1
|
Strashilov S, Yordanov A. Aetiology and Pathogenesis of Cutaneous Melanoma: Current Concepts and Advances. Int J Mol Sci 2021; 22:6395. [PMID: 34203771 PMCID: PMC8232613 DOI: 10.3390/ijms22126395] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/09/2021] [Accepted: 06/11/2021] [Indexed: 12/18/2022] Open
Abstract
Melanoma develops from malignant transformations of the pigment-producing melanocytes. If located in the basal layer of the skin epidermis, melanoma is referred to as cutaneous, which is more frequent. However, as melanocytes are be found in the eyes, ears, gastrointestinal tract, genitalia, urinary system, and meninges, cases of mucosal melanoma or other types (e.g., ocular) may occur. The incidence and morbidity of cutaneous melanoma (cM) are constantly increasing worldwide. Australia and New Zealand are world leaders in this regard with a morbidity rate of 54/100,000 and a mortality rate of 5.6/100,000 for 2015. The aim of this review is to consolidate and present the data related to the aetiology and pathogenesis of cutaneous melanoma, thus rendering them easier to understand. In this article we will discuss these problems and the possible impacts on treatment for this disease.
Collapse
Affiliation(s)
- Strahil Strashilov
- Department of Plastic Restorative, Reconstructive and Aesthetic Surgery, University Hospital “Dr. Georgi Stranski”, Medical University Pleven, 5800 Pleven, Bulgaria
| | - Angel Yordanov
- Clinic of Gynecologic Oncology, University Hospital “Dr. Georgi Stranski”, Medical University Pleven, 5800 Pleven, Bulgaria;
| |
Collapse
|
2
|
Guasch G. [Animal models for the study of cancer stem cells]. Bull Cancer 2017; 104:1064-1067. [PMID: 29157554 DOI: 10.1016/j.bulcan.2017.10.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Accepted: 10/19/2017] [Indexed: 10/18/2022]
Affiliation(s)
- Géraldine Guasch
- Centre de recherche en cancérologie de Marseille (CRCM), Inserm, U1068, 13009 Marseille, France; CNRS, UMR7258, 13009 Marseille, France; Institut Paoli-Calmettes, 13009 Marseille, France; Aix-Marseille université, UM 105, 13284 Marseille, France.
| |
Collapse
|
3
|
Wu JS, Chang JY, Chen CW, Lin MT, Sheu DC, Lee SM. Neokestose suppresses the growth of human melanoma A2058 cells via inhibition of the nuclear factor-κB signaling pathway. Mol Med Rep 2017; 16:295-300. [DOI: 10.3892/mmr.2017.6594] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 02/21/2017] [Indexed: 11/05/2022] Open
|
4
|
Silva JM, Deuker MM, Baguley BC, McMahon M. PIK3CA-mutated melanoma cells rely on cooperative signaling through mTORC1/2 for sustained proliferation. Pigment Cell Melanoma Res 2017; 30:353-367. [PMID: 28233937 DOI: 10.1111/pcmr.12586] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 02/20/2017] [Indexed: 01/01/2023]
Abstract
Malignant conversion of BRAF- or NRAS-mutated melanocytes into melanoma cells can be promoted by PI3'-lipid signaling. However, the mechanism by which PI3'-lipid signaling cooperates with mutationally activated BRAF or NRAS has not been adequately explored. Using human NRAS- or BRAF-mutated melanoma cells that co-express mutationally activated PIK3CA, we explored the contribution of PI3'-lipid signaling to cell proliferation. Despite mutational activation of PIK3CA, melanoma cells were more sensitive to the biochemical and antiproliferative effects of broader spectrum PI3K inhibitors than to an α-selective PI3K inhibitor. Combined pharmacological inhibition of MEK1/2 and PI3K signaling elicited more potent antiproliferative effects and greater inhibition of the cell division cycle compared to single-agent inhibition of either pathway alone. Analysis of signaling downstream of MEK1/2 or PI3K revealed that these pathways cooperate to regulate cell proliferation through mTORC1-mediated effects on ribosomal protein S6 and 4E-BP1 phosphorylation in an AKT-dependent manner. Although PI3K inhibition resulted in cytostatic effects on xenografted NRASQ61H /PIK3CAH1047R melanoma, combined inhibition of MEK1/2 plus PI3K elicited significant melanoma regression. This study provides insights as to how mutationally activated PIK3CA acts in concert with MEK1/2 signaling to cooperatively regulate mTORC1/2 to sustain PIK3CA-mutated melanoma proliferation.
Collapse
Affiliation(s)
- Jillian M Silva
- Helen Diller Family Comprehensive Cancer Center, Department of Cellular & Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA
| | - Marian M Deuker
- Helen Diller Family Comprehensive Cancer Center, Department of Cellular & Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA
| | - Bruce C Baguley
- Auckland Cancer Society Research Centre, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Martin McMahon
- Helen Diller Family Comprehensive Cancer Center, Department of Cellular & Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
5
|
Cicenas J, Tamosaitis L, Kvederaviciute K, Tarvydas R, Staniute G, Kalyan K, Meskinyte-Kausiliene E, Stankevicius V, Valius M. KRAS, NRAS and BRAF mutations in colorectal cancer and melanoma. Med Oncol 2017; 34:26. [DOI: 10.1007/s12032-016-0879-9] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Accepted: 12/29/2016] [Indexed: 01/13/2023]
|
6
|
Teas J, Irhimeh MR. Melanoma and brown seaweed: an integrative hypothesis. JOURNAL OF APPLIED PHYCOLOGY 2017; 29:941-948. [PMID: 28458463 PMCID: PMC5387006 DOI: 10.1007/s10811-016-0979-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 10/01/2016] [Accepted: 10/02/2016] [Indexed: 05/08/2023]
Abstract
Although relatively rare, melanoma accounts for 2 % of cancer diagnoses globally and accounts for about 1 % of all cancer deaths. Worldwide, the annual incidence of melanoma is 272,000 cases which vary hugely, ranging from Japan where it is incredibly infrequent, to Queensland, Australia, where it is nearly 100 times higher. Based on epidemiology and laboratory studies, there is compelling evidence suggesting that seaweed might be protective against different types of cancers such as breast cancer in seaweed consuming populations. By comparing countries where melanoma is more common with countries where it is infrequent, it is possible to construct a hypothesis for how consuming brown seaweeds which may hold clues to the differences in melanoma susceptibility between Japanese and Western nations. Unlike in these other countries, where melanoma incidence has increased dramatically over the last two decades, in Japan, rates have remained remarkably low and stable. There is limited evidence from clinical studies and animal models that have used whole seaweed or isolated fractions from seaweed and measured changes in biomarkers. They have demonstrated the effectiveness of seaweed at inhibiting melanoma development and progression. In this review, the various results will be described. Although there are several effective fractions, it is proposed that consuming whole seaweeds may hold additional benefits that could be lost by consuming only a single extract.
Collapse
Affiliation(s)
- Jane Teas
- Thomas Cooper Library Fellow, University of South Carolina, Columbia, SC 29208 USA
| | - Mohammad R. Irhimeh
- Cell and Tissue Therapies Western Australia, Royal Perth Hospital, Perth, WA 6000 Australia
- Faculty of Medicine, Dentistry and Health Sciences, The University of Western Australia, Crawley, Perth, WA 6009 Australia
| |
Collapse
|
7
|
Beaty MW, Quezado M, Sobel ME, Duray P, Merino MJ. Loss of Heterozygosity on Chromosome 1 and 9 and Hormone Receptor Analysis of Metastatic Malignant Melanoma Presenting in Breast. Int J Surg Pathol 2016; 13:9-18. [PMID: 15735850 DOI: 10.1177/106689690501300102] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Malignant melanoma (MM), the most common metastatic solid tumor to involve the breast, may present as a diagnostic problem, frequently requiring the use of ancillary studies for accurate diagnosis. The implication of hormonal interplay is strong since metastatic MM to the breast is seen nearly always in women. However, the role of hormonal status as a predisposing factor in the development of this entity is largely unresolved. A number of chromosomal loci, including ip36 and 9p2l-22, appear to harbor critical genes important to melanoma tumorigenesis, and additionally chromosome 9q22.3-31. We wanted to know if metastatic MM in breast showed chromosome lp and 9p genetic alterations (loss of heterozygosity) similar to those that occur in primary cutaneous MM, and whether additional 9q LOH changes are present. Hormonal receptor status of the metastatic MM was also determined. We identified 20 patients with known MM metastatic to the breast, which we analyzed with the following genetic markers: D9S12 (9q22.3), D9S171 (9p21), IFNA (9p22), and DlS450 (ip). Visually directed microdissection was performed on archival histologic slides containing both tumor and adjacent normal breast epithelium, followed by single-step DNA extraction and polymerase chain reaction (PCR) amplification for evaluation of loss of heterozygosity (LOH) for the above-listed markers. Immunohistochemical (IHC) stains for estrogen receptor (ER) and progesterone receptor (PR) was performed on 10 of the cases. Twelve of the 20 cases contained DNA suitable for PCR amplification following direct visualization microdissection. Four of 8 (50%) informative cases showed LOH at 9p21 with D9S171. Ten cases were heterozygous for IFNA, with 2 cases (20%) showing LOH at this locus. These particular cases also showed LOH at 9p21. One of 9 (11%) informative cases showed LOH for DlS450 (Ip36). Five cases were heterozygous for D9S12, and 2 1-22 (50%) and lp36 (11%), as previously described in primary cutaneous MM. Additional LOH was observed at the 9q22.3-31 locus (40%). We suggest this locus to be investigated for harboring potential genes important in the tumorigenesis of cutaneous MM.
Collapse
Affiliation(s)
- Michael W Beaty
- Laboratory of Pathology, Surgical Pathology Section, National Cancer Institute, Bethesda, MD 20892, USA
| | | | | | | | | |
Collapse
|
8
|
Carter JH, Deddens JA, Spaulding NR, Lucas D, Colligan BM, Lewis TG, Hawkins E, Jones J, Pemberton JO, Douglass LE, Graff JR. Phosphorylation of eIF4E serine 209 is associated with tumour progression and reduced survival in malignant melanoma. Br J Cancer 2016; 114:444-53. [PMID: 26882068 PMCID: PMC4815768 DOI: 10.1038/bjc.2015.450] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 11/18/2015] [Accepted: 11/30/2015] [Indexed: 02/08/2023] Open
Abstract
Background: Melanoma is a disease that primarily arises in the skin but is a derivative of the neural crest. Eukaryotic translation initiation factor 4E (eIF4E) regulates translation of multiple malignancy-associated mRNAs and is overexpressed in many epithelial tumours. However, expression in human tumours derived from the neural crest is unknown. Here, we determined the association of eIF4E and phospho-eIF4E expression in melanocytic lesions with malignant conversion, metastatic potential and patient survival. Methods: Archived formalin-fixed, paraffin-embedded surgical specimens from 114 patients with melanocytic lesions were stained immunohistochemically for eIF4E and phospho-eIF4E and evaluated semiquantitatively. The relationship between cytoplasmic and nuclear eIF4E and phospho-eIF4E protein expression, melanocytic lesion subtype and tumour progression was determined. Kaplan–Meier survival analyses and Cox proportional hazard regression were performed. Results: Increased eIF4E and phospho-eIF4E expression was highly associated with malignancy (P<0.0001). High nuclear phospho-eIF4E was associated with synchronous or future metastasis (P=0.0059). Kaplan–Meier analyses demonstrated highly significant associations between high histoscores for cytoplasmic and nuclear phospho-eIF4E and reduced survival in all patients (P=0.0003 and 0.0009, respectively). Conclusions: Increased melanoma expression of eIF4E and phospho-eIF4E is associated with metastatic potential, reduced survival and increased risk of death.
Collapse
Affiliation(s)
- Julia H Carter
- Wood Hudson Cancer Research Laboratory, 931 Isabella Street, Newport, KY 41071, USA
| | - James A Deddens
- Department of Mathematical Sciences, University of Cincinnati, 4512 French Hall, ML 25, Cincinnati, OH 45221, USA
| | - Nelson Reed Spaulding
- Department of Pathology and Laboratory Medicine, University of Louisville School of Medicine, 627 South Preston Street, Louisville, KY 40292, USA
| | - Denise Lucas
- Wood Hudson Cancer Research Laboratory, 931 Isabella Street, Newport, KY 41071, USA
| | - Bruce M Colligan
- Wood Hudson Cancer Research Laboratory, 931 Isabella Street, Newport, KY 41071, USA
| | - Thomas Grant Lewis
- Wood Hudson Cancer Research Laboratory, 931 Isabella Street, Newport, KY 41071, USA
| | - Elyse Hawkins
- Wood Hudson Cancer Research Laboratory, 931 Isabella Street, Newport, KY 41071, USA
| | - Jordan Jones
- Wood Hudson Cancer Research Laboratory, 931 Isabella Street, Newport, KY 41071, USA
| | - Jackson O Pemberton
- St Elizabeth Healthcare, Department of Laboratory Medicine, 20 Medical Village Drive, Edgewood, KY 41018, USA
| | - Larry E Douglass
- Wood Hudson Cancer Research Laboratory, 931 Isabella Street, Newport, KY 41071, USA
| | - Jeremy R Graff
- Eli Lilly & Company, Lilly Corporate Center, DC 0546, Indianapolis, IN 46285, USA
| |
Collapse
|
9
|
Lee CH, Lai PS, Lu YP, Chen HY, Chai CY, Tsai RK, Fang KT, Tsai MH, Hsu CY, Hung CC, Wu DC, Yu HS, Chang CH, Tsai DP. Real-time vascular imaging and photodynamic therapy efficacy with micelle-nanocarrier delivery of chlorin e6 to the microenvironment of melanoma. J Dermatol Sci 2015; 80:124-32. [PMID: 26360010 DOI: 10.1016/j.jdermsci.2015.08.005] [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] [Received: 07/08/2015] [Revised: 08/12/2015] [Accepted: 08/28/2015] [Indexed: 01/02/2023]
Abstract
BACKGROUND Strategies combining anti-vascular therapy and vascular imaging may facilitate the prediction of early response and outcome in cancer treatment. OBJECTIVE The aim of this study was to investigate the relationship between the tumor-associated vasculature in melanoma and to develop an approach for melanoma treatment by utilizing the free form and micelle form of the photosensitizer (PS) chlorin e6 in photodynamic therapy (PDT). METHODS Green fluorescence protein (GFP) expressing B16-F10 melanoma cells were implanted into the mouse ear dermis. Ce6 in free form or in micelle form was administered via the tail vein. An OV100 imaging system was used to record the red fluorescence of Ce6 to obtain real-time vascular images in the GFP tumor. RESULTS Compared to free Ce6, Ce6 linked to the micelle-nanocarrier depicted a much clearer vascular image and had an effective vascular destruction by PDT. Micelle Ce6 was localized in lysosomes and in the endoplasmic reticulum of cultured endothelial cells, implying an active endocytosis of the nano-carrier. CONCLUSION Micelle Ce6 can serve as a bifunctional PS for vascular imaging and PDT, which facilitates its delivery in the tumor microenvironment.
Collapse
Affiliation(s)
- Chien-Hsing Lee
- Center for Stem Cell Research, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Department of Nursing, Min-Hwei Junior College of Health Care Management, Tainan 73658, Taiwan
| | - Ping-Shan Lai
- Department of Chemistry, National Chung Hsing University, Taichung 40227, Taiwan
| | - Yen-Pei Lu
- Instrument Technology Research Center, National Applied Research Laboratories, Hsinchu 30076, Taiwan
| | - Hsuan-Ying Chen
- Department of Medicine and Applied Chemistry, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Chee-Yin Chai
- Department of Pathology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Department of Pathology, Kaohsiung Medical University Chung-Ho Memorial Hospital, Kaohsiung 80708, Taiwan
| | - Rong-Kung Tsai
- Institute of Medical Sciences, Tzu Chi University, Hualien 97002, Taiwan
| | - Kang-Tang Fang
- Department of Dermatology, Buddhist Tzu Chi General Hospital, Hualien 97002, Taiwan
| | - Ming-Hsien Tsai
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Chia-Yen Hsu
- Department of Chemistry, National Chung Hsing University, Taichung 40227, Taiwan
| | - Chun-Cheng Hung
- School of Dentistry, College of Dental Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Deng-Chyang Wu
- Center for Stem Cell Research, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Department of Pathology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University, Chung-Ho Memorial Hospital, Kaohsiung 80708, Taiwan; Department of Internal Medicine, Kaohsiung Municipal Hsiao-Kang Hospital, Kaohsiung City 812, Taiwan
| | - Hsin-Su Yu
- Department of Dermatology, Kaohsiung Medical University, Chung-Ho Memorial Hospital, Kaohsiung 80708, Taiwan; National Environmental Health Research Center, National Health Research Institute, Miao-Li, Taiwan
| | - Chung-Hsing Chang
- Center for Stem Cell Research, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Department of Dermatology, Kaohsiung Medical University, Chung-Ho Memorial Hospital, Kaohsiung 80708, Taiwan; Research Center for Applied Sciences, Academia Sinica, Taipei 11529, Taiwan.
| | - Din-Ping Tsai
- Department of Physics, National Taiwan University, Taipei 10617, Taiwan; Research Center for Applied Sciences, Academia Sinica, Taipei 11529, Taiwan.
| |
Collapse
|
10
|
Silva JM, Bulman C, McMahon M. BRAFV600E cooperates with PI3K signaling, independent of AKT, to regulate melanoma cell proliferation. Mol Cancer Res 2014; 12:447-63. [PMID: 24425783 DOI: 10.1158/1541-7786.mcr-13-0224-t] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
UNLABELLED Mutationally activated BRAF(V600E) cooperates with PTEN silencing in the conversion of normal melanocytes to metastatic melanoma cells, but the mechanism underlying this cooperation is poorly understood. Here, the consequences of pharmacologic blockade of BRAF(V600E) or phosphoinositide 3-kinase (PI3K) signaling were explored using pathway-targeted inhibitors and a panel of human BRAF-mutated melanoma-derived cell lines. Blockade of BRAF(V600E) → MEK1/2 → ERK1/2 or class I PI3K inhibited melanoma proliferation, whereas inhibition of AKT had only modest effects, even in cells with mutated or amplified AKT. Although single-agent inhibition of either BRAF(V600E) or PI3K signaling elicited antiproliferative effects, combinatorial inhibition was more potent. Analysis of signaling downstream of BRAF(V600E) or PI3K revealed that these pathways cooperated to regulate protein synthesis through AKT-independent, mTOR complex 1 (mTORC1)-dependent effects on p70(S6K), ribosomal protein S6, and 4E-BP1 phosphorylation. Moreover, inhibition of mTORC1/2 inhibited cell proliferation as profoundly as single-agent inhibition of either BRAF(V600E) or PI3K signaling. These data reveal a mechanism by which BRAF(V600E) and PI3K signaling cooperate to regulate melanoma proliferation through AKT-independent effects on protein translation. Furthermore, this study provides a potential foundation for pathway-targeted combination therapy designed to enhance the therapeutic benefit to patients with melanoma that contain combined alterations in BRAF and PI3K signaling. IMPLICATIONS PI3K, but not AKT, represent potential targets for melanoma therapy.
Collapse
Affiliation(s)
- Jillian M Silva
- Diller Cancer Research Building, MC-0128, 1450 Third Street, Room HD-365, University of California, San Francisco, CA 94158.
| | | | | |
Collapse
|
11
|
Marsh Durban V, Deuker MM, Bosenberg MW, Phillips W, McMahon M. Differential AKT dependency displayed by mouse models of BRAFV600E-initiated melanoma. J Clin Invest 2013; 123:5104-18. [PMID: 24200692 PMCID: PMC3859393 DOI: 10.1172/jci69619] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Accepted: 09/03/2013] [Indexed: 01/09/2023] Open
Abstract
Malignant melanoma is frequently driven by mutational activation of v-raf murine sarcoma viral oncogene homolog B1 (BRAF) accompanied by silencing of the phosphatase and tensin homology (PTEN) tumor suppressor. Despite the implied importance of PI3K signaling in PTENNull melanomas, mutational activation of the gene encoding the catalytic subunit of PI3Kα (PIK3CA), is rarely detected. Since PTEN has both PI3-lipid phosphatase-dependent and -independent tumor suppressor activities, we investigated the contribution of PI3K signaling to BRAFV600E-induced melanomagenesis using mouse models, cultured melanoma cells, and PI3K pathway-targeted inhibitors. These experiments revealed that mutationally activated PIK3CAH1047R cooperates with BRAFV600E for melanomagenesis in mice. Moreover, pharmacological inhibition of PI3Ks prevented growth of BRAFV600E/PTENNull melanomas in vivo and in tissue culture. Combined inhibition of BRAFV600E and PI3K had more potent effects on the regression of established BRAFV600E/PTENNull melanomas and cultured melanoma cells than individual blockade of either pathway. Surprisingly, growth of BRAFV600E/PIK3CAH1047R melanomas was dependent on the protein kinase AKT; however, AKT inhibition had no effect on growth of BRAFV600E/PTENNull melanomas. These data indicate that PTEN silencing contributes a PI3K-dependent, but AKT-independent, function in melanomagenesis. Our findings enhance our knowledge of how BRAFV600E and PI3K signaling cooperate in melanomagenesis and provide preclinical validation for combined pathway-targeted inhibition of PI3K and BRAFV600E in the therapeutic management of BRAFV600E/PTENNull melanomas.
Collapse
Affiliation(s)
- Victoria Marsh Durban
- Helen Diller Family Comprehensive Cancer Center, Department of Cell and Molecular Pharmacology, UCSF, San Francisco, California, USA.
Department of Dermatology, Yale University School of Medicine, New Haven, Connecticut, USA.
Surgical Oncology Research Laboratory, Peter MacCallum Cancer Centre, and Sir Peter MacCallum Department of Oncology, University of Melbourne, Victoria, Australia
| | - Marian M. Deuker
- Helen Diller Family Comprehensive Cancer Center, Department of Cell and Molecular Pharmacology, UCSF, San Francisco, California, USA.
Department of Dermatology, Yale University School of Medicine, New Haven, Connecticut, USA.
Surgical Oncology Research Laboratory, Peter MacCallum Cancer Centre, and Sir Peter MacCallum Department of Oncology, University of Melbourne, Victoria, Australia
| | - Marcus W. Bosenberg
- Helen Diller Family Comprehensive Cancer Center, Department of Cell and Molecular Pharmacology, UCSF, San Francisco, California, USA.
Department of Dermatology, Yale University School of Medicine, New Haven, Connecticut, USA.
Surgical Oncology Research Laboratory, Peter MacCallum Cancer Centre, and Sir Peter MacCallum Department of Oncology, University of Melbourne, Victoria, Australia
| | - Wayne Phillips
- Helen Diller Family Comprehensive Cancer Center, Department of Cell and Molecular Pharmacology, UCSF, San Francisco, California, USA.
Department of Dermatology, Yale University School of Medicine, New Haven, Connecticut, USA.
Surgical Oncology Research Laboratory, Peter MacCallum Cancer Centre, and Sir Peter MacCallum Department of Oncology, University of Melbourne, Victoria, Australia
| | - Martin McMahon
- Helen Diller Family Comprehensive Cancer Center, Department of Cell and Molecular Pharmacology, UCSF, San Francisco, California, USA.
Department of Dermatology, Yale University School of Medicine, New Haven, Connecticut, USA.
Surgical Oncology Research Laboratory, Peter MacCallum Cancer Centre, and Sir Peter MacCallum Department of Oncology, University of Melbourne, Victoria, Australia
| |
Collapse
|
12
|
The FBXO4 tumor suppressor functions as a barrier to BRAFV600E-dependent metastatic melanoma. Mol Cell Biol 2013; 33:4422-33. [PMID: 24019069 DOI: 10.1128/mcb.00706-13] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Cyclin D1-cyclin-dependent kinase 4/6 (CDK4/6) dysregulation is a major contributor to melanomagenesis. Clinical evidence has revealed that p16(INK4A), an allosteric inhibitor of CDK4/6, is inactivated in over half of human melanomas, and numerous animal models have demonstrated that p16(INK4A) deletion promotes melanoma. FBXO4, a specificity factor for the E3 ligase that directs timely cyclin D1 proteolysis, has not been studied in melanoma. We demonstrate that Fbxo4 deficiency induces Braf-driven melanoma and that this phenotype depends on cyclin D1 accumulation in mice, underscoring the importance of this ubiquitin ligase in tumor suppression. Furthermore, we have identified a substrate-binding mutation, FBXO4 I377M, that selectively disrupts cyclin D1 degradation while preserving proteolysis of the other known FBXO4 substrate, TRF1. The I377M mutation and Fbxo4 deficiency result in nuclear accumulation of cyclin D1, a key transforming neoplastic event. Collectively, these data provide evidence that FBXO4 dysfunction, as a mechanism for cyclin D1 overexpression, is a contributor to human malignancy.
Collapse
|
13
|
Chan KT, Jones SW, Brighton HE, Bo T, Cochran SD, Sharpless NE, Bear JE. Intravital imaging of a spheroid-based orthotopic model of melanoma in the mouse ear skin. INTRAVITAL 2013; 2. [PMID: 28748125 DOI: 10.4161/intv.25805] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Multiphoton microscopy is a powerful tool that enables the visualization of fluorescently tagged tumor cells and their stromal interactions within tissues in vivo. We have developed an orthotopic model of implanting multicellular melanoma tumor spheroids into the dermis of the mouse ear skin without the requirement for invasive surgery. Here, we demonstrate the utility of this approach to observe the primary tumor, single cell actin dynamics, and tumor-associated vasculature. These methods can be broadly applied to investigate an array of biological questions regarding tumor cell behavior in vivo.
Collapse
Affiliation(s)
- Keefe T Chan
- Department of Cell Biology and Physiology; Howard Hughes Medical Institute, Lineberger Comprehensive Cancer Center; University of North Carolina-Chapel Hill; Chapel Hill, NC USA
| | - Stephen W Jones
- Department of Cell Biology and Physiology; Howard Hughes Medical Institute, Lineberger Comprehensive Cancer Center; University of North Carolina-Chapel Hill; Chapel Hill, NC USA
| | - Hailey E Brighton
- Department of Cell Biology and Physiology; Howard Hughes Medical Institute, Lineberger Comprehensive Cancer Center; University of North Carolina-Chapel Hill; Chapel Hill, NC USA
| | - Tao Bo
- Department of Cell Biology and Physiology; Howard Hughes Medical Institute, Lineberger Comprehensive Cancer Center; University of North Carolina-Chapel Hill; Chapel Hill, NC USA
| | - Shelly D Cochran
- Department of Biomedical Engineering; North Carolina State University; Raleigh, NC USA
| | - Norman E Sharpless
- Departments of Genetics and Medicine; Lineberger Comprehensive Cancer Center; University of North Carolina-Chapel Hill; Chapel Hill, NC USA
| | - James E Bear
- Department of Cell Biology and Physiology; Howard Hughes Medical Institute, Lineberger Comprehensive Cancer Center; University of North Carolina-Chapel Hill; Chapel Hill, NC USA
| |
Collapse
|
14
|
Lakhter AJ, Sahu RP, Sun Y, Kaufmann WK, Androphy EJ, Travers JB, Naidu SR. Chloroquine promotes apoptosis in melanoma cells by inhibiting BH3 domain-mediated PUMA degradation. J Invest Dermatol 2013; 133:2247-54. [PMID: 23370537 PMCID: PMC3675185 DOI: 10.1038/jid.2013.56] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Revised: 01/10/2013] [Accepted: 01/14/2013] [Indexed: 01/27/2023]
Abstract
The BH3-only protein PUMA counters Bcl-2 family anti-apoptotic proteins and promotes apoptosis. Although PUMA is a key regulator of apoptosis, the post-transcriptional mechanisms that control PUMA protein stability are not understood. We show that a lysosome-independent activity of chloroquine prevents degradation of PUMA protein, promotes apoptosis and reduces the growth of melanoma xenografts in mice. Compared to wild–type PUMA, a BH3 domain deleted PUMA protein showed impaired decay in melanoma cells. Fusion of the BH3 domain to a heterologous protein led to its rapid turnover that was inhibited by chloroquine. While both chloroquine and inhibitors of lysosomal proteases stalled autophagy, only choroquine stabilized PUMA protein and promoted apoptosis. Our results reveal a lysosomal protease independent activity of chloroquine that selectively promotes apoptosis in melanoma cells.
Collapse
Affiliation(s)
- Alexander J Lakhter
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | | | | | | | | | | |
Collapse
|
15
|
Joyce CW, Murphy IG, Rafferty M, Ryan D, McDermott EW, Gallagher WM. Tumor profiling using protein biomarker panels in malignant melanoma: application of tissue microarrays and beyond. Expert Rev Proteomics 2013; 9:415-23. [PMID: 22967078 DOI: 10.1586/epr.12.5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Despite advances in our knowledge of the disease, malignant melanoma remains an unpredictable entity. The revolution in molecular biological techniques, such as DNA sequencing and gene-expression profiling, has uncovered many potential protein targets and biomarkers relevant to melanoma progression. Successful clinical application would be aided significantly by downstream proteomic validation of those candidate markers using a combination of immunohistochemistry and tissue microarrays. Yet, research in this context seems to lag behind the output of genomic data relating to melanoma. In this article, we look at the strengths and pitfalls of tissue microarrays in malignant melanoma. We will show how tissue microarrays have become a vital step in the transition from molecular techniques to useful clinical assays and interventions and look at likely future developments for advances in this field.
Collapse
Affiliation(s)
- Cormac W Joyce
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | | | | | | | | | | |
Collapse
|
16
|
Soengas MS. Mitophagy or how to control the Jekyll and Hyde embedded in mitochondrial metabolism: implications for melanoma progression and drug resistance. Pigment Cell Melanoma Res 2012; 25:721-31. [DOI: 10.1111/pcmr.12021] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- María S. Soengas
- Melanoma Laboratory, Molecular Pathology Programme; Centro Nacional de Investigaciones Oncológicas (Spanish National Cancer Research Centre); Madrid; Spain
| |
Collapse
|
17
|
Abstract
Melanoma is often considered one of the most aggressive and treatment-resistant human cancers. It is a disease that, due to the presence of melanin pigment, was accurately diagnosed earlier than most other malignancies and that has been subjected to countless therapeutic strategies. Aside from early surgical resection, no therapeutic modality has been found to afford a high likelihood of curative outcome. However, discoveries reported in recent years have revealed a near avalanche of breakthroughs in the melanoma field-breakthroughs that span fundamental understanding of the molecular basis of the disease all the way to new therapeutic strategies that produce unquestionable clinical benefit. These discoveries have been born from the successful fruits of numerous researchers working in many-sometimes-related, although also distinct-biomedical disciplines. Discoveries of frequent mutations involving BRAF(V600E), developmental and oncogenic roles for the microphthalmia-associated transcription factor (MITF) pathway, clinical efficacy of BRAF-targeted small molecules, and emerging mechanisms underlying resistance to targeted therapeutics represent just a sample of the findings that have created a striking inflection in the quest for clinically meaningful progress in the melanoma field.
Collapse
Affiliation(s)
- Hensin Tsao
- Department of Dermatology, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
- The Wellman Center for Photomedicine, Boston, Massachusetts 02114, USA
| | - Lynda Chin
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Levi A. Garraway
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts 02115, USA
| | - David E. Fisher
- Department of Dermatology, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
| |
Collapse
|
18
|
Thu YM, Su Y, Yang J, Splittgerber R, Na S, Boyd A, Mosse C, Simons C, Richmond A. NF-κB inducing kinase (NIK) modulates melanoma tumorigenesis by regulating expression of pro-survival factors through the β-catenin pathway. Oncogene 2012; 31:2580-92. [PMID: 21963849 PMCID: PMC3253179 DOI: 10.1038/onc.2011.427] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2011] [Revised: 08/13/2011] [Accepted: 08/20/2011] [Indexed: 02/07/2023]
Abstract
Nuclear factor-κB (NF-κB) inducing kinase (NIK) is a MAP3K that regulates the activation of NF-κB. NIK is often highly expressed in tumor cells, including melanoma, but the significance of this in melanoma progression has been unclear. Tissue microarray analysis of NIK expression reveals that dysplastic nevi (n=22), primary (n=15) and metastatic melanoma (n=13) lesions showed a statistically significant elevation in NIK expression when compared with benign nevi (n=30). Moreover, when short hairpin RNA techniques were used to knock-down NIK, the resultant NIK-depleted melanoma cell lines exhibited decreased proliferation, increased apoptosis, delayed cell cycle progression and reduced tumor growth in a mouse xenograft model. As expected, when NIK was depleted there was decreased activation of the non-canonical NF-κB pathway, whereas canonical NF-κB activation remained intact. NIK depletion also resulted in reduced expression of genes that contribute to tumor growth, including CXCR4, c-MYC and c-MET, and pro-survival factors such as BCL2 and survivin. These changes in gene expression are not fully explained by the attenuation of the non-canonical NF-κB pathway. Shown here for the first time is the demonstration that NIK modulates β-catenin-mediated transcription to promote expression of survivin. NIK-depleted melanoma cells exhibited downregulation of survivin as well as other β-catenin regulated genes including c-MYC, c-MET and CCND2. These data indicate that NIK mediates both β-catenin and NF-κB regulated transcription to modulate melanoma survival and growth. Thus, NIK may be a promising therapeutic target for melanoma.
Collapse
Affiliation(s)
- Yee Mon Thu
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Yingjun Su
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Jinming Yang
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Ryan Splittgerber
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Songqing Na
- Oncology Research, Eli Lily and Co., Indianapolis, IN 46285
| | - Alan Boyd
- Division of Dermatology, Vanderbilt University School of Medicine, Nashville, TN 37204
| | - Claudio Mosse
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Christopher Simons
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Ann Richmond
- Department of Veterans Affairs Medical Center, Nashville, TN
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN 37232
| |
Collapse
|
19
|
|
20
|
Comprehensive analysis of receptor tyrosine kinase activation in human melanomas reveals autocrine signaling through IGF-1R. Melanoma Res 2011; 21:274-84. [PMID: 21654344 DOI: 10.1097/cmr.0b013e328343a1d6] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Melanomas depend on autocrine signals for proliferation and survival; however, no systematic screen of known receptor tyrosine kinases (RTKs) has been performed to identify which autocrine signaling pathways are activated in melanoma. Here, we performed a comprehensive analysis of 42 RTKs in six individual human melanoma tumor specimens as well as 17 melanoma cell lines, some of which were derived from the tumor specimens. We identified five RTKs that were active in almost every one of the melanoma tissue specimens and cell lines, including two previously unreported receptors, insulin-like growth factor receptor 1 (IGF-1R) and macrophage-stimulating protein receptor (MSPR), in addition to three receptors (vascular endothelial growth factor receptor, fibroblast growth factor receptor, and hepatocyte growth factor receptor) known to be autocrine activated in melanoma. We show, by quantitative real time PCR, that all melanoma cell lines expressed genes for the RTK ligands such as HGF, IGF-1, and MSP. Addition of antibodies to either IGF-1 or HGF, but not to MSP, to the culture medium blocked melanoma cell proliferation, and even caused net loss of melanoma cells. Antibody addition deactivated IGF-1R and hepatocyte growth factor receptors, as well as mitogen-activated protein kinase signaling. Thus, IGF-1 is a new growth factor for autocrine driven proliferation of human melanoma in vitro. Our results suggest that IGF-1-IGF-1R autocrine pathway in melanoma is a possible target for therapy in human melanomas.
Collapse
|
21
|
Fernandez AA, Garcia R, Paniker L, Trono D, Mitchell DL. An experimental population study of nucleotide excision repair as a risk factor for UVB-induced melanoma. Photochem Photobiol 2011; 87:335-41. [PMID: 21143485 DOI: 10.1111/j.1751-1097.2010.00875.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Nucleotide excision repair (NER) is the primary defense against the DNA damage implicit in skin cancer formation and is negatively affected by chronic exposure to UVB radiation. However, in situ and in vitro studies consistently yield equivocal results when addressing individual DNA repair capacity and melanoma susceptibility. The primary objective of this study was to determine if individual global NER capacity is a risk factor for melanoma formation in a prominent UVB-inducible melanoma model, hybrid Xiphophorus fishes. After neonatal UVB irradiation, adult tumor-bearing and tumor-free fish were given a challenge UVB dose and (6-4) photoproduct repair was quantified in individual fish at 24 h using radioimmunoassay. Despite considerable inter-individual variation in repair capacity, ranging from 13% to 91%, we found no difference in mean NER capacity between fish with and without melanomas, thus detaching global NER from melanomagenesis. Furthermore, despite epidemiological data indicating that sex and age are important risk factors underlying melanoma susceptibility, we found no difference in mean NER rates among the sexes or as a function of age. We conclude with a discussion of the apparent paradox of how inter-individual variation in NER is not a risk factor given the clear evidence that DNA damage underlies melanoma susceptibility.
Collapse
Affiliation(s)
- André A Fernandez
- Department of Carcinogenesis, University of Texas MD Anderson Cancer Center, Smithville, TX, USA
| | | | | | | | | |
Collapse
|
22
|
Jafarnejad SM, Wani AA, Martinka M, Li G. Prognostic significance of Sox4 expression in human cutaneous melanoma and its role in cell migration and invasion. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 177:2741-52. [PMID: 20952589 DOI: 10.2353/ajpath.2010.100377] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The Sox4 transcription factor is involved in various cellular processes, such as embryonic development and differentiation. Deregulated expression of Sox4 in several human cancers has been reported to date, but its role in melanoma is unknown. We explored the role of Sox4 in melanoma pathogenesis in vivo and in vitro. Using tissue microarray, we evaluated Sox4 expression in 180 melanocytic lesions and investigated its role in melanoma cell migration and invasion. Sox4 expression was remarkably reduced in metastatic melanoma compared with dysplastic nevi (P < 0.05) and primary melanoma (P < 0.01). This reduction was correlated with a poorer disease-specific survival of melanoma patients (P = 0.039). Multivariate Cox regression analysis revealed that reduced Sox4 expression is an independent prognostic factor (P = 0.049). Knockdown of Sox4 enhanced melanoma cell invasion, migration, and stress fiber formation. The increased migration and invasion on Sox4 knockdown depends on the presence of nuclear factor (NF)-κB p50 and is abrogated when p50 is knocked down. We further observed inhibition of NF-κB p50 transcription by Sox4, in addition to a reverse pattern of expression of Sox4 and NF-κB p50 in different stages of melanocytic lesions. Our results suggest that Sox4 regulates melanoma cell migration and invasion in an NF-κB p50-dependent manner and may serve as a prognostic marker and potential therapeutic target for human melanoma.
Collapse
Affiliation(s)
- Seyed Mehdi Jafarnejad
- Department of Dermatology and Skin Science, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | | | | | | |
Collapse
|
23
|
Abstract
Melanoma remains one of the cancers for which a decline in morbidity has not been achieved with current scientific and medical advances. Mono-therapies targeting melanoma have been largely ineffective, increasing the need for identification of new drugable targets. Multiple tumor suppressors and oncogenes that impart genetic predisposition to melanoma have been identified and are being studied in an attempt to provide insight on the development of anti-melanoma therapies. Metabotropic Glutamate Receptor I (GRM1) has recently been implicated as a novel oncogene involved in melanomagenesis. GRM1 (mGlu1, protein) belongs to the G protein coupled receptor (GPCR) super family and is normally functional in the central nervous system. Our group showed in a transgenic mouse model system that ectopic expression of Grm1 in melanocytes is sufficient to induce spontaneous melanoma development in vivo. GPCRs are some of the most important therapeutic drug targets discovered to date and they make up a significant proportion of existing therapies. This super family of transmembrane receptors has wide spread expression and interacts with a diverse array of ligands. Diverse physiological responses can be induced by stimulator(s) or suppressor(s) of GPCRs, which contributes to their attractiveness in existing and emerging therapies. GPCR targeting therapies are employed against a variety of human disorders including those of the central nervous system, cardiovascular, metabolic, urogenital and respiratory systems. In the current review, we will discuss how the identification of the oncogenic properties of GRM1 opens up new strategies for the design of potential novel therapies for the treatment of melanoma.
Collapse
|
24
|
Abstract
The role of ultraviolet radiation (UV) in the pathogenesis has been discussed controversially for many decades. Studies in mice (SCID, HGF/SF, SV40T) which develop malignant melanoma, show a role of UVB in melanomagenesis. In contrast to this, the role of UVA is less clear. We will review the recent in vitro and in vivo data in support of the hypothesis that UVA is also involved in the development of malignant melanoma. The role of UVA in p53 activation, apoptosis, cell cycle arrest and photoproduct formation is discussed.
Collapse
|
25
|
BH3-only proteins and their effects on cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 687:49-63. [PMID: 20919637 DOI: 10.1007/978-1-4419-6706-0_3] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Apoptosis, a form of cellular suicide is a key mechanism involved in the clearance of cells that are dysfunctional, superfluous or infected. For this reason, the cell needs mechanisms o sense death cues and relay death signals to the apoptotic machinery involved in cellular execution. In the intrinsic apoptotic pathway, a subclass of BCL-2 family proteins called the BH3-onlyproteins are responsible for triggering apoptosis in response to varied cellular stress cues. The mechanisms by which they are regulated are tied to the type of cellular stress they sense. Once triggered, they interact with other BCL-2 family proteins to cause mitochondrial outer membrane permeabilization which in turn results in the activation ofserine proteases necessary for cell killing. Failure to properly sense death cues and relay the death signal can have a major impact on cancer. This chapter will discuss our current models of how BH3-only proteins function as well as their impact on carcinogenesis and cancer treatment.
Collapse
|
26
|
Identification and functional validation of therapeutic targets for malignant melanoma. Crit Rev Oncol Hematol 2009; 72:194-214. [DOI: 10.1016/j.critrevonc.2009.02.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2008] [Revised: 02/04/2009] [Accepted: 02/19/2009] [Indexed: 12/12/2022] Open
|
27
|
Martorell-Calatayud A, Requena C, Botella-Estrada R, Sangüeza O. Novedades en biología molecular y su aplicación en el diagnóstico y el tratamiento del melanoma. ACTAS DERMO-SIFILIOGRAFICAS 2009; 100 Suppl 1:52-65. [DOI: 10.1016/s0001-7310(09)73168-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
|
28
|
Lin H, Wong RPC, Martinka M, Li G. Loss of SNF5 expression correlates with poor patient survival in melanoma. Clin Cancer Res 2009; 15:6404-11. [PMID: 19808872 DOI: 10.1158/1078-0432.ccr-09-1135] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Aberrant expression of SWI/SNF chromatin remodeling complex is involved in cancer development. The tumor suppressor SNF5, the core subunit of SWI/SNF complex, has been shown to regulate cell differentiation, cell cycle control, and apoptosis. To investigate the role of SNF5 in the development of melanoma, we examined the expression of SNF5 in melanocytic lesions at different stages and analyzed the correlation between SNF5 expression and clinicopathologic variables and patient survival. EXPERIMENTAL DESIGN Using tissue microarray and immunohistochemistry, we evaluated SNF5 staining in 51 dysplastic nevi, 88 primary melanomas, and 48 metastatic melanomas. We studied chemosensitivity of melanoma cells with reduced SNF5 expression by siRNA using cell survival and apoptosis assays. RESULTS SNF5 expression was reduced in metastatic melanoma compared with dysplastic nevi (P = 0.005), in advanced primary melanoma (Clark's level V) compared with low risk Clark's level II melanoma (P = 0.019), and in melanoma at sun-exposed sites compared with sun-protected sites (P = 0.044). Furthermore, we showed a strong correlation between negative SNF5 expression and a worse 5-year survival in melanoma patients (P = 0.016). Multivariate Cox regression analysis revealed that negative SNF5 expression is an independent prognostic factor to predict patient outcome in primary melanomas (P = 0.031). Finally, we showed that knockdown of SNF5 in melanoma cell lines resulted in significant chemoresistance. CONCLUSIONS Our data indicate that SNF5 may be an important marker for human melanoma progression and prognosis as well as a potential therapeutic target.
Collapse
Affiliation(s)
- Hanyang Lin
- Department of Dermatology, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, BC, Canada
| | | | | | | |
Collapse
|
29
|
Laverty HG, Wakefield LM, Occleston NL, O'Kane S, Ferguson MWJ. TGF-beta3 and cancer: a review. Cytokine Growth Factor Rev 2009; 20:305-17. [PMID: 19656717 DOI: 10.1016/j.cytogfr.2009.07.002] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
With the development of growth factors and growth factor modulators as therapeutics for a range of disorders, it is prudent to consider whether modulating the growth factor profile in a tissue can influence tumour initiation or progression. As recombinant human TGF-beta3 (avotermin) is being developed for the improvement of scarring in the skin it is important to understand the role, if any, of this cytokine in tumour progression. Elevated levels of TGF-beta3 expression detected in late-stage tumours have linked this cytokine with tumourigenesis, although functional data to support a causative role are lacking. While it has proved tempting for researchers to interpret a 'correlation' as a 'cause' of disease, what has often been overlooked is the normal biological role of TGF-beta3 in processes that are often subverted in tumourigenesis. Clarifying the role of this cytokine is complicated by inappropriate extrapolation of the data relating to TGF-beta1 in tumourigenesis, despite marked differences in biology between the TGF-beta isoforms. Indeed, published studies have indicated that TGF-beta3 may actually play a protective role against tumourigenesis in a range of tissues including the skin, breast, oral and gastric mucosa. Based on currently available data it is reasonable to hypothesize that administration of acute low doses of exogenous TGF-beta3 is unlikely to influence tumour initiation or progression.
Collapse
Affiliation(s)
- H G Laverty
- Renovo Group Plc, Core Technology Facility, 48 Grafton Street, Manchester M13 9XX, UK
| | | | | | | | | |
Collapse
|
30
|
Gould Rothberg BE, Bracken MB, Rimm DL. Tissue biomarkers for prognosis in cutaneous melanoma: a systematic review and meta-analysis. J Natl Cancer Inst 2009; 101:452-74. [PMID: 19318635 DOI: 10.1093/jnci/djp038] [Citation(s) in RCA: 130] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
In the clinical management of early-stage cutaneous melanoma, it is critical to determine which patients are cured by surgery alone and which should be treated with adjuvant therapy. To assist in this decision, many groups have made an effort to use molecular information. However, although there are hundreds of studies that have sought to assess the potential prognostic value of molecular markers in predicting the course of cutaneous melanoma, at this time, no molecular method to improve risk stratification is part of recommended clinical practice. To help understand this disconnect, we conducted a systematic review and meta-analysis of the published literature that reported immunohistochemistry-based protein biomarkers of melanoma outcome. Three parallel search strategies were applied to the PubMed database through January 15, 2008, to identify cohort studies that reported associations between immunohistochemical expression and survival outcomes in melanoma that conformed to the REMARK criteria. Of the 102 cohort studies, we identified only 37 manuscripts, collectively describing 87 assays on 62 distinct proteins, which met all inclusion criteria. Promising markers that emerged included melanoma cell adhesion molecule (MCAM)/MUC18 (all-cause mortality [ACM] hazard ratio [HR] = 16.34; 95% confidence interval [CI] = 3.80 to 70.28), matrix metalloproteinase-2 (melanoma-specific mortality [MSM] HR = 2.6; 95% CI = 1.32 to 5.07), Ki-67 (combined ACM HR = 2.66; 95% CI = 1.41 to 5.01), proliferating cell nuclear antigen (ACM HR = 2.27; 95% CI = 1.56 to 3.31), and p16/INK4A (ACM HR = 0.29; 95% CI = 0.10 to 0.83, MSM HR = 0.4; 95% CI = 0.24 to 0.67). We further noted incomplete adherence to the REMARK guidelines: 14 of 27 cohort studies that failed to adequately report their methods and nine studies that failed to either perform multivariable analyses or report their risk estimates were published since 2005.
Collapse
|
31
|
Braf(V600E) cooperates with Pten loss to induce metastatic melanoma. Nat Genet 2009; 41:544-52. [PMID: 19282848 PMCID: PMC2705918 DOI: 10.1038/ng.356] [Citation(s) in RCA: 880] [Impact Index Per Article: 58.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2008] [Accepted: 02/13/2009] [Indexed: 12/22/2022]
Abstract
Mutational activation of BRAF is the earliest and most common genetic alteration in human melanoma. Hence, to build a model of human melanoma, we generated mice with conditional melanocyte-specific expression of BRafV600E. Upon induction of BRafV600E expression, mice developed benign melanocytic hyperplasias that failed to progress to melanoma over 15-20 months. By contrast, expression of BRafV600E combined with Pten tumor suppressor gene silencing elicited development of melanoma with 100% penetrance, short latency and with metastases observed in lymph nodes and lungs. Melanoma was prevented by inhibitors of mTorc1 (Rapamycin) or MEK1/2 (PD325901) but, upon cessation of drug administration, mice developed melanoma indicating the presence of long-lived melanoma-initiating cells in this system. Importantly, combined treatment with Rapamycin and PD325901 led to shrinkage of established melanomas. These mice, engineered with a common genetic profile to human melanoma, provide an excellent system to study melanoma’s cardinal feature of metastasis and for pre-clinical evaluation of agents designed to prevent or treat metastatic disease.
Collapse
|
32
|
Eskandarpour M, Huang F, Reeves KA, Clark E, Hansson J. OncogenicNRAShas multiple effects on the malignant phenotype of human melanoma cells culturedin vitro. Int J Cancer 2009; 124:16-26. [DOI: 10.1002/ijc.23876] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
33
|
Roadcap DW, Clemen CS, Bear JE. The role of mammalian coronins in development and disease. Subcell Biochem 2008; 48:124-35. [PMID: 18925377 DOI: 10.1007/978-0-387-09595-0_12] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Coronins have maintained a high degree of conservation over the roughly 800 million years of eukaryotic evolution.1,2 From its origins as a single gene in simpler eukaryotes, the mammalian Coronin gene family has expanded to include at least six members (see Chapter 4). Increasing evidence indicates that Coronins play critical roles as regulators of actin dependent processes such as cell motility and vesicle trafficking3,4 (see Chapters 6-9). Considering the importance of these processes, it is not surprising that recent findings have implicated the involvement of Coronins in multiple diseases. This review primarily focuses on Coronin 1C (HGNC symbol: CORO1C, also known as Coronin 3) which is a transcriptionally dynamic gene that is up-regulated in multiple types of clinically aggressive cancer. In addition to reviewing the molecular signals and events that lead to Coronin 1C transcription, we summarize the results of several studies describing the possible functional roles of Coronin 1C in development as well as disease progression. Here, the main focus is on brain development and on the progression of melanoma and glioma. Finally, we will also review the role of other mammalian Coronin genes in clinically relevant processes such as neural regeneration and pathogenic bacterial infections (see Chapter 10).
Collapse
Affiliation(s)
- David W Roadcap
- Lineberger Comprehensive Cancer Center and Department of Cell and Developmental Biology, UNC-Chapel Hill, Chapel Hill, NC 27599, USA
| | | | | |
Collapse
|
34
|
Sarikas A, Xu X, Field LJ, Pan ZQ. The cullin7 E3 ubiquitin ligase: a novel player in growth control. Cell Cycle 2008; 7:3154-61. [PMID: 18927510 PMCID: PMC2637179 DOI: 10.4161/cc.7.20.6922] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Cullin7 (CUL7) is a molecular scaffold that organizes an E3 ubiquitin ligase containing the F-box protein Fbw8, Skp1 and the ROC1 RING finger protein. Dysregulation of the CUL7 E3 Ligase has been directly linked to hereditary human diseases as cul7 germline mutations were found in patients with autosomal-recessive 3-M and Yakuts short stature syndromes, which are characterized by profound pre- and postnatal growth retardation. In addition, genetic ablation of CUL7 in mice resulted in intrauterine growth retardation and perinatal lethality, underscoring its importance for growth regulation. The recent identification of insulin receptor substrate 1, a critical mediator of insulin and insulin-like growth factor-1 signaling, as the proteolytic target of the CUL7 E3 ligase, provided a molecular link between CUL7 and a well-established growth regulatory pathway. This result, coupled with other studies demonstrating interactions between CUL7 and the p53 tumor suppressor protein, as well as the simian virus 40 large T antigen oncoprotein, further implicated CUL7 as a novel player in growth control and suggested pathomechanistic insights into CUL7-linked growth retardation syndromes.
Collapse
Affiliation(s)
- Antonio Sarikas
- Department of Oncological Sciences; The Mount Sinai School of Medicine; New York, New York USA
| | - Xinsong Xu
- Department of Oncological Sciences; The Mount Sinai School of Medicine; New York, New York USA
| | - Loren J. Field
- Indiana University School of Medicine; Wells Center for Pediatric Research and Krannert Institute of Cardiology; Indianapolis, Indiana USA
| | - Zhen-Qiang Pan
- Department of Oncological Sciences; The Mount Sinai School of Medicine; New York, New York USA
| |
Collapse
|
35
|
Wajapeyee N, Serra RW, Zhu X, Mahalingam M, Green MR. Oncogenic BRAF induces senescence and apoptosis through pathways mediated by the secreted protein IGFBP7. Cell 2008; 132:363-74. [PMID: 18267069 PMCID: PMC2266096 DOI: 10.1016/j.cell.2007.12.032] [Citation(s) in RCA: 679] [Impact Index Per Article: 42.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2007] [Revised: 10/26/2007] [Accepted: 12/20/2007] [Indexed: 10/22/2022]
Abstract
Expression of an oncogene in a primary cell can, paradoxically, block proliferation by inducing senescence or apoptosis through pathways that remain to be elucidated. Here we perform genome-wide RNA-interference screening to identify 17 genes required for an activated BRAF oncogene (BRAFV600E) to block proliferation of human primary fibroblasts and melanocytes. Surprisingly, we find a secreted protein, IGFBP7, has a central role in BRAFV600E-mediated senescence and apoptosis. Expression of BRAFV600E in primary cells leads to synthesis and secretion of IGFBP7, which acts through autocrine/paracrine pathways to inhibit BRAF-MEK-ERK signaling and induce senescence and apoptosis. Apoptosis results from IGFBP7-mediated upregulation of BNIP3L, a proapoptotic BCL2 family protein. Recombinant IGFBP7 (rIGFBP7) induces apoptosis in BRAFV600E-positive human melanoma cell lines, and systemically administered rIGFBP7 markedly suppresses growth of BRAFV600E-positive tumors in xenografted mice. Immunohistochemical analysis of human skin, nevi, and melanoma samples implicates loss of IGFBP7 expression as a critical step in melanoma genesis.
Collapse
Affiliation(s)
- Narendra Wajapeyee
- Howard Hughes Medical Institute, Programs in Gene Function and Expression and Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | | | | | | | | |
Collapse
|
36
|
Abstract
Cutaneous melanoma is increasing in incidence at one of the highest rates for any form of cancer in the USA, with a current lifetime incidence of 1 in 68. Although early-stage disease is often curable, the survival rate for advanced disease is low, with an average life expectancy of 6–10 months. Knowledge of the molecular alterations associated with melanoma development and progression is expected to lead to improved therapies and outcomes. Major progress in defining the molecular alterations associated with the evolution of melanoma came in 2002, through a systematic genome-wide assessment of cancer-associated pathways. Large-scale sequencing of growth-associated genes in a variety of cancers identified a high frequency (>60%) of activating mutations of the BRAF kinase gene in human melanomas. This discovery has prompted a large number of studies evaluating the biological significance of BRAF kinase mutations in the initiation and progression of melanoma, and their importance for the development of novel melanoma therapies. Here we review the most recent studies of BRAF kinase in the pathogenesis of melanoma and their implications for defining BRAF kinase as a therapeutic point of interest in melanoma.
Collapse
|
37
|
Abstract
Malignant melanoma originates in melanocytes, the pigment-producing cells of the skin and eye, and is one of the most deadly human cancers with no effective cure for metastatic disease. Like many other cancers, melanoma has both environmental and genetic components. For more than 20 years, the melanoma genome has been subject to extensive scrutiny, which has led to the identification of several genes that contribute to melanoma genesis and progression. Three molecular pathways have been found to be nearly invariably dysregulated in melanocytic tumors, including the RAS-RAF-MEK-ERK pathway (through mutation of BRAF, NRAS or KIT), the p16 INK4A-CDK4-RB pathway (through mutation of INK4A or CDK4) and the ARF-p53 pathway (through mutation of ARF or TP53). Less frequently targeted pathways include the PI3K-AKT pathway (through mutation of NRAS, PTEN or PIK3CA) and the canonical Wnt signaling pathway (through mutation of CTNNB1 or APC). Beyond the specific and well-characterized genetic events leading to activation of proto-oncogenes or inactivation of tumor suppressor genes in these pathways, systematic high-resolution genomic analysis of melanoma specimens has revealed recurrent DNA copy number aberrations as well as perturbations of DNA methylation patterns. Melanoma provides one of the best examples of how genomic analysis can lead to a better understanding of tumor biology. We review current knowledge of the genes involved in the development of melanoma and the molecular pathways in which these genes operate.
Collapse
Affiliation(s)
- Christina Dahl
- Institute of Cancer Biology, Danish Cancer Society, Copenhagen, Denmark
| | | |
Collapse
|
38
|
Kwong L, Chin L, Wagner SN. Growth factors and oncogenes as targets in melanoma: lost in translation? ACTA ACUST UNITED AC 2008; 23:99-129. [PMID: 18159898 DOI: 10.1016/j.yadr.2007.07.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Lawrence Kwong
- Department of Medical Oncology, Dana-Farber Cancer Institute, 44 Binney Street, Boston, MA 02115, USA
| | | | | |
Collapse
|
39
|
Woo SK, Lee TS, Kim KM, Kim JY, Jung JH, Kang JH, Cheon GJ, Choi CW, Lim SM. Anesthesia condition for 18F-FDG imaging of lung metastasis tumors using small animal PET. Nucl Med Biol 2008; 35:143-50. [DOI: 10.1016/j.nucmedbio.2007.10.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2007] [Revised: 10/02/2007] [Accepted: 10/09/2007] [Indexed: 12/20/2022]
|
40
|
Muller HK, Malley RC, McGee HM, Scott DK, Wozniak T, Woods GM. Effect of UV Radiation on the Neonatal Skin Immune System- Implications for Melanoma†. Photochem Photobiol 2007; 84:47-54. [DOI: 10.1111/j.1751-1097.2007.00246.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
41
|
Cartlidge RA, Thomas GR, Cagnol S, Jong KA, Molton SA, Finch AJ, McMahon M. Oncogenic BRAF(V600E) inhibits BIM expression to promote melanoma cell survival. Pigment Cell Melanoma Res 2007; 21:534-44. [PMID: 18715233 DOI: 10.1111/j.1755-148x.2008.00491.x] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Somatic activating mutations of BRAF are the earliest and most common genetic abnormality detected in the genesis of human melanoma. However, the mechanism(s) by which activated BRAF promotes melanoma cell cycle progression and/or survival remain unclear. Here we demonstrate that expression of BIM, a pro-apoptotic member of the BCL-2 family, is inhibited by BRAF-->MEK-->ERK signaling in mouse and human melanocytes and in human melanoma cells. Trophic factor deprivation of melanocytes leads to elevated BIM expression. However, re-addition of trophic factors or activation of a conditional form of BRAF(V600E) leads to rapid inhibition of BIM expression. In both cases, inhibition of BIM expression was dependent on the activity of MEK1/2 and the proteasome. Consistent with these observations, pharmacological inhibition of BRAF(V600E) or MEK1/2 in human melanoma cells (using PLX4720 and CI-1040 respectively) led to a striking elevation of BIM expression. Re-activation of BRAF-->MEK-->ERK signaling led to phosphorylation of BIM-EL on serine 69 and its subsequent degradation. Interestingly, endogenous expression of BIM in melanoma cells was insufficient to induce apoptosis unless combined with serum deprivation. Under these circumstances, inhibition of BIM expression by RNA interference provided partial protection from apoptosis. These data suggest that regulation of BIM expression by BRAF-->MEK-->ERK signaling is one mechanism by which oncogenic BRAF(V600E) can influence the aberrant physiology of melanoma cells.
Collapse
Affiliation(s)
- Robert A Cartlidge
- Cancer Research Institute and Department of Cellular and Molecular Pharmacology, University of California, San Francisco, Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, USA
| | | | | | | | | | | | | |
Collapse
|
42
|
Na YJ, Baek HS, Ahn SM, Shin HJ, Chang IS, Hwang JS. [4-t-butylphenyl]-N-(4-imidazol-1-yl phenyl)sulfonamide (ISCK03) inhibits SCF/c-kit signaling in 501mel human melanoma cells and abolishes melanin production in mice and brownish guinea pigs. Biochem Pharmacol 2007; 74:780-6. [PMID: 17658483 DOI: 10.1016/j.bcp.2007.05.028] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2007] [Revised: 05/28/2007] [Accepted: 05/29/2007] [Indexed: 11/28/2022]
Abstract
It is well known that c-kit is related to pigmentation as well as to the oncology target protein. The objective of this study was to discover a skin-whitening agent that regulates c-kit activity. We have developed a high-throughput screening system using recombinant human c-kit protein. Approximately 10,000 synthetic compounds were screened for their effect on c-kit activity. Phenyl-imidazole sulfonamide derivatives showed inhibitory activity on c-kit phosphorylation in vitro. The effects of one derivative, [4-t-butylphenyl]-N-(4-imidazol-1-yl phenyl)sulfonamide (ISCK03), on stem-cell factor (SCF)/c-kit cellular signaling in 501mel human melanoma cells were examined further. Pretreatment of 501mel cells with ISCK03 inhibited SCF-induced c-kit phosphorylation dose dependently. ISCK03 also inhibited p44/42 ERK mitogen-activated protein kinase (MAPK) phosphorylation, which is known to be involved in SCF/c-kit downstream signaling. However ISCK03 did not inhibit hepatocyte growth factor (HGF)-induced phosphorylation of p44/42 ERK proteins. To determine the in vivo potency of ISCK03, it was orally administered to depilated C57BL/6 mice. Interestingly, oral administration of ISCK03 induced the dose-dependent depigmentation of newly regrown hair, and this was reversed with cessation of ISCK03 treatment. Finally, to investigate whether the inhibitory effect of ISCK03 on SCF/c-kit signaling abolished UV-induced pigmentation, ISCK03 was applied to UV-induced pigmented spots on brownish guinea pig skin. The topical application of ISCK03 promoted the depigmentation of UV-induced hyperpigmented spots. Fontana-Masson staining analysis showed epidermal melanin was diminished in spots treated with ISCK03. These results indicate that phenyl-imidazole sulfonamide derivatives are potent c-kit inhibitors and might be used as skin-whitening agents.
Collapse
Affiliation(s)
- Yong Joo Na
- Skin Research Institute, AMOREPACIFIC R&D Center, 314-1 Bora-dong, Giheung-gu, Yongin-si, 446-729, Republic of Korea
| | | | | | | | | | | |
Collapse
|
43
|
Bedogni B, Welford SM, Kwan AC, Ranger-Moore J, Saboda K, Powell MB. Inhibition of phosphatidylinositol-3-kinase and mitogen-activated protein kinase kinase 1/2 prevents melanoma development and promotes melanoma regression in the transgenic TPRas mouse model. Mol Cancer Ther 2007; 5:3071-7. [PMID: 17172409 DOI: 10.1158/1535-7163.mct-06-0269] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
A number of human melanomas show hyperactivation of the Ras pathway due to mutations of the molecule or alteration of upstream or downstream effectors. In this study, we evaluated the effect of blocking the two Ras downstream pathways phosphatidylinositol-3-kinase/Akt and Raf/mitogen-activated protein kinase kinase/extracellular signal-regulated kinase on melanoma development and regression in the TPRas mouse model. The inhibition of these two signaling cascades by topically applied Ly294002 and U0126 significantly delayed melanoma development and significantly decreased the tumor incidence, particularly when the drugs were applied in combination. Treatment with the inhibitors of established melanomas resulted in complete remission in 33% of mice and partial regression in 46% of mice when drugs were delivered in combination. These responses correlated with increased apoptosis and decreased proliferation both in vitro and in vivo and reduced tumor angiogenesis. In conclusion, this study strongly supports the role of the phosphatidylinositol-3-kinase/Akt and Raf/mitogen-activated protein kinase kinase/extracellular signal-regulated kinase pathways in the development and maintenance of Ras-dependent melanomas and supports the notion that specific inhibition of these effectors may represent a very promising avenue for the treatment and prevention of the disease.
Collapse
Affiliation(s)
- Barbara Bedogni
- Division of Radiation and Cancer Biology, 269 Campus Drive, CCSR-S-1230, Stanford 94305, CA, USA
| | | | | | | | | | | |
Collapse
|
44
|
Abstract
Irreversible changes in the DNA sequence, including chromosomal deletions or amplification, activating or inactivating mutations in genes, have been implicated in the development and progression of melanoma. However, increasing attention is being turned towards the participation of 'epigenetic' events in melanoma progression that do not affect DNA sequence, but which nevertheless may lead to stable inherited changes in gene expression. Epigenetic events including histone modifications and DNA methylation play a key role in normal development and are crucial to establishing the correct program of gene expression. In contrast, mistargeting of such epigenetic modifications can lead to aberrant patterns of gene expression and loss of anti-cancer checkpoints. Thus, to date at least 50 genes have been reported to be dysregulated in melanoma by aberrant DNA methylation and accumulating evidence also suggests that mistargetting of histone modifications and altered chromatin remodeling activities will play a key role in melanoma. This review gives an overview of the many different types of epigenetic modifications and their involvement in cancer and especially in melanoma development and progression.
Collapse
Affiliation(s)
- Tanja Rothhammer
- Institute of Pathology, University of Regensburg Medical School, Franz-Josef-Strauss-Allee 11, D-93053 Regensburg, Germany
| | | |
Collapse
|
45
|
Butler AP, Trono D, Coletta LD, Beard R, Fraijo R, Kazianis S, Nairn RS. Regulation of CDKN2A/B and Retinoblastoma genes in Xiphophorus melanoma. Comp Biochem Physiol C Toxicol Pharmacol 2007; 145:145-55. [PMID: 17011832 DOI: 10.1016/j.cbpc.2006.07.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2006] [Revised: 07/12/2006] [Accepted: 07/31/2006] [Indexed: 02/02/2023]
Abstract
Xiphophorus interspecies hybrids provide several well-characterized genetic models of melanoma susceptibility. The Xiphophorus CDKN2A/B gene, homologous to mammalian CDKN2A/B cyclin-dependent kinase inhibitors (p16 and p15), is a candidate tumor susceptibility gene in these models. Using real-time PCR and Western blot analysis, we analyzed expression of CDKN2A/B in spontaneous and UV-induced primary melanomas from individual backcross hybrid fish. We found that CDKN2A/B mRNA is highly expressed in melanomas (18-fold), relative to other fish tissues. Expression is also elevated, to a lesser extent (9.5-fold), in melanized skin from tumor-bearing fish. However, quantitative levels of CDKN2A/B mRNA in tumors varied considerably and positively correlated with expression of the Xmrk oncogene, suggesting possible functional interaction between Xmrk and CDKN2A/B expression. As a homolog corresponding to members of the mammalian CDKN2 family which regulate cell cycle progression at the G1 checkpoint, the CDKN2A/B p13 protein is a putative regulator of the G1 checkpoint apparatus in Xiphophorus. Since CDKN2A is often observed to be inversely regulated compared to RB in some human tumors, and is capable of transcriptionally regulating RB in human ovarian tumors, we cloned the Xiphophorus maculatus RB cDNA and analyzed RB expression by real-time PCR and Western blot analysis in the fish melanomas. These experiments were designed to ascertain whether CDKN2A/B and RB expression were inversely correlated. Our results indicate that RB mRNA was consistently expressed at only a 2-fold higher level in both tumors and melanized skin than in muscle. Qualitatively similar results were obtained for protein expression. These results collectively suggest that (i) Xmrk and CDKN2A/B may be co-regulated at the transcriptional level, and (ii) there is little, if any, alteration of RB expression in Xiphophorus melanomas.
Collapse
Affiliation(s)
- Andrew P Butler
- Virginia Harris Cockrell Cancer Research Center, Department of Carcinogenesis, Science Park-Research Division, University of Texas MD Anderson Cancer Center, Smithville, TX 78957, USA.
| | | | | | | | | | | | | |
Collapse
|
46
|
Carreira S, Goodall J, Denat L, Rodriguez M, Nuciforo P, Hoek KS, Testori A, Larue L, Goding CR. Mitf regulation of Dia1 controls melanoma proliferation and invasiveness. Genes Dev 2007; 20:3426-39. [PMID: 17182868 PMCID: PMC1698449 DOI: 10.1101/gad.406406] [Citation(s) in RCA: 451] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
It is widely held that cells with metastatic properties such as invasiveness and expression of matrix metalloproteinases arise through the stepwise accumulation of genetic lesions arising from genetic instability and "clonal evolution." By contrast, we show here that in melanomas invasiveness can be regulated epigenetically by the microphthalmia-associated transcription factor, Mitf, via regulation of the DIAPH1 gene encoding the diaphanous-related formin Dia1 that promotes actin polymerization and coordinates the actin cytoskeleton and microtubule networks at the cell periphery. Low Mitf levels lead to down-regulation of Dia1, reorganization of the actin cytoskeleton, and increased ROCK-dependent invasiveness, whereas increased Mitf expression leads to decreased invasiveness. Significantly the regulation of Dia1 by Mitf also controls p27(Kip1)-degradation such that reduced Mitf levels lead to a p27(Kip1)-dependent G1 arrest. Thus Mitf, via regulation of Dia1, can both inhibit invasiveness and promote proliferation. The results imply variations in the repertoire of environmental cues that determine Mitf activity will dictate the differentiation, proliferative, and invasive/migratory potential of melanoma cells through a dynamic epigenetic mechanism.
Collapse
Affiliation(s)
- Suzanne Carreira
- Signalling and Development Laboratory, Marie Curie Research Institute, Oxted, Surrey, RH8 0TL, United Kingdom
| | - Jane Goodall
- Signalling and Development Laboratory, Marie Curie Research Institute, Oxted, Surrey, RH8 0TL, United Kingdom
| | - Laurence Denat
- Developmental Genetics of Melanocytes, UMR146 Centre national de la recherche scientifique (CNRS), Institut Curie, 91405 Orsay Cedex, France
| | - Mercedes Rodriguez
- Signalling and Development Laboratory, Marie Curie Research Institute, Oxted, Surrey, RH8 0TL, United Kingdom
| | - Paolo Nuciforo
- Instituto FIRC di Oncologia Molecolare-Fondazione Italiana per la Ricerca sul Cancro Institute of Molecular Oncology, 20139 Milano, Italy
| | - Keith S. Hoek
- Department of Dermatology, University Hospital of Zürich, 8091 Zürich, Switzerland
| | | | - Lionel Larue
- Developmental Genetics of Melanocytes, UMR146 Centre national de la recherche scientifique (CNRS), Institut Curie, 91405 Orsay Cedex, France
| | - Colin R. Goding
- Signalling and Development Laboratory, Marie Curie Research Institute, Oxted, Surrey, RH8 0TL, United Kingdom
- Corresponding author.E-MAIL ; FAX 44-1882-714375
| |
Collapse
|
47
|
Magnoni C, Tenedini E, Ferrari F, Benassi L, Bernardi C, Gualdi G, Bertazzoni G, Roncaglia E, Fantoni L, Manfredini R, Bicciato S, Ferrari S, Giannetti A, Tagliafico E. Transcriptional profiles in melanocytes from clinically unaffected skin distinguish the neoplastic growth pattern in patients with melanoma. Br J Dermatol 2007; 156:62-71. [PMID: 17199568 DOI: 10.1111/j.1365-2133.2006.07564.x] [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: 01/21/2023]
Abstract
BACKGROUND It is generally accepted that sunlight may contribute to the development of melanoma. OBJECTIVES To analyse gene expression of melanocytes obtained from clinically unaffected skin of patients with melanoma and healthy controls before and after exposure to ultraviolet B radiation. METHODS Using GeneChip array technology, the gene expression of melanocytes obtained from the two donor groups was profiled, in order to identify transcriptional differences affecting susceptibility to melanoma. RESULTS The data collected did not show any difference between the expression profiles of melanocytes purified from normal donors and from patients with melanoma that was able to give a statistically significant class separation. However, by means of unsupervised clustering our data could be divided into two main classes. The first class included the transcriptome profiles of melanocytes obtained from skin samples of patients with a vertical growth phase (VGP) melanoma, while the second class included the transcriptome profiles of melanocytes obtained from skin samples of patients with a radial growth phase (RGP) melanoma. CONCLUSIONS These data suggest that melanocytes in patients with VGP and RGP melanomas show significant differences in gene expression profiles, which allow us to classify patients with melanoma also from clinically unaffected skin.
Collapse
Affiliation(s)
- C Magnoni
- Sezione di Chimica Biologica, Dipartimento di Scienze Biomediche, Università di Modena e Reggio Emilia, Via Campi 287, 41100 Modena, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Butler AP, Trono D, Beard R, Fraijo R, Nairn RS. Melanoma susceptibility and cell cycle genes inXiphophorus hybrids. Mol Carcinog 2007; 46:685-91. [PMID: 17477382 DOI: 10.1002/mc.20343] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Xiphophorus interspecies hybrids provide genetically defined models of both spontaneous and inducible melanomagenesis. In both models, backcrossing F(1) hybrids of different strains of X. maculatus and X. helleri to a X. helleri parental fish results in segregation of melanoma susceptibility, fitting a Mendelian two-gene inheritance model. The sex-linked Xmrk oncogene is required for melanoma development in both crosses. The Xiphophorus CDKN2A/B gene, which is homologous to mammalian CDKN2A/B cyclin-dependent kinase inhibitors (p16 and p15), is a candidate melanoma susceptibility gene. In this model, tumor susceptibility segregates with homozgyosity for CDKN2A/B from the recurrent X. helleri parent in backcross hybrids. We found that both CDKN2A/B mRNA and protein are highly overexpressed in melanoma. Because the p13 protein product of CDKN2A/B is a putative regulator of the G1 checkpoint, we investigated expression of other components of Xiphophorus G1 checkpoint control. By real-time PCR analysis, retinoblastoma gene (RB) is consistently expressed twofold higher in both tumors and melanized skin than in normal tissue, indicating that RB is not downregulated by the overexpression of CDKN2A/B in Xiphophorus melanoma. We also found a significant correlation between the quantitative level of CDKN2A/B and Xmrk RNA in tumors, suggesting a functional relationship between Xmrk and CDKN2A/B expression. Although X. helleri CDKN2A/B protein contains a non-conservative substitution, the biochemical function appears to show little overt defect. These studies indicate that in Xiphophorus melanoma, CDKN2A/B is functionally insufficient to mediate cell-cycle arrest in the presence of Xmrk.
Collapse
Affiliation(s)
- Andrew P Butler
- Department of Carcinogenesis, University of Texas, M. D. Anderson Cancer Center, Science Park, Research Division, Smithville, Texas 78957, USA
| | | | | | | | | |
Collapse
|
49
|
Qin JZ, Xin H, Sitailo LA, Denning MF, Nickoloff BJ. Enhanced Killing of Melanoma Cells by Simultaneously Targeting Mcl-1 and NOXA. Cancer Res 2006; 66:9636-45. [PMID: 17018621 DOI: 10.1158/0008-5472.can-06-0747] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
By deciphering the dysregulation of apoptosis in melanoma cells, new treatment approaches exploiting aberrant control mechanisms regulating cell death can be envisioned. Among the Bcl-2 family, a BH3-only member, NOXA, functions in a specific mitochondrial-based cell death pathway when melanoma cells are exposed to a proteasome inhibitor (e.g., bortezomib). Some therapeutic agents, such as bortezomib, not only induce proapoptotic Bcl-2 family members and active conformational changes in Bak and Bax but also are associated with undesirable effects, including accumulation of antiapoptotic proteins, such as Mcl-1. To enhance the bortezomib-mediated killing of melanoma cells, the apoptotic pathway involving NOXA was further investigated, leading to identification of an important target (i.e., the labile Bcl-2 homologue Mcl-1 but not other survival proteins). To reduce Mcl-1 levels, melanoma cells were pretreated with several different agents, including Mcl-1 small interfering RNA (siRNA), UV light, or the purine nucleoside analogue fludarabine. By simultaneously triggering production of NOXA (using bortezomib) as well as reducing Mcl-1 levels (using siRNA, UV light, or fludarabine), significantly enhanced killing of melanoma cells was achieved. These results show binding interactions between distinct Bcl-2 family members, such as NOXA and Mcl-1, in melanoma cells, paving the way for novel and rational therapeutic combination strategies, which target guardians of the proapoptotic Bak- and Bax-mediated pathways, against this highly aggressive and often fatal malignancy.
Collapse
Affiliation(s)
- Jian-Zhong Qin
- Department of Pathology, Loyola University Medical Center, Maywood, IL 60153, USA
| | | | | | | | | |
Collapse
|
50
|
Abstract
Cell for cell, probably no human cancer is as aggressive as melanoma. It is among a handful of cancers whose dimensions are reported in millimeters. Tumor thickness approaching 4 mm presents a high risk of metastasis, and a diagnosis of metastatic melanoma carries with it an abysmal median survival of 6-9 mo. What features of this malignancy account for such aggressive behavior? Is it the migratory history of its cell of origin or the programmed adaptation of its differentiated progeny to environmental stress, particularly ultraviolet radiation? While the answers to these questions are far from complete, major strides have been made in our understanding of the cellular, molecular, and genetic underpinnings of melanoma. More importantly, these discoveries carry profound implications for the development of therapies focused directly at the molecular engines driving melanoma, suggesting that we may have reached the brink of an unprecedented opportunity to translate basic science into clinical advances. In this review, we attempt to summarize our current understanding of the genetics and biology of this disease, drawing from expanding genomic information and lessons from development and genetically engineered mouse models. In addition, we look forward toward how these new insights will impact on therapeutic options for metastatic melanoma in the near future.
Collapse
Affiliation(s)
- Lynda Chin
- Melanoma Program, Dana-Farber Cancer Institute, Boston, Massachusetts 02115, USA.
| | | | | |
Collapse
|