1
|
Shiozaki A, Konishi T, Kosuga T, Kudou M, Kurashima K, Inoue H, Shoda K, Arita T, Konishi H, Morimura R, Komatsu S, Ikoma H, Toma A, Kubota T, Fujiwara H, Okamoto K, Otsuji E. Roles of voltage‑gated potassium channels in the maintenance of pancreatic cancer stem cells. Int J Oncol 2021; 59:76. [PMID: 34414448 PMCID: PMC8425586 DOI: 10.3892/ijo.2021.5256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 06/16/2021] [Indexed: 11/10/2022] Open
Abstract
The targeting of membrane proteins that are activated in cancer stem cells (CSCs) represents one of the key recent strategies in cancer therapy. The present study analyzed ion channel expression profiles and functions in pancreatic CSCs (PCSCs). Cells strongly expressing aldehyde dehydrogenase 1 family member A1 (ALDH1A1) were isolated from the human pancreatic PK59 cell line using fluorescence-activated cell sorting, and PCSCs were identified based on tumorsphere formation. Microarray analysis was performed to investigate the gene expression profiles in PCSCs. ALDH1A1 messenger RNA levels were higher in PCSCs compared with non-PCSCs. PCSCs were resistant to 5-fluorouracil and capable of redifferentiation. The results of the microarray analysis revealed that gene expression related to ion channels, including voltage-gated potassium channels (Kv), was upregulated in PCSCs compared with non-PCSCs. 4-Aminopyridine (4-AP), a potent Kv inhibitor, exhibited greater cytotoxicity in PCSCs compared with non-PCSCs. In a xenograft model in nude mice, tumor volumes were significantly lower in mice inoculated with PK59 cells pre-treated with 4-AP compared with those in mice injected with non-treated cells. The present results identified a role of Kv in the persistence of PCSCs and suggested that the Kv inhibitor 4-AP may have potential as a therapeutic agent for pancreatic carcinoma.
Collapse
Affiliation(s)
- Atsushi Shiozaki
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602‑8566, Japan
| | - Tomoki Konishi
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602‑8566, Japan
| | - Toshiyuki Kosuga
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602‑8566, Japan
| | - Michihiro Kudou
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602‑8566, Japan
| | - Kento Kurashima
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602‑8566, Japan
| | - Hiroyuki Inoue
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602‑8566, Japan
| | - Katsutoshi Shoda
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602‑8566, Japan
| | - Tomohiro Arita
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602‑8566, Japan
| | - Hirotaka Konishi
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602‑8566, Japan
| | - Ryo Morimura
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602‑8566, Japan
| | - Shuhei Komatsu
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602‑8566, Japan
| | - Hisashi Ikoma
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602‑8566, Japan
| | - Atsushi Toma
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602‑8566, Japan
| | - Takeshi Kubota
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602‑8566, Japan
| | - Hitoshi Fujiwara
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602‑8566, Japan
| | - Kazuma Okamoto
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602‑8566, Japan
| | - Eigo Otsuji
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602‑8566, Japan
| |
Collapse
|
2
|
Neurofibromin level directs RAS pathway signaling and mediates sensitivity to targeted agents in malignant peripheral nerve sheath tumors. Oncotarget 2018; 9:22571-22585. [PMID: 29854299 PMCID: PMC5978249 DOI: 10.18632/oncotarget.25181] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 04/04/2018] [Indexed: 12/11/2022] Open
Abstract
Malignant peripheral nerve sheath tumor (MPNST) is a type of soft-tissue sarcoma strongly associated with dysfunction in neurofibromin; an inhibitor of the RAS pathway. We performed high-throughput screening of an array of FDA approved and promising agents in clinical development both alone and in combination at physiologically achievable concentrations against a panel of established MPNST cell line models. We found that drugs targeting a variety of factors in the RAS pathway can effectively lead to cell death in vitro with considerable drug combination synergy in regimens that target MEK or mTOR. We observed that the degree of relative sensitivity to chemotherapeutic agents was associated with the status of neurofibromin in these cell line models. Using a combination of agents that target MEK and mTORC1/2, we effectively silenced RAS/PI3K/MEK/mTOR signaling in vitro. Moreover, we employed RNAi against NF1 to establish that MPNST drug sensitivity is directly proportional to relative level of intracellular neurofibromin. Thus, two-drug combinations that target MEK and mTORC1/2 are most effective in halting the RAS signaling cascade, and the relative success of this and related small molecule interventions in MPNSTs may be predicated upon the molecular status of neurofibromin.
Collapse
|
3
|
Hu XM, Li XQ, Li XT. The Tau-Induced Reduction of mRNA Levels of Kv Channels in Human Neuroblastoma SK-N-SH Cells. J Mol Neurosci 2015; 58:306-11. [PMID: 26576773 DOI: 10.1007/s12031-015-0683-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2015] [Accepted: 11/09/2015] [Indexed: 11/25/2022]
Abstract
Previous findings indicated that microtubule-binding protein tau and voltage-gated K(+) (Kv) channels exhibit a regulatory role in cell proliferation. However, the possible interaction of tau with Kv channels remained obscure. In this report, transfection of tau plasmids into human neuroblastoma SK-N-SH cells caused a significant reduction in the messenger RNA (mRNA) levels of several Kv channels, including Kv2.1, Kv3.1, Kv5.1, Kv9.2, and KCNH4. Correspondingly, the Kv currents recorded using patch-clamp techniques were substantially declined in the tau-transfected SK-N-SH cells. Moreover, tau induction and treatment with the Kv channel blocker TEA (tetraethylammonium) were able to improve proliferation rates of SK-N-SH cells by 43.1 and 66.2%, respectively. These data suggested that the tau-mediated alteration of Kv channels could be involved in its action on neural proliferation.
Collapse
Affiliation(s)
- Xi-Mu Hu
- Graduate Institute of South-Central University for Nationalities, Wuhan, 430074, China
| | - Xiao-Qing Li
- Graduate Institute of South-Central University for Nationalities, Wuhan, 430074, China
| | - Xian-Tao Li
- Department of Neurobiology, College of Life Sciences, South-Central University for Nationalities, Wuhan, 430074, China.
| |
Collapse
|
4
|
Neuroprotective or neurotoxic effects of 4-aminopyridine mediated by KChIP1 regulation through adjustment of Kv 4.3 potassium channels expression and GABA-mediated transmission in primary hippocampal cells. Toxicology 2015; 333:107-117. [PMID: 25917026 DOI: 10.1016/j.tox.2015.04.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2015] [Revised: 04/05/2015] [Accepted: 04/22/2015] [Indexed: 11/21/2022]
Abstract
4-Aminopyridine (4-AP) is a potassium channel blocker used for the treatment of neuromuscular disorders. Otherwise, it has been described to produce a large number of adverse effects among them cell death mediated mainly by blockage of K(+) channels. However, a protective effect against cell death has also been described. On the other hand, Kv channel interacting protein 1 (KChIP1) is a neuronal calcium sensor protein that is predominantly expressed at GABAergic synapses and it has been related with modulation of K(+) channels, GABAergic transmission and cell death. According to this KChIP1 could play a key role in the protective or toxic effects induced by 4-AP. We evaluated, in wild type and KChIP1 silenced primary hippocampal neurons, the effect of 4-AP (0.25μM to 2mM) with or without semicarbazide (0.3M) co-treatment after 24h and after 14 days 4-AP alone exposure on cell viability, the effect of 4-AP (0.25μM to 2mM) on KChIP1 and Kv 4.3 potassium channels gene expression and GABAergic transmission after 24h treatment or after 14 days exposure to 4-AP (0.25μM to1μM). 4-AP induced cell death after 24h (from 1mM) and after 14 days treatment. We observed that 4-AP modulates KChIP1 which regulate Kv 4.3 channels expression and GABAergic transmission. Our study suggests that KChIP1 is a key gene that has a protective effect up to certain concentration after short-term treatment with 4-AP against induced cell injury; but this protection is erased after long term exposure, due to KChIP1 down-regulation predisposing cell to 4-AP induced damages. These data might help to explain protective and toxic effects observed after overdose and long term exposure.
Collapse
|
5
|
Rela L, Piantanida AP, Bordey A, Greer CA. Voltage-dependent K+ currents contribute to heterogeneity of olfactory ensheathing cells. Glia 2015; 63:1646-59. [PMID: 25856239 DOI: 10.1002/glia.22834] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 03/24/2015] [Indexed: 02/03/2023]
Abstract
The olfactory nerve is permissive for axon growth throughout life. This has been attributed in part to the olfactory ensheathing glial cells that encompass the olfactory sensory neuron fascicles. Olfactory ensheathing cells (OECs) also promote axon growth in vitro and when transplanted in vivo to sites of injury. The mechanisms involved remain largely unidentified owing in part to the limited knowledge of the physiological properties of ensheathing cells. Glial cells rely for many functions on the properties of the potassium channels expressed; however, those expressed in ensheathing cells are unknown. Here we show that OECs express voltage-dependent potassium currents compatible with inward rectifier (Kir ) and delayed rectifier (KDR ) channels. Together with gap junction coupling, these contribute to the heterogeneity of membrane properties observed in OECs. The relevance of K(+) currents expressed by ensheathing cells is discussed in relation to plasticity of the olfactory nerve.
Collapse
Affiliation(s)
- Lorena Rela
- Departments of Neurosurgery, Yale University School of Medicine, New Haven, Connecticut.,Systems Neuroscience Section, Department of Physiology and Biophysics, School of Medicine, University of Buenos Aires, Argentina.,Institute of Physiology and Biophysics Bernardo Houssay (IFIBIO Houssay-CONICET), Buenos Aires, Argentina
| | - Ana Paula Piantanida
- Systems Neuroscience Section, Department of Physiology and Biophysics, School of Medicine, University of Buenos Aires, Argentina.,Institute of Physiology and Biophysics Bernardo Houssay (IFIBIO Houssay-CONICET), Buenos Aires, Argentina
| | - Angelique Bordey
- Departments of Neurosurgery, Yale University School of Medicine, New Haven, Connecticut.,Yale University School of Medicine, Departments of Cellular and Molecular Physiology, New Haven, Connecticut
| | - Charles A Greer
- Departments of Neurosurgery, Yale University School of Medicine, New Haven, Connecticut.,Yale University School of Medicine, Departments of Neurobiology, New Haven, Connecticut
| |
Collapse
|
6
|
Parenti R, Cardile V, Graziano ACE, Parenti C, Venuti A, Bertuccio MP, Furno DL, Magro G. Wilms' tumor gene 1 (WT1) silencing inhibits proliferation of malignant peripheral nerve sheath tumor sNF96.2 cell line. PLoS One 2014; 9:e114333. [PMID: 25474318 PMCID: PMC4256418 DOI: 10.1371/journal.pone.0114333] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 11/06/2014] [Indexed: 12/25/2022] Open
Abstract
Wilms' tumor gene 1 (WT1) plays complex roles in tumorigenesis, acting as tumor suppressor gene or an oncogene depending on the cellular context. WT1 expression has been variably reported in both benign and malignant peripheral nerve sheath tumors (MPNSTs) by means of immunohistochemistry. The aim of the present study was to characterize its potential pathogenetic role in these relatively uncommon malignant tumors. Firstly, immunohistochemical analyses in MPNST sNF96.2 cell line showed strong WT1 staining in nuclear and perinuclear areas of neoplastic cells. Thus, we investigated the effects of silencing WT1 by RNA interference. Through Western Blot analysis and proliferation assay we found that WT1 knockdown leads to the reduction of cell growth in a time- and dose-dependent manner. siWT1 inhibited proliferation of sNF96.2 cell lines likely by influencing cell cycle progression through a decrease in the protein levels of cyclin D1 and inhibition of Akt phosphorylation compared to the control cells. These results indicate that WT1 knockdown attenuates the biological behavior of MPNST cells by decreasing Akt activity, demonstrating that WT1 is involved in the development and progression of MPNSTs. Thus, WT1 is suggested to serve as a potential therapeutic target for MPNSTs.
Collapse
Affiliation(s)
- Rosalba Parenti
- Department of Biomedical and Biotechnological Sciences, Physiology Section, University of Catania, 95125 Catania, Italy
| | - Venera Cardile
- Department of Biomedical and Biotechnological Sciences, Physiology Section, University of Catania, 95125 Catania, Italy
| | | | - Carmela Parenti
- Department of Drug Sciences, Pharmacology and Toxicology Section, University of Catania, 95125 Catania, Italy
| | - Assunta Venuti
- Business Unit Oncology, Nerviano Medical Sciences S.r.l., 20014 Nerviano Milano, Italy
| | - Maria Paola Bertuccio
- Business Unit Oncology, Nerviano Medical Sciences S.r.l., 20014 Nerviano Milano, Italy
| | - Debora Lo Furno
- Department of Biomedical and Biotechnological Sciences, Physiology Section, University of Catania, 95125 Catania, Italy
| | - Gaetano Magro
- Department G.F. Ingrassia, Azienda Ospedaliero-Universitaria “Policlinico-Vittorio Emanuele” Anatomic Pathology, University of Catania, 95125 Catania, Italy
| |
Collapse
|
7
|
Frejo MT, del Pino J, Lobo M, García J, Capo MA, Díaz MJ. Liver and kidney damage induced by 4-aminopyridine in a repeated dose (28 days) oral toxicity study in rats: Gene expression profile of hybrid cell death. Toxicol Lett 2014; 225:252-63. [DOI: 10.1016/j.toxlet.2013.12.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Revised: 12/18/2013] [Accepted: 12/19/2013] [Indexed: 10/25/2022]
|
8
|
Urrego D, Tomczak AP, Zahed F, Stühmer W, Pardo LA. Potassium channels in cell cycle and cell proliferation. Philos Trans R Soc Lond B Biol Sci 2014; 369:20130094. [PMID: 24493742 PMCID: PMC3917348 DOI: 10.1098/rstb.2013.0094] [Citation(s) in RCA: 288] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Normal cell-cycle progression is a crucial task for every multicellular organism, as it determines body size and shape, tissue renewal and senescence, and is also crucial for reproduction. On the other hand, dysregulation of the cell-cycle progression leading to uncontrolled cell proliferation is the hallmark of cancer. Therefore, it is not surprising that it is a tightly regulated process, with multifaceted and very complex control mechanisms. It is now well established that one of those mechanisms relies on ion channels, and in many cases specifically on potassium channels. Here, we summarize the possible mechanisms underlying the importance of potassium channels in cell-cycle control and briefly review some of the identified channels that illustrate the multiple ways in which this group of proteins can influence cell proliferation and modulate cell-cycle progression.
Collapse
Affiliation(s)
- Diana Urrego
- Oncophysiology Group, Max Planck Institute of Experimental Medicine, , Hermann-Rein-Strasse 3, Göttingen 37075, Germany
| | | | | | | | | |
Collapse
|
9
|
Peacock JD, Cherba D, Kampfschulte K, Smith MK, Monks NR, Webb CP, Steensma M. Molecular-guided therapy predictions reveal drug resistance phenotypes and treatment alternatives in malignant peripheral nerve sheath tumors. J Transl Med 2013; 11:213. [PMID: 24040940 PMCID: PMC3848568 DOI: 10.1186/1479-5876-11-213] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 09/12/2013] [Indexed: 12/12/2022] Open
Abstract
Background Malignant peripheral nerve sheath tumors (MPNST) are rare highly aggressive sarcomas that affect 8-13% of people with neurofibromatosis type 1. The prognosis for patients with MPNST is very poor. Despite TOP2A overexpression in these tumors, doxorubicin resistance is common, and the mechanisms of chemotherapy resistance in MPNST are poorly understood. Molecular-guided therapy prediction is an emerging strategy for treatment refractory sarcomas that involves identification of therapy response and resistance mechanisms in individual tumors. Here, we report the results from a personalized, molecular-guided therapy analysis of MPNST samples. Methods Established molecular-guided therapy prediction software algorithms were used to analyze published microarray data from human MPNST samples and cell lines, with benign neurofibroma tissue controls. MPNST and benign neurofibroma-derived cell lines were used for confirmatory in vitro experimentation using quantitative real-time PCR and growth inhibition assays. Microarray data was analyzed using Affymetrix expression console MAS 5.0 method. Significance was calculated with Welch’s t-test with non-corrected p-value < 0.05 and validated using permutation testing across samples. Paired Student’s t-tests were used to compare relative EC50 values from independent growth inhibition experiments. Results Molecular guided therapy predictions highlight substantial variability amongst human MPNST samples in expression of drug target and drug resistance pathways, as well as some similarities amongst samples, including common up-regulation of DNA repair mechanisms. In a subset of MPNSTs, high expression of ABCC1 is observed, serving as a predicted contra-indication for doxorubicin and related therapeutics in these patients. These microarray-based results are confirmed with quantitative, real-time PCR and immunofluorescence. The functional effect of drug efflux in MPNST-derived cells is confirmed using in vitro growth inhibition assays. Alternative therapeutics supported by the molecular-guided therapy predictions are reported and tested in MPNST-derived cells. Conclusions These results confirm the substantial molecular heterogeneity of MPNSTs and validate molecular-guided therapy predictions in vitro. The observed molecular heterogeneity in MPNSTs influences therapy prediction. Also, mechanisms involving drug transport and DNA damage repair are primary mediators of MPNST chemotherapy resistance. Together, these findings support the utility of individualized therapy in MPNST as in other sarcomas, and provide initial proof-of concept that individualized therapy prediction can be accomplished.
Collapse
|
10
|
LEE PHILIPR, COHEN JONATHANE, TENDI ELISABETTAA, FARRER ROBERT, DE VRIES GEORGEH, BECKER KEVING, FIELDS RDOUGLAS. Transcriptional profiling in an MPNST-derived cell line and normal human Schwann cells. ACTA ACUST UNITED AC 2012; 1:135-47. [PMID: 16429615 PMCID: PMC1325299 DOI: 10.1017/s1740925x04000274] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
cDNA microarrays were utilized to identify abnormally expressed genes in a malignant peripheral nerve sheath tumor (MPNST)-derived cell line, T265, by comparing the mRNA abundance profiles with that of normal human Schwann cells (nhSCs). The findings characterize the molecular phenotype of this important cell-line model of MPNSTs, and elucidate the contribution of Schwann cells in MPNSTs. In total, 4608 cDNA sequences were screened and hybridizations replicated on custom cDNA microarrays. In order to verify the microarray data, a large selection of differentially expressed mRNA transcripts were subjected to semi-quantitative reverse transcription PCR (LightCycler). Western blotting was performed to investigate a selection of genes and signal transduction pathways, as a further validation of the microarray data. The data generated from multiple microarray screens, semi-quantitative RT-PCR and Western blotting are in broad agreement. This study represents a comprehensive gene-expression analysis of an MPNST-derived cell line and the first comprehensive global mRNA profile of nhSCs in culture. This study has identified ~900 genes that are expressed abnormally in the T265 cell line and detected many genes not previously reported to be expressed in nhSCs. The results provide crucial information on the T265 cells that is essential for investigation using this cell line in experimental studies in neurofibromatosis type I (NF1), and important information on normal human Schwann cells that is applicable to a wide range of studies on Schwann cells in cell culture.
Collapse
Affiliation(s)
- PHILIP R. LEE
- Section on Nervous System Development and Plasticity, NICHD, National Institutes of Health, Bethesda, MD
| | - JONATHAN E. COHEN
- Section on Nervous System Development and Plasticity, NICHD, National Institutes of Health, Bethesda, MD
| | - ELISABETTA A. TENDI
- Section on Nervous System Development and Plasticity, NICHD, National Institutes of Health, Bethesda, MD
| | - ROBERT FARRER
- Department of Cell Biology, Neurobiology and Anatomy, Loyola University of Chicago Stritch School of Medicine, Chicago, IL
| | - GEORGE H. DE VRIES
- Department of Cell Biology, Neurobiology and Anatomy, Loyola University of Chicago Stritch School of Medicine, Chicago, IL
| | - KEVIN G. BECKER
- Gene Expression and Genomics Unit, NIA, National Institutes of Health, Baltimore, MD
| | - R. DOUGLAS FIELDS
- Section on Nervous System Development and Plasticity, NICHD, National Institutes of Health, Bethesda, MD
- Correspondence should be addressed to: R. Douglas Fields, Section on Nervous System Development and Plasticity, Bldg. 35, Rm. 2A211, MSC 3713 NICHD, National Institutes of Health, Bethesda, MD 20892, USA, phone: +1 301 480 3209, fax: +1 301 496 9630,
| |
Collapse
|
11
|
Dai C, Santagata S, Tang Z, Shi J, Cao J, Kwon H, Bronson RT, Whitesell L, Lindquist S. Loss of tumor suppressor NF1 activates HSF1 to promote carcinogenesis. J Clin Invest 2012; 122:3742-54. [PMID: 22945628 DOI: 10.1172/jci62727] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2012] [Accepted: 07/12/2012] [Indexed: 01/25/2023] Open
Abstract
Intrinsic stress response pathways are frequently mobilized within tumor cells. The mediators of these adaptive mechanisms and how they contribute to carcinogenesis remain poorly understood. A striking example is heat shock factor 1 (HSF1), master transcriptional regulator of the heat shock response. Surprisingly, we found that loss of the tumor suppressor gene neurofibromatosis type 1 (Nf1) increased HSF1 levels and triggered its activation in mouse embryonic fibroblasts. As a consequence, Nf1-/- cells acquired tolerance to proteotoxic stress. This activation of HSF1 depended on dysregulated MAPK signaling. HSF1, in turn, supported MAPK signaling. In mice, Hsf1 deficiency impeded NF1-associated carcinogenesis by attenuating oncogenic RAS/MAPK signaling. In cell lines from human malignant peripheral nerve sheath tumors (MPNSTs) driven by NF1 loss, HSF1 was overexpressed and activated, which was required for tumor cell viability. In surgical resections of human MPNSTs, HSF1 was overexpressed, translocated to the nucleus, and phosphorylated. These findings reveal a surprising biological consequence of NF1 deficiency: activation of HSF1 and ensuing addiction to this master regulator of the heat shock response. The loss of NF1 function engages an evolutionarily conserved cellular survival mechanism that ultimately impairs survival of the whole organism by facilitating carcinogenesis.
Collapse
Affiliation(s)
- Chengkai Dai
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts 02142, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Sun D, Tainsky MA, Haddad R. Oncogene Mutation Survey in MPNST Cell Lines Enhances the Dominant Role of Hyperactive Ras in NF1 Associated Pro-Survival and Malignancy. TRANSLATIONAL ONCOGENOMICS 2012; 5:1-7. [PMID: 22346343 PMCID: PMC3273949 DOI: 10.4137/tog.s8830] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Malignant peripheral nerve sheath tumors (MPNST) are a type of soft tissue sarcoma that can be associated with germline mutations in Neurofibromatosis type 1 (NF1) or may occur sporadically. Although the etiology of MPNST is poorly understood, it is clear that a loss of function of the NF1 gene, encoding a Ras-GAP, is an important factor in the tumorigenesis of the inherited form of MPNST. Tumor latency in NF1 patients suggests that additional mutational events are probably required for malignancy. In order to define oncogene mutations associated with 5 MPNST cell lines, we assayed the 238 most frequent mutations in 19 commonly activated oncogenes using mass spectroscopy-based analysis. All 238 mutation sites in the assayed oncogenes were determined to harbor only wild-type sequences. These data suggest that hyperactive Ras resulting from the loss function of neurofibromin may be sufficient to set up the direction of malignant transformation of Schwann cells to MPNST.
Collapse
Affiliation(s)
- Daochun Sun
- Center for Molecluar Medicine and Genetics, Wayne State University School of Medicine, Wayne State University, Detroit, MI 48201
| | | | | |
Collapse
|
13
|
Ma J, Li M, Hock J, Yu X. Hyperactivation of mTOR critically regulates abnormal osteoclastogenesis in neurofibromatosis Type 1. J Orthop Res 2012; 30:144-52. [PMID: 21748792 DOI: 10.1002/jor.21497] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2010] [Accepted: 06/15/2011] [Indexed: 02/05/2023]
Abstract
Individuals with nerofibromatosis Type 1 (NF1) frequently suffer a spectrum of bone pathologies, such as abnormal skeletal development (scoliosis, congenital bowing, and congenital pseudoarthroses, etc), lower bone mineral density with increased fracture risk. These skeletal problems may result, in part, from abnormal osteoclastogenesis. Enhanced RAS/PI3K activity has been reported to contribute to abnormal osteoclastogenesis in Nf1 heterozygous (Nf1+/-) mice. However, the specific downstream pathways linked to NF1 abnormal osteoclastogenesis have not been defined. Our aim was to determine whether mammalian target of rapamycin (mTOR) was a key effector responsible for abnormal osteoclastogenesis in NF1. Primary osteoclast-like cells (OCLs) were cultured from Nf1 wild-type (Nf1+/+) and Nf1+/- mice. Compared to Nf1+/+ controls, there were 20% more OCLs induced from Nf1+/- mice. Nf1+/- OCLs were larger and contained more nuclei. Hyperactive mTOR signaling was detected in Nf1+/- OCLs. Inhibition of mTOR signaling by rapamycin in Nf1+/- OCLs abrogated abnormalities in cellular size and number. Moreover, we found that hyperactive mTOR signaling induced abnormal osteoclastogenesis major through hyper-proliferation. Our research suggests that neurofibromin directly regulates osteoclastogenesis through mTOR signaling pathway. Inhibiting mTOR may represent a viable strategy to treat NF1 bone diseases.
Collapse
Affiliation(s)
- Junrong Ma
- West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | | | | | | |
Collapse
|
14
|
Wang W, Xiao J, Adachi M, Liu Z, Zhou J. 4-aminopyridine induces apoptosis of human acute myeloid leukemia cells via increasing [Ca2+]i through P2X7 receptor pathway. Cell Physiol Biochem 2011; 28:199-208. [PMID: 21865727 DOI: 10.1159/000331731] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/28/2011] [Indexed: 11/19/2022] Open
Abstract
4-AP, a voltage-gated potassium channel blocker, was identified to exert critical pro-apoptotic properties in various types of cancer cells. The present study aims to explore the effect of 4-AP on the apoptosis of human AML cells and the underlying mechanism. We found 4-AP inhibited the proliferation and induces apoptosis in both AML cell lines and primary cultured human AML cells. The apoptosis of AML cells after 4-AP treatment was further confirmed by the disruption of mitochondrial membrane potential (MMP) and activation of caspase 3 and 9. 4-AP inhibited Kv currents in NB(4), HL-60 and THP-1 cells. Furthermore, 4-AP induced significant increment in [Ca(2+)](i), which were inhibited by KN-62, a specific blocker of P(2)X(7) receptors. KN-62 also abrogated 4-AP induced apoptosis. Knockdown of P(2)X(7) receptor by small interfering RNA blocked the effect of 4-AP. Conclusively, this study indicated that 4-AP promotes apoptosis in human AML cells via increasing [Ca(2+)](i) through P(2)X(7) receptor.
Collapse
Affiliation(s)
- Wei Wang
- Department of Hematology, the First Affiliated Hospital of Harbin Medical University, Harbin, P.R. China
| | | | | | | | | |
Collapse
|
15
|
Fei XW, Pan CJ, He YL, Fang YJ, Zhuang JL, Mei YA. Brain natriuretic peptide modulates the delayed rectifier outward K(+) current and promotes the proliferation of mouse Schwann cells. J Cell Physiol 2011; 226:440-9. [PMID: 20672326 DOI: 10.1002/jcp.22352] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Brain natriuretic peptide (BNP) may act as a neuromodulator via its associated receptors (natriuretic peptide receptors, NPRs) in the central nervous system (CNS), but few studies have reported its activity in the peripheral nervous system (PNS). In this study, we observed that BNP increased the tetraethylammonium chloride (TEA)-sensitive delayed rectifier outward potassium current (I(K)) in mouse Schwann cells (SCs) using whole-cell recording techniques. At concentrations of 1-100 nM, BNP reversibly activated I(K) in a dose-dependent manner, with modulating its steady-state activation and inactivation properties. The effect of BNP on I(K) was abolished by preincubation with the specific antagonist of NPR-A, and could not be mimicked by application of NPR-C agonist. These results were supported by immunocytochemical findings indicating that NPR-A was expressed in SCs. The application of 8-Br-guanosine 3',5'-monophosphate (8-Br-cGMP) mimicked the effect of BNP on I(K), but BNP was unable to further increase I(K) after the application of cyclic guanosine monophosphate (cGMP). Genistein blocked I(K) and also completely eliminated the effects of BNP and cGMP on I(K). The selective K(V)2.1 subunit blocker, Jingzhaotoxin-III (JZTX-III), reduced I(K) amplitude by 30%, but did not abolish the increase effect of BNP on I(K) amplitude. In addition, BNP significantly stimulated SCs proliferation and this effect could be partly inhibited by TEA. Together these results suggest that BNP modulated I(K) probably via cGMP- and tyrosine kinase-dependent pathways by activation of NPR-A. This effect of BNP on I(K) in SCs might partly explain its effect on cell proliferation.
Collapse
Affiliation(s)
- Xiao-Wei Fei
- Institutes of Brain Science, School of Life Sciences and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | | | | | | | | | | |
Collapse
|
16
|
Morphological changes do not reflect biochemical and functional differentiation in OLN-93 oligodendroglial cells. J Neurosci Methods 2009; 184:1-9. [DOI: 10.1016/j.jneumeth.2009.07.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2009] [Revised: 07/02/2009] [Accepted: 07/03/2009] [Indexed: 01/31/2023]
|
17
|
Potassium channels: new targets in cancer therapy. ACTA ACUST UNITED AC 2006; 30:375-85. [PMID: 16971052 DOI: 10.1016/j.cdp.2006.06.002] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/26/2006] [Indexed: 01/01/2023]
Abstract
BACKGROUND Potassium channels (KCh) are the most diverse and ubiquitous class of ion channels. KCh control membrane potential and contribute to nerve and cardiac action potentials and neurotransmitter release. KCh are also involved in insulin release, differentiation, activation, proliferation, apoptosis, and several other physiological functions. The aim of this review is to provide an updated overview of the KCh role during the cell growth. Their potential use as pharmacological targets in cancer therapies is also discussed. METHODS We searched PubMed (up to 2005) and identified relevant articles. Reprints were mainly obtained by on line subscription. Additional sources were identified through cross-referencing and obtained from Library services. RESULTS KCh are responsible for some neurological and cardiovascular diseases and for a new medical discipline, channelopathies. Their role in congenital deafness, multiple sclerosis, episodic ataxia, LQT syndrome and diabetes has been proven. Furthermore, a large body of information suggests that KCh play a role in the cell cycle progression, and it is now accepted that cells require KCh to proliferate. Thus, KCh expression has been studied in a number of tumours and cancer cells. CONCLUSIONS Cancer is far from being considered a channelopathy. However, it seems appropriate to take into account the involvement of KCh in cancer progression and pathology when developing new strategies for cancer therapy.
Collapse
|
18
|
Dong DL, Wang QH, Yue P, Jiao JD, Gu RM, Yang BF. Indapamide induces apoptosis of GH3 pituitary cells independently of its inhibition of voltage-dependent K+ currents. Eur J Pharmacol 2006; 536:78-84. [PMID: 16556441 DOI: 10.1016/j.ejphar.2006.02.037] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2006] [Accepted: 02/21/2006] [Indexed: 11/26/2022]
Abstract
Indapamide blocks multiple voltage-dependent K+ currents (Kv) in the heart and Kv have an important role in cell proliferation and apoptosis, so the aim of this work was to study the effects of indapamide on Kv and the viability of GH3 cells. Indapamide inhibited Kv of GH3 cells and the inhibition was irreversible after a 10-min washout when more than 250 microM indapamide was used. Indapamide reduced the viability of GH3 cells in a concentration-dependent manner. The decreased cell viability was because indapamide induced cell apoptosis, or even necrosis at higher concentrations. HepG2 cells, which express no apparent Kv, were used to determine the association between inhibition of Kv and the apoptotic action of indapamide. Indapamide had a similar action on cell viability and apoptosis of HepG2 cells. 4-Aminopyridine, the voltage-dependent K+ channel blocker, inhibited Kv of GH3 cells but did not induce the cell apoptosis. We concluded that while indapamide inhibited Kv and induced apoptosis of GH3 cells, the apoptotic action of indapamide was not associated with its inhibition of Kv.
Collapse
Affiliation(s)
- De-Li Dong
- Department of Pharmacology, Harbin Medical University, PR China.
| | | | | | | | | | | |
Collapse
|
19
|
Dasgupta B, Yi Y, Chen DY, Weber JD, Gutmann DH. Proteomic analysis reveals hyperactivation of the mammalian target of rapamycin pathway in neurofibromatosis 1-associated human and mouse brain tumors. Cancer Res 2005; 65:2755-60. [PMID: 15805275 DOI: 10.1158/0008-5472.can-04-4058] [Citation(s) in RCA: 222] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Individuals with the tumor predisposition syndrome, neurofibromatosis 1 (NF1), are prone to development of nervous system tumors, including neurofibromas and pilocytic astrocytomas. Based on the ability of the NF1 gene product (neurofibromin) to function as a GTPase activating protein for RAS, initial biologically based therapies for NF1-associated tumors focused on the use of RAS inhibitors, but with limited clinical success. In an effort to identify additional targets for therapeutic drug design in NF1, we used an unbiased proteomic approach to uncover unanticipated intracellular signaling pathways dysregulated in Nf1-deficient astrocytes. We found that the expression of proteins involved in promoting ribosome biogenesis was increased in the absence of neurofibromin. In addition, Nf1-deficient astrocytes exhibit high levels of mammalian target of rapamycin (mTOR) pathway activation, which was inhibited by blocking K-RAS or phosphatidylinositol 3-kinase activation. This mTOR pathway hyperactivation was reflected by high levels of ribosomal S6 activation in both Nf1 mutant mouse optic nerve gliomas and in human NF1-associated pilocytic astrocytoma tumors. Moreover, inhibition of mTOR signaling in Nf1-/- astrocytes abrogated their growth advantage in culture, restoring normal proliferative rates. These results suggest that mTOR pathway inhibition may represent a logical and tractable biologically based therapy for brain tumors in NF1.
Collapse
Affiliation(s)
- Biplab Dasgupta
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | | | | | | | | |
Collapse
|
20
|
Dasgupta B, Li W, Perry A, Gutmann DH. Glioma Formation in Neurofibromatosis 1 Reflects Preferential Activation of K-RAS in Astrocytes. Cancer Res 2005. [DOI: 10.1158/0008-5472.236.65.1] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Abstract
Children with the tumor predisposition syndrome, neurofibromatosis 1 (NF1), develop optic pathway gliomas. The NF1 gene product, neurofibromin, functions as a negative regulator of RAS, such that NF1 inactivation results in RAS hyperactivation. Recent studies have highlighted the divergent biological and biochemical properties of the various RAS isoforms, which prompted us to examine the consequence of Nf1 inactivation in astrocytes on RAS isoform activation in vitro and in vivo. In this report, we show that only K-RAS is activated in Nf1−/− astrocytes and that activation of K-RAS, but not H-RAS, accounts for the proliferative advantage and abnormal actin cytoskeleton–mediated processes observed in Nf1−/− astrocytes in vitro. Moreover, dominant inhibitory K-RAS corrects these abnormalities in Nf1−/− astrocytes invitro. Lastly, we show that Nf1+/− mice with astrocyte-specific activated K-RAS expression in vivo develop optic pathway gliomas, similar to our previously reported Nf1+/− mice with astrocyte Nf1 inactivation. Collectively, our results show that K-RAS is the primary target for neurofibromin GTPase-activating protein activity in vitro and in vivo and that K-RAS activation in astrocytes recapitulates the biochemical, biological, and tumorigenic properties of neurofibromin loss.
Collapse
Affiliation(s)
| | - Wen Li
- 1Neurology and Departments of
| | - Arie Perry
- 2Pathology (Neuropathology), Washington University School of Medicine, St. Louis, Missouri
| | | |
Collapse
|
21
|
Fieber LA. Voltage-Gated ion currents of schwann cells in cell culture models of human neurofibromatosis. ACTA ACUST UNITED AC 2004; 300:76-83. [PMID: 14598389 DOI: 10.1002/jez.a.10312] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
K(+) (K) channels play a role in the proliferation of many cell types in normal cells and certain disease states. Several laboratories have studied K currents in cultured Schwann cells from models of the human diseases, neurofibromatosis type 1 (NF1) and neurofibromatosis type 2 (NF2). These diseases are characterized by the growth of Schwann cell tumors. In all cell culture NF models the K current properties differ in tumor-derived and normal Schwann cells. Depending on the model however, the type of K channel abnormality differs. K channels appear to play a role in the proliferation of Schwann cell cultures of these disease models, because a link has been established between K current blockade and the inhibition of Schwann cell proliferation in NF1 and NF2. Differences in the proliferation response of normal Schwann cells to K channel blockers suggest that in vitro regulation of proliferation in neoplastic and normal Schwann cells is complex.
Collapse
Affiliation(s)
- Lynne A Fieber
- Division of Marine Biology and Fisheries, University of Miami Rosentiel School, Miami, Florida, 33149, USA.
| |
Collapse
|