1
|
Richards JR, Shin D, Pryor R, Sorensen LK, Sun Z, So WM, Park G, Wolff R, Truong A, McMahon M, Grossmann AH, Harbour JW, Zhu W, Odelberg SJ, Yoo JH. Activation of NFAT by HGF and IGF-1 via ARF6 and its effector ASAP1 promotes uveal melanoma metastasis. Oncogene 2023; 42:2629-2640. [PMID: 37500798 PMCID: PMC11008337 DOI: 10.1038/s41388-023-02792-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 07/12/2023] [Accepted: 07/19/2023] [Indexed: 07/29/2023]
Abstract
Preventing or effectively treating metastatic uveal melanoma (UM) is critical because it occurs in about half of patients and confers a very poor prognosis. There is emerging evidence that hepatocyte growth factor (HGF) and insulin-like growth factor 1 (IGF-1) promote metastasis and contribute to the striking metastatic hepatotropism observed in UM metastasis. However, the molecular mechanisms by which HGF and IGF-1 promote UM liver metastasis have not been elucidated. ASAP1, which acts as an effector for the small GTPase ARF6, is highly expressed in the subset of uveal melanomas most likely to metastasize. Here, we found that HGF and IGF-1 hyperactivate ARF6, leading to its interaction with ASAP1, which then acts as an effector to induce nuclear localization and transcriptional activity of NFAT1. Inhibition of any component of this pathway impairs cellular invasiveness. Additionally, knocking down ASAP1 or inhibiting NFAT signaling reduces metastasis in a xenograft mouse model of UM. The discovery of this signaling pathway represents not only an advancement in our understanding of the biology of uveal melanoma metastasis but also identifies a novel pathway that could be targeted to treat or prevent metastatic uveal melanoma.
Collapse
Affiliation(s)
- Jackson R Richards
- Department of Oncological Sciences, School of Medicine, University of Utah, 2000 Circle of Hope Drive, Salt Lake City, UT, 84112, USA
- Department of Medicine, Program in Molecular Medicine, University of Utah, 15 North 2030 East, Salt Lake City, UT, 84112, USA
- Department of Psychiatry, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Donghan Shin
- Department of Medicine, Program in Molecular Medicine, University of Utah, 15 North 2030 East, Salt Lake City, UT, 84112, USA
| | - Rob Pryor
- Department of Medicine, Program in Molecular Medicine, University of Utah, 15 North 2030 East, Salt Lake City, UT, 84112, USA
| | - Lise K Sorensen
- Department of Medicine, Program in Molecular Medicine, University of Utah, 15 North 2030 East, Salt Lake City, UT, 84112, USA
| | - Zhonglou Sun
- Department of Medicine, Program in Molecular Medicine, University of Utah, 15 North 2030 East, Salt Lake City, UT, 84112, USA
| | - Won Mi So
- Department of Ophthalmology & Visual Sciences, Truhlsen Eye Institute, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Garam Park
- Department of Ophthalmology & Visual Sciences, Truhlsen Eye Institute, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Roger Wolff
- Department of Pathology, University of Utah, 15 North Medical Drive East, Salt Lake City, UT, 84112, USA
- Huntsman Cancer Institute, University of Utah Health Sciences Center, 2000 Circle of Hope Drive, Salt Lake City, UT, 84112, USA
| | - Amanda Truong
- Department of Oncological Sciences, School of Medicine, University of Utah, 2000 Circle of Hope Drive, Salt Lake City, UT, 84112, USA
- Huntsman Cancer Institute, University of Utah Health Sciences Center, 2000 Circle of Hope Drive, Salt Lake City, UT, 84112, USA
| | - Martin McMahon
- Department of Oncological Sciences, School of Medicine, University of Utah, 2000 Circle of Hope Drive, Salt Lake City, UT, 84112, USA
- Huntsman Cancer Institute, University of Utah Health Sciences Center, 2000 Circle of Hope Drive, Salt Lake City, UT, 84112, USA
- Department of Dermatology, University of Utah, 30 N 1900 E, Salt Lake City, UT, 84132, USA
| | - Allie H Grossmann
- Department of Pathology, University of Utah, 15 North Medical Drive East, Salt Lake City, UT, 84112, USA
- Huntsman Cancer Institute, University of Utah Health Sciences Center, 2000 Circle of Hope Drive, Salt Lake City, UT, 84112, USA
- ARUP Laboratories, University of Utah, 500 Chipeta Way, Salt Lake City, UT, 84112, USA
| | - J William Harbour
- Department of Ophthalmology, Harold C. Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Weiquan Zhu
- Department of Medicine, Program in Molecular Medicine, University of Utah, 15 North 2030 East, Salt Lake City, UT, 84112, USA
- Division of Cardiovascular Medicine, Department of Medicine, University of Utah, 30 North 1900 East, Salt Lake City, UT, 84132, USA
| | - Shannon J Odelberg
- Department of Medicine, Program in Molecular Medicine, University of Utah, 15 North 2030 East, Salt Lake City, UT, 84112, USA.
- Division of Cardiovascular Medicine, Department of Medicine, University of Utah, 30 North 1900 East, Salt Lake City, UT, 84132, USA.
- Department of Neurobiology, University of Utah, 20 South 2030 East, Salt Lake City, UT, 84112, USA.
| | - Jae Hyuk Yoo
- Department of Ophthalmology & Visual Sciences, Truhlsen Eye Institute, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
2
|
Mou K, Zhou Y, Mu X, Zhang J, Wang L, Ge R. PARP1 Is a Prognostic Marker and Targets NFATc2 to Promote Carcinogenesis in Melanoma. Genet Test Mol Biomarkers 2022; 26:503-511. [DOI: 10.1089/gtmb.2021.0214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Affiliation(s)
- Kuanhou Mou
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, P.R. China
| | - Yan Zhou
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, P.R. China
| | - Xin Mu
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, P.R. China
| | - Jian Zhang
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, P.R. China
| | - Lijuan Wang
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, P.R. China
| | - Rui Ge
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, P.R. China
| |
Collapse
|
3
|
Hsieh CH, Huang YW, Tsai TF. Oral Conventional Synthetic Disease-Modifying Antirheumatic Drugs with Antineoplastic Potential: a Review. Dermatol Ther (Heidelb) 2022; 12:835-860. [PMID: 35381976 PMCID: PMC9021342 DOI: 10.1007/s13555-022-00713-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Indexed: 01/17/2023] Open
Abstract
There is an increasing trend of malignancy worldwide. Disease-modifying antirheumatic drugs (DMARDs) are the cornerstones for the treatment of immune-mediated inflammatory diseases (IMIDs), but risk of malignancy is a major concern for patients receiving DMARDs. In addition, many IMIDs already carry higher background risks of neoplasms. Recently, the black box warning of malignancies has been added for Janus kinase inhibitors. Also, the use of biologic DMARDs in patients with established malignancies is usually discouraged owing to exclusion of such patients in pivotal studies and, hence, lack of evidence. In contrast, some conventional synthetic DMARDs (csDMARDs) have been reported to show antineoplastic properties and can be beneficial for patients with cancer. Among the csDMARDs, chloroquine and hydroxychloroquine have been the most extensively studied, and methotrexate is an established chemotherapeutic agent. Even cyclosporine A, a well-known drug associated with cancer risk, can potentiate the effect of some chemotherapeutic agents. We review the possible mechanisms behind and clinical evidence of the antineoplastic activities of csDMARDs, including chloroquine and hydroxychloroquine, cyclosporine, leflunomide, mycophenolate mofetil, mycophenolic acid, methotrexate, sulfasalazine, and thiopurines. This knowledge may guide physicians in the choice of csDMARDs for patients with concurrent IMIDs and malignancies.
Collapse
Affiliation(s)
- Cho-Hsun Hsieh
- Department of Medical Education, National Taiwan University Hospital, Taipei, Taiwan
| | - Yi-Wei Huang
- Department of Dermatology, National Taiwan University Hospital, 7 Chung Shan S Rd, Taipei, 10048, Taiwan
| | - Tsen-Fang Tsai
- Department of Dermatology, National Taiwan University Hospital, 7 Chung Shan S Rd, Taipei, 10048, Taiwan. .,Department of Dermatology, National Taiwan University Hospital & National Taiwan University College of Medicine, Taipei, Taiwan.
| |
Collapse
|
4
|
Understanding Molecular Mechanisms of Phenotype Switching and Crosstalk with TME to Reveal New Vulnerabilities of Melanoma. Cells 2022; 11:cells11071157. [PMID: 35406721 PMCID: PMC8997563 DOI: 10.3390/cells11071157] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/20/2022] [Accepted: 03/21/2022] [Indexed: 12/15/2022] Open
Abstract
Melanoma cells are notorious for their high plasticity and ability to switch back and forth between various melanoma cell states, enabling the adaptation to sub-optimal conditions and therapeutics. This phenotypic plasticity, which has gained more attention in cancer research, is proposed as a new paradigm for melanoma progression. In this review, we provide a detailed and deep comprehensive recapitulation of the complex spectrum of phenotype switching in melanoma, the key regulator factors, the various and new melanoma states, and corresponding signatures. We also present an extensive description of the role of epigenetic modifications (chromatin remodeling, methylation, and activities of long non-coding RNAs/miRNAs) and metabolic rewiring in the dynamic switch. Furthermore, we elucidate the main role of the crosstalk between the tumor microenvironment (TME) and oxidative stress in the regulation of the phenotype switching. Finally, we discuss in detail several rational therapeutic approaches, such as exploiting phenotype-specific and metabolic vulnerabilities and targeting components and signals of the TME, to improve the response of melanoma patients to treatments.
Collapse
|
5
|
薛 晓, 李 忠, 赵 明. [Metformin and lipopolysaccharide regulate transcription of NFATc2 gene via the transcription factor RUNX2]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2022; 42:425-431. [PMID: 35426808 PMCID: PMC9010990 DOI: 10.12122/j.issn.1673-4254.2022.03.16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Indexed: 06/14/2023]
Abstract
OBJECTIVE To construct a luciferase reporter gene vector carrying human nuclear factor of activated T cells 2 (NFATc2) gene promoter and examine the effects of metformin and lipopolysaccharide (LPS) on the transcriptional activity of NFATc2 gene. METHODS The promoter sequence of human NFATc2 gene was acquired from UCSC website for PCR amplification. NFATc2 promoter fragment was inserted into pGL3-basic plasmid double cleaved with Kpn Ⅰ and Hind Ⅲ. The resultant recombinant plasmid pGL3-NFATC2-promoter was co-transfected with the internal reference plasmid pRL-TK in 293F cells, and luciferase activity in the cells was detected. Reporter gene vectors of human NFATc2 gene promoter with different fragment lengths were also constructed and assayed for luciferase activity. The changes in transcription activity of NFATc2 gene were assessed after treatment with different concentrations of metformin and LPS for 24 h. We also examined the effect of mutation in RUNX2-binding site in NFATC2 gene promoter on the regulatory effects of metformin and LPS on NFATc2 transcription. RESULTS We successfully constructed pGL3-NFATc2-promoter plasmids carrying different lengths (2170 bp, 2077 bp, 1802 bp, 1651 bp, 1083 bp, 323 bp) of NFATc2 promoter sequences as verified by enzymatic digestion and sequencing. Transfection of 293F cells with the plasmid carrying a 1651 bp NFATc2 promoter (pGL3-1651 bp) resulted in the highest transcriptional activity of NFATc2 gene, and the luciferase activity was approximately 3.3 times that of pGL3-2170 bp (1.843 ± 0.146 vs 0.547 ± 0.085). Moderate (5 mmol/L) and high (10 mmol/L) concentrations of metformin significantly upregulated the transcriptional activity of pGL3-1651 bp by up to 2.5 and 3 folds, respectively. LPS at different doses also upregulated the transcriptional activity of pGL3-1651 bp by at least 1.6 folds. The mutation in the RUNX2 binding site on pGL3-1651 bp obviously reduced metformin- and LPS-induced enhancement of pGL3-1651bp transcription by 1.7 and 2 folds, respectively. CONCLUSION pGL3-NFATc2-promoter can be transcribed and activated in 293F cells, and LPS and metformin can activate the transcription of pGL3- NFATc2-promoter in a RUNX2-dependent manner.
Collapse
Affiliation(s)
- 晓阳 薛
- 南方医科大学第二临床医学院,广东 广州 510515Second School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China
| | - 忠豪 李
- 广东省医学休克微循环重点实验室,南方医科大学基础医学院病理生理学教研室,广东 广州 510515Key Lab of Medical Shock and Microcirculation Research of Guangdong Province, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - 明 赵
- 广东省医学休克微循环重点实验室,南方医科大学基础医学院病理生理学教研室,广东 广州 510515Key Lab of Medical Shock and Microcirculation Research of Guangdong Province, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
6
|
A Novel Prognostic Four-Gene Signature of Breast Cancer Identified by Integrated Bioinformatics Analysis. DISEASE MARKERS 2022; 2022:5925982. [PMID: 35265226 PMCID: PMC8898848 DOI: 10.1155/2022/5925982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 12/20/2021] [Accepted: 12/27/2021] [Indexed: 11/20/2022]
Abstract
Molecular analysis facilitates the prediction of overall survival (OS) of breast cancer and decision-making of the treatment plan. The current study was designed to identify new prognostic genes for breast cancer and construct an effective prognostic signature with integrated bioinformatics analysis. Differentially expressed genes in breast cancer samples from The Cancer Genome Atlas (TCGA) dataset were filtered by univariate Cox regression analysis. The prognostic model was optimized by the Akaike information criterion and further validated using the TCGA dataset (n = 1014) and Gene Expression Omnibus (GEO) dataset (n = 307). The correlation between the risk score and clinical information was assessed by univariate and multivariate Cox regression analyses. Functional pathways in relation to high-risk and low-risk groups were analyzed using gene set enrichment analysis (GSEA). Four prognostic genes (EXOC6, GPC6, PCK2, and NFATC2) were screened and used to construct a prognostic model, which showed robust performance in classifying the high-risk and low-risk groups. The risk score was significantly related to clinical features and OS. We identified 19 functional pathways significantly associated with the risk score. This study constructed a new prognostic model with a high prediction performance for breast cancer. The four-gene prognostic signature could serve as an effective tool to predict prognosis and assist the management of breast cancer patients.
Collapse
|
7
|
Rohini M, Vairamani M, Selvamurugan N. TGF-β1-stimulation of NFATC2 and ATF3 proteins and their interaction for matrix metalloproteinase 13 expression in human breast cancer cells. Int J Biol Macromol 2021; 192:1325-1330. [PMID: 34687766 DOI: 10.1016/j.ijbiomac.2021.10.099] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 09/30/2021] [Accepted: 10/14/2021] [Indexed: 01/25/2023]
Abstract
Activating transcription factor 3 (ATF3), an inducible stress gene, is stimulated by transforming growth factor-beta1 (TGF-β1) in a protracted and relentless manner in human mammary cancer cells (hBC cells; MDA-MB231). The molecular mechanism behind this stable expression of ATF3 via TGF-β1 in MDA-MB231 cells is unknown. This study found that TGF-β1 stimulated the expression of the nuclear factor of activated T Cells 2 (NFATC2) in MDA-MB231 cells and provided evidence of its interaction with ATF3. The functional characterization of NFATC2 in association with ATF3 was determined by silencing of NFATC2 using siRNA. Knock-down of NFATC2 decreased the expression of both ATF3 and its target gene MMP13 (matrix metalloproteinase 13, a critical invasive gene) in hBC cells. Chromatin immunoprecipitation revealed that TGF-β1 promoted NFATC2 binding and NFATC2-ATF3 complex binding at the MMP13 promoter region, whereas silencing of NFATC2 decreased their binding in hBC cells. Thus, we uncovered the mechanism of interaction between NFATC2 and ATF3 regulated by TGF-β1, and NFATC2 acted as a pivotal factor in providing ATF3 stability and further drove MMP13 transcription. Targeting NFATC2 and blocking its association with ATF3 could therefore help to slow the progression of breast cancer.
Collapse
Affiliation(s)
- M Rohini
- Department of Biotechnology, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - M Vairamani
- Department of Biotechnology, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - N Selvamurugan
- Department of Biotechnology, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India.
| |
Collapse
|
8
|
Arslanbaeva LR, Santoro MM. Adaptive redox homeostasis in cutaneous melanoma. Redox Biol 2020; 37:101753. [PMID: 33091721 PMCID: PMC7578258 DOI: 10.1016/j.redox.2020.101753] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/16/2020] [Accepted: 10/05/2020] [Indexed: 02/07/2023] Open
Abstract
Cutaneous melanoma is the most aggressive type of skin cancer. Although cutaneous melanoma accounts for a minority of all types of skin cancer, it causes the greatest number of skin cancer related deaths worldwide. Oxidative stress and redox homeostasis have been shown to be involved at each stage of a malignant melanocyte transformation, called melanomagenesis, as well as during drug resistance. Reactive oxygen species (ROS) play an important and diverse role that regulate many aspects of skin cell behaviors ranging from proliferation and stemness, to oxidative damage and cell death. On the other hand, antioxidants are associated with melanoma spread and metastasis. Overall, the contribution of redox homeostasis to melanoma development and progression is controversial and highly complex. The aim of this study is to examine the association between redox homeostasis and the melanomagenic process. To this purpose we are presenting what is currently known about the role of ROS in melanoma initiation and progression. In addition, we are discussing the role of antioxidant mechanisms during the spread of the disease and in cases of melanoma drug resistance. Although challenging, targeting redox homeostasis in melanoma progression remains to be a promising therapeutic approach, especially valid during melanoma drug resistance.
Collapse
Affiliation(s)
| | - Massimo M Santoro
- Department of Biology, University of Padua, 35131, Italy; Veneto Institute of Molecular Medicine (VIMM), Via Orus 2, 35129, Padua, Italy.
| |
Collapse
|
9
|
Zhang X, Gibhardt CS, Will T, Stanisz H, Körbel C, Mitkovski M, Stejerean I, Cappello S, Pacheu‐Grau D, Dudek J, Tahbaz N, Mina L, Simmen T, Laschke MW, Menger MD, Schön MP, Helms V, Niemeyer BA, Rehling P, Vultur A, Bogeski I. Redox signals at the ER-mitochondria interface control melanoma progression. EMBO J 2019; 38:e100871. [PMID: 31304984 PMCID: PMC6669928 DOI: 10.15252/embj.2018100871] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 05/21/2019] [Accepted: 05/23/2019] [Indexed: 12/20/2022] Open
Abstract
Reactive oxygen species (ROS) are emerging as important regulators of cancer growth and metastatic spread. However, how cells integrate redox signals to affect cancer progression is not fully understood. Mitochondria are cellular redox hubs, which are highly regulated by interactions with neighboring organelles. Here, we investigated how ROS at the endoplasmic reticulum (ER)-mitochondria interface are generated and translated to affect melanoma outcome. We show that TMX1 and TMX3 oxidoreductases, which promote ER-mitochondria communication, are upregulated in melanoma cells and patient samples. TMX knockdown altered mitochondrial organization, enhanced bioenergetics, and elevated mitochondrial- and NOX4-derived ROS. The TMX-knockdown-induced oxidative stress suppressed melanoma proliferation, migration, and xenograft tumor growth by inhibiting NFAT1. Furthermore, we identified NFAT1-positive and NFAT1-negative melanoma subgroups, wherein NFAT1 expression correlates with melanoma stage and metastatic potential. Integrative bioinformatics revealed that genes coding for mitochondrial- and redox-related proteins are under NFAT1 control and indicated that TMX1, TMX3, and NFAT1 are associated with poor disease outcome. Our study unravels a novel redox-controlled ER-mitochondria-NFAT1 signaling loop that regulates melanoma pathobiology and provides biomarkers indicative of aggressive disease.
Collapse
Affiliation(s)
- Xin Zhang
- Molecular PhysiologyInstitute of Cardiovascular PhysiologyUniversity Medical CenterGeorg‐August‐UniversityGöttingenGermany
- BiophysicsCIPMMSaarland UniversityHomburgGermany
| | - Christine S Gibhardt
- Molecular PhysiologyInstitute of Cardiovascular PhysiologyUniversity Medical CenterGeorg‐August‐UniversityGöttingenGermany
| | - Thorsten Will
- Center for BioinformaticsSaarland UniversitySaarbrückenGermany
| | - Hedwig Stanisz
- Department of Dermatology, Venereology and AllergologyUniversity Medical CenterGeorg‐August‐UniversityGöttingenGermany
| | - Christina Körbel
- Institute for Clinical and Experimental SurgerySaarland UniversityHomburgGermany
| | - Miso Mitkovski
- Light Microscopy FacilityMax Planck Institute for Experimental MedicineGöttingenGermany
| | - Ioana Stejerean
- Molecular PhysiologyInstitute of Cardiovascular PhysiologyUniversity Medical CenterGeorg‐August‐UniversityGöttingenGermany
| | - Sabrina Cappello
- Molecular PhysiologyInstitute of Cardiovascular PhysiologyUniversity Medical CenterGeorg‐August‐UniversityGöttingenGermany
| | - David Pacheu‐Grau
- Department of Cellular BiochemistryUniversity Medical CenterGeorg‐August‐UniversityGöttingenGermany
| | - Jan Dudek
- Department of Cellular BiochemistryUniversity Medical CenterGeorg‐August‐UniversityGöttingenGermany
| | - Nasser Tahbaz
- Department of Cell BiologyUniversity of AlbertaEdmontonABCanada
| | - Lucas Mina
- Department of Cell BiologyUniversity of AlbertaEdmontonABCanada
| | - Thomas Simmen
- Department of Cell BiologyUniversity of AlbertaEdmontonABCanada
| | - Matthias W Laschke
- Institute for Clinical and Experimental SurgerySaarland UniversityHomburgGermany
| | - Michael D Menger
- Institute for Clinical and Experimental SurgerySaarland UniversityHomburgGermany
| | - Michael P Schön
- Department of Dermatology, Venereology and AllergologyUniversity Medical CenterGeorg‐August‐UniversityGöttingenGermany
| | - Volkhard Helms
- Center for BioinformaticsSaarland UniversitySaarbrückenGermany
| | | | - Peter Rehling
- Department of Cellular BiochemistryUniversity Medical CenterGeorg‐August‐UniversityGöttingenGermany
- Max Planck Institute for Biophysical ChemistryGöttingenGermany
| | - Adina Vultur
- Molecular PhysiologyInstitute of Cardiovascular PhysiologyUniversity Medical CenterGeorg‐August‐UniversityGöttingenGermany
| | - Ivan Bogeski
- Molecular PhysiologyInstitute of Cardiovascular PhysiologyUniversity Medical CenterGeorg‐August‐UniversityGöttingenGermany
- BiophysicsCIPMMSaarland UniversityHomburgGermany
| |
Collapse
|
10
|
Hamdan FH, Johnsen SA. Perturbing Enhancer Activity in Cancer Therapy. Cancers (Basel) 2019; 11:cancers11050634. [PMID: 31067678 PMCID: PMC6563029 DOI: 10.3390/cancers11050634] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 04/26/2019] [Accepted: 05/05/2019] [Indexed: 02/07/2023] Open
Abstract
Tight regulation of gene transcription is essential for normal development, tissue homeostasis, and disease-free survival. Enhancers are distal regulatory elements in the genome that provide specificity to gene expression programs and are frequently misregulated in cancer. Recent studies examined various enhancer-driven malignant dependencies and identified different approaches to specifically target these programs. In this review, we describe numerous features that make enhancers good transcriptional targets in cancer therapy and discuss different approaches to overcome enhancer perturbation. Interestingly, a number of approved therapeutic agents, such as cyclosporine, steroid hormones, and thiazolidinediones, actually function by affecting enhancer landscapes by directly targeting very specific transcription factor programs. More recently, a broader approach to targeting deregulated enhancer programs has been achieved via Bromodomain and Extraterminal (BET) inhibition or perturbation of transcription-related cyclin-dependent kinases (CDK). One challenge to enhancer-targeted therapy is proper patient stratification. We suggest that monitoring of enhancer RNA (eRNA) expression may serve as a unique biomarker of enhancer activity that can help to predict and monitor responsiveness to enhancer-targeted therapies. A more thorough investigation of cancer-specific enhancers and the underlying mechanisms of deregulation will pave the road for an effective utilization of enhancer modulators in a precision oncology approach to cancer treatment.
Collapse
Affiliation(s)
- Feda H Hamdan
- Gene Regulatory Mechanisms and Molecular Epigenetics Lab, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905, USA.
| | - Steven A Johnsen
- Gene Regulatory Mechanisms and Molecular Epigenetics Lab, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|
11
|
Liu Z, Jiang L, Li Y, Xie B, Xie J, Wang Z, Zhou X, Jiang H, Fang Y, Pan H, Han W. Cyclosporine A sensitizes lung cancer cells to crizotinib through inhibition of the Ca2 +/calcineurin/Erk pathway. EBioMedicine 2019; 42:326-339. [PMID: 30879923 PMCID: PMC6491942 DOI: 10.1016/j.ebiom.2019.03.019] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 03/03/2019] [Accepted: 03/07/2019] [Indexed: 12/27/2022] Open
Abstract
Background Crizotinib has potent anti-tumor activity in patients with advanced MET-amplified non-small cell lung cancer (NSCLC). However, the therapeutic effect is still not satisfying. Thus, developing approaches that improve the efficacy of crizotinib remains a significant challenge. Methods MET-amplified NSCLC cell lines were treated with crizotinib and cyclosporine A (CsA). Cell viability was determined by MTS assay. The changes of apoptosis, cell cycle and calcineurin-Erk pathways were assessed by western blot. Xenograft mouse model, primary human NSCLC cells and hollow fiber assays were utilized to confirm the effects of CsA. Findings We demonstrated that CsA significantly increased the anti-tumor effect of crizotinib on multiple MET-amplified NSCLC cells in vitro and in vivo. Mechanistically, crizotinib treatment led to the activation of Ca2+-calcineurin (CaN)-Kinase suppressor of Ras 2 (KSR2) signaling, resulting in Erk1/2 activation and enhanced survival of cancer cells. CsA effectively blocked CaN-KSR2-Erk1/2 signaling, promoting crizotinib-induced apoptosis and G2/M arrest. Similarly, pharmacologic or genetic inhibition of Erk1/2 also enhanced crizotinib-induced growth inhibition in vitro. Xenograft studies further confirmed that CsA or Erk1/2 inhibitor PD98059 enhanced the anti-cancer activity of crizotinib through inhibition of CaN-Erk1/2 axis. The results were also validated by primary human NSCLC cells in vitro and hollow fiber assays in vivo. Interpretation This study provides preclinical evidences that combination therapy of CsA and crizotinib is a promising approach for targeted treatment of MET-amplified lung cancer patients. Fund This work was supported by the National Natural Science Foundation of China, the Key Projects of Natural Foundation of Zhejiang Province, the Ten thousand plan youth talent support program of Zhejiang Province, the Zhejiang Natural Sciences Foundation Grant, and the Zhejiang medical innovative discipline construction project-2016.
Collapse
Affiliation(s)
- Zhen Liu
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Zhejiang, Hangzhou, China
| | - Liming Jiang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Zhejiang, Hangzhou, China
| | - Yiran Li
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Zhejiang, Hangzhou, China
| | - Binbin Xie
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Zhejiang, Hangzhou, China
| | - Jiansheng Xie
- Laboratory of Cancer Biology, Institute of Clinical Science, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Zhejiang, Hangzhou, China
| | - Zhanggui Wang
- Department of Radiotherapy, The Second People's Hospital of Anhui Province, Hefei, Anhui, China
| | - Xiaoyun Zhou
- Department of Medical Oncology, Xiasha Branch of Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Zhejiang, Hangzhou, China
| | - Hanliang Jiang
- Department of Respiratory Medicine, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Zhejiang, Hangzhou, China
| | - Yong Fang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Zhejiang, Hangzhou, China
| | - Hongming Pan
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Zhejiang, Hangzhou, China.
| | - Weidong Han
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Zhejiang, Hangzhou, China; Laboratory of Cancer Biology, Institute of Clinical Science, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Zhejiang, Hangzhou, China.
| |
Collapse
|
12
|
Perotti V, Baldassari P, Molla A, Nicolini G, Bersani I, Grazia G, Benigni F, Maurichi A, Santinami M, Anichini A, Mortarini R. An actionable axis linking NFATc2 to EZH2 controls the EMT-like program of melanoma cells. Oncogene 2019; 38:4384-4396. [PMID: 30710146 PMCID: PMC6756060 DOI: 10.1038/s41388-019-0729-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 12/03/2018] [Accepted: 01/21/2019] [Indexed: 01/30/2023]
Abstract
Discovery of new actionable targets and functional networks in melanoma is an urgent need as only a fraction of metastatic patients achieves durable clinical benefit by targeted therapy or immunotherapy approaches. Here we show that NFATc2 expression is associated with an EMT-like transcriptional program and with an invasive melanoma phenotype, as shown by analysis of melanoma cell lines at the mRNA and protein levels, interrogation of the TCGA melanoma dataset and characterization of melanoma lesions by immunohistochemistry. Gene silencing or pharmacological inhibition of NFATc2 downregulated EMT-related genes and AXL, and suppressed c-Myc, FOXM1, and EZH2. Targeting of c-Myc suppressed FOXM1 and EZH2, while targeting of FOXM1 suppressed EZH2. Inhibition of c-Myc, or FOXM1, or EZH2 downregulated EMT-related gene expression, upregulated MITF and suppressed migratory and invasive activity of neoplastic cells. Stable silencing of NFATc2 impaired melanoma cell proliferation in vitro and tumor growth in vivo in SCID mice. In NFATc2+ EZH2+ melanoma cell lines pharmacological co-targeting of NFATc2 and EZH2 exerted strong anti-proliferative and pro-apoptotic activity, irrespective of BRAF or NRAS mutations and of BRAF inhibitor resistance. These results provide preclinical evidence for a role of NFATc2 in shaping the EMT-like melanoma phenotype and reveal a targetable vulnerability associated with NFATc2 and EZH2 expression in melanoma cells belonging to different mutational subsets.
Collapse
Affiliation(s)
- Valentina Perotti
- Department of Research, Human Tumors Immunobiology Unit, Milan, Italy
| | - Paola Baldassari
- Department of Research, Human Tumors Immunobiology Unit, Milan, Italy
| | - Alessandra Molla
- Department of Research, Human Tumors Immunobiology Unit, Milan, Italy
| | | | - Ilaria Bersani
- Department of Research, Human Tumors Immunobiology Unit, Milan, Italy
| | - Giulia Grazia
- Department of Research, Human Tumors Immunobiology Unit, Milan, Italy
| | - Fabio Benigni
- HuMabs Biomed, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Andrea Maurichi
- Melanoma and Sarcoma Unit, Department of Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Mario Santinami
- Melanoma and Sarcoma Unit, Department of Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Andrea Anichini
- Department of Research, Human Tumors Immunobiology Unit, Milan, Italy
| | - Roberta Mortarini
- Department of Research, Human Tumors Immunobiology Unit, Milan, Italy.
| |
Collapse
|
13
|
Liu H, Yang L, Qi M, Zhang J. NFAT1 enhances the effects of tumor-associated macrophages on promoting malignant melanoma growth and metastasis. Biosci Rep 2018; 38:BSR20181604. [PMID: 30459241 PMCID: PMC6435508 DOI: 10.1042/bsr20181604] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 10/31/2018] [Accepted: 11/12/2018] [Indexed: 12/24/2022] Open
Abstract
Tumor-associated macrophages (TAMs) play substantial roles in tumor growth, invasion, and metastasis. Nuclear factor of activated T cell (NFAT1) has been shown to promote melanoma growth and metastasis in vivo We herein aim to investigate whether NFAT1 is capable to promote melanoma growth and metastasis by influencing TAM properties. Melanoma-conditioned TAMs were obtained from human monocytes after incubation with conditioned medium from A375 cell culture. The phenotype of the macrophages was detected. Cell proliferation, migration, and invasion were evaluated. Human malignant melanoma tissues exhibited increased CD68+-macrophage infiltration and NFAT1 expression compared with the normal pigmented nevus tissues. Melanoma-conditioned TAMs displayed M2-like phenotype. Melanoma-conditioned TAMs also promoted proliferation, migration, and invasion of human malignant melanoma cell lines A375 and WM451. Furthermore, NFAT1 expression in TAMs was significantly increased compared with the M0 group. NFAT1 overexpression significantly strengthened the melanoma-conditioned TAM-mediated promotion of cell migration and invasion in A375 and WM451 cells, whereas NFAT1 knockdown exerted the opposite effects. Moreover, NFAT1 overexpression in melanoma-conditioned TAMs promoted CD68+-macrophage infiltration, tumor growth, and metastasis in vivo NFAT1 may play a critical role in enhancing the TAM-mediated promotion of growth and metastasis in malignant melanoma.
Collapse
Affiliation(s)
- Hao Liu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan Province 410008, P.R. China
| | - Liping Yang
- College of Life Sciences, Hunan Normal University, Changsha, Hunan Province 410006, P.R. China
| | - Min Qi
- Department of Plastic and Cosmetic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan Province 410008, P.R. China
| | - Jianglin Zhang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan Province 410008, P.R. China
| |
Collapse
|
14
|
Fane ME, Chhabra Y, Smith AG, Sturm RA. BRN2, a POUerful driver of melanoma phenotype switching and metastasis. Pigment Cell Melanoma Res 2018; 32:9-24. [PMID: 29781575 DOI: 10.1111/pcmr.12710] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 04/18/2018] [Accepted: 04/25/2018] [Indexed: 12/30/2022]
Abstract
The POU domain family of transcription factors play a central role in embryogenesis and are highly expressed in neural crest cells and the developing brain. BRN2 is a class III POU domain protein that is a key mediator of neuroendocrine and melanocytic development and differentiation. While BRN2 is a central regulator in numerous developmental programs, it has also emerged as a major player in the biology of tumourigenesis. In melanoma, BRN2 has been implicated as one of the master regulators of the acquisition of invasive behaviour within the phenotype switching model of progression. As a mediator of melanoma cell phenotype switching, it coordinates the transition to a dedifferentiated, slow cycling and highly motile cell type. Its inverse expression relationship with MITF is believed to mediate tumour progression and metastasis within this model. Recent evidence has now outlined a potential epigenetic switching mechanism in melanoma cells driven by BRN2 expression that induces melanoma cell invasion. We summarize the role of BRN2 in tumour cell dissemination and metastasis in melanoma, while also examining it as a potential metastatic regulator in other tumour models.
Collapse
Affiliation(s)
- Mitchell E Fane
- School of Biomedical Sciences, Institute of Health and Biomedical Innovation at the Translational Research Institute, Queensland University of Technology, Brisbane, QLD, Australia.,Dermatology Research Centre, UQ Diamantina Institute, Translational Research Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Yash Chhabra
- School of Biomedical Sciences, Institute of Health and Biomedical Innovation at the Translational Research Institute, Queensland University of Technology, Brisbane, QLD, Australia.,Dermatology Research Centre, UQ Diamantina Institute, Translational Research Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Aaron G Smith
- School of Biomedical Sciences, Institute of Health and Biomedical Innovation at the Translational Research Institute, Queensland University of Technology, Brisbane, QLD, Australia
| | - Richard A Sturm
- Dermatology Research Centre, UQ Diamantina Institute, Translational Research Institute, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
15
|
Downregulated TRPV1 Expression Contributes to Melanoma Growth via the Calcineurin-ATF3-p53 Pathway. J Invest Dermatol 2018; 138:2205-2215. [PMID: 29580868 DOI: 10.1016/j.jid.2018.03.1510] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 03/05/2018] [Accepted: 03/15/2018] [Indexed: 01/22/2023]
Abstract
Melanoma is the most lethal form of skin cancer with increasing incidence over the years. Because of its rapid proliferative and drastic metastatic capacity, the prognosis of melanoma remains dismal, although the targeted therapy and immunotherapy have gained revolutionary progress recently. Therefore, it is of necessity to further clarify the mechanism of melanoma pathogenesis for developing an alternative treatment strategy. Transient receptor potential vanilloid 1 (TRPV1) is a nonselective Ca2+ channel greatly involved in regulating cell apoptosis, proliferation, metabolism, and cancer development, but its role in melanoma remains unknown. Herein, we first found that TRPV1 expression was significantly decreased in melanoma tissues and cell lines, compared with nevus tissues and normal melanocytes, respectively. We then proved that TRPV1 overexpression or its agonist capsaicin treatment inhibited melanoma growth by activating p53 and inducing cell apoptosis. A subsequent mechanistic study revealed that TRPV1 induced Ca2+ influx to regulate p53 activation via calcineurin-ATF3 transcriptional cascade. Finally, the effect of TRPV1 on melanoma growth was proved in vivo. Altogether, our study demonstrates that TRPV1 is a potential tumor suppressor in melanoma.
Collapse
|
16
|
Senavirathna LK, Huang C, Yang X, Munteanu MC, Sathiaseelan R, Xu D, Henke CA, Liu L. Hypoxia induces pulmonary fibroblast proliferation through NFAT signaling. Sci Rep 2018; 8:2709. [PMID: 29426911 PMCID: PMC5807313 DOI: 10.1038/s41598-018-21073-x] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 01/23/2018] [Indexed: 11/09/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive and typically fatal lung disease with a very low survival rate. Excess accumulation of fibroblasts, myofibroblasts and extracellular matrix creates hypoxic conditions within the lungs, causing asphyxiation. Hypoxia is, therefore, one of the prominent features of IPF. However, there have been few studies concerning the effects of hypoxia on pulmonary fibroblasts. In this study, we investigated the molecular mechanisms of hypoxia-induced lung fibroblast proliferation. Hypoxia increased the proliferation of normal human pulmonary fibroblasts and IPF fibroblasts after exposure for 3–6 days. Cell cycle analysis demonstrated that hypoxia promoted the G1/S phase transition. Hypoxia downregulated cyclin D1 and A2 levels, while it upregulated cyclin E1 protein levels. However, hypoxia had no effect on the protein expression levels of cyclin-dependent kinase 2, 4, and 6. Chemical inhibition of hypoxia-inducible factor (HIF)-2 reduced hypoxia-induced fibroblast proliferation. Moreover, silencing of Nuclear Factor Activated T cell (NFAT) c2 attenuated the hypoxia-mediated fibroblasts proliferation. Hypoxia also induced the nuclear translocation of NFATc2, as determined by immunofluorescence staining. NFAT reporter assays showed that hypoxia-induced NFAT signaling activation is dependent on HIF-2, but not HIF-1. Furthermore, the inhibition or silencing of HIF-2, but not HIF-1, reduced the hypoxia-mediated NFATc2 nuclear translocation. Our studies suggest that hypoxia induces the proliferation of human pulmonary fibroblasts through NFAT signaling and HIF-2.
Collapse
Affiliation(s)
- Lakmini Kumari Senavirathna
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, OK, 74078, USA.,Department of Physiological Sciences, Lundberg-Kienlen Lung Biology and Toxicology Laboratory, Oklahoma State University, Stillwater, OK, 74078, USA
| | - Chaoqun Huang
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, OK, 74078, USA.,Department of Physiological Sciences, Lundberg-Kienlen Lung Biology and Toxicology Laboratory, Oklahoma State University, Stillwater, OK, 74078, USA
| | - Xiaoyun Yang
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, OK, 74078, USA.,Department of Physiological Sciences, Lundberg-Kienlen Lung Biology and Toxicology Laboratory, Oklahoma State University, Stillwater, OK, 74078, USA
| | - Maria Cristina Munteanu
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, OK, 74078, USA.,Department of Physiological Sciences, Lundberg-Kienlen Lung Biology and Toxicology Laboratory, Oklahoma State University, Stillwater, OK, 74078, USA
| | - Roshini Sathiaseelan
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, OK, 74078, USA.,Department of Physiological Sciences, Lundberg-Kienlen Lung Biology and Toxicology Laboratory, Oklahoma State University, Stillwater, OK, 74078, USA
| | - Dao Xu
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, OK, 74078, USA.,Department of Physiological Sciences, Lundberg-Kienlen Lung Biology and Toxicology Laboratory, Oklahoma State University, Stillwater, OK, 74078, USA
| | - Craig A Henke
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Lin Liu
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, OK, 74078, USA. .,Department of Physiological Sciences, Lundberg-Kienlen Lung Biology and Toxicology Laboratory, Oklahoma State University, Stillwater, OK, 74078, USA.
| |
Collapse
|
17
|
Marciel MP, Khadka VS, Deng Y, Kilicaslan P, Pham A, Bertino P, Lee K, Chen S, Glibetic N, Hoffmann FW, Matter ML, Hoffmann PR. Selenoprotein K deficiency inhibits melanoma by reducing calcium flux required for tumor growth and metastasis. Oncotarget 2018; 9:13407-13422. [PMID: 29568366 PMCID: PMC5862587 DOI: 10.18632/oncotarget.24388] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Accepted: 01/23/2018] [Indexed: 11/30/2022] Open
Abstract
Interest has emerged in the therapeutic potential of inhibiting store operated calcium (Ca2+) entry (SOCE) for melanoma and other cancers because malignant cells exhibit a strong dependence on Ca2+ flux for disease progression. We investigated the effects of deleting Selenoprotein K (SELENOK) in melanoma since previous work in immune cells showed SELENOK was required for efficient Ca2+ flux through the endoplasmic reticulum Ca2+ channel protein, inositol 1,4,5-trisphosphate receptor (IP3R), which is due to the role SELENOK plays in palmitoylating and stabilizing the expression of IP3R. CRISPR/Cas9 was used to generate SELENOK-deficiency in human melanoma cells and this led to reduced Ca2+ flux and impaired IP3R function, which inhibited cell proliferation, invasion, and migration. Ca2+-dependent signaling through calcineurin was inhibited with SELENOK-deficiency, and gene array analyses together with evaluation of transcript and protein levels showed altered transcriptional programs that ultimately disrupted stemness and pro-growth properties. In vivo investigations were conducted using the Grm1-Tg transgenic mouse strain that develops spontaneous metastatic melanoma, which was crossed with SELENOK−/− mice to generate the following littermates: Grm1-Tg/SELENOK−/−, Grm1-Tg/SELENOK−/+, Grm1-Tg/SELENOK+/+. SELENOK-deficiency in Grm1-Tg/SELENOK−/− male and female mice inhibited primary tumor growth on tails and ears and reduced metastasis to draining lymph nodes down to levels equivalent to non-tumor control mice. Cancer stem cell pools were also decreased in Grm1-Tg/SELENOK−/− mice compared to littermates. These results suggest that melanoma requires SELENOK expression for IP3R dependent maintenance of stemness, tumor growth and metastasic potential, thus revealing a new potential therapeutic target for treating melanoma and possibly other cancers.
Collapse
Affiliation(s)
- Michael P Marciel
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, U.S.A
| | - Vedbar S Khadka
- Bioinformatics Core in the Department of Complementary and Integrative Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, U.S.A
| | - Youping Deng
- Bioinformatics Core in the Department of Complementary and Integrative Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, U.S.A
| | - Pascal Kilicaslan
- Biotechnology Department, University of Applied Sciences Mannheim, Mannheim, Germany
| | - Andrew Pham
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, U.S.A
| | - Pietro Bertino
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, U.S.A
| | - Katie Lee
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, U.S.A
| | - Suzie Chen
- Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey, U.S.A
| | | | - FuKun W Hoffmann
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, U.S.A
| | | | - Peter R Hoffmann
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, U.S.A
| |
Collapse
|
18
|
FGF5 is expressed in melanoma and enhances malignancy in vitro and in vivo. Oncotarget 2017; 8:87750-87762. [PMID: 29152117 PMCID: PMC5675669 DOI: 10.18632/oncotarget.21184] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 08/17/2017] [Indexed: 11/25/2022] Open
Abstract
Although FGF5 mRNA was previously found expressed in some melanoma cell lines in contrast to normal human melanocytes, neither its contribution to melanoma growth nor its expression in melanoma tissue has been investigated. Here we demonstrate that ectopic overexpression of FGF5 in human melanoma cells with low endogenous FGF5 expression increased clonogenicity and invasion but not short-term growth in vitro. Silencing of FGF5 in melanoma cells with high endogenous FGF5 expression had the opposite effect on clonogenicity. FGF overexpression led to increased signaling along the MAPK and NFAT axis but had no effect on STAT3 signaling. In an in vivo experiment in immunocompromised mice, human melanoma xenografts overexpressing FGF5 showed enhanced tumor growth, a higher Ki-67 proliferation index, decreased apoptosis and enhanced angiogenesis. Immunohistochemistry performed on a tissue microarray demonstrated FGF5 protein expression in more than 50% of samples of melanoma and benign nevi. These data suggest that FGF5 has oncogenic potential in melanoma cells and contributes to melanoma growth in a subset of patients. This highlights the importance of further evaluating FGF5 as potential biomarker and therapy target in melanoma.
Collapse
|
19
|
Zhang X, Kang T, Zhang L, Tong Y, Ding W, Chen S. NFATc3 mediates the sensitivity of gastric cancer cells to arsenic sulfide. Oncotarget 2017; 8:52735-52745. [PMID: 28881766 PMCID: PMC5581065 DOI: 10.18632/oncotarget.17175] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 03/03/2017] [Indexed: 12/23/2022] Open
Abstract
Arsenic sulfide (As4S4) is the main component of Realgar which is widely used in traditional Chinese medicine. Previously we showed that As4S4 inhibited the proliferation of colon cancer cells through regulating nuclear factor of activated T cells (NFAT) pathway. Here we explore the role of NFAT in gastric cancer. We showed that As4S4 inhibited the expression of NFATc1, NFATc3, and NFATc4, and modulated the expression of NFATc2 accompanying with p53. The baseline expression of NFATc3 varied distinctly in gastric cancer cell lines (AGS, MGC803, MKN28, MKN45, and SGC7901) and the sensitivity of these cells to As4S4 was dissimilar, with AGS and MGC803 cells showing higher sensitivity while the SGC7901 cells relatively resistant. Interestingly, the sensitivity to As4S4 was correlated with the level of expression of NFATc3, and the cells relatively sensitivity just showing higher expression of NFATc3. Furthermore, NFATc3 expression was significantly higher in gastric cancer tissues compared with the adjacent normal tissues. Our data also showed that, NFATc3 promoted the proliferation of gastric cancer cells by regulating c-Myc. In conclusion, As4S4 inhibited the proliferation of gastric cancer cells through NFATc3/c-Myc pathway and the diverse sensitivity among different cell lines correlated with the expression level of NFATc3 indicating that NFATc3 may be a potential therapeutic target in gastric cancer.
Collapse
Affiliation(s)
- Xiuli Zhang
- Department of Oncology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ting Kang
- Department of Oncology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lian Zhang
- Department of Oncology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yingying Tong
- Department of Oncology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenping Ding
- Department of Oncology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Siyu Chen
- Department of Oncology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
20
|
Regulation of CacyBP/SIP expression by NFAT1 transcription factor. Immunobiology 2017; 222:872-877. [PMID: 28526484 DOI: 10.1016/j.imbio.2017.05.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Accepted: 05/09/2017] [Indexed: 01/11/2023]
Abstract
In this work we have shown that NFAT1 transcription factor is involved in the regulation of CacyBP/SIP expression. We have demonstrated, by applying Western blot, RT-PCR and luciferase assay that the level of CacyBP/SIP increases upon NFAT1 overexpression. Moreover, inhibition or stimulation of NFAT transcriptional activity exerts a corresponding effect on the expression of CacyBP/SIP gene. Furthermore, EMSA and chromatin immunoprecipitation (ChIP) assay have shown that NFAT1 binds to its specific binding sites within the CacyBP/SIP gene. In conclusion, our data have shown for the first time the regulation of CacyBP/SIP gene expression by NFAT1. Since NFAT transcription factors are involved in processes related to immune response, these results indicate potential involvement of CacyBP/SIP in the immune system.
Collapse
|
21
|
Khodeer S, Era T. Identifying the Biphasic Role of Calcineurin/NFAT Signaling Enables Replacement of Sox2 in Somatic Cell Reprogramming. Stem Cells 2017; 35:1162-1175. [DOI: 10.1002/stem.2572] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 12/15/2016] [Accepted: 12/20/2016] [Indexed: 12/28/2022]
Affiliation(s)
- Sherif Khodeer
- Department of Cell Modulation; Institute of Molecular Embryology and Genetics, Kumamoto University; 2-2-1 Honjo Kumamoto Japan
| | - Takumi Era
- Department of Cell Modulation; Institute of Molecular Embryology and Genetics, Kumamoto University; 2-2-1 Honjo Kumamoto Japan
| |
Collapse
|
22
|
Primary cross-resistance to BRAFV600E-, MEK1/2- and PI3K/mTOR-specific inhibitors in BRAF-mutant melanoma cells counteracted by dual pathway blockade. Oncotarget 2016; 7:3947-65. [PMID: 26678033 PMCID: PMC4826182 DOI: 10.18632/oncotarget.6600] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 11/22/2015] [Indexed: 01/09/2023] Open
Abstract
Intrinsic cross-resistance to inhibition of different signaling pathways may hamper development of combinatorial treatments in melanoma, but the relative frequency of this phenotype and the strategies to overcome this hurdle remain poorly understood. Among 49 BRAF-mutant melanoma cell lines from patients not previously treated with target therapy, 21 (42.9%) showed strong primary resistance (IC50 > 1 μM) to a BRAFV600E inhibitor. Most of the BRAF-inhibitor-resistant cell lines showed also strong or intermediate cross-resistance to MEK1/2- and to PI3K/mTOR-specific inhibitors. Primary cross-resistance was confirmed in an independent set of 23 BRAF-mutant short-term melanoma cell cultures. MEK1/2 and PI3K/mTOR co-targeting was the most effective approach, compared to BRAF and PI3K/mTOR dual blockade, to counteract primary resistance to BRAF inhibition and the cross-resistant phenotype. This was shown by extensive drug interaction analysis, tumor growth inhibition assays in-vivo, p-ERK and p-AKT inhibition, promotion of melanoma apoptosis, apoptosis-related protein modulation, activation of effector caspases and selective modulation of genes involved in melanoma drug resistance and belonging to the ERK/MAPK and PI3K/AKT canonical pathways. Compared to co-targeting of mutant BRAF and PI3K/mTOR, the association of a MEK1/2 and a PI3K/mTOR inhibitor was more effective in the activation of Bax and of caspase-3 and in the induction of caspase-dependent melanoma apoptosis. Furthermore Bax silencing reduced the latter effects. These results suggest that intrinsic resistance to BRAF inhibition is frequently associated with primary cross-resistance to MEK and PI3K/mTOR blockade in BRAF-mutant melanoma and provide pre-clinical evidence for a combinatorial approach to counteract this phenotype.
Collapse
|
23
|
Schummer P, Kuphal S, Vardimon L, Bosserhoff AK, Kappelmann M. Specific c-Jun target genes in malignant melanoma. Cancer Biol Ther 2016; 17:486-97. [PMID: 27050748 DOI: 10.1080/15384047.2016.1156264] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
A fundamental event in the development and progression of malignant melanoma is the de-regulation of cancer-relevant transcription factors. We recently showed that c-Jun is a main regulator of melanoma progression and, thus, is the most important member of the AP-1 transcription factor family in this disease. Surprisingly, no cancer-related specific c-Jun target genes in melanoma were described in the literature, so far. Therefore, we focused on pre-existing ChIP-Seq data (Encyclopedia of DNA Elements) of 3 different non-melanoma cell lines to screen direct c-Jun target genes. Here, a specific c-Jun antibody to immunoprecipitate the associated promoter DNA was used. Consequently, we identified 44 direct c-Jun targets and a detailed analysis of 6 selected genes confirmed their deregulation in malignant melanoma. The identified genes were differentially regulated comparing 4 melanoma cell lines and normal human melanocytes and we confirmed their c-Jun dependency. Direct interaction between c-Jun and the promoter/enhancer regions of the identified genes was confirmed by us via ChIP experiments. Interestingly, we revealed that the direct regulation of target gene expression via c-Jun can be independent of the existence of the classical AP-1 (5´-TGA(C/G)TCA-3´) consensus sequence allowing for the subsequent down- or up-regulation of the expression of these cancer-relevant genes. In summary, the results of this study indicate that c-Jun plays a crucial role in the development and progression of malignant melanoma via direct regulation of cancer-relevant target genes and that inhibition of direct c-Jun targets through inhibition of c-Jun is a potential novel therapeutic option for treatment of malignant melanoma.
Collapse
Affiliation(s)
- Patrick Schummer
- a Institute of Biochemistry (Emil-Fischer Center), Friedrich-Alexander University Erlangen-Nürnberg , Erlangen , Germany
| | - Silke Kuphal
- a Institute of Biochemistry (Emil-Fischer Center), Friedrich-Alexander University Erlangen-Nürnberg , Erlangen , Germany
| | - Lily Vardimon
- b Department of Biochemistry and Molecular Biology , Tel Aviv University , Israel
| | - Anja K Bosserhoff
- a Institute of Biochemistry (Emil-Fischer Center), Friedrich-Alexander University Erlangen-Nürnberg , Erlangen , Germany
| | - Melanie Kappelmann
- a Institute of Biochemistry (Emil-Fischer Center), Friedrich-Alexander University Erlangen-Nürnberg , Erlangen , Germany
| |
Collapse
|
24
|
Eriksson J, Le Joncour V, Nummela P, Jahkola T, Virolainen S, Laakkonen P, Saksela O, Hölttä E. Gene expression analyses of primary melanomas reveal CTHRC1 as an important player in melanoma progression. Oncotarget 2016; 7:15065-92. [PMID: 26918341 PMCID: PMC4924771 DOI: 10.18632/oncotarget.7604] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 01/31/2016] [Indexed: 02/04/2023] Open
Abstract
Melanoma is notorious for its high tendency to metastasize and its refractoriness to conventional treatments after metastasis, and the responses to most targeted therapies are short-lived. A better understanding of the molecular mechanisms behind melanoma development and progression is needed to develop more effective therapies and to identify new markers to predict disease behavior. Here, we compared the gene expression profiles of benign nevi, and non-metastatic and metastatic primary melanomas to identify any common changes in disease progression. We identified several genes associated with inflammation, angiogenesis, and extracellular matrix modification to be upregulated in metastatic melanomas. We selected one of these genes, collagen triple helix repeat containing 1 (CTHRC1), for detailed analysis, and found that CTHRC1 was expressed in both melanoma cells and the associated fibroblasts, as well as in the endothelium of tumor blood vessels. Knockdown of CTHRC1 expression by shRNAs in melanoma cells inhibited their migration in Transwell assays and their invasion in three-dimensional collagen and Matrigel matrices. We also elucidated the possible down-stream effectors of CTHRC1 by gene expression profiling of the CTHRC1-knockdown cells. Our analyses showed that CTHRC1 is regulated coordinately with fibronectin and integrin β3 by the pro-invasive and -angiogenic transcription factor NFATC2. We also found CTHRC1 to be a target of TFGβ and BRAF. These data highlight the importance of tumor stroma in melanoma progression. Furthermore, CTHRC1 was recognized as an important mediator of melanoma cell migration and invasion, providing together with its regulators-NFATC2, TGFβ, and BRAF-attractive therapeutic targets against metastatic melanomas.
Collapse
Affiliation(s)
- Johanna Eriksson
- Department of Pathology, University of Helsinki, FI-00014 Helsinki, Finland
| | - Vadim Le Joncour
- University of Helsinki, Research Programs Unit, Translational Cancer Biology, Biomedicum Helsinki, FI-00014 Helsinki, Finland
| | - Pirjo Nummela
- Department of Pathology, University of Helsinki, FI-00014 Helsinki, Finland
| | - Tiina Jahkola
- Department of Plastic Surgery, Helsinki University Central Hospital, FI-00029 Helsinki, Finland
| | - Susanna Virolainen
- Department of Pathology, University of Helsinki, FI-00014 Helsinki, Finland
| | - Pirjo Laakkonen
- University of Helsinki, Research Programs Unit, Translational Cancer Biology, Biomedicum Helsinki, FI-00014 Helsinki, Finland
| | - Olli Saksela
- Department of Dermatology, Helsinki University Central Hospital, FI-00029 Helsinki, Finland
| | - Erkki Hölttä
- Department of Pathology, University of Helsinki, FI-00014 Helsinki, Finland
| |
Collapse
|
25
|
Sema6A and Mical1 control cell growth and survival of BRAFV600E human melanoma cells. Oncotarget 2015; 6:2779-93. [PMID: 25576923 PMCID: PMC4413617 DOI: 10.18632/oncotarget.2995] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 12/12/2014] [Indexed: 01/13/2023] Open
Abstract
We used whole genome microarray analysis to identify potential candidate genes with differential expression in BRAFV600E vs NRASQ61R melanoma cells. We selected, for comparison, a peculiar model based on melanoma clones, isolated from a single tumor characterized by mutually exclusive expression of BRAFV600E and NRASQ61R in different cells. This effort led us to identify two genes, SEMA6A and MICAL1, highly expressed in BRAF-mutant vs NRAS-mutant clones. Real-time PCR, Western blot and immunohistochemistry confirmed preferential expression of Sema6A and Mical1 in BRAFV600E melanoma. Sema6A is a member of the semaphorin family, and it complexes with the plexins to regulate actin cytoskeleton, motility and cell proliferation. Silencing of Sema6A in BRAF-mutant cells caused cytoskeletal remodeling, and loss of stress fibers, that in turn induced cell death. Furthermore, Sema6A depletion caused loss of anchorage-independent growth, inhibition of chemotaxis and invasion. Forced Sema6A overexpression, in NRASQ61R clones, induced anchorage-independent growth, and a significant increase of invasiveness. Mical1, that links Sema/PlexinA signaling, is also a negative regulator of apoptosis. Indeed, Mical-1 depletion in BRAF mutant cells restored MST-1-dependent NDR phosphorylation and promoted a rapid and massive NDR-dependent apoptosis. Overall, our data suggest that Sema6A and Mical1 may represent new potential therapeutic targets in BRAFV600E melanoma.
Collapse
|
26
|
Identifying Novel Candidate Genes Related to Apoptosis from a Protein-Protein Interaction Network. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2015; 2015:715639. [PMID: 26543496 PMCID: PMC4620916 DOI: 10.1155/2015/715639] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 06/29/2015] [Indexed: 12/31/2022]
Abstract
Apoptosis is the process of programmed cell death (PCD) that occurs in multicellular organisms. This process of normal cell death is required to maintain the balance of homeostasis. In addition, some diseases, such as obesity, cancer, and neurodegenerative diseases, can be cured through apoptosis, which produces few side effects. An effective comprehension of the mechanisms underlying apoptosis will be helpful to prevent and treat some diseases. The identification of genes related to apoptosis is essential to uncover its underlying mechanisms. In this study, a computational method was proposed to identify novel candidate genes related to apoptosis. First, protein-protein interaction information was used to construct a weighted graph. Second, a shortest path algorithm was applied to the graph to search for new candidate genes. Finally, the obtained genes were filtered by a permutation test. As a result, 26 genes were obtained, and we discuss their likelihood of being novel apoptosis-related genes by collecting evidence from published literature.
Collapse
|
27
|
Perotti V, Baldassari P, Molla A, Vegetti C, Bersani I, Maurichi A, Santinami M, Anichini A, Mortarini R. NFATc2 is an intrinsic regulator of melanoma dedifferentiation. Oncogene 2015; 35:2862-72. [DOI: 10.1038/onc.2015.355] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 08/04/2015] [Indexed: 12/20/2022]
|
28
|
Hajibeigi A, Dioum EM, Guo J, Öz OK. Identification of novel regulatory NFAT and TFII-I binding elements in the calbindin-D28k promoter in response to serum deprivation. Biochem Biophys Res Commun 2015; 465:414-420. [PMID: 26260319 DOI: 10.1016/j.bbrc.2015.08.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Accepted: 08/05/2015] [Indexed: 11/18/2022]
Abstract
Calbindin-D28k, a key regulator of calcium homeostasis plays a cytoprotective role in various tissues. We used serum free (SFM) and charcoal stripped serum (csFBS) culture media as models of cellular stress to modulate calbindin D28k expression and identify regulatory cis-elements and trans-acting factors in kidney and beta cells. The murine calbindin-D28k promoter activity was significantly upregulated under SFM or csFBS condition. Promoter analysis revealed evolutionary conserved regulatory cis-elements and deletion of 23 nt from +117/+139 as critical for basal transcription. Bioinformatics analysis of the promoter revealed conserved NFAT and TFII regulators elements. Forced expression of NFAT stimulated promoter activity. Inhibition of NFAT transcriptional activity by FK506 attenuated calbindin-D28k expression. TFII-I was shown to be necessary for basal promoter activity and to act cooperatively with NFAT. Using chromatin immunoprecipitation (ChIP) assays, NFAT was shown to bind to both proximal and distal promoter regions. ChIP assays also revealed recruitment of TFII to the -36/+139 region. Knockdown of TFII-I decreased promoter activity. In summary, calbindin-D28k expression during serum deprivation is partly regulated by NFAT and TF-II. This regulation may be important in vivo during ischemia and growth factor withdrawal to regulate cellular function and maintenance.
Collapse
Affiliation(s)
- Asghar Hajibeigi
- Department of Radiology, UT Southwestern Medical Center, Dallas, Texas 75390-9153, USA
| | - Elhadji M Dioum
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, Texas 75390-9153, USA
| | - Jianfei Guo
- Department of Radiology, UT Southwestern Medical Center, Dallas, Texas 75390-9153, USA
| | - Orhan K Öz
- Department of Radiology, UT Southwestern Medical Center, Dallas, Texas 75390-9153, USA
| |
Collapse
|
29
|
Shou J, Jing J, Xie J, You L, Jing Z, Yao J, Han W, Pan H. Nuclear factor of activated T cells in cancer development and treatment. Cancer Lett 2015; 361:174-84. [PMID: 25766658 DOI: 10.1016/j.canlet.2015.03.005] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 03/04/2015] [Accepted: 03/04/2015] [Indexed: 01/03/2023]
Abstract
Since nuclear factor of activated T cells (NFAT) was first identified as a transcription factor in T cells, various NFAT isoforms have been discovered and investigated. Accumulating studies have suggested that NFATs are involved in many aspects of cancer, including carcinogenesis, cancer cell proliferation, metastasis, drug resistance and tumor microenvironment. Different NFAT isoforms have distinct functions in different cancers. The exact function of NFAT in cancer or the tumor microenvironment is context dependent. In this review, we summarize our current knowledge of NFAT regulation and function in cancer development and treatment. NFATs have emerged as a potential target for cancer prevention and therapy.
Collapse
Affiliation(s)
- Jiawei Shou
- Department of Medical Oncology, Institute of Clinical Science, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jing Jing
- Department of Medical Oncology, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Jiansheng Xie
- Laboratory of Cancer Biology, Institute of Clinical Science, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Liangkun You
- Department of Medical Oncology, Institute of Clinical Science, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Zhao Jing
- Department of Medical Oncology, Institute of Clinical Science, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Junlin Yao
- Department of Medical Oncology, Institute of Clinical Science, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Weidong Han
- Department of Medical Oncology, Institute of Clinical Science, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China; Laboratory of Cancer Biology, Institute of Clinical Science, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Hongming Pan
- Department of Medical Oncology, Institute of Clinical Science, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China; Laboratory of Cancer Biology, Institute of Clinical Science, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
30
|
Kaunisto A, Henry WS, Montaser-Kouhsari L, Jaminet SC, Oh EY, Zhao L, Luo HR, Beck AH, Toker A. NFAT1 promotes intratumoral neutrophil infiltration by regulating IL8 expression in breast cancer. Mol Oncol 2015; 9:1140-54. [PMID: 25735562 DOI: 10.1016/j.molonc.2015.02.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Revised: 02/09/2015] [Accepted: 02/10/2015] [Indexed: 12/26/2022] Open
Abstract
NFAT transcription factors are key regulators of gene expression in immune cells. In addition, NFAT1-induced genes play diverse roles in mediating the progression of various solid tumors. Here we show that NFAT1 induces the expression of the IL8 gene by binding to its promoter and leading to IL8 secretion. Thapsigargin stimulation of breast cancer cells induces IL8 expression in an NFAT-dependent manner. Moreover, we show that NFAT1-mediated IL8 production promotes the migration of primary human neutrophils in vitro and also promotes neutrophil infiltration in tumor xenografts. Furthermore, expression of active NFAT1 effectively suppresses the growth of nascent and established tumors by a non cell-autonomous mechanism. Evaluation of breast tumor tissue reveals that while the levels of NFAT1 are similar in tumor cells and normal breast epithelium, cells in the tumor stroma express higher levels of NFAT1 compared to normal stroma. Elevated levels of NFAT1 also correlate with increased neutrophil infiltrate in breast tumors. These data point to a mechanism by which NFAT1 orchestrates the communication between breast cancer cells and host neutrophils during breast cancer progression.
Collapse
Affiliation(s)
- Aura Kaunisto
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Whitney S Henry
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | | | - Shou-Ching Jaminet
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Eun-Yeong Oh
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Li Zhao
- Department of Laboratory Medicine, Children's Hospital Boston, Boston, MA, USA
| | - Hongbo R Luo
- Department of Laboratory Medicine, Children's Hospital Boston, Boston, MA, USA
| | - Andrew H Beck
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Alex Toker
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, USA.
| |
Collapse
|
31
|
Grazia G, Vegetti C, Benigni F, Penna I, Perotti V, Tassi E, Bersani I, Nicolini G, Canevari S, Carlo-Stella C, Gianni AM, Mortarini R, Anichini A. Synergistic anti-tumor activity and inhibition of angiogenesis by cotargeting of oncogenic and death receptor pathways in human melanoma. Cell Death Dis 2014; 5:e1434. [PMID: 25275595 PMCID: PMC4649516 DOI: 10.1038/cddis.2014.410] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Revised: 08/11/2014] [Accepted: 08/18/2014] [Indexed: 12/11/2022]
Abstract
Improving treatment of advanced melanoma may require the development of effective strategies to overcome resistance to different anti-tumor agents and to counteract relevant pro-tumoral mechanisms in the microenvironment. Here we provide preclinical evidence that these goals can be achieved in most melanomas, by co-targeting of oncogenic and death receptor pathways, and independently of their BRAF, NRAS, p53 and PTEN status. In 49 melanoma cell lines, we found independent susceptibility profiles for response to the MEK1/2 inhibitor AZD6244, the PI3K/mTOR inhibitor BEZ235 and the death receptor ligand TRAIL, supporting the rationale for their association. Drug interaction analysis indicated that a strong synergistic anti-tumor activity could be achieved by the three agents and the AZD6244–TRAIL association on 20/21 melanomas, including cell lines resistant to the inhibitors or to TRAIL. Mechanistically, synergy was explained by enhanced induction of caspase-dependent apoptosis, mitochondrial depolarization and modulation of key regulators of extrinsic and intrinsic cell death pathways, including c-FLIP, BIM, BAX, clusterin, Mcl-1 and several IAP family members. Moreover, silencing experiments confirmed the central role of Apollon downmodulation in promoting the apoptotic response of melanoma cells to the combinatorial treatments. In SCID mice, the AZD6244–TRAIL association induced significant growth inhibition of a tumor resistant to TRAIL and poorly responsive to AZD6244, with no detectable adverse events on body weight and tissue histology. Reduction in tumor volume was associated not only with promotion of tumor apoptosis but also with suppression of the pro-angiogenic molecules HIF1α, VEGFα, IL-8 and TGFβ1 and with inhibition of tumor angiogenesis. These results suggest that synergistic co-targeting of oncogenic and death receptor pathways can not only overcome melanoma resistance to different anti-tumor agents in vitro but can also promote pro-apoptotic effects and inhibition of tumor angiogenesis in vivo.
Collapse
Affiliation(s)
- G Grazia
- Human Tumors Immunobiology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, and Medical Oncology, Università degli Studi di Milano, Milan, Italy
| | - C Vegetti
- Human Tumors Immunobiology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, and Medical Oncology, Università degli Studi di Milano, Milan, Italy
| | - F Benigni
- San Raffaele Scientific Institute, URI, Milan, Italy
| | - I Penna
- Human Tumors Immunobiology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, and Medical Oncology, Università degli Studi di Milano, Milan, Italy
| | - V Perotti
- Human Tumors Immunobiology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, and Medical Oncology, Università degli Studi di Milano, Milan, Italy
| | - E Tassi
- Human Tumors Immunobiology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, and Medical Oncology, Università degli Studi di Milano, Milan, Italy
| | - I Bersani
- Human Tumors Immunobiology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, and Medical Oncology, Università degli Studi di Milano, Milan, Italy
| | - G Nicolini
- Human Tumors Immunobiology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, and Medical Oncology, Università degli Studi di Milano, Milan, Italy
| | - S Canevari
- Functional Genomics Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, and Medical Oncology, Università degli Studi di Milano, Milan, Italy
| | - C Carlo-Stella
- 1] Department of Oncology and Hematology, Humanitas Cancer Center, Humanitas Clinical and Research Center, Rozzano, Italy [2] Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - A M Gianni
- Medical Oncology Unit 2, Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, and Medical Oncology, Università degli Studi di Milano, Milan, Italy
| | - R Mortarini
- Human Tumors Immunobiology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, and Medical Oncology, Università degli Studi di Milano, Milan, Italy
| | - A Anichini
- Human Tumors Immunobiology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, and Medical Oncology, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
32
|
Macleod AC, Klug LR, Patterson J, Griffith DJ, Beadling C, Town A, Heinrich MC. Combination therapy for KIT-mutant mast cells: targeting constitutive NFAT and KIT activity. Mol Cancer Ther 2014; 13:2840-51. [PMID: 25253785 DOI: 10.1158/1535-7163.mct-13-0830] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Resistant KIT mutations have hindered the development of KIT kinase inhibitors for treatment of patients with systemic mastocytosis. The goal of this research was to characterize the synergistic effects of a novel combination therapy involving inhibition of KIT and calcineurin phosphatase, a nuclear factor of activated T cells (NFAT) regulator, using a panel of KIT-mutant mast cell lines. The effects of monotherapy or combination therapy on the cellular viability/survival of KIT-mutant mast cells were evaluated. In addition, NFAT-dependent transcriptional activity was monitored in a representative cell line to evaluate the mechanisms responsible for the efficacy of combination therapy. Finally, shRNA was used to stably knockdown calcineurin expression to confirm the role of calcineurin in the observed synergy. The combination of a KIT inhibitor and a calcineurin phosphatase inhibitor (CNPI) synergized to reduce cell viability and induce apoptosis in six distinct KIT-mutant mast cell lines. Both KIT inhibitors and CNPIs were found to decrease NFAT-dependent transcriptional activity. NFAT-specific inhibitors induced similar synergistic apoptosis induction as CNPIs when combined with a KIT inhibitor. Notably, NFAT was constitutively active in each KIT-mutant cell line tested. Knockdown of calcineurin subunit PPP3R1 sensitized cells to KIT inhibition and increased NFAT phosphorylation and cytoplasmic localization. Constitutive activation of NFAT appears to represent a novel and targetable characteristic of KIT-mutant mast cell disease. Our studies suggest that combining KIT inhibition with NFAT inhibition might represent a new treatment strategy for mast cell disease.
Collapse
Affiliation(s)
- Alison C Macleod
- Portland VA Medical Center, Portland, Oregon. OHSU Knight Cancer Institute, Portland, Oregon
| | - Lillian R Klug
- Portland VA Medical Center, Portland, Oregon. OHSU Knight Cancer Institute, Portland, Oregon
| | - Janice Patterson
- Portland VA Medical Center, Portland, Oregon. OHSU Knight Cancer Institute, Portland, Oregon
| | - Diana J Griffith
- Portland VA Medical Center, Portland, Oregon. OHSU Knight Cancer Institute, Portland, Oregon
| | - Carol Beadling
- Portland VA Medical Center, Portland, Oregon. OHSU Knight Cancer Institute, Portland, Oregon
| | - Ajia Town
- Portland VA Medical Center, Portland, Oregon. OHSU Knight Cancer Institute, Portland, Oregon
| | - Michael C Heinrich
- Portland VA Medical Center, Portland, Oregon. OHSU Knight Cancer Institute, Portland, Oregon.
| |
Collapse
|
33
|
Gene profiling of human VEGF signaling pathways in human endothelial and retinal pigment epithelial cells after anti VEGF treatment. BMC Res Notes 2014; 7:617. [PMID: 25201034 PMCID: PMC4167513 DOI: 10.1186/1756-0500-7-617] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Accepted: 08/27/2014] [Indexed: 11/23/2022] Open
Abstract
Background Ranibizumab (Lucentis®) is a Fab-antibody fragment developed from Bevacizumab, a full-length anti-VEGF antibody. Both compounds are used for treating age-related macular degeneration (AMD). The influence of bevacizumab and ranibizumab on genes involved in signal transduction and cell signaling downstream of VEGF were compared in order to detect possible differences in their mode of action, which are not related to their Fab-antibody fragments. Methods Human umbilical vein cell lines (EA.hy926) and retinal pigment epithelial cells (ARP-19) were exposed to oxidative stress. The cells were treated with therapeutic concentrations of bevacizumab (0.25 mg/mL) and ranibizumab (125 mg/mL) for 24 hours prior to all experiments, and their effects on gene expressions were determined by RT- PCR. Results After exposure to bevacizumab, more genes in the endothelial cells were up-regulated (KDR, NFATc2) and down-regulated (Pla2g12a, Rac2, HgdC, PRKCG) compared to non-treated controls. After exposure to ranibizumab, fewer genes were up-regulated (PTGS2) and down-regulated (NOS3) compared to controls. In comparison between drugs, more genes were up-regulated (NFATc2 and KDR) and more were down-regulated (Pla2g12a, Pla2g1b, Ppp3r2, Rac2) by bevacizumab than by ranibizumab. In RPE cells, NOS3 and PGF were up-regulated and Pla2g12b was down-regulated after exposure to ranibizumab, while PIK3CG was up-regulated and FIGF was down-regulated after exposure to bevacizumab, but the differences in gene expression were minor between drugs (PIK3CGand PGF were down-regulated more by ranibizumab than by bevacizumab). Conclusions The different gene expressions after exposure to ranibizumab and bevacizumab in endothelial and RPE cells may indicate a somewhat different biological activity of the two compounds.
Collapse
|
34
|
Levin-Gromiko U, Koshelev V, Kushnir P, Fedida-Metula S, Voronov E, Fishman D. Amplified lipid rafts of malignant cells constitute a target for inhibition of aberrantly active NFAT and melanoma tumor growth by the aminobisphosphonate zoledronic acid. Carcinogenesis 2014; 35:2555-66. [DOI: 10.1093/carcin/bgu178] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
35
|
Tringali C, Silvestri I, Testa F, Baldassari P, Anastasia L, Mortarini R, Anichini A, López-Requena A, Tettamanti G, Venerando B. Molecular subtyping of metastatic melanoma based on cell ganglioside metabolism profiles. BMC Cancer 2014; 14:560. [PMID: 25085576 PMCID: PMC4132924 DOI: 10.1186/1471-2407-14-560] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Accepted: 07/28/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND In addition to alterations concerning the expression of oncogenes and onco-suppressors, melanoma is characterized by the presence of distinctive gangliosides (sialic acid carrying glycosphingolipids). Gangliosides strongly control cell surface dynamics and signaling; therefore, it could be assumed that these alterations are linked to modifications of cell behavior acquired by the tumor. On these bases, this work investigated the correlations between melanoma cell ganglioside metabolism profiles and the biological features of the tumor and the survival of patients. METHODS Melanoma cell lines were established from surgical specimens of AJCC stage III and IV melanoma patients. Sphingolipid analysis was carried out on melanoma cell lines and melanocytes through cell metabolic labeling employing [3-3H]sphingosine and by FACS. N-glycolyl GM3 was identified employing the 14 F7 antibody. Gene expression was assayed by Real Time PCR. Cell invasiveness was assayed through a Matrigel invasion assay; cell proliferation was determined through the soft agar assay, MTT, and [3H] thymidine incorporation. Statistical analysis was performed using XLSTAT software for melanoma hierarchical clustering based on ganglioside profile, the Kaplan-Meier method, the log-rank (Mantel-Cox) test, and the Mantel-Haenszel test for survival analysis. RESULTS Based on the ganglioside profiles, through a hierarchical clustering, we classified melanoma cells isolated from patients into three clusters: 1) cluster 1, characterized by high content of GM3, mainly in the form of N-glycolyl GM3, and GD3; 2) cluster 2, characterized by the appearance of complex gangliosides and by a low content of GM3; 3) cluster 3, which showed an intermediate phenotype between cluster 1 and cluster 3. Moreover, our data demonstrated that: a) a correlation could be traced between patients' survival and clusters based on ganglioside profiles, with cluster 1 showing the worst survival; b) the expression of several enzymes (sialidase NEU3, GM2 and GM1 synthases) involved in ganglioside metabolism was associated with patients' survival; c) melanoma clusters showed different malignant features such as growth in soft agar, invasiveness, expression of anti-apoptotic proteins. CONCLUSIONS Ganglioside profile and metabolism is strictly interconnected with melanoma aggressiveness. Therefore, the profiling of melanoma gangliosides and enzymes involved in their metabolism could represent a useful prognostic and diagnostic tool.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Bruno Venerando
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Segrate, Milan, Italy.
| |
Collapse
|
36
|
Grazia G, Penna I, Perotti V, Anichini A, Tassi E. Towards combinatorial targeted therapy in melanoma: from pre-clinical evidence to clinical application (review). Int J Oncol 2014; 45:929-49. [PMID: 24920406 PMCID: PMC4121406 DOI: 10.3892/ijo.2014.2491] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Accepted: 04/30/2014] [Indexed: 12/15/2022] Open
Abstract
Over the last few years, clinical trials with BRAF and mitogen-activated protein/extracellular signal-regulated kinase (MEK) inhibitors have shown significant clinical activity in melanoma, but only a fraction of patients respond to these therapies, and development of resistance is frequent. This has prompted a large set of preclinical studies looking at several new combinatorial approaches of pathway- or target-specific inhibitors. At least five main drug association strategies have been verified in vitro and in preclinical models. The most promising include: i) vertical targeting of either MEK or phosphoinositide-3 kinase (PI3K)/mammalian target of rapamycin (mTOR) pathways, or their combined blockade; ii) association of receptor tyrosine kinases (RTKs) inhibitors with other pro-apoptotic strategies; iii) engagement of death receptors in combination with MEK-, mTOR/PI3K-, histone deacetylase (HDAC)-inhibitors, or with anti-apoptotic molecules modulators; iv) strategies aimed at blocking anti-apoptotic proteins belonging to B-cell lymphoma (Bcl-2) or inhibitors of apoptosis (IAP) families associated with MEK/BRAF/p38 inhibition; v) co-inhibition of other molecules important for survival [proteasome, HDAC and Signal transducers and activators of transcription (Stat)3] and the major pathways activated in melanoma; vi) simultaneous targeting of multiple anti-apoptotic molecules. Here we review the anti-melanoma efficacy and mechanism of action of the above-mentioned combinatorial strategies, together with the potential clinical application of the most promising studies that may eventually lead to therapeutic benefit.
Collapse
Affiliation(s)
- Giulia Grazia
- Human Tumors Immunobiology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
| | - Ilaria Penna
- Human Tumors Immunobiology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
| | - Valentina Perotti
- Human Tumors Immunobiology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
| | - Andrea Anichini
- Human Tumors Immunobiology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
| | - Elena Tassi
- Human Tumors Immunobiology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
| |
Collapse
|
37
|
Ando H, Asai T, Koide H, Okamoto A, Maeda N, Tomita K, Dewa T, Minamino T, Oku N. Advanced cancer therapy by integrative antitumor actions via systemic administration of miR-499. J Control Release 2014; 181:32-9. [DOI: 10.1016/j.jconrel.2014.02.019] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2013] [Revised: 02/05/2014] [Accepted: 02/21/2014] [Indexed: 02/01/2023]
|
38
|
Li W, Chen W, Herberman RB, Plotnikoff NP, Youkilis G, Griffin N, Wang E, Lu C, Shan F. Immunotherapy of cancer via mediation of cytotoxic T lymphocytes by methionine enkephalin (MENK). Cancer Lett 2013; 344:212-22. [PMID: 24291668 DOI: 10.1016/j.canlet.2013.10.029] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2013] [Revised: 10/07/2013] [Accepted: 10/27/2013] [Indexed: 01/03/2023]
Abstract
The aim of this study was to investigate the immunological mechanisms by which synthetic methionine enkephalin (MENK) exerts therapeutic effects on tumor growth. Our findings in vivo or in vitro show that MENK treatment either in vivo or in vitro could up-regulate the percentages of CD8+T cells, induce markers of activated T cells, increased cytotoxic activity against mouse S180 tumor cells and increase secretion of IFNγ. In addition, the adoptively transferred CD8+T cells, after either in vitro or in vivo treatment with MENK, result in significantly increased survival of S180 tumor-bearing mice and significant shrinkage in tumor growth. Opioid receptors are detected on normal CD8+T cells and exposure to MENK leads to increased expression of opioid receptors. Interaction between MENK and the opioid receptors on CD8+T cells appears to be essential for the activation of CTL, since the addition of naltrexone (NTX), an opioid receptor antagonist, significantly inhibits all of the effects of MENK. The evidence obtained indicates that the MENK-induced T cell signaling is associated with a significant up-regulation of Ca2+ influx into the cytoplasm and the translocation of NFAT2 into nucleus, and these signaling effects are also inhibited by naltrexone.
Collapse
Affiliation(s)
- Weiwei Li
- Department of Immunology, School of Basic Medical Science, China Medical University, No. 92, North Second Road, Heping District, Shenyang 110001, PR China
| | - Wenna Chen
- Center of Teaching & Research, Liaoning University of Traditional Chinese Medicine, No. 79, Chongshan Eastern Road, Huanggu District, Shenyang 110847, PR China
| | - Ronald B Herberman
- TNI Bio Tech. Inc., 6701 Democracy Blvd., Suite 300, Bethesda, MD 20817, USA
| | | | - Gene Youkilis
- TNI Bio Tech. Inc., 6701 Democracy Blvd., Suite 300, Bethesda, MD 20817, USA
| | - Noreen Griffin
- TNI Bio Tech. Inc., 6701 Democracy Blvd., Suite 300, Bethesda, MD 20817, USA
| | - Enhua Wang
- Institute of Pathology and Pathophysiology, School of Basic Medical Science, China Medical University, No. 92, North Second Road, Heping District, Shenyang 110001, PR China
| | - Changlong Lu
- Department of Immunology, School of Basic Medical Science, China Medical University, No. 92, North Second Road, Heping District, Shenyang 110001, PR China
| | - Fengping Shan
- Department of Immunology, School of Basic Medical Science, China Medical University, No. 92, North Second Road, Heping District, Shenyang 110001, PR China.
| |
Collapse
|
39
|
Vinante F, Tomei P, Zaza G, Zamò A, Lupo A. Hairy cell leukemia in kidney transplantation: lesson from a rare disorder. Exp Hematol Oncol 2013; 2:22. [PMID: 23927433 PMCID: PMC3750510 DOI: 10.1186/2162-3619-2-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Accepted: 08/02/2013] [Indexed: 11/10/2022] Open
Abstract
We report here on the diagnosis and successful treatment of a case of hairy cell leukemia (HCL) that arose 15 years after kidney transplantation in a 51-year-old patient. As soon as the diagnosis was made, HCL was treated with 2-CDA, obtaining complete hematological remission. Immunosuppression with the calcineurin inhibitor cyclosporin was maintained, and the graft was preserved. In kidney transplant recipients supported with immunosuppressive drugs, post-transplant lymphoproliferative diseases (PTLDs) are frequent and typically related to immunosuppression via a loss of control of infectious/EBV-related proliferative stimuli. To date, HCL has not been considered among PTLDs. Recently, however, the oncogenic mutation V600E of the BRAF protein kinase has been found to be a hallmark of HCL, and calcineurin inhibitors have been shown to interfere with signaling downstream of V600E BRAF early on by counteracting senescence-associated mechanisms that protect against the oncogenic potential of the mutated kinase. Such a biochemical link between the oncogene-dependent signaling and calcineurin inhibitor activities suggests that HCL in transplanted patients might be a peculiar type of PTLD based on the presence of a specific mutation. This mechanism might also be involved in other neoplasias bearing the same or similar mutations, such as melanoma and non-melanoma skin cancer.
Collapse
Affiliation(s)
- Fabrizio Vinante
- Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
| | - Paola Tomei
- Renal Unit, Department of Medicine, University Hospital of Verona, Verona, Italy
| | - Gianluigi Zaza
- Renal Unit, Department of Medicine, University Hospital of Verona, Verona, Italy
| | - Alberto Zamò
- Section of Pathological Anatomy, Department of Pathology and Diagnostics, University of Verona, Verona, Italy
| | - Antonio Lupo
- Renal Unit, Department of Medicine, University Hospital of Verona, Verona, Italy
| |
Collapse
|
40
|
Molecular detection and targeting of EWSR1 fusion transcripts in soft tissue tumors. Med Oncol 2013; 30:412. [PMID: 23329308 PMCID: PMC3586390 DOI: 10.1007/s12032-012-0412-8] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Accepted: 11/30/2012] [Indexed: 12/11/2022]
Abstract
Soft tissue tumors are a heterogeneous group of tumors, traditionally classified according to morphology and histogenesis. Molecular classification divides sarcomas into two main categories: (a) sarcomas with specific genetic alterations and (b) sarcomas showing multiple complex karyotypic abnormalities without any specific pattern. Most chromosomal alterations are represented by translocations which are increasingly detected. The identification of fusion transcripts, in fact, not only support the diagnosis but also provides the basis for the development of new therapeutic strategies aimed at blocking aberrant activity of the chimeric proteins. One of the genes most susceptible to breakage/translocation in soft tissue tumors is represented by Ewing sarcoma breakpoint region 1 (EWSR1). This gene has a large number of fusion partners, mainly associated with the pathogenesis of Ewing's sarcoma but with other soft tissue tumors too. In this review, we illustrate the characteristics of this gene/protein, both in normal cellular physiology and in carcinogenesis. We describe the different fusion partners of EWSR1, the molecular pathways in which is involved and the main molecular biology techniques for the identification of fusion transcripts and for their inhibition.
Collapse
|