51
|
Abstract
Treatment options for patients with metastatic melanoma, and especially BRAF-mutant melanoma, have changed dramatically in the past 5 years, with the FDA approval of eight new therapeutic agents. During this period, the treatment paradigm for BRAF-mutant disease has evolved rapidly: the standard-of-care BRAF-targeted approach has shifted from single-agent BRAF inhibition to combination therapy with a BRAF and a MEK inhibitor. Concurrently, immunotherapy has transitioned from cytokine-based treatment to antibody-mediated blockade of the cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4) and, now, the programmed cell-death protein 1 (PD-1) immune checkpoints. These changes in the treatment landscape have dramatically improved patient outcomes, with the median overall survival of patients with advanced-stage melanoma increasing from approximately 9 months before 2011 to at least 2 years - and probably longer for those with BRAF-V600-mutant disease. Herein, we review the clinical trial data that established the standard-of-care treatment approaches for advanced-stage melanoma. Mechanisms of resistance and biomarkers of response to BRAF-targeted treatments and immunotherapies are discussed, and the contrasting clinical benefits and limitations of these therapies are explored. We summarize the state of the field and outline a rational approach to frontline-treatment selection for each individual patient with BRAF-mutant melanoma.
Collapse
|
52
|
Marie Vincent K, Postovit LM. Investigating the utility of human melanoma cell lines as tumour models. Oncotarget 2017; 8:10498-10509. [PMID: 28060736 PMCID: PMC5354675 DOI: 10.18632/oncotarget.14443] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 12/13/2016] [Indexed: 12/31/2022] Open
Abstract
Melanoma researchers utilize cell lines to model many tumour phenomena. It is thus important to understand similarities and differences between cell lines and the tumours that they represent, so that the optimal models can be chosen to answer specific research questions. Herein, we compared the transcriptomes of 42 melanoma cell lines to hundreds of tumours from The Cancer Genome Atlas and thousands of single melanoma cells. Tumour purity was accounted for using the ESTIMATE algorithm, so that differences likely resulting from non-tumour cells could be accounted for. In addition, UV mutational signatures and the expression of skin-associated genes were analyzed in order to identify the putative origin of various cell lines. We found the transcriptional and mutational characteristics of melanoma cell lines to mirror those of the tumours, with the exception of immune-associated transcripts, which were absent in cell culture. We also determined cell lines that highly or poorly recapitulate melanomas and have identified colon (COLO 741) and lung (COLO 699) cancer cell lines that may actually be melanoma. In summary, this study represents a comprehensive comparison of melanoma cell lines and tumours that can be used as a guide for researchers when selecting melanoma cell line models.
Collapse
Affiliation(s)
- Krista Marie Vincent
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, T6G 2E1, Canada
- Department of Anatomy and Cell Biology, Faculty of Medicine and Dentistry, University of Western Ontario, London, ON N6A 3K7, Canada
| | - Lynne-Marie Postovit
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, T6G 2E1, Canada
| |
Collapse
|
53
|
Gene Expression Profile of Dendritic Cell-Tumor Cell Hybrids Determined by Microarrays and Its Implications for Cancer Immunotherapy. J Immunol Res 2015; 2015:789136. [PMID: 26605345 PMCID: PMC4641191 DOI: 10.1155/2015/789136] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 10/08/2015] [Indexed: 12/24/2022] Open
Abstract
Background. Dendritic cell- (DC-) tumor fusion cells stimulate effective in vivo antitumor responses. However, therapeutic approaches are dependent upon the coadministration of exogenous 3rd signals. The purpose of this study was to determine the mechanisms for inadequate 3rd signaling by electrofused DC-tumor cell hybrids. Methods. Murine melanoma cells were fused with DCs derived from C57BL/6 mice. Quantitative real-time PCR (qPCR) was used to determine relative changes in Th (T helper) 1 and Th2 cytokine gene expression. In addition, changes in gene expression of fusion cells were determined by microarray. Last, cytokine secretion by fusion cells upon inhibition of signaling pathways was analyzed by ELISA. Results. qPCR analyses revealed that fusion cells exhibited a downregulation of Th1 associated cytokines IL-12 and IL-15 and an upregulation of the Th2 cytokine IL-4. Microarray studies further showed that the expression of chemokines, costimulatory molecules, and matrix-metalloproteinases was deregulated in fusion cells. Lastly, inhibitor studies demonstrate that inhibition of the PI3K/Akt/mTOR signaling pathway could restore the secretion of bioactive IL-12p70 by fusion cells. Conclusion. Our results suggest that combining fusion cell-based vaccination with administration of inhibitors of the PI3K/Akt/mTOR signaling pathway may enhance antitumor responses in patients.
Collapse
|
54
|
Kuzu OF, Nguyen FD, Noory MA, Sharma A. Current State of Animal (Mouse) Modeling in Melanoma Research. CANCER GROWTH AND METASTASIS 2015; 8:81-94. [PMID: 26483610 PMCID: PMC4597587 DOI: 10.4137/cgm.s21214] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Revised: 08/10/2015] [Accepted: 08/17/2015] [Indexed: 11/16/2022]
Abstract
Despite the considerable progress in understanding the biology of human cancer and technological advancement in drug discovery, treatment failure remains an inevitable outcome for most cancer patients with advanced diseases, including melanoma. Despite FDA-approved BRAF-targeted therapies for advanced stage melanoma showed a great deal of promise, development of rapid resistance limits the success. Hence, the overall success rate of melanoma therapy still remains to be one of the worst compared to other malignancies. Advancement of next-generation sequencing technology allowed better identification of alterations that trigger melanoma development. As development of successful therapies strongly depends on clinically relevant preclinical models, together with the new findings, more advanced melanoma models have been generated. In this article, besides traditional mouse models of melanoma, we will discuss recent ones, such as patient-derived tumor xenografts, topically inducible BRAF mouse model and RCAS/TVA-based model, and their advantages as well as limitations. Although mouse models of melanoma are often criticized as poor predictors of whether an experimental drug would be an effective treatment, development of new and more relevant models could circumvent this problem in the near future.
Collapse
Affiliation(s)
- Omer F Kuzu
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Felix D Nguyen
- The University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Mohammad A Noory
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Arati Sharma
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| |
Collapse
|
55
|
Serrano OK, Parrow NL, Violet PC, Yang J, Zornjak J, Basseville A, Levine M. Antitumor effect of pharmacologic ascorbate in the B16 murine melanoma model. Free Radic Biol Med 2015; 87:193-203. [PMID: 26119785 DOI: 10.1016/j.freeradbiomed.2015.06.032] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 06/18/2015] [Accepted: 06/22/2015] [Indexed: 11/29/2022]
Abstract
Because 5-year survival rates for patients with metastatic melanoma remain below 25%, there is continued need for new therapeutic approaches. For some tumors, pharmacologic ascorbate treatment may have a beneficial antitumor effect and may work synergistically with standard chemotherapeutics. To investigate this possibility in melanoma, we examined the effect of pharmacologic ascorbate on B16-F10 cells. Murine models were employed to compare tumor size following treatment with ascorbate, and the chemotherapeutic agents dacarbazine or valproic acid, alone or in combination with ascorbate. Results indicated that nearly all melanoma cell lines were susceptible to ascorbate-mediated cytotoxicity. Compared to saline controls, pharmacologic ascorbate decreased tumor size in both C57BL/6 (P < 0.0001) and NOD-scid tumor bearing mice (P < 0.0001). Pharmacologic ascorbate was superior or equivalent to dacarbazine as an antitumor agent. Synergy was not apparent when ascorbate was combined with either dacarbazine or valproic acid; the latter combination may have additional toxicities. Pharmacologic ascorbate induced DNA damage in melanoma cells, as evidenced by increased phosphorylation of the histone variant, H2A.X. Differences were not evident in tumor samples from C57BL/6 mice treated with pharmacologic ascorbate compared to tumors from saline-treated controls. Together, these results suggest that pharmacologic ascorbate has a cytotoxic effect against melanoma that is largely independent of lymphocytic immune functions and that continued investigation of pharmacologic ascorbate in cancer treatment is warranted.
Collapse
Affiliation(s)
- Oscar K Serrano
- Department of Surgery, Albert Einstein College of Medicine, Montefiore Medical Center, New York, NY, USA
| | - Nermi L Parrow
- Molecular and Clinical Nutrition Section, Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Pierre-Christian Violet
- Molecular and Clinical Nutrition Section, Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jacqueline Yang
- Molecular and Clinical Nutrition Section, Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jennifer Zornjak
- Molecular and Clinical Nutrition Section, Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Agnes Basseville
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Mark Levine
- Molecular and Clinical Nutrition Section, Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
56
|
Mac Keon S, Ruiz MS, Gazzaniga S, Wainstok R. Dendritic cell-based vaccination in cancer: therapeutic implications emerging from murine models. Front Immunol 2015; 6:243. [PMID: 26042126 PMCID: PMC4438595 DOI: 10.3389/fimmu.2015.00243] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 05/06/2015] [Indexed: 01/29/2023] Open
Abstract
Dendritic cells (DCs) play a pivotal role in the orchestration of immune responses, and are thus key targets in cancer vaccine design. Since the 2010 FDA approval of the first cancer DC-based vaccine (Sipuleucel-T), there has been a surge of interest in exploiting these cells as a therapeutic option for the treatment of tumors of diverse origin. In spite of the encouraging results obtained in the clinic, many elements of DC-based vaccination strategies need to be optimized. In this context, the use of experimental cancer models can help direct efforts toward an effective vaccine design. This paper reviews recent findings in murine models regarding the antitumoral mechanisms of DC-based vaccination, covering issues related to antigen sources, the use of adjuvants and maturing agents, and the role of DC subsets and their interaction in the initiation of antitumoral immune responses. The summary of such diverse aspects will highlight advantages and drawbacks in the use of murine models, and contribute to the design of successful DC-based translational approaches for cancer treatment.
Collapse
Affiliation(s)
- Soledad Mac Keon
- Laboratorio de Cancerología, Fundación Instituto Leloir, Instituto de Investigaciones Bioquímicas de Buenos Aires IIBBA-CONICET , Buenos Aires , Argentina
| | - María Sol Ruiz
- Centro de Investigaciones Oncológicas, Fundación para la Investigación, Docencia y Prevención del Cáncer (FUCA) , Buenos Aires , Argentina
| | - Silvina Gazzaniga
- Laboratorio de Biología Tumoral, Departamento de Química Biológica IQUIBICEN-CONICET, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires , Buenos Aires , Argentina
| | - Rosa Wainstok
- Laboratorio de Cancerología, Fundación Instituto Leloir, Instituto de Investigaciones Bioquímicas de Buenos Aires IIBBA-CONICET , Buenos Aires , Argentina ; Laboratorio de Biología Tumoral, Departamento de Química Biológica IQUIBICEN-CONICET, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires , Buenos Aires , Argentina
| |
Collapse
|
57
|
Morris VL, Toseef T, Nazumudeen FB, Rivoira C, Spatafora C, Tringali C, Rotenberg SA. Anti-tumor properties of cis-resveratrol methylated analogs in metastatic mouse melanoma cells. Mol Cell Biochem 2015; 402:83-91. [PMID: 25567208 DOI: 10.1007/s11010-014-2316-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 12/23/2014] [Indexed: 02/06/2023]
Abstract
Resveratrol (E-3,5,4'-trihydroxystilbene) is a polyphenol found in red wine that has been shown to have multiple anti-cancer properties. Although cis-(Z)- and trans-(E)-isomers of resveratrol occur in nature, the cis form is not biologically active. However, methylation at key positions of the cis form results in more potent anti-cancer properties. This study determined that synthetic cis-polymethoxystilbenes (methylated analogs of cis-resveratrol) inhibited cancer-related phenotypes of metastatic B16 F10 and non-metastatic B16 F1 mouse melanoma cells. In contrast with cis- or trans-resveratrol and trans-polymethoxystilbene which were ineffective at 10 μM, cis-polymethoxystilbenes inhibited motility and proliferation of melanoma cells with low micromolar specificity (IC50 < 10 μM). Inhibitory effects by cis-polymethoxystilbenes were significantly stronger with B16 F10 cells and were accompanied by decreased expression of β-tubulin and pleckstrin homology domain-interacting protein, a marker of metastatic B16 cells. Thus, cis-polymethoxystilbenes have potential as chemotherapeutic agents for metastatic melanoma.
Collapse
Affiliation(s)
- Valery L Morris
- Department of Chemistry and Biochemistry, Queens College of the City University of New York, Flushing, NY, USA
| | | | | | | | | | | | | |
Collapse
|
58
|
Mohammad N, Malvi P, Meena AS, Singh SV, Chaube B, Vannuruswamy G, Kulkarni MJ, Bhat MK. Cholesterol depletion by methyl-β-cyclodextrin augments tamoxifen induced cell death by enhancing its uptake in melanoma. Mol Cancer 2014; 13:204. [PMID: 25178635 PMCID: PMC4175626 DOI: 10.1186/1476-4598-13-204] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 08/22/2014] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Despite modern advances in treatment, skin cancer is still one of the most common causes of death in the western countries. Chemotherapy plays an important role in melanoma management. Tamoxifen has been used either alone or in- combination with other chemotherapeutic agents to treat melanoma. However, response rate of tamoxifen as a single agent has been comparatively low. In the present study, we investigated whether treatment with methyl-β-cyclodextrin (MCD), a cholesterol depleting agent, increases the efficacy of tamoxifen in melanoma cells. METHODS This was a two-part study that incorporated in vitro effects of tamoxifen and MCD combination by analyzing cell survival, apoptosis and cell cycle analysis and in vivo antitumor efficacy on tumor isografts in C57BL/6J mice. RESULTS MCD potentiated tamoxifen induced anticancer effects by causing cell cycle arrest and induction of apoptosis. Sensitization to tamoxifen was associated with down regulation of antiapoptotic protein Bcl-2, up-regulation of proapoptotic protein Bax, reduced caveolin-1 (Cav-1) and decreased pAkt/pERK levels. Co-administration of tamoxifen and MCD caused significant reduction in tumor volume and tumor weight in mice due to enhancement of drug uptake in the tumor. Supplementation with cholesterol abrogated combined effect of tamoxifen and MCD. CONCLUSION Our results emphasize a potential synergistic effect of tamoxifen with MCD, and therefore, may provide a unique therapeutic window for improvement in melanoma treatment.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Manoj Kumar Bhat
- National Centre for Cell Science, NCCS Complex, Ganeshkhind, Pune 411007, India.
| |
Collapse
|
59
|
Matsushita H, Hosoi A, Ueha S, Abe J, Fujieda N, Tomura M, Maekawa R, Matsushima K, Ohara O, Kakimi K. Cytotoxic T lymphocytes block tumor growth both by lytic activity and IFNγ-dependent cell-cycle arrest. Cancer Immunol Res 2014; 3:26-36. [PMID: 25127875 DOI: 10.1158/2326-6066.cir-14-0098] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
To understand global effector mechanisms of CTL therapy, we performed microarray gene expression analysis in a murine model using pmel-1 T-cell receptor (TCR) transgenic T cells as effectors and B16 melanoma cells as targets. In addition to upregulation of genes related to antigen presentation and the MHC class I pathway, and cytotoxic effector molecules, cell-cycle-promoting genes were downregulated in the tumor on days 3 and 5 after CTL transfer. To investigate the impact of CTL therapy on the cell cycle of tumor cells in situ, we generated B16 cells expressing a fluorescent ubiquitination-based cell-cycle indicator (B16-fucci) and performed CTL therapy in mice bearing B16-fucci tumors. Three days after CTL transfer, we observed diffuse infiltration of CTLs into the tumor with a large number of tumor cells arrested at the G1 phase of the cell cycle, and the presence of spotty apoptotic or necrotic areas. Thus, tumor growth suppression was largely dependent on G1 cell-cycle arrest rather than killing by CTLs. Neutralizing antibody to IFNγ prevented both tumor growth inhibition and G1 arrest. The mechanism of G1 arrest involved the downregulation of S-phase kinase-associated protein 2 (Skp2) and the accumulation of its target cyclin-dependent kinase inhibitor p27 in the B16-fucci tumor cells. Because tumor-infiltrating CTLs are far fewer in number than the tumor cells, we propose that CTLs predominantly regulate tumor growth via IFNγ-mediated profound cytostatic effects rather than via cytotoxicity. This dominance of G1 arrest over other mechanisms may be widespread but not universal because IFNγ sensitivity varied among tumors.
Collapse
Affiliation(s)
- Hirokazu Matsushita
- Department of Immunotherapeutics, The University of Tokyo Hospital, Tokyo, Japan
| | - Akihiro Hosoi
- Department of Immunotherapeutics, The University of Tokyo Hospital, Tokyo, Japan. Medinet Co Ltd., Yokohama, Japan
| | - Satoshi Ueha
- Department of Molecular Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Jun Abe
- Department of Molecular Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Nao Fujieda
- Department of Immunotherapeutics, The University of Tokyo Hospital, Tokyo, Japan. Medinet Co Ltd., Yokohama, Japan
| | - Michio Tomura
- Center for Innovation in Immunoregulative Technology and Therapeutics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | | | - Kouji Matsushima
- Department of Molecular Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Osamu Ohara
- Department of Human Genome Research, Kazusa DNA Research Institute, Chiba, Japan
| | - Kazuhiro Kakimi
- Department of Immunotherapeutics, The University of Tokyo Hospital, Tokyo, Japan.
| |
Collapse
|
60
|
Siu KS, Chen D, Zheng X, Zhang X, Johnston N, Liu Y, Yuan K, Koropatnick J, Gillies ER, Min WP. Non-covalently functionalized single-walled carbon nanotube for topical siRNA delivery into melanoma. Biomaterials 2014; 35:3435-42. [PMID: 24424208 DOI: 10.1016/j.biomaterials.2013.12.079] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 12/23/2013] [Indexed: 01/03/2023]
Abstract
RNAi can specifically regulate gene expression, but efficient delivery of siRNA in vivo is difficult while it has been shown that modified carbon nanotubes (CNT) protect siRNA, facilitate entry into cells and enhance transdermal drugs delivery. Single-walled carbon nanotubes (SWCNT) were functionalized non-covalently with succinated polyethyleimine (PEI-SA). In this study, the water soluble CNT, PEI-SA/CNT (IS/C) were isolated and characterized, the gene silencing induced by IS/C/siRNA complexes was achieved in vitro in B16-F10 cells. In vivo delivery was topically applied to shaved mouse skin, as well as topically to a C57BL/6 mice melanoma model. We found significant uptake of Cy3-labeled siRNA specific to Braf (siBraf) and gene silencing in the tumor tissue. Treatment with IS/C/siBraf resulted in attenuation of tumor growth over a 25-day period. This new delivery method has provided a new possibility for future siRNA delivery and therapy, which providing insight for the potential application and development of CNT-based siRNA delivery.
Collapse
Affiliation(s)
- King Sun Siu
- Department of Pathology, Western University, London, Canada; Department of Surgery, Western University, London, Canada; Department of Oncology, Western University, London, Canada; Department of Microbiology and Immunology, Western University, London, Canada; Department of Physiology and Pharmacology, Western University, London, Canada
| | - Di Chen
- Department of Pathology, Western University, London, Canada; Department of Surgery, Western University, London, Canada; Department of Oncology, Western University, London, Canada; Department of Microbiology and Immunology, Western University, London, Canada; Department of Physiology and Pharmacology, Western University, London, Canada
| | - Xiufen Zheng
- Department of Pathology, Western University, London, Canada; Department of Surgery, Western University, London, Canada; Department of Oncology, Western University, London, Canada; Department of Microbiology and Immunology, Western University, London, Canada; Department of Physiology and Pharmacology, Western University, London, Canada
| | - Xusheng Zhang
- Department of Pathology, Western University, London, Canada; Department of Surgery, Western University, London, Canada; Department of Oncology, Western University, London, Canada; Department of Microbiology and Immunology, Western University, London, Canada; Department of Physiology and Pharmacology, Western University, London, Canada
| | - Nathan Johnston
- Department of Pathology, Western University, London, Canada; Department of Surgery, Western University, London, Canada; Department of Oncology, Western University, London, Canada; Department of Microbiology and Immunology, Western University, London, Canada; Department of Physiology and Pharmacology, Western University, London, Canada
| | - Yanling Liu
- Department of Pathology, Western University, London, Canada; Department of Surgery, Western University, London, Canada; Department of Oncology, Western University, London, Canada; Department of Microbiology and Immunology, Western University, London, Canada; Department of Physiology and Pharmacology, Western University, London, Canada; Jiangxi Academy of Medical Sciences, and Institute of Immunomodulation and Immunotherapy of Nanchang University, Nanchang, China
| | - Ken Yuan
- Jiangxi Academy of Medical Sciences, and Institute of Immunomodulation and Immunotherapy of Nanchang University, Nanchang, China
| | - James Koropatnick
- Department of Pathology, Western University, London, Canada; Department of Surgery, Western University, London, Canada; Department of Oncology, Western University, London, Canada; Department of Microbiology and Immunology, Western University, London, Canada; Department of Physiology and Pharmacology, Western University, London, Canada; Lawson Health Research Institute, London, Canada; London Regional Cancer Program, London, Canada
| | - Elizabeth R Gillies
- Department of Chemistry, Chemical and Biochemical Engineering, Western University, London, Canada
| | - Wei-Ping Min
- Department of Pathology, Western University, London, Canada; Department of Surgery, Western University, London, Canada; Department of Oncology, Western University, London, Canada; Department of Microbiology and Immunology, Western University, London, Canada; Department of Physiology and Pharmacology, Western University, London, Canada; Jiangxi Academy of Medical Sciences, and Institute of Immunomodulation and Immunotherapy of Nanchang University, Nanchang, China; Lawson Health Research Institute, London, Canada; London Regional Cancer Program, London, Canada.
| |
Collapse
|
61
|
Lin X, Zheng W, Liu J, Zhang Y, Qin H, Wu H, Xue B, Lu Y, Shen P. Oxidative stress in malignant melanoma enhances tumor necrosis factor-α secretion of tumor-associated macrophages that promote cancer cell invasion. Antioxid Redox Signal 2013; 19:1337-55. [PMID: 23373752 DOI: 10.1089/ars.2012.4617] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
AIMS Malignant melanoma is well known for abundant reactive oxygen species (ROS) that exist in the primary tumor environment. Within this microenvironment, tumor-associated macrophages (TAMs) play substantial roles in multiple steps of tumor development in terms of tumor growth, invasion, and metastasis. We therefore aimed to determine whether this high-level ROS in primary melanoma is capable to promote tumor invasiveness by influencing TAM properties. Moreover, we wanted to further investigate probable underlying mechanisms. RESULTS We characterized malignant melanoma TAMs as a heterogeneous phenotype, which possesses both M1 and M2 markers. We also revealed a role for high-level intracellular ROS in enhancing proinvasion signature of TAMs by strongly increasing their tumor necrosis factor α secretion, which is possibly attributed to ROS-enhanced peroxisome proliferator-activated receptor γ (PPARγ) translocation mediated by MAPK/ERK kinase 1. INNOVATION This is the first study demonstrating that high levels of ROS in the primary melanoma environment can influence TAM behaviors. Furthermore, we are also the first to indentify that nucleus-to-cytoplasm translocation of PPARγ is significantly upregulated by ROS and responsible for the proinvasiveness capacity of melanoma TAMs. CONCLUSION Taken together, our data describe how a high level of ROS plays a critical role in enhancing the proinvasion characteristic of TAMs in malignant melanoma.
Collapse
Affiliation(s)
- Xuzhu Lin
- 1 State Key Laboratory of Pharmaceutical Biotechnology, and Model Animal Research Center (MARC) of Nanjing University, Nanjing University , Nanjing, China
| | | | | | | | | | | | | | | | | |
Collapse
|
62
|
Rodríguez MI, Peralta-Leal A, O'Valle F, Rodriguez-Vargas JM, Gonzalez-Flores A, Majuelos-Melguizo J, López L, Serrano S, de Herreros AG, Rodríguez-Manzaneque JC, Fernández R, del Moral RG, de Almodóvar JM, Oliver FJ. PARP-1 regulates metastatic melanoma through modulation of vimentin-induced malignant transformation. PLoS Genet 2013; 9:e1003531. [PMID: 23785295 PMCID: PMC3681683 DOI: 10.1371/journal.pgen.1003531] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2012] [Accepted: 04/12/2013] [Indexed: 12/30/2022] Open
Abstract
PARP inhibition can induce anti-neoplastic effects when used as monotherapy or in combination with chemo- or radiotherapy in various tumor settings; however, the basis for the anti-metastasic activities resulting from PARP inhibition remains unknown. PARP inhibitors may also act as modulators of tumor angiogenesis. Proteomic analysis of endothelial cells revealed that vimentin, an intermediary filament involved in angiogenesis and a specific hallmark of EndoMT (endothelial to mesenchymal transition) transformation, was down-regulated following loss of PARP-1 function in endothelial cells. VE-cadherin, an endothelial marker of vascular normalization, was up-regulated in HUVEC treated with PARP inhibitors or following PARP-1 silencing; vimentin over-expression was sufficient to drive to an EndoMT phenotype. In melanoma cells, PARP inhibition reduced pro-metastatic markers, including vasculogenic mimicry. We also demonstrated that vimentin expression was sufficient to induce increased mesenchymal/pro-metastasic phenotypic changes in melanoma cells, including ILK/GSK3-β-dependent E-cadherin down-regulation, Snail1 activation and increased cell motility and migration. In a murine model of metastatic melanoma, PARP inhibition counteracted the ability of melanoma cells to metastasize to the lung. These results suggest that inhibition of PARP interferes with key metastasis-promoting processes, leading to suppression of invasion and colonization of distal organs by aggressive metastatic cells. Metastasis is the spread of malignant tumor cells from their original site to other parts of the body and is responsible for the vast majority of solid cancer-related mortality. PARP inhibitors are emerging as promising anticancer therapeutics and are currently undergoing clinical trials. It is therefore important to elucidate the mechanisms underlying the anti-tumor actions of these drugs. In our current study, we elucidated novel anti-neoplastic properties of PARP inhibitors that are responsible for the anti-metastatic effect of these drugs in the context of malignant melanoma. These effects appear to be the result of PARP-1's ability to regulate the expression of key factors, such as vimentin and VE-cadherin, involved in vascular cell dynamics and to limit pro-malignant processes such as vasculogenic mimicry and EMT.
Collapse
Affiliation(s)
- María Isabel Rodríguez
- Instituto de Parasitología y Biomedicina López Neyra, CSIC, Granada, Spain
- IBIMER, Centro de Investigaciones Biomédicas, Universidad de Granada, Granada, Spain
- Departamento de Bioquímica y Biología Molecular I, Universidad de Granada, Granada, Spain
- * E-mail: (MIR); (FJO)
| | | | - Francisco O'Valle
- Departamento de Anatomía Patológica, Universidad de Granada, Granada, Spain
| | | | | | | | - Laura López
- Instituto de Parasitología y Biomedicina López Neyra, CSIC, Granada, Spain
| | - Santiago Serrano
- Instituto de Parasitología y Biomedicina López Neyra, CSIC, Granada, Spain
| | | | | | - Rubén Fernández
- Pfizer-University of Granada-Junta de Andalucía Centre for Genomics and Oncological Research (GENYO), Granada, Spain
| | - Raimundo G. del Moral
- Unidad de Anatomía Patológica, Complejo Hospitalario y Áreas Sur y Noreste de Granada, Granada, Spain
| | | | - F. Javier Oliver
- Instituto de Parasitología y Biomedicina López Neyra, CSIC, Granada, Spain
- * E-mail: (MIR); (FJO)
| |
Collapse
|
63
|
Lee DJ, Kang DH, Choi M, Choi YJ, Lee JY, Park JH, Park YJ, Lee KW, Kang SW. Peroxiredoxin-2 represses melanoma metastasis by increasing E-Cadherin/β-Catenin complexes in adherens junctions. Cancer Res 2013; 73:4744-57. [PMID: 23749642 DOI: 10.1158/0008-5472.can-12-4226] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In melanoma, transition to the vertical growth phase is the critical step in conversion to a deadly malignant disease. Here, we offer the first evidence that an antioxidant enzyme has a key role in this transition. We found that the antioxidant enzyme peroxiredoxin-2 (Prx2) inversely correlated with the metastatic capacity of human melanoma cells. Silencing Prx2 expression stimulated proliferation and migration, whereas ectopic expression of Prx2 produced the opposite effect. Mechanistic investigations indicated that Prx2 negatively regulated Src/ERK activation status, which in turn fortified adherens junctions function by increasing E-cadherin expression and phospho-Y654-dependent retention of β-catenin in the plasma membrane. In murine melanoma cells, Prx2 silencing enhanced lung metastasis in vivo. Interestingly, the natural compound gliotoxin, which is known to exert a Prx-like activity, inhibited proliferation and migration as well as lung metastasis of Prx2-deficient melanoma cells. Overall, our findings reveal that Prx2 is a key regulator of invasion and metastasis in melanoma, and also suggest a pharmacologic strategy to effectively decrease deadly malignant forms of this disease.
Collapse
Affiliation(s)
- Doo Jae Lee
- Division of Life and Pharmaceutical Sciences, Ewha Womans University, Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
64
|
Wein AN, Liu S, Zhang Y, McKenzie AT, Leppla SH. Tumor therapy with a urokinase plasminogen activator-activated anthrax lethal toxin alone and in combination with paclitaxel. Invest New Drugs 2013; 31:206-12. [PMID: 22843210 PMCID: PMC3757568 DOI: 10.1007/s10637-012-9847-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2012] [Accepted: 06/07/2012] [Indexed: 01/03/2023]
Abstract
PA-U2, an engineered anthrax protective antigen that is activated by urokinase was combined with wildtype lethal factor in the treatment of Colo205 colon adenocarcinoma in vitro and B16-BL6 mouse melanoma in vitro and in vivo. This therapy was also tested in combination with the small molecule paclitaxel, based on prior reports suggesting synergy between ERK1/2 inhibition and chemotherapeutics. Colo205 was sensitive to PA-U2/LF while B16-BL6 was not. For the combination treatment of B16-BL6, paclitaxel showed a dose response in vitro, but cells remained resistant to PA-U2/LF even in the presence of paclitaxel. In vivo, each therapy slowed tumor progression, and an additive effect between the two was observed. Since LF targets tumor vasculature while paclitaxel is an antimitotic, it is possible the agents were acting against different cells in the stroma, precluding a synergistic effect. The engineered anthrax toxin PA-U2/LF warrants further development and testing, possibly in combination with an antiangiogenesis therapy such as sunitinib or sorafinib.
Collapse
Affiliation(s)
- Alexander N. Wein
- Microbial Pathogenesis Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 33 North Drive, Bethesda, MD, 20892, USA
| | - Shihui Liu
- Microbial Pathogenesis Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 33 North Drive, Bethesda, MD, 20892, USA
| | - Yi Zhang
- Microbial Pathogenesis Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 33 North Drive, Bethesda, MD, 20892, USA
| | - Andrew T. McKenzie
- Microbial Pathogenesis Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 33 North Drive, Bethesda, MD, 20892, USA
| | - Stephen H. Leppla
- Microbial Pathogenesis Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 33 North Drive, Bethesda, MD, 20892, USA
| |
Collapse
|
65
|
Castle JC, Kreiter S, Diekmann J, Löwer M, van de Roemer N, de Graaf J, Selmi A, Diken M, Boegel S, Paret C, Koslowski M, Kuhn AN, Britten CM, Huber C, Türeci O, Sahin U. Exploiting the mutanome for tumor vaccination. Cancer Res 2012; 72:1081-91. [PMID: 22237626 DOI: 10.1158/0008-5472.can-11-3722] [Citation(s) in RCA: 603] [Impact Index Per Article: 50.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Multiple genetic events and subsequent clonal evolution drive carcinogenesis, making disease elimination with single-targeted drugs difficult. The multiplicity of gene mutations derived from clonal heterogeneity therefore represents an ideal setting for multiepitope tumor vaccination. Here, we used next generation sequencing exome resequencing to identify 962 nonsynonymous somatic point mutations in B16F10 murine melanoma cells, with 563 of those mutations in expressed genes. Potential driver mutations occurred in classical tumor suppressor genes and genes involved in proto-oncogenic signaling pathways that control cell proliferation, adhesion, migration, and apoptosis. Aim1 and Trrap mutations known to be altered in human melanoma were included among those found. The immunogenicity and specificity of 50 validated mutations was determined by immunizing mice with long peptides encoding the mutated epitopes. One-third of these peptides were found to be immunogenic, with 60% in this group eliciting immune responses directed preferentially against the mutated sequence as compared with the wild-type sequence. In tumor transplant models, peptide immunization conferred in vivo tumor control in protective and therapeutic settings, thereby qualifying mutated epitopes that include single amino acid substitutions as effective vaccines. Together, our findings provide a comprehensive picture of the mutanome of B16F10 melanoma which is used widely in immunotherapy studies. In addition, they offer insight into the extent of the immunogenicity of nonsynonymous base substitution mutations. Lastly, they argue that the use of deep sequencing to systematically analyze immunogenicity mutations may pave the way for individualized immunotherapy of cancer patients.
Collapse
Affiliation(s)
- John C Castle
- TRON-Translational Oncology at the University Medical Center Mainz, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
66
|
Jongsma M, Matas-Rico E, Rzadkowski A, Jalink K, Moolenaar WH. LPA is a chemorepellent for B16 melanoma cells: action through the cAMP-elevating LPA5 receptor. PLoS One 2011; 6:e29260. [PMID: 22195035 PMCID: PMC3237609 DOI: 10.1371/journal.pone.0029260] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2011] [Accepted: 11/23/2011] [Indexed: 01/08/2023] Open
Abstract
Lysophosphatidic acid (LPA), a lipid mediator enriched in serum, stimulates cell migration, proliferation and other functions in many cell types. LPA acts on six known G protein-coupled receptors, termed LPA1–6, showing both overlapping and distinct signaling properties. Here we show that, unexpectedly, LPA and serum almost completely inhibit the transwell migration of B16 melanoma cells, with alkyl-LPA(18∶1) being 10-fold more potent than acyl-LPA(18∶1). The anti-migratory response to LPA is highly polarized and dependent on protein kinase A (PKA) but not Rho kinase activity; it is associated with a rapid increase in intracellular cAMP levels and PIP3 depletion from the plasma membrane. B16 cells express LPA2, LPA5 and LPA6 receptors. We show that LPA-induced chemorepulsion is mediated specifically by the alkyl-LPA-preferring LPA5 receptor (GPR92), which raises intracellular cAMP via a noncanonical pathway. Our results define LPA5 as an anti-migratory receptor and they implicate the cAMP-PKA pathway, along with reduced PIP3 signaling, as an effector of chemorepulsion in B16 melanoma cells.
Collapse
Affiliation(s)
- Maikel Jongsma
- Division of Cell Biology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Elisa Matas-Rico
- Division of Cell Biology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Adrian Rzadkowski
- Division of Cell Biology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Kees Jalink
- Division of Cell Biology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Wouter H. Moolenaar
- Division of Cell Biology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- * E-mail:
| |
Collapse
|
67
|
Mijatovic S, Maksimovic-Ivanic D, Mojic M, Timotijevic G, Miljkovic D, Mangano K, Donia M, Di Cataldo A, Al-Abed Y, Cheng KF, Stosic-Grujicic S, Nicoletti F. Cytotoxic and immune-sensitizing properties of nitric oxide-modified saquinavir in iNOS-positive human melanoma cells. J Cell Physiol 2011; 226:1803-12. [DOI: 10.1002/jcp.22513] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
68
|
Cryptotanshinone has diverse effects on cell cycle events in melanoma cell lines with different metastatic capacity. Cancer Chemother Pharmacol 2010; 68:17-27. [PMID: 20820782 DOI: 10.1007/s00280-010-1440-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2010] [Accepted: 08/16/2010] [Indexed: 10/19/2022]
Abstract
PURPOSE Cryptotanshinone is a major active component of Salvia miltiorrhiza, which is often used as Chinese herbal medicine in cancer therapy. Here, we systematically assessed the anti-tumor effect of Cryptotanshinone on two melanoma cell lines with low/high-metastatic capacity (B16/B16BL6). METHODS MTT and LDH assays were used to evaluate cell growth and cytotoxicity. We assessed the effect of Cryptotanshinone on cell apoptosis or proliferation by Annexin V, TUNEL or BrdU assay. Cell cycle distribution was detected by flow cytometry. The integrity of cell cycle checkpoints was determined by mutational analyses of B-RAF and N-RAS, and the expression of cell cycle-associated proteins by western blotting. RESULTS Treatment with Cryptotanshinone had no obvious effect on cell apoptosis but significantly inhibited cell proliferation. Cryptotanshinone slightly increased the expression of p53, Chk1, and Chk2 in both B16 and B16BL6. Interestingly, Cryptotanshinone induced G1 arrest with a concomitant increase in p21 expression in B16BL6 cells. However, in B16 cells, Cryptotanshinone induced the G2/M arrest through its induction of Cdc25c. Regulation of Cyclin A1, Cyclin B1 and Cdk1/cdc2 expression might contribute to the different cell cycle patterns in B16 and B16BL6 after Cryptotanshinone treatment. CONCLUSIONS Cryptotanshinone could have diverse effects on cell cycle events in melanoma cell lines with different metastatic capacity. This property might offer an opportunity to study underlying mechanisms for the different antitumor effects of administered Cryptotanshinone in B16 and B16BL6 cells.
Collapse
|
69
|
Vance KW, Shaw HM, Rodriguez M, Ott S, Goding CR. The retinoblastoma protein modulates Tbx2 functional specificity. Mol Biol Cell 2010; 21:2770-9. [PMID: 20534814 PMCID: PMC2912361 DOI: 10.1091/mbc.e09-12-1029] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
This study demonstrates that Tbx2 binds Rb1. The interaction with Rb1 increases Tbx2 DNA-binding activity and enhances the ability of Tbx2 to repress transcription. The results show that Tbx2 regulates the expression of genes involved in cell division and DNA replication and that Rb1 modulates Tbx2 target gene recognition and specificity. Tbx2 is a member of a large family of transcription factors defined by homology to the T-box DNA-binding domain. Tbx2 plays a key role in embryonic development, and in cancer through its capacity to suppress senescence and promote invasiveness. Despite its importance, little is known of how Tbx2 is regulated or how it achieves target gene specificity. Here we show that Tbx2 specifically associates with active hypophosphorylated retinoblastoma protein (Rb1), a known regulator of many transcription factors involved in cell cycle progression and cellular differentiation, but not with the Rb1-related proteins p107 or p130. The interaction with Rb1 maps to a domain immediately carboxy-terminal to the T-box and enhances Tbx2 DNA binding and transcriptional repression. Microarray analysis of melanoma cells expressing inducible dominant-negative Tbx2, comprising the T-box and either an intact or mutated Rb1 interaction domain, shows that Tbx2 regulates the expression of many genes involved in cell cycle control and that a mutation which disrupts the Rb1-Tbx2 interaction also affects Tbx2 target gene selectivity. Taken together, the data show that Rb1 is an important determinant of Tbx2 functional specificity.
Collapse
Affiliation(s)
- Keith W Vance
- Department of Systems Biology, Biomedical Research Institute, University of Warwick, Coventry, CV4 7AL, United Kingdom.
| | | | | | | | | |
Collapse
|
70
|
Kundu S, Fan K, Cao M, Lindner DJ, Zhao ZJ, Borden E, Yi T. Novel SHP-1 inhibitors tyrosine phosphatase inhibitor-1 and analogs with preclinical anti-tumor activities as tolerated oral agents. THE JOURNAL OF IMMUNOLOGY 2010; 184:6529-36. [PMID: 20421638 DOI: 10.4049/jimmunol.0903562] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Src homology region 2 domain-containing phosphatase 1 (SHP-1) has been implicated as a potential cancer therapeutic target by its negative regulation of immune cell activation and the activity of the SHP-1 inhibitor sodium stibogluconate that induced IFN-gamma(+) cells for anti-tumor action. To develop more potent SHP-1-targeted anti-cancer agents, inhibitory leads were identified from a library of 34,000 drug-like compounds. Among the leads and active at low nM for recombinant SHP-1, tyrosine phosphatase inhibitor-1 (TPI-1) selectively increased SHP-1 phospho-substrates (pLck-pY394, pZap70, and pSlp76) in Jurkat T cells but had little effects on pERK1/2 or pLck-pY505 regulated by phosphatases SHP-2 or CD45, respectively. TPI-1 induced mouse splenic-IFN-gamma(+) cells in vitro, approximately 58-fold more effective than sodium stibogluconate, and increased mouse splenic-pLck-pY394 and -IFN-gamma(+) cells in vivo. TPI-1 also induced IFN-gamma(+) cells in human peripheral blood in vitro. Significantly, TPI-1 inhibited ( approximately 83%, p < 0.002) the growth of B16 melanoma tumors in mice at a tolerated oral dose in a T cell-dependent manner but had little effects on B16 cell growth in culture. TPI-1 also inhibited B16 tumor growth and prolonged tumor mice survival as a tolerated s.c. agent. TPI-1 analogs were identified with improved activities in IFN-gamma(+) cell induction and in anti-tumor actions. In particular, analog TPI-1a4 as a tolerated oral agent completely inhibited the growth of K1735 melanoma tumors and was more effective than the parental lead against MC-26 colon cancer tumors in mice. These results designate TPI-1 and the analogs as novel SHP-1 inhibitors with anti-tumor activity likely via an immune mechanism, supporting SHP-1 as a novel target for cancer treatment.
Collapse
Affiliation(s)
- Suman Kundu
- Department of Cancer Biology, Lerner Research Institute, The Cleveland Clinic, Cleveland, OH 44195, USA
| | | | | | | | | | | | | |
Collapse
|
71
|
Chen RY, Chen HX, Lin JX, She WB, Jiang P, Xu L, Tu YT. In-vivo transfection of pcDNA3.1-IGFBP7 inhibits melanoma growth in mice through apoptosis induction and VEGF downexpression. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2010; 29:13. [PMID: 20158915 PMCID: PMC2844372 DOI: 10.1186/1756-9966-29-13] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2009] [Accepted: 02/16/2010] [Indexed: 02/05/2023]
Abstract
BACKGROUND Genome-wide RNA interference screening study revealed that loss of expression of insulin-like growth factor binding protein 7 (IGFBP7) is a critical step in development of a malignant melanoma (MM), and this secreted protein plays a central role in apoptosis of MM. In this study we constructed pcDNA3.1-IGFBP7 to obtain high expression of IGBPF7 and to inhibit the growth of MM in C57BL/6J mice. METHODS pcDNA3.1-IGFBP7 was transfected into B16-F10 cell, the expression of IGFBP7 was detected by RT-PCR and western blot. The proliferations and apoptosis rates of transfected and control cells were measured by CCK8 and FCM, respectively. The tumorigenicity and tumor growth in both pcDNA3.1-IGFBP7 group and control groups were studied in C57BL/6J mice model. IGFBP7, caspase-3, and VEGF expressions in tumor tissue were measured by immunohistochemistry. Apoptosis of tumors were detected by TUNEL. RESULTS We demonstrated this plasmid inhibited proliferation of B16-F10 melanoma cells efficiently in vivo, exploiting the high expression of IGFBP7. More importantly, in-vivo transfection of pcDNA3.1-IGFBP7 inhibited MM growth in C57BL/6J mice. The inhibition of MM growth was proved owing to apoptosis and reduced expression of VEGF induced by pcDNA3.1-IGFBP7. CONCLUSIONS These results suggest a potential new clinical strategy for MM gene treatment.
Collapse
Affiliation(s)
- Rong-Yi Chen
- Department of Dermatology, Affiliated Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 PR China
| | | | | | | | | | | | | |
Collapse
|
72
|
p53 Protein and Pathogenesis of Melanoma and Nonmelanoma Skin Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2009; 624:265-82. [DOI: 10.1007/978-0-387-77574-6_21] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
73
|
Estler M, Boskovic G, Denvir J, Miles S, Primerano DA, Niles RM. Global analysis of gene expression changes during retinoic acid-induced growth arrest and differentiation of melanoma: comparison to differentially expressed genes in melanocytes vs melanoma. BMC Genomics 2008; 9:478. [PMID: 18847503 PMCID: PMC2572629 DOI: 10.1186/1471-2164-9-478] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2008] [Accepted: 10/11/2008] [Indexed: 11/28/2022] Open
Abstract
Background The incidence of malignant melanoma has significantly increased over the last decade. Some of these malignancies are susceptible to the growth inhibitory and pro-differentiating effects of all-trans-retinoic acid (RA). The molecular changes responsible for the biological activity of RA in melanoma are not well understood. Results In an analysis of sequential global gene expression changes during a 4–48 h RA treatment of B16 mouse melanoma cells, we found that RA increased the expression of 757 genes and decreased the expression of 737 genes. We also compared the gene expression profile (no RA treatment) between non-malignant melan-a mouse melanocytes and B16 melanoma cells. Using the same statistical test, we found 1495 genes whose expression was significantly higher in melan-a than in B16 cells and 2054 genes whose expression was significantly lower in melan-a than in B16 cells. By intersecting these two gene sets, we discovered a common set of 233 genes whose RNA levels were significantly different between B16 and melan-a cells and whose expression was altered by RA treatment. Within this set, RA treatment altered the expression of 203 (87%) genes toward the melan-a expression level. In addition, hierarchical clustering showed that after 48 h of RA treatment expression of the 203 genes was more closely related to the melan-a gene set than any other RA treatment time point. Functional analysis of the 203 gene set indicated that RA decreased expression of mRNAs that encode proteins involved in cell division/cell cycle, DNA replication, recombination and repair, and transcription regulation. Conversely, it stimulated genes involved in cell-cell signaling, cell adhesion and cell differentiation/embryonic development. Pathway analysis of the 203 gene set revealed four major hubs of connectivity: CDC2, CHEK1, CDC45L and MCM6. Conclusion Our analysis of common genes in the 48 h RA-treatment of B16 melanoma cells and untreated B16 vs. melan-a data set show that RA "normalized" the expression of genes involved in energy metabolism, DNA replication, DNA repair and differentiation. These results are compatible with the known growth inhibitory and pro-differentiating effects of RA. Pathway analysis suggests that CDC2, CHEK1, CDC45L and MCM6 are key players in mediating the biological activity of RA in B16 melanoma cells.
Collapse
Affiliation(s)
- Mary Estler
- Department of Biochemistry and Microbiology, Joan C. Edwards School of Medicine, Marshall University, One John Marshall Drive - BBSC, Huntington, WV 25755, USA.
| | | | | | | | | | | |
Collapse
|
74
|
Acid Sphingomyelinase Overexpression Enhances the Antineoplastic Effects of Irradiation In Vitro and In Vivo. Mol Ther 2008; 16:1565-71. [DOI: 10.1038/mt.2008.145] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
|
75
|
Liao YH, Hsu SM, Huang PH. ARMS Depletion Facilitates UV Irradiation Induced Apoptotic Cell Death in Melanoma. Cancer Res 2007; 67:11547-56. [DOI: 10.1158/0008-5472.can-07-1930] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
76
|
Abstract
Human melanoma represents the fastest growing malignancy in the US. The etiology of melanoma is highly debated as is the role of ultraviolet (UV) radiation in the initiation and progression of melanoma. This article discusses data from UV exposure and its relationship to the development of melanoma from various models of melanoma as well as various genetic alterations seen in oncogenic transformation of melanocytes. Genetic alterations such as the p16(INK4a) deletion, melanocortin 1 receptor (MC1R), RAS, and v-raf murine sarcoma viral oncogene homolog B1 (BRAF) may be indicative of a predisposition to melanoma development. Historical research as well as current data on the significance of the hot spot mutation in BRAF is discussed in its relative potential to the activating mutation in RAS.
Collapse
Affiliation(s)
- Cara L Benjamin
- Department of Immunology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030, USA
| | | | | |
Collapse
|
77
|
Chiu KP, Ariyaratne P, Xu H, Tan A, Ng P, Liu ETB, Ruan Y, Wei CL, Sung WKK. Pathway aberrations of murine melanoma cells observed in Paired-End diTag transcriptomes. BMC Cancer 2007; 7:109. [PMID: 17594473 PMCID: PMC1929113 DOI: 10.1186/1471-2407-7-109] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2007] [Accepted: 06/26/2007] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Melanoma is the major cause of skin cancer deaths and melanoma incidence doubles every 10 to 20 years. However, little is known about melanoma pathway aberrations. Here we applied the robust Gene Identification Signature Paired End diTag (GIS-PET) approach to investigate the melanoma transcriptome and characterize the global pathway aberrations. METHODS GIS-PET technology directly links 5' mRNA signatures with their corresponding 3' signatures to generate, and then concatenate, PETs for efficient sequencing. We annotated PETs to pathways of KEGG database and compared the murine B16F1 melanoma transcriptome with three non-melanoma murine transcriptomes (Melan-a2 melanocytes, E14 embryonic stem cells, and E17.5 embryo). Gene expression levels as represented by PET counts were compared across melanoma and melanocyte libraries to identify the most significantly altered pathways and investigate the expression levels of crucial cancer genes. RESULTS Melanin biosynthesis genes were solely expressed in the cells of melanocytic origin, indicating the feasibility of using the PET approach for transcriptome comparison. The most significantly altered pathways were metabolic pathways, including upregulated pathways: purine metabolism, aminophosphonate metabolism, tyrosine metabolism, selenoamino acid metabolism, galactose utilization, nitrobenzene degradation, and bisphenol A degradation; and downregulated pathways: oxidative phosphorylation, ATPase synthesis, TCA cycle, pyruvate metabolism, and glutathione metabolism. The downregulated pathways concurrently indicated a slowdown of mitochondrial activities. Mitochondrial permeability was also significantly altered, as indicated by transcriptional activation of ATP/ADP, citrate/malate, Mg++, fatty acid and amino acid transporters, and transcriptional repression of zinc and metal ion transporters. Upregulation of cell cycle progression, MAPK, and PI3K/Akt pathways were more limited to certain region(s) of the pathway. Expression levels of c-Myc and Trp53 were also higher in melanoma. Moreover, transcriptional variants resulted from alternative transcription start sites or alternative polyadenylation sites were found in Ras and genes encoding adhesion or cytoskeleton proteins such as integrin, beta-catenin, alpha-catenin, and actin. CONCLUSION The highly correlated results unmistakably point to a systematic downregulation of mitochondrial activities, which we hypothesize aims to downgrade the mitochondria-mediated apoptosis and the dependency of cancer cells on angiogenesis. Our results also demonstrate the advantage of using the PET approach in conjunction with KEGG database for systematic pathway analysis.
Collapse
Affiliation(s)
- Kuo Ping Chiu
- Genome Institute of Singapore, 60 Biopolis Street, Genome #02-01, 138672, Singapore
| | - Pramila Ariyaratne
- Genome Institute of Singapore, 60 Biopolis Street, Genome #02-01, 138672, Singapore
| | - Han Xu
- Genome Institute of Singapore, 60 Biopolis Street, Genome #02-01, 138672, Singapore
| | - Adrian Tan
- Genome Institute of Singapore, 60 Biopolis Street, Genome #02-01, 138672, Singapore
| | - Patrick Ng
- Genome Institute of Singapore, 60 Biopolis Street, Genome #02-01, 138672, Singapore
| | - Edison Tak-Bun Liu
- Genome Institute of Singapore, 60 Biopolis Street, Genome #02-01, 138672, Singapore
| | - Yijun Ruan
- Genome Institute of Singapore, 60 Biopolis Street, Genome #02-01, 138672, Singapore
| | - Chia-Lin Wei
- Genome Institute of Singapore, 60 Biopolis Street, Genome #02-01, 138672, Singapore
| | - Wing-Kin Ken Sung
- Genome Institute of Singapore, 60 Biopolis Street, Genome #02-01, 138672, Singapore
- Department of Computer Science, National University of Singapore, 3 Science Drive 2, 117543, Singapore
| |
Collapse
|
78
|
Oppitz M, Busch C, Schriek G, Metzger M, Just L, Drews U. Non-malignant migration of B16 mouse melanoma cells in the neural crest and invasive growth in the eye cup of the chick embryo. Melanoma Res 2007; 17:17-30. [PMID: 17235238 DOI: 10.1097/cmr.0b013e3280114f49] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Melanocytes originate from the neural crest. In a previous study, we observed that human SK-Mel 28 human melanoma cells resumed neural crest cell migration after transplantation into the chick embryo neural tube. Here, we used transgenic mouse B16-F1 melanoma cells transfected with green fluorescent protein-vasodilator-stimulated phosphoprotein construct to extend these observations. After the injection of a cell suspension into the trunk neural tube of E2 chick embryos, the migration of melanoma cells was followed by live fluorescence microscopy. Within 12 h, the melanoma cells formed clusters in the neural tube at the levels of the intersegmental clefts between somites. After 24 h, a segmental pattern of emigration was visible. Emigrated melanoma cells were identified in serial paraffin sections by immunohistochemistry with ab732 as a marker for melanoma cells and by in-situ hybridization of mouse-specific repetitive genomic sequence mL1. After 24 h, melanoma cells were found along the medial neural crest pathway and in the sympathetic trunk ganglia and, after 48 h, also in the lateral melanocytic pathway. During migration along the neural crest pathways, mouse melanoma cells underwent apoptosis, which was assessed by anti-caspase 3 and terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate-biotin nick-end labeling staining. To prove the ablation of malignant behavior after back-transplantation into the original embryonic neural crest environment, we injected the same cell suspension into the eye cup of the E3 embryo. In this location, invasive melanomas formed.
Collapse
Affiliation(s)
- Matthias Oppitz
- Department of Experimental Embryology, Institute of Anatomy, University of Tübingen, Tübingen, Germany
| | | | | | | | | | | |
Collapse
|
79
|
Murphy DA, Makonnen S, Lassoued W, Feldman MD, Carter C, Lee WMF. Inhibition of tumor endothelial ERK activation, angiogenesis, and tumor growth by sorafenib (BAY43-9006). THE AMERICAN JOURNAL OF PATHOLOGY 2006; 169:1875-85. [PMID: 17071608 PMCID: PMC1780219 DOI: 10.2353/ajpath.2006.050711] [Citation(s) in RCA: 121] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Activation of the Raf-MEK-ERK signal transduction pathway in endothelial cells is required for angiogenesis. Raf is the kinase most efficiently inhibited by the multikinase inhibitor sorafenib, which has shown activity against certain human cancers in clinical trials. To understand the mechanisms underlying this activity, we studied how it controlled growth of K1735 murine melanomas. Therapy caused massive regional tumor cell death accompanied by severe tumor hypoxia, decreased microvessel density, increased percentage of pericyte-covered vessels, and increased caliber and decreased arborization of vessels. These signs of K1735 angiogenesis inhibition, along with its ability to inhibit Matrigel neovascularization, showed that sorafenib is an effective anti-angiogenic agent. Extracellular signal-regulated kinase (ERK) activation in tumor endothelial cells, revealed by immunostaining for phospho-ERK and CD34, was inhibited, whereas AKT activation, revealed by phospho-AKT immunostaining, was not inhibited in K1735 and two other tumor types treated with sorafenib. Treatment decreased endothelial but not tumor cell proliferation and increased both endothelial cell and tumor cell apoptosis. These data indicate that sorafenib's anti-tumor efficacy may be primarily attributable to angiogenesis inhibition resulting from its inhibition of Raf-MEK-ERK signaling in endothelial cells. Assessing endothelial cell ERK activation in tumor bio-psies may provide mechanistic insights into and allow monitoring of sorafenib's activity in patients in clinical trials.
Collapse
Affiliation(s)
- Danielle A Murphy
- Biomedical Graduate Program, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | | | | | | | | |
Collapse
|
80
|
Culp WD, Neal R, Massey R, Egevad L, Pisa P, Garland D. Proteomic analysis of tumor establishment and growth in the B16-F10 mouse melanoma model. J Proteome Res 2006; 5:1332-43. [PMID: 16739985 DOI: 10.1021/pr060059q] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The B16-F10 mouse model of melanoma is a widely used model to study many aspects of cancer biology and therapeutics in a solid tumor. Melanomas aggressively progress within a dynamic microenvironment containing in addition to tumor cells, stroma cells and components such as fibroblasts, immune cells, vascular cells, extracellular matrix (ECM) and extracellular molecules. The goal of this study was to elucidate the processes of tumor progression by identifying differentially expressed proteins in the tumor mass during specific stages of tumor growth. A comparative proteome analysis was performed on B16-F10 derived tumors in C57BL/6 mice at days 3, 5, 7, and 10. Statistical approaches were used to determine quantitative differential protein expression at each tumor time stage. Hierarchical clustering of 44 protein spots (p < 0.01) revealed a progressive change in the tumor mass when all 4 time stages were classified together, but there was a clear switch in expression of these proteins between the day 5 and the day 7 tumors. A trend analysis showed 53 protein spots (p < 0.001) following 6 predominant kinetic paths of expression as the tumor progressed. The protein spots were then identified using MALDI-TOF mass spectrometry. Proteins involved in glycolysis, inflammation, wounding, superoxide metabolism, and chemotaxis increased during tumorigenesis. From day 3 to day 7 VEGF and active cathepsin D were induced 7-fold and 4-fold, respectively. Proteins involved in electron transport, protein folding, blood coagulation, and transport decreased during tumorigenesis. This work illustrates changes in the biology of the B16-F10 tumor mass during tumor progression.
Collapse
Affiliation(s)
- W David Culp
- Protein Biochemistry Section, National Eye Institute, National Institutes of Health, Bethesda, Maryland 20892, USA.
| | | | | | | | | | | |
Collapse
|
81
|
Hölsken A, Eyüpoglu IY, Lueders M, Tränkle C, Dieckmann D, Buslei R, Hahnen E, Blümcke I, Siebzehnrübl FA. Ex vivo therapy of malignant melanomas transplanted into organotypic brain slice cultures using inhibitors of histone deacetylases. Acta Neuropathol 2006; 112:205-15. [PMID: 16773328 DOI: 10.1007/s00401-006-0082-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2005] [Revised: 04/28/2006] [Accepted: 04/28/2006] [Indexed: 12/17/2022]
Abstract
Disease progression in patients suffering from malignant melanomas is often determined by metastatic spreading into brain parenchyma. Systemic chemotherapy regimens are, therefore, mandatory for successful treatment. Most recently, inhibitors of histone deacetylases (HDACi) have been shown to significantly inhibit melanoma progression. Here, mouse as well as human melanoma cells were transplanted into rodent hippocampal slice cultures in order to translate and microscopically confirm promising in vitro chemotherapeutic propensities of HDACi within the organotypic brain environment. In our ex vivo model, tumor progression was significantly inhibited by administration of low micromolar concentrations of second generation HDACi MS-275 over a period of 8 days. In contrast, HDACi treatment with suberoylanilide hydroxamic acid was less efficient ex vivo, although both compounds were successful in the treatment of tumor cell monolayer cultures. Protein levels of the cell cycle inhibitor p21(WAF1) were significantly increased after HDACi treatment, which points to enhanced G1 arrest of tumor cells as confirmed by cytofluorometric analysis. Considering the ability of MS-275 to cross the blood-brain barrier, our experimental model identifies the benzamide MS-275 as a promising therapeutic compound for targeting epigenetic chromatin modulation as systemic treatment of metastatic melanomas.
Collapse
Affiliation(s)
- Annett Hölsken
- Department of Neuropathology, University of Erlangen-Nuremberg, Krankenhausstr. 8-10, 91054, Erlangen, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
82
|
Arai K, Matsumoto Y, Nagashima Y, Yagasaki K. Regulation of Class II β-Tubulin Expression by Tumor Suppressor p53 Protein in Mouse Melanoma Cells in Response toVincaAlkaloid. Mol Cancer Res 2006; 4:247-55. [PMID: 16603638 DOI: 10.1158/1541-7786.mcr-05-0183] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The continuous exposure of antimicrotubule drugs to tumors often results in the emergence of drug-resistant tumor cells with altered expression of several beta-tubulin isotypes. We found that Vinca alkaloid enhanced expression of class II beta-tubulin isotype (mTUBB2) in mouse B16F10 melanoma cells via alteration of the tumor suppressor p53 protein. Vincristine treatment stimulated an increase in mTUBB2 mRNA expression and promoted accumulation of this isotype around the nuclei. Transient transfection assays employing a reporter construct, together with site-directed mutagenesis studies, suggested that the p53-binding site found in the first intron was a critical region for mTUBB2 expression. Electrophoretic mobility shift assay and associated antibody supershift experiments showed that vincristine promoted release of p53 protein from the binding site. In addition, exogenous induction of TAp63gamma (p51A), a homologue of p53, canceled the effect of vincristine on mTUBB2 expression. These results suggest that p53 protein may function as a suppressor of mTUBB2 expression and vincristine-mediated inhibition of p53 binding results in enhanced mTUBB2 expression. This phenomenon could be related with the emergence of drug-resistant tumor cells induced by Vinca alkaloid and may participate in determining the fate of these cells.
Collapse
Affiliation(s)
- Katsuhiko Arai
- Department of Tissue Physiology, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Saiwai-cho 3-5-8, Fuchu, Tokyo 183-8509, Japan.
| | | | | | | |
Collapse
|
83
|
Abstract
Protein kinase C (PKC) is activated by diacylglycerol generated by receptor-mediated hydrolysis of membrane phospholipids to mediate signals for cell growth and plays as a target of tumor-promoting phorbol esters in malignant transformation. PKC is a family of enzymes and their expression profiles have been examined in the normal melanocytes and melanoma cells, and studies have been carried out on the functions of PKC isoforms in proliferation, transformation, and metastasis of melanoma cells. Here, we summarize current knowledge of the expression and possible roles of the PKC family in melanoma in comparison with those of normal melanocytes.
Collapse
Affiliation(s)
- Masahiro Oka
- Division of Dermatology, Department of Clinical Molecular Medicine, Kobe University Graduate School of Medicine, Kobe, 650-0017, Japan.
| | | |
Collapse
|
84
|
Vance KW, Carreira S, Brosch G, Goding CR. Tbx2 Is Overexpressed and Plays an Important Role in Maintaining Proliferation and Suppression of Senescence in Melanomas. Cancer Res 2005; 65:2260-8. [PMID: 15781639 DOI: 10.1158/0008-5472.can-04-3045] [Citation(s) in RCA: 171] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The INK4a and ARF genes found at the CDKN2A locus are key effectors of cellular senescence that is believed to act as a powerful anticancer mechanism. Accordingly, mutations in these genes are present in a wide variety of spontaneous human cancers and CDKN2A germ line mutations are found in familial melanoma. The TBX2 gene encoding a key developmental transcription factor is amplified in pancreatic cancer cell lines and preferentially amplified and overexpressed in BRCA1 and BRCA2 mutated breast tumors. Overexpression of Tbx2 and the related factor Tbx3, which is also overexpressed in breast cancer and melanomas, can suppress senescence in defined experimental systems through repression of ARF expression. However, it is not known how Tbx2 mediates its repressive effect nor whether endogenous Tbx2 or Tbx3 perform a similar antisenescence function in transformed cells. This is a particularly important question because the loss of CDKN2A in many human cancers would, in principle, bypass the requirement for Tbx2/3-mediated repression of ARF in suppressing senescence. We show here that Tbx2 is overexpressed in melanoma cell lines and that Tbx2 targets histone deacetylase 1 to the p21Cip1 (CDKN1A) initiator. Strikingly, expression of an inducible dominant-negative Tbx2 (dnTbx2) leads to displacement of histone deacetylase 1, up-regulation of p21(Cip1) expression, and the induction of replicative senescence in CDKN2A-null B16 melanoma cells. In human melanoma cells, expression of dnTbx2 leads to severely reduced growth and induction of senescence-associated heterochromatin foci. The results suggest that the activity of endogenous Tbx2 is critically required to maintain proliferation and suppress senescence in melanomas.
Collapse
Affiliation(s)
- Keith W Vance
- Signaling and Development Laboratory, Marie Curie Research Institute, Surrey, United Kingdom
| | | | | | | |
Collapse
|
85
|
Hussein MR. The TP53 Tumor Suppressor Gene and Melanoma Tumorigenesis: Is There a Relationship? Tumour Biol 2004; 25:200-7. [PMID: 15557758 DOI: 10.1159/000081103] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2004] [Accepted: 04/05/2004] [Indexed: 01/26/2023] Open
Abstract
Mutations in the TP53 gene are found in about 11% of melanomas. Although nearly 600 papers have been published with varying degrees of consensus, there does not appear to be any comparable analysis that facilitates more than a glimpse into the role of p53 in melanomagenesis. This article reviews p53 alterations (at the gene and protein levels) in melanocytic skin lesions and discusses the following points: (i) p53 alterations commence as early as at the stage of benign and dysplastic nevi; (ii) these alterations are frequent in melanomas, and gradually increase with their progression; (iii) there is no concordance between the frequent p53 protein expression and the rarity of both TP53 gene mutations in melanomas, and (iv) the entire p53 pathway is a more critical determinant of the fate of the melanocytic skin lesions than the status of the p53 protein or the gene itself.
Collapse
Affiliation(s)
- Mahmoud R Hussein
- Department of Pathology, Faculty of Medicine, Assuit University, Assuit City, Egypt.
| |
Collapse
|