1
|
Krchniakova M, Paukovcekova S, Chlapek P, Neradil J, Skoda J, Veselska R. Thiosemicarbazones and selected tyrosine kinase inhibitors synergize in pediatric solid tumors: NDRG1 upregulation and impaired prosurvival signaling in neuroblastoma cells. Front Pharmacol 2022; 13:976955. [PMID: 36160437 PMCID: PMC9490180 DOI: 10.3389/fphar.2022.976955] [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: 06/23/2022] [Accepted: 08/01/2022] [Indexed: 11/21/2022] Open
Abstract
Tyrosine kinase inhibitors (TKIs) are frequently used in combined therapy to enhance treatment efficacy and overcome drug resistance. The present study analyzed the effects of three inhibitors, sunitinib, gefitinib, and lapatinib, combined with iron-chelating agents, di-2-pyridylketone-4,4-dimethyl-3-thiosemicarbazone (Dp44mT) or di-2-pyridylketone-4-cyclohexyl-4-methyl-3-thiosemicarbazone (DpC). Simultaneous administration of the drugs consistently resulted in synergistic and/or additive activities against the cell lines derived from the most frequent types of pediatric solid tumors. The results of a detailed analysis of cell signaling in the neuroblastoma cell lines revealed that TKIs inhibited the phosphorylation of the corresponding receptor tyrosine kinases, and thiosemicarbazones downregulated the expression of epidermal growth factor receptor, platelet-derived growth factor receptor, and insulin-like growth factor-1 receptor, leading to a strong induction of apoptosis. Marked upregulation of the metastasis suppressor N-myc downstream regulated gene-1 (NDRG1), which is known to be activated and upregulated by thiosemicarbazones in adult cancers, was also detected in thiosemicarbazone-treated neuroblastoma cells. Importantly, these effects were more pronounced in the cells treated with drug combinations, especially with the combinations of lapatinib with thiosemicarbazones. Therefore, these results provide a rationale for novel strategies combining iron-chelating agents with TKIs in therapy of pediatric solid tumors.
Collapse
Affiliation(s)
- Maria Krchniakova
- Laboratory of Tumor Biology, Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czechia
- International Clinical Research Center, St. Anne’s University Hospital, Brno, Czechia
| | - Silvia Paukovcekova
- Laboratory of Tumor Biology, Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czechia
| | - Petr Chlapek
- Laboratory of Tumor Biology, Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czechia
- International Clinical Research Center, St. Anne’s University Hospital, Brno, Czechia
| | - Jakub Neradil
- Laboratory of Tumor Biology, Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czechia
- International Clinical Research Center, St. Anne’s University Hospital, Brno, Czechia
| | - Jan Skoda
- Laboratory of Tumor Biology, Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czechia
- International Clinical Research Center, St. Anne’s University Hospital, Brno, Czechia
- *Correspondence: Jan Skoda, ; Renata Veselska,
| | - Renata Veselska
- Laboratory of Tumor Biology, Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czechia
- International Clinical Research Center, St. Anne’s University Hospital, Brno, Czechia
- *Correspondence: Jan Skoda, ; Renata Veselska,
| |
Collapse
|
2
|
Macsek P, Skoda J, Krchniakova M, Neradil J, Veselska R. Iron-Chelation Treatment by Novel Thiosemicarbazone Targets Major Signaling Pathways in Neuroblastoma. Int J Mol Sci 2021; 23:ijms23010376. [PMID: 35008802 PMCID: PMC8745636 DOI: 10.3390/ijms23010376] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 12/24/2021] [Accepted: 12/27/2021] [Indexed: 01/23/2023] Open
Abstract
Despite constant advances in the field of pediatric oncology, the survival rate of high-risk neuroblastoma patients remains poor. The molecular and genetic features of neuroblastoma, such as MYCN amplification and stemness status, have established themselves not only as potent prognostic and predictive factors but also as intriguing targets for personalized therapy. Novel thiosemicarbazones target both total level and activity of a number of proteins involved in some of the most important signaling pathways in neuroblastoma. In this study, we found that di-2-pyridylketone 4-cyclohexyl-4-methyl-3-thiosemicarbazone (DpC) potently decreases N-MYC in MYCN-amplified and c-MYC in MYCN-nonamplified neuroblastoma cell lines. Furthermore, DpC succeeded in downregulating total EGFR and phosphorylation of its most prominent tyrosine residues through the involvement of NDRG1, a positive prognostic marker in neuroblastoma, which was markedly upregulated after thiosemicarbazone treatment. These findings could provide useful knowledge for the treatment of MYC-driven neuroblastomas that are unresponsive to conventional therapies.
Collapse
Affiliation(s)
- Peter Macsek
- Laboratory of Tumor Biology, Department of Experimental Biology, Faculty of Science, Masaryk University, 601 77 Brno, Czech Republic; (P.M.); (J.S.); (M.K.); (R.V.)
- International Clinical Research Center, St. Anne’s University Hospital, 656 91 Brno, Czech Republic
| | - Jan Skoda
- Laboratory of Tumor Biology, Department of Experimental Biology, Faculty of Science, Masaryk University, 601 77 Brno, Czech Republic; (P.M.); (J.S.); (M.K.); (R.V.)
- International Clinical Research Center, St. Anne’s University Hospital, 656 91 Brno, Czech Republic
| | - Maria Krchniakova
- Laboratory of Tumor Biology, Department of Experimental Biology, Faculty of Science, Masaryk University, 601 77 Brno, Czech Republic; (P.M.); (J.S.); (M.K.); (R.V.)
| | - Jakub Neradil
- Laboratory of Tumor Biology, Department of Experimental Biology, Faculty of Science, Masaryk University, 601 77 Brno, Czech Republic; (P.M.); (J.S.); (M.K.); (R.V.)
- International Clinical Research Center, St. Anne’s University Hospital, 656 91 Brno, Czech Republic
- Department of Pediatric Oncology, Faculty of Medicine, University Hospital Brno, Masaryk University, 662 63 Brno, Czech Republic
- Correspondence: ; Tel.: +420-549-49-6003
| | - Renata Veselska
- Laboratory of Tumor Biology, Department of Experimental Biology, Faculty of Science, Masaryk University, 601 77 Brno, Czech Republic; (P.M.); (J.S.); (M.K.); (R.V.)
- International Clinical Research Center, St. Anne’s University Hospital, 656 91 Brno, Czech Republic
- Department of Pediatric Oncology, Faculty of Medicine, University Hospital Brno, Masaryk University, 662 63 Brno, Czech Republic
| |
Collapse
|
3
|
Damen PJJ, Bulthuis VJ, Hanssens PEJ, Lie ST, Fleischeuer R, Melotte V, Wouters KA, Ruland A, Beckervordersandforth J, Speel EJM. WHO grade I meningiomas that show regrowth after gamma knife radiosurgery often show 1p36 loss. Sci Rep 2021; 11:16432. [PMID: 34385566 PMCID: PMC8361078 DOI: 10.1038/s41598-021-95956-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 07/21/2021] [Indexed: 12/12/2022] Open
Abstract
WHO grade I meningiomas occasionally show regrowth after radiosurgical treatment, which cannot be predicted by clinical features. There is increasing evidence that certain biomarkers are associated with regrowth of meningiomas. The aim of this retrospective study was to asses if these biomarkers could be of value to predict regrowth of WHO grade I meningiomas after additive radiosurgery. Forty-four patients with WHO grade I meningiomas who underwent additive radiosurgical treatment between 2002 and 2015 after Simpson IV resection were included in this study, of which 8 showed regrowth. Median follow-up time was 64 months (range 24–137 months). Tumors were analyzed for the proliferation marker Ki-67 by immunohistochemistry and for deletion of 1p36 by fluorescence in situ hybridization (FISH). Furthermore, genomic DNA was analyzed for promoter hypermethylation of the genes NDRG1–4, SFRP1, HOXA9 and MGMT. Comparison of meningiomas with and without regrowth after radiosurgery revealed that loss of 1p36 (p = 0.001) and hypermethylation of NDRG1 (p = 0.046) were correlated with regrowth free survival. Loss of 1p36 was the only parameter that was significantly associated with meningioma regrowth after multivariate analysis (p = 0.01). Assessment of 1p36 loss in tumor tissue prior to radiosurgery might be considered an indicator of prognosis/regrowth. However, this finding has to be validated in an independent larger set of tumors.
Collapse
Affiliation(s)
- Pim J J Damen
- Department of Pathology, GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, P. Debyelaan 25, Postbox 5800, 6202 AZ, Maastricht, The Netherlands
| | - Vincent J Bulthuis
- Department of Neurosurgery, Maastricht University Medical Center, Maastricht, The Netherlands
| | | | - Suan Te Lie
- Gamma Knife Center Tilburg, ETZ-Elisabeth Hospital, Tilburg, The Netherlands
| | - Ruth Fleischeuer
- Department of Pathology, ETZ-Elisabeth Hospital, Tilburg, The Netherlands
| | - Veerle Melotte
- Department of Pathology, GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, P. Debyelaan 25, Postbox 5800, 6202 AZ, Maastricht, The Netherlands
| | - Kim A Wouters
- Department of Pathology, GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, P. Debyelaan 25, Postbox 5800, 6202 AZ, Maastricht, The Netherlands
| | - Andrea Ruland
- Department of Pathology, GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, P. Debyelaan 25, Postbox 5800, 6202 AZ, Maastricht, The Netherlands
| | - Jan Beckervordersandforth
- Department of Pathology, GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, P. Debyelaan 25, Postbox 5800, 6202 AZ, Maastricht, The Netherlands
| | - Ernst Jan M Speel
- Department of Pathology, GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, P. Debyelaan 25, Postbox 5800, 6202 AZ, Maastricht, The Netherlands.
| |
Collapse
|
4
|
Abstract
Cancer cells accumulate iron to supplement their aberrant growth and metabolism. Depleting cells of iron by iron chelators has been shown to be selectively cytotoxic to cancer cells in vitro and in vivo. Iron chelators are effective at combating a range of cancers including those which are difficult to treat such as androgen insensitive prostate cancer and cancer stem cells. This review will evaluate the impact of iron chelation on cancer cell survival and the underlying mechanisms of action. A plethora of studies have shown iron chelators can reverse some of the major hallmarks and enabling characteristics of cancer. Iron chelators inhibit signalling pathways that drive proliferation, migration and metastasis as well as return tumour suppressive signalling. In addition to this, iron chelators stimulate apoptotic and ER stress signalling pathways inducing cell death even in cells lacking a functional p53 gene. Iron chelators can sensitise cancer cells to PARP inhibitors through mimicking BRCAness; a feature of cancers trademark genomic instability. Iron chelators target cancer cell metabolism, attenuating oxidative phosphorylation and glycolysis. Moreover, iron chelators may reverse the major characteristics of oncogenic transformation. Iron chelation therefore represent a promising selective mode of cancer therapy.
Collapse
|
5
|
Park KC, Paluncic J, Kovacevic Z, Richardson DR. Pharmacological targeting and the diverse functions of the metastasis suppressor, NDRG1, in cancer. Free Radic Biol Med 2020; 157:154-175. [PMID: 31132412 DOI: 10.1016/j.freeradbiomed.2019.05.020] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 04/24/2019] [Accepted: 05/16/2019] [Indexed: 12/18/2022]
Abstract
N-myc downstream regulated gene-1 (NDRG1) is a potent metastasis suppressor that is regulated by hypoxia, metal ions including iron, the free radical nitric oxide (NO.), and various stress stimuli. This intriguing molecule exhibits diverse functions in cancer, inhibiting epithelial-mesenchymal transition (EMT), cell migration and angiogenesis by modulation of a plethora of oncogenes via cellular signaling. Thus, pharmacological targeting of NDRG1 signaling in cancer is a promising therapeutic strategy. Of note, novel anti-tumor agents of the di-2-pyridylketone thiosemicarbazone series, which exert the "double punch" mechanism by binding metal ions to form redox-active complexes, have been demonstrated to markedly up-regulate NDRG1 expression in cancer cells. This review describes the mechanisms underlying NDRG1 modulation by the thiosemicarbazones and the diverse effects NDRG1 exerts in cancer. As a major induction mechanism, iron depletion appears critical, with NO. also inducing NDRG1 through its ability to bind iron and generate dinitrosyl-dithiol iron complexes, which are then effluxed from cells. Apart from its potent anti-metastatic role, several studies have reported a pro-oncogenic role of NDRG1 in a number of cancer-types. Hence, it has been suggested that NDRG1 plays pleiotropic roles depending on the cancer-type. The molecular mechanism(s) underlying NDRG1 pleiotropy remain elusive, but are linked to differential regulation of WNT signaling and potentially differential interaction with the tumor suppressor, PTEN. This review discusses NDRG1 induction mechanisms by metal ions and NO. and both the anti- and possible pro-oncogenic functions of NDRG1 in multiple cancer-types and compares the opposite effects this protein exerts on cancer progression.
Collapse
Affiliation(s)
- Kyung Chan Park
- Molecular Pharmacology and Pathology Program, Discipline of Pathology and Bosch Institute, Medical Foundation Building (K25), The University of Sydney, Sydney, New South Wales, 2006, Australia
| | - Jasmina Paluncic
- Molecular Pharmacology and Pathology Program, Discipline of Pathology and Bosch Institute, Medical Foundation Building (K25), The University of Sydney, Sydney, New South Wales, 2006, Australia
| | - Zaklina Kovacevic
- Molecular Pharmacology and Pathology Program, Discipline of Pathology and Bosch Institute, Medical Foundation Building (K25), The University of Sydney, Sydney, New South Wales, 2006, Australia.
| | - Des R Richardson
- Molecular Pharmacology and Pathology Program, Discipline of Pathology and Bosch Institute, Medical Foundation Building (K25), The University of Sydney, Sydney, New South Wales, 2006, Australia.
| |
Collapse
|
6
|
Schonkeren SL, Massen M, van der Horst R, Koch A, Vaes N, Melotte V. Nervous NDRGs: the N-myc downstream-regulated gene family in the central and peripheral nervous system. Neurogenetics 2019; 20:173-186. [PMID: 31485792 PMCID: PMC6754360 DOI: 10.1007/s10048-019-00587-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 08/22/2019] [Indexed: 02/07/2023]
Abstract
The N-Myc downstream-regulated gene (NDRG) family consists of four members (NDRG1, NDRG2, NDRG3, NDRG4) that are differentially expressed in various organs and function in important processes, like cell proliferation and differentiation. In the last couple of decades, interest in this family has risen due to its connection with several disorders of the nervous system including Charcot-Marie-Tooth disease and dementia, as well as nervous system cancers. By combining a literature review with in silico data analysis of publicly available datasets, such as the Mouse Brain Atlas, BrainSpan, the Genotype-Tissue Expression (GTEx) project, and Gene Expression Omnibus (GEO) datasets, this review summarizes the expression and functions of the NDRG family in the healthy and diseased nervous system. We here show that the NDRGs have a differential, relatively cell type-specific, expression pattern in the nervous system. Even though NDRGs share functionalities, like a role in vesicle trafficking, stress response, and neurite outgrowth, other functionalities seem to be unique to a specific member, e.g., the role of NDRG1 in myelination. Furthermore, mutations, phosphorylation, or changes in expression of NDRGs are related to nervous system diseases, including peripheral neuropathy and different forms of dementia. Moreover, NDRG1, NDRG2, and NDRG4 are all involved in cancers of the nervous system, such as glioma, neuroblastoma, or meningioma. All in all, our review elucidates that although the NDRGs belong to the same gene family and share some functional features, they should be considered unique in their expression patterns and functional importance for nervous system development and neuronal diseases.
Collapse
Affiliation(s)
- Simone L Schonkeren
- Department of Pathology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
| | - Maartje Massen
- Department of Pathology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
| | - Raisa van der Horst
- Department of Pathology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
| | - Alexander Koch
- Department of Pathology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
| | - Nathalie Vaes
- Department of Pathology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
| | - Veerle Melotte
- Department of Pathology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, P.O. Box 616, 6200 MD, Maastricht, The Netherlands.
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
7
|
Potential role of the N-MYC downstream-regulated gene family in reprogramming cancer metabolism under hypoxia. Oncotarget 2018; 7:57442-57451. [PMID: 27447861 PMCID: PMC5303000 DOI: 10.18632/oncotarget.10684] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2016] [Accepted: 06/13/2016] [Indexed: 12/19/2022] Open
Abstract
Metabolic reprogramming toward aerobic glycolysis and lactate fermentation supplies cancer cells with intermediate metabolites, which are used as macromolecule precursors. The oncogene MYC contributes to such aerobic metabolism by activating the expression of numerous genes essential for glycolysis and mitochondrial biogenesis. However, to survive and evolve in a hypoxic tumor milieu, cancer cells must revise MYC-driven metabolism because the mitochondrial respiratory chain provides free electrons to generate oxygen free radicals with inefficient production of ATP due to oxygen depletion. Instead, hypoxia-inducible transcription factor hypoxia-inducible factor 1 (HIF-1) takes over the role of MYC in glycolysis, but suppresses mitochondrial biogenesis and activity to protect cells from such threats. Recently, the N-MYC downstream-regulated gene (NDRG) family has received attention as potential biomarkers of cancer prognosis. NDRGs are repressed MYC-dependently in various cancers, but induced under hypoxia because HIF-1 directly activates their promoters and indirectly de-represses them by antagonizing MYC. In this review, we summarize the current understanding of the reprogramming of cancer metabolism via the counterbalance between MYC and HIF-1, and discuss the proven and putative roles of the NDRG family in adjusting cancer metabolism according to the ambient oxygen level.
Collapse
|
8
|
Ambrosio S, Amente S, Saccà CD, Capasso M, Calogero RA, Lania L, Majello B. LSD1 mediates MYCN control of epithelial-mesenchymal transition through silencing of metastatic suppressor NDRG1 gene. Oncotarget 2018; 8:3854-3869. [PMID: 27894074 PMCID: PMC5354800 DOI: 10.18632/oncotarget.12924] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 10/14/2016] [Indexed: 12/30/2022] Open
Abstract
Neuroblastoma (NB) with MYCN amplification is a highly aggressive and metastatic tumor in children. The high recurrence rate and resistance of NB cells to drugs urgently demands a better therapy for this disease. We have recently found that MYCN interacts with the lysine-specific demethylase 1 (LSD1), a histone modifier that participates in key aspects of gene transcription. In cancer cells, LSD1 contributes to the genetic reprogramming that underlies to Epithelial-Mesenchymal Transition (EMT) and tumor metastasis. Here, we show that LSD1 affects motility and invasiveness of NB cells by modulating the transcription of the metastasis suppressor NDRG1 (N-Myc Downstream-Regulated Gene 1). At mechanistic level, we found that LSD1 co-localizes with MYCN at the promoter region of the NDRG1 gene and inhibits its expression. Pharmacological inhibition of LSD1 relieves repression of NDRG1 by MYCN and affects motility and invasiveness of NB cells. These effects were reversed by overexpressing NDRG1. In NB tissues, high levels of LSD1 correlate with low levels of NDRG1 and reduced patients survival. Collectively, our findings elucidate a mechanism of how MYCN/LSD1 control motility and invasiveness of NB cells through transcription regulation of NDRG1 expression and suggest that pharmacological targeting of LSD1 represents a valuable approach for NB therapy.
Collapse
Affiliation(s)
- Susanna Ambrosio
- Department of Biology, University of Naples 'Federico II', Naples, Italy
| | - Stefano Amente
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples, 'Federico II', Naples, Italy
| | - Carmen D Saccà
- Department of Biology, University of Naples 'Federico II', Naples, Italy
| | - Mario Capasso
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples, 'Federico II', Naples, Italy.,CEINGE Biotecnologie Avanzate, Napoli, Italy
| | - Raffaele A Calogero
- Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Torino, Turin, Italy
| | - Luigi Lania
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples, 'Federico II', Naples, Italy
| | - Barbara Majello
- Department of Biology, University of Naples 'Federico II', Naples, Italy
| |
Collapse
|
9
|
Chaudhari N, Talwar P, Lefebvre D'hellencourt C, Ravanan P. CDDO and ATRA Instigate Differentiation of IMR32 Human Neuroblastoma Cells. Front Mol Neurosci 2017; 10:310. [PMID: 29018329 PMCID: PMC5623017 DOI: 10.3389/fnmol.2017.00310] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2017] [Accepted: 09/14/2017] [Indexed: 12/15/2022] Open
Abstract
Neuroblastoma is the most common solid extra cranial tumor in infants. Improving the clinical outcome of children with aggressive tumors undergoing one of the multiple treatment options has been a major concern. Differentiating neuroblastoma cells holds promise in inducing tumor growth arrest and treating minimal residual disease. In this study, we investigated the effect of partial PPARγ agonist 2-cyano-3,12-dioxooleana-1,9(11)-dien-28-oic acid (CDDO) on human neuroblastoma IMR32 cells. Our results demonstrate that treatment with low concentration of CDDO and particularly in combination with all trans retinoic acid (ATRA) induced neurite outgrowth, increased the percentage of more than two neurites bearing cells, and decreased viability in IMR32 cells. These morphological changes were associated with an increase in expression of bonafide differentiation markers like β3-tubulin and Neuron Specific Enolase (NSE). The differentiation was accompanied by a decrease in the expression of MYCN whose amplification is known to contribute to the pathogenesis of neuroblastoma. MYCN is known to negatively regulate NMYC downstream-regulated gene 1 (NDRG1) in neuroblastomas. MYCN down-regulation induced by CDDO correlated with increased expression of NDRG1. CDDO decreased Anaplastic Lymphoma Kinase (ALK) mRNA expression without affecting its protein level, while ATRA significantly down-regulated ALK. Antagonism of PPARγ receptor by T0070907 meddled with differentiation inducing effects of CDDO as observed by stunted neurite growth, increased viability and decreased expression of differentiation markers. Our findings indicate that IMR32 differentiation induced by CDDO in combination with ATRA enhances, differentiation followed by cell death via cAMP-response-element binding protein (CREB) independent and PPARγ dependent signaling mechanisms.
Collapse
Affiliation(s)
- Namrata Chaudhari
- Apoptosis and Cell Survival Research Lab, Department of Biosciences, School of Biosciences and Technology, VIT University, Vellore, India
| | - Priti Talwar
- Apoptosis and Cell Survival Research Lab, Department of Biosciences, School of Biosciences and Technology, VIT University, Vellore, India
| | - Christian Lefebvre D'hellencourt
- Université de La Réunion, Institut National de la Santé et de la Recherche Médicale, UMR Diabète Athérothombose Thérapies Réunion Océan Indien, Saint-Denis de La Réunion, France
| | - Palaniyandi Ravanan
- Apoptosis and Cell Survival Research Lab, Department of Biosciences, School of Biosciences and Technology, VIT University, Vellore, India
| |
Collapse
|
10
|
Chang X, Xu X, Xue X, Ma J, Li Z, Deng P, Chen J, Zhang S, Zhi Y, Dai D. NDRG1 Controls Gastric Cancer Migration and Invasion through Regulating MMP-9. Pathol Oncol Res 2016; 22:789-96. [PMID: 27154576 DOI: 10.1007/s12253-016-0071-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2015] [Accepted: 04/27/2016] [Indexed: 01/16/2023]
Abstract
The purpose of this study is to detect the clinical significance of NDRG1 and its relationship with MMP-9 in gastric cancer metastatic progression. 101 cases of gastric cancer specimens were utilized to identify the protein expression of NDRG1 and MMP-9 by immunohistochemistry, their clinical significance was also analyzed. The suppression by siRNA-NDRG1 was employed to detect the role of NDRG1 in gastric cancer progression and its relationship with MMP-9. NDRG1 expression was correlated inversely with the degree of tumor cell differentiation (p < 0.01), invasion depth (p < 0.05), lymph node metastasis (p < 0.05) and TNM stage (p < 0.05), whereas MMP-9 was positive correlated with the degree of tumor cell differentiation (p < 0.01), lymph node metastasis (p < 0.05) and TNM stage (p < 0.05), but not correlated with invasion depth (p>0.05). Furthermore, cell proliferation and invasion effect were remarkably enhanced when NDRG1 was silencing, but MMP-9 expression was increased. NDRG1 silencing enhances gastric cancer cells progression through upregulating MMP-9. It suggests that NDRG1 may inhibit the metastasis of gastric cancer via regulating MMP-9.
Collapse
Affiliation(s)
- Xiaojing Chang
- Department of Gastrointestinal Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, China
- Cancer Center, The Fourth Affiliated Hospital, China Medical University, Shenyang, China
- Department of Radiotherapy, The Second Hospital, Hebei Medical University, Shijiazhuang, China
| | - Xiaoyang Xu
- Cancer Center, The Fourth Affiliated Hospital, China Medical University, Shenyang, China
| | - Xiaoying Xue
- Department of Radiotherapy, The Second Hospital, Hebei Medical University, Shijiazhuang, China
| | - Jinguo Ma
- Cancer Center, The Fourth Affiliated Hospital, China Medical University, Shenyang, China
| | - Zhenhua Li
- Cancer Center, The Fourth Affiliated Hospital, China Medical University, Shenyang, China
| | - Peng Deng
- Cancer Center, The Fourth Affiliated Hospital, China Medical University, Shenyang, China
| | - Jing Chen
- Department of Gastrointestinal Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, China
| | - Shuanglong Zhang
- Cancer Center, The Fourth Affiliated Hospital, China Medical University, Shenyang, China
| | - Yu Zhi
- Cancer Center, The Fourth Affiliated Hospital, China Medical University, Shenyang, China
| | - Dongqiu Dai
- Department of Gastrointestinal Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, China.
- Cancer Center, The Fourth Affiliated Hospital, China Medical University, Shenyang, China.
- Department of Gastrointestinal Surgery and Cancer Center, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China.
| |
Collapse
|
11
|
Redox cycling metals: Pedaling their roles in metabolism and their use in the development of novel therapeutics. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:727-48. [PMID: 26844773 DOI: 10.1016/j.bbamcr.2016.01.026] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 01/29/2016] [Indexed: 12/12/2022]
Abstract
Essential metals, such as iron and copper, play a critical role in a plethora of cellular processes including cell growth and proliferation. However, concomitantly, excess of these metal ions in the body can have deleterious effects due to their ability to generate cytotoxic reactive oxygen species (ROS). Thus, the human body has evolved a very well-orchestrated metabolic system that keeps tight control on the levels of these metal ions. Considering their very high proliferation rate, cancer cells require a high abundance of these metals compared to their normal counterparts. Interestingly, new anti-cancer agents that take advantage of the sensitivity of cancer cells to metal sequestration and their susceptibility to ROS have been developed. These ligands can avidly bind metal ions to form redox active metal complexes, which lead to generation of cytotoxic ROS. Furthermore, these agents also act as potent metastasis suppressors due to their ability to up-regulate the metastasis suppressor gene, N-myc downstream regulated gene 1. This review discusses the importance of iron and copper in the metabolism and progression of cancer, how they can be exploited to target tumors and the clinical translation of novel anti-cancer chemotherapeutics.
Collapse
|
12
|
Zhang D, Jia J, Zhao G, Yue M, Yang H, Wang J. NDRG1 promotes the multidrug resistance of neuroblastoma cells with upregulated expression of drug resistant proteins. Biomed Pharmacother 2015; 76:46-51. [PMID: 26653549 DOI: 10.1016/j.biopha.2015.10.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 10/16/2015] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Resistance to chemotherapeutic drugs and recurrence are two major causes of poor prognosis in many tumors including neuroblastoma. This study aimed to investigate the effect of the elevated intracellular NDRG1 expression on drug resistance of human neuroblastoma cells to chemotherapy, for exploring novel approaches for biotherapy of neuroblastoma. METHODS Human neuroblastoma KP-N-Ns cell lines were transfected with the lentivirus vector containing human NDRG1 cDNA, with empty vector-transfected or blank cells as controls. Transfection status was confirmed under fluorescence microscope, while PCR assay and western blot analysis were performed to determine the expression changes. MTT and TUNEL assays were used to detect the resistance of the cells to anticancer drugs, including vincristine, cyclophosphamide and so on. Additionally, the expression of drug resistant proteins was detected. RESULTS Stable lentiviral transfection cell line was successfully established with significantly upregulated NDRG1 expression. MTT assay revealed greater cell growth under NDRG1 overexpression with drugs stimulation, as compared to controls. TUNEL assay also showed less apoptosis of NDRG1 overexpressing cells than those of controls when exposed to these drugs, suggesting the increasing drug resistance through NDRG1 overexpression. Besides, the expression of MDR, LRP-1 and MRP-1 was also increased in NDRG1 overexpressing cells, implying NDRG1-mediated pathways in multidrug resistance of neuroblastoma. CONCLUSION NDRG1 could increase the resistance of neuroblastoma cells to chemotherapeutic drugs, with its positive regulation on drug resistant proteins. This study provided new insights for exploring the mechanism of the resistance to chemotherapeutic drugs and also novel approach for biotherapy in neuroblastoma.
Collapse
Affiliation(s)
- Da Zhang
- Department of Pediatric Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Jia Jia
- Department of Pediatric Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Ge Zhao
- Department of Pediatric Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Min Yue
- Department of Pediatric Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Heying Yang
- Department of Pediatric Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Jiaxiang Wang
- Department of Pediatric Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China.
| |
Collapse
|
13
|
Identification of DRG-1 As a Melanoma-Associated Antigen Recognized by CD4+ Th1 Cells. PLoS One 2015; 10:e0124094. [PMID: 25993655 PMCID: PMC4439028 DOI: 10.1371/journal.pone.0124094] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 03/02/2015] [Indexed: 01/08/2023] Open
Abstract
Immunotherapy has emerged as a promising strategy for the treatment of metastatic melanoma. Clinical studies have demonstrated the feasibility of cancer immunotherapy using tumor antigens recognized by CD8+ T cells. However, the overall immune responses induced by these antigens are too weak and transient to induce tumor regression in the majority of patients who received immunization. A growing body of evidence suggests that CD4+ T helper (Th) cells play an important role in antitumor immunity. Therefore, the identification of MHC class II-restricted tumor antigens capable of stimulating CD4+ T cells may provide opportunities for developing effective cancer vaccines. To this end, we describe the identification of developmentally regulated GTP-binding protein 1 (DRG-1) as a melanoma-associated antigen recognized by HLA-DR11-restricted CD4+ Th1 cells. Epitope mapping analysis showed that the DRG1248-268 epitope of DRG-1 was required for T cell recognition. Reverse transcription-polymerase chain reaction revealed that DRG-1 was highly expressed in melanoma cell lines but not in normal tissues. DRG-1 knockdown by lentiviral-based shRNA suppressed melanoma cell proliferation and soft agar colony formation. Taken together, these data suggest that DRG-1 plays an important role in melanoma cell growth and transformation, indicating that DRG1 may represent a novel target for CD4+ T cell-mediated immunotherapy in melanoma.
Collapse
|
14
|
Li D, Mei H, Qi M, Yang D, Zhao X, Xiang X, Pu J, Huang K, Zheng L, Tong Q. FOXD3 is a novel tumor suppressor that affects growth, invasion, metastasis and angiogenesis of neuroblastoma. Oncotarget 2014; 4:2021-44. [PMID: 24269992 PMCID: PMC3875767 DOI: 10.18632/oncotarget.1579] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The transcription factor forkhead box D3 (FOXD3) plays a crucial role in the development of neural crest cells. However, the function and underlying mechanisms of FOXD3 in the progression of neuroblastoma (NB), an embryonal tumor that is derived from the neural crest, still remain largely unknown. Here, we report that FOXD3 is an important oncosuppressor of NB tumorigenicity and aggressiveness. We found that FOXD3 was down-regulated in NB tissues and cell lines. Patients with high FOXD3 expression have greater survival probability. Over-expression or knockdown of FOXD3 responsively altered both the protein and mRNA levels of N-myc downstream regulated 1 (NDRG1) and its downstream genes, vascular endothelial growth factor and matrix metalloproteinase 9, in cultured NB cell lines SH-SY5Y and SK-N-SH. Luciferase reporter and chromatin immunoprecipitation assays indicated that FOXD3 directly targeted the binding site within NDRG1 promoter to facilitate its transcription. Ectopic expression of FOXD3 suppressed the growth, invasion, metastasis and angiogenesis of SH-SY5Y and SK-N-SH cells in vitro and in vivo. Conversely, knockdown of FOXD3 promoted the growth, migration, invasion and angiogenesis of NB cells. In addition, rescue experiments in FOXD3 over-expressed or silenced NB cells showed that restoration of NDRG1 expression prevented the tumor cells from FOXD3-mediated changes in these biological features. Our results indicate that FOXD3 exhibits tumor suppressive activity that affects the growth, aggressiveness and angiogenesis of NB through transcriptional regulation of NDRG1.
Collapse
Affiliation(s)
- Dan Li
- Department of Pediatric Surgery, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Metastasis suppressors in breast cancers: mechanistic insights and clinical potential. J Mol Med (Berl) 2013; 92:13-30. [PMID: 24311119 DOI: 10.1007/s00109-013-1109-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Revised: 11/14/2013] [Accepted: 11/19/2013] [Indexed: 01/20/2023]
Abstract
For the most part, normal epithelial cells do not disseminate to other parts of the body and proliferate, as do metastatic cells. Presumably, a class of molecules-termed metastasis suppressors-are involved in this homeostatic control. Metastasis suppressors are, by definition, cellular factors that, when re-expressed in metastatic cells, functionally inhibit metastasis without significantly inhibiting tumor growth. In this brief review, we catalog known metastasis suppressors, what is known about their mechanism(s) of action, and experimental and clinical associations to date.
Collapse
|