1
|
Huang C, Lau TWS, Smoller BR. Diagnosing Cutaneous Melanocytic Tumors in the Molecular Era: Updates and Review of Literature. Dermatopathology (Basel) 2024; 11:26-51. [PMID: 38247727 PMCID: PMC10801542 DOI: 10.3390/dermatopathology11010005] [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: 10/31/2023] [Revised: 01/08/2024] [Accepted: 01/09/2024] [Indexed: 01/23/2024] Open
Abstract
Over the past decade, molecular and genomic discoveries have experienced unprecedented growth, fundamentally reshaping our comprehension of melanocytic tumors. This review comprises three main sections. The first part gives an overview of the current genomic landscape of cutaneous melanocytic tumors. The second part provides an update on the associated molecular tests and immunohistochemical stains that are helpful for diagnostic purposes. The third section briefly outlines the diverse molecular pathways now utilized for the classification of cutaneous melanomas. The primary goal of this review is to provide a succinct overview of the molecular pathways involved in melanocytic tumors and demonstrate their practical integration into the realm of diagnostic aids. As the molecular and genomic knowledge base continues to expand, this review hopes to serve as a valuable resource for healthcare professionals, offering insight into the evolving molecular landscape of cutaneous melanocytic tumors and its implications for patient care.
Collapse
Affiliation(s)
- Chelsea Huang
- Department of Pathology, Loma Linda University Medical Center, Loma Linda, CA 92354, USA
| | | | - Bruce R. Smoller
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA;
| |
Collapse
|
2
|
Silverman RB. Inactivators of Ornithine Aminotransferase for the Treatment of Hepatocellular Carcinoma. ACS Med Chem Lett 2021; 13:38-49. [PMID: 35059122 PMCID: PMC8762738 DOI: 10.1021/acsmedchemlett.1c00526] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 11/22/2021] [Indexed: 01/16/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the second or third leading cause of cancer mortality worldwide (depending on which statistics are used), yet there is no effective treatment. Currently, there are nine FDA-approved drugs for HCC, five monoclonal antibodies and four tyrosine kinase inhibitors. Ornithine aminotransferase (OAT) has been validated as a target in preclinical studies, which demonstrates that it is a potential target to treat HCC. Currently, there are no OAT inactivators in clinical trials for HCC. This Innovation describes evidence to support inhibition of OAT as a novel approach for HCC tumor growth inhibition. After the mechanism of OAT is discussed, the origins of our involvement in OAT inactivation, based on our previous work on mechanism-based inactivation of GABA-AT, are described. Once it was demonstrated that OAT inactivation does lead to HCC tumor growth inhibition, new selective OAT inactivators were designed and their inactivation mechanisms were elucidated. A summary of these mechanistic studies is presented. Inactivators of OAT provide the potential for treatment of HCC, targeting the Wnt/β-catenin pathway.
Collapse
|
3
|
Del Bosque-Plata L, Hernández-Cortés EP, Gragnoli C. The broad pathogenetic role of TCF7L2 in human diseases beyond type 2 diabetes. J Cell Physiol 2021; 237:301-312. [PMID: 34612510 PMCID: PMC9292842 DOI: 10.1002/jcp.30581] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 08/16/2021] [Accepted: 08/18/2021] [Indexed: 12/20/2022]
Abstract
The TCF7L2 protein is a key transcriptional effector of the Wnt/β‐catenin signaling pathway, regulating gene expression. It was initially identified in cancer research and embryologic developmental studies. Later, the TCF7L2 gene was linked to type 2 diabetes (T2D), implicating TCF7L2 and Wnt‐signaling in metabolic disorders and homeostasis. In fact, TCF7L2‐T2D variants confer the greatest relative risk for T2D, unquestionably predicting conversion to T2D in individuals with impaired glucose tolerance. We aim to describe the relevance of TCF7L2 in other human disorders. The TCF7L2‐single nucleotide polymorphisms (SNPs) and T2D‐risk association have been replicated in numerous follow‐up studies, and research has now been performed in several other diseases. In this article, we discuss common TCF7L2‐T2D variants within the framework of their association with human diseases. The TCF7L2 functional regions need to be further investigated because the molecular and cellular mechanisms through which TCF7L2 contributes to risk associations with different diseases are still not fully elucidated. In this review, we show the association of common TCF7L2‐T2D variants with many types of diseases. However, the role of rare genetic variations in the TCF7L2 gene in distinct diseases and ethnic groups has not been explored, and understanding their impact on specific phenotypes will be of clinical relevance. This offers an excellent opportunity to gain a clearer picture of the role that the TCF7L2 gene plays in the pathophysiology of human diseases. The potential pleiotropic role of TCF7L2 may underlie a possible pathway for comorbidity in human disorders.
Collapse
Affiliation(s)
- Laura Del Bosque-Plata
- Laboratorio de Nutrigenética y Nutrigenómica, Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| | | | - Claudia Gragnoli
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolic Disease, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania.,Division of Endocrinology, Creighton University School of Medicine, Omaha, Nebraska, USA.,Department of Public Health Sciences, Penn State College of Medicine, Hershey, Pennsylvania, USA.,Molecular Biology Laboratory, Bios Biotech Multi-Diagnostic Health Center, Rome, Italy
| |
Collapse
|
4
|
Kahn M. Taking the road less traveled - the therapeutic potential of CBP/β-catenin antagonists. Expert Opin Ther Targets 2021; 25:701-719. [PMID: 34633266 PMCID: PMC8745629 DOI: 10.1080/14728222.2021.1992386] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 10/08/2021] [Indexed: 10/20/2022]
Abstract
AREAS COVERED This perspective discusses the challenges of targeting the Wnt signaling cascade, the safety, efficacy, and therapeutic potential of specific CBP/β-catenin antagonists and a rationale for the pleiotropic effects of CBP/β-catenin antagonists beyond Wnt signaling. EXPERT OPINION CBP/β-catenin antagonists can correct lineage infidelity, enhance wound healing, both normal and aberrant (e.g. fibrosis) and force the differentiation and lineage commitment of stem cells and cancer stem cells by regulating enhancer and super-enhancer coactivator occupancy. Small molecule CBP/β-catenin antagonists rebalance the equilibrium between CBP/β-catenin versus p300/β-catenin dependent transcription and may be able to treat or prevent many diseases of aging, via maintenance of our somatic stem cell pool, and regulating mitochondrial function and metabolism involved in differentiation and immune cell function.
Collapse
Affiliation(s)
- Michael Kahn
- Department of Molecular Medicine, City of Hope, Beckman Research Institute, 1500 East Duarte Road Flower Building, Duarte, CA, USA
| |
Collapse
|
5
|
The role of Hypoxia-Inducible Factor-1alpha and its signaling in melanoma. Biomed Pharmacother 2021; 141:111873. [PMID: 34225012 DOI: 10.1016/j.biopha.2021.111873] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/25/2021] [Accepted: 06/28/2021] [Indexed: 12/11/2022] Open
Abstract
Adaptation to the loss of O2 is regulated via the activity of hypoxia-inducible factors such as Hypoxia-Inducible Factor-1 (HIF-1). HIF-1 acts as a main transcriptional mediator in the tissue hypoxia response that regulates over 1000 genes related to low oxygen tension. The role of HIF-1α in oncogenic processes includes angiogenesis, tumor metabolism, cell proliferation, and metastasis, which has been examined in various malignancies, such as melanoma. Melanoma is accompanied by a high death rate and a cancer type whose incidence has risen over the last decades. The linkage between O2 loss and melanogenesis had extensively studied over decades. Recent studies revealed that HIF-1α contributes to melanoma progression via different signaling pathways such as PI3K/Akt/mTOR, RAS/RAF/MEK/ERK, JAK/STAT, Wnt/β-catenin, Notch, and NF-κB. Also, various microRNAs (miRs) are known to mediate the HIF-1α role in melanoma. Therefore, HIF-1α offers a diagnostic/prognostic biomarker and a candidate for targeted therapy in melanoma.
Collapse
|
6
|
Del Bosque-Plata L, Martínez-Martínez E, Espinoza-Camacho MÁ, Gragnoli C. The Role of TCF7L2 in Type 2 Diabetes. Diabetes 2021; 70:1220-1228. [PMID: 34016596 PMCID: PMC8275893 DOI: 10.2337/db20-0573] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 03/19/2021] [Indexed: 12/16/2022]
Abstract
TCF7L2 is the most potent locus for type 2 diabetes (T2D) risk and the first locus to have been robustly reported by genomic linkage studies. TCF7L2 is a transcription factor that forms a basic part of the Wnt signaling pathway. This gene has highly conserved sequence regions that correspond to functional domains. The association of TCF7L2 with T2D is one of the most powerful genetically discovered in studies of complex diseases, as it has been consistently replicated in multiple populations with diverse genetic origins. The mechanisms over which TCF7L2 exerts its effect on T2D are still not well understood. In this article, we describe the main molecular mechanisms of how TCF7L2 is related to T2D. TCF7L2 variants associated with T2D risk exert an influence on the initial therapeutic success of the hypoglycemic oral agent sulfonylurea. Thus, it is important to know whether there are other TCF7L2 variants associated with T2D that can influence treatment with oral hypoglycemic agents. Resequencing of the TCF7L2 gene in diverse ethnic groups is required to reveal common and rare variations and their role in different pathologies and in adverse reactions to drugs. Identification of TCF7L2-susceptibility disease variants will permit, at a given moment, offering of therapies to patients according to their genotype.
Collapse
Affiliation(s)
- Laura Del Bosque-Plata
- Laboratory of Nutrigenetics and Nutrigenomics, National Institute of Genomic Medicine, Mexico City, Mexico
| | - Eduardo Martínez-Martínez
- Laboratory of Cell Communication and Extracellular Vesicles, National Institute of Genomic Medicine, Mexico City, Mexico
| | | | - Claudia Gragnoli
- Division of Endocrinology, Department of Medicine, School of Medicine, Creighton University, Omaha, NE
- Division of Endocrinology, Diabetes, and Metabolic Disease, Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, PA
- Molecular Biology Laboratory, Bios Biotech Multi-Diagnostic Health Center, Rome, Italy
| |
Collapse
|
7
|
Shami Shah A, Cao X, White AC, Baskin JM. PLEKHA4 Promotes Wnt/β-Catenin Signaling-Mediated G 1-S Transition and Proliferation in Melanoma. Cancer Res 2021; 81:2029-2043. [PMID: 33574086 PMCID: PMC8137570 DOI: 10.1158/0008-5472.can-20-2584] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 12/29/2020] [Accepted: 02/05/2021] [Indexed: 11/16/2022]
Abstract
Despite recent promising advances in targeted therapies and immunotherapies, patients with melanoma incur substantial mortality. In particular, inhibitors targeting BRAF-mutant melanoma can lead to resistance, and no targeted therapies exist for NRAS-mutant melanoma, motivating the search for additional therapeutic targets and vulnerable pathways. Here we identify a regulator of Wnt/β-catenin signaling, PLEKHA4, as a factor required for melanoma proliferation and survival. PLEKHA4 knockdown in vitro decreased Dishevelled levels, attenuated Wnt/β-catenin signaling, and blocked progression through the G1-S cell-cycle transition. In mouse xenograft and allograft models, inducible PLEKHA4 knockdown attenuated tumor growth in BRAF- and NRAS-mutant melanomas and exhibited an additive effect with the clinically used inhibitor encorafenib in a BRAF-mutant model. As an E3 ubiquitin ligase regulator with both lipid- and protein-binding partners, PLEKHA4 presents several opportunities for targeting with small molecules. Our work identifies PLEKHA4 as a promising drug target for melanoma and clarifies a controversial role for Wnt/β-catenin signaling in the control of melanoma proliferation. SIGNIFICANCE: This study establishes that melanoma cell proliferation requires the protein PLEKHA4 to promote pathologic Wnt signaling for proliferation, highlighting PLEKHA4 inhibition as a new avenue for the development of targeted therapies.
Collapse
Affiliation(s)
- Adnan Shami Shah
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, New York
| | - Xiaofu Cao
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, New York
| | - Andrew C White
- Department of Biomedical Sciences, Cornell University, Ithaca, New York
| | - Jeremy M Baskin
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York.
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, New York
| |
Collapse
|
8
|
Sayedyahossein S, Huang K, Li Z, Zhang C, Kozlov AM, Johnston D, Nouri-Nejad D, Dagnino L, Betts DH, Sacks DB, Penuela S. Pannexin 1 binds β-catenin to modulate melanoma cell growth and metabolism. J Biol Chem 2021; 296:100478. [PMID: 33647315 PMCID: PMC8027267 DOI: 10.1016/j.jbc.2021.100478] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 02/11/2021] [Accepted: 02/24/2021] [Indexed: 01/05/2023] Open
Abstract
Melanoma is the most aggressive skin malignancy with increasing incidence worldwide. Pannexin1 (PANX1), a member of the pannexin family of channel-forming glycoproteins, regulates cellular processes in melanoma cells including proliferation, migration, and invasion/metastasis. However, the mechanisms responsible for coordinating and regulating PANX1 function remain unclear. Here, we demonstrated a direct interaction between the C-terminal region of PANX1 and the N-terminal portion of β-catenin, a key transcription factor in the Wnt pathway. At the protein level, β-catenin was significantly decreased when PANX1 was either knocked down or inhibited by two PANX1 blockers, Probenecid and Spironolactone. Immunofluorescence imaging showed a disrupted pattern of β-catenin localization at the cell membrane in PANX1-deficient cells, and transcription of several Wnt target genes, including MITF, was suppressed. In addition, a mitochondrial stress test revealed that the metabolism of PANX1-deficient cells was impaired, indicating a role for PANX1 in the regulation of the melanoma cell metabolic profile. Taken together, our data show that PANX1 directly interacts with β-catenin to modulate growth and metabolism in melanoma cells. These findings provide mechanistic insight into PANX1-mediated melanoma progression and may be applicable to other contexts where PANX1 and β-catenin interact as a potential new component of the Wnt signaling pathway.
Collapse
Affiliation(s)
- Samar Sayedyahossein
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Kenneth Huang
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Zhigang Li
- Department of Laboratory Medicine, National Institutes of Health, Bethesda, Maryland, USA
| | - Christopher Zhang
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Alexandra M Kozlov
- Department of Biology, Faculty of Science, University of Western Ontario, London, Ontario, Canada
| | - Danielle Johnston
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Daniel Nouri-Nejad
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Lina Dagnino
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentristry, University of Western Ontario, London, Ontario, Canada; Division of Experimental Oncology, Department of Oncology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Dean H Betts
- Department of Biology, Faculty of Science, University of Western Ontario, London, Ontario, Canada; Department of Physiology and Pharmacology, Schulich School of Medicine and Dentristry, University of Western Ontario, London, Ontario, Canada
| | - David B Sacks
- Department of Laboratory Medicine, National Institutes of Health, Bethesda, Maryland, USA
| | - Silvia Penuela
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada; Division of Experimental Oncology, Department of Oncology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada.
| |
Collapse
|
9
|
Wang R, Cai J, Xie S, Zhao C, Wang Y, Cao D, Li G. T Cell Factor 4 Is Involved in Papillary Thyroid Carcinoma via Regulating Long Non-Coding RNA HCP5. Technol Cancer Res Treat 2020; 19:1533033820983290. [PMID: 33371788 PMCID: PMC7780308 DOI: 10.1177/1533033820983290] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The annual incidence of papillary thyroid carcinoma has increased dramatically. T cell factor 4 (TCF4) is an important component of Wnt signaling pathway.However, the role of TCF4 in PTC remains unknown. In this study, TCF4 was observed to overexpress in PTC patients and cells by qRT-PCR assay. The colony formation assay, Edu staining and transwell assay indicated thatoverexpression of TCF4 promoted cell proliferation and invasion of TCP-1 cells, whereas knockdown of TCF4 inhibited cell proliferation and invasion of IHH-4 cells. To investigate the mechanism of TCF4 in PTC cells, the luciferase assay demonstrated that TCF4 could modulate HCP5 expression. Besides, GLuc-ON promoter reporter assayproved that TCF4 could bind to HCP5 promoter. Further, knockdown of HCP5 could significantly up-regulated miR-15a, miR-216a-5p, miR-22-3p, miR-139-5p, miR-203, miR-27a-3p and miR-320, and down-regulated miR-186-5p in IHH-4 cells, which might be potential downstream of TFC4/HCP5 axis. In conclusion, up-regulation TCF4 can promote HCP5 expression via binding to HCP5 promoter. It may be the first time to prove that TCF4 regulates HCP5 in PTC, which provides a novel sight for treatment of PTC.
Collapse
Affiliation(s)
- Rui Wang
- Surgical Oncology, Hangzhou Cancer Hospital, Hangzhou City, Zhejiang Province, China
| | - Jidong Cai
- Surgical Oncology, Hangzhou Cancer Hospital, Hangzhou City, Zhejiang Province, China
| | - Shangnao Xie
- Surgical Oncology, Hangzhou Cancer Hospital, Hangzhou City, Zhejiang Province, China
| | - Chunlei Zhao
- Department of Nuclear Medicine, Hangzhou Cancer Hospital, Hangzhou City, Zhejiang Province, China
| | - Yi Wang
- Surgical Oncology, Hangzhou Cancer Hospital, Hangzhou City, Zhejiang Province, China
| | - Deming Cao
- Surgical Oncology, Hangzhou Cancer Hospital, Hangzhou City, Zhejiang Province, China
| | - Gang Li
- Surgical Oncology, Hangzhou Cancer Hospital, Hangzhou City, Zhejiang Province, China
| |
Collapse
|
10
|
Zhu GX, Gao D, Shao ZZ, Chen L, Ding WJ, Yu QF. Wnt/β‑catenin signaling: Causes and treatment targets of drug resistance in colorectal cancer (Review). Mol Med Rep 2020; 23:105. [PMID: 33300082 PMCID: PMC7723170 DOI: 10.3892/mmr.2020.11744] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 10/23/2020] [Indexed: 02/07/2023] Open
Abstract
Colorectal cancer (CRC) is the third most common malignant tumor in humans. Chemotherapy is used for the treatment of CRC. However, the effect of chemotherapy remains unsatisfactory due to drug resistance. Growing evidence has shown that the presence of highly metastatic tumor stem cells, regulation of non-coding RNAs and the tumor microenvironment contributes to drug resistance mechanisms in CRC. Wnt/β-catenin signaling mediates the chemoresistance of CRC in these three aspects. Therefore, the present study analyzed the abundant evidence of the contribution of Wnt/β-catenin signaling to the development of drug resistance in CRC and discussed its possible role in improving the chemosensitivity of CRC, which may provide guidelines for its clinical treatment.
Collapse
Affiliation(s)
- Gui-Xian Zhu
- Department of Gastroenterology and Hepatology, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Dian Gao
- Department of Pathogen Biology and Immunology, Medical College of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Zhao-Zhao Shao
- Department of Gastroenterology and Hepatology, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Li Chen
- Department of Gastroenterology and Hepatology, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Wen-Jie Ding
- Department of Gastroenterology and Hepatology, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Qiong-Fang Yu
- Department of Gastroenterology and Hepatology, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
11
|
Bellei B, Migliano E, Picardo M. A Framework of Major Tumor-Promoting Signal Transduction Pathways Implicated in Melanoma-Fibroblast Dialogue. Cancers (Basel) 2020; 12:cancers12113400. [PMID: 33212834 PMCID: PMC7697272 DOI: 10.3390/cancers12113400] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 11/11/2020] [Accepted: 11/13/2020] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Melanoma cells reside in a complex stromal microenvironment, which is a critical component of disease onset and progression. Mesenchymal or fibroblastic cell type are the most abundant cellular element of tumor stroma. Factors secreted by melanoma cells can activate non-malignant associated fibroblasts to become melanoma associate fibroblasts (MAFs). MAFs promote tumorigenic features by remodeling the extracellular matrix, supporting tumor cells proliferation, neo-angiogenesis and drug resistance. Additionally, environmental factors may contribute to the acquisition of pro-tumorigenic phenotype of fibroblasts. Overall, in melanoma, perturbed tissue homeostasis contributes to modulation of major oncogenic intracellular signaling pathways not only in tumor cells but also in neighboring cells. Thus, targeted molecular therapies need to be considered from the reciprocal point of view of melanoma and stromal cells. Abstract The development of a modified stromal microenvironment in response to neoplastic onset is a common feature of many tumors including cutaneous melanoma. At all stages, melanoma cells are embedded in a complex tissue composed by extracellular matrix components and several different cell populations. Thus, melanomagenesis is not only driven by malignant melanocytes, but also by the altered communication between melanocytes and non-malignant cell populations, including fibroblasts, endothelial and immune cells. In particular, cancer-associated fibroblasts (CAFs), also referred as melanoma-associated fibroblasts (MAFs) in the case of melanoma, are the most abundant stromal cells and play a significant contextual role in melanoma initiation, progression and metastasis. As a result of dynamic intercellular molecular dialogue between tumor and the stroma, non-neoplastic cells gain specific phenotypes and functions that are pro-tumorigenic. Targeting MAFs is thus considered a promising avenue to improve melanoma therapy. Growing evidence demonstrates that aberrant regulation of oncogenic signaling is not restricted to transformed cells but also occurs in MAFs. However, in some cases, signaling pathways present opposite regulation in melanoma and surrounding area, suggesting that therapeutic strategies need to carefully consider the tumor–stroma equilibrium. In this novel review, we analyze four major signaling pathways implicated in melanomagenesis, TGF-β, MAPK, Wnt/β-catenin and Hyppo signaling, from the complementary point of view of tumor cells and the microenvironment.
Collapse
Affiliation(s)
- Barbara Bellei
- Laboratory of Cutaneous Physiopathology and Integrated Center of Metabolomics Research, San Gallicano Dermatological Institute, IRCCS, 00144 Rome, Italy;
- Correspondence: ; Tel.: +39-0652666246
| | - Emilia Migliano
- Department of Plastic and Regenerative Surgery, San Gallicano Dermatological Institute, IRCCS, 00144 Rome, Italy;
| | - Mauro Picardo
- Laboratory of Cutaneous Physiopathology and Integrated Center of Metabolomics Research, San Gallicano Dermatological Institute, IRCCS, 00144 Rome, Italy;
| |
Collapse
|
12
|
Zhang W, Mao K, Liu S, Xu Y, Ren J. miR-942-5p promotes the proliferation and invasion of human melanoma cells by targeting DKK3. J Recept Signal Transduct Res 2020; 41:180-187. [PMID: 32772782 DOI: 10.1080/10799893.2020.1804280] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
OBJECTIVE The purpose of this study was to figure out the dysregulation of miR-942-5p in melanoma and its role in melanoma pathogenesis. METHODS Quantitative real-time PCR (qRT-PCR) assay was used to determine the change of RNA expression. Protein expression was examined by Western blotting. miRNA target was validated through TargetScan and luciferase assay. Cell migration and invasion were detected by wound healing and transwell assay, respectively. RESULTS Results of qRT-PCR manifested miR-942-5p were upregulated in melanoma cell. High expression of miR-942-5p in melanoma patients presented a poor prognosis. Upregulation of miR-942-5p accelerated cell proliferation, migration, and invasion in melanoma cells. Cell apoptosis was inhibited by miR-942-5p mimics. Suppression of miR-942-5p by its inhibitor showed the opposite effects in melanoma cells. TargetScan and luciferase assay showed that miR-942-5p directly targeted to the 3'-untranslated region (3'-UTR) of DKK3. Overexpression of DKK3 inhibited GSK-3β phosphorylation and reduced the expression of β-catenin in both cytoplasm and nucleus, which were induced by miR-942-5p mimics leading to the activation of Wnt/β-catenin pathway. CONCLUSION Upregulation of miR-942-5p was observed in melanoma cells and tissues and significantly associated with a poor prognosis. Though targeting 3'-UTR of DKK3, miR-942-5p could activate Wnt/β-catenin pathway, resulting in melanoma cell proliferation, migration, and invasion, which promoted the development of melanoma. These results showed that miR-942-5p might be a diagnosis and prognosis biomarker in melanoma.
Collapse
Affiliation(s)
- Weina Zhang
- Department of Plastic and Cosmetic Surgery, The Affiliated Hospital of Qingdao University, Qingdao City, China
| | - Kaiping Mao
- Department of Thoracic Surgery, The Affiliated Hospital of Qingdao University, Qingdao City, China
| | - Sumei Liu
- Clinical Teaching and Research Office, Qingdao Health School, Qingdao City, China
| | - Yujiao Xu
- Department of Hemodialysis, Shandong Qingdao Hospital of Intergrated Traditional and Western Medicine, Qingdao City, China
| | - Jizhen Ren
- Department of Plastic and Cosmetic Surgery, The Affiliated Hospital of Qingdao University, Qingdao City, China
| |
Collapse
|
13
|
Gajos-Michniewicz A, Czyz M. WNT Signaling in Melanoma. Int J Mol Sci 2020; 21:E4852. [PMID: 32659938 PMCID: PMC7402324 DOI: 10.3390/ijms21144852] [Citation(s) in RCA: 130] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 07/06/2020] [Accepted: 07/07/2020] [Indexed: 12/12/2022] Open
Abstract
WNT-signaling controls important cellular processes throughout embryonic development and adult life, so any deregulation of this signaling can result in a wide range of pathologies, including cancer. WNT-signaling is classified into two categories: β-catenin-dependent signaling (canonical pathway) and β-catenin-independent signaling (non-canonical pathway), the latter can be further divided into WNT/planar cell polarity (PCP) and calcium pathways. WNT ligands are considered as unique directional growth factors that contribute to both cell proliferation and polarity. Origin of cancer can be diverse and therefore tissue-specific differences can be found in WNT-signaling between cancers, including specific mutations contributing to cancer development. This review focuses on the role of the WNT-signaling pathway in melanoma. The current view on the role of WNT-signaling in cancer immunity as well as a short summary of WNT pathway-related drugs under investigation are also provided.
Collapse
Affiliation(s)
| | - Malgorzata Czyz
- Department of Molecular Biology of Cancer, Medical University of Lodz, 6/8 Mazowiecka Street, 92–215 Lodz, Poland;
| |
Collapse
|
14
|
Jin Y, Liu J, Liu Y, Liu Y, Guo G, Yu S, An R. Oxymatrine Inhibits Renal Cell Carcinoma Progression by Suppressing β-Catenin Expression. Front Pharmacol 2020; 11:808. [PMID: 32581789 PMCID: PMC7289957 DOI: 10.3389/fphar.2020.00808] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 05/18/2020] [Indexed: 01/17/2023] Open
Abstract
Aims Oxymatrine (OMT) has been identified to possess immunomodulatory, antiinflammatory and anticancer properties. This study aimed to investigate its precise function and the underlying molecular mechanisms in renal cell carcinoma progression. Methods The antineoplastic effect of oxymatrine was investigated by CCK-8 assay, cell cycle analysis, apoptosis assay, wound healing experiment, transwell assay, and drug-sensitivity analysis in renal cancer cells following oxymatrine treatment. The modulation of oxymatrine on β-catenin was analyzed through western blot and immunofluorescence assay. β-catenin overexpression was employed to determine the key role of β-catenin in oxymatrine-inhibited renal cell carcinoma in vitro. In addition, animal model was established to investigate the effect of oxymatrine on tumor growth in vivo. Results Oxymatrine inhibited renal cell carcinoma progression in vitro, including cell proliferation, apoptosis, migration, invasion and chemotherapy sensitivity. Further mechanistic studies demonstrated that oxymatrine exerted its antineoplastic effect through suppressing the expression of β-catenin. Moreover, in nude mice model, oxymatrine exhibited remarkable inhibition of tumor growth, which was consistent with our in vitro results. Conclusions Our findings illuminate oxymatrine as an effective antitumor agent in renal cell carcinoma, and suggest it a promising therapeutic application in renal cell carcinoma treatment.
Collapse
Affiliation(s)
- Yinshan Jin
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jiannan Liu
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yadong Liu
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yang Liu
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Guiying Guo
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shiliang Yu
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Ruihua An
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
15
|
Abstract
Developmental signaling pathways control a vast array of biological processes during embryogenesis and in adult life. The WNT pathway was discovered simultaneously in cancer and development. Recent advances have expanded the role of WNT to a wide range of pathologies in humans. Here, we discuss the WNT pathway and its role in human disease and some of the advances in WNT-related treatments.
Collapse
|
16
|
Shukla S, Milewski D, Pradhan A, Rama N, Rice K, Le T, Flick MJ, Vaz S, Zhao X, Setchell KD, Logarinho E, Kalinichenko VV, Kalin TV. The FOXM1 Inhibitor RCM-1 Decreases Carcinogenesis and Nuclear β-Catenin. Mol Cancer Ther 2019; 18:1217-1229. [PMID: 31040162 PMCID: PMC7341442 DOI: 10.1158/1535-7163.mct-18-0709] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 12/13/2018] [Accepted: 04/25/2019] [Indexed: 12/13/2022]
Abstract
The oncogenic transcription factor FOXM1 has been previously shown to play a critical role in carcinogenesis by inducing cellular proliferation in multiple cancer types. A small-molecule compound, Robert Costa Memorial drug-1 (RCM-1), has been recently identified from high-throughput screen as an inhibitor of FOXM1 in vitro and in mouse model of allergen-mediated lung inflammation. In the present study, we examined antitumor activities of RCM-1 using tumor models. Treatment with RCM-1 inhibited tumor cell proliferation as evidenced by increased cell-cycle duration. Confocal imaging of RCM-1-treated tumor cells indicated that delay in cellular proliferation was concordant with inhibition of FOXM1 nuclear localization in these cells. RCM-1 reduced the formation and growth of tumor cell colonies in the colony formation assay. In animal models, RCM-1 treatment inhibited growth of mouse rhabdomyosarcoma Rd76-9, melanoma B16-F10, and human H2122 lung adenocarcinoma. RCM-1 decreased FOXM1 protein in the tumors, reduced tumor cell proliferation, and increased tumor cell apoptosis. RCM-1 decreased protein levels and nuclear localization of β-catenin, and inhibited protein-protein interaction between β-catenin and FOXM1 in cultured tumor cells and in vivo Altogether, our study provides important evidence of antitumor potential of the small-molecule compound RCM-1, suggesting that RCM-1 can be a promising candidate for anticancer therapy.
Collapse
Affiliation(s)
- Samriddhi Shukla
- Perinatal Institute, Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - David Milewski
- Perinatal Institute, Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Arun Pradhan
- Perinatal Institute, Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
- Center for Lung Regenerative Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Nihar Rama
- Perinatal Institute, Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Kathryn Rice
- College of Medicine, University of Cincinnati, Cincinnati, Ohio
| | - Tien Le
- Perinatal Institute, Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Matthew J Flick
- Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Sara Vaz
- Instituto de Biologia Molecular e Celular (IBMC), Instituto de Inovação e Investigação em Saúde (i3S), Universidade do Porto, Rua Alfredo Allen, Porto, Portugal
| | - Xueheng Zhao
- Mass Spectrometry Facility, Pathology and Laboratory Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Kenneth D Setchell
- Mass Spectrometry Facility, Pathology and Laboratory Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Elsa Logarinho
- Instituto de Biologia Molecular e Celular (IBMC), Instituto de Inovação e Investigação em Saúde (i3S), Universidade do Porto, Rua Alfredo Allen, Porto, Portugal
| | - Vladimir V Kalinichenko
- Perinatal Institute, Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
- Center for Lung Regenerative Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Tanya V Kalin
- Perinatal Institute, Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio.
| |
Collapse
|
17
|
Moschitto MJ, Doubleday PF, Catlin DS, Kelleher NL, Liu D, Silverman RB. Mechanism of Inactivation of Ornithine Aminotransferase by (1 S,3 S)-3-Amino-4-(hexafluoropropan-2-ylidenyl)cyclopentane-1-carboxylic Acid. J Am Chem Soc 2019; 141:10711-10721. [PMID: 31251613 DOI: 10.1021/jacs.9b03254] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The inhibition of ornithine aminotransferase (OAT), a pyridoxal 5'-phosphate-dependent enzyme, has been implicated as a treatment for hepatocellular carcinoma (HCC), the most common form of liver cancer, for which there is no effective treatment. From a previous evaluation of our aminotransferase inhibitors, (1S,3S)-3-amino-4-(perfluoropropan-2-ylidene)cyclopentane-1-carboxylic acid hydrochloride (1) was found to be a selective and potent inactivator of human OAT (hOAT), which inhibited the growth of HCC in athymic mice implanted with human-derived HCC, even at a dose of 0.1 mg/kg. Currently, investigational new drug (IND)-enabling studies with 1 are underway. The inactivation mechanism of 1, however, has proved to be elusive. Here we propose three possible mechanisms, based on mechanisms of known aminotransferase inactivators: Michael addition, enamine addition, and fluoride ion elimination followed by conjugate addition. On the basis of crystallography and intact protein mass spectrometry, it was determined that 1 inactivates hOAT through fluoride ion elimination to an activated 1,1'-difluoroolefin, followed by conjugate addition and hydrolysis. This result was confirmed with additional studies, including the detection of the cofactor structure by mass spectrometry and through the identification of turnover metabolites. On the basis of this inactivation mechanism and to provide further evidence for the mechanism, analogues of 1 (19, 20) were designed, synthesized, and demonstrated to have the predicted selective inactivation mechanism. These analogues highlight the importance of the trifluoromethyl group and provide a basis for future inactivator design.
Collapse
Affiliation(s)
| | | | - Daniel S Catlin
- Department of Chemistry and Biochemistry , Loyola University Chicago , Chicago , Illinois 60660 , United States
| | | | - Dali Liu
- Department of Chemistry and Biochemistry , Loyola University Chicago , Chicago , Illinois 60660 , United States
| | | |
Collapse
|
18
|
Dickkopf-1: Current knowledge and related diseases. Life Sci 2018; 209:249-254. [PMID: 30102902 DOI: 10.1016/j.lfs.2018.08.019] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 08/02/2018] [Accepted: 08/07/2018] [Indexed: 02/07/2023]
Abstract
Dickkopf-1(DKK-1) has been identified as a secretory protein that can inhibit the Wnt signaling transduction pathway. It is well known that the Wnt signaling pathway plays an important role in embryogenesis, organogenesis and homeostasis. This signaling cascade is essential for many normal physiological processes such as cellular proliferation, tissue regeneration, embryonic development and many other systemic and local effects, and it can be regulated at different levels. Therefore, defects in the pathway may lead to some complicated effects. In addition, it has been demonstrated that defects in this pathway are closely linked to some diseases including cancer, rheumatism, bone disease, diabetes, and Alzheimer disease. Since DKK-1 is an antagonist of the Wnt pathway, it may be related to these diseases; in fact, many studies have identified this fact. This review will summarize the current knowledge of DKK-1 and DKK-1-mediated regulation of Wnt signaling in the development of these related diseases.
Collapse
|
19
|
Yang S, Liu B, Ji K, Fan R, Dong C. MicroRNA-5110 regulates pigmentation by cotargeting melanophilin and WNT family member 1. FASEB J 2018; 32:5405-5412. [PMID: 29733692 PMCID: PMC6133708 DOI: 10.1096/fj.201800040r] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Mammalian pigmentation requires the production of melanin by melanocytes and its transfer to neighboring keratinocytes. These complex processes are regulated by several molecular pathways. Melanophilin ( MLPH) and WNT family member 1 ( WNT1), known to be involved in melanin transfer and melanin production, respectively, were predicted to be targets of microRNA-5110 using bioinformatics. In the current study, we investigated the effects of microRNA-5110 on pigmentation in alpaca ( Vicugna pacos) melanocytes. In situ hybridization identified high levels of microRNA-5110 in the cytoplasm of alpaca melanocytes. Luciferase activity assays confirmed that MLPH and WNT1 were targeted by microRNA-5110 in these cells. Overexpression and knockdown of microRNA-5110 in alpaca melanocytes downregulated and upregulated MLPH and WNT1 expression at the mRNA and protein levels, respectively. In addition, overexpression and knockdown of microRNA-5110 in alpaca melanocytes decreased and increased, respectively, the mRNA levels of the melanin transfer-related genes, rat sarcoma (RAS)-associated binding ( RAB27a) and myosin 5a ( MYO5a); the mRNA levels of microphthalmia-associated transcription factor ( MITF), tyrosinase ( TYR), and tyrosinase-related protein ( TYRP) 1; and the production of total alkali melanin and pheomelanin. In contrast, overexpression and knockdown of microRNA-5110 increased and decreased the mRNA levels of TYRP2, respectively. Overexpression of microRNA-5110 also increased eumelanin. These results indicate that microRNA-5110 regulates pigmentation in alpaca melanocytes by directly targeting MLPH and WNT1 to affect eumelanin production and transfer.-Yang, S., Liu, B., Ji, K., Fan, R., Dong, C. MicroRNA-5110 regulates pigmentation by cotargeting melanophilin and WNT family member 1.
Collapse
Affiliation(s)
- Shanshan Yang
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, China
| | - Bo Liu
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, China
| | - Kaiyuan Ji
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, China
| | - Ruiwen Fan
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, China
| | - Changsheng Dong
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, China
| |
Collapse
|
20
|
Min W, Liu X, Lu Y, Gong Z, Wang M, Lin S, Kang H, Jin T, Wang X, Ma X, Liu K, Dai C, Zheng Y, Li S, Ma Q, Dai Z. Association of transcription factor 7-like 2 gene polymorphisms with breast cancer risk in northwest Chinese women. Oncotarget 2018; 7:77175-77182. [PMID: 27738320 PMCID: PMC5363578 DOI: 10.18632/oncotarget.12591] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 09/29/2016] [Indexed: 01/08/2023] Open
Abstract
Genetic variations in transcription factor 7-like 2 (TCF7L2) are associated with cancer risk. This study was conducted to establish the relationship between TCF7L2 polymorphisms (rs1225404, rs7003146, and rs7903146) and clinical features and risk of breast cancer in Northwest Chinese Han women. In this study, three polymorphisms of TCF7L2 (rs1225404, rs7003146, and rs7903146) were genotyped in 458 patients with breast cancer and 500 healthy controls using the Sequenom MassARRAY-iPLEX system. We evaluated the associations between the polymorphisms and breast cancer using odds ratios (ORs) and corresponding 95% confidence intervals (95% CIs). The C allele of rs1225404 was associated with increased breast cancer risk (OR = 1.58, P = 0.0004, PC= 0.0012), whereas the G allele of rs7003146 was associated with decreased breast cancer risk (OR = 0.71, P = 0.01, PC= 0.03). Furthermore, the rs1225404 polymorphism positively correlated with negative progesterone receptor status. A positive correlation with positive estrogen receptor (ER) status was observed for the rs7003146 polymorphism. Our results suggest that TCF7L2 polymorphisms rs1225404 and rs7003146, but not rs7903146, may affect breast cancer risk in Northwest Chinese women. Additionally, the tag polymorphisms in TCF7L2 are associated with the clinical features of breast cancer, which may provide us novel insight into the pathogenesis of breast cancer.
Collapse
Affiliation(s)
- Weili Min
- Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Xinghan Liu
- Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Ye Lu
- Department of Student Affairs, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Zhuoqing Gong
- Department of Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Meng Wang
- Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Shuai Lin
- Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Huafeng Kang
- Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Tianbo Jin
- Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Xijing Wang
- Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Xiaobin Ma
- Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Kang Liu
- Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Cong Dai
- Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Yi Zheng
- Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Shanli Li
- Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Qingyong Ma
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Zhijun Dai
- Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| |
Collapse
|
21
|
Sinnberg T, Levesque MP, Krochmann J, Cheng PF, Ikenberg K, Meraz-Torres F, Niessner H, Garbe C, Busch C. Wnt-signaling enhances neural crest migration of melanoma cells and induces an invasive phenotype. Mol Cancer 2018; 17:59. [PMID: 29454361 PMCID: PMC5816360 DOI: 10.1186/s12943-018-0773-5] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 01/29/2018] [Indexed: 01/04/2023] Open
Abstract
Background During embryonic development Wnt family members and bone morphogenetic proteins (BMPs) cooperatively induce epithelial-mesenchymal transition (EMT) in the neural crest. Wnt and BMPs are reactivated during malignant transformation in melanoma. We previously demonstrated that the BMP-antagonist noggin blocked the EMT phenotype of melanoma cells in the neural crest and malignant invasion of melanoma cells in the chick embryo; vice-versa, malignant invasion was induced in human melanocytes in vivo by pre-treatment with BMP-2. Results Although there are conflicting results in the literature about the role of β-catenin for invasion of melanoma cells, we found Wnt/β-catenin signaling to be analogously important for the EMT-like phenotype of human metastatic melanoma cells in the neural crest and during invasion: β-catenin was frequently expressed at the invasive front of human primary melanomas and Wnt3a expression was inversely correlated with survival of melanoma patients. Accordingly, cytoplasmic β-catenin levels were increased during invasion of melanoma cells in the rhombencephalon of the chick embryo. Fibroblast derived Wnt3a reduced melanoma cell adhesion and enhanced migration, while the β-catenin inhibitor PKF115–584 increased adhesion and reduced migration in vitro and in the chick embryonic neural crest environment in vivo. Similarly, knockdown of β-catenin impaired intradermal melanoma cell invasion and PKF115–584 efficiently reduced liver metastasis in a chick chorioallantoic membrane model. Our observations were accompanied by specific alterations in gene expression which are linked to overall survival of melanoma patients. Conclusion We present a novel role for Wnt-signaling in neural crest like melanoma cell invasion and metastasis, stressing the crucial role of embryonic EMT-inducing neural crest signaling for the spreading of malignant melanoma. Electronic supplementary material The online version of this article (10.1186/s12943-018-0773-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tobias Sinnberg
- Center for Dermatooncology, Department of Dermatology, University Hospital Tübingen, University of Tübingen, Liebermeisterstr.25, 72076, Tübingen, Germany.
| | - Mitchell P Levesque
- Department of Dermatology, Universitaets Spital Zürich, Gloriastrasse 31, 8091, Zürich, Switzerland
| | - Jelena Krochmann
- Center for Dermatooncology, Department of Dermatology, University Hospital Tübingen, University of Tübingen, Liebermeisterstr.25, 72076, Tübingen, Germany
| | - Phil F Cheng
- Department of Dermatology, Universitaets Spital Zürich, Gloriastrasse 31, 8091, Zürich, Switzerland
| | - Kristian Ikenberg
- Institute of Clinical Pathology, University Hospital Zürich, Schmelzbergstrasse 12, 8091, Zürich, Switzerland
| | - Francisco Meraz-Torres
- Center for Dermatooncology, Department of Dermatology, University Hospital Tübingen, University of Tübingen, Liebermeisterstr.25, 72076, Tübingen, Germany
| | - Heike Niessner
- Center for Dermatooncology, Department of Dermatology, University Hospital Tübingen, University of Tübingen, Liebermeisterstr.25, 72076, Tübingen, Germany
| | - Claus Garbe
- Center for Dermatooncology, Department of Dermatology, University Hospital Tübingen, University of Tübingen, Liebermeisterstr.25, 72076, Tübingen, Germany
| | - Christian Busch
- Center for Dermatooncology, Department of Dermatology, University Hospital Tübingen, University of Tübingen, Liebermeisterstr.25, 72076, Tübingen, Germany. .,Dermateam, Bankstrasse 4, 8400, Winterthur, Switzerland.
| |
Collapse
|
22
|
Duffy DJ, Krstic A, Schwarzl T, Halasz M, Iljin K, Fey D, Haley B, Whilde J, Haapa-Paananen S, Fey V, Fischer M, Westermann F, Henrich KO, Bannert S, Higgins DG, Kolch W. Wnt signalling is a bi-directional vulnerability of cancer cells. Oncotarget 2018; 7:60310-60331. [PMID: 27531891 PMCID: PMC5312386 DOI: 10.18632/oncotarget.11203] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 07/26/2016] [Indexed: 12/30/2022] Open
Abstract
Wnt signalling is involved in the formation, metastasis and relapse of a wide array of cancers. However, there is ongoing debate as to whether activation or inhibition of the pathway holds the most promise as a therapeutic treatment for cancer, with conflicting evidence from a variety of tumour types. We show that Wnt/β-catenin signalling is a bi-directional vulnerability of neuroblastoma, malignant melanoma and colorectal cancer, with hyper-activation or repression of the pathway both representing a promising therapeutic strategy, even within the same cancer type. Hyper-activation directs cancer cells to undergo apoptosis, even in cells oncogenically driven by β-catenin. Wnt inhibition blocks proliferation of cancer cells and promotes neuroblastoma differentiation. Wnt and retinoic acid co-treatments synergise, representing a promising combination treatment for MYCN-amplified neuroblastoma. Additionally, we report novel cross-talks between MYCN and β-catenin signalling, which repress normal β-catenin mediated transcriptional regulation. A β-catenin target gene signature could predict patient outcome, as could the expression level of its DNA binding partners, the TCF/LEFs. This β-catenin signature provides a tool to identify neuroblastoma patients likely to benefit from Wnt-directed therapy. Taken together, we show that Wnt/β-catenin signalling is a bi-directional vulnerability of a number of cancer entities, and potentially a more broadly conserved feature of malignant cells.
Collapse
Affiliation(s)
- David J Duffy
- Systems Biology Ireland, University College Dublin, Belfield, Dublin, Ireland.,Current address: The Whitney Laboratory for Marine Bioscience, University of Florida, St. Augustine, Florida, USA
| | - Aleksandar Krstic
- Systems Biology Ireland, University College Dublin, Belfield, Dublin, Ireland
| | - Thomas Schwarzl
- Systems Biology Ireland, University College Dublin, Belfield, Dublin, Ireland.,Current address: European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Melinda Halasz
- Systems Biology Ireland, University College Dublin, Belfield, Dublin, Ireland
| | | | - Dirk Fey
- Systems Biology Ireland, University College Dublin, Belfield, Dublin, Ireland
| | - Bridget Haley
- Systems Biology Ireland, University College Dublin, Belfield, Dublin, Ireland
| | - Jenny Whilde
- Systems Biology Ireland, University College Dublin, Belfield, Dublin, Ireland
| | | | - Vidal Fey
- VTT Technical Research Centre of Finland, Espoo, Finland
| | - Matthias Fischer
- Department of Paediatric Haematology and Oncology and Center for Molecular Medicine Cologne (CMMC), University Hospital Cologne, Cologne, Germany
| | - Frank Westermann
- Division of NB Genomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Kai-Oliver Henrich
- Division of NB Genomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Steffen Bannert
- Division of NB Genomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Desmond G Higgins
- Systems Biology Ireland, University College Dublin, Belfield, Dublin, Ireland.,Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin, Ireland.,School of Medicine, University College Dublin, Belfield, Dublin, Ireland
| | - Walter Kolch
- Systems Biology Ireland, University College Dublin, Belfield, Dublin, Ireland.,Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin, Ireland.,School of Medicine, University College Dublin, Belfield, Dublin, Ireland
| |
Collapse
|
23
|
Kovacs D, Migliano E, Muscardin L, Silipo V, Catricalà C, Picardo M, Bellei B. The role of Wnt/β-catenin signaling pathway in melanoma epithelial-to-mesenchymal-like switching: evidences from patients-derived cell lines. Oncotarget 2017; 7:43295-43314. [PMID: 27175588 PMCID: PMC5190024 DOI: 10.18632/oncotarget.9232] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 04/10/2016] [Indexed: 12/13/2022] Open
Abstract
Deregulations or mutations of WNT/β-catenin signaling have been associated to both tumour formation and progression. However, contradictory results concerning the role of β-catenin in human melanoma address an open question on its oncogenic nature and prognostic value in this tumour. Changes in WNT signaling pathways have been linked to phenotype switching of melanoma cells between a highly proliferative/non-invasive and a slow proliferative/metastatic condition. We used a novel panel of cell lines isolated from melanoma specimens, at initial passages, to investigate phenotype differences related to the levels and activity of WNT/β-catenin signaling pathway. This in vitro cell system revealed a marked heterogeneity that comprises, in some cases, two distinct tumour-derived subpopulations of cells presenting a different activation level and cellular distribution of β-catenin. In cells derived from the same tumor, we demonstrated that the prevalence of LEF1 (high β-catenin expressing cells) or TCF4 (low β-catenin expressing cells) as β-catenin partner for DNA binding, is associated to the expression of two distinct profiles of WNT-responsive genes. Interestingly, melanoma cells expressing relative low level of β-catenin and an invasive markers signature were associated to the TNF-α-induced pro-inflammatory pathway and to the chemotherapy resistance, suggesting that the co-existence of melanoma subpopulations with distinct biological properties could influence the impact of chemo- and immunotherapy.
Collapse
Affiliation(s)
- Daniela Kovacs
- Laboratory of Cutaneous Physiopathology and Integrated Center of Metabolomics Research, San Gallicano Dermatologic Institute, IRCCS, Rome, Italy
| | - Emilia Migliano
- Department of Plastic and Reconstructive Surgery, San Gallicano Dermatologic Institute, IRCCS, Rome, Italy
| | - Luca Muscardin
- Dermatopathological Laboratory, San Gallicano Dermatologic Institute, IRCCS, Rome, Italy
| | - Vitaliano Silipo
- Department of Oncologic Dermatology, San Gallicano Dermatologic Institute, IRCCS, Rome, Italy
| | - Caterina Catricalà
- Department of Oncologic Dermatology, San Gallicano Dermatologic Institute, IRCCS, Rome, Italy
| | - Mauro Picardo
- Laboratory of Cutaneous Physiopathology and Integrated Center of Metabolomics Research, San Gallicano Dermatologic Institute, IRCCS, Rome, Italy
| | - Barbara Bellei
- Laboratory of Cutaneous Physiopathology and Integrated Center of Metabolomics Research, San Gallicano Dermatologic Institute, IRCCS, Rome, Italy
| |
Collapse
|
24
|
Activation of PPARα by clofibrate sensitizes pancreatic cancer cells to radiation through the Wnt/β-catenin pathway. Oncogene 2017; 37:953-962. [PMID: 29059162 DOI: 10.1038/onc.2017.401] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 08/18/2017] [Accepted: 09/13/2017] [Indexed: 02/08/2023]
Abstract
Radiotherapy is emerging as an important modality for the local control of pancreatic cancer, but pancreatic cancer cell radioresistance remains a serious concern. Peroxisome proliferator-activated receptor α (PPARα) is a member of the PPAR nuclear hormone receptor superfamily, which can be activated by fibrate ligands. The clinical relevance of PPARα and its biological function in pancreatic cancer radiosensitivity have not been previously described. In this study, we examined PPARα expression in tissue samples of pancreatic cancer patients. We found significantly higher expression of PPARα in pancreatic cancer tissues than in tumor-adjacent tissues and that the PPARα expression level is inversely associated with higher overall patient survival rate. We further observed that PPARα activation by its agonist clofibrate sensitizes pancreatic cancer cells to radiation by modulating cell cycle progression and apoptosis in several pancreatic cancer cell lines. Small interfering RNA-mediated PPARα silencing and PPARα blockade by the antagonist GW6471 abolish the effect of clofibrate on radiosensitization. An in vivo study showed that PANC1 xenografts treated with clofibrate are more sensitive to radiation than untreated xenografts. mRNA profiling by microarray analysis revealed that the expression of PTPRZ1 and Wnt8a, two core components of the β-catenin pathway, is downregulated by clofibrate. Chromatin immunoprecipitation analysis confirmed that clofibrate abrogates the binding of nuclear factor-κB to the PTPRZ1 and Wnt8a promoters, ultimately decreasing Wnt/β-catenin signaling activity, which is associated with radiosensitivity. Overall, we demonstrate that PPARα is overexpressed in pancreatic cancer tissues and clofibrate-mediated PPARα activation sensitizes pancreatic cancer cells to radiation through the Wnt/β-catenin pathway.
Collapse
|
25
|
Effects of microRNA-136 on melanoma cell proliferation, apoptosis, and epithelial-mesenchymal transition by targetting PMEL through the Wnt signaling pathway. Biosci Rep 2017; 37:BSR20170743. [PMID: 28724603 PMCID: PMC5587917 DOI: 10.1042/bsr20170743] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 07/16/2017] [Accepted: 07/18/2017] [Indexed: 12/17/2022] Open
Abstract
The study aims to evaluate the effects of miR-136 on the proliferation, apoptosis, and epithelial–mesenchymal transition (EMT) of melanoma cells by targetting premelanosome protein (PMEL) through the Wnt signaling pathway. After establishment of melanoma mouse models, melanoma (model group) and normal tissues (normal group) were collected. Immunohistochemistry was performed to determine PMEL protein concentration. Mouse melanoma cells were assigned into control, blank, negative control (NC), miR-136 mimics, miR-136 inhibitors, siRNA-PMEL, and miR-136 inhibitors + siRNA-PMEL, LiC1 (Wnt signaling pathway activator), and siRNA-PMEL+ LiCl groups. MTT, Scratch test, Transwell assay, and flow cytometry were performed to measure cell proliferation, migration, invasion, and apoptosis. Quantitative real-time PCR (qRT-PCR) and Western blotting were performed to evaluate miR-136, PMEL, β-catenin, Wnt3a, Bcl-2, Bax, Caspase, E-cadherin, and N-cadherin expressions. PMEL is highly expressed in melanoma tissues. MiR-136, Bax, Caspase, and E-cadherin expressions decreased in the model group, whereas PMEL, β-catenin, Bcl-2, Wnt3a, and N-cadherin expressions increased. Bax, Caspase, and E-cadherin expressions increased in the miR-136 mimics and siRNA-PMEL groups, whereas the expressions decreased in the miR-136 inhibitors group and LiC1 group. PMEL, β-catenin, Bcl-2, Wnt3a, and N-cadherin expressions, cell proliferation, migration, and invasion decreased, and the apoptosis rate inceased in the miR-136 mimics and siRNA-PMEL groups; whereas the tendencies were opposite to those in the miR-136 inhibitors group and LiC1 group. In the siRNA-PMEL+ LiCl group, PMEL expression decreased. These findings indicated that overexpression of miR-136 inhibits melanoma cell EMT, proliferation, migration, invasion, and promotes apoptosis by targetting PMEL through down-regulation of the Wnt signaling pathway.
Collapse
|
26
|
The Effect of Pyrroloquinoline Quinone on the Expression of WISP1 in Traumatic Brain Injury. Stem Cells Int 2017; 2017:4782820. [PMID: 28883836 PMCID: PMC5573117 DOI: 10.1155/2017/4782820] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2017] [Accepted: 05/30/2017] [Indexed: 11/24/2022] Open
Abstract
WISP1, as a member of the CCN4 protein family, has cell protective effects of promoting cell proliferation and inhibiting cell apoptosis. Although some studies have confirmed that WISP1 is concerned with colon cancer and lung cancer, there is little report about the influence of WISP1 in traumatic brain injury. Here, we found that the expression of WISP1 mRNA and protein decreased at 3 d and then increased at 5 d after traumatic brain injury (TBI). Meanwhile, immunofluorescence demonstrated that there was little colocation of WISP1 with GFAP, Iba1, and WISP1 colocalized with NeuN partly. WISP1 colocalized with LC3, but there was little of colocation about WISP1 with cleaved caspase-3. Subsequent study displayed that the expression of β-catenin protein was identical to that of WISP1 after TBI. WISP1 was mainly located in cytoplasm of PC12 or SHSY5Y cells. Compared with the negative control group, WISP1 expression reduced obviously in SHSY5Y cells transfected with WISP1 si-RNA. CCK-8 assay showed that pyrroloquinoline quinone (PQQ) had little influence on viability of PC12 and SHSY5Y cells. These results suggested that WISP1 played a protective role after traumatic brain injury in rats, and this effect might be relative to autophagy caused by traumatic brain injury.
Collapse
|
27
|
Palmitoylation of proteins in cancer. Biochem Soc Trans 2017; 45:409-416. [DOI: 10.1042/bst20160233] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 01/30/2017] [Accepted: 02/06/2017] [Indexed: 12/14/2022]
Abstract
Post-translational modification of proteins by attachment of palmitate serves as a mechanism to regulate protein localization and function in both normal and malignant cells. Given the essential role that palmitoylation plays in cancer cell signaling, approaches that target palmitoylated proteins and palmitoyl acyltransferases (PATs) have the potential for therapeutic intervention in cancer. Highlighted here are recent advances in understanding the importance of protein palmitoylation in tumorigenic pathways. A new study has uncovered palmitoylation sites within the epidermal growth factor receptor that regulate receptor trafficking, signaling and sensitivity to tyrosine kinase inhibitors. Global data analysis from nearly 150 cancer studies reveals genomic alterations in several PATs that may account for their ability to function as tumor suppressors or oncogenes. Selective inhibitors have recently been developed that target hedgehog acyltransferase (Hhat) and Porcupine (Porcn), the acyltransferases that modify hedgehog and Wnt proteins, respectively. These inhibitors, coupled with targeted knockdown of Hhat and Porcn, reveal the essential functions of fatty acylation of secreted morphogens in a wide variety of human tumors.
Collapse
|
28
|
Wang E, Wang D, Li B, Ma H, Wang C, Guan L, Zhang H, Yi L, Li S. Capn4 promotes epithelial-mesenchymal transition in human melanoma cells through activation of the Wnt/β-catenin pathway. Oncol Rep 2016; 37:379-387. [PMID: 27878263 DOI: 10.3892/or.2016.5247] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 07/01/2016] [Indexed: 11/06/2022] Open
Abstract
Melanoma, as one of the most highly metastatic types of cancer, is resistant to current treatment methods, including popular targeted molecular therapy. Consequently, it is essential to develop a deeper understanding of the mechanisms involved in melanoma progression so that alternative treatments may be identified. To date, accumulating evidence supports the use of calpains, including calpain small subunit 1 (also known as Capn4 or CAPNS1), which affect cancer progression through many pathways, such as epithelial‑mesenchymal transition (EMT), the Wnt/β-catenin (β-catenin) and the nuclear factor κB (NF-κB) signaling pathways. The EMT pathway is well known as one of the most important events in tumor metastasis. The present study observed cross-talk among the EMT, β-catenin and NF-κB pathways. To identify the underlying mechanisms of Capn4 activity in melanoma cells, we determined Capn4 expression by gene chip and immunohistochemistral analyses in melanoma tissues and cells in vitro. The extent of apoptosis as determined by TUNEL assay, DAPI staining, and cleaved-caspase-3 assay was increased in human melanoma cells in which Capn4 expression had been knocked down when compared with untreated cells. Transwell assays and xenograft tumorigenicity studies were also performed to assess the effects of Capn4 on migration and invasion in vitro and tumor growth in vivo, respectively. The levels of β-catenin, vimentin, E-cadherin and N-cadherin were altered in human melanoma cells as determined by western blot analysis assay. Our study demonstrated that Capn4 is an underlying target for melanoma treatment.
Collapse
Affiliation(s)
- Enwen Wang
- Department of Radiology, College of Basic Medicine, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Donglin Wang
- Department of Medical Oncology, Chongqing Cancer Hospital and Institute and Cancer Center, Chongqing 400030, P.R. China
| | - Bing Li
- Department of Ear-Nose-Throat, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Huiwen Ma
- Department of Medical Oncology, Chongqing Cancer Hospital and Institute and Cancer Center, Chongqing 400030, P.R. China
| | - Chunmei Wang
- Department of Medical Oncology, Chongqing Cancer Hospital and Institute and Cancer Center, Chongqing 400030, P.R. China
| | - Lili Guan
- Department of Nuclear Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Haiwei Zhang
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing Cancer Hospital and Institute and Cancer Center, Chongqing 400030, P.R. China
| | - Lin Yi
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing Cancer Hospital and Institute and Cancer Center, Chongqing 400030, P.R. China
| | - Shaolin Li
- Department of Radiology, College of Basic Medicine, Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
29
|
Dar MS, Singh P, Singh G, Jamwal G, Hussain SS, Rana A, Akhter Y, Monga SP, Dar MJ. Terminal regions of β-catenin are critical for regulating its adhesion and transcription functions. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:2345-57. [DOI: 10.1016/j.bbamcr.2016.06.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 05/31/2016] [Accepted: 06/27/2016] [Indexed: 11/25/2022]
|
30
|
Yin X, Yu XW, Zhu P, Zhang YM, Zhang XH, Wang F, Zhang JJ, Yan W, Xi Y, Wan JB, Kang JX, Zou ZQ, Bu SZ. Endogenously synthesized n-3 fatty acids in fat-1 transgenic mice prevent melanoma progression by increasing E-cadherin expression and inhibiting β-catenin signaling. Mol Med Rep 2016; 14:3476-84. [PMID: 27573698 DOI: 10.3892/mmr.2016.5639] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2015] [Accepted: 07/13/2016] [Indexed: 11/06/2022] Open
Abstract
Malignant melanoma is the most lethal form of skin cancer. Although preclinical studies have shown that n-3 polyunsaturated fatty acids (PUFAs) are beneficial for prevention of melanoma, the molecular mechanisms underlying the protective effects of n‑3 PUFAs on melanoma remain largely unknown. In the present study, endogenously increased levels of n-3 PUFAs in the tumor tissues of omega‑3 fatty acid desaturase (fat‑1) transgenic mice was associated with a reduction in the growth rate of melanoma xenografts. This reduction in tumor growth in fat‑1 mice compared with wild‑type controls may have been associated, in part, to the: i) Increased expression of E‑cadherin and the reduced expression of its transcriptional repressors, the zinc finger E‑box binding homeobox 1 and snail family transcriptional repressor 1; ii) significant repression of the epidermal growth factor receptor/Akt/β‑catenin signaling pathway; and iii) formation of significant levels of n‑3 PUFA‑derived lipid mediators, particularly resolvin D2 and E1, maresin 1 and 15‑hydroxyeicosapentaenoic acid. In addition, vitamin E administration counteracted n‑3 PUFA‑induced lipid peroxidation and enhanced the antitumor effect of n‑3 PUFAs, which suggests that the protective role of n‑3 PUFAs against melanoma is not mediated by n‑3 PUFAs‑induced lipid peroxidation. These results highlight a potential role of n‑3 PUFAs supplementation for the chemoprevention of melanoma in high‑risk individuals, and as a putative adjuvant agent in the treatment of malignant melanoma.
Collapse
Affiliation(s)
- Xuan Yin
- Medical School, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Xiong-Wei Yu
- Medical School, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Pan Zhu
- Medical School, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Yuan-Ming Zhang
- Medical School, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Xiao-Hong Zhang
- Medical School, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Feng Wang
- Clinical Laboratory, Lihuili Hospital, Ningbo, Zhejiang 315040, P.R. China
| | - Jin-Jie Zhang
- Maritime Faculty, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Wang Yan
- Neurosurgery Department, Second Hospital of Ningbo, Ningbo, Zhejiang 315010, P.R. China
| | - Yang Xi
- Medical School, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Jian-Bo Wan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, SAR 519000, P.R. China
| | - Jing-Xuan Kang
- Laboratory for Lipid Medicine and Technology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Zu-Quan Zou
- Medical School, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Shi-Zhong Bu
- Medical School, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| |
Collapse
|
31
|
Gsk3β and Tomm20 are substrates of the SCFFbxo7/PARK15 ubiquitin ligase associated with Parkinson's disease. Biochem J 2016; 473:3563-3580. [PMID: 27503909 PMCID: PMC5260939 DOI: 10.1042/bcj20160387] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 08/08/2016] [Indexed: 12/15/2022]
Abstract
Fbxo7 is a clinically relevant F-box protein, associated with both cancer and Parkinson's disease (PD). Additionally, SNPs within FBXO7 are correlated with alterations in red blood cell parameters. Point mutations within FBXO7 map within specific functional domains, including near its F-box domain and its substrate recruiting domains, suggesting that deficiencies in SCFFbxo7/PARK15 ubiquitin ligase activity are mechanistically linked to early-onset PD. To date, relatively few substrates of the ligase have been identified. These include HURP (hepatoma up-regulated protein), whose ubiquitination results in proteasome-mediated degradation, and c-IAP1 (inhibitor of apoptosis protein 1), TNF receptor-associated factor 2 (TRAF2), and NRAGE, which are not destabilized as a result of ubiquitination. None of these substrates have been linked directly to PD, nor has it been determined whether they would directly engage neuronal cell death pathways. To discover ubiquitinated substrates of SCFFbxo7 implicated more directly in PD aetiology, we conducted a high-throughput screen using protein arrays to identify new candidates. A total of 338 new targets were identified and from these we validated glycogen synthase kinase 3β (Gsk3β), which can phosphorylate α-synuclein, and translocase of outer mitochondrial membrane 20 (Tomm20), a mitochondrial translocase that, when ubiquitinated, promotes mitophagy, as SCFFbxo7 substrates both in vitro and in vivo. Ubiquitin chain restriction analyses revealed that Fbxo7 modified Gsk3β using K63 linkages. Our results indicate that Fbxo7 negatively regulates Gsk3β activity, rather than its levels or localization. In addition, Fbxo7 ubiquitinated Tomm20, and its levels correlated with Fbxo7 expression, indicating a stabilizing effect. None of the PD-associated mutations in Fbxo7 impaired Tomm20 ubiquitination. Our findings demonstrate that SCFFbxo7 has an impact directly on two proteins implicated in pathological processes leading to PD.
Collapse
|
32
|
Intestinal knockout of Nedd4 enhances growth of Apc min tumors. Oncogene 2016; 35:5839-5849. [PMID: 27086928 DOI: 10.1038/onc.2016.125] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 03/07/2016] [Accepted: 03/11/2016] [Indexed: 12/24/2022]
Abstract
Nedd4 (Nedd4-1) is an E3 ubiquitin ligase that belongs to the HECT family and comprises a C2-WW(n)-HECT domain architecture. Although it has been reported to regulate growth factor receptors and cellular signaling, its role in cancer development has been controversial, with some studies proposing that it promotes cancer while others suggest it inhibits tumor growth. Here, we tested the effect of Nedd4 on intestinal tumor formation and growth using Nedd4-knockout mice (Nedd4 floxed (fl) mice crossed to villin-Cre mice). Although we find that knockout of Nedd4 on its own does not cause tumor growth, its knockout in the context of Apc+/min-derived colorectal tumors leads to augmentation of tumor growth, suggesting that Nedd4 normally suppresses intestinal WNT signaling and growth of colonic tumors. WNT signaling microarray, immunoblotting and immunohistochemistry analyses of tumors derived from the Villin-Cre;Nedd4fl/fl;Apc+/min colons demonstrated elevated expression of the WNT upstream effectors LEF1 (full length) and YY1 in these tumors relative to control (Apc+/min alone) tumors. Together, these results suggest that Nedd4 suppresses colonic WNT signaling and tumor growth, at least in part, by suppressing the transcription factors LEF1 and YY1.
Collapse
|
33
|
Gajos-Michniewicz A, Czyz M. Modulation of WNT/β-catenin pathway in melanoma by biologically active components derived from plants. Fitoterapia 2016; 109:283-92. [DOI: 10.1016/j.fitote.2016.02.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 01/28/2016] [Accepted: 02/01/2016] [Indexed: 01/06/2023]
|
34
|
FRIZZLED7 Is Required for Tumor Initiation and Metastatic Growth of Melanoma Cells. PLoS One 2016; 11:e0147638. [PMID: 26808375 PMCID: PMC4726610 DOI: 10.1371/journal.pone.0147638] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 01/05/2016] [Indexed: 01/28/2023] Open
Abstract
Metastases are thought to arise from cancer stem cells and their tumor initiating abilities are required for the establishment of metastases. Nevertheless, in metastatic melanoma, the nature of cancer stem cells is under debate and their contribution to metastasis formation remains unknown. Using an experimental metastasis model, we discovered that high levels of the WNT receptor, FZD7, correlated with enhanced metastatic potentials of melanoma cell lines. Knocking down of FZD7 in a panel of four melanoma cell lines led to a significant reduction in lung metastases in animal models, arguing that FZD7 plays a causal role during metastasis formation. Notably, limiting dilution analyses revealed that FZD7 is essential for the tumor initiation of melanoma cells and FZD7 knockdown impeded the early expansion of metastatic melanoma cells shortly after seeding, in accordance with the view that tumor initiating ability of cancer cells is required for metastasis formation. FZD7 activated JNK in melanoma cell lines in vitro and the expression of a dominant negative JNK suppressed metastasis formation in vivo, suggesting that FZD7 may promote metastatic growth of melanoma cells via activation of JNK. Taken together, our findings uncovered a signaling pathway that regulates the tumor initiation of melanoma cells and contributes to metastasis formation in melanoma.
Collapse
|
35
|
Ferretti R, Bhutkar A, McNamara MC, Lees JA. BMI1 induces an invasive signature in melanoma that promotes metastasis and chemoresistance. Genes Dev 2016; 30:18-33. [PMID: 26679841 PMCID: PMC4701976 DOI: 10.1101/gad.267757.115] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 11/18/2015] [Indexed: 01/17/2023]
Abstract
Melanoma can switch between proliferative and invasive states, which have identifying gene expression signatures that correlate with good and poor prognosis, respectively. However, the mechanisms controlling these signatures are poorly understood. In this study, we identify BMI1 as a key determinant of melanoma metastasis by which its overexpression enhanced and its deletion impaired dissemination. Remarkably, in this tumor type, BMI1 had no effect on proliferation or primary tumor growth but enhanced every step of the metastatic cascade. Consistent with the broad spectrum of effects, BMI1 activated widespread gene expression changes, which are characteristic of melanoma progression and also chemoresistance. Accordingly, we showed that up-regulation or down-regulation of BMI1 induced resistance or sensitivity to BRAF inhibitor treatment and that induction of noncanonical Wnt by BMI1 is required for this resistance. Finally, we showed that our BMI1-induced gene signature encompasses all of the hallmarks of the previously described melanoma invasive signature. Moreover, our signature is predictive of poor prognosis in human melanoma and is able to identify primary tumors that are likely to become metastatic. These data yield key insights into melanoma biology and establish BMI1 as a compelling drug target whose inhibition would suppress both metastasis and chemoresistance of melanoma.
Collapse
Affiliation(s)
- Roberta Ferretti
- David H. Koch Institute for Integrative Cancer Research, Cambridge, Massachusetts 02139, USA
| | - Arjun Bhutkar
- David H. Koch Institute for Integrative Cancer Research, Cambridge, Massachusetts 02139, USA
| | - Molly C McNamara
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Jacqueline A Lees
- David H. Koch Institute for Integrative Cancer Research, Cambridge, Massachusetts 02139, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| |
Collapse
|
36
|
Vaid M, Singh T, Prasad R, Katiyar SK. Bioactive proanthocyanidins inhibit growth and induce apoptosis in human melanoma cells by decreasing the accumulation of β-catenin. Int J Oncol 2015; 48:624-34. [PMID: 26676402 PMCID: PMC4725457 DOI: 10.3892/ijo.2015.3286] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 10/23/2015] [Indexed: 11/22/2022] Open
Abstract
Melanoma is a highly aggressive form of skin cancer with poor survival rate. Aberrant activation of Wnt/β-catenin has been observed in nearly one-third of human melanoma cases thereby indicating that targeting Wnt/β-catenin signaling could be a promising strategy against melanoma development. In the present study, we determined chemotherapeutic effect of grape seed proanthocyanidins (GSPs) on the growth of melanoma cells and validated their protective effects in vivo using a xenograft mouse model, and assessed if β-catenin is the target of GSP chemotherapeutic effect. Our in vitro data show that treatment of A375 and Hs294t human melanoma cells with GSPs inhibit the growth of melanoma cells, which was associated with the reduction in the levels of β-catenin. Administration of dietary GSPs (0.2 and 0.5%, w/w) in supplementation with AIN76A control diet significantly inhibited the growth of melanoma tumor xenografts in nude mice. Furthermore, dietary GSPs inhibited the xenograft growth of Mel928 (β-catenin-activated), while did not inhibit the xenograft growth of Mel1011 (β-catenin-inactivated) cells. These observations were further verified by siRNA knockdown of β-catenin and forced overexpression of β-catenin in melanoma cells using a cell culture model.
Collapse
Affiliation(s)
- Mudit Vaid
- Department of Dermatology, University of Alabama at Birmingham, AL 35294, USA
| | - Tripti Singh
- Department of Dermatology, University of Alabama at Birmingham, AL 35294, USA
| | - Ram Prasad
- Department of Dermatology, University of Alabama at Birmingham, AL 35294, USA
| | - Santosh K Katiyar
- Department of Dermatology, University of Alabama at Birmingham, AL 35294, USA
| |
Collapse
|
37
|
Freeman J, Smith D, Latinkic B, Ewan K, Samuel L, Zollo M, Marino N, Tyas L, Jones N, Dale TC. A functional connectome: regulation of Wnt/TCF-dependent transcription by pairs of pathway activators. Mol Cancer 2015; 14:206. [PMID: 26643252 PMCID: PMC4672529 DOI: 10.1186/s12943-015-0475-1] [Citation(s) in RCA: 13] [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: 07/20/2015] [Accepted: 11/23/2015] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Wnt/β-catenin signaling is often portrayed as a simple pathway that is initiated by Wnt ligand at the cell surface leading, via linear series of interactions between 'core pathway' members, to the induction of nuclear transcription from genes flanked by β-catenin/TCF transcription factor binding sites. Wnt/β-catenin signaling is also regulated by a much larger set of 'non-core regulators'. However the relationship between 'non-core regulators' is currently not well understood. Aberrant activation of the pathway has been shown to drive tumorgenesis in a number of different tissues. METHODS Mammalian cells engineered to have a partially-active level of Wnt/β-catenin signaling were screened by transfection for proteins that up or down-regulated a mid-level of TCF-dependent transcription induced by transient expression of an activated LRP6 Wnt co-receptor (∆NLRP). RESULTS 141 novel regulators of TCF-dependent transcription were identified. Surprisingly, when tested without ∆NLRP activation, most up-regulators failed to alter TCF-dependent transcription. However, when expressed in pairs, 27 % (466/1170) functionally interacted to alter levels of TCF-dependent transcription. When proteins were displayed as nodes connected by their ability to co-operate in the regulation of TCF-dependent transcription, a network of functional interactions was revealed. In this network, 'core pathway' components (Eg. β-catenin, GSK-3, Dsh) were found to be the most highly connected nodes. Activation of different nodes in this network impacted on the sensitivity to Wnt pathway small molecule antagonists. CONCLUSIONS The 'functional connectome' identified here strongly supports an alternative model of the Wnt pathway as a complex context-dependent network. The network further suggests that mutational activation of highly connected Wnt signaling nodes predisposed cells to further context-dependent alterations in levels of TCF-dependent transcription that may be important during tumor progression and treatment.
Collapse
Affiliation(s)
- Jamie Freeman
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff, CF10 3AX, Wales, UK
| | - David Smith
- Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, Keppel Street, London, WC1E 7HT, UK
| | - Branko Latinkic
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff, CF10 3AX, Wales, UK
| | - Ken Ewan
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff, CF10 3AX, Wales, UK
| | - Lee Samuel
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff, CF10 3AX, Wales, UK
| | - Massimo Zollo
- Department of Molecular Medicine and Biotechnology and Centro di Ingegneria Genetica e Biotecnologia Avanzate, Federico II, Via Pansini 5, 80131, Naples, Italy
| | - Natascia Marino
- Department of Molecular Medicine and Biotechnology and Centro di Ingegneria Genetica e Biotecnologia Avanzate, Federico II, Via Pansini 5, 80131, Naples, Italy
| | - Lorraine Tyas
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff, CF10 3AX, Wales, UK
| | - Nick Jones
- Department of Mathematics, Imperial College, London, SW7 2AZ, UK
| | - Trevor C Dale
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff, CF10 3AX, Wales, UK.
| |
Collapse
|
38
|
Zigmond E, Ya’acov AB, Lee H, Lichtenstein Y, Shalev Z, Smith Y, Zolotarov L, Ziv E, Kalman R, Le HV, Lu H, Silverman RB, Ilan Y. Suppression of Hepatocellular Carcinoma by Inhibition of Overexpressed Ornithine Aminotransferase. ACS Med Chem Lett 2015; 6:840-4. [PMID: 26288681 DOI: 10.1021/acsmedchemlett.5b00153] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2015] [Accepted: 05/29/2015] [Indexed: 12/24/2022] Open
Abstract
Hepatocellular carcinoma is the second leading cause of cancer death worldwide. DNA microarray analysis identified the ornithine aminotransferase (OAT) gene as a prominent gene overexpressed in hepatocellular carcinoma (HCC) from Psammomys obesus. In vitro studies demonstrated inactivation of OAT by gabaculine (1), a neurotoxic natural product, which suppressed in vitro proliferation of two HCC cell lines. Alpha-fetoprotein (AFP) secretion, a biomarker for HCC, was suppressed by gabaculine in both cell lines, but not significantly. Because of the active site similarity between GABA aminotransferase (GABA-AT) and OAT, a library of 24 GABA-AT inhibitors was screened to identify a more selective inhibitor of OAT. (1S,3S)-3-Amino-4-(hexafluoropropan-2-ylidene)cyclopentane-1-carboxylic acid (2) was found to be an inactivator of OAT that only weakly inhibits GABA-AT, l-aspartate aminotransferase, and l-alanine aminotransferase. In vitro administration of 2 significantly suppressed AFP secretion in both Hep3B and HepG2 HCC cells; in vivo, 2 significantly suppressed AFP serum levels and tumor growth in HCC-harboring mice, even at 0.1 mg/kg. Overexpression of the OAT gene in HCC and the ability to block the growth of HCC by OAT inhibitors support the role of OAT as a potential therapeutic target to inhibit HCC growth. This is the first demonstration of suppression of HCC by an OAT inactivator.
Collapse
Affiliation(s)
| | | | - Hyunbeom Lee
- Department of Chemistry,
Department of Molecular Biosciences, Chemistry of Life Processes Institute,
and Center for Molecular Innovation and Drug Discovery, Northwestern University, Evanston, Illinois 60208, United States
| | | | | | | | | | | | | | - Hoang V. Le
- Department of Chemistry,
Department of Molecular Biosciences, Chemistry of Life Processes Institute,
and Center for Molecular Innovation and Drug Discovery, Northwestern University, Evanston, Illinois 60208, United States
| | - Hejun Lu
- Department of Chemistry,
Department of Molecular Biosciences, Chemistry of Life Processes Institute,
and Center for Molecular Innovation and Drug Discovery, Northwestern University, Evanston, Illinois 60208, United States
| | - Richard B. Silverman
- Department of Chemistry,
Department of Molecular Biosciences, Chemistry of Life Processes Institute,
and Center for Molecular Innovation and Drug Discovery, Northwestern University, Evanston, Illinois 60208, United States
| | | |
Collapse
|
39
|
Bongiovanni L, D'Andrea A, Porcellato I, Ciccarelli A, Malatesta D, Romanucci M, Della Salda L, Mechelli L, Brachelente C. Canine cutaneous melanocytic tumours: significance of β-catenin and survivin immunohistochemical expression. Vet Dermatol 2015; 26:270-e59. [DOI: 10.1111/vde.12211] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/02/2015] [Indexed: 12/13/2022]
Affiliation(s)
- Laura Bongiovanni
- Faculty of Veterinary Medicine; University of Teramo; Piazza A. Moro 45 Teramo 64100 Italy
| | - Alessandra D'Andrea
- Faculty of Veterinary Medicine; University of Teramo; Piazza A. Moro 45 Teramo 64100 Italy
| | - Ilaria Porcellato
- Department of Veterinary Medicine; University of Perugia; Via San Costanzo 4 Perugia 06126 Italy
| | - Andrea Ciccarelli
- Faculty of Political Science; University of Teramo; Campus Coste Sant'Agostino Teramo 64100 Italy
| | - Daniela Malatesta
- Faculty of Veterinary Medicine; University of Teramo; Piazza A. Moro 45 Teramo 64100 Italy
| | - Mariarita Romanucci
- Faculty of Veterinary Medicine; University of Teramo; Piazza A. Moro 45 Teramo 64100 Italy
| | - Leonardo Della Salda
- Faculty of Veterinary Medicine; University of Teramo; Piazza A. Moro 45 Teramo 64100 Italy
| | - Luca Mechelli
- Department of Veterinary Medicine; University of Perugia; Via San Costanzo 4 Perugia 06126 Italy
| | - Chiara Brachelente
- Department of Veterinary Medicine; University of Perugia; Via San Costanzo 4 Perugia 06126 Italy
| |
Collapse
|
40
|
Atkinson JM, Rank KB, Zeng Y, Capen A, Yadav V, Manro JR, Engler TA, Chedid M. Activating the Wnt/β-Catenin Pathway for the Treatment of Melanoma--Application of LY2090314, a Novel Selective Inhibitor of Glycogen Synthase Kinase-3. PLoS One 2015; 10:e0125028. [PMID: 25915038 PMCID: PMC4411090 DOI: 10.1371/journal.pone.0125028] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 03/19/2015] [Indexed: 12/12/2022] Open
Abstract
It has previously been observed that a loss of β-catenin expression occurs with melanoma progression and that nuclear β-catenin levels are inversely proportional to cellular proliferation, suggesting that activation of the Wnt/β-catenin pathway may provide benefit for melanoma patients. In order to further probe this concept we tested LY2090314, a potent and selective small-molecule inhibitor with activity against GSK3α and GSK3β isoforms. In a panel of melanoma cell lines, nM concentrations of LY2090314 stimulated TCF/LEF TOPFlash reporter activity, stabilized β-catenin and elevated the expression of Axin2, a Wnt responsive gene and marker of pathway activation. Cytotoxicity assays revealed that melanoma cell lines are very sensitive to LY2090314 in vitro (IC50 ~10 nM after 72hr of treatment) in contrast to other solid tumor cell lines (IC50 >10 uM) as evidenced by caspase activation and PARP cleavage. Cell lines harboring mutant B-RAF or N-RAS were equally sensitive to LY2090314 as were those with acquired resistance to the BRAF inhibitor Vemurafenib. shRNA studies demonstrated that β-catenin stabilization is required for apoptosis following treatment with the GSK3 inhibitor since the sensitivity of melanoma cell lines to LY290314 could be overcome by β-catenin knockdown. We further demonstrate that in vivo, LY2090314 elevates Axin2 gene expression after a single dose and produces tumor growth delay in A375 melanoma xenografts with repeat dosing. The activity of LY2090314 in preclinical models suggests that the role of Wnt activators for the treatment of melanoma should be further explored.
Collapse
Affiliation(s)
- Jennifer M. Atkinson
- Oncology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, 46285, United States of America
| | - Kenneth B. Rank
- Oncology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, 46285, United States of America
| | - Yi Zeng
- Oncology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, 46285, United States of America
| | - Andrew Capen
- Oncology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, 46285, United States of America
| | - Vipin Yadav
- Oncology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, 46285, United States of America
| | - Jason R. Manro
- Statistics Discovery, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, 46285, United States of America
| | - Thomas A. Engler
- Discovery Chemistry Research and Technologies, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, 46285, United States of America
| | - Marcio Chedid
- Oncology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, 46285, United States of America
- * E-mail:
| |
Collapse
|
41
|
MITF drives endolysosomal biogenesis and potentiates Wnt signaling in melanoma cells. Proc Natl Acad Sci U S A 2015; 112:E420-9. [PMID: 25605940 DOI: 10.1073/pnas.1424576112] [Citation(s) in RCA: 184] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Canonical Wnt signaling plays an important role in development and disease, regulating transcription of target genes and stabilizing many proteins phosphorylated by glycogen synthase kinase 3 (GSK3). We observed that the MiT family of transcription factors, which includes the melanoma oncogene MITF (micropthalmia-associated transcription factor) and the lysosomal master regulator TFEB, had the highest phylogenetic conservation of three consecutive putative GSK3 phosphorylation sites in animal proteomes. This finding prompted us to examine the relationship between MITF, endolysosomal biogenesis, and Wnt signaling. Here we report that MITF expression levels correlated with the expression of a large subset of lysosomal genes in melanoma cell lines. MITF expression in the tetracycline-inducible C32 melanoma model caused a marked increase in vesicular structures, and increased expression of late endosomal proteins, such as Rab7, LAMP1, and CD63. These late endosomes were not functional lysosomes as they were less active in proteolysis, yet were able to concentrate Axin1, phospho-LRP6, phospho-β-catenin, and GSK3 in the presence of Wnt ligands. This relocalization significantly enhanced Wnt signaling by increasing the number of multivesicular bodies into which the Wnt signalosome/destruction complex becomes localized upon Wnt signaling. We also show that the MITF protein was stabilized by Wnt signaling, through the novel C-terminal GSK3 phosphorylations identified here. MITF stabilization caused an increase in multivesicular body biosynthesis, which in turn increased Wnt signaling, generating a positive-feedback loop that may function during the proliferative stages of melanoma. The results underscore the importance of misregulated endolysosomal biogenesis in Wnt signaling and cancer.
Collapse
|
42
|
Epithelial Notch signaling is a limiting step for pancreatic carcinogenesis. BMC Cancer 2014; 14:862. [PMID: 25416148 PMCID: PMC4289235 DOI: 10.1186/1471-2407-14-862] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 11/13/2014] [Indexed: 11/21/2022] Open
Abstract
Background Pancreatic cancer is one of the deadliest human malignancies, with few therapeutic options. Re-activation of embryonic signaling pathways is commonly in human pancreatic cancer and provided rationale to explore inhibition of these pathways therapeutically. Notch signaling is important during pancreatic development, and it is re-activated in pancreatic cancer. The functional role of Notch signaling during pancreatic carcinogenesis has been previously characterized using both genetic and drug-based approaches. However, contrasting findings were reported based on the study design. In fact, Notch signaling has been proposed to act as tumor-promoter or tumor-suppressor. Given the availability of Notch inhibitors in the clinic, understanding how this signaling pathway contributes to pancreatic carcinogenesis has important therapeutic implications. Here, we interrogated the role of Notch signaling specifically in the epithelial compartment of the pancreas, in the context of a genetically engineered mouse model of pancreatic cancer. Methods To inhibit Notch signaling in the pancreas epithelium, we crossed a mouse model of pancreatic cancer based on pancreas-specific expression of mutant Kras with a transgenic mouse that conditionally expresses a dominant negative form of the Mastermind-like 1 gene. MAML is an essential co-activator of the canonical Notch signaling-mediated transcription. DNMAML encodes a truncated MAML protein that represses all canonical Notch mediated transcription in a cell autonomous manner, independent of which Notch receptor is activated. As a result, in mice co-expressing mutant Kras and DNMAML, Notch signaling is inhibited specifically in the epithelium upon Cre-mediated recombination. We explored the effect of epithelial-specific DNMAML expression on Kras-driven carcinogenesis both during normal aging and following the induction of acute pancreatitis. Results We find that DNMAML expression efficiently inhibits epithelial Notch signaling and delays PanIN formation. However, over time, loss of Notch inhibition allows PanIN formation and progression. Conclusions Epithelial-specific Notch signaling is important for PanIN initiation. Our findings indicate that PanIN formation can only occur upon loss of epithelial Notch inhibition, thus supporting an essential role of this signaling pathway during pancreatic carcinogenesis. Electronic supplementary material The online version of this article (doi:10.1186/1471-2407-14-862) contains supplementary material, which is available to authorized users.
Collapse
|
43
|
Biological functions of casein kinase 1 isoforms and putative roles in tumorigenesis. Mol Cancer 2014; 13:231. [PMID: 25306547 PMCID: PMC4201705 DOI: 10.1186/1476-4598-13-231] [Citation(s) in RCA: 166] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Accepted: 09/26/2014] [Indexed: 12/13/2022] Open
Abstract
Isoforms of the casein kinase 1 (CK1) family have been shown to phosphorylate key regulatory molecules involved in cell cycle, transcription and translation, the structure of the cytoskeleton, cell-cell adhesion and receptor-coupled signal transduction. They regulate key signaling pathways known to be critically involved in tumor progression. Recent results point to an altered expression or activity of different CK1 isoforms in tumor cells. This review summarizes the expression and biological function of CK1 family members in normal and malignant cells and the evidence obtained so far about their role in tumorigenesis.
Collapse
|
44
|
Salaroli R, Ronchi A, Buttarelli FR, Cortesi F, Marchese V, Della Bella E, Renna C, Baldi C, Giangaspero F, Cenacchi G. Wnt activation affects proliferation, invasiveness and radiosensitivity in medulloblastoma. J Neurooncol 2014; 121:119-27. [PMID: 25261924 DOI: 10.1007/s11060-014-1621-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2013] [Accepted: 09/21/2014] [Indexed: 12/26/2022]
Abstract
Medulloblastomas (MBs) associated with the Wnt activation represent a subgroup with a favorable prognosis, but it remains unclear whether Wnt activation confers a less aggressive phenotype and/or enhances radiosensitivity. To investigate this issue, we evaluated the biological behavior of an MB cell line, UW228-1, stably transfected with human β-catenin cDNA encoding a nondegradable form of β-catenin (UW-B) in standard culture conditions and after radiation treatment. We evaluated the expression, transcriptional activity, and localization of β-catenin in the stably transfected cells using immunofluorescence and WB. We performed morphological analysis using light and electron microscopy. We then analyzed changes in the invasiveness, growth, and mortality in standard culture conditions and after radiation. We demonstrated that (A) Wnt activation inhibited 97 % of the invasion capability of the cells, (B) the growth of the UW-B cells was statistically significantly lower than that of all the other control cells (p < 0.01), (C) the mortality of irradiated UW-B cells was statistically significantly higher than that of the controls and their nonirradiated counterparts (p < 0.05), and (D) morphological features of neuronal differentiation were observed in the Wnt-activated cells. In tissue samples, the Ki-67 labeling index (LI) was lower in β-catenin-positive samples compared to non-β-catenin positive ones. The Ki-67 LI median (LI = 40) of the nuclear β-catenin-positive tumor samples was lower than that of non-nuclear β-catenin-positive samples (LI = 50), but the difference was not statistically significant. Overall, our data suggest that activation of the Wnt pathway reduces the proliferation and invasion of MBs and increases the tumor's radiosensitivity.
Collapse
Affiliation(s)
- Roberta Salaroli
- Department of Biomedical and Neuromotor Sciences, "Alma Mater Studiorum" University of Bologna, Via Massarenti 9, 40138, Bologna, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Fan C, Jiang G, Zhang X, Miao Y, Lin X, Luan L, Xu Z, Zhang Y, Zhao H, Liu D, Wang E. Zbed3 contributes to malignant phenotype of lung cancer via regulating β-catenin and P120-catenin 1. Mol Carcinog 2014; 54 Suppl 1:E138-47. [PMID: 25263389 DOI: 10.1002/mc.22216] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Revised: 07/02/2014] [Accepted: 07/30/2014] [Indexed: 12/19/2022]
Affiliation(s)
- Chuifeng Fan
- Department of Pathology; First Affiliated Hospital and College of Basic Medical Sciences of China Medical University; Shenyang China
- Institute of Pathology and Pathophysiology; China Medical University; Shenyang China
| | - Guiyang Jiang
- Department of Pathology; First Affiliated Hospital and College of Basic Medical Sciences of China Medical University; Shenyang China
- Institute of Pathology and Pathophysiology; China Medical University; Shenyang China
| | - Xiupeng Zhang
- Department of Pathology; First Affiliated Hospital and College of Basic Medical Sciences of China Medical University; Shenyang China
- Institute of Pathology and Pathophysiology; China Medical University; Shenyang China
| | - Yuan Miao
- Department of Pathology; First Affiliated Hospital and College of Basic Medical Sciences of China Medical University; Shenyang China
- Institute of Pathology and Pathophysiology; China Medical University; Shenyang China
| | - Xuyong Lin
- Department of Pathology; First Affiliated Hospital and College of Basic Medical Sciences of China Medical University; Shenyang China
- Institute of Pathology and Pathophysiology; China Medical University; Shenyang China
| | - Lan Luan
- Department of Pathology; First Affiliated Hospital and College of Basic Medical Sciences of China Medical University; Shenyang China
- Institute of Pathology and Pathophysiology; China Medical University; Shenyang China
| | - Zhonghai Xu
- Department of Pathology; First Affiliated Hospital and College of Basic Medical Sciences of China Medical University; Shenyang China
- Institute of Pathology and Pathophysiology; China Medical University; Shenyang China
| | - Yijun Zhang
- Department of Pathology; First Affiliated Hospital and College of Basic Medical Sciences of China Medical University; Shenyang China
- Institute of Pathology and Pathophysiology; China Medical University; Shenyang China
| | - Huanyu Zhao
- Department of Pathology; First Affiliated Hospital and College of Basic Medical Sciences of China Medical University; Shenyang China
- Institute of Pathology and Pathophysiology; China Medical University; Shenyang China
| | - Di Liu
- Department of Pathology; First Affiliated Hospital and College of Basic Medical Sciences of China Medical University; Shenyang China
- Institute of Pathology and Pathophysiology; China Medical University; Shenyang China
| | - Enhua Wang
- Department of Pathology; First Affiliated Hospital and College of Basic Medical Sciences of China Medical University; Shenyang China
- Institute of Pathology and Pathophysiology; China Medical University; Shenyang China
| |
Collapse
|
46
|
Qi XH, Wu D, Cui HX, Ma N, Su J, Wang YT, Jiang YH. Silencing of the glypican-3 gene affects the biological behavior of human hepatocellular carcinoma cells. Mol Med Rep 2014; 10:3177-84. [PMID: 25270552 DOI: 10.3892/mmr.2014.2600] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Accepted: 06/05/2014] [Indexed: 11/05/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the leading causes of cancer death in the world. The gene glypican-3 (GPC3) is reported to be a potential therapeutic target for HCC. In this study, we use RNA interference with lentiviral vectors to explore the effect of GPC3 silencing on the biological behavior of HCC cells and the potential role of the GPC3 protein in the activation of epithelial-mesenchymal transition (EMT), which relates to HCC cell invasion and migration. Our data suggest that GPC3 silencing leads to a decrease in HCC cell proliferation and to an increase in apoptosis. We demonstrated that GPC3 silencing regulates cell invasion and migration, most probably through the activation of the EMT cellular program. In conclusion, GPC3 is associated with the HCC cell biological behavior, while the relationship between GPC3 and EMT in tumorigenesis of HCC deserves future investigation.
Collapse
Affiliation(s)
- Xin-Hui Qi
- Cancer Research Institute, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Di Wu
- Cancer Research Institute, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Hui-Xia Cui
- Cancer Research Institute, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Nan Ma
- Cancer Research Institute, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Jia Su
- Cancer Research Institute, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Yu-Tong Wang
- Cancer Research Institute, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - You-Hong Jiang
- Cancer Research Institute, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
47
|
Serio RN. Wnt of the Two Horizons: Putting Stem Cell Self-Renewal and Cell Fate Determination into Context. Stem Cells Dev 2014; 23:1975-90. [DOI: 10.1089/scd.2014.0055] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Affiliation(s)
- Ryan N. Serio
- Graduate School of Pharmacology, Weill Cornell Medical College, New York, New York
| |
Collapse
|
48
|
Developmental pathways activated in melanocytes and melanoma. Arch Biochem Biophys 2014; 563:13-21. [PMID: 25109840 DOI: 10.1016/j.abb.2014.07.023] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 07/14/2014] [Accepted: 07/17/2014] [Indexed: 12/25/2022]
Abstract
Cutaneous malignant melanomas originate primarily within epidermal melanocytic cells. Melanoma cells share many characteristics with melanocyte precursors, suggesting that melanoma cells utilize the developmental programs of their normal counterpart for their own progression. The pigmentation system provides an advantageous model to assess survival pathway interactions in the melanocytic lineage, as genetic alterations controlling melanocyte development can be easily detectable by coat color phenotype that do not affect the viability of an animal. By integrating combinatorial gene knockout approaches, cell-based assays and immunohistochemical observations, recent studies have illustrated several genes and pathways that play important roles both in melanocyte specification and maintenance and in melanoma formation and progression. We are reviewing those genes and pathways to understand the connection between normal and cancerous development and to reveal therapeutic potential of targeting developmental pathways for melanoma therapy.
Collapse
|
49
|
Rochefort GY. The osteocyte as a therapeutic target in the treatment of osteoporosis. Ther Adv Musculoskelet Dis 2014; 6:79-91. [PMID: 24891879 DOI: 10.1177/1759720x14523500] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Osteoporosis is characterized by a low bone-mineral density associated with skeletal fractures. The decrease in bone-mineral density is the consequence of an unbalanced bone-remodeling process, with higher bone resorption than bone formation. The orchestration of the bone-remodeling process is under the control of the most abundant cell in bone, the osteocyte. Functioning as an endocrine cell, osteocytes are also a source of soluble factors that not only target cells on the bone surface, but also target distant organs. Therefore, any drugs targeting the osteocyte functions and signaling pathways will have a major impact on the bone-remodeling process. This review discusses potential advances in drug therapy for osteoporosis, including novel osteocyte-related antiresorptive and anabolic agents that may become available in the coming years.
Collapse
Affiliation(s)
- Gaël Y Rochefort
- EA 2496, Faculté de Chirurgie Dentaire, Université Paris Descartes, 1 rue Maurice Arnoux, 92120 Montrouge, France
| |
Collapse
|
50
|
Melanoma Development and Progression Are Associated with Rad6 Upregulation and β -Catenin Relocation to the Cell Membrane. J Skin Cancer 2014; 2014:439205. [PMID: 24891954 PMCID: PMC4033428 DOI: 10.1155/2014/439205] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Revised: 04/02/2014] [Accepted: 04/15/2014] [Indexed: 12/21/2022] Open
Abstract
We have previously demonstrated that Rad6 and β -catenin enhance each other's expression through a positive feedback loop to promote breast cancer development/progression. While β -catenin has been implicated in melanoma pathogenesis, Rad6 function has not been investigated. Here, we examined the relationship between Rad6 and β -catenin in melanoma development and progression. Eighty-eight cutaneous tumors, 30 nevi, 29 primary melanoma, and 29 metastatic melanomas, were immunostained with anti- β -catenin and anti-Rad6 antibodies. Strong expression of Rad6 was observed in only 27% of nevi as compared to 100% of primary and 96% of metastatic melanomas. β -Catenin was strongly expressed in 97% of primary and 93% of metastatic melanomas, and unlike Rad6, in 93% of nevi. None of the tumors expressed nuclear β -catenin. β -Catenin was exclusively localized on the cell membrane of 55% of primary, 62% of metastatic melanomas, and only 10% of nevi. Cytoplasmic β -catenin was detected in 90% of nevi, 17% of primary, and 8% of metastatic melanoma, whereas 28% of primary and 30% of metastatic melanomas exhibited β -catenin at both locations. These data suggest that melanoma development and progression are associated with Rad6 upregulation and membranous redistribution of β -catenin and that β -catenin and Rad6 play independent roles in melanoma development.
Collapse
|