1
|
Li K, Xia Y, He J, Wang J, Li J, Ye M, Jin X. The SUMOylation and ubiquitination crosstalk in cancer. J Cancer Res Clin Oncol 2023; 149:16123-16146. [PMID: 37640846 DOI: 10.1007/s00432-023-05310-z] [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: 07/17/2023] [Accepted: 08/16/2023] [Indexed: 08/31/2023]
Abstract
BACKGROUND The cancer occurrence and progression are largely affected by the post-translational modifications (PTMs) of proteins. Currently, it has been shown that the relationship between ubiquitination and SUMOylation is highly complex and interactive. SUMOylation affects the process of ubiquitination and degradation of substrates. Contrarily, SUMOylation-related proteins are also regulated by the ubiquitination process thus altering their protein levels or activity. Emerging evidence suggests that the abnormal regulation between this crosstalk may lead to tumorigenesis. PURPOSE In this review, we have discussed the study of the relationship between ubiquitination and SUMOylation, as well as the possibility of a corresponding application in tumor therapy. METHODS The relevant literatures from PubMed have been reviewed for this article. CONCLUSION The interaction between ubiquitination and SUMOylation is crucial for the occurrence and development of cancer. A greater understanding of the crosstalk of SUMOylation and ubiquitination may be more conducive to the development of more selective and effective SUMOylation inhibitors, as well as a promotion of synergy with other tumor treatment strategies.
Collapse
Affiliation(s)
- Kailang Li
- Department of Oncology, The First Hospital of Ningbo University, Ningbo, 315020, China
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Yongming Xia
- Department of Oncology, Yuyao People's Hospital of Zhejiang, Yuyao, 315400, Zhejiang, China
| | - Jian He
- Department of Oncology, The First Hospital of Ningbo University, Ningbo, 315020, China
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Jie Wang
- Department of Oncology, The First Hospital of Ningbo University, Ningbo, 315020, China
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Jingyun Li
- Department of Oncology, The First Hospital of Ningbo University, Ningbo, 315020, China
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Meng Ye
- Department of Oncology, The First Hospital of Ningbo University, Ningbo, 315020, China.
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, 315211, China.
| | - Xiaofeng Jin
- Department of Oncology, The First Hospital of Ningbo University, Ningbo, 315020, China.
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, 315211, China.
| |
Collapse
|
2
|
Vidal S, Bouzaher YH, El Motiam A, Seoane R, Rivas C. Overview of the regulation of the class IA PI3K/AKT pathway by SUMO. Semin Cell Dev Biol 2022; 132:51-61. [PMID: 34753687 DOI: 10.1016/j.semcdb.2021.10.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/22/2021] [Accepted: 10/26/2021] [Indexed: 12/14/2022]
Abstract
The phosphatidylinositol-3-kinase (PI3K)/AKT pathway is a major regulator of metabolism, migration, survival, proliferation, and antiviral immunity. Both an overactivation and an inhibition of the PI3K/AKT pathway are related to different pathologies. Activation of this signaling pathway is tightly controlled through a multistep process and its deregulation can be associated with aberrant post-translational modifications including SUMOylation. Here, we review the complex modulation of the PI3K/AKT pathway by SUMOylation and we discuss its putative incvolvement in human disease.
Collapse
Affiliation(s)
- Santiago Vidal
- Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), Universidade de Santiago de Compostela, Instituto de Investigaciones Sanitarias (IDIS), 15706 Santiago de Compostela, Spain
| | - Yanis Hichem Bouzaher
- Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), Universidade de Santiago de Compostela, Instituto de Investigaciones Sanitarias (IDIS), 15706 Santiago de Compostela, Spain
| | - Ahmed El Motiam
- Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), Universidade de Santiago de Compostela, Instituto de Investigaciones Sanitarias (IDIS), 15706 Santiago de Compostela, Spain; Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health Systems, Department of Ophthalmology and Vision Science, and Department of Lab Medicine and Pathobiology, University of Toronto, Toronto, ON M5G 1X5, Canada
| | - Rocío Seoane
- Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), Universidade de Santiago de Compostela, Instituto de Investigaciones Sanitarias (IDIS), 15706 Santiago de Compostela, Spain
| | - Carmen Rivas
- Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), Universidade de Santiago de Compostela, Instituto de Investigaciones Sanitarias (IDIS), 15706 Santiago de Compostela, Spain; Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Cantoblanco, 28049 Madrid, Spain.
| |
Collapse
|
3
|
Nishida T. SUMO-specific protease SENP3 enhances MDM2-mediated ubiquitination of PARIS/ZNF746 in HeLa cells. Biochem Biophys Res Commun 2022; 615:150-156. [PMID: 35623300 DOI: 10.1016/j.bbrc.2022.05.065] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 05/17/2022] [Indexed: 11/19/2022]
Abstract
The transcriptional repressor PARIS, a substrate of the ubiquitin E3 ligase parkin, represses the expression of the transcriptional co-activator, PGC-1α gene, and is involved in several pathological processes, including neurodegenerative disease and cancers. We have previously shown that SUMOylation of PARIS play an important role in its transcriptional repression activity. In addition, RNF4-mediated ubiquitination of SUMO2/3-conjugated PARIS is required for the control of PARIS-mediated transcriptional repression in HeLa cells that lack parkin expression. However, little is known about how PARIS ubiquitination and degradation are regulated in parkin-deficient cells. Here, we report that the deSUMOylase SENP3 interacted with PARIS and enhanced the ubiquitination of PARIS independently of its SUMOylation in HeLa cells. SENP3-enhanced PARIS ubiquitination mainly contributed to its proteasomal degradation, and required the oncogenic E3 ubiquitin ligase MDM2. MDM2 knockdown by small interfering RNA or expression of a dominant-negative MDM2 mutant inhibited the ubiquitination of PARIS. We further found that MDM2 activation via the PI3K/AKT pathway was involved in PARIS ubiquitination. Taken together, these results suggest that PARIS ubiquitination through SENP3-mediated MDM2 activation may control its functions in parkin-deficient cells.
Collapse
Affiliation(s)
- Tamotsu Nishida
- Department of Human Functional Genomics, Advanced Science Research Promotion Center, Mie University, 1577 Kurima-machiya, Tsu, 514-8507, Japan
| |
Collapse
|
4
|
Lara-Ureña N, Jafari V, García-Domínguez M. Cancer-Associated Dysregulation of Sumo Regulators: Proteases and Ligases. Int J Mol Sci 2022; 23:8012. [PMID: 35887358 PMCID: PMC9316396 DOI: 10.3390/ijms23148012] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/14/2022] [Accepted: 07/19/2022] [Indexed: 02/04/2023] Open
Abstract
SUMOylation is a post-translational modification that has emerged in recent decades as a mechanism involved in controlling diverse physiological processes and that is essential in vertebrates. The SUMO pathway is regulated by several enzymes, proteases and ligases being the main actors involved in the control of sumoylation of specific targets. Dysregulation of the expression, localization and function of these enzymes produces physiological changes that can lead to the appearance of different types of cancer, depending on the enzymes and target proteins involved. Among the most studied proteases and ligases, those of the SENP and PIAS families stand out, respectively. While the proteases involved in this pathway have specific SUMO activity, the ligases may have additional functions unrelated to sumoylation, which makes it more difficult to study their SUMO-associated role in cancer process. In this review we update the knowledge and advances in relation to the impact of dysregulation of SUMO proteases and ligases in cancer initiation and progression.
Collapse
Affiliation(s)
| | | | - Mario García-Domínguez
- Andalusian Centre for Molecular Biology and Regenerative Medicine (CABIMER), CSIC-Universidad de Sevilla-Universidad Pablo de Olavide, Av. Américo Vespucio 24, 41092 Seville, Spain; (N.L.-U.); (V.J.)
| |
Collapse
|
5
|
Gomarasca M, Lombardi G, Maroni P. SUMOylation and NEDDylation in Primary and Metastatic Cancers to Bone. Front Cell Dev Biol 2022; 10:889002. [PMID: 35465332 PMCID: PMC9020829 DOI: 10.3389/fcell.2022.889002] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 03/25/2022] [Indexed: 12/22/2022] Open
Abstract
Post-translational modifications comprise series of enzymatically-driven chemical modifications, virtually involving the entire cell proteome, that affect the fate of a target protein and, in turn, cell activity. Different classes of modifications can be established ranging from phosphorylation, glycosylation, ubiquitination, acetylation, methylation, lipidation and their inverse reactions. Among these, SUMOylation and NEDDylation are ubiquitin-like multi-enzymatic processes that determine the bound of SUMOs and NEDD8 labels, respectively, on defined amino acidic residues of a specific protein and regulate protein function. As fate-determinants of several effectors and mediators, SUMOylation and NEDDylation play relevant roles in many aspects of tumor cell biology. Bone represents a preferential site of metastasis for solid tumors (e.g., breast and prostate cancers) and the primary site of primitive tumors (e.g., osteosarcoma, chondrosarcoma). Deregulation of SUMOylation and NEDDylation affects different aspects of neoplastic transformation and evolution such as epithelial-mesenchymal transition, adaptation to hypoxia, expression and action of tumor suppressors and oncogenic mediators, and drug resistance. Thereby, they represent potential therapeutic targets. This narrative review aims at describing the involvement and regulation of SUMOylation and NEDDylation in tumor biology, with a specific focus on primary and secondary bone tumors, and to summarize and highlight their potentiality in diagnostics and therapeutic strategies.
Collapse
Affiliation(s)
- Marta Gomarasca
- Laboratory of Experimental Biochemistry and Molecular Biology, IRCCS Istituto Ortopedico Galeazzi, Milano, Italy
| | - Giovanni Lombardi
- Laboratory of Experimental Biochemistry and Molecular Biology, IRCCS Istituto Ortopedico Galeazzi, Milano, Italy
- Department of Athletics, Strength and Conditioning, Poznań University of Physical Education, Poznań, Polska
- *Correspondence: Giovanni Lombardi,
| | - Paola Maroni
- Laboratory of Experimental Biochemistry and Molecular Biology, IRCCS Istituto Ortopedico Galeazzi, Milano, Italy
| |
Collapse
|
6
|
Wang L, Qian J, Yang Y, Gu C. Novel insights into the impact of the SUMOylation pathway in hematological malignancies (Review). Int J Oncol 2021; 59:73. [PMID: 34368858 PMCID: PMC8360622 DOI: 10.3892/ijo.2021.5253] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 07/26/2021] [Indexed: 12/17/2022] Open
Abstract
The small ubiquitin-like modifier (SUMO) system serves an important role in the regulation of protein stability and function. SUMOylation sustains the homeostatic equilibrium of protein function in normal tissues and numerous types of tumor. Accumulating evidence has revealed that SUMO enzymes participate in carcinogenesis via a series of complex cellular or extracellular processes. The present review outlines the physiological characteristics of the SUMOylation pathway and provides examples of SUMOylation participation in different cancer types, including in hematological malignancies (leukemia, lymphoma and myeloma). It has been indicated that the SUMO pathway may influence chromosomal instability, cell cycle progression, apoptosis and chemical drug resistance. The present review also discussed the possible relationship between SUMOylation and carcinogenic mechanisms, and evaluated their potential as biomarkers and therapeutic targets in the diagnosis and treatment of hematological malignancies. Developing and investigating inhibitors of SUMO conjugation in the future may offer promising potential as novel therapeutic strategies.
Collapse
Affiliation(s)
- Ling Wang
- Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210022, P.R. China
| | - Jinjun Qian
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Ye Yang
- Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210022, P.R. China
| | - Chunyan Gu
- Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210022, P.R. China
| |
Collapse
|
7
|
Meng Y, Li X. Expression and Prognosis Analysis of SUMOylation Regulators in Oral Squamous Cell Carcinoma Based on High-Throughput Sequencing. Front Genet 2021; 12:671392. [PMID: 34267779 PMCID: PMC8277238 DOI: 10.3389/fgene.2021.671392] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 05/26/2021] [Indexed: 11/16/2022] Open
Abstract
Introduction Oral squamous cell carcinoma (OSCC) originates from oral mucosal epithelial cells, accounting for more than 90% of oral cancers. The relationship between the expression and prognostic role of SUMOylation regulators in OSCC is rarely studied. Materials and methods The expression and survival data of OSCC were derived from TCGA and GEO databases. Wilcoxon test was used to determine the differential expression of the SUMOylation regulators. A prognostic model based on SUMOylation regulator-related genes was constructed by Cox regression. Gene set enrichment analysis was applied to predict the potential biological functions that the genes might be involved in. Results RANBP2 and SENP6 had the highest SNV frequency. Eleven genes including PIAS3, RANBP2, USPL1, SENP6, SENP2, SENP5, SAE1, UBA2, PIAS4, UBE2I, and SENP3 were highly expressed in OSCC. The prognostic model based on nine SUMOylation-regulated genes (TRIM37, UFM1, FUBP1, CCNT1, FXR1, HMG20A, RANBP3, SPATA5, and DDX23) had a strong ability to predict the prognosis of OSCC. Conclusion This study might provide targets for prognostic evaluation and targeted therapy of patients with OSCC.
Collapse
Affiliation(s)
- Yutong Meng
- Department of Stomatology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiaozhi Li
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
8
|
Kroonen JS, Vertegaal ACO. Targeting SUMO Signaling to Wrestle Cancer. Trends Cancer 2020; 7:496-510. [PMID: 33353838 DOI: 10.1016/j.trecan.2020.11.009] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 11/20/2020] [Accepted: 11/23/2020] [Indexed: 01/16/2023]
Abstract
The small ubiquitin-like modifier (SUMO) signaling cascade is critical for gene expression, genome integrity, and cell cycle progression. In this review, we discuss the important role SUMO may play in cancer and how to target SUMO signaling. Recently developed small molecule inhibitors enable therapeutic targeting of the SUMOylation pathway. Blocking SUMOylation not only leads to reduced cancer cell proliferation but also to an increased antitumor immune response by stimulating interferon (IFN) signaling, indicating that SUMOylation inhibitors have a dual mode of action that can be employed in the fight against cancer. The search for tumor types that can be treated with SUMOylation inhibitors is ongoing. Employing SUMO conjugation inhibitory drugs in the years to come has potential as a new therapeutic strategy.
Collapse
Affiliation(s)
- Jessie S Kroonen
- Department of Cell and Chemical Biology, Leiden University Medical Center, Albinusdreef 2, 2333, ZA, Leiden, The Netherlands
| | - Alfred C O Vertegaal
- Department of Cell and Chemical Biology, Leiden University Medical Center, Albinusdreef 2, 2333, ZA, Leiden, The Netherlands.
| |
Collapse
|
9
|
Zhao Q, Ma Y, Li Z, Zhang K, Zheng M, Zhang S. The Function of SUMOylation and Its Role in the Development of Cancer Cells under Stress Conditions: A Systematic Review. Stem Cells Int 2020; 2020:8835714. [PMID: 33273928 PMCID: PMC7683158 DOI: 10.1155/2020/8835714] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 10/17/2020] [Accepted: 10/22/2020] [Indexed: 02/06/2023] Open
Abstract
Malignant tumors still pose serious threats to human health due to their high morbidity and mortality. Recurrence and metastasis are the most important factors affecting patient prognosis. Chemotherapeutic drugs and radiation used to treat these tumors mainly interfere with tumor metabolism, destroy DNA integrity, and inhibit protein synthesis. The upregulation of small ubiquitin-like modifier (SUMO) is a prevalent posttranslational modification (PTM) in various cancers and plays a critical role in tumor development. The dysregulation of SUMOylation can protect cancer cells from stresses exerted by external or internal stimuli. SUMOylation is a dynamic process finely regulated by SUMOylation enzymes and proteases to maintain a balance between SUMOylation and deSUMOylation. An increasing number of studies have reported that SUMOylation imbalance may contribute to cancer development, including metastasis, angiogenesis, invasion, and proliferation. High level of SUMOylation is required for cancer cells to survive internal or external stresses. Downregulation of SUMOylation may inhibit the development of cancer, making it an important potential clinical therapeutic target. Some studies have already begun to treat tumors by inhibiting the expression of SUMOylation family members, including SUMO E1 or E2. The tumor cells become more aggressive under internal and external stresses. The prevention of tumor development, metastasis, recurrence, and radiochemotherapy resistance by attenuating SUMOylation requires further exploration. This review focused on SUMOylation in tumor cells to discuss its effects on tumor suppressor proteins and oncoproteins as well as classical tumor pathways to identify new insights for cancer clinical therapy.
Collapse
Affiliation(s)
- Qi Zhao
- Department of Pathology, Tianjin Union Medical Center, Tianjin, China
- Tianjin Medical University, Tianjin, China
| | - Ying Ma
- Department of Spine Center, Tianjin Union Medical Center, Tianjin, China
| | - Zugui Li
- Department of Pathology, Tianjin Union Medical Center, Tianjin, China
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Kexin Zhang
- Department of Pathology, Tianjin Union Medical Center, Tianjin, China
- Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Minying Zheng
- Department of Pathology, Tianjin Union Medical Center, Tianjin, China
| | - Shiwu Zhang
- Department of Pathology, Tianjin Union Medical Center, Tianjin, China
| |
Collapse
|
10
|
Celen AB, Sahin U. Sumoylation on its 25th anniversary: mechanisms, pathology, and emerging concepts. FEBS J 2020; 287:3110-3140. [DOI: 10.1111/febs.15319] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 03/04/2020] [Accepted: 03/30/2020] [Indexed: 12/26/2022]
Affiliation(s)
- Arda B. Celen
- Department of Molecular Biology and Genetics Center for Life Sciences and Technologies Bogazici University Istanbul Turkey
| | - Umut Sahin
- Department of Molecular Biology and Genetics Center for Life Sciences and Technologies Bogazici University Istanbul Turkey
| |
Collapse
|
11
|
Fox BM, Janssen A, Estevez-Ordonez D, Gessler F, Vicario N, Chagoya G, Elsayed G, Sotoudeh H, Stetler W, Friedman GK, Bernstock JD. SUMOylation in Glioblastoma: A Novel Therapeutic Target. Int J Mol Sci 2019; 20:ijms20081853. [PMID: 30991648 PMCID: PMC6514907 DOI: 10.3390/ijms20081853] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Revised: 04/11/2019] [Accepted: 04/11/2019] [Indexed: 12/22/2022] Open
Abstract
Protein SUMOylation is a dynamic post-translational modification which is involved in a diverse set of physiologic processes throughout the cell. Of note, SUMOylation also plays a role in the pathobiology of a myriad of cancers, one of which is glioblastoma (GBM). Accordingly, herein, we review core aspects of SUMOylation as it relates to GBM and in so doing highlight putative methods/modalities capable of therapeutically engaging the pathway for treatment of this deadly neoplasm.
Collapse
Affiliation(s)
- Brandon M Fox
- Department of Neurosurgery, University of Alabama at Birmingham, 1060 Faculty Office Tower, 510 20th Street South, Birmingham, AL 35223, USA.
- Medical Scientist Training Program, University of Alabama at Birmingham, 1825 University Boulevard, SHEL 121, Birmingham, AL 35294, USA.
| | - Andrew Janssen
- Department of Neurosurgery, University of Alabama at Birmingham, 1060 Faculty Office Tower, 510 20th Street South, Birmingham, AL 35223, USA.
| | - Dagoberto Estevez-Ordonez
- Department of Neurosurgery, University of Alabama at Birmingham, 1060 Faculty Office Tower, 510 20th Street South, Birmingham, AL 35223, USA.
| | - Florian Gessler
- Department of Neurosurgery, University Hospital Frankfurt, Goethe-University, Schleusenweg 2-16, 60528 Frankfurt, Germany.
| | - Nunzio Vicario
- Department of Biomedical and Biotechnological Sciences, Physiology Section, University of Catania, Via S. Sofia n. 97, Torre Biologica, 95123 Catania, Italy.
| | - Gustavo Chagoya
- Department of Neurosurgery, University of Alabama at Birmingham, 1060 Faculty Office Tower, 510 20th Street South, Birmingham, AL 35223, USA.
| | - Galal Elsayed
- Department of Neurosurgery, University of Alabama at Birmingham, 1060 Faculty Office Tower, 510 20th Street South, Birmingham, AL 35223, USA.
| | - Houman Sotoudeh
- Division of Neuroradiology, Department of Radiology, University of Alabama at Birmingham, Jefferson Tower N419-619 19th Street South, Birmingham, AL 35223, USA.
| | - William Stetler
- Department of Neurosurgery, University of Alabama at Birmingham, 1060 Faculty Office Tower, 510 20th Street South, Birmingham, AL 35223, USA.
| | - Gregory K Friedman
- Department of Neurosurgery, University of Alabama at Birmingham, 1060 Faculty Office Tower, 510 20th Street South, Birmingham, AL 35223, USA.
- Division of Neuroradiology, Department of Radiology, University of Alabama at Birmingham, Jefferson Tower N419-619 19th Street South, Birmingham, AL 35223, USA.
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, University of Alabama at Birmingham, Lowder 512, 1600 7th Avenue South, Birmingham, AL 35223, USA.
| | - Joshua D Bernstock
- Department of Neurosurgery, University of Alabama at Birmingham, 1060 Faculty Office Tower, 510 20th Street South, Birmingham, AL 35223, USA.
- Medical Scientist Training Program, University of Alabama at Birmingham, 1825 University Boulevard, SHEL 121, Birmingham, AL 35294, USA.
| |
Collapse
|
12
|
Chen Y, Xu H, Shi Q, Gu M, Wan X, Chen Q, Wang Z. Hypoxia-inducible factor 1α (HIF-1α) mediates the epithelial-mesenchymal transition in benign prostatic hyperplasia. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2019; 12:295-304. [PMID: 31933745 PMCID: PMC6944022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Accepted: 09/27/2018] [Indexed: 06/10/2023]
Abstract
BACKGROUND Epithelial-mesenchymal transition (EMT) based cancer cell invasion and metastasis has been thoroughly studied in prostate cancer. It was well known that EMT markers which have been found in benign prostatic hyperplasia (BPH) tissues, but system descriptions have not been described. METHODS First, in order to construct the epithelial cells to mesenchymal cell transformation model, BPH-1 cells were cultured with supernatant of prostate matrix normal prostate stromal WPMY-1 cells, after obtaining the culture medium through a filter. After that, we observed the morphology of cells cultured for a period of time by microscopy, detected cell invasion ability by transwell assay, detected cell proliferation ability by MTT, and detected EMT marker expression by western. Finally, we treated the cells with anti-HIF-1α drugs to study their effects on EMT, and then tested several related proteins simultaneously. RESULTS The results showed that the morphology of BPH-1 cells gradually changed to fusiform after cultured with WSCM. At the same time, E-cadherin and cytokeratin levels were significantly lower than those in normal medium. Simultaneous detection of vimentin (SMA) and Snail was positive compared to normal cultured cells. At the same time, the cells were cultured with WSCM and the invasive ability was up-regulated. After treatment with anti-HIF-1α drug, E-cadherin and CK5/8 protein expression was up-regulated, but vimentin, α-SMA, and Snail expression was down-regulated, and in addition, p-Smad3 protein expression was also down-regulated after anti-HIF-1α drug was added. CONCLUSION The above results indicated that WSCM-1 stromal cell supernatant WSCM can induced BPH-1 cell interstitialization, and at the same time, by inducing EMT, secreting HIF-1α activates Smad3 signaling. Our study shows that inhibition of HIF-1α expression provides a new reference for clinical treatment of BPH.
Collapse
|
13
|
Tecalco-Cruz AC, Ríos-López DG, Vázquez-Victorio G, Rosales-Alvarez RE, Macías-Silva M. Transcriptional cofactors Ski and SnoN are major regulators of the TGF-β/Smad signaling pathway in health and disease. Signal Transduct Target Ther 2018; 3:15. [PMID: 29892481 PMCID: PMC5992185 DOI: 10.1038/s41392-018-0015-8] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Revised: 02/16/2018] [Accepted: 03/15/2018] [Indexed: 12/19/2022] Open
Abstract
The transforming growth factor-β (TGF-β) family plays major pleiotropic roles by regulating many physiological processes in development and tissue homeostasis. The TGF-β signaling pathway outcome relies on the control of the spatial and temporal expression of >500 genes, which depend on the functions of the Smad protein along with those of diverse modulators of this signaling pathway, such as transcriptional factors and cofactors. Ski (Sloan-Kettering Institute) and SnoN (Ski novel) are Smad-interacting proteins that negatively regulate the TGF-β signaling pathway by disrupting the formation of R-Smad/Smad4 complexes, as well as by inhibiting Smad association with the p300/CBP coactivators. The Ski and SnoN transcriptional cofactors recruit diverse corepressors and histone deacetylases to repress gene transcription. The TGF-β/Smad pathway and coregulators Ski and SnoN clearly regulate each other through several positive and negative feedback mechanisms. Thus, these cross-regulatory processes finely modify the TGF-β signaling outcome as they control the magnitude and duration of the TGF-β signals. As a result, any alteration in these regulatory mechanisms may lead to disease development. Therefore, the design of targeted therapies to exert tight control of the levels of negative modulators of the TGF-β pathway, such as Ski and SnoN, is critical to restore cell homeostasis under the specific pathological conditions in which these cofactors are deregulated, such as fibrosis and cancer. Proteins that repress molecular signaling through the transforming growth factor-beta (TGF-β) pathway offer promising targets for treating cancer and fibrosis. Marina Macías-Silva and colleagues from the National Autonomous University of Mexico in Mexico City review the ways in which a pair of proteins, called Ski and SnoN, interact with downstream mediators of TGF-β to inhibit the effects of this master growth factor. Aberrant levels of Ski and SnoN have been linked to diverse range of diseases involving cell proliferation run amok, and therapies that regulate the expression of these proteins could help normalize TGF-β signaling to healthier physiological levels. For decades, drug companies have tried to target the TGF-β pathway, with limited success. Altering the activity of these repressors instead could provide a roundabout way of remedying pathogenic TGF-β activity in fibrosis and oncology.
Collapse
Affiliation(s)
- Angeles C Tecalco-Cruz
- 1Instituto de Investigaciones Biomédicas at Universidad Nacional Autónoma de México, Mexico city, 04510 Mexico
| | - Diana G Ríos-López
- 2Instituto de Fisiología Celular at Universidad Nacional Autónoma de México, Mexico city, 04510 Mexico
| | | | - Reyna E Rosales-Alvarez
- 2Instituto de Fisiología Celular at Universidad Nacional Autónoma de México, Mexico city, 04510 Mexico
| | - Marina Macías-Silva
- 2Instituto de Fisiología Celular at Universidad Nacional Autónoma de México, Mexico city, 04510 Mexico
| |
Collapse
|
14
|
Tomasi ML, Ramani K. SUMOylation and phosphorylation cross-talk in hepatocellular carcinoma. Transl Gastroenterol Hepatol 2018; 3:20. [PMID: 29780898 DOI: 10.21037/tgh.2018.04.04] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 04/11/2018] [Indexed: 01/22/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a primary malignancy of the liver and occurs predominantly in patients with underlying chronic liver disease and cirrhosis. The large spectrum of protein post-translational modification (PTM) includes numerous critical signaling events that occur during neoplastic transformation. PTMs occur to nearly all proteins and increase the functional diversity of proteins. We have reviewed the role of two major PTMs, SUMOylation and phosphorylation, in the altered signaling of key players in HCC. SUMOylation is a PTM that involves addition of a small ubiquitin-like modifiers (SUMO) group to proteins. It is known to regulate protein stability, protein-protein interactions, trafficking and transcriptional activity. The major pathways that are regulated by SUMOylation and may influence HCC are regulation of transcription, cell growth pathways associated with B-cell lymphoma 2 (Bcl-2) and methionine adenosyltransferases (MAT), oxidative stress pathways [nuclear erythroid 2-related factor 2 (Nrf2)], tumor suppressor pathways (p53), hypoxia-inducible signaling [hypoxia-inducible factor-1 (HIF-1)], glucose and lipid metabolism, nuclear factor kappa B (NF-κB) and β-Catenin signaling. Phosphorylation is an extensively studied PTM in HCC. The mitogen-activated protein kinase (MAPK), phosphatidyl inositol/AK-strain transforming (PI3K/AKT), and C-SRC pathways have been extensively studied for deregulation of kinases and alteration in signaling of targets through phosphorylation of their substrates. Cross-talk between phosphorylation and SUMOylation is known to influence transcriptional activity of proteins and protein-protein interactions. In HCC, several SUMOylation-dependent phosphorylation events have been studied such as MAPK activation and c-SRC activity that have been reviewed in this work. The drastic effects of site-specific phosphorylation or SUMOylation on enzyme activity of signaling players and its effect on growth and tumorigenesis suggests that these PTMs are novel targets for therapeutic intervention in HCC.
Collapse
Affiliation(s)
- Maria Lauda Tomasi
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Komal Ramani
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
15
|
Abstract
Post-translational protein modification by small ubiquitin-like modifier (SUMO), termed sumoylation, is an important mechanism in cellular responses to stress and one that appears to be upregulated in many cancers. Here, we examine the role of sumoylation in tumorigenesis as a possibly necessary safeguard that protects the stability and functionality of otherwise easily misregulated gene expression programmes and signalling pathways of cancer cells.
Collapse
Affiliation(s)
- Jacob-Sebastian Seeler
- Nuclear Organization and Oncogenesis Unit, INSERM U993, Institut Pasteur, 28 rue de Dr Roux, 75724 Paris Cedex 15, France
| | - Anne Dejean
- Nuclear Organization and Oncogenesis Unit, INSERM U993, Institut Pasteur, 28 rue de Dr Roux, 75724 Paris Cedex 15, France
| |
Collapse
|
16
|
Broster SA, Kyprianou N. Epithelial–mesenchymal transition in prostatic disease. Future Oncol 2015; 11:3197-206. [DOI: 10.2217/fon.15.253] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
A fully differentiated epithelium of the normal prostate gland allows epithelial cells to de-differentiate into mesenchymal-like derivatives via the process of epithelial–mesenchymal transition (EMT) and redifferentiate via the reverse process, mesenchymal–epithelial transition. This review discusses the phenotypic changes associated with EMT and its programming in the development of the two growth disorders of the aging prostate gland, benign prostatic hyperplasia and prostate adenocarcinoma. Considering the cellular heterogeneity that characterizes both conditions, identifying the transcriptional programming of the phenotypic framework defining EMT and its reverse process mesenchymal–epithelial transition in their pathological landscape will enable novel platforms for biomarker-driven therapeutics and their implementation in benign prostatic hyperplasia and prostate cancer.
Collapse
Affiliation(s)
- Seth A Broster
- Departments of Urology, Molecular Biochemistry, Pathology, Toxicology & Cancer Biology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Natasha Kyprianou
- Departments of Urology, Molecular Biochemistry, Pathology, Toxicology & Cancer Biology, University of Kentucky College of Medicine, Lexington, KY, USA
| |
Collapse
|
17
|
Chicooree N, Unwin RD, Griffiths JR. The application of targeted mass spectrometry-based strategies to the detection and localization of post-translational modifications. MASS SPECTROMETRY REVIEWS 2015; 34:595-626. [PMID: 24737647 DOI: 10.1002/mas.21421] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2013] [Accepted: 12/10/2013] [Indexed: 06/03/2023]
Abstract
This review describes some of the more interesting and imaginative ways in which mass spectrometry has been utilized to study a number of important post-translational modifications over the past two decades; from circa 1990 to 2013. A diverse range of modifications is covered, including citrullination, sulfation, hydroxylation and sumoylation. A summary of the biological role of each modification described, along with some brief mechanistic detail, is also included. Emphasis has been placed on strategies specifically aimed at detecting target modifications, as opposed to more serendipitous modification discovery approaches, which rely upon straightforward product ion scanning methods. The authors have intentionally excluded from this review both phosphorylation and glycosylation since these major modifications have been extensively reviewed elsewhere.
Collapse
Affiliation(s)
- Navin Chicooree
- CRUK Manchester Institute, University of Manchester, Wilmslow Road, Manchester, M20 4BX, UK
- School of Chemistry, University of Manchester, Brunswick Street, Manchester, M13 9SU, UK
| | - Richard D Unwin
- Centre for Advanced Discovery and Experimental Therapeutics (CADET), Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Sciences Centre, Manchester, M13 9WL, UK
- Institute of Human Development, Faculty of Medical and Human Sciences, University of Manchester, Manchester, UK
| | - John R Griffiths
- CRUK Manchester Institute, University of Manchester, Wilmslow Road, Manchester, M20 4BX, UK
| |
Collapse
|
18
|
Stastna M, Van Eyk JE. Posttranslational modifications of lysine and evolving role in heart pathologies-recent developments. Proteomics 2015; 15:1164-80. [PMID: 25430483 DOI: 10.1002/pmic.201400312] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Revised: 09/29/2014] [Accepted: 11/24/2014] [Indexed: 02/06/2023]
Abstract
The alteration in proteome composition induced by environmental changes and various pathologies is accompanied by the modifications of proteins by specific cotranslational and PTMs. The type and site stoichiometry of PTMs can affect protein functions, alter cell signaling, and can have acute and chronic effects. The particular interest is drawn to those amino acid residues that can undergo several different PTMs. We hypothesize that these selected amino acid residues are biologically rare and act within the cell as molecular switches. There are, at least, 12 various lysine modifications currently known, several of them have been shown to be competitive and they influence the ability of a particular lysine to be modified by a different PTM. In this review, we discuss the PTMs that occur on lysine, specifically neddylation and sumoylation, and the proteomic approaches that can be applied for the identification and quantification of these PTMs. Of interest are the emerging roles for these modifications in heart disease and what can be inferred from work in other cell types and organs.
Collapse
Affiliation(s)
- Miroslava Stastna
- Institute of Analytical Chemistry of the Academy of Sciences of the Czech Republic, v. v. i, Brno, Czech Republic
| | | |
Collapse
|
19
|
Bogachek MV, De Andrade JP, Weigel RJ. Regulation of epithelial-mesenchymal transition through SUMOylation of transcription factors. Cancer Res 2014; 75:11-5. [PMID: 25524900 DOI: 10.1158/0008-5472.can-14-2824] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Carcinoma cells can transition from an epithelial-to-mesenchymal differentiation state through a process known as epithelial-mesenchymal transition (EMT). The process of EMT is characterized by alterations in the pattern of gene expression and is associated with a loss of cell polarity, an increase in invasiveness, and an increase in cells expressing cancer stem cell (CSC) markers. The reverse process of mesenchymal-to-epithelial transition (MET) can also occur, though the transitions characterizing EMT and MET can be incomplete. A growing number of transcription factors have been identified that influence the EMT/MET processes. Interestingly, SUMOylation regulates the functional activity of many of the transcription factors governing transitions between epithelial and mesenchymal states. In some cases, the transcription factor is a small ubiquitin-like modifier conjugated directly, thus altering its transcriptional activity or cell trafficking. In other cases, SUMOylation alters transcriptional mechanisms through secondary effects. This review explores the role of SUMOylation in controlling transcriptional mechanisms that regulate EMT/MET in cancer. Developing new drugs that specifically target SUMOylation offers a novel therapeutic approach to block tumor growth and metastasis.
Collapse
Affiliation(s)
| | | | - Ronald J Weigel
- Department of Surgery, University of Iowa, Iowa City, Iowa. Department of Anatomy and Cell Biology, University of Iowa, Iowa City, Iowa. Department of Biochemistry, University of Iowa, Iowa City, Iowa.
| |
Collapse
|
20
|
Qin Y, Bao H, Pan Y, Yin M, Liu Y, Wu S, Li H. SUMOylation alterations are associated with multidrug resistance in hepatocellular carcinoma. Mol Med Rep 2014; 9:877-81. [PMID: 24399357 DOI: 10.3892/mmr.2014.1882] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 12/12/2013] [Indexed: 12/29/2022] Open
Abstract
The development of multidrug resistance (MDR) in hepatocellular carcinoma (HCC) may markedly reduce the efficacy of its chemotherapeutic treatment. However, the mechanism regulating the development of MDR in these tumors remains unknown. Given the emerging role of small ubiquitin‑like modifier (SUMO)ylation in tumorigenesis, the possibility that it may also be involved in MDR development was investigated. The expression of SUMO‑1 was first analyzed using immunohistochemistry in 20 cases of HCC. Nuclear SUMO‑1 immunostaining was observed to be significantly increased in HCC specimens compared with matched adjacent non‑neoplastic controls. To further investigate the potential role of SUMOylation in MDR in HCC, a multidrug‑resistant HCC cell line, HepG2/R, was established by exposing HCC cells to gradually increasing concentrations of 5‑fluorouracil. Western blot analysis revealed that the total levels of SUMO‑1‑conjugated proteins were markedly increased in HepG2/R cells compared with parental HepG2 cells. Furthermore, the expression of ubiquitin‑like modifier activating enzyme 2 and sentrin‑specific protease 1, important enzymes in the SUMOylation cascade, were markedly upregulated in the HepG2/R cell line. These findings support the hypothesis that SUMOylation is important in the development of MDR in HCC.
Collapse
Affiliation(s)
- Yu Qin
- Department of Diagnostics, College of Basic Medical Science, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Huijing Bao
- School of Laboratory Medicine, Tianjin Medical University, Tianjin 300203, P.R. China
| | - Yi Pan
- Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, P.R. China
| | - Meilin Yin
- School of Laboratory Medicine, Tianjin Medical University, Tianjin 300203, P.R. China
| | - Yunde Liu
- School of Laboratory Medicine, Tianjin Medical University, Tianjin 300203, P.R. China
| | - Shangwei Wu
- School of Laboratory Medicine, Tianjin Medical University, Tianjin 300203, P.R. China
| | - Huikai Li
- Department of Hepatobiliary Surgery, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, P.R. China
| |
Collapse
|
21
|
Nag S, Zhang X, Srivenugopal K, Wang MH, Wang W, Zhang R. Targeting MDM2-p53 interaction for cancer therapy: are we there yet? Curr Med Chem 2014; 21:553-74. [PMID: 24180275 PMCID: PMC6690199 DOI: 10.2174/09298673113206660325] [Citation(s) in RCA: 112] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Revised: 10/02/2013] [Accepted: 10/22/2013] [Indexed: 11/22/2022]
Abstract
Inactivation of the tumor suppressor p53 and/or overexpression of the oncogene MDM2 frequently occur in human cancers, and are associated with poor prognosis, advanced forms of the disease, and chemoresistance. MDM2, the major negative regulator of p53, induces p53 degradation and inactivates its tumor suppressing activity. In turn, p53 regulates MDM2 expression. This MDM2-p53 negative feedback loop has been widely studied and presents an attractive target for cancer therapy, with a few of the inhibitors of this interaction already having advanced into clinical trials. Additionally, there is an increasing interest in understanding MDM2's p53-independent activities in carcinogenesis and cancer progression, which may also have implications for cancer therapy. This review aims to highlight the various roles that the MDM2-p53 interaction plays in cancer, the p53 independent oncogenic activities of MDM2 and the various strategies that may be used to target MDM2 and the MDM2-p53 interaction. We will summarize the major preclinical and clinical evidences of MDM2 inhibitors for human cancer treatment and make suggestions to further improve efficacy and safety of this interesting class of cancer therapeutics.
Collapse
Affiliation(s)
- S. Nag
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - X. Zhang
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - K.S. Srivenugopal
- Department of Biomedical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
- Cancer Biology Center, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - M.-H. Wang
- Department of Biomedical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
- Cancer Biology Center, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - W. Wang
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
- Cancer Biology Center, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - R. Zhang
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
- Cancer Biology Center, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| |
Collapse
|
22
|
Chicooree N, Griffiths JR, Connolly Y, Smith DL. Chemically facilitating the generation of diagnostic ions from SUMO(2/3) remnant isopeptides. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2013; 27:2108-2114. [PMID: 23943332 DOI: 10.1002/rcm.6670] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Revised: 06/25/2013] [Accepted: 06/25/2013] [Indexed: 06/02/2023]
Abstract
RATIONALE Mapping sites of wild-type SUMO modification is a challenging endeavour. Here we postulate that a combination of chemical derivatistation and collision-induced dissociation (CID) could be used to generate SUMO remnant diagnostic ions to aid both detection of these isopeptides and increase the analytical value of the product ion spectra required to characterize the nature and position of modification. METHODS SUMO(2/3)ylated proteins were digested with trypsin to generate isopeptides bearing TGG and QTGG isotags. The resulting digests were then dimethyl labelled followed by liquid chromatography/tandem mass spectrometry (LC/MS/MS) utilising CID in a data-dependent acquisition on a QSTAR XL. Product ion spectra were interrogated for the presence of iso-N-terminal fragment ions in addition to backbone sequence ions. The ability to diagnostically detect these isopeptides was tested by generation of co-XICs of the iso-N-terminal fragments in a semi-complex background. RESULTS Dimethyl labelling facilitated the robust detection of a1', b2' & b3' (TGG isotag) and a1', b2' & b4' (QTGG isotag) ions. The abundance of both N-terminal and iso-N-terminal fragment ions, supported by dimethyl labelling, facilitated the generation of information-rich product ion spectra of these isopeptides to aid confident site assignment. Moreover, the diagnostic nature of the combined XICs of the iso-N-terminal fragments supported detection of the isopeptide signals from a semi-complex background. CONCLUSIONS A combination of dimethyl labelling and CID does indeed lead to the generation of SUMO remnant isopeptide product ion spectra which are more analytically rich. This enables an improvement in characterization of both the isotag and backbone sequences and the site of modification. The diagnostic value of iso-N-terminal fragment ions allows for post-acquisition XIC interrogation to detect putative isopeptides of interest.
Collapse
Affiliation(s)
- Navin Chicooree
- Paterson Institute for Cancer Research, University of Manchester, Wilmslow Road, Manchester, M20 4BX, UK
| | | | | | | |
Collapse
|
23
|
Wang L, Hou Y, Sun Y, Zhao L, Tang X, Hu P, Yang J, Zeng Z, Yang G, Cui X, Liu M. c-Ski activates cancer-associated fibroblasts to regulate breast cancer cell invasion. Mol Oncol 2013; 7:1116-28. [PMID: 24011664 DOI: 10.1016/j.molonc.2013.08.007] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2013] [Revised: 08/09/2013] [Accepted: 08/12/2013] [Indexed: 12/14/2022] Open
Abstract
Aberrant expression of c-Ski oncoprotein in some tumor cells has been shown to be associated with cancer development. However, the role of c-Ski in cancer-associated fibroblasts (CAFs) of tumor microenvironment has not been characterized. In the current study, we found that c-Ski is highly expressed in CAFs derived from breast carcinoma microenvironment and this CAF-associated c-Ski expression is associated with invasion and metastasis of human breast tumors. We showed that c-Ski overexpression in immortalized breast normal fibroblasts (NFs) induces conversion to breast CAFs by repressing p53 and thereby upregulating SDF-1 in NFs. SDF-1 treatment or p53 knockdown in NFs had similar effects on the activation of NFs as c-Ski overexpression. The c-Ski-activated CAFs show increased proliferation, migration, invasion and contraction compared with NFs. Furthermore, c-Ski-activated CAFs facilitated the migration and invasion of MDA-MB-231 breast cancer cells. Our data suggest that c-Ski is an important regulator in the activation of CAFs and may serve as a potential therapeutic target to block breast cancer progression.
Collapse
Affiliation(s)
- Liyang Wang
- Key Laboratory of Laboratory Medical Diagnostics, Chinese Ministry of Education, Chongqing Medical University, Chongqing 400016, China
| | - Yixuan Hou
- Key Laboratory of Laboratory Medical Diagnostics, Chinese Ministry of Education, Chongqing Medical University, Chongqing 400016, China
- Experimental teaching center of Basic Medicine Science, Chongqing Medical University, Chongqing 400016, China
| | - Yan Sun
- Key Laboratory of Laboratory Medical Diagnostics, Chinese Ministry of Education, Chongqing Medical University, Chongqing 400016, China
| | - Liuyang Zhao
- Key Laboratory of Laboratory Medical Diagnostics, Chinese Ministry of Education, Chongqing Medical University, Chongqing 400016, China
| | - Xi Tang
- Key Laboratory of Laboratory Medical Diagnostics, Chinese Ministry of Education, Chongqing Medical University, Chongqing 400016, China
| | - Ping Hu
- Key Laboratory of Laboratory Medical Diagnostics, Chinese Ministry of Education, Chongqing Medical University, Chongqing 400016, China
| | - Jiajia Yang
- Key Laboratory of Laboratory Medical Diagnostics, Chinese Ministry of Education, Chongqing Medical University, Chongqing 400016, China
| | - Zongyue Zeng
- Key Laboratory of Laboratory Medical Diagnostics, Chinese Ministry of Education, Chongqing Medical University, Chongqing 400016, China
| | - Guanglun Yang
- Department of Endocrine and Breast Surgery, the First Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Xiaojiang Cui
- Department of Surgery, Department of Obstetrics and Gynecology, Women's Cancer Program, Samuel Oschin Comprehensive Cancer Institute, Cedars Sinai Medical Center. Los Angeles, CA 91006, USA
| | - Manran Liu
- Key Laboratory of Laboratory Medical Diagnostics, Chinese Ministry of Education, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
24
|
Sun L, Li H, Chen J, Dehennaut V, Zhao Y, Yang Y, Iwasaki Y, Kahn-Perles B, Leprince D, Chen Q, Shen A, Xu Y. A SUMOylation-dependent pathway regulates SIRT1 transcription and lung cancer metastasis. J Natl Cancer Inst 2013; 105:887-98. [PMID: 23704280 DOI: 10.1093/jnci/djt118] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Epithelial-to-mesenchymal transition (EMT) plays a pivotal role in lung cancer metastasis. The class III deacetylase sirtuin 1 (SIRT1) possesses both pro- and anticarcinogenic properties. The role of SIRT1 in lung cancer EMT is largely undefined. METHODS The effect of SIRT1 on migration of lung cancer cells was evaluated by wound healing assay in vitro and metastasis assay in nude mice in vivo. Protein expression in human lung cancers and cultured lung cancer cells was assessed by western blotting and immunohistochemistry. Interaction between protein and DNA was measured by chromatin immunoprecipitation assay. SIRT1 promoter activity was determined by reporter assay. RESULTS SIRT1 activation antagonized migration of lung cancer cells by suppressing EMT in vitro. Activation of SIRT1 by resveratrol also statistically significantly hampered (by 68.33%; P < .001, two-sided test) lung cancer cell metastasis in vivo. Hypoxia repressed SIRT1 transcription through promoting the competition between Sp1 and HIC1 on the SIRT1 proximal promoter in a SUMOylation-dependent manner. Disruption of SUMOylation by targeting either Ubc9 or PIASy restored SIRT1 expression in and favored an epithelial-like phenotype of cancer cells, thereby preventing metastasis. Decreased SIRT1 combined with elevated PIASy expression was implicated in more-invasive types of lung cancers in humans. CONCLUSIONS We have identified a novel pathway that links SIRT1 down-regulation to hypoxia-induced EMT in lung cancer cells and may shed light on the development of novel antitumor therapeutics.
Collapse
Affiliation(s)
- Lina Sun
- State Key Laboratory of Reproductive Medicine, Department of Pathophysiology, Provincial Key Laboratory of Cardiovascular Disease, Nanjing Medical University, Nanjing, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Gautam M, Mathur A, Khan MA, Majumdar SS, Rai U. Transcriptome analysis of spermatogenically regressed, recrudescent and active phase testis of seasonally breeding wall lizards Hemidactylus flaviviridis. PLoS One 2013; 8:e58276. [PMID: 23536792 PMCID: PMC3594293 DOI: 10.1371/journal.pone.0058276] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Accepted: 02/01/2013] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Reptiles are phylogenically important group of organisms as mammals have evolved from them. Wall lizard testis exhibits clearly distinct morphology during various phases of a reproductive cycle making them an interesting model to study regulation of spermatogenesis. Studies on reptile spermatogenesis are negligible hence this study will prove to be an important resource. METHODOLOGY/PRINCIPAL FINDINGS Histological analyses show complete regression of seminiferous tubules during regressed phase with retracted Sertoli cells and spermatognia. In the recrudescent phase, regressed testis regain cellular activity showing presence of normal Sertoli cells and developing germ cells. In the active phase, testis reaches up to its maximum size with enlarged seminiferous tubules and presence of sperm in seminiferous lumen. Total RNA extracted from whole testis of regressed, recrudescent and active phase of wall lizard was hybridized on Mouse Whole Genome 8×60 K format gene chip. Microarray data from regressed phase was deemed as control group. Microarray data were validated by assessing the expression of some selected genes using Quantitative Real-Time PCR. The genes prominently expressed in recrudescent and active phase testis are cytoskeleton organization GO 0005856, cell growth GO 0045927, GTpase regulator activity GO: 0030695, transcription GO: 0006352, apoptosis GO: 0006915 and many other biological processes. The genes showing higher expression in regressed phase belonged to functional categories such as negative regulation of macromolecule metabolic process GO: 0010605, negative regulation of gene expression GO: 0010629 and maintenance of stem cell niche GO: 0045165. CONCLUSION/SIGNIFICANCE This is the first exploratory study profiling transcriptome of three drastically different conditions of any reptilian testis. The genes expressed in the testis during regressed, recrudescent and active phase of reproductive cycle are in concordance with the testis morphology during these phases. This study will pave the way for deeper insight into regulation and evolution of gene regulatory mechanisms in spermatogenesis.
Collapse
Affiliation(s)
- Mukesh Gautam
- Comparative Immuno-Endocrinology Laboratory, Department of Zoology, University of Delhi, Delhi, India
| | - Amitabh Mathur
- Comparative Immuno-Endocrinology Laboratory, Department of Zoology, University of Delhi, Delhi, India
| | - Meraj Alam Khan
- Department of Physiology, All India Institute of Medical Sciences, New Delhi, India
| | - Subeer S. Majumdar
- Cellular Endocrinology Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, India
| | - Umesh Rai
- Comparative Immuno-Endocrinology Laboratory, Department of Zoology, University of Delhi, Delhi, India
- * E-mail:
| |
Collapse
|
26
|
Chicooree N, Griffiths JR, Connolly Y, Tan CT, Malliri A, Eyers CE, Smith DL. A novel approach to the analysis of SUMOylation with the independent use of trypsin and elastase digestion followed by database searching utilising consecutive residue addition to lysine. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2013; 27:127-34. [PMID: 23239325 DOI: 10.1002/rcm.6425] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2012] [Revised: 08/28/2012] [Accepted: 09/28/2012] [Indexed: 06/01/2023]
Abstract
RATIONALE Identification of sites of protein SUMOylation is of great importance due its functional diversity within the cell. To date, most approaches to this problem rely on site-directed mutagenesis and/or highly specialised mass spectrometry approaches. We present a novel alternative approach to the site mapping of SUMOylation using trypsin and elastase digestion, routine mass spectrometry and an unbiased isotag database searching strategy. METHODS SUMOylated protein samples were digested with a number of enzymes and the resulting peptides separated using liquid chromatography. Analysis was carried out on both linear ion trap Orbitrap and quadrupole-time-of-flight (Q-TOF)-based mass spectrometers equipped with electrospray ionisation. The data files were subsequently searched using the Mascot algorithm with multiple variable tag modifications corresponding to SUMO-derived fragments. The utility of this approach was demonstrated with di-SUMO 2, di-SUMO 3, SUMO 1-RanGap(418-587) 1 and an enriched population of SUMOylated proteins. RESULTS Unbiased database searches led to the identification of a number of analytically useful isotags ranging in length from two to four residues. Isopeptide fragments were generated including QTGG (di-SUMO-2/3), TGG (di-SUMO-2/3) and GG (SUMO-1). The method was validated by successfully mapping a number of sites of SUMO modification on SUMO-modified proteins enriched from a cell lysate. CONCLUSIONS This combination of relaxed enzyme specificity, shortened isotag generation and unbiased database searching enabled confident identification of novel analytically useful SUMOylated isopeptides without a requirement for mutagenesis.
Collapse
Affiliation(s)
- Navin Chicooree
- Paterson Institute for Cancer Research, University of Manchester, Wilmslow Road, Manchester, M20 4BX, UK
| | | | | | | | | | | | | |
Collapse
|