1
|
Hu Y. Hypoxia-induced S-phase kinase-interacting protein 2 knockdown repressed the progression of melanoma through extracellular signal-regulated kinase 1/2 pathway. Cytojournal 2025; 22:9. [PMID: 39958883 PMCID: PMC11829309 DOI: 10.25259/cytojournal_117_2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 12/19/2024] [Indexed: 02/18/2025] Open
Abstract
Objective Hypoxia intensely drives the development of malignant tumors, including skin cutaneous melanoma (SKCM). S-phase kinase-interacting protein 2 (SKP2) is known to participate in the progression of human tumors. The purpose of this study is to explore whether SKP2 acts as a hypoxic response gene during SKCM progression. Material and Methods SKP2 expression in SKCM tissues was analyzed using The Cancer Genome Atlas database. Anoxic experiments were conducted to simulate an anoxic environment. 5-Ethynyl-2'-deoxyuridine and colony formation assays were used to evaluate SKCM cell growth. Scratch healing and Transwell assays were applied to measure the migration and invasion abilities of SKCM cells. An immunoblotting assay was used to detect the levels of extracellular signal-regulated kinase (ERK)1/2 pathway proteins. In addition, the ERK-specific agonist LM22B-10 was added to confirm whether the ERK1/2 signaling pathway is required for SKP2-mediated SKCM progression under hypoxic conditions. Results SKP2 was significantly upregulated in SKCM tissues and closely related to adverse outcomes in patients. Moreover, SKP2 levels increased in SKCM cells under normoxic conditions and further elevated under hypoxic conditions. SKP2 deficiency led to the reduced proliferation, migration, and invasion potential of cells under hypoxic conditions. Mechanically, SKP2 silencing blocked the ERK1/2 pathway in hypoxic cells, and the activation of the ERK1/2 pathway rescued the suppression effect of SKP2 on the hypoxia-induced progression of SKCM. Conclusion SKP2 deficiency repressed the hypoxic-induced progression of SKCM through the ERK1/2 pathway. This novel discovery regarding the SKP2/ERK1/2 axis might provide new insights into the pathogenesis of SKCM.
Collapse
Affiliation(s)
- Yong Hu
- Department of Dermatology and Venereal Diseases, Yihe Women’s and Children’s Hospital, Chaoyang District, Beijing, China
| |
Collapse
|
2
|
Moralev A, Zenkova MA, Markov AV. Complex Inhibitory Activity of Pentacyclic Triterpenoids against Cutaneous Melanoma In Vitro and In Vivo: A Literature Review and Reconstruction of Their Melanoma-Related Protein Interactome. ACS Pharmacol Transl Sci 2024; 7:3358-3384. [PMID: 39539268 PMCID: PMC11555519 DOI: 10.1021/acsptsci.4c00422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 09/13/2024] [Accepted: 10/14/2024] [Indexed: 11/16/2024]
Abstract
Pentacyclic triterpenoids (PTs) are a class of plant metabolites with a wide range of pharmacological activities, including strong antitumor potential against skin malignancies. By acting on multiple signaling pathways that control key cellular processes, PTs are able to exert complex effects on melanoma progression in vitro and in vivo. In this review, we have analyzed the works published in the past decade and devoted to the effects of PTs, both natural and semisynthetic, on cutaneous melanoma pathogenesis, including not only their direct action on melanoma cells but also their influence on the tumor microenvironment and abberant melanogenesis, often associated with melanoma aggressiveness. Special attention will be paid to the molecular basis of the pronounced antimelanoma potency of PTs, including a detailed consideration of the pathways sensitive to PTs in melanoma cells, as well as the reconstruction of the melanoma-related protein interactome of PTs using a network pharmacology approach based on previously published experimentally verified protein targets of PTs. The information collected on the primary targets of PTs was compiled in the Protein Interactome of PTs (PIPTs) database, freely available at http://www.pipts-db.ru/, which can be used to further optimize the mechanistic studies of PTs in the context of melanoma and other malignancies. By summarizing recent research findings, this review provides valuable information to scientists working in the fields related to the evaluation of melanoma pathogenesis and development of PTs-based drug candidates.
Collapse
Affiliation(s)
- Arseny
D. Moralev
- Institute of Chemical Biology and Fundamental
Medicine, Siberian Branch of the Russian
Academy of Sciences, 630090, Lavrent’ev avenue 8, Novosibirsk, Russia
| | - Marina A. Zenkova
- Institute of Chemical Biology and Fundamental
Medicine, Siberian Branch of the Russian
Academy of Sciences, 630090, Lavrent’ev avenue 8, Novosibirsk, Russia
| | - Andrey V. Markov
- Institute of Chemical Biology and Fundamental
Medicine, Siberian Branch of the Russian
Academy of Sciences, 630090, Lavrent’ev avenue 8, Novosibirsk, Russia
| |
Collapse
|
3
|
Targeting the untargetable: RB1-deficient tumours are vulnerable to Skp2 ubiquitin ligase inhibition. Br J Cancer 2022; 127:969-975. [PMID: 35752713 PMCID: PMC9470583 DOI: 10.1038/s41416-022-01898-0] [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: 03/22/2022] [Revised: 06/08/2022] [Accepted: 06/14/2022] [Indexed: 11/08/2022] Open
Abstract
Proteins that regulate the cell cycle are accumulated and degraded in a coordinated manner during the transition from one cell cycle phase to the next. The rapid loss of a critical protein, for example, to allow the cell to move from G1/G0 to S phase, is often regulated by its ubiquitination and subsequent proteasomal degradation. Protein ubiquitination is mediated by a series of three ligases, of which the E3 ligases provide the specificity for a particular protein substrate. One such E3 ligase is SCFSkp1/Cks1, which has a substrate recruiting subunit called S-phase kinase-associated protein 2 (Skp2). Skp2 regulates cell proliferation, apoptosis, and differentiation, can act as an oncogene, and is overexpressed in human cancer. A primary target of Skp2 is the cyclin-dependent kinase inhibitor p27 (CDKN1b) that regulates the cell cycle at several points. The RB1 tumour suppressor gene regulates Skp2 activity by two mechanisms: by controlling its mRNA expression, and by an effect on Skp2's enzymatic activity. For the latter, the RB1 protein (pRb) directly binds to the substrate-binding site on Skp2, preventing protein substrates from being ubiquitinated and degraded. Inactivating mutations in RB1 are common in human cancer, becoming more frequent in aggressive, metastatic, and drug-resistant tumours. Hence, RB1 mutation leads to the loss of pRb, an unrestrained increase in Skp2 activity, the unregulated decrease in p27, and the loss of cell cycle control. Because RB1 mutations lead to the loss of a functional protein, its direct targeting is not possible. This perspective will discuss evidence validating Skp2 as a therapeutic target in RB1-deficient cancer.
Collapse
|
4
|
Thompson LL, Rutherford KA, Lepage CC, McManus KJ. The SCF Complex Is Essential to Maintain Genome and Chromosome Stability. Int J Mol Sci 2021; 22:8544. [PMID: 34445249 PMCID: PMC8395177 DOI: 10.3390/ijms22168544] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 07/29/2021] [Accepted: 08/05/2021] [Indexed: 12/20/2022] Open
Abstract
The SKP1, CUL1, F-box protein (SCF) complex encompasses a group of 69 SCF E3 ubiquitin ligase complexes that primarily modify protein substrates with poly-ubiquitin chains to target them for proteasomal degradation. These SCF complexes are distinguishable by variable F-box proteins, which determine substrate specificity. Although the function(s) of each individual SCF complex remain largely unknown, those that have been characterized regulate a wide array of cellular processes, including gene transcription and the cell cycle. In this regard, the SCF complex regulates transcription factors that modulate cell signaling and ensures timely degradation of primary cell cycle regulators for accurate replication and segregation of genetic material. SCF complex members are aberrantly expressed in a myriad of cancer types, with altered expression or function of the invariable core SCF components expected to have a greater impact on cancer pathogenesis than that of the F-box proteins. Accordingly, this review describes the normal roles that various SCF complexes have in maintaining genome stability before discussing the impact that aberrant SCF complex expression and/or function have on cancer pathogenesis. Further characterization of the SCF complex functions is essential to identify and develop therapeutic approaches to exploit aberrant SCF complex expression and function.
Collapse
Affiliation(s)
- Laura L. Thompson
- CancerCare Manitoba Research Institute, CancerCare Manitoba, Winnipeg, MB R3E 0V9, Canada; (L.L.T.); (K.A.R.); (C.C.L.)
- Department of Biochemistry & Medical Genetics, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Kailee A. Rutherford
- CancerCare Manitoba Research Institute, CancerCare Manitoba, Winnipeg, MB R3E 0V9, Canada; (L.L.T.); (K.A.R.); (C.C.L.)
- Department of Biochemistry & Medical Genetics, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Chloe C. Lepage
- CancerCare Manitoba Research Institute, CancerCare Manitoba, Winnipeg, MB R3E 0V9, Canada; (L.L.T.); (K.A.R.); (C.C.L.)
- Department of Biochemistry & Medical Genetics, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Kirk J. McManus
- CancerCare Manitoba Research Institute, CancerCare Manitoba, Winnipeg, MB R3E 0V9, Canada; (L.L.T.); (K.A.R.); (C.C.L.)
- Department of Biochemistry & Medical Genetics, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| |
Collapse
|
5
|
Soysouvanh F, Giuliano S, Habel N, El-Hachem N, Pisibon C, Bertolotto C, Ballotti R. An Update on the Role of Ubiquitination in Melanoma Development and Therapies. J Clin Med 2021; 10:jcm10051133. [PMID: 33800394 PMCID: PMC7962844 DOI: 10.3390/jcm10051133] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 02/24/2021] [Accepted: 02/25/2021] [Indexed: 12/27/2022] Open
Abstract
The ubiquitination system plays a critical role in regulation of large array of biological processes and its alteration has been involved in the pathogenesis of cancers, among them cutaneous melanoma, which is responsible for the most deaths from skin cancers. Over the last decades, targeted therapies and immunotherapies became the standard therapeutic strategies for advanced melanomas. However, despite these breakthroughs, the prognosis of metastatic melanoma patients remains unoptimistic, mainly due to intrinsic or acquired resistances. Many avenues of research have been investigated to find new therapeutic targets for improving patient outcomes. Because of the pleiotropic functions of ubiquitination, and because each step of ubiquitination is amenable to pharmacological targeting, much attention has been paid to the role of this process in melanoma development and resistance to therapies. In this review, we summarize the latest data on ubiquitination and discuss the possible impacts on melanoma treatments.
Collapse
Affiliation(s)
- Frédéric Soysouvanh
- Inserm U1065, C3M, Team 1, Biology, and Pathologies of Melanocytes, University of Nice Côte d’Azur, 06200 Nice, France; (F.S.); (S.G.); (N.H.); (C.P.); (C.B.)
| | - Serena Giuliano
- Inserm U1065, C3M, Team 1, Biology, and Pathologies of Melanocytes, University of Nice Côte d’Azur, 06200 Nice, France; (F.S.); (S.G.); (N.H.); (C.P.); (C.B.)
| | - Nadia Habel
- Inserm U1065, C3M, Team 1, Biology, and Pathologies of Melanocytes, University of Nice Côte d’Azur, 06200 Nice, France; (F.S.); (S.G.); (N.H.); (C.P.); (C.B.)
| | - Najla El-Hachem
- Laboratory of Cancer Signaling, University of Liège, 4020 Liège, Belgium;
| | - Céline Pisibon
- Inserm U1065, C3M, Team 1, Biology, and Pathologies of Melanocytes, University of Nice Côte d’Azur, 06200 Nice, France; (F.S.); (S.G.); (N.H.); (C.P.); (C.B.)
| | - Corine Bertolotto
- Inserm U1065, C3M, Team 1, Biology, and Pathologies of Melanocytes, University of Nice Côte d’Azur, 06200 Nice, France; (F.S.); (S.G.); (N.H.); (C.P.); (C.B.)
- Equipe labellisée Fondation ARC 2019, 06200 Nice, France
| | - Robert Ballotti
- Inserm U1065, C3M, Team 1, Biology, and Pathologies of Melanocytes, University of Nice Côte d’Azur, 06200 Nice, France; (F.S.); (S.G.); (N.H.); (C.P.); (C.B.)
- Equipe labellisée Ligue Contre le Cancer 2020, 06200 Nice, France
- Correspondence: ; Tel.: +33-4-89-06-43-32
| |
Collapse
|
6
|
Brożyna AA, Aplin A, Cohen C, Carlson G, Page AJ, Murphy M, Slominski AT, Carlson JA. CKS1 expression in melanocytic nevi and melanoma. Oncotarget 2018; 9:4173-4187. [PMID: 29423113 PMCID: PMC5790530 DOI: 10.18632/oncotarget.23648] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 12/16/2017] [Indexed: 12/22/2022] Open
Abstract
Cyclin-dependent kinase subunit 1 (Cks1) regulates the degradation of p27, an important G1-S inhibitor, which is up regulated by MAPK pathway activation. In this study, we sought to determine whether Cks1 expression is increased in melanocytic tumors and correlates with outcome and/or other clinicopathologic prognostic markers. Cks1 expression was assessed by immunohistochemistry in 298 melanocytic lesions. The frequency and intensity of cytoplasmic and nuclear expression was scored as a labeling index and correlated with clinico-pathological data. Nuclear Cks1 protein was found in 63% of melanocytic nevi, 89% primary and 90% metastatic melanomas with mean labeling index of 7 ± 16, 19 ± 20, and 30 ± 29, respectively. While cytoplasmic Cks1 was found in 41% of melanocytic nevi, 84% primary and 95% metastatic melanomas with mean labeling index of 18 ± 34, 35 ± 34, and 52 ± 34, accordingly. Histologic stepwise model of tumor progression, defined as progression from benign nevi to primary melanomas, to melanoma metastases, revealed a significant increase in nuclear and cytoplasmic Cks1 expression with tumor progression. Nuclear and cytoplasmic Cks1 expression correlated with the presence of ulceration, increased mitotic rate, Breslow depth, Clark level, tumor infiltrating lymphocytes and gender. However, other well-known prognostic factors (age, anatomic site, and regression) did not correlate with any type of Cks1 expression. Similarly, increasing nuclear expression of Cks1 significantly correlated with worse overall survival. Thus, Cks1 expression appears to play a role in the progression of melanoma, where high levels of expression are associated with poor outcome. Cytoplasmic expression of Cks1 might represent high turnover of protein via the ubiquination/proteosome pathway.
Collapse
Affiliation(s)
- Anna A Brożyna
- Department of Tumor Pathology and Pathomorphology, Faculty of Health Sciences, Nicolaus Copernicus University Collegium Medicum in Bydgoszcz, Oncology Centre - Prof. Franciszek Łukaszczyk Memorial Hospital, Bydgoszcz 85-796, Poland
| | - Andrew Aplin
- Department of Cancer Biology, BLSB 524, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Cynthia Cohen
- Winship Cancer Institute, Emory University Hospital, Atlanta, GA 30322, USA
| | - Grant Carlson
- Winship Cancer Institute, Emory University Hospital, Atlanta, GA 30322, USA
| | - Andrew Joseph Page
- Pancreas, Liver, and Cancer Surgery, Piedmont Healthcare, Atlanta, GA 30309, USA
| | - Michael Murphy
- Department of Dermatology, UConn Health, Farmington, CT 06030, USA
| | - Andrzej T Slominski
- Department of Dermatology, Comprehensive Cancer Center, Cancer Chemoprevention Program, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - J Andrew Carlson
- Department of Pathology and Laboratory Medicine, Albany Medical College MC-81, Albany, NY 12208, USA
| |
Collapse
|
7
|
Ma J, Guo W, Li C. Ubiquitination in melanoma pathogenesis and treatment. Cancer Med 2017; 6:1362-1377. [PMID: 28544818 PMCID: PMC5463089 DOI: 10.1002/cam4.1069] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Revised: 03/07/2017] [Accepted: 03/10/2017] [Indexed: 12/13/2022] Open
Abstract
Melanoma is one of the most aggressive skin cancers with fiercely increasing incidence and mortality. Since the progressive understanding of the mutational landscape and immunologic pathogenic factors in melanoma, the targeted therapy and immunotherapy have been recently established and gained unprecedented improvements for melanoma treatment. However, the prognosis of melanoma patients remains unoptimistic mainly due to the resistance and nonresponse to current available drugs. Ubiquitination is a posttranslational modification which plays crucial roles in diverse cellular biological activities and participates in the pathogenesis of various cancers, including melanoma. Through the regulation of multiple tumor promoters and suppressors, ubiquitination is emerging as the key contributor and therefore a potential therapeutic target for melanoma. Herein, we summarize the current understanding of ubiquitination in melanoma, from mechanistic insights to clinical progress, and discuss the prospect of ubiquitination modification in melanoma treatment.
Collapse
Affiliation(s)
- Jinyuan Ma
- Department of Dermatology, Xijing hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Weinan Guo
- Department of Dermatology, Xijing hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Chunying Li
- Department of Dermatology, Xijing hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| |
Collapse
|
8
|
Wu S, Yu L. Targeting cullin-RING ligases for cancer treatment: rationales, advances and therapeutic implications. Cytotechnology 2015; 68:1-8. [PMID: 25899169 DOI: 10.1007/s10616-015-9870-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Accepted: 03/26/2015] [Indexed: 02/02/2023] Open
Abstract
New therapeutic intervention strategies for the treatment of human malignancies are always desired. Approval of bortezomib as a front-line treatment for multiple myeloma highlighted the significance of ubiquitin-proteasome system (UPS) as a promising therapeutic target. However, due to the broad impact of proteasome inhibition, deleterious side effects have been reported with bortezomib treatment. Cullin RING ligases (CRLs)-mediated ubiquitin conjugation process is responsible for the ubiquitin conjugation of 20 % cellular proteins that are designated for degradation through the UPS, most of them are critical proteins involved in cell cycle progression, signaling transduction and apoptosis. Studies have depicted the upstream NEDDylation pathway that controls the CRL activity by regulating the conjugation of an ubiquitin-like-protein NEDD8 to the cullin protein in the complex. A specific pharmaceutical inhibitor of NEDD8 activating enzyme (NAE; E1) MLN4924 was recently developed and has been promoted to Phase I clinical trials for the treatment of several human malignancies. This article summarizes the most recent understanding about the process of NEDD8 conjugation, its relevance for cancer therapy and molecular mechanisms responsible for the potent anti-tumor activity of MLN4924.
Collapse
Affiliation(s)
- Shuju Wu
- School of Life Science and Technology, Harbin Normal University, Harbin, 150025, People's Republic of China.
| | - Lijie Yu
- School of Life Science and Technology, Harbin Normal University, Harbin, 150025, People's Republic of China
| |
Collapse
|
9
|
PCTAIRE1 regulates p27 stability, apoptosis and tumor growth in malignant melanoma. Oncoscience 2014; 1:624-33. [PMID: 25593992 PMCID: PMC4278280 DOI: 10.18632/oncoscience.86] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 10/05/2014] [Indexed: 12/27/2022] Open
Abstract
PCTAIRE1 is a cyclin-dependent kinase family protein that has been implicated in spermatogenesis. Although we recently revealed the function of PCTAIRE1 in tumorigenesis of epithelial carcinoma cells, its tumorigenic function in melanoma remains unclear. Interrogation of the Oncomine database revealed that malignant melanoma showed up-regulation of PCTAIRE1 mRNA compared to normal skin and benign melanocytic nevus tissues. In the melanoma cell lines A2058 and SK-MEL-28, PCTAIRE1 gene knockdown using siRNA or shRNA diminished melanoma cell proliferation as assessed by cellular ATP levels, cell counting and clonogenic assays. Moreover, FACS analyses of annexin V-PI staining and DNA content showed that PCTAIRE1 knockdown caused apoptosis in A2058 cells. In contrast, PCTAIRE1 does not appear to be involved in the proliferation of immortalized human keratinocyte HaCaT cells. Depletion of PCTAIRE1 by siRNA/shRNA led to p27 accumulation in melanoma cells but not HaCaT cells. In tumor xenografts of melanoma A2058 cells, conditional knockdown of PCTAIRE1 restored p27 protein expression and suppressed tumor growth. Our findings reveal a crucial role for PCTAIRE1 in regulating p27 protein levels and tumor growth in melanoma cells, suggesting that PCTAIRE1 could provide a target for melanoma treatment.
Collapse
|
10
|
Fagan-Solis KD, Pentecost BT, Gozgit JM, Bentley BA, Marconi SM, Otis CN, Anderton DL, Schneider SS, Arcaro KF. SKP2 overexpression is associated with increased serine 10 phosphorylation of p27 (pSer10p27) in triple-negative breast cancer. J Cell Physiol 2014; 229:1160-9. [PMID: 24443386 DOI: 10.1002/jcp.24545] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Accepted: 12/18/2013] [Indexed: 01/05/2023]
Abstract
S-phase kinase-associated protein 2 (SKP2) is an important cell cycle regulator, targeting the cyclin-dependent kinase (CDK) inhibitor p27 for degradation, and is frequently overexpressed in breast cancer. p27 regulates G1 /S transition by abrogating the activity of cyclin/CDK complexes. p27 can undergo phosphorylation at serine 10 (pSer10p27). This phosphorylation event is associated with increased cell proliferation and poor prognosis in patients with glioma. The relationship between SKP2 and pSer10p27 in breast cancer has not been previously investigated. Immunohistochemistry (IHC) of SKP2, p27, pSer10p27, and other genes involved in this pathway, was analyzed in 188 breast tumors and 50 benign reduction mammoplasty samples. IHC showed SKP2 to be more highly expressed in estrogen receptor α (ERα)-negative breast cancers and demonstrated that triple-negative tumors were more likely to have high expression of SKP2 than were non-triple negative, ERα-negative tumors. A significant positive relationship was discovered for SKP2 and pSer10p27. High levels of SKP2 and pSer10p27 were observed significantly more often in ERα-negative and triple-negative than in ERα-positive breast cancers. Use of the triple-negative TMX2-28 breast cancer cell line to address the role of SKP2 in cell cycle progression confirmed that SKP2 contributes to a more rapid cell cycle progression and may regulates pSer10p27 levels. Together, the results indicate that presence of high SKP2 plus high pSer10p27 levels in triple-negative breast cancers is associated with aggressive growth, and highlight the validity of using SKP2 inhibitors as a therapeutic approach for treating this subset of breast cancers.
Collapse
|
11
|
Qu X, Shen L, Zheng Y, Cui Y, Feng Z, Liu F, Liu J. A signal transduction pathway from TGF-β1 to SKP2 via Akt1 and c-Myc and its correlation with progression in human melanoma. J Invest Dermatol 2014; 134:159-167. [PMID: 23792459 DOI: 10.1038/jid.2013.281] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2012] [Revised: 10/12/2012] [Accepted: 10/29/2012] [Indexed: 12/31/2022]
Abstract
Both SKP2 (S-phase kinase-associated protein 2) and transforming growth factor-β1 (TGF-β1) play important roles in cancer metastasis through different mechanisms: TGF-β1 via induction of epithelial-mesenchymal transition (EMT) and SKP2 via downregulating p27(kip1). Recent studies indicated that c-Myc and Akt1 were active players in metastasis. In this study we demonstrated a crosstalk between these pathways. Specifically, we found that TGF-β1 treatment increased SKP2 expression accompanied with increased phosphorylation of Akt1 and c-Myc protein accumulation during EMT. We demonstrated that Akt1 was required for TGF-β1-mediated SKP2 upregulation and that c-Myc transcription factor specifically bound to the promoter of SKP2 for its enhanced transcription. Analysis of 25 samples of normal human skin, nevi, and melanomas revealed a positive correlation between c-Myc and SKP2 accumulation. Furthermore, accumulation of SKP2 and c-Myc proteins was significantly higher in metastatic melanoma samples as compared with that in primary melanomas, which again was higher than that in normal skin or nevi. In summary, our results integrated TGF-β1 signals to SKP2 via Akt1 and c-Myc during EMT, and provided, to our knowledge, a previously unreported mechanistic molecular event for TGF-β1-induced metastasis in human melanoma.
Collapse
Affiliation(s)
- Xuan Qu
- Institute of Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Xi'an Jiaotong University School of Life Science and Technology, Xi'an, China
| | - Liangliang Shen
- Department of Biochemistry and Molecular Biology, The State Key Laboratory of Cancer Biology, The Fourth Military Medical University, Xi'an, China
| | - Yan Zheng
- Department of Dermatology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Yang Cui
- Institute of Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Xi'an Jiaotong University School of Life Science and Technology, Xi'an, China
| | - Zhihui Feng
- Center for Mitochondrial Biology and Medicine, Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, China.
| | - Feng Liu
- Department of Medicine, University of California Irvine Medical School, Irvine, California, USA; Chao Family Comprehensive Cancer Center, University of California Irvine Medical School, Irvine, California, USA.
| | - Jiankang Liu
- Institute of Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Xi'an Jiaotong University School of Life Science and Technology, Xi'an, China
| |
Collapse
|
12
|
Wu L, Grigoryan AV, Li Y, Hao B, Pagano M, Cardozo TJ. Specific small molecule inhibitors of Skp2-mediated p27 degradation. ACTA ACUST UNITED AC 2013; 19:1515-24. [PMID: 23261596 DOI: 10.1016/j.chembiol.2012.09.015] [Citation(s) in RCA: 183] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Revised: 09/17/2012] [Accepted: 09/24/2012] [Indexed: 10/27/2022]
Abstract
In the ubiquitin proteasome system, the E3 ligase SCF-Skp2 and its accessory protein, Cks1, promote proliferation largely by inducing the degradation of the CDK inhibitor p27. Overexpression of Skp2 in human cancers correlates with poor prognosis, and deregulation of SCF-Skp2-Cks1 promotes tumorigenesis in animal models. We identified small molecule inhibitors specific to SCF-Skp2 activity using in silico screens targeted to the binding interface for p27. These compounds selectively inhibited Skp2-mediated p27 degradation by reducing p27 binding through key compound-receptor contacts. In cancer cells, the compounds induced p27 accumulation in a Skp2-dependent manner and promoted cell-type-specific blocks in the G1 or G2/M phases. Designing SCF-Skp2-specific inhibitors may be a novel strategy to treat cancers dependent on the Skp2-p27 axis.
Collapse
Affiliation(s)
- Lily Wu
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY 10016, USA
| | | | | | | | | | | |
Collapse
|
13
|
Xie CM, Wei W, Sun Y. Role of SKP1-CUL1-F-box-protein (SCF) E3 ubiquitin ligases in skin cancer. J Genet Genomics 2013; 40:97-106. [PMID: 23522382 PMCID: PMC3861240 DOI: 10.1016/j.jgg.2013.02.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Revised: 01/30/2013] [Accepted: 02/04/2013] [Indexed: 11/25/2022]
Abstract
Many biological processes such as cell proliferation, differentiation, and cell death depend precisely on the timely synthesis and degradation of key regulatory proteins. While protein synthesis can be regulated at multiple levels, protein degradation is mainly controlled by the ubiquitin-proteasome system (UPS), which consists of two distinct steps: (1) ubiquitylation of targeted protein by E1 ubiquitin-activating enzyme, E2 ubiquitin-conjugating enzyme and E3 ubiquitin ligase, and (2) subsequent degradation by the 26S proteasome. Among all E3 ubiquitin ligases, the SCF (SKP1-CUL1-F-box protein) E3 ligases are the largest family and are responsible for the turnover of many key regulatory proteins. Aberrant regulation of SCF E3 ligases is associated with various human diseases, such as cancers, including skin cancer. In this review, we provide a comprehensive overview of all currently published data to define a promoting role of SCF E3 ligases in the development of skin cancer. The future directions in this area of research are also discussed with an ultimate goal to develop small molecule inhibitors of SCF E3 ligases as a novel approach for the treatment of human skin cancer. Furthermore, altered components or substrates of SCF E3 ligases may also be developed as the biomarkers for early diagnosis or predicting prognosis.
Collapse
Affiliation(s)
- Chuan-Ming Xie
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Yi Sun
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, University of Michigan, MI 48109, USA
| |
Collapse
|
14
|
Bassermann F, Eichner R, Pagano M. The ubiquitin proteasome system - implications for cell cycle control and the targeted treatment of cancer. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2013; 1843:150-62. [PMID: 23466868 DOI: 10.1016/j.bbamcr.2013.02.028] [Citation(s) in RCA: 208] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Revised: 02/07/2013] [Accepted: 02/22/2013] [Indexed: 01/21/2023]
Abstract
Two families of E3 ubiquitin ligases are prominent in cell cycle regulation and mediate the timely and precise ubiquitin-proteasome-dependent degradation of key cell cycle proteins: the SCF (Skp1/Cul1/F-box protein) complex and the APC/C (anaphase promoting complex or cyclosome). While certain SCF ligases drive cell cycle progression throughout the cell cycle, APC/C (in complex with either of two substrate recruiting proteins: Cdc20 and Cdh1) orchestrates exit from mitosis (APC/C(Cdc20)) and establishes a stable G1 phase (APC/C(Cdh1)). Upon DNA damage or perturbation of the normal cell cycle, both ligases are involved in checkpoint activation. Mechanistic insight into these processes has significantly improved over the last ten years, largely due to a better understanding of APC/C and the functional characterization of multiple F-box proteins, the variable substrate recruiting components of SCF ligases. Here, we review the role of SCF- and APC/C-mediated ubiquitylation in the normal and perturbed cell cycle and discuss potential clinical implications of SCF and APC/C functions. This article is part of a Special Issue entitled: Ubiquitin-Proteasome System. Guest Editors: Thomas Sommer and Dieter H. Wolf.
Collapse
Affiliation(s)
- Florian Bassermann
- Department of Medicine III, Klinikum rechts der Isar, Technische Universität München, Ismaninger Strasse 22, 81675 Munich, Germany.
| | | | | |
Collapse
|
15
|
Yang S, Chen Y, Ahmadie R, Ho EA. Advancements in the field of intravaginal siRNA delivery. J Control Release 2013; 167:29-39. [PMID: 23298612 DOI: 10.1016/j.jconrel.2012.12.023] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2012] [Revised: 12/14/2012] [Accepted: 12/17/2012] [Indexed: 12/17/2022]
Abstract
The vaginal tract is a suitable site for the administration of both local and systemic acting drugs. There are numerous vaginal products on the market such as those approved for contraception, treatment of yeast infection, hormonal replacement therapy, and feminine hygiene. Despite the potential in drug delivery, the vagina is a complex and dynamic organ that requires greater understanding. The recent discovery that injections of double stranded RNA (dsRNA) in Caenorhabditis elegans (C. elegans) results in potent gene specific silencing, was a major scientific revolution. This phenomenon known as RNA interference (RNAi), is believed to protect host genome against invasion by mobile genetic elements such as transposons and viruses. Gene silencing or RNAi has opened new potential opportunities to study the function of a gene in an organism. Furthermore, its therapeutic potential is being investigated in the field of sexually transmitted infections such as human immunodeficiency virus (HIV) and other diseases such as age-related macular degeneration (AMD), diabetes, hypercholesterolemia, respiratory disease, and cancer. This review will focus on the therapeutic potential of siRNA for the treatment and/or prevention of infectious diseases such as HIV, HPV, and HSV within the vaginal tract. Specifically, formulation design parameters to improve siRNA stability and therapeutic efficacy in the vaginal tract will be discussed along with challenges, advancements, and future directions of the field.
Collapse
Affiliation(s)
- Sidi Yang
- Faculty of Pharmacy, University of Manitoba, 750 McDermot Ave, Winnipeg, Manitoba, Canada
| | | | | | | |
Collapse
|
16
|
Chen L, Tweddle DA. p53, SKP2, and DKK3 as MYCN Target Genes and Their Potential Therapeutic Significance. Front Oncol 2012; 2:173. [PMID: 23226679 PMCID: PMC3508619 DOI: 10.3389/fonc.2012.00173] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Accepted: 11/01/2012] [Indexed: 12/15/2022] Open
Abstract
Neuroblastoma is the most common extra-cranial solid tumor of childhood. Despite significant advances, it currently still remains one of the most difficult childhood cancers to cure, with less than 40% of patients with high-risk disease being long-term survivors. MYCN is a proto-oncogene implicated to be directly involved in neuroblastoma development. Amplification of MYCN is associated with rapid tumor progression and poor prognosis. Novel therapeutic strategies which can improve the survival rates whilst reducing the toxicity in these patients are therefore required. Here we discuss genes regulated by MYCN in neuroblastoma, with particular reference to p53, SKP2, and DKK3 and strategies that may be employed to target them.
Collapse
Affiliation(s)
- Lindi Chen
- Newcastle Cancer Centre, Northern Institute for Cancer Research, Newcastle University Newcastle, UK
| | | |
Collapse
|
17
|
Kruck S, Merseburger AS, Hennenlotter J, Scharpf M, Eyrich C, Amend B, Sievert KD, Stenzl A, Bedke J. High cytoplasmic expression of p27(Kip1) is associated with a worse cancer-specific survival in clear cell renal cell carcinoma. BJU Int 2011; 109:1565-70. [PMID: 21981759 DOI: 10.1111/j.1464-410x.2011.10649.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
UNLABELLED What's known on the subject? and What does the study add? The loss of p27(Kip1) correlates with poor prognosis in various human cancers, and was postulated as a biomarker in RCC. Up to now p27(Kip1) analysis in RCC was focused on its nuclear localization. We confirmed higher p27(Kip1) expression in the nucleus and cytoplasm of RCC and correlated high cytoplasmic p27(Kip1) with an unfavourable clinic and a reduced survival. OBJECTIVES To analyse the cytoplasmic and nuclear differences of p27(Kip1) expression and localization in benign and clear cell renal cell carcinoma (ccRCC) with regard to overall survival (OS) and cancer-specific survival (CSS). p27(Kip1) is considered to contribute to the progression of ccRCC and is targeted by next generation dual-therapies. PATIENTS AND METHODS In 140 patients, ccRCC and corresponding benign kidney tissue were analyzed for nuclear and cytoplasmic staining of p27(Kip1) by immunohistochemistry using a tissue microarray technique. Staining intensity and percentage of positive stained cells are given as expression scores. p27(Kip1) expression was categorized as high if ccRCC tissue stained stronger than the respective level of the corresponding benign tissue and categorized as low if ccRCC tissue stained less than or equal to the corresponding benign tissue. Differences in OS and CSS were analyzed by log-rank analysis and expression levels were correlated with tumour and patient characteristics using Fisher's exact test. RESULTS Cytoplasmatic (mean [sd]: 13.8% [1.2%] vs 10.7% [1.7%]; P= 0.04) and nuclear (mean [sd]: 75.6% [2.7%] vs 13.6% [2.1%]; P < 0.001) staining of p27(Kip1) were significantly stronger in ccRCC tissues compared to benign tissue. High cytoplasmic p27(Kip1) expression was significantly associated with a higher T and N stage, Fuhrman grade and the presence of metastatic disease (P < 0.001). The median follow-up time was 38.2 months. There was no difference in OS between the low and high expression groups, although CSS was significantly lower in patients with high cytoplasmic p27(Kip1) (P < 0.001) and CSS heavily tended to be lower in the nuclear low expression group (P= 0.069). CONCLUSIONS High cytoplasmic p27(Kip1) levels in renal cancer tissues are associated with advanced disease and reduced cancer specific survival, whereas low nuclear expression levels appear to be beneficial. The present study corroborates the consideration of cytoplasmic p27(Kip1) for future diagnostic and targeted therapeutic approaches in RCC establishing a potential protective shift of p27(Kip1) from the cytoplasm to the nucleus.
Collapse
Affiliation(s)
- Stephan Kruck
- Department of Urology Pathology, Eberhard-Karls-University Tuebingen, Tuebingen, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Chen G, Cheng Y, Zhang Z, Martinka M, Li G. Prognostic significance of cytoplasmic p27 expression in human melanoma. Cancer Epidemiol Biomarkers Prev 2011; 20:2212-21. [PMID: 21828232 DOI: 10.1158/1055-9965.epi-11-0472] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND The cyclin-dependent kinase inhibitor p27 plays important roles in cell proliferation, cell motility, and apoptosis. Interestingly, the nuclear and cytoplasmic p27 exert opposite biological functions. In this study, we investigated the prognostic impact of subcellular p27 expression. METHODS We constructed melanoma tissue microarrays in a large series of melanoma patients, including 29 normal nevi, 52 dysplastic nevi, 270 primary melanomas, and 148 metastatic melanomas. The expression level of subcellular p27 in different stages of melanocytic lesions and its prognostic significance were evaluated. RESULTS Compared with dysplastic nevi, nuclear p27 expression was remarkably reduced in primary melanomas and further reduced in metastatic melanoma (P < 0.001 for both), whereas cytoplasmic p27 expression is significantly increased from dysplastic nevi to primary melanomas (P = 0.032) and further increased in melanoma metastases (P = 0.037). Although loss of nuclear p27 expression is correlated with a worse 5-year survival of primary melanoma patients in Kaplan-Meier analysis (P = 0.046), it is not a prognostic factor by multivariate Cox regression analysis. On the contrary, Kaplan-Meier analysis showed that gain of cytoplasmic p27 was associated with a poor 5-year survival of metastatic melanoma patients (P < 0.001). Multivariate Cox regression analysis revealed that positive cytoplasmic p27 expression is an independent prognostic factor to predict metastatic melanoma patient outcome. CONCLUSION Cytoplasmic p27 may serve as a promising prognostic marker for metastatic melanoma. IMPACT Because there is no reliable prognostic marker for metastatic melanoma, our finding may have important clinical implications using cytoplasmic p27 as a prognostic biomarker for advanced melanoma.
Collapse
Affiliation(s)
- Guangdi Chen
- Department of Dermatology and Skin Science, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | | | | | | | | |
Collapse
|
19
|
Jia L, Sun Y. SCF E3 ubiquitin ligases as anticancer targets. Curr Cancer Drug Targets 2011; 11:347-56. [PMID: 21247385 DOI: 10.2174/156800911794519734] [Citation(s) in RCA: 118] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2010] [Accepted: 12/27/2010] [Indexed: 11/22/2022]
Abstract
The SCF multisubunit complex (Skp1, Cullins, F-box proteins) E3 ubiquitin ligase, also known as CRL (Cullin-RING ubiquitin Ligase) is the largest E3 ubiquitin ligase family that promotes the ubiquitination of various regulatory proteins for targeted degradation, thus regulating many biological processes, including cell cycle progression, signal transduction, and DNA replication. The efforts to discover small molecule inhibitors of a SCF-type ligase or its components were expedited by the FDA approval of Bortezomib (also known as Velcade or PS-341), the first (and only) class of general proteasome inhibitor, for the treatment of relapsed/refractory multiple myeloma and mantle cell lymphoma. Although Bortezomib has demonstrated a certain degree of cancer cell selectivity with measurable therapeutic index, the drug is, in general, cytotoxic due to its inhibition of overall protein degradation. An alternative and ideal approach is to target a specific E3 ligase, known to be activated in human cancer, for a high level of specificity and selectivity with less associated toxicity, since such inhibitors would selectively stabilize a specific set of cellular proteins regulated by this E3. Here, we review recent advances in validation of SCF E3 ubiquitin ligase complex as an attractive anti-cancer target and discuss how MLN4924, a small molecule inhibitor of NEDD8-activating enzyme, can be developed as a novel class of anticancer agents by inhibiting SCF E3 ligase complex via removal of cullin neddylation. Finally, we discuss under future perspective how basic research on SCF biology will direct the drug discovery efforts surrounding this target.
Collapse
Affiliation(s)
- L Jia
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, University of Michigan Comprehensive Cancer Center, 4424B Medical Science-I, 1301 Catherine Street, Ann Arbor, MI 48109, USA
| | | |
Collapse
|
20
|
Huang HC, Lin CL, Lin JK. 1,2,3,4,6-penta-O-galloyl-β-D-glucose, quercetin, curcumin and lycopene induce cell-cycle arrest in MDA-MB-231 and BT474 cells through downregulation of Skp2 protein. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2011; 59:6765-6775. [PMID: 21598989 DOI: 10.1021/jf201096v] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
The F-box protein S-phase kinase-associated protein 2 (Skp2), which acts as an oncogene through targeting p27 for degradation, is overexpressed in many different human cancers. Skp2 can play an important role in breast cancer progression and may also be a novel molecular target for the treatment of breast cancer, especially estrogen receptor (ER)/human epidermal growth factor 2 (HER2) negative breast cancers. Unfortunately, specific drugs that target Skp2 are unavailable at present. Therefore, it is important to explore whether commonly used chemopreventive agents may downregulate Skp2 expression. In this study, we examined the effects of 1,2,3,4,6-penta-O-galloyl-β-D-glucose (pentagalloylglucose, 5gg), quercetin, curcumin and lycopene on the expression of Skp2 in MDA-MB-231 (ER/HER2-negative) and BT474 (ER-negative/HER2-positive) cells. We found that all four phytochemicals studied induced cell growth inhibition in MDA-MB-231 cells. The mechanism of the initial growth inhibitory events involves blocking the cell cycle progression. Further, we found that quercetin and curcumin induced growth arrest by inhibition of Skp2, and induced p27 expression in MDA-MB-231 cells. However, the decrease in Skp2 levels in cells treated with 5gg or lycopene did not translate to p27 upregulation. Consequently, the downregulation of Skp2 did not always correlate with the upregulation of p27, suggesting that phytochemical-dependent downregulation of Skp2 can influence cell growth in several ways. Several studies have demonstrated that Skp2 directs the ubiquitylation and subsequent degradation of forkhead box protein O1 (FoxO1). Furthermore, our results reveal that FoxO1 protein was increased after 5gg, quercetin, curcumin and lycopene treatment. The therapeutic strategies designed to reduce Skp2 may therefore play an important clinical role in treatment of breast cancer cells, especially ER/HER2-negative breast cancers.
Collapse
Affiliation(s)
- Hsiu-Chen Huang
- Department of Applied Science, National Hsinchu University of Education, Hsinchu 30014, Taiwan
| | | | | |
Collapse
|
21
|
Redpath M, Xu B, van Kempen LC, Spatz A. The dual role of the X-linked FoxP3 gene in human cancers. Mol Oncol 2011; 5:156-63. [PMID: 21489891 DOI: 10.1016/j.molonc.2011.03.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2011] [Accepted: 03/11/2011] [Indexed: 01/18/2023] Open
Abstract
The FoxP3 (forkhead box P3) gene is an X-linked gene that is submitted to inactivation. It is an essential transcription factor in CD4(+)CD25(+)FoxP3 regulatory T cells, which are therapeutic targets in disseminated cutaneous melanoma. Moreover, FoxP3 is an important tumor suppressor gene in carcinomas and has putative cancer suppressor gene function in cutaneous melanoma as well. Therefore understanding the structure and function of the FoxP3 gene is crucial to gaining insight into the biology of melanoma to better develop immunotherapeutics and future therapeutic strategies.
Collapse
Affiliation(s)
- Margaret Redpath
- Department of Pathology, McGill University, Montreal, QC, Canada
| | | | | | | |
Collapse
|
22
|
Cytoplasmic Skp2 expression is increased in human melanoma and correlated with patient survival. PLoS One 2011; 6:e17578. [PMID: 21386910 PMCID: PMC3046256 DOI: 10.1371/journal.pone.0017578] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2010] [Accepted: 02/09/2011] [Indexed: 12/05/2022] Open
Abstract
Background S-phase kinase protein 2 (Skp2), an F-box protein, targets cell cycle regulators via ubiquitin-mediated degradation. Skp2 is frequently overexpressed in a variety of cancers and associated with patient survival. In melanoma, however, the prognostic significance of subcellular Skp2 expression remains controversial. Methods To investigate the role of Skp2 in melanoma development, we constructed tissue microarrays and examined Skp2 expression in melanocytic lesions at different stages, including 30 normal nevi, 61 dysplastic nevi, 290 primary melanomas and 146 metastatic melanomas. The TMA was assessed for cytoplasmic and nuclear Skp2 expression by immunohistochemistry. The Kaplan-Meier method was used to evaluate the patient survival. The univariate and multivariate Cox regression models were performed to estimate the harzard ratios (HR) at five-year follow-up. Results Cytoplasmic but not nuclear Skp2 expression was gradually increased from normal nevi, dysplastic nevi, primary melanomas to metastatic melanomas. Cytoplasmic Skp2 expression correlated with AJCC stages (I vs II–IV, P<0.001), tumor thickness (≤2.00 vs >2.00 mm, P<0.001) and ulceration (P = 0.005). Increased cytoplasmic Skp2 expression was associated with a poor five-year disease-specific survival of patients with primary melanoma (P = 0.018) but not metastatic melanoma (P>0.05). Conclusion This study demonstrates that cytoplasmic Skp2 plays an important role in melanoma pathogenesis and its expression correlates with patient survival. Our data indicate that cytoplasmic Skp2 may serve as a potential biomarker for melanoma progression and a therapeutic target for this disease.
Collapse
|
23
|
Rose AE, Wang G, Hanniford D, Monni S, Tu T, Shapiro RL, Berman RS, Pavlick AC, Pagano M, Darvishian F, Mazumdar M, Hernando E, Osman I. Clinical relevance of SKP2 alterations in metastatic melanoma. Pigment Cell Melanoma Res 2010; 24:197-206. [PMID: 20883453 DOI: 10.1111/j.1755-148x.2010.00784.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
In this study, we investigated the mechanism(s) of altered expression of protooncogene SKP2 in metastatic melanoma and its clinical relevance in patients with metastatic melanoma. The genomic status of SKP2 was assessed in cell lines by sequencing, single nucleotide polymorphism array, and genomic PCR. Copy number status was then evaluated for concordance with SKP2 mRNA and protein expression. SKP2 protein was further evaluated by immunohistochemistry in 93 human metastatic tissues. No mutations were identified in SKP2. Increased copy number at the SKP2 locus was observed in 6/14 (43%) metastatic cell lines and in 9/22 (41%) human metastatic tissues which was associated with overexpression of SKP2 protein. Overexpression of SKP2 protein in human tissues was associated with worse survival in a multivariate model controlling for the site of metastasis. Copy number gain is a major contributing mechanism of SKP2 overexpression in metastatic melanoma. Results may have implications for the development of therapeutics that target SKP2.
Collapse
Affiliation(s)
- Amy E Rose
- Ronald O. Perelman Department of Dermatology, New York University School of Medicine, New York, NY, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Nakayama K. Growth and progression of melanoma and non-melanoma skin cancers regulated by ubiquitination. Pigment Cell Melanoma Res 2010; 23:338-51. [DOI: 10.1111/j.1755-148x.2010.00692.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
25
|
Chen G, Wang Y, Garate M, Zhou J, Li G. The tumor suppressor ING3 is degraded by SCFSkp2-mediated ubiquitin–proteasome system. Oncogene 2009; 29:1498-508. [DOI: 10.1038/onc.2009.424] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
26
|
Xiao J, Yin S, Li Y, Xie S, Nie D, Ma L, Wang X, Wu Y, Feng J. SKP2 siRNA inhibits the degradation of P27kip1 and down-regulates the expression of MRP in HL-60/A cells. Acta Biochim Biophys Sin (Shanghai) 2009; 41:699-708. [PMID: 19657571 DOI: 10.1093/abbs/gmp058] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
S-phase kinase-associated protein 2 (SKP2) gene is a tumor suppressor gene, and is involved in the ubiquitin-mediated degradation of P27kip1. SKP2 and P27kip1 affect the proceeding and prognosis of leukemia through regulating the proliferation, apoptosis and differentiation of leukemia cells. In this study, we explored the mechanism of reversing of HL-60/A drug resistance through SKP2 down-regulation. HL-60/A cells were nucleofected by Amaxa Nucleofector System with SKP2 siRNA. The gene and protein expression levels of Skp2, P27kip1, and multi-drug resistance associated protein (MRP) were determined by reverse transcription-polymerase chain reaction and western blot analysis, respectively. The cell cycle was analyzed by flow cytometry. The 50% inhibitory concentration value was calculated using cytotoxic analysis according to the death rate of these two kinds of cells under different concentrations of chemotherapeutics to compare the sensitivity of the cells. HL-60/A cells showed multi-drug resistance phenotype characteristic by cross-resistance to adriamycin, daunorubicin, and arabinosylcytosine, due to the expression of MRP. We found that the expression of SKP2 was higher in HL-60/A cells than in HL-60 cells, but the expression of P27kip1 was lower. The expression of SKP2 in HL-60/A cells nucleofected by SKP2 siRNA was down-regulated whereas the protein level of P27kip1 was up-regulated. Compared with the MRP expression level in the control group (nucleofected by control siRNA), the mRNA and protein expression levels of MRP in HL-60/A cells nucleofected by SKP2 siRNA were lower, and the latter cells were more sensitive to adriamycin, daunorubicin, and arabinosylcytosine. Down-regulating the SKP2 expression and arresting cells in the G0/G1 phase improve drug sensitivity of leukemia cells with down-regulated MRP expression.
Collapse
Affiliation(s)
- Jie Xiao
- Department of Hematology, Second Afflicted Hospital of Sun Yat-Sen University, Guangzhou, China
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Shen C, Gu M, Liang D, Miao L, Hu L, Zheng C, Chen J. Establishment and characterization of three new human breast cancer cell lines derived from Chinese breast cancer tissues. Cancer Cell Int 2009; 9:2. [PMID: 19121212 PMCID: PMC2646685 DOI: 10.1186/1475-2867-9-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2008] [Accepted: 01/02/2009] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Breast cancer is a major malignancy affecting females worldwide. It is the most common cause of death from cancer in women. Cell lines are widely used in laboratory research and particularly as in vitro models in cancer research. But we found that the routinely used breast cancer cell lines were mostly derived from Caucasians or African-Americans. There were few standard models to study the pathogenic mechanism at molecular level and cell signaling pathway of breast cancer for Asian patients. It is quite necessary to establish new breast cancer cell lines from xanthoderm to study the pathogenic mechanism and therapeutic methods. RESULTS Three new breast cancer cell lines, designated BC-019, BC-020 and BC-021, were successfully established and characterized from breast invasive ductal carcinoma tissues of three Chinese female patients. These new cell lines growing as adherent monolayer with characteristic epithelial morphology could be maintained continuously in vitro, and they were ER-, PR- and C-erbB-2-positive. Their chromosomes showed high hyperdiploidy and complex rearrangements, and they displayed aggressive tumorigencity in tumorigenesis test. CONCLUSION The three newly established breast cancer cell lines from Chinese patients were tested for a number of, and the results indicate that the cell lines were in good quality and could be served as new cell models in breast cancer study.
Collapse
Affiliation(s)
- Chao Shen
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, PR China
- China Center for Type Culture Collection, Wuhan University, Wuhan 430072, PR China
| | - Meijia Gu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, PR China
- China Center for Type Culture Collection, Wuhan University, Wuhan 430072, PR China
| | - Dan Liang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, PR China
- China Center for Type Culture Collection, Wuhan University, Wuhan 430072, PR China
| | - Lixia Miao
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, PR China
| | - Liu Hu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, PR China
| | - Congyi Zheng
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, PR China
- China Center for Type Culture Collection, Wuhan University, Wuhan 430072, PR China
| | - Jiakuan Chen
- Zhongnan Hospital, Wuhan University, Wuhan 430072, PR China
| |
Collapse
|
28
|
He S, Zhang D, Cheng F, Gong F, Guo Y. Applications of RNA interference in cancer therapeutics as a powerful tool for suppressing gene expression. Mol Biol Rep 2009; 36:2153-63. [PMID: 19117119 DOI: 10.1007/s11033-008-9429-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2008] [Accepted: 12/08/2008] [Indexed: 01/07/2023]
Abstract
Cancer poses a tremendous therapeutic challenge worldwide, highlighting the critical need for developing novel therapeutics. A promising cancer treatment modality is gene therapy, which is a form of molecular medicine designed to introduce into target cells genetic material with therapeutic intent. The history of RNA interference (RNAi) has only a dozen years, however, further studies have revealed that it is a potent method of gene silencing that has developed rapidly over the past few years as a result of its extensive importance in the study of genetics, molecular biology and physiology. RNAi is a natural process by which small interfering RNA (siRNA) duplex directs sequence specific post-transcriptional silencing of homologous genes by binding to its complementary mRNA and triggering its elimination. RNAi has been extensively used as a novel and effective gene silencing tool for the fundamental research of cancer therapeutics, and has displayed great potential in clinical treatment.
Collapse
Affiliation(s)
- Song He
- Molecular Medicine & Tumor Research Center, Chongqing Medical University, Chongqing, China.
| | | | | | | | | |
Collapse
|
29
|
Miranda-Carboni GA, Krum SA, Yee K, Nava M, Deng QE, Pervin S, Collado-Hidalgo A, Galić Z, Zack JA, Nakayama K, Nakayama KI, Lane TF. A functional link between Wnt signaling and SKP2-independent p27 turnover in mammary tumors. Genes Dev 2008; 22:3121-34. [PMID: 19056892 PMCID: PMC2593606 DOI: 10.1101/gad.1692808] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2008] [Accepted: 09/12/2008] [Indexed: 11/24/2022]
Abstract
Loss of the CDK inhibitor p27(KIP1) is widely linked with poor prognosis in human cancer. In Wnt10b-expressing mammary tumors, levels of p27(KIP1) were extremely low; conversely, Wnt10b-null mammary cells expressed high levels of this protein, suggesting Wnt-dependent regulation of p27(KIP1). Interestingly we found that Wnt-induced turnover of p27(KIP1) was independent from classical SCF(SKP2)-mediated degradation in both mouse and human cells. Instead, turnover required Cullin 4A and Cullin 4B, components of an alternative E3 ubiquitin ligase induced in response to active Wnt signaling. We found that CUL4A was a novel Wnt target gene in both mouse and human cells and that CUL4A physically interacted with p27(KIP1) in Wnt-responding cells. We further demonstrated that both Cul4A and Cul4B were required for Wnt-induced p27(KIP1) degradation and S-phase progression. CUL4A and CUL4B are therefore components of a conserved Wnt-induced proteasome targeting (WIPT) complex that regulates p27(KIP1) levels and cell cycle progression in mammalian cells.
Collapse
Affiliation(s)
- Gustavo A. Miranda-Carboni
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California 90095, USA
| | - Susan A. Krum
- Department of Orthopaedic Surgery, University of California at Los Angeles-Orthopaedic Hospital, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California 90095, USA
| | - Kathleen Yee
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California 90095, USA
| | - Miguel Nava
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California 90095, USA
| | - Qiming E. Deng
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California 90095, USA
| | - Shehla Pervin
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California 90095, USA
| | - Alicia Collado-Hidalgo
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California 90095, USA
| | - Zoran Galić
- Department of Medicine, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California 90095, USA
| | - Jerome A. Zack
- Department of Medicine, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California 90095, USA
- Department of Microbiology, Immunology and Molecular Genetics, University of California at Los Angeles, Los Angeles, California 90095, USA
| | - Keiko Nakayama
- Department of Developmental Biology, Center for Translational and Advance Animal Research, Graduate School of Medicine Tohoku University, Aoba-ku Sendai 980-8575, Japan
| | - Keiichi I. Nakayama
- Department of Developmental Biology, Center for Translational and Advance Animal Research, Graduate School of Medicine Tohoku University, Aoba-ku Sendai 980-8575, Japan
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Higashi-ku, Fukuoka 812-8582, Japan
| | - Timothy F. Lane
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California 90095, USA
- Department of Orthopaedic Surgery, University of California at Los Angeles-Orthopaedic Hospital, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California 90095, USA
- Department of Biological Chemistry, University of California at Los Angeles, Los Angeles, California 90095, USA
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California 90095, USA
| |
Collapse
|
30
|
Fang L, Hu Q, Hua Z, Li S, Dong W. Growth inhibition of a tongue squamous cell carcinoma cell line (Tca8113) in vitro and in vivo via siRNA-mediated down-regulation of skp2. Int J Oral Maxillofac Surg 2008; 37:847-52. [DOI: 10.1016/j.ijom.2008.05.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2007] [Revised: 01/02/2008] [Accepted: 05/29/2008] [Indexed: 10/21/2022]
|
31
|
Abstract
Targeted proteasomal degradation mediated by E3 ubiquitin ligases controls cell cycle progression, and alterations in their activities likely contribute to malignant cell proliferation. S phase kinase-associated protein 2 (Skp2) is the F-box component of an E3 ubiquitin ligase complex that targets p27(Kip1) and cyclin E1 to the proteasome. In human melanoma, Skp2 is highly expressed, regulated by mutant B-RAF, and required for cell growth. We show that Skp2 depletion in melanoma cells resulted in a tetraploid cell cycle arrest. Surprisingly, co-knockdown of p27(Kip1) or cyclin E1 failed to prevent the tetraploid arrest induced by Skp2 knockdown. Enhanced Aurora A phosphorylation and repression of G2/M regulators cyclin B1, cyclin-dependent kinase 1, and cyclin A indicated a G2/early M phase arrest in Skp2-depleted cells. Furthermore, expression of nuclear localized cyclin B1 prevented tetraploid accumulation after Skp2 knockdown. The p53 status is most frequently wild type in melanoma, and the tetraploid arrest and down-regulation of G2/M regulatory genes were strongly dependent on wild-type p53 expression. In mutant p53 melanoma lines, Skp2 depletion did not induce cell cycle arrest despite up-regulation of p27(Kip1). These data indicate that elevated Skp2 expression may overcome p53-dependent cell cycle checkpoints in melanoma cells and highlight Skp2 actions that are independent of p27(Kip1) degradation.
Collapse
Affiliation(s)
- Rong Hu
- Center for Cell Biology and Cancer Research, Albany Medical College, Albany, NY 12208, USA
| | | |
Collapse
|
32
|
|
33
|
Frescas D, Pagano M. Deregulated proteolysis by the F-box proteins SKP2 and beta-TrCP: tipping the scales of cancer. Nat Rev Cancer 2008; 8:438-49. [PMID: 18500245 PMCID: PMC2711846 DOI: 10.1038/nrc2396] [Citation(s) in RCA: 753] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The maintenance and preservation of distinct phases during the cell cycle is a highly complex and coordinated process. It is regulated by phosphorylation--through the activity of cyclin-dependent kinases (CDKs)--and protein degradation, which occurs through ubiquitin ligases such as SCF (SKP1-CUL1-F-box protein) complexes and APC/C (anaphase-promoting complex/cyclosome). Here, we explore the functionality and biology of the F-box proteins, SKP2 (S-phase kinase-associated protein 2) and beta-TrCP (beta-transducin repeat-containing protein), which are emerging as important players in cancer biogenesis owing to the deregulated proteolysis of their substrates.
Collapse
Affiliation(s)
- David Frescas
- Department of Pathology, NYU Cancer Institute, New York University School of Medicine, 550 First Avenue, New York, New York 10016, USA
| | | |
Collapse
|
34
|
Abstract
The secreted glycoprotein osteopontin (OPN) sets into motion an astounding variety of activities that range from bone remodeling via immunomodulation to the inhibition of apoptosis. In the current issue of the European Journal of Immunology, OPN now also enters mast cell biology and the regulation of IgE-dependent immune responses since it is reported that connective tissue-type mast cells from fetal murine skin constitutively secrete biologically active OPN. Moreover, it is shown that, in vitro, OPN augments IgE-mediated mast cell degranulation and migration via ligand binding to cognate OPN receptors on the mast cell surface (CD44, alpha v integrin) and that the magnitude of an IgE-mediated passive cutaneous anaphylaxis reaction is augmented by OPN in vivo. Here, we discuss why this newly discovered property of OPN fits well into the emerging concept that OPN may serve as a multi-purpose environmental damage-response protein.
Collapse
Affiliation(s)
- Silvia Bulfone-Paus
- Department of Immunology and Cell Biology, Research Center Borstel, Borstel, Germany.
| | | |
Collapse
|
35
|
Shen C, Gu M, Song C, Miao L, Hu L, Liang D, Zheng C. The tumorigenicity diversification in human embryonic kidney 293 cell line cultured in vitro. Biologicals 2008; 36:263-8. [PMID: 18378163 DOI: 10.1016/j.biologicals.2008.02.002] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2007] [Revised: 02/12/2008] [Accepted: 02/21/2008] [Indexed: 11/25/2022] Open
Abstract
The Human Embryonic Kidney (HEK) 293 cell line is widely used in research work such as vaccine production, adenovirus and adeno-associated viral vectors, and gene therapy. However, little attention was drawn to the passage level of 293 cells. We first claim that the tumorigenicity of the HEK 293 cell line reached 100% when the passage exceeded 65, whereas using low-passage (<52) HEK 293 cell line no tumor could be induced under the same condition. Results from nude mice assay, tumor tissue histological examination, primary culture, PCR and isoenzyme analysis showed that the tumor in nude mice could only be induced by viable high-passage 293 cells. This suggests that more attention should be paid to the passage level of the HEK 293 cell line, especially for vaccine production but the low-passage HEK 293 cell line should be acceptable to regulatory authorities for recombinant virus vector, vaccines, and gene therapy. Meanwhile, we also find that high-passage HEK 293 can be employed as a highly malignant tumor model as its tumorigenicity increases significantly.
Collapse
Affiliation(s)
- Chao Shen
- State key laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | | | | | | | | | | | | |
Collapse
|
36
|
Targeting the p27 E3 ligase SCF(Skp2) results in p27- and Skp2-mediated cell-cycle arrest and activation of autophagy. Blood 2008; 111:4690-9. [PMID: 18305219 DOI: 10.1182/blood-2007-09-112904] [Citation(s) in RCA: 221] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Decreased p27(Kip1) levels are a poor prognostic factor in many malignancies, and can occur through up-regulation of SCF(Skp2) E3 ligase function, resulting in enhanced p27 ubiquitination and proteasome-mediated degradation. While proteasome inhibitors stabilize p27(Kip1), agents inhibiting SCF(Skp2) may represent more directly targeted drugs with the promise of enhanced efficacy and reduced toxicity. Using high-throughput screening, we identified Compound A (CpdA), which interfered with SCF(Skp2) ligase function in vitro, and induced specific accumulation of p21 and other SCF(Skp2) substrates in cells without activating a heat-shock protein response. CpdA prevented incorporation of Skp2 into the SCF(Skp2) ligase, and induced G(1)/S cell-cycle arrest as well as SCF(Skp2)- and p27-dependent cell killing. This programmed cell death was caspase-independent, and instead occurred through activation of autophagy. In models of multiple myeloma, CpdA overcame resistance to dexamethasone, doxorubicin, and melphalan, as well as to bortezomib, and also acted synergistically with this proteasome inhibitor. Importantly, CpdA was active against patient-derived plasma cells and both myeloid and lymphoblastoid leukemia blasts, and showed preferential activity against neoplastic cells while relatively sparing other marrow components. These findings provide a rational framework for further development of SCF(Skp2) inhibitors as a novel class of antitumor agents.
Collapse
|
37
|
Nagasaka A, Matsue H, Matsushima H, Aoki R, Nakamura Y, Kambe N, Kon S, Uede T, Shimada S. Osteopontin is produced by mast cells and affects IgE-mediated degranulation and migration of mast cells. Eur J Immunol 2008; 38:489-99. [DOI: 10.1002/eji.200737057] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
38
|
RNAi silencing of the WT1 gene inhibits cell proliferation and induces apoptosis in the B16F10 murine melanoma cell line. Melanoma Res 2007; 17:341-8. [DOI: 10.1097/cmr.0b013e3282efd3ae] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
39
|
Lejeune FJ, Rimoldi D, Speiser D. New approaches in metastatic melanoma: biological and molecular targeted therapies. Expert Rev Anticancer Ther 2007; 7:701-13. [PMID: 17492933 DOI: 10.1586/14737140.7.5.701] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Classical metastatic melanoma therapy is disappointing but important progress has been made in the understanding of melanoma biology. Genetic lesions and several intracellular signaling pathways that could serve as targets for novel therapy have been identified and a number of new agents are under evaluation. Promising tumor cell targets were identified in the cell membrane, cytoplasm and nucleus. New therapeutic approaches, besides monoclonal antibodies and vaccination, include an increasing number of small molecules that have been shown to interfere restrictively with intracellular signaling pathways in melanoma and decrease proliferation, survival, migration or invasion. Other agents can interfere with stromal components of melanoma, such as angiogenesis and components of the immune system.
Collapse
Affiliation(s)
- Ferdy J Lejeune
- Ludwig Institute for Cancer Research, Division of Clinical Onco-Immunology and Centre Hospitalier Universitaire Vaudois, Department of Visceral Surgery, Lausanne, Switzerland.
| | | | | |
Collapse
|
40
|
Abstract
Ubiquitin and ubiquitin-like proteins (Ubls) are signalling messengers that control many cellular functions, such as cell proliferation, apoptosis, the cell cycle and DNA repair. It is becoming apparent that the deregulation of ubiquitin pathways results in the development of human diseases, including many types of tumours. Here we summarize the common principles and specific features of ubiquitin and Ubls in the regulation of cancer-relevant pathways, and discuss new strategies to target ubiquitin signalling in drug discovery.
Collapse
Affiliation(s)
- Daniela Hoeller
- Institute of Biochemistry II, Goethe University School of Medicine, University Hospital, Building 75, Theodour-Stern-Kai 7, D-60590 Frankfurt, Germany
| | | | | |
Collapse
|
41
|
Abstract
E3 ubiquitin ligases are a large family of proteins that are engaged in the regulation of the turnover and activity of many target proteins. Together with ubiquitin-activating enzyme E1 and ubiquitin-conjugating enzyme E2, E3 ubiquitin ligases catalyze the ubiquitination of a variety of biologically significant protein substrates for targeted degradation through the 26S proteasome, as well as for nonproteolytic regulation of their functions or subcellular localizations. E3 ubiquitin ligases, therefore, play an essential role in the regulation of many biologic processes. Increasing amounts of evidence strongly suggest that the abnormal regulation of some E3 ligases is involved in cancer development. Furthermore, some E3 ubiquitin ligases are frequently overexpressed in human cancers, which correlates well with increased chemoresistance and poor clinic prognosis. In this review, E3 ubiquitin ligases (such as murine double minute 2, inhibitor of apoptosis protein, and Skp1-Cullin-F-box protein) will be evaluated as potential cancer drug targets and prognostic biomarkers. Extensive study in this field would lead to a better understanding of the molecular mechanism by which E3 ligases regulate cellular processes and of how their deregulations contribute to carcinogenesis. This would eventually lead to the development of a novel class of anticancer drugs targeting specific E3 ubiquitin ligases, as well as the development of sensitive biomarkers for cancer treatment, diagnosis, and prognosis.
Collapse
Affiliation(s)
- Yi Sun
- Division of Cancer Biology, Department of Radiation Oncology, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI 48109-0936, USA.
| |
Collapse
|
42
|
Bhatt KV, Hu R, Spofford LS, Aplin AE. Mutant B-RAF signaling and cyclin D1 regulate Cks1/S-phase kinase-associated protein 2-mediated degradation of p27Kip1 in human melanoma cells. Oncogene 2006; 26:1056-66. [PMID: 16924241 DOI: 10.1038/sj.onc.1209861] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Levels of cyclins and cyclin-dependent kinase (Cdk) inhibitors are tightly controlled during normal cell proliferation and are frequently dysregulated in cancerous cells. In melanoma, cyclin D1 is highly expressed and downregulation of the Cdk inhibitor, p27(Kip1), is associated with a poor prognosis. Mutant B-RAF is frequently expressed in melanoma and overrides growth factor and matrix adhesion control of cyclin D1 and p27(Kip1) levels in human melanocytes. Here, we demonstrate that p27(Kip1) expression is regulated by multiple mechanisms in melanoma cells. B-RAF regulates p27(Kip1) mRNA abundance independently of cyclin D1. Additionally, B-RAF and cyclin D1 control the levels of S-phase kinase-associated protein 2 (Skp2) that directs ubiquitin-mediated proteolysis of p27(Kip1). The cofactor for Skp2, Cdc kinase subunit 1 (Cks1) controls levels of Skp2 in melanoma cells and acts jointly with Skp2 to regulate p27(Kip1) levels. Importantly, expression of Cks1 is regulated by B-RAF and cyclin D1 at the mRNA level. Reduced Cks1 or Skp2 expression and enhanced p27(Kip1) levels inhibit melanoma cell growth. In summary, p27(Kip1) expression in melanoma is regulated by B-RAF at the mRNA level, and via B-RAF and cyclin D1 control of Cks1/Skp2-mediated proteolysis.
Collapse
Affiliation(s)
- K V Bhatt
- Center for Cell Biology and Cancer Research, 47 New Scotland Avenue, Albany Medical College, Albany, NY 12208, USA
| | | | | | | |
Collapse
|
43
|
Abstract
Regulated protein turnover via the ubiquitin-proteasome system (UPS) underlies a wide variety of signalling pathways, from cell-cycle control and transcription to development. Recent evidence that pharmacological inhibition of the proteasome can be efficacious in the treatment of human cancers has set the stage for attempts to selectively inhibit the activities of disease-specific components of the UPS. Here, we review recent advances linking UPS components with specific human diseases, most prominently cancer and neurodegenerative disorders, and emphasize potential sites of therapeutic intervention along the regulated protein-degradation pathway.
Collapse
Affiliation(s)
- Grzegorz Nalepa
- Department of Pathology, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, Massachusetts 02115, USA
| | | | | |
Collapse
|