1
|
Montazersaheb S, Farahzadi R, Fathi E, Alizadeh M, Abdolalizadeh Amir S, Khodaei Ardakan A, Jafari S. Investigation the apoptotic effect of silver nanoparticles (Ag-NPs) on MDA-MB 231 breast cancer epithelial cells via signaling pathways. Heliyon 2024; 10:e26959. [PMID: 38455550 PMCID: PMC10918200 DOI: 10.1016/j.heliyon.2024.e26959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 01/29/2024] [Accepted: 02/22/2024] [Indexed: 03/09/2024] Open
Abstract
Background The discovery of novel cancer therapeutic strategies leads to the development of nanotechnology-based methods for cancer treatment. Silver nanoparticles (Ag-NPs) have garnered considerable interest owing to their size, shape, and capacity to modify chemical, optical, and photonic properties. This study aimed to investigate the impact of Ag-NPs on inducing of apoptosis in MDA-MB 231 cells by examining specific signaling pathways. Materials and methods The cytotoxicity of Ag-NPs was determined using an MTT assay in MDA-MB 231 cells. The apoptotic effects were assessed using the Annexin-V/PI assay. Real-time PCR and western blotting were conducted to analyze the expression of apoptosis-related genes and proteins, respectively. Levels of ERK1/2 and cyclin D1 were measured using ELISA. Cell cycle assay was determined by flow cytometry. Cell migration was evaluated by scratch assay. Results The results revealed that Ag-NPs triggered apoptosis and cell cycle arrest in MDA-MB 231 cells. The expression level of Bax (pro-apoptotic gene) was increased, while Bcl-2 (anti-apoptotic gene) expression was decreased. Increased apoptosis was correlated with increased levels of p53 and PTEN. Additionally, notable alterations were observed in protein expression related to the Janus kinase/Signal transducers (JAK/STAT) pathway, including p-AKT. Additionally, reduced expression of h-TERT was observed following exposure to Ag-NPs. ELISA results demonstrated a significant reduction in p-ERK/Total ERK and cyclin D1 levels in Ag-NPs-exposed MDA-MB 231 cells. Western blotting analysis also confirmed the reduction of p-ERK/Total ERK and cyclin D1. Decreased level of cyclin D is associated with suppression of cell cycle progression. The migratory ability of MDA-MB-231 cells was reduced upon treatment with Ag-NPs. Conclusions Our findings revealed that Ag-NPs influenced the proliferation, apoptosis, cell cycle, and migration in MDA-MB 231 cells, possibly by modulating protein expression of the AKT/ERK/Cyclin D1 axis.
Collapse
Affiliation(s)
- Soheila Montazersaheb
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Raheleh Farahzadi
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ezzatollah Fathi
- Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Mahsan Alizadeh
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Tabriz Medical Sciences, Islamic Azad University, Tabriz, Iran
| | - Shahabaddin Abdolalizadeh Amir
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Tabriz Medical Sciences, Islamic Azad University, Tabriz, Iran
| | - Alireza Khodaei Ardakan
- Faculty of Veterinary Medicine, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Sevda Jafari
- Nutrition Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
2
|
de Kort WWB, de Ruiter EJ, Haakma WE, Driehuis E, Devriese LA, van Es RJJ, Willems SM. p-mTOR, p-ERK and PTEN Expression in Tumor Biopsies and Organoids as Predictive Biomarkers for Patients with HPV Negative Head and Neck Cancer. Head Neck Pathol 2023; 17:697-707. [PMID: 37486536 PMCID: PMC10514008 DOI: 10.1007/s12105-023-01576-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 07/11/2023] [Indexed: 07/25/2023]
Abstract
BACKGROUND Survival rates of head and neck squamous cell carcinoma (HNSCC) have only marginally improved in the last decades. Hence there is a need for predictive biomarkers for long-time survival that can help to guide treatment decisions and might lead to the development of new therapies. The phosphatidylinositol 3-kinase (PI3K)/AKT/mTOR signaling pathway is the most frequently altered pathway in HNSCC, genes are often mutated, amplificated and overexpressed causing aberrant signaling affecting cell growth and differentiation. Numerous genetic alterations of upstream and downstream factors have currently been clarified. However, their predictive value has yet to be established. Therefore we assess the predictive value of p-mTOR, p-ERK and PTEN expression. METHODS Tissue microarrays (TMA's) of HPV-negative patients with oropharyngeal (n = 48), hypopharyngeal (n = 16) or laryngeal (n = 13) SCC, treated with primary chemoradiation (cisplatin/carboplatin/cetuximab and radiotherapy), were histologically stained for p-mTOR, PTEN and p-ERK. Expression was correlated to overall survival (OS), disease free survival (DFS) and locoregional control (LRC). Also p-mTOR was histologically stained in a separate cohort of HNSCC organoids (n = 8) and correlated to mTOR-inhibitor everolimus response. RESULTS High p-mTOR expression correlated significantly with worse OS in multivariate analysis in the whole patient cohort [Hazar Ratio (HR) 1.06, 95%CI 1.01-1.11, p = 0.03] and in the cisplatin/carboplatin group with both worse OS (HR 1.09, 95%CI 1.02-1.16, p = 0.02) and DFS (HR 1.06, 95%CI 1.00-1.12, p = 0,04). p-ERK expression correlated significantly with DFS in univariate analysis in the whole patient cohort (HR 1.03, 95%CI 1.00-1.05, p = 0.04) and cisplatin/carboplatin group (HR 1.03, 95%CI 1.00-1.07, p = 0.04). PTEN-expression did not correlate with OS/DFS/LRC. Better organoid response to everolimus correlated significantly to higher p-mTOR expression (Rs = - 0.731, p = 0.04). CONCLUSIONS High p-mTOR expression predicts and high p-ERK expression tends to predict worse treatment outcome in HPV negative HNSCC patients treated with chemoradiation, providing additional evidence that these markers are candidate prognostic biomarkers for survival in this patient population. Also this study shows that the use of HNSCC organoids for biomarker research has potential. The role of PTEN expression as prognostic biomarker remains unclear, as consistent evidence on its prognostic and predictive value is lacking.
Collapse
Affiliation(s)
- W. W. B. de Kort
- Department of Pathology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
- Department of Oral and Maxillofacial Surgery, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - E. J. de Ruiter
- Department of Pathology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - W. E. Haakma
- Department of Pathology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - E. Driehuis
- Oncode Institute, Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, 3584 CT Utrecht, The Netherlands
| | - L. A. Devriese
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - R. J. J. van Es
- Department of Oral and Maxillofacial Surgery, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
- Department of Head and Neck Surgical Oncology, Utrecht Cancer Center, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - S. M. Willems
- Department of Pathology, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| |
Collapse
|
3
|
Jung JM, Lee MY, Won CH, Chang SE, Lee MW, Lee WJ. Performance of PRAME immunohistochemistry compared with that of c-Kit, c-Myc, or cyclin D1 for the diagnosis of acral melanocytic tumors. Pathol Int 2023; 73:27-36. [PMID: 36468840 DOI: 10.1111/pin.13292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 11/09/2020] [Indexed: 12/12/2022]
Abstract
The diagnostic role of preferentially expressed antigen in melanoma (PRAME) immunohistochemistry has not been thoroughly evaluated for acral melanocytic tumors. The objective of this study was to evaluate the utility of this modality for the diagnosis of acral melanocytic tumors compared with other potential markers. Melanocytic tumors were classified as either acral nevi, challenging melanocytic tumors (superficial atypical melanocytic proliferation of uncertain significance (SAMPUS)-favor benign (SAMPUS-FB), SAMPUS-favor malignant (SAMPUS-FM)) or acral melanomas. A total of 106 acral melanocytic tumors including acral nevi (n = 32), SAMPUS-FB (n = 17), SAMPUS-FM (n = 20), and acral melanomas (n = 37) were included. Diagnostic power, assessed using an area under the receiver operating characteristic curve (AUC) for distinguishing acral melanomas and acral nevi, was highest for PRAME (AUC = 0.997), followed by c-Myc (AUC = 0.755), cyclin D1 (AUC = 0.652), and c-Kit (AUC = 0.573). At a PRAME expression level ≥30% as a positive test for acral melanoma, the sensitivity and specificity of this marker for discriminating acral melanoma from acral nevus were 100% and 96.9%, respectively. PRAME immunohistochemistry also discriminated SAMPUS-FM from SAMPUS-FB with a sensitivity and specificity of 90.0% and 76.5%, respectively. In conclusion, PRAME immunohistochemistry can be used effectively to distinguish between various spectra of acral melanocytic neoplasms.
Collapse
Affiliation(s)
- Joon Min Jung
- Department of Dermatology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Mi Young Lee
- Department of Dermatology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Chong Hyun Won
- Department of Dermatology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Sung Eun Chang
- Department of Dermatology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Mi Woo Lee
- Department of Dermatology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Woo Jin Lee
- Department of Dermatology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
4
|
Cai G, Zou R, yang H, Xie J, Chen X, Zheng C, Luo S, Wei N, Liu S, Chen R. Circ_0084043-miR-134-5p axis regulates PCDH9 to suppress melanoma. Front Oncol 2022; 12:891476. [PMID: 36387162 PMCID: PMC9641620 DOI: 10.3389/fonc.2022.891476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 09/20/2022] [Indexed: 11/29/2022] Open
Abstract
The low survival rates, poor responses, and drug resistance of patients with melanoma make it urgent to find new therapeutic targets. This study investigated whether the circ_0084043-miR-134-5p axis regulates the antitumor effect of protocadherin 9 (PCDH9) in melanoma. Ectopic expression or knock down (KD) of PCDH9 with a lentivirus vector, we explored its effects on the proliferation, invasion, and apoptosis of melanoma and verified its regulatory effect on ras-related C3 botulinum toxin substrate 1 (RAC1), proline-rich tyrosine kinase 2 (Pyk2), Cyclin D1, matrix metalloproteinase 2 (MMP2), and MMP9. We further observed the effect of KD circ_0084043 on the malignant behavior of melanoma and studied whether circ_0084043 sponged miR-134-5p and regulated PCDH9. We found that circ_0084043 was overexpressed in melanoma and associated with the malignant phenotype. PCDH9 was poorly expressed in human melanoma tissues, and overexpression of PCDH9 inhibited melanoma progression. Quantitative real-time PCR and Western blotting results showed that overexpression of PCDH9 could downregulate RAC1, MMP2, and MMP9 and upregulate Pyk2 and Cyclin D1. Circ_0084043 KD inhibited invasion and promoted apoptosis in melanoma cells. Circ_0084043 could sponge miR-134-5p and thus indirectly regulate PCDH9. Furthermore, we discovered that inhibiting circ_0084043 had an anti–PD-Ll effect. In vivo, PCDH9 overexpression inhibited melanoma tumor growth, but PCDH9 KD promoted it. In conclusion, PCDH9, which is regulated by the circ 0084043-miR-134-5p axis, can suppress malignant biological behavior in melanoma and influence the expression levels of Pyk2, RAC1, Cyclin D1, MMP2, and MMP9.
Collapse
Affiliation(s)
- Guiyue Cai
- Dermatology Department, Dermatology Hospital, Southern Medical University, Guangzhou, China
- Clinical School, Guangdong Medical University, Zhanjiang, China
| | - Ruitao Zou
- Dermatology Department, Dermatology Hospital, Southern Medical University, Guangzhou, China
- Clinical School, Guangdong Medical University, Zhanjiang, China
| | - Huizhi yang
- Dermatology Department, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jiahao Xie
- Dermatology Department, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiaoxuan Chen
- Clinical School, Guangdong Medical University, Zhanjiang, China
| | - Chunchan Zheng
- Dermatology Department, Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Sujun Luo
- Dermatology Department, Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Na Wei
- Dermatology Department, Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Shuang Liu
- Dermatology Department, Dermatology Hospital, Southern Medical University, Guangzhou, China
- *Correspondence: Shuang Liu, ; Rongyi Chen,
| | - Rongyi Chen
- Dermatology Department, Dermatology Hospital, Southern Medical University, Guangzhou, China
- Clinical School, Guangdong Medical University, Zhanjiang, China
- *Correspondence: Shuang Liu, ; Rongyi Chen,
| |
Collapse
|
5
|
Tsutsumi R, Yoshida Y, Yamamoto O. Minute Acral Melanoma. JAMA Dermatol 2021; 157:1371. [PMID: 34586339 DOI: 10.1001/jamadermatol.2021.3298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Affiliation(s)
- Reiko Tsutsumi
- Division of Dermatology, Department of Medicine of Sensory and Motor Organs, Faculty of Medicine, Tottori University, Tottori, Japan
| | - Yuichi Yoshida
- Division of Dermatology, Department of Medicine of Sensory and Motor Organs, Faculty of Medicine, Tottori University, Tottori, Japan
| | - Osamu Yamamoto
- Division of Dermatology, Department of Medicine of Sensory and Motor Organs, Faculty of Medicine, Tottori University, Tottori, Japan
| |
Collapse
|
6
|
Aprilliantina YS, Novita HD, Sadono EG, Aldina R. Protective Effect of Genistein on Cyclin D1 Expression in Malignant Ocular Melanoma Cells. Med Arch 2021; 75:180-183. [PMID: 34483446 PMCID: PMC8385747 DOI: 10.5455/medarh.2021.75.180-183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 06/26/2021] [Indexed: 11/23/2022] Open
Abstract
Background: Ocular melanoma is a disorder that is rarely found but is deadly. Four tissues in the eye that can be attacked by melanoma include the uveal tract, conjunctiva, eyelids, and orbit. Uveal melanoma is the most common case, while melanoma conjunctiva is very rare. Objective: This study aimed to investigate the effect of giving genistein on cyclin D1 expression in malignant melanoma. Methods: When confluent, CRL1872 malignant melanoma cells will be divided into treatment groups, the group giving genistein dose 25 μM, the group giving genistein a dose of 50 μM, and the group giving genistein a dose of 100 μM. Cyclin D1 analysis was measured by immunofluorescence using confocal laser scan microscopy. Results: There was a significant increase in the expression of cyclin D1, in the group given genistein 25 μM and 50 μM (p < 0.05). For the administration of the genistein dose of 100 μM, cyclin D1 expression decreased significantly compared to the control group (p < 0.05). Conclusion: It was concluded that genistein had a biphasic effect on cyclin D1 expression in malignant melanoma cells. Thus, genistein at the right dose can be a treatment of malignant melanoma.
Collapse
Affiliation(s)
- Yasmin Sani Aprilliantina
- Department of Ophtalmology, Dr. Saiful Anwar General Hospital/Faculty of Medicine, Universitas Brawijaya, Malang, East Java, Indonesia
| | - Hera Dwi Novita
- Department of Ophtalmology, Dr. Saiful Anwar General Hospital/Faculty of Medicine, Universitas Brawijaya, Malang, East Java, Indonesia
| | - Elfina G Sadono
- Department of Ophtalmology, Dr. Saiful Anwar General Hospital/Faculty of Medicine, Universitas Brawijaya, Malang, East Java, Indonesia
| | - Rosy Aldina
- Department of Ophtalmology, Dr. Saiful Anwar General Hospital/Faculty of Medicine, Universitas Brawijaya, Malang, East Java, Indonesia
| |
Collapse
|
7
|
Volgareva GM, Zavalishina LE, Kuznetsova OA, Burova OS. [Reed nevus and cutaneous melanoma in children: an immunohistochemical study]. Arkh Patol 2021; 83:30-34. [PMID: 34041893 DOI: 10.17116/patol20218303130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Certain difficulties arise in the differential diagnosis between Reed nevus that is common in children and adolescents and cutaneous melanoma that is extremely rare in patients of this age group. In addition to the classical histological examination, an immunohistochemical test for marker proteins is carried out to improve the accuracy of the diagnosis of pigmented skin neoplasms. OBJECTIVE To evaluate the suitability of a set of proteins (cyclin D1, p16INK4a, and HLA class I antigens) in the differential diagnosis of Reed nevus and cutaneous melanoma in children and adolescents. MATERIAL AND METHODS Three samples of pigmented skin neoplasms were taken during surgical treatment in patients at the Blokhin National Medical Research Center of Oncology: two samples of Reed nevus (from a 9-year-old girl and a 15-year-old boy) and a sample of cutaneous melanoma that had developed in an 8-year-old boy. The materials were presented by paraffin blocks. Immunohistochemical staining was performed using antibodies against cyclin D1, p16INK4a, and HLA class I antigens. RESULTS Differences were found between nevus and melanoma in the expression of all three markers. Thus, the nuclear expression of cyclin D1, a proliferation activator, occurred in some cells in the nevus samples and in the vast majority of cells in the melanoma samples. The nuclear expression of the cell cycle inhibitor p16INK4a was dramatically reduced in melanoma compared to nevus. HLA class I antigens were detected on the surface of individual nevus cells, but were completely absent on the membranes and in the cytoplasm of melanoma cells, which could promote the evasion of this tumor from the body's immunological supervision. CONCLUSION Immunohistochemical test for the proteins cyclin D1, p16INK4a, and HLA class 1 antigens is a promising approach to differentially diagnosing between Reed nevus and cutaneous melanoma in children and adolescents.
Collapse
Affiliation(s)
- G M Volgareva
- Blokhin National Medical Research Center of Oncology, Moscow, Russia
| | - L E Zavalishina
- Russian Medical Academy for Continuous Professional Education, Moscow, Russia
| | - O A Kuznetsova
- Russian Medical Academy for Continuous Professional Education, Moscow, Russia
| | - O S Burova
- Blokhin National Medical Research Center of Oncology, Moscow, Russia
| |
Collapse
|
8
|
Smedley RC, Thaiwong T, Deeth LE, Kiupel M. Correlation Between KIT Expression and c-Kit Mutations in 2 Subtypes of Canine Oral Melanocytic Neoplasms. Vet Pathol 2021; 58:683-691. [PMID: 33910439 DOI: 10.1177/03009858211009784] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
c-Kit mutations have been reported in 15% to 40% of certain human melanoma subtypes, including those histologically similar to canine oral malignant melanomas. Therapeutic response to tyrosine kinase inhibitors has been demonstrated in those human patients. As canine oral malignant melanomas tend to have a poor prognosis despite aggressive surgical removal, evaluation of KIT expression and identification of c-Kit mutations in canine oral melanocytic neoplasms was performed to determine if there is any indication that tyrosine kinase inhibitor drugs might effectively treat any of these cases. This study evaluated 27 canine oral malignant melanomas and 12 canine histologically well-differentiated oral melanocytic neoplasms for activating c-Kit mutations, determined differences in immunohistochemical expression of KIT and c-Kit mutation status, and determined if KIT expression could predict c-Kit mutation status. Among samples that contained intraepithelial nests of neoplastic melanocytes in the KIT-labeled sections, KIT was expressed within cells in these nests in 22/23 (96%) malignant melanomas and 5/7 histologically well-differentiated neoplasms. KIT was expressed in 10% to 30% of neoplastic melanocytes in the lamina propria in 3/24 (13%) malignant melanomas, but 0/9 (0%) histologically well-differentiated neoplasms. Next-generation sequencing identified 85 variants in c-Kit, including 9 nonsynonymous mutations that resulted in amino acid changes predicted to affect protein function. c-Kit mutations with predicted deleterious protein effects were more common in malignant melanomas (8/27 [30%] vs 1/12 [8%]). There was no apparent relationship between detected c-Kit mutations and KIT expression. These results do not support the use of therapies that target c-Kit.
Collapse
|
9
|
Tseng YS, Wu PR, Lu JW, Wang YF, Yeh KT, Lin SH. Cytoplasmic phosphorylated ERK1/2 expression in patients with melanoma is associated with tumor stage and metastasis. Biotech Histochem 2021; 97:118-125. [PMID: 33902381 DOI: 10.1080/10520295.2021.1912827] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Melanoma is the cause of most deaths from skin cancer. The extracellular signal-regulated kinase 1/2 (ERK1/2) pathway has been reported to participate in progression of melanoma in fair skinned populations. ERK1/2 is found in both the cytoplasm and nucleus of cells, and phosphorylated ERK1/2 has been implicated in tumor progression. We investigated the relation between melanoma progression and expression of cytoplasmic and nuclear phosphorylated ERK1/2. We examined 34 surgically resected melanomas and investigated their clinicopathologic characteristics. We found immunostaining of phosphorylated ERK1/2 in all melanomas and faint staining in benign nevi. We found expression of cytoplasmic phosphorylated ERK1/2 in most melanomas; however, nuclear phosphorylated ERK1/2 expression was found in only five melanomas. Expression of cytoplasmic phosphorylated ERK1/2 was related to the tumor stage in melanoma. Nine of 10 cases of distant metastasis were positive for cytoplasmic phosphorylated ERK1/2. Our findings suggest that phosphorylated ERK1/2 expression is relevant to clinical pathology and that in melanoma patients, phosphorylated ERK1/2 expression is found in both the cytoplasm and nucleus. Our findings suggest that cytoplasmic phosphorylated ERK1/2 participates in progression of melanoma and that it could be a useful target for clinical treatment of melanoma.
Collapse
Affiliation(s)
- Yen-Shuo Tseng
- Department of Dermatology, Changhua Christian Hospital, Changhua, Taiwan
| | - Pei-Ru Wu
- Department of Surgical Pathology, Cheng Ching Hospital, Taichung, Taiwan
| | - Jeng-Wei Lu
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Yu-Fen Wang
- Cancer Research Center, Changhua Christian Hospital, Changhua, Taiwan
| | - Kun-Tu Yeh
- Department of Surgical Pathology, Changhua Christian Hospital, Changhua, Taiwan.,School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Shu-Hui Lin
- Department of Surgical Pathology, Changhua Christian Hospital, Changhua, Taiwan.,Department of Medical Laboratory Science and Biotechnology, Central Taiwan University of Science and Technology, Taichung, Taiwan
| |
Collapse
|
10
|
Kaufmann C, Kempf W, Mangana J, Cheng P, Emberger M, Lang R, Kaiser AK, Lattmann E, Levesque M, Dummer R, Koelblinger P. The role of cyclin D1 and Ki-67 in the development and prognostication of thin melanoma. Histopathology 2020; 77:460-470. [PMID: 32374893 PMCID: PMC7540531 DOI: 10.1111/his.14139] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Accepted: 05/03/2020] [Indexed: 12/16/2022]
Abstract
AIMS Despite their low individual metastatic potential, thin melanomas (≤1 mm Breslow thickness) contribute significantly to melanoma mortality overall. Therefore, identification of prognostic biomarkers is particularly important in this subgroup of melanoma. Prompted by preclinical results, we investigated cyclin D1 protein and Ki-67 expression in in-situ, metastatic and non-metastatic thin melanomas. METHODS AND RESULTS Immunohistochemistry was performed on 112 melanoma specimens, comprising 22 in situ, 48 non-metastatic and 42 metastatic thin melanomas. Overall, epidermal and dermal cyclin D1 and Ki-67 expression were semiquantitatively evaluated by three independent investigators and compared between groups. Epidermal Ki-67 expression did not differ statistically in in-situ and invasive melanoma (P = 0.7). Epidermal cyclin D1 expression was significantly higher in thin invasive than in in-situ melanoma (P = 0.003). No difference was found in cyclin D1 expression between metastatic and non-metastatic invasive tumours. Metastatic and non-metastatic thin melanomas did not show significant differences in epidermal expression of Ki-67 and cyclin D1 (P = 0.148 and P = 0.611, respectively). In contrast, strong dermal expression of Ki-67 was more frequent in metastatic than non-metastatic samples (28.6 versus 8.3%, respectively, P = 0.001). The prognostic value of dermal Ki-67 expression was confirmed by multivariate analysis (P = 0.047). CONCLUSION We found an increased expression of cyclin D1 in invasive thin melanomas compared to in-situ melanomas, which supports a potential role of this protein in early invasion in melanoma, as suggested by preclinical findings. Moreover, our results confirm that high dermal Ki-67 expression is associated with an increased risk of development of metastasis in thin melanoma and could possibly serve as a prognostic biomarker in clinical practice, especially if combined with additional methods.
Collapse
Affiliation(s)
- Corina Kaufmann
- Department of DermatologyUniversity Hospital ZurichZurichSwitzerland
- University of ZurichZurichSwitzerland
| | - Werner Kempf
- Kempf and Pfaltz Histological DiagnosticsZurichSwitzerland
| | - Joanna Mangana
- Department of DermatologyUniversity Hospital ZurichZurichSwitzerland
| | - Phil Cheng
- Department of DermatologyUniversity Hospital ZurichZurichSwitzerland
| | | | - Roland Lang
- Department of Dermatology and AllergologyParacelsus Medical UniversitySalzburgAustria
| | - Andreas K Kaiser
- Department of Clinical PsychologyChristian‐Doppler‐KlinikParacelsus Medical UniversitySalzburgAustria
| | | | - Mitchell Levesque
- Department of DermatologyUniversity Hospital ZurichZurichSwitzerland
| | - Reinhard Dummer
- Department of DermatologyUniversity Hospital ZurichZurichSwitzerland
| | - Peter Koelblinger
- Department of DermatologyUniversity Hospital ZurichZurichSwitzerland
- Department of Dermatology and AllergologyParacelsus Medical UniversitySalzburgAustria
| |
Collapse
|
11
|
González-Ruiz L, González-Moles MÁ, González-Ruiz I, Ruiz-Ávila I, Ayén Á, Ramos-García P. An update on the implications of cyclin D1 in melanomas. Pigment Cell Melanoma Res 2020; 33:788-805. [PMID: 32147907 DOI: 10.1111/pcmr.12874] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 02/03/2020] [Accepted: 03/02/2020] [Indexed: 12/13/2022]
Abstract
Cyclin D1 is a protein encoded by the CCND1 gene, located on 11q13 chromosome, which is a key component of the physiological regulation of the cell cycle. CCND1/cyclin D1 is upregulated in several types of human tumors including melanoma and is currently classified as an oncogene that promotes uncontrolled cell proliferation. Despite the demonstrated importance of CCND1/cyclin D1 as a central oncogene in several types of human tumors, its knowledge in melanoma is still limited. This review examines data published on upregulation of the CCND1 gene and cyclin D1 protein in the melanoma setting, focusing on the pathways and molecular mechanisms involved in the activation of the gene and on the clinical and therapeutic implications.
Collapse
Affiliation(s)
- Lucia González-Ruiz
- Dermatology Service, Ciudad Real General University Hospital, Ciudad Real, Spain
| | | | | | - Isabel Ruiz-Ávila
- Biohealth Research Institute, Granada, Spain.,Pathology Service, San Cecilio Hospital Complex, Granada, Spain
| | - Ángela Ayén
- Dermatology Service, San Cecilio Hospital Complex, Granada, Spain
| | | |
Collapse
|
12
|
Moradi Binabaj M, Bahrami A, Khazaei M, Ryzhikov M, Ferns GA, Avan A, Mahdi Hassanian S. The prognostic value of cyclin D1 expression in the survival of cancer patients: A meta-analysis. Gene 2019; 728:144283. [PMID: 31838249 DOI: 10.1016/j.gene.2019.144283] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 11/30/2019] [Accepted: 12/05/2019] [Indexed: 01/21/2023]
Abstract
BACKGROUND The relationship between the expression of cyclin D1 and cancer prognosis and outcomes in different malignancies has not been fully elucidated. AIMS In the presented meta-analysis, we assessed the association between the expression level of cyclin D1 with overall survival (OS) in several cancers. METHODS Eligible studies were identified using PubMed, EMBase, Scopus, Web of Sciences and Cochrane Library databases. For the prognostic meta-analysis, study-specific hazard ratios (HRs) of tissue cyclin D1 for survival were obtained. Finally we pooled data derived from one hundred and eight studies comprising 19,224 patients with 10 different cancer types. RESULTS In the pooled analysis, high expression of cyclin D1 was significantly related to a poor OS with a pooled HR of 1.11 (95% CI: 1.02-1.20, P = 0.015; random-effects). Sub-group analysis revealed that high expression of cyclin D1 was related to worse OS of head and neck cancers (HR = 2.08, 95% CI: 1.75-2.47; P < 0.001), but not in breast (HR = 1.033, 95% CI: 0.873-1.223, P = 0.702), gastrointestinal (HR = 1.025, 95% CI:0.824-1.275; P = 0.825), bladder (HR = 0.937, CI: 0.844-1.041; P = 0.225) and in lung cancer patients (HR = 1.092, CI: 0.819-1.455; P = 0.549). CONCLUSION Further large, prospective, and well-designed trials are warranted to elucidate the precise clinical importance of cyclin D1 overexpression in the prognosis of cancer patients receiving different treatment regimens.
Collapse
Affiliation(s)
- Maryam Moradi Binabaj
- Department of Medical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Afsane Bahrami
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Majid Khazaei
- Department of Medical Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mikhail Ryzhikov
- Division of Pulmonary and Critical Care Medicine, Washington University, School of Medicine, Saint Louis, MO, USA
| | - Gordon A Ferns
- Brighton & Sussex Medical School, Division of Medical Education, Falmer, Brighton, Sussex BN1 9PH, UK
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Cancer Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Modern Sciences and Technologies, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mahdi Hassanian
- Department of Medical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
13
|
Hahn HM, Lee KG, Choi W, Cheong SH, Myung KB, Hahn HJ. An updated review of mucosal melanoma: Survival meta-analysis. Mol Clin Oncol 2019; 11:116-126. [PMID: 31281645 PMCID: PMC6589937 DOI: 10.3892/mco.2019.1870] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 04/10/2019] [Indexed: 12/12/2022] Open
Abstract
Mucosal melanoma (MM) is a highly lethal variant of melanoma that carries a poor prognosis. Extremely low incidence and survival rates have led to few clinical trials, and a lack of protocols and guidelines. The present study performed a survival meta-analysis for the quantitative synthesis of available evidence to search for key patterns that would help clinicians tailor optimal therapeutic strategies in MM. PubMed, EMBASE, Cochrane, MEDLINE, Google Scholar and other databases were searched. Hazard ratios, in disease-specific and overall survival, were calculated for each of the survival-determining variables. MM was 2.25 times more lethal than cutaneous melanoma (CM). The most significant threats to survival were advanced Tumor-Node-Metastasis stage, sino-nasal location, and old age. Chemotherapy was the most effective form of adjuvant therapy. Disease-specific survival, the primary measure of the effect sizes, can fluctuate depending on the accuracy of the reported cause of mortality. In conclusion, MM is a peculiar type of melanoma, with clinical and molecular profile vastly different from the much-familiar CM. In the wake of the era of precision oncology, further studies on driver mutations and oncogenic pathways would likely lead to improved patient survival.
Collapse
Affiliation(s)
- Hyung Min Hahn
- Department of Plastic and Reconstructive Surgery, Ajou University School of Medicine, Suwon, Gyeonggi 16499, Republic of Korea
| | - Kyoung Geun Lee
- Department of Dermatology, College of Medicine, Konyang University, Daejeon, Chungcheongnam 35365, Republic of Korea
| | - Won Choi
- Department of Dermatology, College of Medicine, Konyang University, Daejeon, Chungcheongnam 35365, Republic of Korea
| | - Seung Hyun Cheong
- Department of Dermatology, College of Medicine, Konyang University, Daejeon, Chungcheongnam 35365, Republic of Korea
| | - Ki Bum Myung
- Department of Dermatology, College of Medicine, Konyang University, Daejeon, Chungcheongnam 35365, Republic of Korea
| | - Hyung Jin Hahn
- Department of Dermatology, College of Medicine, Konyang University, Daejeon, Chungcheongnam 35365, Republic of Korea.,Myunggok Medical Research Institute, College of Medicine, Konyang University, Daejeon, Chungcheongnam 35365, Republic of Korea
| |
Collapse
|
14
|
Beretti F, Bertoni L, Farnetani F, Pellegrini C, Gorelli G, Cesinaro AM, Reggiani Bonetti L, Di Nardo L, Kaleci S, Chester J, Longo C, Massi D, Fargnoli MC, Pellacani G. Melanoma types by in vivo reflectance confocal microscopy correlated with protein and molecular genetic alterations: A pilot study. Exp Dermatol 2019; 28:254-260. [PMID: 30636079 DOI: 10.1111/exd.13877] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 12/21/2018] [Accepted: 01/07/2019] [Indexed: 12/21/2022]
Abstract
Cutaneous melanoma (CM) is one of the most prevalent skin cancers, which lacks both a prognostic marker and a specific and lasting treatment, due to the complexity of the disease and heterogeneity of patients. Reflectance confocal microscopy (RCM) in vivo analysis is a versatile approach offering immediate morphological information, enabling the identification of four primary cutaneous RCM CM types. Whether RCM CM types are associated with a specific protein and molecular genetic profiles at the tissue level remains unclear. The current pilot study was designed to identify potential correlations between RCM CM types and specific biological characteristics, combining immunohistochemistry (IHC) and molecular analyses. Eighty primary CMs evaluated at patient bedside with RCM (type 1 [19, 24%], type 2 [12, 15%], type 3 [7, 9%] and type 4 [42, 52%]) were retrospectively evaluated by IHC stains (CD271, CD20, CD31, cyclin D1), fluorescence in situ hybridization FISH for MYC gain and CDKN2A loss and molecular analysis for somatic mutations (BRAF, NRAS and KIT). RCM CM types correlated with markers of stemness property, density of intra-tumoral lymphocytic B infiltrate and cyclin D1 expression, while no significant association was found with blood vessel density nor molecular findings. RCM CM types show a different marker profile expression, suggestive of a progression and an increase in aggressiveness, according to RCM morphologies.
Collapse
Affiliation(s)
- Francesca Beretti
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Laura Bertoni
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Francesca Farnetani
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
- Department of Dermatology, University of Modena and Reggio Emilia, Modena, Italy
| | | | - Greta Gorelli
- Division of Pathological Anatomy, Department of Surgery and Translational Medicine, University of Florence, Florence, Italy
| | - Anna Maria Cesinaro
- Anatomic Pathology, Azienda Ospedaliero-Universitaria Policlinico, Modena, Italy
| | - Luca Reggiani Bonetti
- Department of Medical and Surgical Sciences of Children & Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Lucia Di Nardo
- Department of Dermatology, University of L'Aquila, L'Aquila, Italy
| | - Shaniko Kaleci
- Department of Dermatology, University of Modena and Reggio Emilia, Modena, Italy
| | - Johanna Chester
- Department of Dermatology, University of Modena and Reggio Emilia, Modena, Italy
| | - Caterina Longo
- Department of Dermatology, University of Modena and Reggio Emilia, Modena, Italy
- Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Centro Oncologico ad Alta Tecnologia Diagnostica, Reggio Emilia, Italy
| | - Daniela Massi
- Division of Pathological Anatomy, Department of Surgery and Translational Medicine, University of Florence, Florence, Italy
| | - Maria C Fargnoli
- Department of Dermatology, University of L'Aquila, L'Aquila, Italy
| | - Giovanni Pellacani
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
- Department of Dermatology, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
15
|
Harris AL, Lee SE, Dawson LK, Marlow LA, Edenfield BH, Durham WF, Flotte TJ, Thompson M, Small DL, Synnott AJ, Markovic SN, Copland JA. Targeting the cyclin dependent kinase and retinoblastoma axis overcomes standard of care resistance in BRAF V600E -mutant melanoma. Oncotarget 2017. [PMID: 29541385 PMCID: PMC5834273 DOI: 10.18632/oncotarget.23649] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Patient-derived tumor xenograft (PDTX) mouse models were used to discover new therapies for naïve and drug resistant BRAFV600E -mutant melanoma. Tumor histology, oncogenic protein expression, and antitumor activity were comparable between patient and PDTX-matched models thereby validating PDTXs as predictive preclinical models of therapeutic response in patients. PDTX models responsive and non-responsive to BRAF/MEK standard of care (SOC) therapy were used to identify efficacious combination therapies. One such combination includes a CDK4/6 inhibitor that blocks cell cycle progression. The rationale for this is that the retinoblastoma protein (pRb) is 95% wildtype in BRAF mutant melanoma. We discovered that 77/77 stage IV metastatic melanoma tissues were positive for inactive phosphorylated pRb (pRb-Ser780). Rb is hyperphosphorylated and inactivated by CDK4/6:cyclin D1 and when restored to its hypophosphorylated active form blocks cell cycle progression. The addition of a CDK4/6 inhibitor to SOC therapy was superior to SOC. Importantly, triple therapy in an upfront treatment and salvage therapy setting provided sustained durable response. We also showed that CDK4/6 blockade resensitized drug resistant melanoma to SOC therapy. Durable response was associated with sustained suppression of pRb-Ser780. Thus, reactivation of pRb may prove to be a clinical biomarker of response and the mechanism responsible for durable response. In light of recent clinical trial data using this triple therapy against BRAFV600E -mutant melanoma, our findings demonstrating superior and prolonged durable response in PDTX models portend use of this therapeutic strategy against naïve and SOC resistant BRAFV600E -mutant metastatic melanoma coupled with pRB-Ser780 as a biomarker of response.
Collapse
Affiliation(s)
| | | | | | - Laura A Marlow
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | | | | | - Thomas J Flotte
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | | | | | | | | | - John A Copland
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| |
Collapse
|
16
|
Treatment of melanoma with selected inhibitors of signaling kinases effectively reduces proliferation and induces expression of cell cycle inhibitors. Med Oncol 2017; 35:7. [PMID: 29214525 PMCID: PMC5719123 DOI: 10.1007/s12032-017-1069-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 11/30/2017] [Indexed: 01/01/2023]
Abstract
Cancer treatment often tends to involve direct targeting enzymes essential for the growth and proliferation of cancer cells. The aim of this study was the recognition of the possible role of selected protein kinases: PI3K, ERK1/2, and mTOR in cell proliferation and cell cycle in malignant melanoma. We investigated the role of protein kinase inhibitors: U0126 (ERK1/2), LY294002 (PI3K), rapamycin (mTOR), everolimus (mTOR), GDC-0879 (B-RAF), and CHIR-99021 (GSK3beta) in cell proliferation and expression of crucial regulatory cell cycle proteins in human melanoma cells: WM793 (VGP) and Lu1205 (metastatic). They were used either individually or in various combinations. The study on the effect of signaling kinases inhibitors on proliferation—BrdU ELISA test after 48–72 h. Their effect on the expression of cell cycle regulatory proteins: cyclin D1 and D3, cyclin-dependent kinase CDK4 and CDK6, and cell cycle inhibitors: p16, p21, and p27, was studied at the protein level (western blot). Treatment of melanoma cells with protein kinase inhibitors led to significantly decreased cell proliferation except the use of a GSK-3β kinase inhibitors—CHIR-99021. The significant decrease in the expression of selected cyclins and cyclin-dependent kinases (CDKs) with parallel increase in the expression of some of cyclin-dependent kinases inhibitors and in consequence meaningful reduction in melanoma cell proliferation by the combinations of inhibitors of signaling kinases clearly showed the crucial role of AKT, ERK 1/2, and mTOR signal transduction in melanoma progression. The results unanimously indicate those pathways as an important target for treatment of melanoma.
Collapse
|
17
|
Chagani S, Wang R, Carpenter EL, Löhr CV, Ganguli-Indra G, Indra AK. Ablation of epidermal RXRα in cooperation with activated CDK4 and oncogenic NRAS generates spontaneous and acute neonatal UVB induced malignant metastatic melanomas. BMC Cancer 2017; 17:736. [PMID: 29121869 PMCID: PMC5679438 DOI: 10.1186/s12885-017-3714-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 10/30/2017] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Understanding the underlying molecular mechanisms involved in the formation of cutaneous malignant melanoma is critical for improved diagnosis and treatment. Keratinocytic nuclear receptor Retinoid X Receptor α (RXRα) has a protective role against melanomagenesis and is involved in the regulation of keratinocyte and melanocyte homeostasis subsequent acute ultraviolet (UV) irradiation. METHODS We generated a trigenic mouse model system (RXRα ep-/- | Tyr-NRAS Q61K | CDK4 R24C/R24C ) harboring an epidermal knockout of Retinoid X Receptor α (RXRα ep-/- ), combined with oncogenic NRAS Q61K (constitutively active RAS) and activated CDK4 R24C/R24C (constitutively active CDK4). Those mice were subjected to a single neonatal dose of UVB treatment and the role of RXR α was evaluated by characterizing the molecular and cellular changes that took place in the untreated and UVB treated trigenic RXRα ep-/- mice compared to the control mice with functional RXRα. RESULTS Here we report that the trigenic mice develops spontaneous melanoma and exposure to a single neonatal UVB treatment reduces the tumor latency in those mice compared to control mice with functional RXRα. Melanomas from the trigenic RXRα ep-/- mice are substantial in size, show increased proliferation, exhibit increased expression of malignant melanoma markers and exhibit enhanced vascularization. Altered expression of several biomarkers including increased expression of activated AKT, p21 and cyclin D1 and reduced expression of pro-apoptotic marker BAX was observed in the tumor adjacent normal (TAN) skin of acute ultraviolet B treated trigenic RXRα ep-/- mice. Interestingly, we observed a significant increase in p21 and Cyclin D1 in the TAN skin of un-irradiated trigenic RXRα ep-/- mice, suggesting that those changes might be consequences of loss of functional RXRα in the melanoma microenvironment. Loss of RXRα in the epidermal keratinocytes in combination with oncogenic NRAS Q61K and CDK4 R24C/R24C mutations in trigenic mice led to significant melanoma invasion into the draining lymph nodes as compared to controls with functional RXRα. CONCLUSIONS Our study demonstrates the protective role of keratinocytic RxRα in (1) suppressing spontaneous and acute UVB-induced melanoma, and (2) preventing progression of the melanoma to malignancy in the presence of driver mutations like activated CDK4 R24C/R24C and oncogenic NRAS Q61K .
Collapse
Affiliation(s)
- Sharmeen Chagani
- Molecular and Cellular Biology Program, OSU, Corvallis, 97331, OR, USA
- Department of Pharmaceutical Sciences, College of Pharmacy, OSU, Corvallis, 97331, OR, USA
| | - Rong Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, OSU, Corvallis, 97331, OR, USA
- Linus Pauling Institute, OSU, Corvallis, OR, USA
| | - Evan L Carpenter
- Department of Pharmaceutical Sciences, College of Pharmacy, OSU, Corvallis, 97331, OR, USA
| | - Christiane V Löhr
- College of Veterinary Medicine, Oregon State University, Corvallis, Oregon, 97331, USA
| | - Gitali Ganguli-Indra
- Molecular and Cellular Biology Program, OSU, Corvallis, 97331, OR, USA
- Department of Pharmaceutical Sciences, College of Pharmacy, OSU, Corvallis, 97331, OR, USA
- Knight Cancer Institute, Oregon Health & Science University (OHSU), Portland, 97239, OR, USA
| | - Arup K Indra
- Molecular and Cellular Biology Program, OSU, Corvallis, 97331, OR, USA.
- Department of Pharmaceutical Sciences, College of Pharmacy, OSU, Corvallis, 97331, OR, USA.
- Linus Pauling Institute, OSU, Corvallis, OR, USA.
- Knight Cancer Institute, Oregon Health & Science University (OHSU), Portland, 97239, OR, USA.
- Department of Dermatology, OHSU, Portland, 97239, OR, USA.
| |
Collapse
|
18
|
Zhu L, Kalimuthu S, Gangadaran P, Oh JM, Lee HW, Baek SH, Jeong SY, Lee SW, Lee J, Ahn BC. Exosomes Derived From Natural Killer Cells Exert Therapeutic Effect in Melanoma. Theranostics 2017; 7:2732-2745. [PMID: 28819459 PMCID: PMC5558565 DOI: 10.7150/thno.18752] [Citation(s) in RCA: 318] [Impact Index Per Article: 45.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 05/08/2017] [Indexed: 12/13/2022] Open
Abstract
Objective: Exosomes are nanovesicles that are released from normal and tumor cells and are detectable in cell culture supernatant and human biological fluids. Although previous studies have explored exosomes released from cancer cells, little is understood regarding the functions of exosomes released by normal cells. Natural killer (NK) cells display rapid immunity to metastatic or hematological malignancies, and efforts have been undertaken to clinically exploit the antitumor properties of NK cells. However, the characteristics and functions of exosomes derived from NK cells remain unknown. In this study, we explored NK cell-derived exosome-mediated antitumor effects against aggressive melanoma in vitro and in vivo. Methods: B16F10 cells were transfected with enhanced firefly luciferase (effluc) and thy1.1 genes, and thy1.1-positive cells were immunoselected using microbeads. The resulting B16F10/effluc cells were characterized using reverse transcriptase polymerase chain reaction (RT-PCR), western blotting, and luciferase activity assays. Exosomes derived from NK-92MI cells (NK-92 Exo) were isolated by ultracentrifugation and density gradient ultracentrifugation. NK-92 Exo were characterized by transmission electron microscopy and western blotting. We also performed an enzyme-linked immunosorbent assay to measure cytokines retained in NK-92 Exo cells. The in vitro cytotoxicity of NK-92 Exo against the cancer cells was determined using a bioluminescence imaging system (BLI) and CCK-8 assays. To investigate the possible side effects of NK-92 Exo on healthy cells, we also performed the BLI and CCK-8 assays using the human kidney Phoenix™-Ampho cell line. Flow cytometry and western blotting confirmed that NK-92 Exo induced apoptosis in the B16F10/effluc cells. In vivo, we used a B16F10/effluc cell xenograft model to detect the immunotherapeutic effect of NK-92 Exo. We injected NK-92 Exo into tumors, and tumor growth progression was monitored using the IVIS Lumina imaging system and ultrasound imaging. Tumor mass was monitored after in vivo experiments. Results: RT-PCR and western blotting confirmed effluc gene expression and protein levels in B16F10/effluc cells. B16F10/effluc activity was found to increase with increasing cell numbers, using BLI assay. For NK-92 Exo characterization, western blotting was performed on both ultracentrifuged and density gradient-isolated exosomes. The results confirmed that NK cell-derived exosomes express two typical exosome proteins, namely CD63 and ALIX. We demonstrated by western blot analysis that NK-92 Exo presented two functional NK proteins, namely perforin and FasL. Moreover, we confirmed the membrane expression of FasL. The enzyme-linked immunosorbent assay results indicated that NK-92 Exo can secrete tumor necrosis factor (TNF)-α, which affected the cell proliferation signaling pathway. The antitumor effect of NK-92 Exo against B16F10/effluc cells in vitro was confirmed by BLI (p < 0.001) and CCK-8 assays (p < 0.001). Furthermore, in normal healthy cells, even after 24 h of co-culture, NK-92 Exo did not exhibit significant side effects. In the in vivo experiments, tumors in the vehicle control group were significantly increased, compared with those in the NK-92 Exo-treated group (p < 0.05). Conclusion: The results of the current study suggest that exosomes derived from NK cells exert cytotoxic effects on melanoma cells and thus warrant further development as a potential immunotherapeutic strategy for cancer.
Collapse
|
19
|
Luo C, Shen J. Research progress in advanced melanoma. Cancer Lett 2017; 397:120-126. [DOI: 10.1016/j.canlet.2017.03.037] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2017] [Revised: 03/27/2017] [Accepted: 03/28/2017] [Indexed: 12/12/2022]
|
20
|
Melanocytic nevi and melanoma: unraveling a complex relationship. Oncogene 2017; 36:5771-5792. [PMID: 28604751 DOI: 10.1038/onc.2017.189] [Citation(s) in RCA: 120] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 05/09/2017] [Accepted: 05/10/2017] [Indexed: 12/11/2022]
Abstract
Approximately 33% of melanomas are derived directly from benign, melanocytic nevi. Despite this, the vast majority of melanocytic nevi, which typically form as a result of BRAFV600E-activating mutations, will never progress to melanoma. Herein, we synthesize basic scientific insights and data from mouse models with common observations from clinical practice to comprehensively review melanocytic nevus biology. In particular, we focus on the mechanisms by which growth arrest is established after BRAFV600E mutation. Means by which growth arrest can be overcome and how melanocytic nevi relate to melanoma are also considered. Finally, we present a new conceptual paradigm for understanding the growth arrest of melanocytic nevi in vivo termed stable clonal expansion. This review builds upon the canonical hypothesis of oncogene-induced senescence in growth arrest and tumor suppression in melanocytic nevi and melanoma.
Collapse
|
21
|
Maiques O, Macià A, Moreno S, Barceló C, Santacana M, Vea A, Herreros J, Gatius S, Ortega E, Valls J, Chen BJ, Llobet-Navas D, Matias-Guiu X, Cantí C, Marti RM. Immunohistochemical analysis of T-type calcium channels in acquired melanocytic naevi and melanoma. Br J Dermatol 2017; 176:1247-1258. [PMID: 27718503 DOI: 10.1111/bjd.15121] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/26/2016] [Indexed: 11/29/2022]
Abstract
BACKGROUND Cutaneous malignant melanoma arises from transformed melanocytes de novo or from congenital or acquired melanocytic naevi. We have recently reported that T-type Ca2+ channels (TT-Cs) are upregulated in human melanoma and play an important role in cell proliferation. OBJECTIVES To describe for the first time in formalin-fixed paraffin-embedded tissue the immunoexpression of TT-Cs in biopsies of normal skin, acquired melanocytic naevi and melanoma, in order to evaluate their role in melanomagenesis and/or tumour progression, their utility as prognostic markers and their possible use in targeted therapies. METHODS Tissue samples from normal skin, melanocytic naevi and melanoma were subjected to immunohistochemistry for two TT-Cs (Cav3.1, Cav3.2); markers of proliferation (Ki67), the cell cycle (cyclin D1), hypoxia (Glut1), vascularization (CD31) and autophagy (LC3); BRAF V600E mutation (VE1) and phosphatase and tensin homologue (PTEN). Immunostaining was evaluated by histoscore. In silico analysis was used to assess the prognostic value of TT-C overexpression. RESULTS TT-C immunoexpression increased gradually from normal skin to common naevi, dysplastic naevi and melanoma samples, but with differences in the distribution of both isoforms. Particularly, Cav3.2 expression was significantly higher in metastatic melanoma than in primary melanoma. Statistical correlation showed a linear interaction between PTEN loss/BRAF V600E/Cav3.1/LC3/ Ki67/cyclin D1/Cav3.2/Glut1. Disease-free survival (DFS) and overall survival correlated inversely with overexpression of Cav3.2. DFS also correlated inversely with overexpression of Cav3.1. CONCLUSIONS TT-C immunoexpression on melanocytic neoplasms is consistent with our previous in vitro studies and appears to be related to tumour progression. TT-C upregulation can be considered as a prognostic marker using The Cancer Genome Atlas database. The high expression of Cav3.2 in metastatic melanoma encourages the investigation of the use of TT-C blockers in targeted therapies.
Collapse
Affiliation(s)
- O Maiques
- University of Lleida, IRBLleida, Lleida, Spain
| | - A Macià
- University of Lleida, IRBLleida, Lleida, Spain
| | - S Moreno
- Department of Dermatology, Hospital Universitari Arnau de Vilanova; University of Lleida, IRBLleida, Lleida, Spain
| | - C Barceló
- University of Lleida, IRBLleida, Lleida, Spain
| | - M Santacana
- Department of Pathology and Molecular Genetics, Hospital Universitari Arnau de Vilanova; University of Lleida, IRBLleida, Lleida, Spain
| | - A Vea
- Department of Dermatology, Hospital Universitari Arnau de Vilanova; University of Lleida, IRBLleida, Lleida, Spain
| | - J Herreros
- University of Lleida, IRBLleida, Lleida, Spain
| | - S Gatius
- Department of Pathology and Molecular Genetics, Hospital Universitari Arnau de Vilanova; University of Lleida, IRBLleida, Lleida, Spain
| | - E Ortega
- Department of Oncology, Hospital Universitari Arnau de Vilanova; University of Lleida, IRBLleida, Lleida, Spain
| | - J Valls
- Biostatistics Unit, IRBLleida, Lleida, Spain
| | - B J Chen
- New York Genome Center, New York, NY, U.S.A
| | - D Llobet-Navas
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, U.K
| | - X Matias-Guiu
- Department of Pathology and Molecular Genetics, Hospital Universitari Arnau de Vilanova; University of Lleida, IRBLleida, Lleida, Spain
| | - C Cantí
- University of Lleida, IRBLleida, Lleida, Spain
| | - R M Marti
- Department of Dermatology, Hospital Universitari Arnau de Vilanova; University of Lleida, IRBLleida, Lleida, Spain
| |
Collapse
|
22
|
Furue M, Kadono T. Melanoma therapy: Check the checkpoints. J Dermatol 2017; 43:121-4. [PMID: 26813076 DOI: 10.1111/1346-8138.13257] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2015] [Accepted: 11/10/2015] [Indexed: 11/27/2022]
Abstract
Recent mutational and translational studies have revealed that the Ras/Raf/mitogen-activated protein kinase kinase (MEK)/extracellular signal-regulated kinase (ERK) pathway plays a key role in melanomagenesis. Mutations in NRAS and BRAF are found in the majority of melanomas resulting in the formation of constitutively active NRAS and BRAF molecules, which leads to the proliferation and survival of melanoma cells through the activation of MEK/ERK signals. Inhibitors of BRAF or MEK significantly extend the progression-free survival and overall survival of melanoma patients compared with conventional chemotherapies. Combining BRAF and MEK inhibitors further enhances the clinical effectiveness. Cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) is an immune checkpoint molecule that downregulates T-cell activation by binding to B7 (CD80/CD86) molecules on antigen-presenting cells. Programmed death receptor ligand 1 on melanoma cells negatively regulates T-cell function by binding to the programmed death-1 (PD-1) receptor on T cells. Antibodies against CTLA-4 and PD-1 also enhance the survival of melanoma patients. In this review, we summarize the clinical effectiveness and adverse events of the BRAF inhibitors, MEK inhibitors and anti-immune checkpoint antibodies in melanoma treatment.
Collapse
Affiliation(s)
- Masutaka Furue
- Department of Dermatology, Kyushu University, Fukuoka, Japan
| | - Takafumi Kadono
- Department of Dermatology, St Marianna University School of Medicine, Kanagawa, Japan
| |
Collapse
|
23
|
Brasanac D, Stojkovic-Filipovic J, Bosic M, Tomanovic N, Manojlovic-Gacic E. Expression of G1/S-cyclins and cyclin-dependent kinase inhibitors in actinic keratosis and squamous cell carcinoma. J Cutan Pathol 2015; 43:200-10. [PMID: 26349899 DOI: 10.1111/cup.12623] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2015] [Revised: 08/09/2015] [Accepted: 09/07/2015] [Indexed: 01/28/2023]
Abstract
BACKGROUND Actinic keratosis (AK) and Bowen's disease (squamous cell carcinoma in situ, SCCIS) are pre-invasive stages in the development of squamous cell carcinoma (SCC). METHODS Immunohistochemical study of cyclin D1, cyclin E, p16(INK4a) and p21(Cip1) (/Waf1) in AK (53 cases), SCCIS (16 cases) and SCC (40 cases), in relation to the type of the lesion and SCC prognostic parameters (grade, diameter and thickness). RESULTS Diffuse cyclin D1 distribution was more frequent in SCCIS and SCC than in AK (p = 0.03) and similar pattern was observed for p16(INK4a) . For cyclin E, central distribution dominated in SCC compared with the AK (p = 0.001) and SCCIS (p = 0.03). p21(Cip1) (/Waf1) displayed suprabasal distribution more frequently in AK than in SCCIS (p = 0.001) and SCC (p = 0.0004). Semiquantitative assessment showed more positive cells in AK (p = 0.04) and SCCIS (p = 0.04) than in SCC for cyclin E. SCC with diameter over 20 mm and those thicker than 6 mm revealed higher labeling index with p16(INK4a) and p21(Cip1) (/Waf1) , respectively. CONCLUSIONS Our results suggest different alterations for p16(INK4a) and p21(Cip1) (/Waf1) in AK, SCCIS and SCC. Immunostaining distribution showed closer correlation with the type of the lesion, whereas percentage of positive cells displayed better association with the SCC prognostic parameters.
Collapse
Affiliation(s)
- Dimitrije Brasanac
- Institute of Pathology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Jelena Stojkovic-Filipovic
- Clinic of Dermatovenereology, Clinical Center of Serbia, and Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Martina Bosic
- Institute of Pathology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Nada Tomanovic
- Institute of Pathology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | | |
Collapse
|
24
|
Ohi Y, Kosuge H, Tanese K. Case of syringomatous carcinoma: Positive immunohistochemical staining of c-KIT and phosphorylated-extracellular signal-regulated kinase 1/2. J Dermatol 2015; 42:1191-2. [PMID: 26282337 DOI: 10.1111/1346-8138.13080] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Yumiko Ohi
- Department of Dermatology, Hino Municipal Hospital, Tokyo, Japan.,Department of Dermatology, Keio University School of Medicine, Tokyo, Japan
| | - Haruhiko Kosuge
- Department of Dermatology, Hino Municipal Hospital, Tokyo, Japan
| | - Keiji Tanese
- Department of Dermatology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
25
|
Coleman DJ, Chagani S, Hyter S, Sherman AM, Löhr CV, Liang X, Ganguli-Indra G, Indra AK. Loss of keratinocytic RXRα combined with activated CDK4 or oncogenic NRAS generates UVB-induced melanomas via loss of p53 and PTEN in the tumor microenvironment. Mol Cancer Res 2014; 13:186-96. [PMID: 25189354 DOI: 10.1158/1541-7786.mcr-14-0164] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
UNLABELLED Understanding the molecular mechanisms behind formation of melanoma, the deadliest form of skin cancer, is crucial for improved diagnosis and treatment. One key is to better understand the cross-talk between epidermal keratinocytes and pigment-producing melanocytes. Here, using a bigenic mouse model system combining mutant oncogenic NRAS(Q61K) (constitutively active RAS) or mutant activated CDK4(R24C/R24C) (prevents binding of CDK4 by kinase inhibitor p16(INK4A)) with an epidermis-specific knockout of the nuclear retinoid X receptor alpha (RXRα(ep-/-)) results in increased melanoma formation after chronic ultraviolet-B (UVB) irradiation compared with control mice with functional RXRα. Melanomas from both groups of bigenic RXRα(ep-/-) mice are larger in size with higher proliferative capacity, and exhibit enhanced angiogenic properties and increased expression of malignant melanoma markers. Analysis of tumor adjacent normal skin from these mice revealed altered expression of several biomarkers indicative of enhanced melanoma susceptibility, including reduced expression of tumor suppressor p53 and loss of PTEN, with concomitant increase in activated AKT. Loss of epidermal RXRα in combination with UVB significantly enhances invasion of melanocytic cells to draining lymph nodes in bigenic mice expressing oncogenic NRAS(Q61K) compared with controls with functional RXRα. These results suggest a crucial role of keratinocytic RXRα to suppress formation of UVB-induced melanomas and their progression to malignant cancers in the context of driver mutations such as activated CDK4(R24C/R24C) or oncogenic NRAS(Q61K). IMPLICATIONS These findings suggest that RXRα may serve as a clinical diagnostic marker and therapeutic target in melanoma progression and metastasis.
Collapse
Affiliation(s)
- Daniel J Coleman
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, Oregon. Molecular and Cellular Biology Program, Oregon State University, Corvallis, Oregon
| | - Sharmeen Chagani
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, Oregon. Molecular and Cellular Biology Program, Oregon State University, Corvallis, Oregon
| | - Stephen Hyter
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, Oregon. Molecular and Cellular Biology Program, Oregon State University, Corvallis, Oregon
| | - Anna M Sherman
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, Oregon. BioResource Research Program, College of Agricultural Sciences, Oregon State University, Corvallis, Oregon
| | - Christiane V Löhr
- College of Veterinary Medicine, Oregon State University, Corvallis, Oregon
| | - Xiaobo Liang
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, Oregon
| | - Gitali Ganguli-Indra
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, Oregon. Molecular and Cellular Biology Program, Oregon State University, Corvallis, Oregon
| | - Arup K Indra
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, Oregon. Molecular and Cellular Biology Program, Oregon State University, Corvallis, Oregon. Environmental Health Science Center, Oregon State University, Corvallis, Oregon. Department of Dermatology, Oregon Health and Science University, Portland, Oregon.
| |
Collapse
|
26
|
Abstract
From 1976 to 2010, only 2 medications were approved for treating metastatic melanoma. Between 2011 and 2013, 4 agents were approved and other therapies have shown great promise in clinical trials. Fundamental discoveries, such as the identification of oncogenic mutations in most melanomas, the elucidation of the molecular signaling resulting from these mutations, and the revelation that several cell surface molecules serve as regulators of immune activation, have been instrumental in this progress. This article summarizes the molecular pathogenesis of melanoma, describes the current efforts to target oncogene-driven signaling, and presents the rationale for combining immune and molecular targeting.
Collapse
Affiliation(s)
- Ryan J Sullivan
- Center for Melanoma, Massachusetts General Hospital Cancer Center, Harvard Medical School, 55 Fruit Street, Boston, MA 02114, USA
| | - David E Fisher
- Department of Dermatology, Massachusetts General Hospital Cancer Center, Harvard Medical School, Bartlett 6, 55 Fruit Street, Boston, MA 02114, USA.
| |
Collapse
|
27
|
Barysch MJ, Levesque MP, Cheng P, Karpova MB, Mihic-Probst D, Civenni G, Shakhova O, Sommer L, Biedermann T, Schiestl C, Dummer R. Coexpression of SOX10/CD271 (p75(NTR)) and β-Galactosidase in Large to Giant Congenital Melanocytic Nevi of Pediatric Patients. Dermatopathology (Basel) 2014; 1:35-46. [PMID: 27047921 PMCID: PMC4772933 DOI: 10.1159/000362490] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Background Congenital melanocytic nevi (CMNs) are melanocytic neoplasms that can transform into melanoma. However, this development is impeded in the majority of cases and mostly affects patients with large or giant CMNs. Methods To elucidate mechanisms that keep CMNs from malignant transformation, CMN tissue biopsies were investigated for p-ERK and senescence markers by immunohistochemistry and for SOX10/CD271 (p75NTR) by immunofluorescence. CMN cells were cultivated, and MTT assays were performed for evaluating cell viability. Mutation status for NRAS and BRAF was performed by real-time PCR. Results 13 CMNs (from patients aged 0.5-11.8 years, mean: 2.7) showed immunoreactivity for SOX10/CD271 (p75NTR) in 34.2%. p-ERK was immunoreactive in 80% (4/5); β-galactosidase was significantly stronger expressed in CMNs compared to melanocytic nevi of patients over 70 years (p = 0.0085). The 5 CMN cultures were immunoreactive for SOX10/CD271 (p75NTR) in 36.7%. By silencing SOX10 by siRNA in 2 CMN cell cultures, cell viability decreased significantly. NRASQ61K mutation was found in 91.7% (11/12) and BRAFV600E in 6.3% of all analyzable CMNs (1/16). Conclusions Oncogene-induced senescence might prevent malignant transformation through activation of the mitogen-activated protein kinase pathway. SOX10 is necessary for the viability of human CMN cell cultures and may be responsible for clinical changes during aging.
Collapse
Affiliation(s)
- Marjam J Barysch
- Department of Dermatology, University Hospital Zurich, Switzerland
| | | | - Phil Cheng
- Department of Dermatology, University Hospital Zurich, Switzerland
| | - Maria B Karpova
- Department of Dermatology, University Hospital Zurich, Switzerland
| | | | - Gianluca Civenni
- Laboratories of Experimental Oncology, Department of Anatomy, Institute of Oncology Research, Bellinzona, Switzerland
| | - Olga Shakhova
- Institute of Anatomy, University Zurich, Switzerland
| | - Lukas Sommer
- Institute of Anatomy, University Zurich, Switzerland
| | - Thomas Biedermann
- Pediatric Burn Centre, Plastic and Reconstructive Surgery, Department of Surgery, University Children's Hospital Zurich, Switzerland; Children's Research Center, Zurich, Department of Anatomy, Institute of Oncology Research, Bellinzona, Switzerland
| | - Clemens Schiestl
- Pediatric Burn Centre, Plastic and Reconstructive Surgery, Department of Surgery, University Children's Hospital Zurich, Switzerland; Children's Research Center, Zurich, Department of Anatomy, Institute of Oncology Research, Bellinzona, Switzerland
| | - Reinhard Dummer
- Department of Dermatology, University Hospital Zurich, Switzerland
| |
Collapse
|
28
|
Rapozzi V, Zorzet S, Zacchigna M, Della Pietra E, Cogoi S, Xodo LE. Anticancer activity of cationic porphyrins in melanoma tumour-bearing mice and mechanistic in vitro studies. Mol Cancer 2014; 13:75. [PMID: 24684778 PMCID: PMC4021972 DOI: 10.1186/1476-4598-13-75] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Accepted: 03/20/2014] [Indexed: 01/07/2023] Open
Abstract
Background Porphyrin TMPyP4 (P4) and its C14H28-alkyl derivative (C14) are G-quadruplex binders and singlet oxygen (1O2) generators. In contrast, TMPyP2 (P2) produces 1O2 but it is not a G-quadruplex binder. As their photosensitizing activity is currently undefined, we report in this study their efficacy against a melanoma skin tumour and describe an in vitro mechanistic study which gives insights into their anticancer activity. Methods Uptake and antiproliferative activity of photoactivated P2, P4 and C14 have been investigated in murine melanoma B78-H1 cells by FACS, clonogenic and migration assays. Apoptosis was investigated by PARP-1 cleavage and annexin-propidium iodide assays. Biodistribution and in vivo anticancer activity were tested in melanoma tumour-bearing mice. Porphyrin binding and photocleavage of G-rich mRNA regions were investigated by electrophoresis and RT-PCR. Porphyrin effect on ERK pathway was explored by Western blots. Results Thanks to its higher lipophylicity C14 was taken up by murine melanoma B78-H1 cells up to 30-fold more efficiently than P4. When photoactivated (7.2 J/cm2) in B78-H1 melanoma cells, P4 and C14, but not control P2, caused a strong inhibition of metabolic activity, clonogenic growth and cell migration. Biodistribution studies on melanoma tumour-bearing mice showed that P4 and C14 localize in the tumour. Upon irradiation (660 nm, 193 J/cm2), P4 and C14 retarded tumour growth and increased the median survival time of the treated mice by ~50% (P <0.01 by ANOVA), whereas porphyrin P2 did not. The light-dependent mechanism mediated by P4 and C14 is likely due to the binding to and photocleavage of G-rich quadruplex-forming sequences within the 5′-untranslated regions of the mitogenic ras genes. This causes a decrease of RAS protein and inhibition of downstream ERK pathway, which stimulates proliferation. Annexin V/propidium iodide and PARP-1 cleavage assays showed that the porphyrins arrested tumour growth by apoptosis and necrosis. C14 also showed an intrinsic light-independent anticancer activity, as recently reported for G4-RNA binders. Conclusions Porphyrins P4 and C14 impair the clonogenic growth and migration of B78-H1 melanoma cells and inhibit melanoma tumour growth in vivo. Evidence is provided that C14 acts through light-dependent (mRNA photocleavage) and light-independent (translation inhibition) mechanisms.
Collapse
Affiliation(s)
| | | | | | | | | | - Luigi E Xodo
- Department of Medical and Biological Sciences, School of Medicine, P,le Kolbe 4, 33100 Udine, Italy.
| |
Collapse
|
29
|
Lade-Keller J, Riber-Hansen R, Guldberg P, Schmidt H, Hamilton-Dutoit SJ, Steiniche T. Immunohistochemical analysis of molecular drivers in melanoma identifies p16 as an independent prognostic biomarker. J Clin Pathol 2014; 67:520-8. [DOI: 10.1136/jclinpath-2013-202127] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
30
|
Lopez-Bergami P. The role of mitogen- and stress-activated protein kinase pathways in melanoma. Pigment Cell Melanoma Res 2014; 24:902-21. [PMID: 21914141 DOI: 10.1111/j.1755-148x.2011.00908.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Recent discoveries have increased our comprehension of the molecular signaling events critical for melanoma development and progression. Many oncogenes driving melanoma have been identified, and most of them exert their oncogenic effects through the activation of the RAF/MEK/ERK mitogen-activated protein kinase (MAPK) pathway. The c-Jun N-terminal kinase (JNK) and p38 MAPK pathways are also important in melanoma, but their precise role is not clear yet. This review summarizes our current knowledge on the role of the three main MAPK pathways, extracellular regulated kinase (ERK), JNK, and p38, and their impact on melanoma biology. Although the results obtained with BRAF inhibitors in melanoma patients are impressive, several mechanisms of acquired resistance have emerged. To overcome this obstacle constitutes the new challenge in melanoma therapy. Given the major role that MAPKs play in melanoma, understanding their functions and the interconnection among them and with other signaling pathways represents a step forward toward this goal.
Collapse
Affiliation(s)
- Pablo Lopez-Bergami
- Instituto de Medicina y Biología Experimental, CONICET, Buenos Aires, Argentina.
| |
Collapse
|
31
|
Simpson RM, Bastian BC, Michael HT, Webster JD, Prasad ML, Conway CM, Prieto VM, Gary JM, Goldschmidt MH, Esplin DG, Smedley RC, Piris A, Meuten DJ, Kiupel M, Lee CCR, Ward JM, Dwyer JE, Davis BJ, Anver MR, Molinolo AA, Hoover SB, Rodriguez-Canales J, Hewitt SM. Sporadic naturally occurring melanoma in dogs as a preclinical model for human melanoma. Pigment Cell Melanoma Res 2014; 27:37-47. [PMID: 24128326 PMCID: PMC4066658 DOI: 10.1111/pcmr.12185] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Revised: 10/11/2013] [Accepted: 10/11/2013] [Indexed: 12/20/2022]
Abstract
Melanoma represents a significant malignancy in humans and dogs. Different from genetically engineered models, sporadic canine melanocytic neoplasms share several characteristics with human disease that could make dogs a more relevant preclinical model. Canine melanomas rarely arise in sun-exposed sites. Most occur in the oral cavity, with a subset having intra-epithelial malignant melanocytes mimicking the in situ component of human mucosal melanoma. The spectrum of canine melanocytic neoplasia includes benign lesions with some analogy to nevi, as well as invasive primary melanoma, and widespread metastasis. Growing evidence of distinct subtypes in humans, differing in somatic and predisposing germ-line genetic alterations, cell of origin, epidemiology, relationship to ultraviolet radiation and progression from benign to malignant tumors, may also exist in dogs. Canine and human mucosal melanomas appear to harbor BRAF, NRAS, and c-kit mutations uncommonly, compared with human cutaneous melanomas, although both species share AKT and MAPK signaling activation. We conclude that there is significant overlap in the clinical and histopathological features of canine and human mucosal melanomas. This represents opportunity to explore canine oral cavity melanoma as a preclinical model.
Collapse
Affiliation(s)
- R Mark Simpson
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Evans MS, Madhunapantula SV, Robertson GP, Drabick JJ. Current and future trials of targeted therapies in cutaneous melanoma. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 779:223-55. [PMID: 23288642 DOI: 10.1007/978-1-4614-6176-0_10] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
In order to effectively treat melanoma, targeted inhibition of key m-echanistic events regulating melanoma development such as cell proliferation, survival, angiogenesis and invasion or metastasis needs to be accomplished. The Mitogen Activated Protein Kinase (MAPK) pathway has been identified as a key player in melanoma development making this cascade an important therapeutic target. However, identification of the ideal pathway member to therapeutically target for maximal clinical benefit remains a challenge. In normal cells, the MAPK pathway relays extracellular signals from the cell membrane to the nucleus via a cascade of phosphorylation events, which promote cancer development. Dysregulation of the MAPK pathway occurs frequently in many human cancers including melanoma. Mutations in the B-RAF and RAS genes, genetic or epigenetic modifications are the key aberrations observed in this signaling cascade. Constitutive activation of this pathway causes oncogenic transformation of cells by promoting cell proliferation, invasion, metastasis, migration, survival and angiogenesis. This review provides an overview of (a) key members of MAPK signaling regulating melanoma development; (b) key proteins which can serve as biomarkers to assess disease progression; (c) the clinical efficacy of various pharmacological agents targeting MAPK pathway; (d) current clinical trials evaluating downstream targets of the MAPK pathway; (e) issues associated with pharmacological agents such as drug resistance, induction of cancers; and finally (e) various strategies overcoming drug resistance.
Collapse
Affiliation(s)
- Matthew S Evans
- Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA.
| | | | | | | |
Collapse
|
33
|
Vízkeleti L, Ecsedi S, Rákosy Z, Orosz A, Lázár V, Emri G, Koroknai V, Kiss T, Ádány R, Balázs M. The role of CCND1 alterations during the progression of cutaneous malignant melanoma. Tumour Biol 2012; 33:2189-99. [PMID: 23001925 DOI: 10.1007/s13277-012-0480-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2012] [Accepted: 08/01/2012] [Indexed: 11/29/2022] Open
Abstract
It is well demonstrated that CCND1 amplification is a frequent event in the acral subtype of cutaneous malignant melanoma; however, its role in the other subtypes of the disease is still controversial. The objectives of this study were to evaluate genetic and expression alterations of CCND1 with a focus on primary cutaneous melanomas, to define BRAF and NRAS mutation status, and correlate the data with clinical-pathological parameters. CCND1 amplification was associated with ulceration and the localization of the metastasis. After correction for the mutation state of BRAF and NRAS genes, CCND1 amplification in samples without such mutations was associated with ulceration and sun exposure. The cyclin D1 (CCND1) mRNA level decreased in lesions with multiple metastases and was correlated with both the mRNA levels and mutation state of BRAF and NRAS genes. Primary melanomas with BRAF(V600) or NRAS(Q61 ) mutations exhibited lower CCND1 mRNA level. CCND1 protein expression was associated with Breslow thickness, metastasis formation, and shorter survival time. These observations suggest that CCND1 alterations are linked to melanoma progression and are modified by BRAF and NRAS mutations. Our data show that CCND1 amplification could have a prognostic relevance in cutaneous melanoma and highlight that altered CCND1 gene expression may influence the metastatic progression, survival, and the localization of metastases.
Collapse
Affiliation(s)
- Laura Vízkeleti
- Department of Preventive Medicine, Faculty of Public Health, Medical and Health Science Center, University of Debrecen, Debrecen, Hungary
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
|