cells. Using multiple neuroblastoma cell models, we demonstrate that TRPM7 expression closely associates with the migratory and metastatic properties of neuroblastoma cells in vitro and in vivo. Moreover, microarray-based expression profiling on control and TRPM7 shRNA transduced neuroblastoma cells indicates that TRPM7 controls a developmental transcriptional program involving the transcription factor SNAI2. Overall, our data indicate that TRPM7 contributes to neuroblastoma progression by maintaining progenitor-like features.
Collapse
Affiliation(s)
- Jeroen Middelbeek
- Laboratory of Pediatric Oncology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands
| | - Daan Visser
- Division of Cell Biology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Linda Henneman
- Division of Cell Biology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Alwin Kamermans
- Laboratory of Pediatric Oncology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands
| | - Arthur J Kuipers
- Laboratory of Pediatric Oncology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands
| | - Peter M Hoogerbrugge
- Laboratory of Pediatric Oncology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands.,Princes Maxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Kees Jalink
- Division of Cell Biology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Frank N van Leeuwen
- Laboratory of Pediatric Oncology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands
| |
Collapse
|
37
|
Yco LP, Geerts D, Mocz G, Koster J, Bachmann AS. Effect of sulfasalazine on human neuroblastoma: analysis of sepiapterin reductase (SPR) as a new therapeutic target. BMC Cancer 2015; 15:477. [PMID: 26093909 PMCID: PMC4475614 DOI: 10.1186/s12885-015-1447-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 05/19/2015] [Indexed: 12/21/2022] Open
Abstract
Background Neuroblastoma (NB) is an aggressive childhood malignancy in children up to 5 years of age. High-stage tumors frequently relapse even after aggressive multimodal treatment, and then show therapy resistance, typically resulting in patient death. New molecular-targeted compounds that effectively suppress tumor growth and prevent relapse with more efficacy are urgently needed. We and others previously showed that polyamines (PA) like spermidine and spermine are essential for NB tumorigenesis and that DFMO, an inhibitor of the key PA synthesis gene product ODC, is effective both in vitro and in vivo, securing its evaluation in NB clinical trials. To find additional compounds interfering with PA biosynthesis, we tested sulfasalazine (SSZ), an FDA-approved salicylate-based anti-inflammatory and immune-modulatory drug, recently identified to inhibit sepiapterin reductase (SPR). We earlier presented evidence for a physical interaction between ODC and SPR and we showed that RNAi-mediated knockdown of SPR expression significantly reduced native ODC enzyme activity and impeded NB cell proliferation. Methods Human NB mRNA expression datasets in the public domain were analyzed using the R2 platform. Cell viability, isobologram, and combination index analyses as a result of SSZ treatment with our without DFMO were carried out in NB cell cultures. Molecular protein-ligand docking was achieved using the GRAMM algorithm. Statistical analyses were performed with the Kruskal-Wallis test, 2log Pearson test, and Student’s t test. Results In this study, we show the clinical relevance of SPR in human NB tumors. We found that high SPR expression is significantly correlated to unfavorable NB characteristics like high age at diagnosis, MYCN amplification, and high INSS stage. SSZ inhibits the growth of NB cells in vitro, presumably due to the inhibition of SPR as predicted by computational docking of SSZ into SPR. Importantly, the combination of SSZ with DFMO produces synergistic antiproliferative effects in vitro. Conclusions The results suggest the use of SSZ in combination with DFMO for further experiments, and possible prioritization as a novel therapy for the treatment of NB patients.
Collapse
Affiliation(s)
- Lisette P Yco
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, 301 Michigan Street, NE, Grand Rapids, MI, 49503, USA. .,Department of Pharmaceutical Sciences, The Daniel K. Inouye College of Pharmacy, University of Hawaii at Hilo, Hilo, HI, 96720, USA. .,Department of Molecular Biosciences and Bioengineering, College of Tropical Agriculture and Human Resources, University of Hawaii at Manoa, Honolulu, HI, 96822, USA.
| | - Dirk Geerts
- Department of Pediatric Oncology/Hematology, Sophia Children's Hospital, Erasmus University Medical Center, Rotterdam, GE, 3015, The Netherlands.
| | - Gabor Mocz
- Pacific Biosciences Research Center, University of Hawaii at Manoa, Honolulu, HI, 96822, USA.
| | - Jan Koster
- Department of Oncogenomics, Academic Medical Center, University of Amsterdam, Amsterdam, AZ, 1105, The Netherlands.
| | - André S Bachmann
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, 301 Michigan Street, NE, Grand Rapids, MI, 49503, USA. .,Department of Pharmaceutical Sciences, The Daniel K. Inouye College of Pharmacy, University of Hawaii at Hilo, Hilo, HI, 96720, USA. .,Department of Molecular Biosciences and Bioengineering, College of Tropical Agriculture and Human Resources, University of Hawaii at Manoa, Honolulu, HI, 96822, USA.
| |
Collapse
|
38
|
Bandino A, Geerts D, Koster J, Bachmann AS. Deoxyhypusine synthase (DHPS) inhibitor GC7 induces p21/Rb-mediated inhibition of tumor cell growth and DHPS expression correlates with poor prognosis in neuroblastoma patients. Cell Oncol (Dordr) 2014; 37:387-98. [PMID: 25315710 DOI: 10.1007/s13402-014-0201-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/22/2014] [Indexed: 11/29/2022] Open
Abstract
PURPOSE Neuroblastoma (NB) is an aggressive pediatric malignancy that typically occurs in infants and children under the age of 5 years. High-stage tumors relapse frequently even after aggressive multimodal treatment, resulting in therapy resistance and eventually in patient death. Clearly, new biologically-targeted drugs are needed that more efficiently suppress tumor growth and prevent relapse. We and others previously showed that polyamines such as spermidine play an essential role in NB tumorigenesis and that DFMO, an inhibitor of the central polyamine synthesis gene ODC, is effective in vitro and in vivo, prompting its evaluation in NB clinical trials. However, the specific molecular actions of polyamines remain poorly defined. Spermidine and deoxyhypusine synthase (DHPS) are essential components in the hypusination-driven post-translational activation of eukaryotic initiation factor 5A (eIF5A). METHODS We assessed the role of DHPS in NB and the impact of its inhibition by N(1)-guanyl-1,7-diaminoheptane (GC7) on tumor cell growth using cell proliferation assays, Western blot, immunofluorescence microscopy, and Affymetrix micro-array mRNA expression analyses in NB tumor samples. RESULTS We found that GC7 inhibits NB cell proliferation in a dose-dependent manner, through induction of the cell cycle inhibitor p21 and reduction of total and phosphorylated Rb proteins. Strikingly, high DHPS mRNA expression correlated significantly with unfavorable clinical parameters, including poor patient survival, in a cohort of 88 NB tumors (all P < 0.04). CONCLUSIONS These results suggest that spermidine and DHPS are key contributing factors in NB tumor proliferation through regulation of the p21/Rb signaling axis.
Collapse
Affiliation(s)
- Andrea Bandino
- Department of Pharmaceutical Sciences, The Daniel K. Inouye College of Pharmacy, University of Hawaii at Hilo, 34 Rainbow Drive, Hilo, HI, 96720, USA
| | | | | | | |
Collapse
|
39
|
Weichhaus M, Segaran P, Renaud A, Geerts D, Connelly L. Osteoprotegerin expression in triple-negative breast cancer cells promotes metastasis. Cancer Med 2014; 3:1112-25. [PMID: 24976340 PMCID: PMC4302663 DOI: 10.1002/cam4.277] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Revised: 04/29/2014] [Accepted: 05/01/2014] [Indexed: 01/12/2023] Open
Abstract
Osteoprotegerin (OPG) is a secreted member of the tumor necrosis factor (TNF) receptor superfamily that has been well characterized as a negative regulator of bone remodeling. OPG is also expressed in human breast cancer tissues and cell lines. In vitro studies suggest that OPG exerts tumor-promoting effects by binding to TNF-related apoptosis inducing ligand (TRAIL), thereby preventing induction of apoptosis. However, the in vivo effect of OPG expression by primary breast tumors has not been characterized. We knocked down OPG expression in MDA-MB-231 and MDA-MB-436 human breast cancer cells using shRNA and siRNA to investigate impact on metastasis in the chick embryo model. We observed a reduction in metastasis with OPG knockdown cells. We found that lowering OPG expression did not alter sensitivity to TRAIL-induced apoptosis; however, the OPG knockdown cells had a reduced level of invasion. In association with this we observed reduced expression of the proteases Cathepsin D and Matrix Metalloproteinase-2 upon OPG knockdown, indicating that OPG may promote metastasis via modulation of protease expression and invasion. We conclude that OPG has a metastasis-promoting effect in breast cancer cells.
Collapse
Affiliation(s)
- Michael Weichhaus
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Hawaii at Hilo, Hilo, Hawaii
| | | | | | | | | |
Collapse
|
40
|
Xu B, Geerts D, Bu Z, Ai J, Jin L, Li Y, Zhang H, Zhu G. Regulation of endometrial receptivity by the highly expressed HOXA9, HOXA11 and HOXD10 HOX-class homeobox genes. Hum Reprod 2014; 29:781-90. [PMID: 24549215 DOI: 10.1093/humrep/deu004] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
STUDY QUESTION Are other HOX genes, in addition to HOXA10, involved in endometrial receptivity? SUMMARY ANSWER The highly expressed HOXA9, HOXA11 and HOXD10 genes also appear to be involved in endometrial receptivity. WHAT IS KNOWN ALREADY Within the HOX family of homeobox transcription factor genes are the leading candidates for the regulation of embryonic implantation. A crucial role of HOXA10 in endometrial receptivity has been well established. STUDY DESIGN, SIZE, DURATION To identify HOX candidate genes, we performed data mining on all 39 human HOX genes in the 'Human body index' gene expression database of normal human tissue. The temporal and spatial expression pattern of four highly expressed HOX genes in the human endometrium was determined. To further investigate the function of these Hox genes, we used a robust in vivo mouse model in which we blocked maternal Hox gene expression. PARTICIPANTS/MATERIALS, SETTING AND METHODS Analysis of a gene expression profile set in the public domain consisting of 504 samples representing 95 different normal human tissues, showed that in addition to HOXA10, also HOXA9, HOXA11, HOXB6 and HOXD10 mRNA showed increased expression in the human endometrium (16 samples). The temporal and spatial expression pattern of these four HOX genes throughout the menstrual cycle was determined in the endometrium from 27 female patients eligible for IVF-embryo transfer with a normal cycle by quantitative real-time PCR (qRT-PCR), western blot and immunohistochemistry. The role of maternal Hoxa9, Hoxa11 and Hoxd10 was assessed in a mouse implantation model by expression knockdown using RNA interference. Forty mice were transfected with Hoxa9-, Hoxa11- or Hoxd10-specific small hairpin RNA (shRNA) constructs or a vector control by injection into the uterine horn at Day 2 after vaginal plug detection (Day 1) (160 mice in total). The effects were examined by qRT-PCR and western blot at Day 4 and litter sizes counted at Day 9 of pregnancy. MAIN RESULTS AND THE ROLE OF CHANCE HOXA10, HOXA9, HOXA11 and HOXD10 all showed increased expression during the mid-secretory phase of the menstrual cycle (P < 0.01). Knockdown of Hoxa9, Hoxa11 and Hoxd10 in the murine uterus resulted in significantly reduced average implantation rates (P < 0.01) and, with regard to four Hox target genes, also correlated with a significantly increased empty spiracles homolog 2 (Emx2) and insulin-like growth factor binding protein-1 (Igfbp1), and decreased integrin β3 (Itgb3) and leukemia inhibitory factor (Lif), expression (P < 0.01). LIMITATIONS, REASONS FOR CAUTION Menstrual cycle stage was not confirmed by serum hormone analysis. We verified the absence of significant differences in stage-specific expression of the reference genes used in our study (ACTB/Actb and GAPDH/Gapdh) and therefore possible limitations of this approach were minimized. In addition, the translatability of our data from a mouse model to patients needs to be investigated further. WIDER IMPLICATIONS OF THE FINDINGS We provide evidence that three other HOX genes in addition to HOXA10 are involved in endometrial receptivity, and that part of their function is asserted through several known HOX target genes, suggesting the presence of a central HOX signal transduction pathway.
Collapse
Affiliation(s)
- Bei Xu
- Reproductive Medicine Center, Tongji Hospital, Tongji Medicine College, Huazhong University of Science and Technology, 1095 JieFang Avenue, Wuhan 430030, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Lange I, Geerts D, Feith DJ, Mocz G, Koster J, Bachmann AS. Novel interaction of ornithine decarboxylase with sepiapterin reductase regulates neuroblastoma cell proliferation. J Mol Biol 2013; 426:332-46. [PMID: 24096079 DOI: 10.1016/j.jmb.2013.09.037] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Revised: 09/21/2013] [Accepted: 09/25/2013] [Indexed: 01/24/2023]
Abstract
Ornithine decarboxylase (ODC) is the sentinel enzyme in polyamine biosynthesis. Both ODC and polyamines regulate cell division, proliferation, and apoptosis. Sepiapterin reductase (SPR) catalyzes the last step in the biosynthesis of tetrahydrobiopterin (BH4), an essential cofactor of nitric oxide synthase, and has been implicated in neurological diseases but not yet in cancer. In this study, we present compelling evidence that native ODC and SPR physically interact, and we defined the individual amino acid residues involved in both enzymes using in silico protein-protein docking simulations. The resulting heterocomplex is a surprisingly compact structure, featuring two energetically and structurally equivalent binding modes both in monomer and in dimer conformations. The novel interaction between ODC and SPR proteins was confirmed under physiological conditions by co-immunoprecipitation and co-localization in neuroblastoma (NB) cells. Importantly, we showed that siRNA (small interfering RNA)-mediated knockdown of SPR expression significantly reduced endogenous ODC enzyme activity in NB cells, thus demonstrating the biological relevance of the ODC-SPR interaction. Finally, in a cohort of 88 human NB tumors, we found that high SPR mRNA expression correlated significantly with poor survival prognosis using a Kaplan-Meier analysis (log-rank test, P=5 × 10(-4)), suggesting an oncogenic role for SPR in NB tumorigenesis. In conclusion, we showed that ODC binds SPR and thus propose a new concept in which two well-characterized biochemical pathways converge via the interaction of two enzymes. We identified SPR as a novel regulator of ODC enzyme activity and, based on clinical evidence, present a model in which SPR drives ODC-mediated malignant progression in NB.
Collapse
Affiliation(s)
- Ingo Lange
- Department of Pharmaceutical Sciences, The Daniel K. Inouye College of Pharmacy, University of Hawaii at Hilo, Hilo, HI 96720, USA
| | - Dirk Geerts
- Department of Pediatric Oncology/Hematology, Sophia Children's Hospital, Erasmus University Medical Center, 3015 GE Rotterdam, The Netherlands
| | - David J Feith
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Gabor Mocz
- Pacific Biosciences Research Center, University of Hawaii at Manoa, Honolulu, HI 96822, USA
| | - Jan Koster
- Department of Oncogenomics, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - André S Bachmann
- Department of Pharmaceutical Sciences, The Daniel K. Inouye College of Pharmacy, University of Hawaii at Hilo, Hilo, HI 96720, USA; Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96813, USA.
| |
Collapse
|
42
|
Wilzén A, Krona C, Sveinbjörnsson B, Kristiansson E, Dalevi D, Øra I, De Preter K, Stallings RL, Maris J, Versteeg R, Nilsson S, Kogner P, Abel F. ERBB3 is a marker of a ganglioneuroblastoma/ganglioneuroma-like expression profile in neuroblastic tumours. Mol Cancer 2013; 12:70. [PMID: 23835063 PMCID: PMC3766266 DOI: 10.1186/1476-4598-12-70] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Accepted: 06/25/2013] [Indexed: 12/21/2022] Open
Abstract
Background Neuroblastoma (NB) tumours are commonly divided into three cytogenetic subgroups. However, by unsupervised principal components analysis of gene expression profiles we recently identified four distinct subgroups, r1-r4. In the current study we characterized these different subgroups in more detail, with a specific focus on the fourth divergent tumour subgroup (r4). Methods Expression microarray data from four international studies corresponding to 148 neuroblastic tumour cases were subject to division into four expression subgroups using a previously described 6-gene signature. Differentially expressed genes between groups were identified using Significance Analysis of Microarray (SAM). Next, gene expression network modelling was performed to map signalling pathways and cellular processes representing each subgroup. Findings were validated at the protein level by immunohistochemistry and immunoblot analyses. Results We identified several significantly up-regulated genes in the r4 subgroup of which the tyrosine kinase receptor ERBB3 was most prominent (fold change: 132–240). By gene set enrichment analysis (GSEA) the constructed gene network of ERBB3 (n = 38 network partners) was significantly enriched in the r4 subgroup in all four independent data sets. ERBB3 was also positively correlated to the ErbB family members EGFR and ERBB2 in all data sets, and a concurrent overexpression was seen in the r4 subgroup. Further studies of histopathology categories using a fifth data set of 110 neuroblastic tumours, showed a striking similarity between the expression profile of r4 to ganglioneuroblastoma (GNB) and ganglioneuroma (GN) tumours. In contrast, the NB histopathological subtype was dominated by mitotic regulating genes, characterizing unfavourable NB subgroups in particular. The high ErbB3 expression in GN tumour types was verified at the protein level, and showed mainly expression in the mature ganglion cells. Conclusions Conclusively, this study demonstrates the importance of performing unsupervised clustering and subtype discovery of data sets prior to analyses to avoid a mixture of tumour subtypes, which may otherwise give distorted results and lead to incorrect conclusions. The current study identifies ERBB3 as a clear-cut marker of a GNB/GN-like expression profile, and we suggest a 7-gene expression signature (including ERBB3) as a complement to histopathology analysis of neuroblastic tumours. Further studies of ErbB3 and other ErbB family members and their role in neuroblastic differentiation and pathogenesis are warranted.
Collapse
Affiliation(s)
- Annica Wilzén
- Department of Clinical Genetics, Institution of Biomedicine, Box 413, S- 405 30, Gothenburg University, Gothenburg, Sweden.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Kaaij LTJ, van de Wetering M, Fang F, Decato B, Molaro A, van de Werken HJG, van Es JH, Schuijers J, de Wit E, de Laat W, Hannon GJ, Clevers HC, Smith AD, Ketting RF. DNA methylation dynamics during intestinal stem cell differentiation reveals enhancers driving gene expression in the villus. Genome Biol 2013; 14:R50. [PMID: 23714178 PMCID: PMC4053812 DOI: 10.1186/gb-2013-14-5-r50] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Accepted: 05/28/2013] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND DNA methylation is of pivotal importance during development. Previous genome-wide studies identified numerous differentially methylated regions upon differentiation of stem cells, many of them associated with transcriptional start sites. RESULTS We present the first genome-wide, single-base-resolution view into DNA methylation dynamics during differentiation of a mammalian epithelial stem cell: the mouse small intestinal Lgr5+ stem cell. Very little change was observed at transcriptional start sites and our data suggest that differentiation-related genes are already primed for expression in the stem cell. Genome-wide, only 50 differentially methylated regions were identified. Almost all of these loci represent enhancers driving gene expression in the differentiated part of the small intestine. Finally, we show that binding of the transcription factor Tcf4 correlates with hypo-methylation and demonstrate that Tcf4 is one of the factors contributing to formation of differentially methylated regions. CONCLUSIONS Our results reveal limited DNA methylation dynamics during small intestine stem cell differentiation and an impact of transcription factor binding on shaping the DNA methylation landscape during differentiation of stem cells in vivo.
Collapse
|
44
|
Abadie C, Lechaix B, Gandemer V, Bonnaure-Mallet M. Neuroblastoma and tooth abnormalities: a common history? Oral Oncol 2013; 49:e11-3. [PMID: 23352669 DOI: 10.1016/j.oraloncology.2012.12.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Revised: 12/20/2012] [Accepted: 12/23/2012] [Indexed: 10/27/2022]
|
45
|
Koomoa DLT, Geerts D, Lange I, Koster J, Pegg AE, Feith DJ, Bachmann AS. DFMO/eflornithine inhibits migration and invasion downstream of MYCN and involves p27Kip1 activity in neuroblastoma. Int J Oncol 2013; 42:1219-28. [PMID: 23440295 PMCID: PMC3622674 DOI: 10.3892/ijo.2013.1835] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Accepted: 09/03/2012] [Indexed: 01/02/2023] Open
Abstract
Neuroblastoma (NB) is the most common extracranial pediatric tumor. NB patients over 18 months of age at the time of diagnosis are often in the later stages of the disease, present with widespread dissemination, and often possess MYCN tumor gene amplification. MYCN is a transcription factor that regulates the expression of a number of genes including ornithine decarboxylase (ODC), a rate-limiting enzyme in the biosynthesis of polyamines. Inhibiting ODC in NB cells produces many deleterious effects including G1 cell cycle arrest, inhibition of cell proliferation, and decreased tumor growth, making ODC a promising target for drug interference. DFMO treatment leads to the accumulation of the cyclin-dependent kinase inhibitor p27Kip1 protein and causes p27Kip1/Rb-coupled G1 cell cycle arrest in MYCN-amplified NB tumor cells through a process that involves p27Kip1 phosphorylation at residues Ser10 and Thr198. While p27Kip1 is well known for its role as a cyclin-dependent kinase inhibitor, recent studies have revealed a novel function of p27Kip1 as a regulator of cell migration and invasion. In the present study we found that p27Kip1 regulates the migration and invasion in NB and that these events are dependent on the state of phosphorylation of p27Kip1. DFMO treatments induced MYCN protein downregulation and phosphorylation of Akt/PKB (Ser473) and GSK3-β (Ser9), and polyamine supplementation alleviated the DFMO-induced effects. Importantly, we provide strong evidence that p27Kip1 mRNA correlates with clinical features and the survival probability of NB patients.
Collapse
Affiliation(s)
- Dana-Lynn T Koomoa
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Hawaii at Hilo, Hilo, HI 96720, USA
| | | | | | | | | | | | | |
Collapse
|
46
|
PRAF2 stimulates cell proliferation and migration and predicts poor prognosis in neuroblastoma. Int J Oncol 2013; 42:1408-16. [PMID: 23440329 DOI: 10.3892/ijo.2013.1836] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2012] [Accepted: 09/07/2012] [Indexed: 11/05/2022] Open
Abstract
Prenylated Rab acceptor 1 domain family, member 2 (PRAF2) is a novel 19-kDa protein with four transmembrane-spanning domains that belongs to the PRAF protein family. Neuroblastoma (NB) is the most common malignant extracranial solid tumor of childhood that originates in primitive cells of the developing sympathetic nervous system. We investigated the correlation of PRAF2 mRNA expression to NB clinical and genetic parameters using Affymetrix expression analysis of a series of 88 NB tumors and examined the functional role of PRAF2 in an NB cell line using RNA interference. We found that high PRAF2 expression is significantly correlated to several unfavorable NB characteristics: MYCN amplification, high age at diagnosis, poor outcome and high INSS stage. The shRNA-mediated PRAF2 downregulation in the SK-N-SH NB cell line resulted in decreased cellular proliferation, migration and matrix-attachment. These findings were confirmed in NB patient tumor samples, where high PRAF2 expression is significantly correlated to bone and bone marrow metastasis, the main cause of death in NB patients. The present study shows that PRAF2 plays an essential role in NB tumorigenesis and metastasis.
Collapse
|
47
|
DNAJB6 governs a novel regulatory loop determining Wnt/β-catenin signalling activity. Biochem J 2012; 444:573-80. [PMID: 22455953 DOI: 10.1042/bj20120205] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
DKK1 (dickkopf 1 homologue) is a secreted inhibitor of the Wnt signalling pathway and a critical modulator of tumour promotion and the tumour microenvironment. However, mechanisms regulating DKK1 expression are understudied. DNAJB6 {DnaJ [HSP40 (heat-shock protein 40 kDa)] homologue, subfamily B, member 6} is an HSP40 family member whose expression is compromised during progression of breast cancer and melanoma. Inhibition of the Wnt/β-catenin signalling pathway by up-regulation of DKK1 is one of the key mechanisms by which DNAJB6 suppresses tumour metastasis and EMT (epithelial-mesenchymal transition). Analysis of the DKK1 promoter to define the cis-site responsible for its up-regulation by DNAJB6 revealed the presence of two binding sites for a transcriptional repressor, MSX1 (muscle segment homeobox 1). Our investigations showed that MSX1 binds the DKK1 promoter and inhibits DKK1 transcription. Interestingly, silencing DNAJB6 resulted in up-regulation of MSX1 concomitant with increased stabilization of β-catenin. ChIP (chromatin immunoprecipitation) studies revealed that β-catenin binds the MSX1 promoter and stabilization of β-catenin elevates MSX1 transcription, indicating that β-catenin works as a transcription co-activator for MSX1. Functionally, exogenous expression of MSX1 in DNAJB6-expressing cells promotes the mesenchymal phenotype by suppression of DKK1. Thus we have identified a novel regulatory mechanism of DNAJB6-mediated DKK1 transcriptional up-regulation that can influence EMT. DKK1 is a feedback regulator of β-catenin levels and thus our studies also define an additional negative control of this β-catenin/DKK1 feedback loop by MSX1, which may potentially contribute to excessive stabilization of β-catenin.
Collapse
|
48
|
Sulzmaier FJ, Valmiki MKG, Nelson DA, Caliva MJ, Geerts D, Matter ML, White EP, Ramos JW. PEA-15 potentiates H-Ras-mediated epithelial cell transformation through phospholipase D. Oncogene 2012; 31:3547-60. [PMID: 22105357 PMCID: PMC3295902 DOI: 10.1038/onc.2011.514] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2011] [Revised: 09/18/2011] [Accepted: 10/09/2011] [Indexed: 01/22/2023]
Abstract
The small GTPase H-Ras is a proto-oncogene that activates a variety of different pathways including the extracellular-signal-regulated kinase (ERK)/mitogen-activated protein kinase pathway. H-Ras is mutated in many human malignancies, and these mutations cause the protein to be constitutively active. Phosphoprotein enriched in astrocytes, 15 kDa (PEA-15) blocks ERK-dependent gene transcription and inhibits proliferation by sequestering ERK in the cytoplasm. We therefore investigated whether PEA-15 influences H-Ras-mediated transformation. We found that PEA-15 does not block H-Ras-activated proliferation when H-Ras is constitutively active. We show instead that in H-Ras-transformed mouse kidney epithelial cells, co-expression of PEA-15 resulted in enhanced soft agar colony growth and increased tumor growth in vivo. Overexpression of both H-Ras and PEA-15 resulted in accelerated G1/S cell cycle transition and increased activation of the ERK signaling pathway. PEA-15 mediated these effects through activation of its binding partner phospholipase D1 (PLD1). Inhibition of PLD1 or interference with PEA-15/PLD1 binding blocked PEA-15's ability to increase ERK activation. Our findings reveal a novel mechanism by which PEA-15 positively regulates Ras/ERK signaling and increases the proliferation of H-Ras-transformed epithelial cells through enhanced PLD1 expression and activation. Thus, our work provides a surprising mechanism by which PEA-15 augments H-Ras-driven transformation. These data reveal that PEA-15 not only suppresses ERK signaling and tumorigenesis but also alternatively enhances tumorigenesis in the context of active Ras.
Collapse
Affiliation(s)
- F J Sulzmaier
- Cancer Biology Program, University of Hawaii Cancer Center, University of Hawaii at Manoa, Honolulu, HI 96813, USA
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Gawecka JE, Geerts D, Koster J, Caliva MJ, Sulzmaier FJ, Opoku-Ansah J, Wada RK, Bachmann AS, Ramos JW. PEA15 impairs cell migration and correlates with clinical features predicting good prognosis in neuroblastoma. Int J Cancer 2012; 131:1556-68. [PMID: 22213050 DOI: 10.1002/ijc.27415] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2011] [Accepted: 12/09/2011] [Indexed: 12/29/2022]
Abstract
ERK and RSK2 drive proliferation and invasion of many cancers. Phosphoprotein enriched in astrocytes 15 (PEA15) binds ERK and RSK2 and high PEA15 levels can impair ERK- and RSK2-dependent transcription. PEA15 expression also inversely correlates with cell motility and invasiveness. We therefore tested PEA15 effects on neuroblastoma cells in vitro. We further analyzed PEA15 expression in the context of clinical and genetic features of neuroblastoma in tumor samples to determine its correlation with disease progression. Affymetrix microarray analysis was performed using 24 different neuroblastoma cell lines. Cell lines expressing low to intermediate levels of PEA15 were chosen for in vitro functional studies. The cell line results were verified by Affymetrix analysis of three different neuroblastic tumor types (total of 110 samples) PEA15 overexpression inhibited neuroblastoma migration in vitro. We verified that inhibition of motility required PEA15 interaction with its binding partners ERK and RSK2. Additionally, synthetic inhibitors of RSK2 suppressed integrin-dependent migration. PEA15 expression correlates with clinical parameters and a 25% increase in patient survival rate. The highest PEA15 levels were found in low stage, more differentiated and less metastatic neuroblastic tumors, and correlated with lack of MYCN amplification. PEA15 blocks neuroblastoma migration through inhibition of ERK/RSK2 signaling. PEA15 expression levels correlate with favorable clinical features suggesting that PEA15 limits metastatic progression of neuroblastoma. Thus, PEA15 and its partners ERK and RSK2 are potential targets for the development of new therapeutics to impede progression of minimal residual disease in patients with high-risk neuroblastoma.
Collapse
Affiliation(s)
- Joanna E Gawecka
- Cancer Biology Program, University of Hawai'i Cancer Center, University of Hawai'i at Manoa, Honolulu, HI 96813, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
Tiling array and novel sequencing technologies have made available the transcription profile of the entire human genome. However, the extent of transcription and the function of genetic elements that occur outside of protein-coding genes, particularly those involved in disease, are still a matter of debate. In this review, we focus on long non-coding RNAs (lncRNAs) that are involved in cancer. We define lncRNAs and present a cancer-oriented list of lncRNAs, list some tools (for example, public databases) that classify lncRNAs or that scan genome spans of interest to find whether known lncRNAs reside there, and describe some of the functions of lncRNAs and the possible genetic mechanisms that underlie lncRNA expression changes in cancer, as well as current and potential future applications of lncRNA research in the treatment of cancer.
Collapse
|