1
|
Matskova L, Zheng S, Kashuba E, Ernberg I, Aspenström P. MTSS1: beyond the integration of actin and membrane dynamics. Cell Mol Life Sci 2024; 81:472. [PMID: 39625546 PMCID: PMC11615175 DOI: 10.1007/s00018-024-05511-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 11/08/2024] [Accepted: 11/12/2024] [Indexed: 12/06/2024]
Abstract
MTSS1 is a ubiquitously expressed intracellular protein known mainly for its involvement in basic cellular processes, such as the regulation of actin organization and membrane architecture. MTSS1 has attracted much attention for its role as a tumor suppressor, being absent or expressed at reduced levels in advanced and metastasizing cancers. Occasionally, MTSS1 is, instead, upregulated in metastasis and, in some cases, even in primary tumors. In addition to these well-established functions of MTSS1 linked to its I-BAR- and WH2-domains, the protein is involved in modulating cell-cell contacts, cell differentiation, lipid metabolism, and vesicle formation and acts as a scaffolding protein for several E3 ubiquitin ligases. MTSS1 is classified as a housekeeping protein and is never mutated despite the several pathologic phenotypes linked to its dysregulation. Despite MTSS1's involvement in fundamental signaling pathways, MTSS1 gene ablation is not ubiquitously lethal, although it affects embryonic development. Due to MTSS1´s involvement in many seemingly disparate processes, with many cases lacking mechanistic explanations, we found it timely to review the recent data on MTSS1's role at the cellular level, as well as in health and disease, to direct further studies on this interesting multifunctional protein.
Collapse
Affiliation(s)
- Liudmila Matskova
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, FE 280, 17177, Sweden
| | - Shixing Zheng
- ENT Institute, Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, 200031, China
| | - Elena Kashuba
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, FE 280, 17177, Sweden
- RE Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology of National Academy of Sciences of Ukraine, Kyiv, 03022, Ukraine
| | - Ingemar Ernberg
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, FE 280, 17177, Sweden.
| | - Pontus Aspenström
- Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, 75185, Sweden.
| |
Collapse
|
2
|
Sanati M, Afshari AR, Ahmadi SS, Moallem SA, Sahebkar A. Modulation of the ubiquitin-proteasome system by phytochemicals: Therapeutic implications in malignancies with an emphasis on brain tumors. Biofactors 2023; 49:782-819. [PMID: 37162294 DOI: 10.1002/biof.1958] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 04/20/2023] [Indexed: 05/11/2023]
Abstract
Regarding the multimechanistic nature of cancers, current chemo- or radiotherapies often fail to eradicate disease pathology, and frequent relapses or resistance to therapies occur. Brain malignancies, particularly glioblastomas, are difficult-to-treat cancers due to their highly malignant and multidimensional biology. Unfortunately, patients suffering from malignant tumors often experience poor prognoses and short survival periods. Thus far, significant efforts have been conducted to discover novel and more effective modalities. To that end, modulation of the ubiquitin-proteasome system (UPS) has attracted tremendous interest since it affects the homeostasis of proteins critically engaged in various cell functions, for example, cell metabolism, survival, proliferation, and differentiation. With their safe and multimodal actions, phytochemicals are among the promising therapeutic tools capable of turning the operation of various UPS elements. The present review, along with an updated outline of the role of UPS dysregulation in multiple cancers, provided a detailed discussion on the impact of phytochemicals on the UPS function in malignancies, especially brain tumors.
Collapse
Affiliation(s)
- Mehdi Sanati
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Birjand University of Medical Sciences, Birjand, Iran
- Experimental and Animal Study Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Amir R Afshari
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
- Department of Physiology and Pharmacology, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Seyed Sajad Ahmadi
- Department of Ophthalmology, Khatam-Ol-Anbia Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Adel Moallem
- Department of Pharmacology and Toxicology, College of Pharmacy, Al-Zahraa University for Women, Karbala, Iraq
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
3
|
Fhu CW, Ali A. Dysregulation of the Ubiquitin Proteasome System in Human Malignancies: A Window for Therapeutic Intervention. Cancers (Basel) 2021; 13:cancers13071513. [PMID: 33805973 PMCID: PMC8037609 DOI: 10.3390/cancers13071513] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/22/2021] [Accepted: 03/22/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary The ubiquitin proteasome system (UPS) governs the non-lysosomal degradation of oxidized, damaged, or misfolded proteins in eukaryotic cells. Dysregulation of the UPS results in loss of ability to maintain protein quality through proteolysis, and is closely related to the development of various malignancies and tumorigenesis. Here, we provide a comprehensive general overview on the regulation and roles of UPS and discuss the mechanisms linking dysregulated UPS to human malignancies. Inhibitors developed against components of the UPS, which include U.S. Food and Drug Administration FDA-approved and those currently undergoing clinical trials, are also presented in this review. Abstract The ubiquitin proteasome system (UPS) governs the non-lysosomal degradation of oxidized, damaged, or misfolded proteins in eukaryotic cells. This process is tightly regulated through the activation and transfer of polyubiquitin chains to target proteins which are then recognized and degraded by the 26S proteasome complex. The role of UPS is crucial in regulating protein levels through degradation to maintain fundamental cellular processes such as growth, division, signal transduction, and stress response. Dysregulation of the UPS, resulting in loss of ability to maintain protein quality through proteolysis, is closely related to the development of various malignancies and tumorigenesis. Here, we provide a comprehensive general overview on the regulation and roles of UPS and discuss functional links of dysregulated UPS in human malignancies. Inhibitors developed against components of the UPS, which include U.S. Food and Drug Administration FDA-approved and those currently undergoing clinical trials, are also presented in this review.
Collapse
|
4
|
Carrara GFA, Evangelista AF, Scapulatempo-Neto C, Abrahão-Machado LF, Morini MA, Kerr LM, Folgueira MAAK, da Costa Vieira RA. Analysis of RPL37A, MTSS1, and HTRA1 expression as potential markers for pathologic complete response and survival. Breast Cancer 2021; 28:307-320. [PMID: 32951185 DOI: 10.1007/s12282-020-01159-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 09/11/2020] [Indexed: 02/05/2023]
Abstract
BACKGROUND Non-metastatic locally advanced breast carcinoma (LABC) treatment involves neoadjuvant chemotherapy (NCT). We evaluated the association of clinical-pathological data and immunoexpression of hormone receptors, HER2 and Ki67, and new biomarkers, RPL37A, MTSS1 and HTRA1, with pathological complete response (PCR) or tumour resistance (stable disease or disease progression), disease-free survival (DFS) and cancer-specific survival (CSS). METHODS This is a retrospective study of 333 patients with LABC who underwent NCT. Expression of MTSS1, RPL37A and HTRA1/PRSS11 was evaluated by immunohistochemistry in TMA slides. Cutoff values were established for low and high tumour expression. ROC plotter evaluated response to NCT. Chi-square test for factors related to PCR, and Kaplan-Meier test and Cox model for factors related to DFS and CSS were prformed. RESULTS The mean follow-up was 70.0 months and PCR rate was 15.6%. At 120 months, DFS rate was 32.5% and CSS rate was 67.1%. In multivariate analysis, there was an association between: (1) necrosis presence, intense inflammatory infiltrate, ER absence, HER2 molecular subtype and high RPL3A expression with increased odds of PCR; (2) lymph node involvement (LNI), high Ki67, low RPL37A and high HTRA1 expression with increased risk for NCT non-response; (3) LNI, high proliferation, necrosis absence, low RPL37A and high HTRA1 expression with increased recurrence risk; (4) advanced LNI, ER negative tumours, high HTRA1, low RPL37A expression and desmoplasia presence with higher risk of cancer death. CONCLUSION RPL37A is a potential biomarker for response to NCT and for prognosis. Additional studies evaluating HTRA1 and MTSS1 prognostic value are needed.
Collapse
Affiliation(s)
- Guilherme Freire Angotti Carrara
- Programa de Pós-Graduação em Oncologia, Hospital de Câncer de Barretos, Rua Antenor Duarte Villela, 1331, Bairro Dr Paulo Prata, Barretos, São Paulo, 14.784-400, Brasil
| | - Adriane Feijo Evangelista
- Programa de Pós-Graduação em Oncologia, Hospital de Câncer de Barretos, Rua Antenor Duarte Villela, 1331, Bairro Dr Paulo Prata, Barretos, São Paulo, 14.784-400, Brasil
- Centro de Pesquisa Molecular em Oncologia, Hospital de Câncer de Barretos, Barretos, Brasil
| | - Cristovam Scapulatempo-Neto
- Programa de Pós-Graduação em Oncologia, Hospital de Câncer de Barretos, Rua Antenor Duarte Villela, 1331, Bairro Dr Paulo Prata, Barretos, São Paulo, 14.784-400, Brasil
- Departamento de Patologia, Hospital de Câncer de Barretos, Barretos, Brasil
| | | | | | - Ligia Maria Kerr
- Departamento de Patologia, Hospital de Câncer de Barretos, Barretos, Brasil
| | - Maria Aparecida Azevedo Koike Folgueira
- Programa de Pós-Graduação em Oncologia, Departamento de Radiologia e Oncologia, Faculdade de Medicina, FMUSP Universidade de São Paulo, FMUSP, São Paulo, Brasil
| | - René Aloisio da Costa Vieira
- Programa de Pós-Graduação em Oncologia, Hospital de Câncer de Barretos, Rua Antenor Duarte Villela, 1331, Bairro Dr Paulo Prata, Barretos, São Paulo, 14.784-400, Brasil.
- Programa de Pós-Graduação em Ginecologia, Obstetricia e Mastologia, Faculdade de Medicina de Botucatu/UNESP, Botucatu, São Paulo, Brasil.
| |
Collapse
|
5
|
Samaržija I. Post-Translational Modifications That Drive Prostate Cancer Progression. Biomolecules 2021; 11:247. [PMID: 33572160 PMCID: PMC7915076 DOI: 10.3390/biom11020247] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 02/04/2021] [Accepted: 02/06/2021] [Indexed: 02/07/2023] Open
Abstract
While a protein primary structure is determined by genetic code, its specific functional form is mostly achieved in a dynamic interplay that includes actions of many enzymes involved in post-translational modifications. This versatile repertoire is widely used by cells to direct their response to external stimuli, regulate transcription and protein localization and to keep proteostasis. Herein, post-translational modifications with evident potency to drive prostate cancer are explored. A comprehensive list of proteome-wide and single protein post-translational modifications and their involvement in phenotypic outcomes is presented. Specifically, the data on phosphorylation, glycosylation, ubiquitination, SUMOylation, acetylation, and lipidation in prostate cancer and the enzymes involved are collected. This type of knowledge is especially valuable in cases when cancer cells do not differ in the expression or mutational status of a protein, but its differential activity is regulated on the level of post-translational modifications. Since their driving roles in prostate cancer, post-translational modifications are widely studied in attempts to advance prostate cancer treatment. Current strategies that exploit the potential of post-translational modifications in prostate cancer therapy are presented.
Collapse
Affiliation(s)
- Ivana Samaržija
- Laboratory for Epigenomics, Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia
| |
Collapse
|
6
|
Yamashita M, Owa T, Shiraishi R, Adachi T, Ichijo K, Taya S, Miyashita S, Hoshino M. The role of SCF Skp2 and SCF β-TrCP1/2 in the cerebellar granule cell precursors. Genes Cells 2020; 25:796-810. [PMID: 33020978 DOI: 10.1111/gtc.12813] [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: 07/09/2020] [Revised: 09/25/2020] [Accepted: 09/28/2020] [Indexed: 12/01/2022]
Abstract
A proper balance between proliferation and differentiation of cerebellar granule cell precursors (GCPs) is required for appropriate cerebellar morphogenesis. The Skp1-Cullin1-F-box (SCF) complex, an E3 ubiquitin ligase complex, is involved in polyubiquitination and subsequent degradation of various cell cycle regulators and transcription factors. However, it remains unknown how the SCF complex affects proliferation and differentiation of GCPs. In this study, we found that the scaffold protein Cullin1, and F-box proteins Skp2, β-TrCP1 and β-TrCP2 are expressed in the external granule layer (EGL). Knockdown of these molecules in the EGL showed that Cullin1, Skp2 and β-TrCP2 enhanced differentiation of GCPs. We also observed accumulation of cyclin-dependent kinase inhibitor p27 in GCPs when treated with a Cullin1 inhibitor or proteasome inhibitor. Furthermore, knockdown of p27 rescued enhancement of differentiation by Cullin1 knockdown. These results suggest that the SCF complex is involved in the maintenance of the proliferative state of GCPs through p27 degradation. In addition, inhibition of Cullin1 activity also prevented cell proliferation and enhanced accumulation of p27 in Daoy cells, a cell line derived from the sonic hedgehog subtype of medulloblastoma. This suggested that excess degradation of p27 through the SCF complex causes overproliferation of medulloblastoma cells.
Collapse
Affiliation(s)
- Mariko Yamashita
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, NCNP, Tokyo, Japan
| | - Tomoo Owa
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, NCNP, Tokyo, Japan
| | - Ryo Shiraishi
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, NCNP, Tokyo, Japan.,Department of NCNP Brain Function and Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, TMDU, Tokyo, Japan
| | - Toma Adachi
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, NCNP, Tokyo, Japan.,Department of Life Science and Medical Bioscience, Graduate School of Advance Science and Engineering, TWIns, Waseda University, Tokyo, Japan
| | - Kentaro Ichijo
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, NCNP, Tokyo, Japan.,Department of Otolaryngology and Head and Neck Surgery, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shinichiro Taya
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, NCNP, Tokyo, Japan
| | - Satoshi Miyashita
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, NCNP, Tokyo, Japan
| | - Mikio Hoshino
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, NCNP, Tokyo, Japan
| |
Collapse
|
7
|
Celebi G, Kesim H, Ozer E, Kutlu O. The Effect of Dysfunctional Ubiquitin Enzymes in the Pathogenesis of Most Common Diseases. Int J Mol Sci 2020; 21:ijms21176335. [PMID: 32882786 PMCID: PMC7503467 DOI: 10.3390/ijms21176335] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 07/17/2020] [Accepted: 07/18/2020] [Indexed: 12/14/2022] Open
Abstract
Ubiquitination is a multi-step enzymatic process that involves the marking of a substrate protein by bonding a ubiquitin and protein for proteolytic degradation mainly via the ubiquitin–proteasome system (UPS). The process is regulated by three main types of enzymes, namely ubiquitin-activating enzymes (E1), ubiquitin-conjugating enzymes (E2), and ubiquitin ligases (E3). Under physiological conditions, ubiquitination is highly reversible reaction, and deubiquitinases or deubiquitinating enzymes (DUBs) can reverse the effect of E3 ligases by the removal of ubiquitin from substrate proteins, thus maintaining the protein quality control and homeostasis in the cell. The dysfunction or dysregulation of these multi-step reactions is closely related to pathogenic conditions; therefore, understanding the role of ubiquitination in diseases is highly valuable for therapeutic approaches. In this review, we first provide an overview of the molecular mechanism of ubiquitination and UPS; then, we attempt to summarize the most common diseases affecting the dysfunction or dysregulation of these mechanisms.
Collapse
Affiliation(s)
- Gizem Celebi
- Faculty of Engineering and Natural Sciences, Molecular Biology, Genetics, and Bioengineering Program, Sabanci University, Istanbul 34956, Turkey; (G.C.); (H.K.); (E.O.)
| | - Hale Kesim
- Faculty of Engineering and Natural Sciences, Molecular Biology, Genetics, and Bioengineering Program, Sabanci University, Istanbul 34956, Turkey; (G.C.); (H.K.); (E.O.)
| | - Ebru Ozer
- Faculty of Engineering and Natural Sciences, Molecular Biology, Genetics, and Bioengineering Program, Sabanci University, Istanbul 34956, Turkey; (G.C.); (H.K.); (E.O.)
| | - Ozlem Kutlu
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Istanbul 34956, Turkey
- Center of Excellence for Functional Surfaces and Interfaces for Nano Diagnostics (EFSUN), Sabanci University, Istanbul 34956, Turkey
- Correspondence: ; Tel.: +90-216-483-9000 (ext. 2413)
| |
Collapse
|
8
|
Lin YC, Chen MC, Hsieh TH, Liou JP, Chen CH. CK1δ as a potential therapeutic target to treat bladder cancer. Aging (Albany NY) 2020; 12:5764-5780. [PMID: 32282334 PMCID: PMC7185098 DOI: 10.18632/aging.102966] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Accepted: 01/27/2020] [Indexed: 02/06/2023]
Abstract
Bladder cancer is the second most common genitourinary malignancy in the world. However, only immune-checkpoint inhibitors and erdafitinib are available to treat advanced bladder cancer. Our previous study reported that 4-((4-(4-ethylpiperazin-1-yl) phenyl)amino)-N-(3,4,5-trichlorophenyl)-7H-pyrrolo-[2, 3-d]pyrimidine-7-carboxamide hydrochloride (13i HCl) is a potent CK1δ inhibitor showing significant anti-bladder cancer activity. In this study, we elucidated the pharmacological mechanisms underlying 13i HCl’s inhibition of human bladder cancer. Our results demonstrate that expression of the CSNK1D gene, which codes for CK1δ, is upregulated in superficial and infiltrating bladder cancer patients in two independent datasets. CK1δ knockdown decreased β-catenin expression in bladder cancer cells and inhibited their growth. Additionally, 13i HCl suppressed bladder cancer cell proliferation and increased apoptosis. We also observed that inhibition of CK1δ using 13i HCl or PF-670462 triggers necroptosis in bladder cancer cells. Finally, 13i HCl inhibited bladder cancer cell migration and reversed their mesenchymal characteristics. These findings suggest further development of 13i HCl as a potential therapeutic agent to treat bladder cancer is warranted.
Collapse
Affiliation(s)
- Yu-Chen Lin
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Mei-Chuan Chen
- Ph.D. Program in Clinical Drug Development of Herbal Medicine, College of Pharmacy, Taipei Medical University, Taipei, Taiwan.,Traditional Herbal Medicine Research Center of Taipei Medical University Hospital, Taipei, Taiwan
| | - Tsung-Han Hsieh
- Joint Biobank, Office of Human Research, Taipei Medical University, Taipei, Taiwan
| | - Jing-Ping Liou
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Chun-Han Chen
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.,TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
9
|
Lou Y, Han M, Song Y, Zhong J, Zhang W, Chen YH, Wang H. The SCF β-TrCP E3 Ubiquitin Ligase Regulates Immune Receptor Signaling by Targeting the Negative Regulatory Protein TIPE2. THE JOURNAL OF IMMUNOLOGY 2020; 204:2122-2132. [PMID: 32188758 DOI: 10.4049/jimmunol.1901142] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 02/07/2020] [Indexed: 12/11/2022]
Abstract
TNFAIP8-like 2 (TIPE2) is a negative regulator of immune receptor signaling that maintains immune homeostasis. Dysregulated TIPE2 expression has been observed in several types of human immunological disorders. However, how TIPE2 expression is regulated remains to be determined. We report in this study that the SCFβ-TrCP E3 ubiquitin ligase regulates TIPE2 protein abundance by targeting it for ubiquitination and subsequent degradation via the 26S proteasome. Silencing of either cullin-1 or β-TrCP1 resulted in increased levels of TIPE2 in immune cells. TAK1 phosphorylated the Ser3 in the noncanonical degron motif of TIPE2 to trigger its interaction with β-TrCP for subsequent ubiquitination and degradation. Importantly, the amount of TIPE2 protein in immune cells determined the strength of TLR 4-induced signaling and downstream gene expression. Thus, our study has uncovered a mechanism by which SCFβ-TrCP E3 ubiquitin ligase regulates TLR responses.
Collapse
Affiliation(s)
- Yunwei Lou
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, Henan 453003, People's Republic of China.,Henan Key Laboratory of Immunology and Targeted Drugs, Xinxiang Medical University, Xinxiang, Henan 453003, People's Republic of China
| | - Meijuan Han
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, Henan 453003, People's Republic of China.,Henan Key Laboratory of Immunology and Targeted Drugs, Xinxiang Medical University, Xinxiang, Henan 453003, People's Republic of China
| | - Yaru Song
- Department of Pulmonary Medicine, The Affiliated Renmin Hospital of Xinxiang Medical University, Xinxiang, Henan 453100, People's Republic of China
| | - Jiateng Zhong
- Department of Pathology, Xinxiang Medical University, Xinxiang, Henan 453003, People's Republic of China; and
| | - Wen Zhang
- Henan Key Laboratory of Immunology and Targeted Drugs, Xinxiang Medical University, Xinxiang, Henan 453003, People's Republic of China
| | - Youhai H Chen
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
| | - Hui Wang
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, Henan 453003, People's Republic of China; .,Henan Key Laboratory of Immunology and Targeted Drugs, Xinxiang Medical University, Xinxiang, Henan 453003, People's Republic of China
| |
Collapse
|
10
|
Tekcham DS, Chen D, Liu Y, Ling T, Zhang Y, Chen H, Wang W, Otkur W, Qi H, Xia T, Liu X, Piao HL, Liu H. F-box proteins and cancer: an update from functional and regulatory mechanism to therapeutic clinical prospects. Am J Cancer Res 2020; 10:4150-4167. [PMID: 32226545 PMCID: PMC7086354 DOI: 10.7150/thno.42735] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 02/04/2020] [Indexed: 12/16/2022] Open
Abstract
E3 ubiquitin ligases play a critical role in cellular mechanisms and cancer progression. F-box protein is the core component of the SKP1-cullin 1-F-box (SCF)-type E3 ubiquitin ligase and directly binds to substrates by various specific domains. According to the specific domains, F-box proteins are further classified into three sub-families: 1) F-box with leucine rich amino acid repeats (FBXL); 2) F-box with WD 40 amino acid repeats (FBXW); 3) F-box only with uncharacterized domains (FBXO). Here, we summarize the substrates of F-box proteins, discuss the important molecular mechanism and emerging role of F-box proteins especially from the perspective of cancer development and progression. These findings will shed new light on malignant tumor progression mechanisms, and suggest the potential role of F-box proteins as cancer biomarkers and therapeutic targets for future cancer treatment.
Collapse
|
11
|
Cai N, Chen Z, Huang Y, Shao S, Yu H, Wang Y, He S. β-TrCP1 promotes cell proliferation via TNF-dependent NF-κB activation in diffuse large B cell lymphoma. Cancer Biol Ther 2019; 21:241-247. [PMID: 31731887 DOI: 10.1080/15384047.2019.1683332] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
Diffuse large B cell lymphoma (DLBCL), a heterogeneous group of invasive disease, is the most common type of B-cell non-Hodgkin's lymphomas. The mechanism of its development is closely related to the constitutive activation of NF-κB. In this study, we investigated the function and the mechanism of β-TRCP1 in DLBCL. CCK8 and EdU assays showed that β-TRCP1 could promote the growth of DLBCL cells under the stimulation of TNFα. Furthermore, overexpression of β-TRCP1 enhanced NF-κB activation in the presence of TNFα. Moreover, ectopic expression of β-TRCP1 decreased IκB-α expression but increased phospho-p65 expression. In addition, β-TRCP1 promoted cell cycle progression by accelerating G1-S phase transition. We also found that silencing of β-TrCP1 increased mitoxantrone-induced cell growth arrest and apoptosis. Based on these, we proposed that the expression of β-TRCP1 promoted cell proliferation via TNF-dependent NF-κB activation in DLBCL cells.
Collapse
Affiliation(s)
- Nannan Cai
- Department of Pathogenic Biology, Medical College, Nantong University, Nantong, Jiangsu, People's Republic of China.,Department of Pathology, Affiliated Tumor Hospital of Nantong University, Nantong, Jiangsu, People's Republic of China.,Department of gynaecology and obstetrics, Nantong Tongzhou People's Hospital, Nantong, Jiangsu, China
| | - Zhuolin Chen
- Department of Pathology, Affiliated Tumor Hospital of Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Yuejiao Huang
- Department of Pathology, Affiliated Tumor Hospital of Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Shan Shao
- Department of Hematology, Shanghai General Hospital, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | - Haiyan Yu
- Department of Pathology, Affiliated Tumor Hospital of Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Yuchan Wang
- Department of Pathogenic Biology, Medical College, Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Song He
- Department of Pathology, Affiliated Tumor Hospital of Nantong University, Nantong, Jiangsu, People's Republic of China
| |
Collapse
|
12
|
Xu P, Ianes C, Gärtner F, Liu C, Burster T, Bakulev V, Rachidi N, Knippschild U, Bischof J. Structure, regulation, and (patho-)physiological functions of the stress-induced protein kinase CK1 delta (CSNK1D). Gene 2019; 715:144005. [PMID: 31376410 PMCID: PMC7939460 DOI: 10.1016/j.gene.2019.144005] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 07/19/2019] [Accepted: 07/23/2019] [Indexed: 12/11/2022]
Abstract
Members of the highly conserved pleiotropic CK1 family of serine/threonine-specific kinases are tightly regulated in the cell and play crucial regulatory roles in multiple cellular processes from protozoa to human. Since their dysregulation as well as mutations within their coding regions contribute to the development of various different pathologies, including cancer and neurodegenerative diseases, they have become interesting new drug targets within the last decade. However, to develop optimized CK1 isoform-specific therapeutics in personalized therapy concepts, a detailed knowledge of the regulation and functions of the different CK1 isoforms, their various splice variants and orthologs is mandatory. In this review we will focus on the stress-induced CK1 isoform delta (CK1δ), thereby addressing its regulation, physiological functions, the consequences of its deregulation for the development and progression of diseases, and its potential as therapeutic drug target.
Collapse
Affiliation(s)
- Pengfei Xu
- Department of General and Visceral Surgery, Surgery Center, Ulm University Hospital, Albert-Einstein-Allee 23, 89081 Ulm, Germany.
| | - Chiara Ianes
- Department of General and Visceral Surgery, Surgery Center, Ulm University Hospital, Albert-Einstein-Allee 23, 89081 Ulm, Germany.
| | - Fabian Gärtner
- Department of General and Visceral Surgery, Surgery Center, Ulm University Hospital, Albert-Einstein-Allee 23, 89081 Ulm, Germany.
| | - Congxing Liu
- Department of General and Visceral Surgery, Surgery Center, Ulm University Hospital, Albert-Einstein-Allee 23, 89081 Ulm, Germany.
| | - Timo Burster
- Department of Biology, School of Science and Technology, Nazarbayev University, 53 Kabanbay Batyr Ave, Nur-Sultan 020000, Kazakhstan.
| | - Vasiliy Bakulev
- Ural Federal University named after the first President of Russia B. N. Eltsin, Technology for Organic Synthesis Laboratory, 19 Mirastr., 620002 Ekaterinburg, Russia.
| | - Najma Rachidi
- Unité de Parasitologie Moléculaire et Signalisation, Department of Parasites and Insect Vectors, Institut Pasteur and INSERM U1201, 25-28 Rue du Dr Roux, 75015 Paris, France.
| | - Uwe Knippschild
- Department of General and Visceral Surgery, Surgery Center, Ulm University Hospital, Albert-Einstein-Allee 23, 89081 Ulm, Germany.
| | - Joachim Bischof
- Department of General and Visceral Surgery, Surgery Center, Ulm University Hospital, Albert-Einstein-Allee 23, 89081 Ulm, Germany.
| |
Collapse
|
13
|
Chen J, Huang L, Zhu Q, Wang Z, Tang Z. MTSS1 hypermethylation is associated with prostate cancer progression. J Cell Physiol 2019; 235:2687-2697. [PMID: 31541465 DOI: 10.1002/jcp.29172] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 08/23/2019] [Indexed: 01/08/2023]
Abstract
This study was conducted to evaluate the influence of DNA methylation of metastasis suppressor 1 (MTSS1) on prostate cancer (PCa) progression. Forty-nine paired PCa tissue samples and normal tissue samples from The Cancer Genome Atlas were analyzed. Methylome analysis, CpG island arrays and Hierarchical clustering were used to analyze methylation profiles of PCa tissues. MTSS1 methylation level was detected by methylation-specific PCR. Relative messenger RNA and the expression level of MTSS1 protein were identified by quantitative real-time PCR (qRT-PCR) and western blot analysis. The migration, invasion, proliferation, and cell cycle were detected separately by wound-healing assay, transwell chamber assay, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide assay and flow cytometry. The roles of MTSS1 in PCa progression were demonstrated in vivo by tumor formation assays in nude mice. MTSS1 expression was decreased in PCa tissues in comparison with paired adjacent normal prostate tissues. Compared to the methylation of MTSS1 in normal prostate tissues based on the MethHC website, the MTSS1 in PCa tissues was hypermethylated. The expression of MTSS1 detected by qRT-PCR and western blot analysis was found to be downregulated in PCa cells and tissues. The reduced expression of MTSS1 by small interfering RNA-MTSS1 was recovered by 5-aza-2'-deoxycytidine treatment. Besides, MTSS1 demethylation inhibited migration, invasion, and proliferation of PCa cells, and induced cell cycle to be arrested at G0/G1 phase. Furthermore, it was shown by tumor xenograft assay that MTSS1 inhibited the growth of tumor in vivo. Hypermethylated MTSS1 promoted PCa cells migration, invasion, and proliferation, and suppressed cell cycle arrest at the G0/G1 phase.
Collapse
Affiliation(s)
- Junjie Chen
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Liang Huang
- Department of Urology, Hunan Provincial People's Hospital, Changsha, Hunan, China
| | - Quan Zhu
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhao Wang
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhengyan Tang
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Hunan Engineering Laboratory for Diagnosis and Treatment Technology of Urogenital Diseases, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
14
|
Hung MS, Chen YC, Lin P, Li YC, Hsu CC, Lung JH, You L, Xu Z, Mao JH, Jablons DM, Yang CT. Cul4A Modulates Invasion and Metastasis of Lung Cancer Through Regulation of ANXA10. Cancers (Basel) 2019; 11:618. [PMID: 31052599 PMCID: PMC6562482 DOI: 10.3390/cancers11050618] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 04/15/2019] [Accepted: 04/24/2019] [Indexed: 02/06/2023] Open
Abstract
: Cullin 4A (Cul4A) is overexpressed in a number of cancers and has been established as an oncogene. This study aimed to elucidate the role of Cul4A in lung cancer invasion and metastasis. We observed that Cul4A was overexpressed in non-small cell lung cancer (NSCLC) tissues and the overexpression of Cul4A was associated with poor prognosis after surgical resection and it also decreased the expression of the tumor suppressor protein annexin A10 (ANXA10). The knockdown of Cul4A was associated with the upregulation of ANXA10, and the forced expression of Cul4A was associated with the downregulation of ANXA10 in lung cancer cells. Further studies showed that the knockdown of Cul4A inhibited the invasion and metastasis of lung cancer cells, which was reversed by the further knockdown of ANXA10. In addition, the knockdown of Cul4A inhibited lung tumor metastasis in mouse tail vein injection xenograft models. Notably, Cul4A regulated the degradation of ANXA10 through its interaction with ANXA10 and ubiquitination in lung cancer cells. Our findings suggest that Cul4A is a prognostic marker in NSCLC patients, and Cul4A plays important roles in lung cancer invasion and metastasis through the regulation of the ANXA10 tumor suppressor.
Collapse
Affiliation(s)
- Ming-Szu Hung
- Division of Thoracic Oncology, Department of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Chiayi branch 61363, Taiwan.
- Department of Medicine, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan.
- Department of Respiratory Care, Chang Gung University of Science and Technology, Chiayi Campus, Chiayi 61363, Taiwan.
| | - Yi-Chuan Chen
- Department of Emergency Medicine, Chang Gung Memorial Hospital, Chiayi branch 61363, Taiwan.
| | - PaulYann Lin
- Department of Anatomic Pathology, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi 62247, Taiwan.
| | - Ya-Chin Li
- Division of Thoracic Oncology, Department of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Chiayi branch 61363, Taiwan.
| | - Chia-Chen Hsu
- Department of Hematology and Oncology, Chang Gung Memorial Hospital, Chiayi branch 61363, Taiwan.
- Department of Medical Research and Development, Chang Gung Memorial Hospital, Chiayi branch 61363, Taiwan.
| | - Jr-Hau Lung
- Department of Medical Research and Development, Chang Gung Memorial Hospital, Chiayi branch 61363, Taiwan.
| | - Liang You
- Thoracic Oncology Laboratory, Department of Surgery, Comprehensive Cancer Center, University of California, San Francisco, CA 94143, USA.
| | - Zhidong Xu
- Thoracic Oncology Laboratory, Department of Surgery, Comprehensive Cancer Center, University of California, San Francisco, CA 94143, USA.
| | - Jian-Hua Mao
- Life Sciences Division, Lawrence Berkeley National Laboratory, One Cyclotron Road, Berkeley, CA 94720, USA.
| | - David M Jablons
- Thoracic Oncology Laboratory, Department of Surgery, Comprehensive Cancer Center, University of California, San Francisco, CA 94143, USA.
| | - Cheng-Ta Yang
- Department of Respiratory Care, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan.
- Department of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Taoyuan branch 33378, Taiwan.
| |
Collapse
|
15
|
Petrov P, Sarapulov AV, Eöry L, Scielzo C, Scarfò L, Smith J, Burt DW, Mattila PK. Computational analysis of the evolutionarily conserved Missing In Metastasis/Metastasis Suppressor 1 gene predicts novel interactions, regulatory regions and transcriptional control. Sci Rep 2019; 9:4155. [PMID: 30858428 PMCID: PMC6411742 DOI: 10.1038/s41598-019-40697-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 02/21/2019] [Indexed: 12/25/2022] Open
Abstract
Missing in Metastasis (MIM), or Metastasis Suppressor 1 (MTSS1), is a highly conserved protein, which links the plasma membrane to the actin cytoskeleton. MIM has been implicated in various cancers, however, its modes of action remain largely enigmatic. Here, we performed an extensive in silico characterisation of MIM to gain better understanding of its function. We detected previously unappreciated functional motifs including adaptor protein (AP) complex interaction site and a C-helix, pointing to a role in endocytosis and regulation of actin dynamics, respectively. We also identified new functional regions, characterised with phosphorylation sites or distinct hydrophilic properties. Strong negative selection during evolution, yielding high conservation of MIM, has been combined with positive selection at key sites. Interestingly, our analysis of intra-molecular co-evolution revealed potential regulatory hotspots that coincided with reduced potentially pathogenic polymorphisms. We explored databases for the mutations and expression levels of MIM in cancer. Experimentally, we focused on chronic lymphocytic leukaemia (CLL), where MIM showed high overall expression, however, downregulation on poor prognosis samples. Finally, we propose strong conservation of MTSS1 also on the transcriptional level and predict novel transcriptional regulators. Our data highlight important targets for future studies on the role of MIM in different tissues and cancers.
Collapse
Affiliation(s)
- Petar Petrov
- Institute of Biomedicine, and MediCity Research Laboratories, University of Turku, Tykistökatu 6A, 20520, Turku, Finland.
| | - Alexey V Sarapulov
- Institute of Biomedicine, and MediCity Research Laboratories, University of Turku, Tykistökatu 6A, 20520, Turku, Finland
| | - Lel Eöry
- Division of Genetics and Genomics, The Roslin Institute and R(D)SVS, University of Edinburgh, Roslin, Easter Bush campus, Midlothian, EH25 9RG, United Kingdom
| | - Cristina Scielzo
- Unit of B Cell Neoplasia, Division of Molecular Oncology, IRCCS, San Raffaele Scientific Institute, Milano, Italy.,Università Vita-Salute San Raffaele, Milan, Italy
| | - Lydia Scarfò
- Unit of B Cell Neoplasia, Division of Molecular Oncology, IRCCS, San Raffaele Scientific Institute, Milano, Italy.,Università Vita-Salute San Raffaele, Milan, Italy.,Strategic Research Program on CLL, Division of Experimental Oncology, IRCCS, San Raffaele Scientific Institute, Milano, Italy
| | - Jacqueline Smith
- Division of Genetics and Genomics, The Roslin Institute and R(D)SVS, University of Edinburgh, Roslin, Easter Bush campus, Midlothian, EH25 9RG, United Kingdom
| | - David W Burt
- University of Queensland, St. Lucia, QLD, 4072, Australia
| | - Pieta K Mattila
- Institute of Biomedicine, and MediCity Research Laboratories, University of Turku, Tykistökatu 6A, 20520, Turku, Finland.
| |
Collapse
|
16
|
Fu H, Yang Y, Wang X, Wang H, Xu Y. DeepUbi: a deep learning framework for prediction of ubiquitination sites in proteins. BMC Bioinformatics 2019; 20:86. [PMID: 30777029 PMCID: PMC6379983 DOI: 10.1186/s12859-019-2677-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 02/12/2019] [Indexed: 01/22/2023] Open
Abstract
Background Protein ubiquitination occurs when the ubiquitin protein binds to a target protein residue of lysine (K), and it is an important regulator of many cellular functions, such as signal transduction, cell division, and immune reactions, in eukaryotes. Experimental and clinical studies have shown that ubiquitination plays a key role in several human diseases, and recent advances in proteomic technology have spurred interest in identifying ubiquitination sites. However, most current computing tools for predicting target sites are based on small-scale data and shallow machine learning algorithms. Results As more experimentally validated ubiquitination sites emerge, we need to design a predictor that can identify lysine ubiquitination sites in large-scale proteome data. In this work, we propose a deep learning predictor, DeepUbi, based on convolutional neural networks. Four different features are adopted from the sequences and physicochemical properties. In a 10-fold cross validation, DeepUbi obtains an AUC (area under the Receiver Operating Characteristic curve) of 0.9, and the accuracy, sensitivity and specificity exceeded 85%. The more comprehensive indicator, MCC, reaches 0.78. We also develop a software package that can be freely downloaded from https://github.com/Sunmile/DeepUbi. Conclusion Our results show that DeepUbi has excellent performance in predicting ubiquitination based on large data. Electronic supplementary material The online version of this article (10.1186/s12859-019-2677-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hongli Fu
- Department of Information and Computing Science, University of Science and Technology Beijing, Beijing, 100083, China
| | - Yingxi Yang
- Department of Information and Computing Science, University of Science and Technology Beijing, Beijing, 100083, China
| | - Xiaobo Wang
- Department of Information and Computing Science, University of Science and Technology Beijing, Beijing, 100083, China
| | - Hui Wang
- Institute of Computing Technology, Chinese Academy of Sciences, Beijing, 100190, China
| | - Yan Xu
- Department of Information and Computing Science, University of Science and Technology Beijing, Beijing, 100083, China. .,Beijing Key Laboratory for Magneto-photoelectrical Composite and Interface Science, University of Science and Technology Beijing, Beijing, 100083, China.
| |
Collapse
|
17
|
Xu H, Wang Y, Lin S, Deng W, Peng D, Cui Q, Xue Y. PTMD: A Database of Human Disease-associated Post-translational Modifications. GENOMICS PROTEOMICS & BIOINFORMATICS 2018; 16:244-251. [PMID: 30244175 PMCID: PMC6205080 DOI: 10.1016/j.gpb.2018.06.004] [Citation(s) in RCA: 125] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 06/04/2018] [Accepted: 06/25/2018] [Indexed: 12/20/2022]
Abstract
Various posttranslational modifications (PTMs) participate in nearly all aspects of biological processes by regulating protein functions, and aberrant states of PTMs are frequently implicated in human diseases. Therefore, an integral resource of PTM–disease associations (PDAs) would be a great help for both academic research and clinical use. In this work, we reported PTMD, a well-curated database containing PTMs that are associated with human diseases. We manually collected 1950 known PDAs in 749 proteins for 23 types of PTMs and 275 types of diseases from the literature. Database analyses show that phosphorylation has the largest number of disease associations, whereas neurologic diseases have the largest number of PTM associations. We classified all known PDAs into six classes according to the PTM status in diseases and demonstrated that the upregulation and presence of PTM events account for a predominant proportion of disease-associated PTM events. By reconstructing a disease–gene network, we observed that breast cancers have the largest number of associated PTMs and AKT1 has the largest number of PTMs connected to diseases. Finally, the PTMD database was developed with detailed annotations and can be a useful resource for further analyzing the relations between PTMs and human diseases. PTMD is freely accessible at http://ptmd.biocuckoo.org.
Collapse
Affiliation(s)
- Haodong Xu
- Department of Bioinformatics & Systems Biology, MOE Key Laboratory of Molecular Biophysics, College of Life Science and Technology and the Collaborative Innovation Center for Biomedical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Yongbo Wang
- Department of Bioinformatics & Systems Biology, MOE Key Laboratory of Molecular Biophysics, College of Life Science and Technology and the Collaborative Innovation Center for Biomedical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Shaofeng Lin
- Department of Bioinformatics & Systems Biology, MOE Key Laboratory of Molecular Biophysics, College of Life Science and Technology and the Collaborative Innovation Center for Biomedical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Wankun Deng
- Department of Bioinformatics & Systems Biology, MOE Key Laboratory of Molecular Biophysics, College of Life Science and Technology and the Collaborative Innovation Center for Biomedical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Di Peng
- Department of Bioinformatics & Systems Biology, MOE Key Laboratory of Molecular Biophysics, College of Life Science and Technology and the Collaborative Innovation Center for Biomedical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Qinghua Cui
- Department of Biomedical Informatics, School of Basic Medical Sciences, MOE Key Laboratory of Molecular Cardiovascular Sciences, Center for Non-coding RNA Medicine, Peking University, Beijing 100191, China.
| | - Yu Xue
- Department of Bioinformatics & Systems Biology, MOE Key Laboratory of Molecular Biophysics, College of Life Science and Technology and the Collaborative Innovation Center for Biomedical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China.
| |
Collapse
|
18
|
MicroRNA-324-5p suppresses the migration and invasion of MM cells by inhibiting the SCF β-TrCP E3 ligase. Oncol Lett 2018; 16:5331-5338. [PMID: 30250603 DOI: 10.3892/ol.2018.9245] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 02/20/2018] [Indexed: 12/12/2022] Open
Abstract
Multiple myeloma (MM) is a cytogenetically heterogeneous malignancy of plasma cells in bone marrow. Among the cytogenetic abnormalities of MM, del(17p) is a well-recognized high-risk genetic lesion associated with the late stage and progression of the disease. MicroRNA (miR)-324-5p, located at 17p13.1, was identified to be involved in the dysregulation of a number of types of malignant disease. However, whether miR-324-5p is associated with the development and progression of MM remains unknown. In the present study, the expression status of miR-324-5p in MM, and its effect on the migratory and invasive ability of MM cells were investigated. Using ubiquitination pathway polymerase chain reaction array, the inhibitory effect of miR-324-5p on the ubiquitinated proteins was investigated. It was identified that miR-324-5p levels were decreased in samples from patients with MM and MM cell lines. Increased expression of miR-324-5p by transfection of miR-324-5p mimic suppressed the proliferative, migratory and invasive abilities of MM.1R cells. Furthermore, increased expression of miR-324-5p in MM.1R cells inhibited the ubiquitination pathway and decreased the levels of ubiquitination-associated proteins, particularly the Skp1-Cullin1-F-box β-transducin repeat-containing protein (SCFβ-TrCP) E3 ligase. In addition, the results of the present study demonstrated that the SCFβ-TrCP E3 ligase may contribute to the suppression of MM cell motility by inhibiting the expression of metastasis-associated genes, including metastasis suppressor 1. In conclusion, the results of the present study suggested that miR-324-5p may act as a tumor suppressor by impairing the motility of MM cells by suppressing the ubiquitination pathway.
Collapse
|
19
|
Behzad MM, Shahrabi S, Jaseb K, Bertacchini J, Ketabchi N, Saki N. Aberrant DNA Methylation in Chronic Myeloid Leukemia: Cell Fate Control, Prognosis, and Therapeutic Response. Biochem Genet 2018; 56:149-175. [DOI: 10.1007/s10528-018-9841-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 01/18/2018] [Indexed: 01/24/2023]
|
20
|
Bracalente C, Ibañez IL, Berenstein A, Notcovich C, Cerda MB, Klamt F, Chernomoretz A, Durán H. Reprogramming human A375 amelanotic melanoma cells by catalase overexpression: Upregulation of antioxidant genes correlates with regression of melanoma malignancy and with malignant progression when downregulated. Oncotarget 2018; 7:41154-41171. [PMID: 27206673 PMCID: PMC5173049 DOI: 10.18632/oncotarget.9273] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 04/02/2016] [Indexed: 12/18/2022] Open
Abstract
Reactive oxygen species (ROS) are implicated in tumor transformation. The antioxidant system (AOS) protects cells from ROS damage. However, it is also hijacked by cancers cells to proliferate within the tumor. Thus, identifying proteins altered by redox imbalance in cancer cells is an attractive prognostic and therapeutic tool. Gene expression microarrays in A375 melanoma cells with different ROS levels after overexpressing catalase were performed. Dissimilar phenotypes by differential compensation to hydrogen peroxide scavenging were generated. The melanotic A375-A7 (A7) upregulated TYRP1, CNTN1 and UCHL1 promoting melanogenesis. The metastatic A375-G10 (G10) downregulated MTSS1 and TIAM1, proteins absent in metastasis. Moreover, differential coexpression of AOS genes (EPHX2, GSTM3, MGST1, MSRA, TXNRD3, MGST3 and GSR) was found in A7 and G10. Their increase in A7 improved its AOS ability and therefore, oxidative stress response, resembling less aggressive tumor cells. Meanwhile, their decrease in G10 revealed a disruption in the AOS and therefore, enhanced its metastatic capacity. These gene signatures, not only bring new insights into the physiopathology of melanoma, but also could be relevant in clinical prognostic to classify between non aggressive and metastatic melanomas.
Collapse
Affiliation(s)
- Candelaria Bracalente
- Departamento de Micro y Nanotecnología, Comisión Nacional de Energía Atómica, San Martín, Buenos Aires, Argentina.,Consejo Nacional de Investigaciones Científicas y Tecnológicas, Buenos Aires, Argentina
| | - Irene L Ibañez
- Departamento de Micro y Nanotecnología, Comisión Nacional de Energía Atómica, San Martín, Buenos Aires, Argentina.,Consejo Nacional de Investigaciones Científicas y Tecnológicas, Buenos Aires, Argentina
| | - Ariel Berenstein
- Fundación Instituto Leloir and Departamento de Física, Facultad Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Cintia Notcovich
- Departamento de Micro y Nanotecnología, Comisión Nacional de Energía Atómica, San Martín, Buenos Aires, Argentina
| | - María B Cerda
- Departamento de Micro y Nanotecnología, Comisión Nacional de Energía Atómica, San Martín, Buenos Aires, Argentina
| | - Fabio Klamt
- Laboratório de Bioquímica Celular, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brasil
| | - Ariel Chernomoretz
- Fundación Instituto Leloir and Departamento de Física, Facultad Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Hebe Durán
- Departamento de Micro y Nanotecnología, Comisión Nacional de Energía Atómica, San Martín, Buenos Aires, Argentina.,Consejo Nacional de Investigaciones Científicas y Tecnológicas, Buenos Aires, Argentina.,Escuela de Ciencia y Tecnología, Universidad Nacional de San Martín, San Martín, Buenos Aires, Argentina
| |
Collapse
|
21
|
Xie W, Sun F, Chen L, Cao X. miR-96 promotes breast cancer metastasis by suppressing MTSS1. Oncol Lett 2018; 15:3464-3471. [PMID: 29456723 PMCID: PMC5795871 DOI: 10.3892/ol.2018.7728] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2016] [Accepted: 10/13/2017] [Indexed: 11/13/2022] Open
Abstract
Novel, non-invasive biomarkers with high sensitivity and specificity are critical for breast cancer treatment, and prognosis. MicroRNA (miR)-96 has been demonstrated to be highly expressed in several solid malignancies, including breast cancer. However, its expression and function in the metastasis and prognosis of breast cancer have not been fully explored, and its regulation mechanisms remain unclear. In the present study, the serum miR-96 expression in healthy controls, benign and malignant breast cancer types was compared by using reverse transcription-quantitative polymerase chain reaction. The effect of chemotherapy on miR-96 expression in breast cancer was also investigated. Result revealed that miR-96 expression was increased in malignant breast cancer types and reduced in patients following chemotherapy treatment. The effect of miR-96 manipulation on the migration of breast cancer cells was also investigated by using wound healing, and Transwell migration assays. These results revealed that the induced expression of miR96 led to enhanced wound closing and trans-membrane cell numbers. By using bioinformatics analysis, western blotting and immunohistochemical staining, the metastasis suppressor-1 (MTSS1) gene was identified to be the functional target of miR-96 in the promotion of cell migration. In conclusion, it was identified that miR-96 exhibited an increased level in serum samples of patients with malignant breast cancer in comparison with benign breast tumor types and health controls and may be substantially reduced by chemotherapy treatment, implying that it may be used as a prognostic marker in breast cancer. miR-96 overexpression may inhibit migration of breast cancer cells by downregulating MTSS1 expression.
Collapse
Affiliation(s)
- Wei Xie
- Department of Laboratory Medicine, Nantong First People's Hospital, The Second Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Feng Sun
- Department of Laboratory Medicine, Nantong Tumor Hospital, Nantong, Jiangsu 226361, P.R. China
| | - Lin Chen
- Department of Laboratory Medicine, Nantong Third People's Hospital, Nantong, Jiangsu 226000, P.R. China
| | - Xinjian Cao
- Department of Laboratory Medicine, Nantong First People's Hospital, The Second Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| |
Collapse
|
22
|
Zeleniak AE, Huang W, Fishel ML, Hill R. PTEN-Dependent Stabilization of MTSS1 Inhibits Metastatic Phenotype in Pancreatic Ductal Adenocarcinoma. Neoplasia 2017; 20:12-24. [PMID: 29175021 PMCID: PMC5714254 DOI: 10.1016/j.neo.2017.10.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 10/10/2017] [Accepted: 10/23/2017] [Indexed: 12/19/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) presents at metastatic stage in over 50% of patients. With a survival rate of just 2.7% for patients presenting with distant disease, it is imperative to uncover novel mechanisms capable of suppressing metastasis in PDAC. Previously, we reported that the loss of metastasis suppressor protein 1 (MTSS1) in PDAC cells results in significant increase in cellular migration and invasion. Conversely, we also found that overexpressing MTSS1 in metastatic PDAC cell lines corresponds with not only decreased metastatic phenotype, but also greater overall survival. While it is known that MTSS1 is downregulated in late-stage PDAC, the mechanism behind that loss has not yet been elucidated. Here, we build off our previous findings to present a novel regulatory mechanism for the stabilization of MTSS1 via the tumor suppressor protein phosphatase and tensin homolog (PTEN). We show that PTEN loss in PDAC cells results in a decrease in MTSS1 expression and increased metastatic potential. Additionally, we demonstrate that PTEN forms a complex with MTSS1 in order to stabilize and protect it from proteasomal degradation. Finally, we show that the inflammatory tumor microenvironment, which makes up over 90% of PDAC tumor bulk, is capable of downregulating PTEN expression through secretion of miRNA-23b, potentially uncovering a novel extrinsic mechanism of MTSS1 regulation. Collectively, these data offer new insight into the role and regulation of MTSS1in suppressing tumor cell invasion and migration and help shed light as to what molecular mechanisms could be leading to early cell dissemination in PDAC.
Collapse
Affiliation(s)
- Ann E Zeleniak
- Harper Cancer Research Institute, University of Notre Dame, South Bend, IN 46556, USA; Integrated Biomedical Sciences Program, University of Notre Dame, South Bend, IN 46556, USA
| | - Wei Huang
- Harper Cancer Research Institute, University of Notre Dame, South Bend, IN 46556, USA; Department of Biological Sciences, University of Notre Dame, South Bend, IN 46556, USA
| | - Melissa L Fishel
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Pediatrics, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Pancreatic Cancer Signature Center, Indianapolis, IN 46202, USA
| | - Reginald Hill
- Harper Cancer Research Institute, University of Notre Dame, South Bend, IN 46556, USA; Department of Biological Sciences, University of Notre Dame, South Bend, IN 46556, USA.
| |
Collapse
|
23
|
Zeleniak AE, Huang W, Brinkman MK, Fishel ML, Hill R. Loss of MTSS1 results in increased metastatic potential in pancreatic cancer. Oncotarget 2017; 8:16473-16487. [PMID: 28146435 PMCID: PMC5369978 DOI: 10.18632/oncotarget.14869] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Accepted: 01/19/2017] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has a 5-year survival rate of 7%. This dismal prognosis is largely due to the inability to diagnose the disease before metastasis occurs. Tumor cell dissemination occurs early in PDAC. While it is known that inflammation facilitates this process, the underlying mechanisms responsible for this progression have not been fully characterized. Here, we functionally test the role of metastasis suppressor 1 (MTSS1) in PDAC. Despite evidence showing that MTSS1 could be important for regulating metastasis in many different cancers, its function in PDAC has not been studied. Here, we show that loss of MTSS1 leads to increased invasion and migration in PDAC cell lines. Moreover, PDAC cells treated with cancer-associated fibroblast-conditioned media also have increased metastatic potential, which is augmented by loss of MTSS1. Finally, overexpression of MTSS1 in PDAC cell lines leads to a loss of migratory potential in vitro and an increase in overall survival in vivo. Collectively, our data provide insight into an important role for MTSS1 in suppressing tumor cell invasion and migration driven by the tumor microenvironment and suggest that therapeutic strategies aimed at increasing MTSS1 levels may effectively slow the development of metastatic lesions, increasing survival of patients with PDAC.
Collapse
Affiliation(s)
- Ann E Zeleniak
- Integrated Biomedical Sciences Program, University of Notre Dame, South Bend, Indiana, USA.,Harper Cancer Research Institute, University of Notre Dame, South Bend, Indiana, USA
| | - Wei Huang
- Harper Cancer Research Institute, University of Notre Dame, South Bend, Indiana, USA.,Department of Biological Sciences, University of Notre Dame, South Bend, Indiana, USA
| | - Mary K Brinkman
- Harper Cancer Research Institute, University of Notre Dame, South Bend, Indiana, USA.,Department of Biological Sciences, University of Notre Dame, South Bend, Indiana, USA
| | - Melissa L Fishel
- Indiana University School of Medicine, Department of Pharmacology and Toxicology, Indianapolis, Indiana, USA.,Indiana University School of Medicine, Department of Pediatrics, Wells Center for Pediatric Research, Indianapolis, Indiana, USA.,Pancreatic Cancer Signature Center, Indianapolis, Indiana, USA
| | - Reginald Hill
- Harper Cancer Research Institute, University of Notre Dame, South Bend, Indiana, USA.,Department of Biological Sciences, University of Notre Dame, South Bend, Indiana, USA
| |
Collapse
|
24
|
Tatarskiy VV, Simonov YP, Shcherbinin DS, Brechalov AV, Georgieva SG, Soshnikova NV. Stability of the PHF10 subunit of PBAF signature module is regulated by phosphorylation: role of β-TrCP. Sci Rep 2017; 7:5645. [PMID: 28717195 PMCID: PMC5514133 DOI: 10.1038/s41598-017-05944-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 06/06/2017] [Indexed: 11/13/2022] Open
Abstract
The PBAF chromatin-remodeling complexes are multi-protein machines, regulating expression of genes involved in proliferation and differentiation. PHF10 is a subunit of the PBAF essential for its association with chromatin. Mammalian PHF10 is expressed as four ubiquitous isoforms, which are alternatively incorporated in the complex and differ by their influence on transcription of target genes. PHF10 have different domain structure and two of them (PHF10-S isoforms) lack C-terminal PHD domains, which enables their phosphorylation by CK-1. Here we have found that PBAF subunits have low turnover rate, except for PHF10 which has much lower half-life, and is degraded by β-TrCP. The β-TrCP knockdown stabilizes PBAF core subunits - BRG1 and BAF155 and specific subunits - PHF10, BAF200, BAF180 and BRD7. PHF10 isoforms contain two non-canonical β-TrCP degrons and are degraded by β-TrCP in a phospho-dependent manner. But phosphorylation of PHF10-S degrons by CK-1, contrary to previously described degrons, prevents their degradation. Targeted molecular docking demonstrated that phosphorylated forms of PHF10 bind to β-TrCP with much lower affinity than non-phosphorylated ones, contrary to previously described degrons. This unorthodox mechanism proposes that phosphorylation of β-TrCP degrons by CK-1 could not only degrade a set of proteins, but also stabilize a different set of targets.
Collapse
Affiliation(s)
- Victor V Tatarskiy
- Laboratory of Tumor Cell Death, N.N. Blokhin Russian Cancer Research Center, Kashirskoye Shosse 24, Moscow, 115478, Russia
| | - Yuriy P Simonov
- Department of Transcription Factors, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov Str. 32, Moscow, 119991, Russia
| | - Dmitrii S Shcherbinin
- Laboratory of Structure Bioinformatics, Institute of Biomedical Chemistry (IBMC), Pogodinskaya street 10 building 8, Moscow, 119121, Russia
| | - Alexander V Brechalov
- Department of Transcription Factors, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov Str. 32, Moscow, 119991, Russia
| | - Sofia G Georgieva
- Department of Eukariotic Transcription Factors, Institute of Gene Biology, Russian Academy of Sciences, Vavilov Str. 34/5, Moscow, 119991, Russia. .,Department of Transcription Factors, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov Str. 32, Moscow, 119991, Russia.
| | - Nataliya V Soshnikova
- Department of Eukariotic Transcription Factors, Institute of Gene Biology, Russian Academy of Sciences, Vavilov Str. 34/5, Moscow, 119991, Russia.
| |
Collapse
|
25
|
Liang J, Wang WF, Xie S, Zhang XL, Qi WF, Zhou XP, Hu JX, Shi Q, Yu RT. β-transducin repeat-containing E3 ubiquitin protein ligase inhibits migration, invasion and proliferation of glioma cells. Oncol Lett 2017; 14:3131-3135. [PMID: 28928851 DOI: 10.3892/ol.2017.6533] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2015] [Accepted: 05/11/2017] [Indexed: 11/06/2022] Open
Abstract
β-transducin repeat-containing E3 ubiquitin protein ligase (β-TrCP) serves as the substrate recognition subunit for the Skp1-Cullin1-F-box protein E3 ubiquitin ligase, which recognizes the double phosphorylated DSG (X)2+nS destruction motif in various substrates that are essential for numerous aspects of tumorigenesis and regulates several important signaling pathways. However, the biological significance of β-TrCP in glioma progression remains largely unknown. A previous study by the authors demonstrated that the levels of β-TrCP protein expression in brain glioma tissues were significantly lower compared with non-tumorous tissues and that higher grades of gliomas exhibited lower levels of β-TrCP expression in comparison with lower glioma grades. In addition, low β-TrCP expression was associated with poor prognosis in patients with glioma. Subsequently, the present study aimed to investigate the effect of β-TrCP on migratory, invasive and proliferative abilities of glioma cells. β-TrCP plasmids were transfected into cultured U251 and U87 glioma cells, and changes in migration, invasion and proliferation were analyzed using wound healing, Transwell and EdU assays. It was identified that the overexpression of β-TrCP inhibited migration, invasion and proliferation in glioma cells. In summary, these results indicate that β-TrCP may serve a protective role against the progression of glioma by suppressing cell migration, invasion and proliferation. The potential mechanism of β-TrCP I glioma cells requires additional investigation.
Collapse
Affiliation(s)
- Jun Liang
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China.,Laboratory of Neurosurgery, Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China
| | - Wei-Feng Wang
- Laboratory of Neurosurgery, Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China.,Department of Neurosurgery, Graduate School, Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China
| | - Shao Xie
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China.,Laboratory of Neurosurgery, Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China
| | - Xian-Li Zhang
- Laboratory of Neurosurgery, Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China.,Department of Neurosurgery, Graduate School, Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China
| | - Wei-Feng Qi
- Laboratory of Neurosurgery, Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China.,Department of Neurosurgery, Graduate School, Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China
| | - Xiu-Ping Zhou
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China.,Laboratory of Neurosurgery, Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China
| | - Jin-Xia Hu
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China.,Laboratory of Neurosurgery, Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China
| | - Qiong Shi
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China.,Laboratory of Neurosurgery, Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China
| | - Ru-Tong Yu
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China.,Laboratory of Neurosurgery, Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China
| |
Collapse
|
26
|
Zhang Y, Wang W, Cai S, Chen Y, Wang Q, Pan Q, Sun F, Wang J. Reciprocal regulation between βTrCP and Smurf1 suppresses proliferative capacity of liver cancer cells. J Cell Physiol 2017; 232:3347-3359. [PMID: 28063214 DOI: 10.1002/jcp.25780] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 01/04/2017] [Accepted: 01/05/2017] [Indexed: 12/30/2022]
Abstract
We previously reported that both the ubiquitin E3 ligases βTrCP (beta-transducin repeat-containing E3 ubiquitin protein ligase) and Smurf1 (SMAD-specific E3 ubiquitin protein ligase 1) play similar antitumorigenic roles in liver cancer cells. However, whether and how they are reciprocally regulated remains elusive. Here, we show that βTrCP interacts with Smurf1 through the 7 × tryptophan (W) aspartic acid (D)(WD) 40 and the region homologous to the E6-AP carboxyl terminus (HECT) domains, which are the E3 ligase domains of βTrCP and Smurf1, respectively. The E3 ligase domains of βTrCP and Smurf1 are also critical for maintaining the protein expressions of Smurf1 and βTrCP. Moreover, a positive correlation between βTrCP and Smurf1 was also revealed by tissue microarray analysis, indicating that this relationship might be important in liver cancer. Further, we found that Smurf1 increases the protein stability of βTrCP, possibly by reducing autoubiquitination of βTrCP, and vice versa. Interestingly, such effects depended on the presence of E3 ligase domains. Importantly, depletion of Smurf1- or βTrCP-enhanced proliferative capacity of liver cancer cells could be partially reversed by overexpression of wild-type βTrCP or Smurf1 but not their E3 ligase-dead mutants. Collectively, a reciprocal post-translational regulation between βTrCP and Smurf1 has been uncovered in this study. Simultaneous enhancement of βTrCP and Smurf1 functions might be helpful in the treatment of liver cancer.
Collapse
Affiliation(s)
- Yue Zhang
- Department of Central Laboratory, Shanghai Tenth People's Hospital of Tongji University, Shanghai, China
| | - Wenhua Wang
- Department of Neurosurgery, Shanghai Tenth People's Hospital of Tongji University, Shanghai, China
| | - Si Cai
- Department of Laboratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yan Chen
- Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Shanghai, China
| | - Qinwan Wang
- Department of Central Laboratory, Shanghai Tenth People's Hospital of Tongji University, Shanghai, China
| | - Qiuhui Pan
- Department of Central Laboratory, Shanghai Tenth People's Hospital of Tongji University, Shanghai, China
| | - Fenyong Sun
- Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Shanghai, China
| | - Jiayi Wang
- Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Shanghai, China.,Advanced Institute of Translational Medicine, Tongji University, Shanghai, China
| |
Collapse
|
27
|
Shimizu K, Fukushima H, Ogura K, Lien EC, Nihira NT, Zhang J, North BJ, Guo A, Nagashima K, Nakagawa T, Hoshikawa S, Watahiki A, Okabe K, Yamada A, Toker A, Asara JM, Fukumoto S, Nakayama KI, Nakayama K, Inuzuka H, Wei W. The SCFβ-TRCP E3 ubiquitin ligase complex targets Lipin1 for ubiquitination and degradation to promote hepatic lipogenesis. Sci Signal 2017; 10:10/460/eaah4117. [PMID: 28049764 DOI: 10.1126/scisignal.aah4117] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The SCFβ-TRCP E3 ubiquitin ligase complex plays pivotal roles in normal cellular physiology and in pathophysiological conditions. Identification of β-transducin repeat-containing protein (β-TRCP) substrates is therefore critical to understand SCFβ-TRCP biology and function. We used a β-TRCP-phosphodegron motif-specific antibody in a β-TRCP substrate screen coupled with tandem mass spectrometry and identified multiple β-TRCP substrates. One of these substrates was Lipin1, an enzyme and suppressor of the family of sterol regulatory element-binding protein (SREBP) transcription factors, which activate genes encoding lipogenic factors. We showed that SCFβ-TRCP specifically interacted with and promoted the polyubiquitination of Lipin1 in a manner that required phosphorylation of Lipin1 by mechanistic target of rapamycin 1 (mTORC1) and casein kinase I (CKI). β-TRCP depletion in HepG2 hepatocellular carcinoma cells resulted in increased Lipin1 protein abundance, suppression of SREBP-dependent gene expression, and attenuation of triglyceride synthesis. Moreover, β-TRCP1 knockout mice showed increased Lipin1 protein abundance and were protected from hepatic steatosis induced by a high-fat diet. Together, these data reveal a critical physiological function of β-TRCP in regulating hepatic lipid metabolic homeostasis in part through modulating Lipin1 stability.
Collapse
Affiliation(s)
- Kouhei Shimizu
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.,Center for Advanced Stem Cell and Regenerative Research, Tohoku University Graduate School of Dentistry, Sendai 980-8575, Japan
| | - Hidefumi Fukushima
- Center for Advanced Stem Cell and Regenerative Research, Tohoku University Graduate School of Dentistry, Sendai 980-8575, Japan
| | - Kohei Ogura
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.,Department of Infectious Diseases, National Center for Global Health and Medicine, Tokyo 162-8655, Japan
| | - Evan C Lien
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Naoe Taira Nihira
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Jinfang Zhang
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Brian J North
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Ailan Guo
- Cell Signaling Technology Inc., Danvers, MA 01923, USA
| | - Katsuyuki Nagashima
- Department of Physiological Science and Molecular Biology, Fukuoka Dental College, Fukuoka 814-0193, Japan
| | - Tadashi Nakagawa
- Division of Cell Proliferation, United Centers for Advanced Research and Translational Medicine, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Seira Hoshikawa
- Center for Advanced Stem Cell and Regenerative Research, Tohoku University Graduate School of Dentistry, Sendai 980-8575, Japan.,Division of Pediatric Dentistry, Department of Oral Health and Development Sciences, Tohoku University Graduate School of Dentistry, Sendai 980-8575, Japan
| | - Asami Watahiki
- Center for Advanced Stem Cell and Regenerative Research, Tohoku University Graduate School of Dentistry, Sendai 980-8575, Japan
| | - Koji Okabe
- Department of Physiological Science and Molecular Biology, Fukuoka Dental College, Fukuoka 814-0193, Japan
| | - Aya Yamada
- Division of Pediatric Dentistry, Department of Oral Health and Development Sciences, Tohoku University Graduate School of Dentistry, Sendai 980-8575, Japan
| | - Alex Toker
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - John M Asara
- Division of Signal Transduction, Beth Israel Deaconess Medical Center and Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Satoshi Fukumoto
- Center for Advanced Stem Cell and Regenerative Research, Tohoku University Graduate School of Dentistry, Sendai 980-8575, Japan.,Division of Pediatric Dentistry, Department of Oral Health and Development Sciences, Tohoku University Graduate School of Dentistry, Sendai 980-8575, Japan
| | - Keiichi I Nakayama
- Division of Cell Regulation Systems, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Keiko Nakayama
- Division of Cell Proliferation, United Centers for Advanced Research and Translational Medicine, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Hiroyuki Inuzuka
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA. .,Center for Advanced Stem Cell and Regenerative Research, Tohoku University Graduate School of Dentistry, Sendai 980-8575, Japan
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
28
|
miR-96 promotes the growth of prostate carcinoma cells by suppressing MTSS1. Tumour Biol 2016; 37:12023-12032. [DOI: 10.1007/s13277-016-5058-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 05/01/2016] [Indexed: 12/14/2022] Open
|
29
|
Zhou L, Li J, Shao QQ, Guo JC, Liang ZY, Zhou WX, Zhang TP, You L, Zhao YP. Expression and Significances of MTSS1 in Pancreatic Cancer. Pathol Oncol Res 2016. [PMID: 26198729 DOI: 10.1007/s12253-015-9963-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Thus far, expression of metastasis suppressor 1 (MTSS1), its clinicopathologic and prognostic significances in pancreatic cancer (PC) remain unknown. Expression of MTSS1 was detected by Western blotting in PC cell lines, and by tissue microarray-based immunohistochemical staining in paired tumor and non-tumor samples from 242 patients with PC. Furthermore, the correlations between MTSS1 expression and clinicopathologic variables as well as overall survival were evaluated. In PC cell lines, MTSS1 was differentially expressed. In addition, MTSS1 expression was significantly lower in tumor than in non-tumor tissues (P < 0.001 in both McNemar and Mann-Whitney U tests). High tumoral expression of MTSS1 was closely associated with absence of lymph node metastasis (P = 0.023). Univariate analysis found that high MTSS1 expression in tumor tissues was a strong predictor of favorable overall survival in the whole cohort (P < 0.001). Besides, its impacts on prognosis were also observed in nine out of fourteen subgroups. Finally, MTSS1 expression was identified as an independent prognostic marker in the whole cohort (P = 0.031) as well as in six subgroups (P < 0.05), as shown by multivariate Cox regression test. Down-regulation of MTSS1 expression is evident in PC, and is associated with lymph node metastasis and poor prognosis.
Collapse
Affiliation(s)
- Li Zhou
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, 100730, China
| | - Jian Li
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, 100730, China
| | - Qian-Qian Shao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, 100730, China
| | - Jun-Chao Guo
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, 100730, China.
| | - Zhi-Yong Liang
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, 100730, China
| | - Wei-Xun Zhou
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, 100730, China
| | - Tai-Ping Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, 100730, China
| | - Lei You
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, 100730, China
| | - Yu-Pei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
30
|
Zhao P, Cao M, Song L, Wu H, Hu K, Chen B, Wang Q, Gu N. Downregulation of MIM protein inhibits the cellular endocytosis process of magnetic nanoparticles in macrophages. RSC Adv 2016. [DOI: 10.1039/c6ra21530k] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
MIM plays a positive role in the RAW 264.7 cellular endocytosis process of iron oxide nanoparticles mainly in clathrin-mediated pathway, which is a meaningful molecular basis for biomedical applications of nanomaterials.
Collapse
Affiliation(s)
- Peng Zhao
- State Key Laboratory of Bioelectronics
- Jiangsu Key Laboratory for Biomaterials and Devices
- School of Biological Science and Medical Engineering
- Southeast University
- Nanjing 210096
| | - Meng Cao
- State Key Laboratory of Bioelectronics
- Jiangsu Key Laboratory for Biomaterials and Devices
- School of Biological Science and Medical Engineering
- Southeast University
- Nanjing 210096
| | - Lina Song
- State Key Laboratory of Bioelectronics
- Jiangsu Key Laboratory for Biomaterials and Devices
- School of Biological Science and Medical Engineering
- Southeast University
- Nanjing 210096
| | - Hao Wu
- State Key Laboratory of Bioelectronics
- Jiangsu Key Laboratory for Biomaterials and Devices
- School of Biological Science and Medical Engineering
- Southeast University
- Nanjing 210096
| | - Ke Hu
- State Key Laboratory of Bioelectronics
- Jiangsu Key Laboratory for Biomaterials and Devices
- School of Biological Science and Medical Engineering
- Southeast University
- Nanjing 210096
| | - Bo Chen
- State Key Laboratory of Bioelectronics
- Jiangsu Key Laboratory for Biomaterials and Devices
- School of Biological Science and Medical Engineering
- Southeast University
- Nanjing 210096
| | - Qiwei Wang
- State Key Laboratory of Bioelectronics
- Jiangsu Key Laboratory for Biomaterials and Devices
- School of Biological Science and Medical Engineering
- Southeast University
- Nanjing 210096
| | - Ning Gu
- State Key Laboratory of Bioelectronics
- Jiangsu Key Laboratory for Biomaterials and Devices
- School of Biological Science and Medical Engineering
- Southeast University
- Nanjing 210096
| |
Collapse
|
31
|
Schemionek M, Herrmann O, Reher MM, Chatain N, Schubert C, Costa IG, Hänzelmann S, Gusmao EG, Kintsler S, Braunschweig T, Hamilton A, Helgason GV, Copland M, Schwab A, Müller-Tidow C, Li S, Holyoake TL, Brümmendorf TH, Koschmieder S. Mtss1 is a critical epigenetically regulated tumor suppressor in CML. Leukemia 2015; 30:823-32. [DOI: 10.1038/leu.2015.329] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 10/23/2015] [Accepted: 11/16/2015] [Indexed: 12/22/2022]
|
32
|
Shi W, Hasimu G, Wang Y, Li N, Chen M, Zhang H. MTSS1 is an independent prognostic biomarker for survival in intrahepatic cholangiocarcinoma patients. Am J Transl Res 2015; 7:1974-1983. [PMID: 26692940 PMCID: PMC4656773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 08/24/2015] [Indexed: 06/05/2023]
Abstract
MTSS1 is a possible metastasis suppressor which has been proved to play a key role in metastasis of various tumors, yet its role in intrahepatic cholangiocarcinoma (ICC) remains unclear. In present study, we reported detection of MTSS1 expression in ICC and explored its clinical significances. Tissue microarrays containing 93 cases with ICC were constructed and immunohistochemistry was performed to detect MTSS1 expression on these arrays. PcDNA3.1-MTSS1 was transfected into QBC939 cell lines and cell function was measured by transwell assay. Data showed that MTSS1 expression was barely detectable in 56 cases (60.0%) of the 93 primary tumors and that lacking MTSS1 expression was significantly associated with tumor size, nodal metastases and advanced disease stage. In addition, survival analysis demonstrated that lacking MTSS1 expression also correlated significantly with tumor recurrence and poor outcome of patients with ICC. Meanwhile, enhanced expression of MTSS1 leaded to inhibition of the migration of QBC939 cell lines in vitro. These findings together support that MTSS1 may serve as a useful biomarker in predicting tumor recurrence and prognosis of ICC.
Collapse
Affiliation(s)
- Wei Shi
- Department of General Surgery, Huashan Hospital, Fudan UniversityShanghai 200040, China
| | - Gulimire Hasimu
- Department of General Surgery, Huashan Hospital, Fudan UniversityShanghai 200040, China
| | - Yan Wang
- Department of General Surgery, Huashan Hospital, Fudan UniversityShanghai 200040, China
| | - Ning Li
- Department of Infectious Diseases, Huashan Hospital, Fudan UniversityShanghai 200040, China
| | - Mingquan Chen
- Department of Infectious Diseases, Huashan Hospital, Fudan UniversityShanghai 200040, China
| | - Hao Zhang
- Department of General Surgery, Huashan Hospital, Fudan UniversityShanghai 200040, China
| |
Collapse
|
33
|
Guo Y, Ren MS, Shang C, Zhu L, Zhong M. MTSS1 gene regulated by miR-96 inhibits cell proliferation and metastasis in tongue squamous cellular carcinoma Tca8113 cell line. Int J Clin Exp Med 2015; 8:15441-15449. [PMID: 26629033 PMCID: PMC4658922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 09/07/2015] [Indexed: 06/05/2023]
Abstract
BACKGROUND Metastasis suppressor-1 (MTSS1) is a novel potential metastasis suppressor gene in several types of human cancers. However, the exact function and regulatory mechanism of MTSS1 in Tongue squamous cellular carcinoma (TSCC) have not been elucidated. MATERIAL/METHODS We first confirmed the MTSS1 gene expression by using quantitative real time-PCR (qRT-PCR) and immunohistochemical staining. Then we detected the effect of MTSS1 gene on Tca8113 cells proliferation and invasion ability by using MTT, wound healing and invasion assay. Finally by using bioinformatics analysis, luciferase reporter assay and a serial method, we analyzed the targeting of miR-96 on MTSS1 and the ability of miR-96 on MTSS1 gene mediated biological alterations in Tca8113 cells. RESULTS Our findings showed that the expression of MTSS1 was down-regulated in both TSCC tissues and Tca8113 cells. Forced expression of MTSS1 led to inhibited cell proliferation ability, retarded wound closing and reduced trans-membrane cell numbers. MiR-96 is confirmed to be a direct target of MTSS1 gene and could regulate MTSS1 mediated Tca8113 cells proliferation and metastasis. But miR-96 could not completely restore the invasion ability of Tca8113 cells. CONCLUSIONS MiR-96 targeting and promoting MTSS1 repression may precipitate in the TSCC tumorigenesis through bypassing cell proliferation and metastasis control.
Collapse
Affiliation(s)
- Yan Guo
- Key Laboratory of Liaoning Province Oral Disease, School of Stomatology, China Medical UniversityShenyang 110002, P. R. China
| | - Mei-Si Ren
- Department of Pathology, School of Stomatology, China Medical UniversityShenyang 110002, P. R. China
| | - Chao Shang
- Department of Neurobiology, China Medical UniversityShenyang 110001, P. R. China
| | - Li Zhu
- Key Laboratory of Liaoning Province Oral Disease, School of Stomatology, China Medical UniversityShenyang 110002, P. R. China
| | - Ming Zhong
- Key Laboratory of Liaoning Province Oral Disease, School of Stomatology, China Medical UniversityShenyang 110002, P. R. China
- Department of Pathology, School of Stomatology, China Medical UniversityShenyang 110002, P. R. China
| |
Collapse
|
34
|
LIU RONG, MARTIN TRACEYA, JORDAN NICOLAJ, RUGE FIONA, YE LIN, JIANG WENG. Metastasis suppressor 1 expression in human ovarian cancer: The impact on cellular migration and metastasis. Int J Oncol 2015; 47:1429-39. [DOI: 10.3892/ijo.2015.3121] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 05/26/2015] [Indexed: 11/05/2022] Open
|
35
|
Liang J, Wang WF, Xie S, Zhang XL, Qi WF, Zhou XP, Hu JX, Shi Q, Yu RT. Expression of β-transducin repeat-containing E3 ubiquitin protein ligase in human glioma and its correlation with prognosis. Oncol Lett 2015; 9:2651-2656. [PMID: 26137122 DOI: 10.3892/ol.2015.3113] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 03/09/2015] [Indexed: 11/05/2022] Open
Abstract
β-transducin repeat-containing E3 ubiquitin protein ligase (β-TrCP) targets a number of substrates essential for specific aspects of tumorigenesis. In addition, β-TrCP regulates various important signaling pathways. As β-TrCP is involved in regulating the ubiquitination and degradation of multiple oncogenes and tumor suppressors, the function of β-TrCP varies between cancer types. At present, the association between β-TrCP expression and clinicopathological factors in glioma is unknown. Therefore, the current study used western blotting and immunohistochemistry to investigate the expression of β-TrCP protein in glioma tissue specimens. It was identified that β-TrCP protein expression levels were significantly lower in glioma compared with non-tumorous human brain tissues. Furthermore, the higher the grade of glioma, the lower the level of β-TrCP expression. Kaplan-Meier analysis demonstrated that patients with low β-TrCP expression experienced significantly worse overall survival compared with patients with high β-TrCP expression. The results indicate that downregulation of β-TrCP may be associated with poor survival in patients with glioma. Together, the current data indicates that β-TrCP may be applied as a useful indicator of glioma prognosis and may serve as an anticancer therapeutic target for glioma, however further investigation is required.
Collapse
Affiliation(s)
- Jun Liang
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China ; Laboratory of Neurosurgery, Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China
| | - Wei-Feng Wang
- Laboratory of Neurosurgery, Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China ; Graduate School, Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China
| | - Shao Xie
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China ; Laboratory of Neurosurgery, Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China
| | - Xian-Li Zhang
- Laboratory of Neurosurgery, Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China ; Graduate School, Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China
| | - Wei-Feng Qi
- Laboratory of Neurosurgery, Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China ; Graduate School, Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China
| | - Xiu-Ping Zhou
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China ; Laboratory of Neurosurgery, Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China
| | - Jin-Xia Hu
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China ; Laboratory of Neurosurgery, Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China
| | - Qiong Shi
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China ; Laboratory of Neurosurgery, Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China
| | - Ru-Tong Yu
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China ; Laboratory of Neurosurgery, Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China
| |
Collapse
|
36
|
Kedmi M, Ben-Chetrit N, Körner C, Mancini M, Ben-Moshe NB, Lauriola M, Lavi S, Biagioni F, Carvalho S, Cohen-Dvashi H, Schmitt F, Wiemann S, Blandino G, Yarden Y. EGF induces microRNAs that target suppressors of cell migration: miR-15b targets MTSS1 in breast cancer. Sci Signal 2015; 8:ra29. [PMID: 25783158 DOI: 10.1126/scisignal.2005866] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Growth factors promote tumor growth and metastasis. We found that epidermal growth factor (EGF) induced a set of 22 microRNAs (miRNAs) before promoting the migration of mammary cells. These miRNAs were more abundant in human breast tumors relative to the surrounding tissue, and their abundance varied among breast cancer subtypes. One of these miRNAs, miR-15b, targeted the 3' untranslated region of MTSS1 (metastasis suppressor protein 1). Although xenografts in which MTSS1 was knocked down grew more slowly in mice initially, longer-term growth was unaffected. Knocking down MTSS1 increased migration and Matrigel invasion of nontransformed mammary epithelial cells. Overexpressing MTSS1 in an invasive cell line decreased cell migration and invasiveness, decreased the formation of invadopodia and actin stress fibers, and increased the formation of cellular junctions. In tissues from breast cancer patients with the aggressive basal subtype, an inverse correlation occurred with the high expression of miRNA-15b and the low expression of MTSS1. Furthermore, low abundance of MTSS1 correlated with poor patient prognosis. Thus, growth factor-inducible miRNAs mediate mechanisms underlying the progression of cancer.
Collapse
Affiliation(s)
- Merav Kedmi
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Nir Ben-Chetrit
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Cindy Körner
- Division of Molecular Genome Analysis, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
| | - Maicol Mancini
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Noa Bossel Ben-Moshe
- Department of Physics of Complex Systems, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Mattia Lauriola
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Sara Lavi
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Francesca Biagioni
- Translational Oncogenomics Unit, Italian National Cancer Institute "Regina Elena," Rome 00144, Italy
| | - Silvia Carvalho
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Hadas Cohen-Dvashi
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Fernando Schmitt
- Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto and Department of Pathology, University Health Network, Toronto, Ontario M5C 2C4, Canada. IPATIMUP, University of Porto, Porto 4200-465, Portugal
| | - Stefan Wiemann
- Division of Molecular Genome Analysis, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
| | - Giovanni Blandino
- Translational Oncogenomics Unit, Italian National Cancer Institute "Regina Elena," Rome 00144, Italy
| | - Yosef Yarden
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel.
| |
Collapse
|
37
|
Roles of ubiquitination and SUMOylation on prostate cancer: mechanisms and clinical implications. Int J Mol Sci 2015; 16:4560-80. [PMID: 25734985 PMCID: PMC4394435 DOI: 10.3390/ijms16034560] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 02/09/2015] [Accepted: 02/12/2015] [Indexed: 12/30/2022] Open
Abstract
The initiation and progression of human prostate cancer are highly associated with aberrant dysregulations of tumor suppressors and proto-oncogenes. Despite that deletions and mutations of tumor suppressors and aberrant elevations of oncogenes at the genetic level are reported to cause cancers, emerging evidence has revealed that cancer progression is largely regulated by posttranslational modifications (PTMs) and epigenetic alterations. PTMs play critical roles in gene regulation, cellular functions, tissue development, diseases, malignant progression and drug resistance. Recent discoveries demonstrate that ubiquitination and SUMOylation are complicated but highly-regulated PTMs, and make essential contributions to diseases and cancers by regulation of key factors and signaling pathways. Ubiquitination and SUMOylation pathways can be differentially modulated under various stimuli or stresses in order to produce the sustained oncogenic potentials. In this review, we discuss some new insights about molecular mechanisms on ubiquitination and SUMOylation, their associations with diseases, oncogenic impact on prostate cancer (PCa) and clinical implications for PCa treatment.
Collapse
|
38
|
Yi YW, Kang HJ, Bae EJ, Oh S, Seong YS, Bae I. β-TrCP1 degradation is a novel action mechanism of PI3K/mTOR inhibitors in triple-negative breast cancer cells. Exp Mol Med 2015; 47:e143. [PMID: 25721419 PMCID: PMC4346488 DOI: 10.1038/emm.2014.127] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 12/04/2014] [Accepted: 12/10/2014] [Indexed: 12/20/2022] Open
Abstract
An F-box protein, β-TrCP recognizes substrate proteins and destabilizes them through ubiquitin-dependent proteolysis. It regulates the stability of diverse proteins and functions as either a tumor suppressor or an oncogene. Although the regulation by β-TrCP has been widely studied, the regulation of β-TrCP itself is not well understood yet. In this study, we found that the level of β-TrCP1 is downregulated by various protein kinase inhibitors in triple-negative breast cancer (TNBC) cells. A PI3K/mTOR inhibitor PI-103 reduced the level of β-TrCP1 in a wide range of TNBC cells in a proteasome-dependent manner. Concomitantly, the levels of c-Myc and cyclin E were also downregulated by PI-103. PI-103 reduced the phosphorylation of β-TrCP1 prior to its degradation. In addition, knockdown of β-TrCP1 inhibited the proliferation of TNBC cells. We further identified that pharmacological inhibition of mTORC2 was sufficient to reduce the β-TrCP1 and c-Myc levels. These results suggest that mTORC2 regulates the stability of β-TrCP1 in TNBC cells and targeting β-TrCP1 is a potential approach to treat human TNBC.
Collapse
Affiliation(s)
- Yong Weon Yi
- 1] Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA [2] Department of Nanobiomedical Science & BK21 PLUS Research Center for Regenerative Medicine, Dankook University, Cheonan, Korea
| | - Hyo Jin Kang
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Edward Jeong Bae
- 1] Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA [2] Department of Nursing and Health Studies, Georgetown University, Washington, DC, USA
| | - Seunghoon Oh
- Department of Physiology, College of Medicine, Dankook University, Cheonan, Korea
| | - Yeon-Sun Seong
- 1] Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA [2] Department of Nanobiomedical Science & BK21 PLUS Research Center for Regenerative Medicine, Dankook University, Cheonan, Korea
| | - Insoo Bae
- 1] Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA [2] Department of Nanobiomedical Science & BK21 PLUS Research Center for Regenerative Medicine, Dankook University, Cheonan, Korea
| |
Collapse
|
39
|
Liu J, Wan L, Liu P, Inuzuka H, Liu J, Wang Z, Wei W. SCF(β-TRCP)-mediated degradation of NEDD4 inhibits tumorigenesis through modulating the PTEN/Akt signaling pathway. Oncotarget 2015; 5:1026-37. [PMID: 24657926 PMCID: PMC4011580 DOI: 10.18632/oncotarget.1675] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The HECT domain-containing ubiquitin E3 ligase NEDD4 is widely expressed in mammalian tissues and plays a crucial role in governing a wide spectrum of cellular processes including cell growth, tissue development and homeostasis. Recent reports have indicated that NEDD4 might facilitate tumorigenesis through targeted degradation of multiple tumor suppressor proteins including PTEN. However, the molecular mechanism by which NEDD4 stability is regulated has not been fully elucidated. Here we report that SCF(β-TRCP) governs NEDD4 protein stability by targeting it for ubiquitination and subsequent degradation in a Casein Kinase-I (CKI) phosphorylation-dependent manner. Specifically, depletion of β-TRCP, or inactivation of CKI, stabilized NEDD4, leading to down-regulation of its ubiquitin target PTEN and subsequent activation of the mTOR/Akt oncogenic pathway. Furthermore, we found that CKIδ-mediated phosphorylation of Ser347 and Ser348 on NEDD4 promoted its interaction with SCF(β-TRCP) for subsequent ubiquitination and degradation. As a result, compared to ectopic expression of wild-type NEDD4, introducing a non-degradable NEDD4 (S347A/S348A-NEDD4) promoted cancer cell growth and migration. Hence, our findings revealed the CKI/SCF(β-TRCP) signaling axis as the upstream negative regulator of NEDD4, and further suggested that enhancing NEDD4 degradation, presumably with CKI or SCF(β-TRCP) agonists, could be a promising strategy for treating human cancers.
Collapse
Affiliation(s)
- Jia Liu
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Frontier Institute of Life Science, FIST, Xi'an Jiaotong University, Xi'an, China
| | | | | | | | | | | | | |
Collapse
|
40
|
Downregulation of MTSS1 expression is an independent prognosticator in squamous cell carcinoma of the lung. Br J Cancer 2015; 112:866-73. [PMID: 25625275 PMCID: PMC4453956 DOI: 10.1038/bjc.2015.2] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 12/22/2014] [Indexed: 12/29/2022] Open
Abstract
Background: The metastasis suppressor 1 (MTSS1) is a newly discovered protein putatively involved in tumour progression and metastasis. Material and Methods: Immunohistochemical expression of MTSS1 was analysed in 264 non-small-cell lung carcinomas (NSCLCs). Results: The metastasis suppressor 1 was significantly overexpressed in NSCLC compared with normal lung (P=0.01). Within NSCLC, MTSS1 expression was inversely correlated with pT-stage (P=0.019) and histological grading (P<0.001). NSCLC with MTSS1 downregulation (<20%) showed a significantly worse outcome (P=0.007). This proved to be an independent prognostic factor in squamous cell carcinomas (SCCs; P=0.041), especially in early cancer stages (P=0.006). Conclusion: The metastasis suppressor 1 downregulation could thus serve as a stratifying marker for adjuvant therapy in early-stage SCC of the lung.
Collapse
|
41
|
Abstract
Deletion of ovarian carcinoma 2/disabled homolog 2 (DOC-2/DAB2) interacting protein (DAB2IP), is a tumor suppressor that serves as a scaffold protein involved in coordinately regulating cell proliferation, survival and apoptotic pathways. DAB2IP is epigenetically down-regulated in a variety of tumors through the action of the histone methyltransferase EZH2. Although DAB2IP is transcriptionally down-regulated in a variety of tumors, it remains unclear if other mechanisms contribute to functional inactivation of DAB2IP. Here we demonstrate that DAB2IP can be functionally down-regulated by two independent mechanisms. First, we identified that Akt1 can phosphorylate DAB2IP on S847, which regulates the interaction between DAB2IP and its effector molecules H-Ras and TRAF2. Second, we demonstrated that DAB2IP can be degraded in part through ubiquitin-proteasome pathway by SCFFbw7. DAB2IP harbors two Fbw7 phosho-degron motifs, which can be regulated by the kinase, CK1δ. Our data hence indicate that in addition to epigenetic down-regulation, two additional pathways can functional inactivate DAB2IP. Given that DAB2IP has previously been identified to possess direct causal role in tumorigenesis and metastasis, our data indicate that a variety of pathways may pass through DAB2IP to govern cancer development, and therefore highlight DAB2IP agonists as potential therapeutic approaches for future anti-cancer drug development.
Collapse
Affiliation(s)
- Xiangping Dai
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | | | | |
Collapse
|
42
|
Liu J, Shaik S, Dai X, Wu Q, Zhou X, Wang Z, Wei W. Targeting the ubiquitin pathway for cancer treatment. Biochim Biophys Acta Rev Cancer 2014; 1855:50-60. [PMID: 25481052 DOI: 10.1016/j.bbcan.2014.11.005] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 11/24/2014] [Accepted: 11/25/2014] [Indexed: 12/15/2022]
Abstract
Proteasome-mediated degradation is a common mechanism by which cells renew their intracellular proteins and maintain protein homeostasis. In this process, the E3 ubiquitin ligases are responsible for targeting specific substrates (proteins) for ubiquitin-mediated degradation. However, in cancer cells, the stability and the balance between oncoproteins and tumor suppressor proteins are disturbed in part due to deregulated proteasome-mediated degradation. This ultimately leads to either stabilization of oncoprotein(s) or increased degradation of tumor suppressor(s), contributing to tumorigenesis and cancer progression. Therefore, E3 ubiquitin ligases including the SCF types of ubiquitin ligases have recently evolved as promising therapeutic targets for the development of novel anti-cancer drugs. In this review, we highlighted the critical components along the ubiquitin pathway including E1, E2, various E3 enzymes and DUBs that could serve as potential drug targets and also described the available bioactive compounds that target the ubiquitin pathway to control various cancers.
Collapse
Affiliation(s)
- Jia Liu
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Shavali Shaik
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Xiangpeng Dai
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Qiong Wu
- Department of Medical Oncology, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, China
| | - Xiuxia Zhou
- The Cyrus Tang Hematology Center and Collaborative Innovation Center of Hematology, Jiangsu Institute of Hematology, The First Affiliated Hospital, Soochow University, Suzhou 215123, China
| | - Zhiwei Wang
- The Cyrus Tang Hematology Center and Collaborative Innovation Center of Hematology, Jiangsu Institute of Hematology, The First Affiliated Hospital, Soochow University, Suzhou 215123, China.
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
43
|
Knippschild U, Krüger M, Richter J, Xu P, García-Reyes B, Peifer C, Halekotte J, Bakulev V, Bischof J. The CK1 Family: Contribution to Cellular Stress Response and Its Role in Carcinogenesis. Front Oncol 2014; 4:96. [PMID: 24904820 PMCID: PMC4032983 DOI: 10.3389/fonc.2014.00096] [Citation(s) in RCA: 201] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Accepted: 04/18/2014] [Indexed: 12/19/2022] Open
Abstract
Members of the highly conserved and ubiquitously expressed pleiotropic CK1 family play major regulatory roles in many cellular processes including DNA-processing and repair, proliferation, cytoskeleton dynamics, vesicular trafficking, apoptosis, and cell differentiation. As a consequence of cellular stress conditions, interaction of CK1 with the mitotic spindle is manifold increased pointing to regulatory functions at the mitotic checkpoint. Furthermore, CK1 is able to alter the activity of key proteins in signal transduction and signal integration molecules. In line with this notion, CK1 is tightly connected to the regulation and degradation of β-catenin, p53, and MDM2. Considering the importance of CK1 for accurate cell division and regulation of tumor suppressor functions, it is not surprising that mutations and alterations in the expression and/or activity of CK1 isoforms are often detected in various tumor entities including cancer of the kidney, choriocarcinomas, breast carcinomas, oral cancer, adenocarcinomas of the pancreas, and ovarian cancer. Therefore, scientific effort has enormously increased (i) to understand the regulation of CK1 and its involvement in tumorigenesis- and tumor progression-related signal transduction pathways and (ii) to develop CK1-specific inhibitors for the use in personalized therapy concepts. In this review, we summarize the current knowledge regarding CK1 regulation, function, and interaction with cellular proteins playing central roles in cellular stress-responses and carcinogenesis.
Collapse
Affiliation(s)
- Uwe Knippschild
- Department of General and Visceral Surgery, Surgery Center, Ulm University Hospital , Ulm , Germany
| | - Marc Krüger
- Department of General and Visceral Surgery, Surgery Center, Ulm University Hospital , Ulm , Germany
| | - Julia Richter
- Department of General and Visceral Surgery, Surgery Center, Ulm University Hospital , Ulm , Germany
| | - Pengfei Xu
- Department of General and Visceral Surgery, Surgery Center, Ulm University Hospital , Ulm , Germany
| | - Balbina García-Reyes
- Department of General and Visceral Surgery, Surgery Center, Ulm University Hospital , Ulm , Germany
| | - Christian Peifer
- Institute for Pharmaceutical Chemistry, Christian Albrechts University , Kiel , Germany
| | - Jakob Halekotte
- Institute for Pharmaceutical Chemistry, Christian Albrechts University , Kiel , Germany
| | - Vasiliy Bakulev
- Department of Organic Synthesis, Ural Federal University , Ekaterinburg , Russia
| | - Joachim Bischof
- Department of General and Visceral Surgery, Surgery Center, Ulm University Hospital , Ulm , Germany
| |
Collapse
|