1
|
Guay KP, Chou WC, Canniff NP, Paul KB, Hebert DN. N-glycan-dependent protein maturation and quality control in the ER. Nat Rev Mol Cell Biol 2025:10.1038/s41580-025-00855-y. [PMID: 40389697 DOI: 10.1038/s41580-025-00855-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/11/2025] [Indexed: 05/21/2025]
Abstract
The vast majority of proteins that traverse the mammalian secretory pathway become N-glycosylated in the endoplasmic reticulum (ER). The bulky glycan protein modifications, which are conserved in fungi and humans, act as maturation and quality-control tags. In this Review, we discuss findings published in the past decade that have rapidly expanded our understanding of the transfer and processing of N-glycans, as well as their role in protein maturation, quality control and trafficking in the ER, facilitated by structural insights into the addition of N-glycans by the oligosaccharyltransferases A and B (OST-A and OST-B). These findings suggest that N-glycans serve as reporters of the folding status of secretory proteins as they traverse the ER, enabling the lectin chaperones to guide their maturation. We also explore how the emergence of co-translational glycosylation and the expansion of the glycoproteostasis network in metazoans has expanded the role of N-glycans in early protein-maturation events and quality control.
Collapse
Affiliation(s)
- Kevin P Guay
- Program in Molecular and Cellular Biology, University of Massachusetts, Amherst, MA, USA.
- Department of Biochemistry and Molecular Biology, University of Massachusetts, Amherst, MA, USA.
| | - Wen-Chuan Chou
- Program in Molecular and Cellular Biology, University of Massachusetts, Amherst, MA, USA
| | - Nathan P Canniff
- Program in Molecular and Cellular Biology, University of Massachusetts, Amherst, MA, USA
| | - Kylie B Paul
- Program in Molecular and Cellular Biology, University of Massachusetts, Amherst, MA, USA
| | - Daniel N Hebert
- Program in Molecular and Cellular Biology, University of Massachusetts, Amherst, MA, USA
- Department of Biochemistry and Molecular Biology, University of Massachusetts, Amherst, MA, USA
| |
Collapse
|
2
|
Zhang M, Li M, Li H, Wan Y, Yang S, Ji S, Zhang H, Liu C, Lu G, Jiang X, Liu H. Dysregulation of N-glycosylation by Rpn1 knockout in spermatocytes induces male infertility via endoplasmic reticulum stress in mice. Int J Biol Sci 2025; 21:2360-2379. [PMID: 40083683 PMCID: PMC11900820 DOI: 10.7150/ijbs.106468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 02/21/2025] [Indexed: 03/16/2025] Open
Abstract
N-glycosylation protein modification plays a crucial regulatory role in numerous biological processes, although their contribution to male reproduction in mammals remains largely undefined. Here, we found that Ribophorin I (RPN1), a subunit of oligosaccharyltransferase complex, is indispensable for spermatogenesis in male germ cells. Germ cell-specific Rpn1 knockout results in significant inhibition of the progression of meiosis, consequently disrupting homologous chromosome pairing, meiotic recombination, and DNA double strand breaks repair during meiosis. N-glycoproteomic profiling revealed that glycosylation levels are reduced in endoplasmic reticulum-associated proteins, while functional analyses showed that Rpn1 deficiency could inhibit endoplasmic reticulum function and trigger endoplasmic reticulum stress during meiosis and increasing apoptosis levels in mice. These findings highlight the essential physiological functions of N-glycosylation modification in male spermatogenesis and expand our understanding of its role in male fertility.
Collapse
Affiliation(s)
- Mingyu Zhang
- Institute of Women, Children and Reproductive Health, Shandong University, 250012, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, 250012, China
- Key Laboratory of Reproductive Endocrinoligy (Shandong University), Ministry of Education, Jinan, Shandong, 250012, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, 250012, China
- Shandong Key Laboratory of Reproductive Research and Birth Defect Prevention, Jinan, Shandong, 250012, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No.2021RU001), Jinan, Shandong, 250012, China
- School of Basic Medical Sciences, Shandong University, Jinan 250012, China
| | - Mengjing Li
- Institute of Women, Children and Reproductive Health, Shandong University, 250012, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, 250012, China
- Key Laboratory of Reproductive Endocrinoligy (Shandong University), Ministry of Education, Jinan, Shandong, 250012, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, 250012, China
- Shandong Key Laboratory of Reproductive Research and Birth Defect Prevention, Jinan, Shandong, 250012, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No.2021RU001), Jinan, Shandong, 250012, China
| | - Hanzhen Li
- Institute of Women, Children and Reproductive Health, Shandong University, 250012, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, 250012, China
- Key Laboratory of Reproductive Endocrinoligy (Shandong University), Ministry of Education, Jinan, Shandong, 250012, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, 250012, China
- Shandong Key Laboratory of Reproductive Research and Birth Defect Prevention, Jinan, Shandong, 250012, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No.2021RU001), Jinan, Shandong, 250012, China
- School of Basic Medical Sciences, Shandong University, Jinan 250012, China
| | - Yanling Wan
- Institute of Women, Children and Reproductive Health, Shandong University, 250012, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, 250012, China
- Key Laboratory of Reproductive Endocrinoligy (Shandong University), Ministry of Education, Jinan, Shandong, 250012, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, 250012, China
- Shandong Key Laboratory of Reproductive Research and Birth Defect Prevention, Jinan, Shandong, 250012, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No.2021RU001), Jinan, Shandong, 250012, China
| | - Shuang Yang
- School of Basic Medical Sciences, Shandong University, Jinan 250012, China
| | - Shuhui Ji
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Haobo Zhang
- Institute of Women, Children and Reproductive Health, Shandong University, 250012, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, 250012, China
- Key Laboratory of Reproductive Endocrinoligy (Shandong University), Ministry of Education, Jinan, Shandong, 250012, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, 250012, China
- Shandong Key Laboratory of Reproductive Research and Birth Defect Prevention, Jinan, Shandong, 250012, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No.2021RU001), Jinan, Shandong, 250012, China
- Center for Reproductive Medicine, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Chao Liu
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Gang Lu
- CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, the Chinese University of Hong Kong, Hong Kong, China
| | - Xiaohua Jiang
- Center for Reproduction and Genetics, Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Hongbin Liu
- Institute of Women, Children and Reproductive Health, Shandong University, 250012, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, 250012, China
- Key Laboratory of Reproductive Endocrinoligy (Shandong University), Ministry of Education, Jinan, Shandong, 250012, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, 250012, China
- Shandong Key Laboratory of Reproductive Research and Birth Defect Prevention, Jinan, Shandong, 250012, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No.2021RU001), Jinan, Shandong, 250012, China
- School of Basic Medical Sciences, Shandong University, Jinan 250012, China
- CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, the Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
3
|
Kodali S, Proietti L, Valcarcel G, López-Rubio AV, Pessina P, Eder T, Shi J, Jen A, Lupión-Garcia N, Starner AC, Bartels MD, Cui Y, Sands CM, Planas-Riverola A, Martínez A, Velasco-Hernandez T, Tomás-Daza L, Alber B, Manhart G, Mayer IM, Kollmann K, Fatica A, Menendez P, Shishkova E, Rau RE, Javierre BM, Coon J, Chen Q, Van Nostrand EL, Sardina JL, Grebien F, Di Stefano B. RNA sequestration in P-bodies sustains myeloid leukaemia. Nat Cell Biol 2024; 26:1745-1758. [PMID: 39169219 PMCID: PMC12042958 DOI: 10.1038/s41556-024-01489-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 07/18/2024] [Indexed: 08/23/2024]
Abstract
Post-transcriptional mechanisms are fundamental safeguards of progenitor cell identity and are often dysregulated in cancer. Here, we identified regulators of P-bodies as crucial vulnerabilities in acute myeloid leukaemia (AML) through genome-wide CRISPR screens in normal and malignant haematopoietic progenitors. We found that leukaemia cells harbour aberrantly elevated numbers of P-bodies and show that P-body assembly is crucial for initiation and maintenance of AML. Notably, P-body loss had little effect upon homoeostatic haematopoiesis but impacted regenerative haematopoiesis. Molecular characterization of P-bodies purified from human AML cells unveiled their critical role in sequestering messenger RNAs encoding potent tumour suppressors from the translational machinery. P-body dissolution promoted translation of these mRNAs, which in turn rewired gene expression and chromatin architecture in leukaemia cells. Collectively, our findings highlight the contrasting and unique roles of RNA sequestration in P-bodies during tissue homoeostasis and oncogenesis. These insights open potential avenues for understanding myeloid leukaemia and future therapeutic interventions.
Collapse
MESH Headings
- Humans
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- RNA, Messenger/metabolism
- RNA, Messenger/genetics
- Animals
- Hematopoiesis/genetics
- Cell Line, Tumor
- Mice
- Gene Expression Regulation, Leukemic
- Hematopoietic Stem Cells/metabolism
- Hematopoietic Stem Cells/pathology
- Mice, Inbred C57BL
Collapse
Affiliation(s)
- Srikanth Kodali
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Center for Cancer Epigenetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ludovica Proietti
- Institute for Medical Biochemistry, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Gemma Valcarcel
- Josep Carreras Leukaemia Research Institute, Badalona, Spain
| | | | - Patrizia Pessina
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Center for Cancer Epigenetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Thomas Eder
- Institute for Medical Biochemistry, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Junchao Shi
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, USA
| | - Annie Jen
- Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI, USA
| | - Núria Lupión-Garcia
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Center for Cancer Epigenetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anne C Starner
- Verna & Marrs McLean Department of Biochemistry & Molecular Biology and Therapeutic Innovation Center, Baylor College of Medicine, Houston, TX, USA
| | - Mason D Bartels
- Verna & Marrs McLean Department of Biochemistry & Molecular Biology and Therapeutic Innovation Center, Baylor College of Medicine, Houston, TX, USA
| | - Yingzhi Cui
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Center for Cancer Epigenetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Caroline M Sands
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Center for Cancer Epigenetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Alba Martínez
- Josep Carreras Leukaemia Research Institute, Badalona, Spain
| | | | | | - Bernhard Alber
- Institute for Medical Biochemistry, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Gabriele Manhart
- Institute for Medical Biochemistry, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Isabella Maria Mayer
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Karoline Kollmann
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Alessandro Fatica
- Department of Biology and Biotechnology 'Charles Darwin', Sapienza University of Rome, Rome, Italy
| | - Pablo Menendez
- Josep Carreras Leukaemia Research Institute, Badalona, Spain
| | - Evgenia Shishkova
- Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI, USA
- National Center for Quantitative Biology of Complex Systems, Madison, WI, USA
| | - Rachel E Rau
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, USA
| | | | - Joshua Coon
- Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI, USA
- National Center for Quantitative Biology of Complex Systems, Madison, WI, USA
- Department of Chemistry, University of Wisconsin, Madison, WI, USA
- Morgridge Institute for Research, Madison, WI, USA
| | - Qi Chen
- Molecular Medicine Program, Division of Urology, Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Eric L Van Nostrand
- Verna & Marrs McLean Department of Biochemistry & Molecular Biology and Therapeutic Innovation Center, Baylor College of Medicine, Houston, TX, USA
| | - Jose L Sardina
- Josep Carreras Leukaemia Research Institute, Badalona, Spain.
| | - Florian Grebien
- Institute for Medical Biochemistry, University of Veterinary Medicine Vienna, Vienna, Austria.
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria.
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria.
| | - Bruno Di Stefano
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA.
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA.
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA.
- Center for Cancer Epigenetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
4
|
Wu Q, Berglund AE, Macaulay RJ, Etame AB. Epigenetic Activation of TUSC3 Sensitizes Glioblastoma to Temozolomide Independent of MGMT Promoter Methylation Status. Int J Mol Sci 2023; 24:15179. [PMID: 37894860 PMCID: PMC10606804 DOI: 10.3390/ijms242015179] [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: 09/14/2023] [Revised: 10/10/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023] Open
Abstract
Temozolomide (TMZ) is an important first-line treatment for glioblastoma (GBM), but there are limitations to TMZ response in terms of durability and dependence on the promoter methylation status of the DNA repair gene O6-methylguanine DNA methyltransferase (MGMT). MGMT-promoter-hypermethylated (MGMT-M) GBMs are more sensitive to TMZ than MGMT-promoter-hypomethylated (MGMT-UM) GBMs. Moreover, TMZ resistance is inevitable even in TMZ-sensitive MGMT-M GBMs. Hence, epigenetic reprogramming strategies are desperately needed in order to enhance TMZ response in both MGMT-M and MGMT-UM GBMs. In this study, we present novel evidence that the epigenetic reactivation of Tumor Suppressor Candidate 3 (TUSC3) can reprogram sensitivity of GBM stem cells (GSCs) to TMZ irrespective of MGMT promoter methylation status. Interrogation of TCGA patient GBM datasets confirmed TUSC3 promoter regulation of TUSC3 expression and also revealed a strong positive correlation between TUSC3 expression and GBM patient survival. Using a combination of loss-of-function, gain-of-function and rescue studies, we demonstrate that TUSC3 reactivation is associated with enhanced TMZ response in both MGMT-M and MGMT-UM GSCs. Further, we provide novel evidence that the demethylating agent 5-Azacitidine (5-Aza) reactivates TUSC3 expression in MGMT-M GSCs, whereas the combination of 5-Aza and MGMT inhibitor Lomeguatrib is necessary for TUSC3 reactivation in MGMT-UM GSCs. Lastly, we propose a pharmacological epigenetic reactivation strategy involving TUSC3 that leads to significantly prolonged survival in MGMT-M and MGMT-UM orthotopic GSCs models. Collectively, our findings provide a framework and rationale to further explore TUSC3-mediated epigenetic reprogramming strategies that could enhance TMZ sensitivity and outcomes in GBM. Mechanistic and translational evidence gained from such studies could contribute towards optimal design of impactful trials for MGMT-UM GBMs that currently do not have good treatment options.
Collapse
Affiliation(s)
- Qiong Wu
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - Anders E. Berglund
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - Robert J. Macaulay
- Department of Anatomic Pathology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - Arnold B. Etame
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA
| |
Collapse
|
5
|
Siri G, Mosallaei M, Ehtesham N, Rahimi H, Mazarei M, Nasrollahzadeh Sabet M, Behroozi J. TUSC3 Methylation in Peripheral Blood Cells as a Biomarker for Diagnosis of Colorectal Cancer. Adv Biomed Res 2023; 12:174. [PMID: 37564442 PMCID: PMC10410437 DOI: 10.4103/abr.abr_396_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 02/14/2023] [Accepted: 02/21/2023] [Indexed: 08/12/2023] Open
Abstract
Background Several case-control studies have suggested that global and loci-specific deoxyribonucleic acid (DNA) methylation in peripheral blood mononuclear cells (PBMCs) of DNA might be potential biomarkers of cancer diagnosis and prognosis. In this study, for the first time, we intended to assess the diagnostic power of the methylation level of tumor suppressor candidate 3 (TUSC3) gene promoter in patients with colorectal cancer (CRC). Materials and Methods In the current study, we quantitatively assessed the promoter methylation level of TUSC3 in PBMCs of 70 CRC cases and 75 non-cancerous subjects via methylation quantification of endonuclease-resistant DNA (MethyQESD) method. Results The methylation level of the TUSC3 was meaningfully higher in CRC cases than in non-CRC subjects (43.55 ± 21.80% vs. 16.07 ± 13.63%, respectively; P < 0.001). The sensitivity and specificity of this gene for the detection of CRC were 88.6% and 76.0%, respectively. The receiver operating characteristic (ROC) curve examination discovered an area under the curve (AUC) of 0.880, representing a very high accuracy of the TUSC3 methylation marker in distinguishing CRC subjects from healthy individuals. However, there was no substantial diversity in methylation level between various CRC stages (P: 0.088). Conclusion For CRC screening, PBMCs are a reliable source for DNA methylation analysis and TUSC3 promoter methylation can be utilized as a hopeful biomarker for early and non-invasive diagnosis of CRC.
Collapse
Affiliation(s)
- Goli Siri
- Department of Internal Medicine, Amir-Alam Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Meysam Mosallaei
- Department of Genetics and Advanced Medical Technology, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Naeim Ehtesham
- School of Medicine, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | - Hasan Rahimi
- Faculty of Biostatistics, Tarbiat Modares University, Tehran, Iran
| | - Madineh Mazarei
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Science, Shahrekord, Iran
| | - Mehrdad Nasrollahzadeh Sabet
- Department of Genetics and Advanced Medical Technology, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Javad Behroozi
- Department of Genetics and Advanced Medical Technology, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran
- Department of Genetics and Advanced Medical Technology, Faculty of Medicine, AJA University of Medical Sciences; Research Center for Cancer Screening and Epidemiology, AJA University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Deng R, Lu X, Hong C, Cai R, Wang P, Xiong L, Wang X, Chen Q, Lin J. Downregulation of TUSC3 promotes EMT and hepatocellular carcinoma progression through LIPC/AKT axis. Lab Invest 2022; 20:485. [PMID: 36274132 PMCID: PMC9590144 DOI: 10.1186/s12967-022-03690-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 10/06/2022] [Indexed: 11/26/2022]
Abstract
Background Hepatocellular carcinoma (HCC) is one of the most common and malignant tumors in the digestive tract. Tumor Suppressor Candidate 3 (TUSC3) is one subunit of the endoplasmic reticulum Oligosaccharyl transferase (OST) complex, which plays an important role in N-glycosylation during the protein folding process. However, the role of TUSC3 in the initiation and progression of HCC has not been mentioned yet. In the present study, we aim to investigate the effects of TUSC3 on the initiation and progression of HCC. Methods Immunohistochemical assay and qRT-PCR were used to detect the expression of TUSC3 and lipase C hepatic type (LIPC) in HCC tissue and cells. Loss-of-function and gain-of-function were applied to detect the function of TUSC3 and LIPC in vivo and in vitro. Immunofluorescence assay and co-immunoprecipitation were used to detect the relationship between TUSC3 and LPC. Western blot was applied to detect the expression of epithelial–mesenchymal transition (EMT) markers and the Akt signaling pathway. Results TUSC3 was aberrantly decreased in hepatocellular carcinoma tissues compared to the matched adjacent normal tissues, which resulted in bigger size of tumor (P = 0.001, Table 2), worse differentiation (P = 0.006, Table 2) and an advanced BCLC stage. Down-regulation of TUSC3 led to the enhanced proliferation and migration of hepatocellular carcinoma cells in vivo and vitro, whereas the opposite effect could be observed in the TUSC3-overexpression group. The analysis of TUSC3 microarray showed that LIPC, a glycoprotein primarily synthesized and secreted by hepatocytes, was a downstream target of TUSC3, and it negatively modulated the development of HCC. The morphological changes in HCC cells indicated that TUSC3 regulated the epithelial-mesenchymal transition (EMT). Mechanistically, TUSC3 inhibited EMT progression through the LIPC/AKT axis. Conclusion Down-regulation of TUSC3 promotes EMT progression by activating AKT signaling via targeting LIPC in HCC, which is probably the possible mechanism driving TUSC3-deficient hepatocellular carcinoma cells toward a malignant phenotype.
Collapse
|
7
|
Sun F, Jie Q, Li Q, Wei Y, Li H, Yue X, Ma Y. TUSC3 inhibits cell proliferation and invasion in cervical squamous cell carcinoma via suppression of the AKT signalling pathway. J Cell Mol Med 2022; 26:1629-1642. [PMID: 35137520 PMCID: PMC8899155 DOI: 10.1111/jcmm.17204] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 12/28/2021] [Accepted: 01/10/2022] [Indexed: 12/24/2022] Open
Abstract
The decreased expression of tumour suppressor candidate 3 (TUSC3) is associated with proliferation in several types of cancer, leading to an unfavourable prognosis. The present study aimed to assess the cellular and molecular function of TUSC3 in patients with cervical squamous cell carcinoma (CSCC). Levels of mRNA expressions of TUSC3 were analysed in CSCC tissues and six cell lines using qRT-PCR. Immunohistochemistry(IHC) was used to evaluate the protein expression level of TUSC3 in four paired specimens, 220 paraffin-embedded CSCC specimens and 60 cases of normal cervical tissues(NCTs), respectively. Short hairpin RNA interference was employed for TUSC3 knockdown. Cell proliferation, migration and invasion were evaluated using growth curve, MTT assay, wound healing, transwell assay and xenograft tumour model, respectively. The results demonstrated that TUSC3 mRNA and protein expression levels were downregulated in CSCC samples. Multivariate and univariate analyses indicated that TUSC3 was an independent prognostic factor for patients with CSCC. Decreased TUSC3 expression levels were significantly associated with proliferation and an aggressive phenotype of cervical cancer cells both in vitro and in vivo. Moreover, the knockdown of TUSC3 promoted migration and invasion of cancer cells, while the increased expression of TUSC3 exhibited the opposite effects. The downregulation of TUSC3 facilitated proliferation and invasion of CSCC cells through the activation of the AKT signalling pathway. Our data demonstrated that the downregulation of TUSC3 promoted CSCC cell metastasis via the AKT signalling pathway. Therefore, TUSC3 may serve as a novel prognostic marker and potential target for CSCC.
Collapse
Affiliation(s)
- Fei Sun
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic ResearchHainan Provincial Clinical Research Center for Thalassemiathe Key Laboratory of Tropical Translational Medicine of Ministry of EducationDepartment of Reproductive Medicinethe First Affiliated Hospital of Hainan Medical UniversityHainan Medical UniversityHaikouHainanP.R. China
- Department of Obstetrics and GynecologyNanfang HospitalSouthern Medical UniversityGuangdongChina
- Haikou Key Laboratory for Preservation of Human Genetic Resourcethe First Affiliated Hospital of Hainan Medical UniversityHaikouHainanChina
| | - Qiuling Jie
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic ResearchHainan Provincial Clinical Research Center for Thalassemiathe Key Laboratory of Tropical Translational Medicine of Ministry of EducationDepartment of Reproductive Medicinethe First Affiliated Hospital of Hainan Medical UniversityHainan Medical UniversityHaikouHainanP.R. China
- Department of Obstetrics and GynecologyNanfang HospitalSouthern Medical UniversityGuangdongChina
- Haikou Key Laboratory for Preservation of Human Genetic Resourcethe First Affiliated Hospital of Hainan Medical UniversityHaikouHainanChina
| | - Qi Li
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic ResearchHainan Provincial Clinical Research Center for Thalassemiathe Key Laboratory of Tropical Translational Medicine of Ministry of EducationDepartment of Reproductive Medicinethe First Affiliated Hospital of Hainan Medical UniversityHainan Medical UniversityHaikouHainanP.R. China
- Haikou Key Laboratory for Preservation of Human Genetic Resourcethe First Affiliated Hospital of Hainan Medical UniversityHaikouHainanChina
- Hainan Modern Women and Children’s HospitialReproductive MedicineHaikouHainanChina
| | - Yunjian Wei
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic ResearchHainan Provincial Clinical Research Center for Thalassemiathe Key Laboratory of Tropical Translational Medicine of Ministry of EducationDepartment of Reproductive Medicinethe First Affiliated Hospital of Hainan Medical UniversityHainan Medical UniversityHaikouHainanP.R. China
- Department of Obstetrics and GynecologyNanfang HospitalSouthern Medical UniversityGuangdongChina
- Haikou Key Laboratory for Preservation of Human Genetic Resourcethe First Affiliated Hospital of Hainan Medical UniversityHaikouHainanChina
| | - Hong Li
- Department of Obstetrics and GynecologyNanfang HospitalSouthern Medical UniversityGuangdongChina
| | - Xiaojing Yue
- Department of Obstetrics and GynecologyNanfang HospitalSouthern Medical UniversityGuangdongChina
| | - Yanlin Ma
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic ResearchHainan Provincial Clinical Research Center for Thalassemiathe Key Laboratory of Tropical Translational Medicine of Ministry of EducationDepartment of Reproductive Medicinethe First Affiliated Hospital of Hainan Medical UniversityHainan Medical UniversityHaikouHainanP.R. China
- Department of Obstetrics and GynecologyNanfang HospitalSouthern Medical UniversityGuangdongChina
- Haikou Key Laboratory for Preservation of Human Genetic Resourcethe First Affiliated Hospital of Hainan Medical UniversityHaikouHainanChina
| |
Collapse
|
8
|
Lin H, Wang J, Wang T, Wu J, Wang P, Huo X, Zhang J, Pan H, Fan Y. The LncRNA MIR503HG/miR-224-5p/TUSC3 Signaling Cascade Suppresses Gastric Cancer Development via Modulating ATF6 Branch of Unfolded Protein Response. Front Oncol 2021; 11:708501. [PMID: 34381729 PMCID: PMC8352579 DOI: 10.3389/fonc.2021.708501] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 07/08/2021] [Indexed: 12/29/2022] Open
Abstract
Background Unfolded protein response (UPR)-mediated tumor-promoting functions have been identified in multiple cancers, and this study focused on investigating the role and molecular mechanisms of UPR in modulating gastric cancer (GC) pathogenesis. Methods The bioinformatics analysis was performed to examine the expression status of cancer associated genes in patients with stomach adenocarcinoma (STAD) and predict the targeting sites of miR-224-5p with LncRNA MIR503HG and TUSC3. Genes expressions were quantified by Real-Time qPCR, Western Blot and immunohistochemistry (IHC). Cell proliferation, viability, apoptosis and mobility were evaluated by MTT assay, trypan blue staining assay, flow cytometer and transwell assay, respectively. The binding sites were validated by dual-luciferase reporter gene system assay. Results LncRNA MIR503HG and TUSC3 were downregulated, but miR-224-5p was upregulated in GC tissues and cells, in contrast with their normal counterparts. Further gain- and loss-of-function experiments validated that the malignant phenotypes in GC cells, including cell proliferation, invasion, epithelial-mesenchymal transition (EMT) and tumorigenesis, were negatively regulated by LncRNA MIR503HG. Mechanistically, LncRNA MIR503HG upregulated TUSC3 in GC cells through sponging miR-224-5p, resulting in the repression of GC progression. Finally, we validated that knock-down of ATF6, but not other two branches of UPR (PERK1 and IRE1), partially rescued cell proliferation and EMT in the GC cells with LncRNA MIR503HG overexpression. Conclusions Targeting the LncRNA MIR503HG/miR-224-5p/TUSC3 signaling cascade suppressed ATF6-mediated UPR, resulting in the blockage of GC development.
Collapse
Affiliation(s)
- Han Lin
- Department of General Surgery, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China.,Department of Formulas of Chinese Medicine of Basic Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Jinge Wang
- The Second Affiliated Hospital & College of Nursing, Harbin Medical University, Harbin, China
| | - Tong Wang
- Department of General Surgery, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jiaming Wu
- Department of General Surgery, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Peng Wang
- Department of General Surgery, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiaoyan Huo
- Department of General Surgery, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jun Zhang
- Department of General Surgery, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Huayang Pan
- Department of General Surgery, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yuying Fan
- The Second Affiliated Hospital & College of Nursing, Harbin Medical University, Harbin, China
| |
Collapse
|
9
|
Ren Y, Deng R, Cai R, Lu X, Luo Y, Wang Z, Zhu Y, Yin M, Ding Y, Lin J. TUSC3 induces drug resistance and cellular stemness via Hedgehog signaling pathway in colorectal cancer. Carcinogenesis 2021; 41:1755-1766. [PMID: 32338281 DOI: 10.1093/carcin/bgaa038] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 04/08/2020] [Accepted: 04/16/2020] [Indexed: 12/14/2022] Open
Abstract
Tumor suppressor candidate 3 (TUSC3) is a coding gene responsible for N-glycosylation of many critical proteins. TUSC3 gene plays an oncogenic role in colorectal cancer (CRC), however, the role of TUSC3 in drug resistance of CRC is still unclear. The aim of this study is to investigate the biological function and molecular mechanism of TUSC3 in CRC drug resistance. The expression of TUSC3 in CRC is positively correlated to tumor stage in 90 paired clinical samples, and negatively associated with overall survival and disease-free survival of CRC patients. In vitro, TUSC3 promotes the formation of stemness and induces the drug resistance to 5-fluorouracil and cis-dichlorodiammineplatinum(II) in CRC cells. The tissue microarray assay and bioinformatic analysis indicate that TUSC3 may promote the expression of CD133 and ABCC1 via Hedgehog signaling pathway. Treatment of Hedgehog signaling pathway agonist or inhibitor in TUSC3-silenced or TUSC3-overexpressed cells reverse the effects of TUSC3 in cellular stemness phenotype and drug resistance. Meanwhile, coimmunoprecipitation and immunofluorescence assays indicate a tight relationship between TUSC3 and SMO protein. Our data suggest that TUSC3 promotes the formation of cellular stemness and induces drug resistance via Hedgehog signaling pathway in CRC.
Collapse
Affiliation(s)
- Yansong Ren
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, PR China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, PR China.,Department of Pathology, Guangdong Provincial Key Laboratory of Molecular Oncologic Pathology, Guangzhou, Guangdong Province, PR China
| | - Ruxia Deng
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, PR China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, PR China.,Department of Pathology, Guangdong Provincial Key Laboratory of Molecular Oncologic Pathology, Guangzhou, Guangdong Province, PR China
| | - Rui Cai
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, PR China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, PR China.,Department of Pathology, Guangdong Provincial Key Laboratory of Molecular Oncologic Pathology, Guangzhou, Guangdong Province, PR China
| | - Xiansheng Lu
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, PR China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, PR China.,Department of Pathology, Guangdong Provincial Key Laboratory of Molecular Oncologic Pathology, Guangzhou, Guangdong Province, PR China
| | - Yuejun Luo
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, PR China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, PR China.,Department of Pathology, Guangdong Provincial Key Laboratory of Molecular Oncologic Pathology, Guangzhou, Guangdong Province, PR China
| | - Ziyuan Wang
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, PR China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, PR China.,Department of Pathology, Guangdong Provincial Key Laboratory of Molecular Oncologic Pathology, Guangzhou, Guangdong Province, PR China
| | - Yuchen Zhu
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, PR China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, PR China.,Department of Pathology, Guangdong Provincial Key Laboratory of Molecular Oncologic Pathology, Guangzhou, Guangdong Province, PR China
| | - Mengyuan Yin
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, PR China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, PR China.,Department of Pathology, Guangdong Provincial Key Laboratory of Molecular Oncologic Pathology, Guangzhou, Guangdong Province, PR China
| | - Yanqing Ding
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, PR China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, PR China.,Department of Pathology, Guangdong Provincial Key Laboratory of Molecular Oncologic Pathology, Guangzhou, Guangdong Province, PR China
| | - Jie Lin
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, PR China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, PR China.,Department of Pathology, Guangdong Provincial Key Laboratory of Molecular Oncologic Pathology, Guangzhou, Guangdong Province, PR China
| |
Collapse
|
10
|
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
|
11
|
Kawahara R, Recuero S, Srougi M, Leite KRM, Thaysen-Andersen M, Palmisano G. The Complexity and Dynamics of the Tissue Glycoproteome Associated With Prostate Cancer Progression. Mol Cell Proteomics 2021; 20:100026. [PMID: 33127837 PMCID: PMC8010466 DOI: 10.1074/mcp.ra120.002320] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 10/19/2020] [Accepted: 10/30/2020] [Indexed: 12/30/2022] Open
Abstract
The complexity and dynamics of the immensely heterogeneous glycoproteome of the prostate cancer (PCa) tumor microenvironment remain incompletely mapped, a knowledge gap that impedes our molecular-level understanding of the disease. To this end, we have used sensitive glycomics and glycoproteomics to map the protein-, cell-, and tumor grade-specific N- and O-glycosylation in surgically removed PCa tissues spanning five histological grades (n = 10/grade) and tissues from patients with benign prostatic hyperplasia (n = 5). Quantitative glycomics revealed PCa grade-specific alterations of the oligomannosidic-, paucimannosidic-, and branched sialylated complex-type N-glycans, and dynamic remodeling of the sialylated core 1- and core 2-type O-glycome. Deep quantitative glycoproteomics identified ∼7400 unique N-glycopeptides from 500 N-glycoproteins and ∼500 unique O-glycopeptides from nearly 200 O-glycoproteins. With reference to a recent Tissue and Blood Atlas, our data indicate that paucimannosidic glycans of the PCa tissues arise mainly from immune cell-derived glycoproteins. Furthermore, the grade-specific PCa glycosylation arises primarily from dynamics in the cellular makeup of the PCa tumor microenvironment across grades involving increased oligomannosylation of prostate-derived glycoproteins and decreased bisecting GlcNAcylation of N-glycans carried by the extracellular matrix proteins. Furthermore, elevated expression of several oligosaccharyltransferase subunits and enhanced N-glycoprotein site occupancy were observed associated with PCa progression. Finally, correlations between the protein-specific glycosylation and PCa progression were observed including increased site-specific core 2-type O-glycosylation of collagen VI. In conclusion, integrated glycomics and glycoproteomics have enabled new insight into the complexity and dynamics of the tissue glycoproteome associated with PCa progression generating an important resource to explore the underpinning disease mechanisms.
Collapse
Affiliation(s)
- Rebeca Kawahara
- Departamento de Parasitologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, USP, São Paulo, Brazil; Department of Molecular Sciences, Macquarie University, Sydney, NSW, Australia; Biomolecular Discovery Research Centre, Macquarie University, Sydney, NSW, Australia
| | - Saulo Recuero
- Laboratório de Investigação Médica da Disciplina de Urologia da Faculdade de Medicina da USP, São Paulo, Brazil
| | - Miguel Srougi
- Laboratório de Investigação Médica da Disciplina de Urologia da Faculdade de Medicina da USP, São Paulo, Brazil
| | - Katia R M Leite
- Laboratório de Investigação Médica da Disciplina de Urologia da Faculdade de Medicina da USP, São Paulo, Brazil
| | - Morten Thaysen-Andersen
- Department of Molecular Sciences, Macquarie University, Sydney, NSW, Australia; Biomolecular Discovery Research Centre, Macquarie University, Sydney, NSW, Australia.
| | - Giuseppe Palmisano
- Departamento de Parasitologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, USP, São Paulo, Brazil.
| |
Collapse
|
12
|
Patel C, Saad H, Shenkman M, Lederkremer GZ. Oxidoreductases in Glycoprotein Glycosylation, Folding, and ERAD. Cells 2020; 9:cells9092138. [PMID: 32971745 PMCID: PMC7563561 DOI: 10.3390/cells9092138] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/17/2020] [Accepted: 09/18/2020] [Indexed: 12/17/2022] Open
Abstract
N-linked glycosylation and sugar chain processing, as well as disulfide bond formation, are among the most common post-translational protein modifications taking place in the endoplasmic reticulum (ER). They are essential modifications that are required for membrane and secretory proteins to achieve their correct folding and native structure. Several oxidoreductases responsible for disulfide bond formation, isomerization, and reduction have been shown to form stable, functional complexes with enzymes and chaperones that are involved in the initial addition of an N-glycan and in folding and quality control of the glycoproteins. Some of these oxidoreductases are selenoproteins. Recent studies also implicate glycan machinery–oxidoreductase complexes in the recognition and processing of misfolded glycoproteins and their reduction and targeting to ER-associated degradation. This review focuses on the intriguing cooperation between the glycoprotein-specific cell machineries and ER oxidoreductases, and highlights open questions regarding the functions of many members of this large family.
Collapse
Affiliation(s)
- Chaitanya Patel
- The Shmunis School of Biomedicine and Cancer Research, Cell Biology Division, George Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel; (C.P.); (H.S.); (M.S.)
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel
| | - Haddas Saad
- The Shmunis School of Biomedicine and Cancer Research, Cell Biology Division, George Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel; (C.P.); (H.S.); (M.S.)
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel
| | - Marina Shenkman
- The Shmunis School of Biomedicine and Cancer Research, Cell Biology Division, George Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel; (C.P.); (H.S.); (M.S.)
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel
| | - Gerardo Z. Lederkremer
- The Shmunis School of Biomedicine and Cancer Research, Cell Biology Division, George Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel; (C.P.); (H.S.); (M.S.)
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel
- Correspondence:
| |
Collapse
|
13
|
Kong L, Sun Y, Chen M, Dai Y, Liu Z. Downregulation of microRNA-320a inhibits proliferation and induces apoptosis of retinoblastoma cells via targeting TUSC3. Exp Ther Med 2020; 20:9. [PMID: 32934674 PMCID: PMC7471862 DOI: 10.3892/etm.2020.9137] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Accepted: 05/20/2020] [Indexed: 02/07/2023] Open
Abstract
MicroRNA (miR)-320a is specific to vertebrates and has been indicated to serve a role in a number of cancer types, such as gastric, colorectal, pancreatic and ovarian cancer. miR-320a has been reported to be expressed at high levels in retinoblastoma tissues; however its role and mechanism of function in retinoblastoma remain to be elucidated. The aim of the present study was to investigate the role of miR-320a in retinoblastoma cells and the underlying mechanisms. The expression of miR-320a in retinoblastoma cell lines Y79 and WERI-Rb-1, and normal human retinal pigment epithelial cell line ARPE-19 was examined via reverse transcription-quantitative PCR (RT-qPCR). TargetScan bioinformatics analysis and dual-luciferase reporter assay were used to predict and reveal the target gene of miR-320a. Target gene expression was detected via RT-qPCR in retinoblastoma cell lines and ARPE-19 cells. Subsequently, gain- or loss-of-function experiments for miR-320a and tumor suppressor candidate 3 (TUSC3) were performed to study the role of miR-320a/TUSC3 in retinoblastoma cells. Cell viability and apoptosis were assessed via MTT and flow cytometry analysis, respectively. Compared with ARPE-19 cells, miR-320a was prominently expressed in retinoblastoma cell lines. TUSC3 was predicted to be a target gene of miR-320a. Compared with ARPE-19 cells, the expression of TUSC3 in retinoblastoma cell lines was reduced. The results of MTT and flow cytometry analysis revealed that overexpression of TUSC3 reduced the viability of retinoblastoma cells and induced apoptosis. Additional analysis indicated that miR-320a inhibitor enhanced the expression of the target gene TUSC3, thereby inhibiting retinoblastoma cell viability and inducing apoptosis. The effects of miR-320a inhibitor on retinoblastoma cells were inhibited by TUSC3-short hairpin RNA. miR-320a regulated the viability and apoptosis of retinoblastoma cells via targeting TUSC3. Therefore, the present study provided a reference for investigating a potential target for the clinical treatment of retinoblastoma.
Collapse
Affiliation(s)
- Li Kong
- Department of Ophthalmology, Chongqing Aier Eye Hospital, Chongqing 400020, P.R. China
| | - Yang Sun
- Department of Ophthalmology, Chongqing Aier Eye Hospital, Chongqing 400020, P.R. China
| | - Maosheng Chen
- Department of Ophthalmology, Chongqing Aier Eye Hospital, Chongqing 400020, P.R. China
| | - Yan Dai
- Department of Ophthalmology, Mianyang Central Hospital, Mianyang, Sichuan 621000, P.R. China
| | - Zhen Liu
- Department of Ophthalmology, Chongqing Aier Eye Hospital, Chongqing 400020, P.R. China
| |
Collapse
|
14
|
Wang S, Zhu W. Tumour suppressor candidate 3 inhibits biological function and increases endoplasmic reticulum stress of melanoma cells WM451 by regulating AKT/GSK3-β/β-catenin pathway. Cell Biochem Funct 2020; 38:604-612. [PMID: 32090352 DOI: 10.1002/cbf.3515] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 02/02/2020] [Accepted: 02/05/2020] [Indexed: 12/17/2022]
Abstract
Melanoma is a highly malignant and is a life-threatening disease with no effective treatment currently. This study aims to evaluate the significance of TUSC3, an endoplasmic reticulum stress (ERS)-inducible gene and explore its relationship with AKT/GSK3-β/β-catenin signalling pathway in melanoma cell WM451. We investigated TUSC3 expression in melanoma cell by qRT-PCR, CCK-8 and clonal formation assays were utilized to evaluate cell proliferation. Wound healing and transwell experiments detected cell migration and invasion. Flow cytometry detected the level of apoptosis. Western blot analysed MMP2, MMP9, p-AKT, p-GSK3-β, β-catenin and AKT, GSK3-β, ERS-related proteins and apoptosis-related proteins in WM451 cells. The results revealed that TUSC3 was remarkably decreased in melanoma cell lines. Overexpression of TUSC3 significantly inhibits melanoma cell WM451 biological functions and promotes expression of ERS-related proteins in WM451 cells, increases ERS in WM451 cells by inhibiting AKT/GSK3-β/β-catenin pathway. These finding suggest that TUSC3 regulates biological functions of melanoma cells WM451 and increases ERS in melanoma cells WM451 via the inhibition of the AKT/GSK3-β/β-catenin signalling pathway. SIGNIFICANCE OF THE STUDY: Melanoma is a highly malignant and is a life-threatening disease with no effective treatment currently. Therefore, studying the molecular mechanism of melanoma occurrence and metastasis is essential for the treatment of melanoma. Meanwhile, mounting studies suggest that TUSC3 is considered to be closely associated with the development of various malignancies. TUSC3 regulates proliferation, migration and epithelial-to-mesenchymal transition, but the molecular mechanism of the tumour suppressor effects of TUSC3 on melanoma cells is not well understood. Our study demonstrates that TUSC3 inhibits biological function of melanoma cells and increases ERS in melanoma cells by inhibiting AKT/GSK3-β/β-catenin pathway. And this is expected to be a new target and method for the treatment of melanoma.
Collapse
Affiliation(s)
- Shoujie Wang
- Department of Plastic Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Zhejiang, China
| | - Weifang Zhu
- Department of Dermatology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Zhejiang, China
| |
Collapse
|
15
|
Harada Y, Ohkawa Y, Kizuka Y, Taniguchi N. Oligosaccharyltransferase: A Gatekeeper of Health and Tumor Progression. Int J Mol Sci 2019; 20:ijms20236074. [PMID: 31810196 PMCID: PMC6929149 DOI: 10.3390/ijms20236074] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 11/28/2019] [Accepted: 11/28/2019] [Indexed: 02/06/2023] Open
Abstract
Oligosaccharyltransferase (OST) is a multi-span membrane protein complex that catalyzes the addition of glycans to selected Asn residues within nascent polypeptides in the lumen of the endoplasmic reticulum. This process, termed N-glycosylation, is a fundamental post-translational protein modification that is involved in the quality control, trafficking of proteins, signal transduction, and cell-to-cell communication. Given these crucial roles, N-glycosylation is essential for homeostasis at the systemic and cellular levels, and a deficiency in genes that encode for OST subunits often results in the development of complex genetic disorders. A growing body of evidence has also demonstrated that the expression of OST subunits is cell context-dependent and is frequently altered in malignant cells, thus contributing to tumor cell survival and proliferation. Importantly, a recently developed inhibitor of OST has revealed this enzyme as a potential target for the treatment of incurable drug-resistant tumors. This review summarizes our current knowledge regarding the functions of OST in the light of health and tumor progression, and discusses perspectives on the clinical relevance of inhibiting OST as a tumor treatment.
Collapse
Affiliation(s)
- Yoichiro Harada
- Department of Glyco-Oncology and Medical Biochemistry, Osaka International Cancer Institute, 3-1-69 Otemae, Chuo-ku, Osaka 541-8567, Japan; (Y.H.); (Y.O.)
| | - Yuki Ohkawa
- Department of Glyco-Oncology and Medical Biochemistry, Osaka International Cancer Institute, 3-1-69 Otemae, Chuo-ku, Osaka 541-8567, Japan; (Y.H.); (Y.O.)
| | - Yasuhiko Kizuka
- Center for Highly Advanced Integration of Nano and Life Sciences (G-CHAIN), Gifu University, Gifu 501-1193, Japan;
| | - Naoyuki Taniguchi
- Department of Glyco-Oncology and Medical Biochemistry, Osaka International Cancer Institute, 3-1-69 Otemae, Chuo-ku, Osaka 541-8567, Japan; (Y.H.); (Y.O.)
- Correspondence: ; Tel.: +81-6-6945-1181
| |
Collapse
|
16
|
Chen S, Liu Y, Wang Y, Xue Z. LncRNA CCAT1 Promotes Colorectal Cancer Tumorigenesis Via A miR-181b-5p/TUSC3 Axis. Onco Targets Ther 2019; 12:9215-9225. [PMID: 31807005 PMCID: PMC6842281 DOI: 10.2147/ott.s216718] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 09/12/2019] [Indexed: 01/17/2023] Open
Abstract
AIM The aim was to determine the function and molecular mechanism of long non-coding RNA colon cancer associated transcript-1(lncRNA CCAT1) in the development of colorectal cancer (CRC). METHODS CCAT1 mRNA expression levels were determined in CRC tissues and cells using reverse transcription-quantitative polymerase chain reaction. Cell Counting Kit-8 and colony formation assays were used to examine the effects of CCAT1 on the proliferation of CRC cells. Luciferase reporter gene analysis was used to confirm the target gene of microRNA-181b-5p (miR-181b-5p) in CRC cells. Tumor xenografts were subsequently used to investigate the role of CCAT1 in CRC growth in vivo. RESULTS The relative mRNA expression levels of CCAT1 were significantly higher in CRC tissues and cell lines compared with the normal tissues or cells. CCAT1 knockdown significantly inhibited CRC cell proliferation in vitro and in vivo. Bioinformatics and luciferase reporter assays showed that miR-181b-5p was a direct target of CCAT1, and the expression of miR-181b-5p was negatively correlated with the expression of CCAT1 in CRC tissues. Furthermore, CCAT1 positively regulated the level of tumor suppressor candidate 3 (TUSC3) by competing with miR-181b-5p in CRC cells. CONCLUSION These data suggested that lncRNA CCAT1 promoted colorectal cancer tumorigenesis via a miR-181b-5p/TUSC3 axis.
Collapse
Affiliation(s)
- Si Chen
- Department of Colorectal and Anal Surgery
| | | | | | - Zhaoping Xue
- Anaesthesia Wake Room, First Hospital of Jilin University, Changchun, Jilin130021, People’s Republic of China
| |
Collapse
|
17
|
Identifying small molecule probes of ENTPD5 through high throughput screening. PLoS One 2019; 14:e0210305. [PMID: 31242188 PMCID: PMC6594577 DOI: 10.1371/journal.pone.0210305] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 06/15/2019] [Indexed: 12/18/2022] Open
Abstract
Ectonucleoside Triphosphate Diphosphohydrolase 5 (ENTPD5) has been shown to be important in maintaining cellular function in cancer, and its expression is upregulated through multiple, unique pathways in certain cancers, including laryngeal, glioblastoma multiforme, breast, testicular, and prostate. ENTPD5 supports cancer growth by promoting the import of UDP-glucose, a metabolite used for protein glycosylation and hence proper glycoprotein folding, into the ER by providing the counter molecule, UMP, to the ER antiporter. Despite its cancer-supporting function, no small molecule inhibitors of ENTPD5 are commercially available, and few studies have been performed in tissue culture to understand the effects of chemical inhibition of ENTPD5. We performed a high-throughput screen (HTS) of 21,120 compounds to identify small molecule inhibitors of ENPTD5 activity. Two hits were identified, and we performed a structure activity relationship (SAR) screen around these hits. Further validation of these probes were done in an orthogonal assay and then assayed in cell culture to assess their effect on prostate cancer cell lines. Notably, treatment with the novel ENTPD5 inhibitor reduced the amount of glycoprotein produced in treated cells, consistent with the hypothesis that ENTPD5 is important for glycoprotein folding. This work serves as an important step in designing new molecular probes for ENTPD5 as well as further probing the utility of targeting ENTPD5 to combat cancer cell proliferation.
Collapse
|
18
|
Yan Y, Chen Z, Liao Y, Zhou J. TUSC3 as a potential biomarker for prognosis in clear cell renal cell carcinoma. Oncol Lett 2019; 17:5073-5079. [PMID: 31186719 PMCID: PMC6507427 DOI: 10.3892/ol.2019.10161] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 02/26/2019] [Indexed: 12/31/2022] Open
Abstract
The aim of the present study was to explore the expression levels of tumor suppressor candidate 3 (TUSC3) in human clear cell renal cell carcinoma (ccRCC) and its clinical value. Immunohistochemical staining, western blotting and reverse transcription-quantitative polymerase chain reaction were used to detect TUSC3 expression in paracancerous normal tissues and ccRCC tissues. The tissues were derived from the pathological specimens of 54 patients with ccRCC. Additionally, associations among TUSC3 expression and histological grade and clinicopathological staging of ccRCC were investigated. The results of these comparisons revealed that TUSC3 expression in ccRCC tissues was significantly lower than that in paracancerous tissues (P<0.05). TUSC3 expression in the high differentiation group was higher than that in the median and low differentiation groups (P<0.05). Expression levels of TUSC3 in stage I and II tissues were higher than those in stage III and IV tissues (P<0.05). The expression levels of TUSC3 in the lymph node metastasis group were lower than those in the non-lymph node metastasis group (P<0.05). In conclusion, the expression levels of TUSC3 in human ccRCC tissues were downregulated compared with those found in normal human renal tissue, and TUSC3 may inhibit the progression of ccRCC. Furthermore, the TUSC3 gene may be used as a promising tumor marker for the early diagnosis and prognosis of ccRCC.
Collapse
Affiliation(s)
- Youji Yan
- Department of Urology, Jingzhou Central Hospital, The Second Clinical College, Yangtze University, Jingzhou, Hubei 434000, P.R. China
| | - Zhongjun Chen
- Department of Urology, Jingzhou Central Hospital, The Second Clinical College, Yangtze University, Jingzhou, Hubei 434000, P.R. China
| | - Yixiang Liao
- Department of Urology, Jingzhou Central Hospital, The Second Clinical College, Yangtze University, Jingzhou, Hubei 434000, P.R. China
| | - Jiajie Zhou
- Department of Urology, Jingzhou Central Hospital, The Second Clinical College, Yangtze University, Jingzhou, Hubei 434000, P.R. China
| |
Collapse
|
19
|
Wilk G, Braun R. regQTLs: Single nucleotide polymorphisms that modulate microRNA regulation of gene expression in tumors. PLoS Genet 2018; 14:e1007837. [PMID: 30557297 PMCID: PMC6343932 DOI: 10.1371/journal.pgen.1007837] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 01/23/2019] [Accepted: 11/17/2018] [Indexed: 02/07/2023] Open
Abstract
Genome-wide association studies (GWAS) have identified single nucleotide polymorphisms (SNPs) associated with trait diversity and disease susceptibility, yet their functional properties often remain unclear. It has been hypothesized that SNPs in microRNA binding sites may disrupt gene regulation by microRNAs (miRNAs), short non-coding RNAs that bind to mRNA and downregulate the target gene. While several studies have predicted the location of SNPs in miRNA binding sites, to date there has been no comprehensive analysis of their impact on miRNA regulation. Here we investigate the functional properties of genetic variants and their effects on miRNA regulation of gene expression in cancer. Our analysis is motivated by the hypothesis that distinct alleles may cause differential binding (from miRNAs to mRNAs or from transcription factors to DNA) and change the expression of genes. We previously identified pathways—systems of genes conferring specific cell functions—that are dysregulated by miRNAs in cancer, by comparing miRNA–pathway associations between healthy and tumor tissue. We draw on these results as a starting point to assess whether SNPs on dysregulated pathways are responsible for miRNA dysregulation of individual genes in tumors. Using an integrative regression analysis that incorporates miRNA expression, mRNA expression, and SNP genotype data, we identify functional SNPs that we term “regulatory QTLs (regQTLs)”: loci whose alleles impact the regulation of genes by miRNAs. We apply the method to breast, liver, lung, and prostate cancer data from The Cancer Genome Atlas, and provide a tool to explore the findings. Genomics studies have identified single nucleotide polymorphisms (SNPs) associated with trait diversity and disease susceptibility, yet the mechanism of action of many genetic variants remains unclear. MicroRNAs (miRNAs) are a class of small non-coding RNA molecules that base-pair coding mRNAs to regulate gene transcription. We hypothesize that SNP variants may affect the ability of miRNAs to bind their target genes, thus influencing gene regulation. To identify these “regulatory QTLs” (regQTLs), we integrate miRNA expression, mRNA expression, and SNP data to identify miRNAs that are associated with pathway dysregulation in tumors, and assess whether SNPs on these pathways are responsible for disrupted miRNA-gene regulation. This data-driven approach enables the discovery of SNPs whose alleles impact gene regulation by miRNAs, with functional consequences for tumor biology. We detail the method, apply it to data from The Cancer Genome Atlas, and provide a tool to explore the findings.
Collapse
Affiliation(s)
- Gary Wilk
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, Illinois, United States of America
- Biostatistics Division, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Rosemary Braun
- Biostatistics Division, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
- Department of Engineering Sciences and Applied Mathematics, Northwestern University, Evanston, Illinois, United States of America
- * E-mail:
| |
Collapse
|
20
|
Jeon YJ, Kim T, Park D, Nuovo GJ, Rhee S, Joshi P, Lee BK, Jeong J, Suh SS, Grotzke JE, Kim SH, Song J, Sim H, Kim Y, Peng Y, Jeong Y, Garofalo M, Zanesi N, Kim J, Liang G, Nakano I, Cresswell P, Nana-Sinkam P, Cui R, Croce CM. miRNA-mediated TUSC3 deficiency enhances UPR and ERAD to promote metastatic potential of NSCLC. Nat Commun 2018; 9:5110. [PMID: 30504895 PMCID: PMC6269493 DOI: 10.1038/s41467-018-07561-8] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 11/09/2018] [Indexed: 02/05/2023] Open
Abstract
Non-small cell lung carcinoma (NSCLC) is leading cause of cancer-related deaths in the world. The Tumor Suppressor Candidate 3 (TUSC3) at chromosome 8p22 known to be frequently deleted in cancer is often found to be deleted in advanced stage of solid tumors. However, the role of TUSC3 still remains controversial in lung cancer and context-dependent in several cancers. Here we propose that miR-224/-520c-dependent TUSC3 deficiency enhances the metastatic potential of NSCLC through the alteration of three unfolded protein response pathways and HRD1-dependent ERAD. ATF6α-dependent UPR is enhanced whereas the affinity of HRD1 to its substrates, PERK, IRE1α and p53 is weakened. Consequently, the alteration of UPRs and the suppressed p53-NM23H1/2 pathway by TUSC3 deficiency is ultimately responsible for enhancing metastatic potential of lung cancer. These findings provide mechanistic insight of unrecognized roles of TUSC3 in cancer progression and the oncogenic role of HRD1-dependent ERAD in cancer metastasis.
Collapse
Affiliation(s)
- Young-Jun Jeon
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Taewan Kim
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
| | - Dongju Park
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
| | - Gerard J Nuovo
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
| | - Siyeon Rhee
- Department of Biology, Stanford University, Stanford, CA, 94305, USA
| | - Pooja Joshi
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
| | - Bum-Kyu Lee
- Institute for Cellular and Molecular Biology, Center for Systems and Synthetic Biology, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Johan Jeong
- Department of Pathology, Stanford University, Stanford, CA, 94305, USA
| | - Sung-Suk Suh
- Department of Biosciences, Mokpo National University, Muan, 58554, South Korea
| | - Jeff E Grotzke
- Departments of Immunobiology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Sung-Hak Kim
- Department of Animal Science, College of Agriculture and Life Sciences, Chonnam National University, Gwangju, 61186, Korea
- Gwangju Center, Korea Basic Science Institute, Gwangju, 61186, Korea
| | - Jieun Song
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
| | - Hosung Sim
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
| | - Yonghwan Kim
- Department of Life System, Sookmyung Woman's University, Seoul, 140-742, Republic of Korea
| | - Yong Peng
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
- Department of Thoracic Surgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, 610041, Chengdu, China
| | - Youngtae Jeong
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Michela Garofalo
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
- Transcriptional Networks in Lung Cancer Group, Cancer Research United Kingdom Manchester Institute, University of Manchester, Manchester, M20 4BX, United Kingdom
| | - Nicola Zanesi
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, 43210, USA
| | - Jonghwan Kim
- Institute for Cellular and Molecular Biology, Center for Systems and Synthetic Biology, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Guang Liang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Ichiro Nakano
- Department of Neurosurgery UAB Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Peter Cresswell
- Departments of Immunobiology, Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Patrick Nana-Sinkam
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Medical Oncology, The Ohio State University, Columbus, OH, 43210, USA
| | - Ri Cui
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA.
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
| | - Carlo M Croce
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA.
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, 43210, USA.
| |
Collapse
|
21
|
Feng S, Zhai J, Lu D, Lin J, Dong X, Liu X, Wu H, Roden AC, Brandi G, Tavolari S, Bille A, Cai K. TUSC3 accelerates cancer growth and induces epithelial-mesenchymal transition by upregulating claudin-1 in non-small-cell lung cancer cells. Exp Cell Res 2018; 373:44-56. [PMID: 30098333 DOI: 10.1016/j.yexcr.2018.08.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 08/01/2018] [Accepted: 08/07/2018] [Indexed: 12/23/2022]
Abstract
Lung cancer is the most frequent cause of cancer-related deaths worldwide, but its molecular pathogenesis is poorly understood. The tumor suppressor candidate 3 (TUSC3) gene is located on chromosome 8p22 and is universally acknowledged as a cancer suppressor. However, our research has demonstrated that TUSC3 expression is significantly upregulated in non-small-cell lung cancer compared to benign controls. In this study, we analyzed the consequences of TUSC3 knockdown or overexpression on the biological functions of non-small-cell lung cancer cell lines. To identify the molecules and signaling pathways with which TUSC3 might interact, we completed immunoblotting, quantitative polymerase chain reaction, microarray, co-immunoprecipitation, and immunofluorescence assays. We demonstrated that TUSC3 knockdown leads to decreased proliferation, migration, and invasion, and reduced xenograft tumor growth of non-small-cell lung cancer cell lines, whereas opposite results were observed with overexpression of TUSC3. In addition, TUSC3 knockdown suppressed epithelial-mesenchymal transition by downregulating the expression of claudin-1, which plays an indispensable role in EMT progress. On the contrary, overexpression of TUSC3 significantly enhanced EMT progress by upregulating claudin-1 expression. Overall, our observations suggest that TUSC3 accelerates cancer growth and induces the epithelial-mesenchymal transition in non-small-cell lung cancer cells; we also identified claudin-1 as a target of TUSC3.
Collapse
Affiliation(s)
- Siyang Feng
- Department of Thoracic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China
| | - Jianxue Zhai
- Department of Thoracic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China
| | - Di Lu
- Department of Thoracic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China
| | - Jie Lin
- Department of Pathology, Nanfang Hospital & School of Basic Medicine, Southern Medical University, 1838 Guangzhou Avenue, Guangzhou 510515, PR China
| | - Xiaoying Dong
- Department of Thoracic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China
| | - Xiguang Liu
- Department of Thoracic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China
| | - Hua Wu
- Department of Thoracic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China
| | - Anja C Roden
- Department of Laboratory Medicine and Pathology, Mayo Clinic Rochester, MN, USA
| | - Giovanni Brandi
- Department of Experimental, Diagnostic and Specialty Medicine, University Hospital S. Orsola, Malpighi Bologna, via Massarenti 9, 40138, Italy
| | - Simona Tavolari
- Department of Experimental, Diagnostic and Specialty Medicine, University Hospital S. Orsola, Malpighi Bologna, via Massarenti 9, 40138, Italy
| | - Andrea Bille
- Department of Thoracic Surgery, Guy's Hospital, London, UK
| | - Kaican Cai
- Department of Thoracic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China.
| |
Collapse
|
22
|
Alleviation of endoplasmic reticulum stress by tauroursodeoxycholic acid delays senescence of mouse ovarian surface epithelium. Cell Tissue Res 2018; 374:643-652. [PMID: 30066106 DOI: 10.1007/s00441-018-2888-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 07/04/2018] [Indexed: 12/16/2022]
Abstract
Ovarian surface epithelium (OSE) forms a single layer of mostly cuboidal cells on surface of mammalian ovaries that is inherently exposed to cell stress evoked by tissue damage every ovulation and declines morphologically after menopause. Endoplasmic reticulum (ER) is a principal cell organelle involved in proteosynthesis, but also integrating various stress signals. ER stress evokes a conserved signaling pathway, the unfolded protein response (UPR), leading to cell death or adaptation to stress conditions. In this work, we document that mouse OSE suffers from ER stress during replicative senescence in vitro, develops abnormalities in ER and initiates UPR. Attenuation of ER stress in senescent OSE by tauroursodeoxycholic acid (TUDCA) reconditions ER architecture and leads to delayed onset of senescence. In summary, we show for the first time a mutual molecular link between ER stress response and replicative senescence leading to phenotypic changes of non-malignant ovarian surface epithelium.
Collapse
|
23
|
Vašíčková K, Horak P, Vaňhara P. TUSC3: functional duality of a cancer gene. Cell Mol Life Sci 2018; 75:849-857. [PMID: 28929175 PMCID: PMC11105401 DOI: 10.1007/s00018-017-2660-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 09/13/2017] [Indexed: 02/06/2023]
Abstract
Two decades ago, following a systematic screening of LOH regions on chromosome 8p22, TUSC3 has been identified as a candidate tumor suppressor gene in ovarian, prostate and pancreatic cancers. Since then, a growing body of evidence documented its clinical importance in various other types of cancers, and first initial insights into its molecular function and phenotypic effects have been gained, though the precise role of TUSC3 in different cancers remains unclear. As a part of the oligosaccharyltransferase complex, TUSC3 localizes to the endoplasmic reticulum and functions in final steps of N-glycosylation of proteins, while its loss evokes the unfolded protein response. We are still trying to figure out how this mechanistic function is reconcilable with its varied effects on cancer promotion. In this review, we focus on cancer-related effects of TUSC3 and envisage a possible role of TUSC3 beyond endoplasmic reticulum.
Collapse
Affiliation(s)
- Kateřina Vašíčková
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 126/3, 625 00, Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital Brno, Pekařská 53, 65691, Brno, Czech Republic
| | - Peter Horak
- Department of Translational Oncology, National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 460, 69120, Heidelberg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Petr Vaňhara
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 126/3, 625 00, Brno, Czech Republic.
- International Clinical Research Center, St. Anne's University Hospital Brno, Pekařská 53, 65691, Brno, Czech Republic.
| |
Collapse
|
24
|
Sheng XR, Xing SG, Wang RD, Chen K, Jia WD. Low levels of tumor suppressor candidate 3 predict poor prognosis of patients with hepatocellular carcinoma. Onco Targets Ther 2018; 11:909-917. [PMID: 29503566 PMCID: PMC5825997 DOI: 10.2147/ott.s153381] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Purpose The tumor suppressor candidate 3 (TUSC3) has been considered to be closely associated with the occurrence, development and invasion of various malignant tumors. However, the expression of TUSC3 in hepatocellular carcinoma (HCC) tissues remains ambiguous. The purpose of this research was to investigate the expression of TUSC3 in HCC tissues and analyze the relationship between TUSC3 levels and clinicopathological characteristics and prognosis of HCC patients. Materials and methods Immunohistochemistry was used to detect the expression of TUSC3 in HCC and the corresponding para-cancerous tissues from 92 samples of HCC patients. mRNA and protein expression levels of TUSC3 were evaluated by quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot assays in 25 paired HCC and corresponding adjacent nontumor tissues. Furthermore, statistical analysis was applied to evaluate the correlation between TUSC3 level and the clinicopathological features and prognosis of HCC patients. Results Immunohistochemical assay indicated that the expression of TUSC3 was significantly lower in HCC tissues when compared with the corresponding para-cancerous tissues (χ2=11.512, P=0.001). The analysis of clinicopathological characteristics showed that low expression of TUSC3 in HCC tissues was significantly associated with Edmondson grade, Barcelona Clinic Liver Cancer stage and tumor size (P=0.008, 0.009 and 0.020, respectively). Univariate analysis showed that the expression of TUSC3 was strongly correlated with overall survival (OS) and disease-free survival (DFS) after radical surgery in HCC patients (P<0.001, P<0.001, respectively). Multivariate analysis revealed that the TUSC3 level was an independent risk factor for OS and DFS in HCC patients (P=0.001, P<0.001, respectively). Results of qRT-PCR and Western blot assays indicated that the level of TUSC3 in HCC tissues was significantly lower than that in the corresponding adjacent noncancerous tissues (P<0.01, P<0.001). Conclusion The expression of TUSC3 in HCC was significantly downregulated and was correlated with tumor progression and prognosis, which could be used as an independent predictor of prognosis in HCC patients.
Collapse
Affiliation(s)
- Xu-Ren Sheng
- Department of Liver Surgery, Affiliated Provincial Hospital, Anhui Medical University.,Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery
| | - Song-Ge Xing
- Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery.,CAS Key Laboratory of Innate Immunity and Chronic Disease, Innovation Center for Cell Signaling Network, School of Life Sciences, University of Science and Technology of China, Hefei, People's Republic of China
| | - Run-Dong Wang
- Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery
| | - Kang Chen
- Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery
| | - Wei-Dong Jia
- Department of Liver Surgery, Affiliated Provincial Hospital, Anhui Medical University.,Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery
| |
Collapse
|
25
|
Abdullahi A, Stanojcic M, Parousis A, Patsouris D, Jeschke MG. Modeling Acute ER Stress in Vivo and in Vitro. Shock 2018; 47:506-513. [PMID: 27755507 DOI: 10.1097/shk.0000000000000759] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The endoplasmic reticulum (ER) is a critical organelle that synthesizes secretory proteins and serves as the main calcium storage site of the cell. The accumulation of unfolded proteins at the ER results in ER stress. Although the association between ER stress and the pathogenesis of many metabolic conditions have been well characterized using both in vivo and in vitro models, no standardized model concerning ER stress exists. Here, we report a standardized model of ER stress using two well-characterized ER stress-inducing agents, thapsigargin and tunicamycin. Our aim in this current study was 2-fold: to characterize and establish which agent is optimal for in vitro use to model acute ER stress and to evaluate which agent is optimal for in vivo use. To study the first aim we used two well-established metabolic cell lines; human hepatocellular carcinoma (HepG2s) and differentiated mouse adipocytes (3T3-L1). In the second aim we utilized C57BL/6J mice that were randomized into three treatment groups of sham, thapsigargin, and tunicamycin. Our in vitro results showed that tunicamycin worked as a rapid and efficacious inducer of ER stress in adipocytes consistently, whereas thapsigargin and tunicamycin were equally effective in inducing ER stress in hepatocytes. In regards to our in vivo results, we saw that tunicamycin was superior in not only inducing ER stress but also recapturing the metabolic alterations associated with ER stress. Thus, our findings will help guide and inform researchers as to which ER stress agent is appropriate with regards to their model.
Collapse
Affiliation(s)
- Abdikarim Abdullahi
- *Biological Sciences, Sunnybrook Research Institute, Toronto, ON, Canada †Ross Tilley Burn Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada ‡Faculty of Medicine, University of Toronto, Toronto, ON, Canada §Department of Surgery, Division of Plastic Surgery and Department of Immunology, University of Toronto, Toronto, ON, Canada
| | | | | | | | | |
Collapse
|
26
|
Tumor suppressor candidate 3: A novel grading tool and predictor of clinical malignancy in human gliomas. Oncol Lett 2018; 15:5655-5661. [PMID: 29556302 PMCID: PMC5844021 DOI: 10.3892/ol.2018.8082] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 11/21/2017] [Indexed: 01/29/2023] Open
Abstract
For several years, the cause of autosomal recessive mental retardation has been attributed to the deletion or mutation of a gene named tumor suppressor candidate 3 (TUSC3). Previous research has identified that TUSC3 is a potential tumor suppressor gene in oral epidermoid carcinoma, lung cancer and esophageal cancer. However, to the best of our knowledge, no previously published data has existed on the expression of TUSC3 in gliomas. The present study focused on the expression of TUSC3 in brain gliomas. Additionally, the present study sought to identify he association between TUSC3 expression and the typical clinical and pathological disease manifestations of gliomas. TUSC3 levels were evaluated using a western blot assay and immunohistochemistry on tissue microarray slides. Results indicated a significant decrease in TUSC3 expression in glioma tissues compared with the normal adjacent tissues. Furthermore, TUSC3 expression and World Health Organization grade demonstrated an inverse association in patients with glioma. This revealed that lower levels of TUSC3 in gliomas may be associated with a poorly-differentiated (high grade) tumor and thus a higher malignancy. Through the combination of the results of the present study and future research projects, TUSC3 may be a novel grading tool that assists with evaluating tumor malignancy and consequently a more active therapeutic regimen may be used in patients with glioma.
Collapse
|
27
|
Vu T, Datta PK. Regulation of EMT in Colorectal Cancer: A Culprit in Metastasis. Cancers (Basel) 2017; 9:cancers9120171. [PMID: 29258163 PMCID: PMC5742819 DOI: 10.3390/cancers9120171] [Citation(s) in RCA: 375] [Impact Index Per Article: 46.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 12/05/2017] [Accepted: 12/05/2017] [Indexed: 12/12/2022] Open
Abstract
Epithelial to mesenchymal transition (EMT) is a process during which cells lose their epithelial characteristics, for instance cell polarity and cell-cell contact, and gain mesenchymal properties, such as increased motility. In colorectal cancer (CRC), EMT is associated with an invasive or metastatic phenotype. In this review, we discuss recent studies exploring novel regulation mechanisms of EMT in CRC, including the identification of new CRC EMT regulators. Upregulation of inducers can promote EMT, leading to increased invasiveness and metastasis in CRC. These inducers can downregulate E-cadherin and upregulate N-cadherin and vimentin (VIM) through modulating EMT-related signaling pathways, for instance WNT/β-catenin and TGF-β, and EMT transcription factors, such as zinc finger E-box binding homeobox 1 (ZEB1) and ZEB2. In addition, several microRNAs (miRNAs), including members of the miR-34 and miR-200 families, are found to target mRNAs of EMT-transcription factors, for example ZEB1, ZEB2, or SNAIL. Downregulation of these miRNAs is associated with distant metastasis and advanced stage tumors. Furthermore, the role of EMT in circulating tumor cells (CTCs) is also discussed. Mesenchymal markers on the surface of EMT CTCs were found to be associated with metastasis and could serve as potential biomarkers for metastasis. Altogether, these studies indicate that EMT is orchestrated by a complicated network, involving regulators of different signaling pathways. Further studies are required to understand the mechanisms underlying EMT in CRC.
Collapse
Affiliation(s)
- Trung Vu
- Division of Hematology and Oncology, Department of Medicine, Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35233, USA.
| | - Pran K Datta
- Division of Hematology and Oncology, Department of Medicine, Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35233, USA.
- Birmingham Veterans Affairs Medical Center, Birmingham, AL 35233, USA.
| |
Collapse
|
28
|
Liang G, Fang X, Yang Y, Song Y. Knockdown of CEMIP suppresses proliferation and induces apoptosis in colorectal cancer cells: downregulation of GRP78 and attenuation of unfolded protein response. Biochem Cell Biol 2017; 96:332-341. [PMID: 29024602 DOI: 10.1139/bcb-2017-0151] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
It has been suggested that cell migration inducing hyaluronan binding protein (CEMIP) contributes to the carcinogenesis of colorectal cancer (CRC). Cancer cells can adapt to endoplasmic reticulum (ER) stress by initiating an unfolded protein response (UPR). This study aimed to investigate whether CEMIP affects the UPR of CRC cells, with a focus on 78 kDa glucose-regulated protein (GRP78, a major ER chaperone). We found that knockdown of CEMIP inhibited cell proliferation and induced a G1 arrest in SW480 CRC cells. The levels of cyclin D1 and cyclin E1 and phospho-retinoblastoma, which are known to promote the cell cycle progression from G0 or G1 into S phase, were decreased in CEMIP-silenced cells. CEMIP shRNA induced apoptosis and inhibited GRP78 expression in SW480 and Colo205 cells. The basal UPR of cancer cells was attenuated by CEMIP shRNA, as evidenced by the decreased expression of UPR sensors, protein kinase R-like endoplasmic reticulum kinase (PERK), inositol requiring enzyme 1 (IRE1), and activating transcription factor 6 (ATF6). Furthermore, CEMIP silencing sensitized CRC cells to thapsigargin-induced apoptosis. Our study demonstrates that the in-vitro anti-proliferative and pro-apoptotic effects in CRC cells that were induced by silencing CEMIP may be associated with GRP78 repression and UPR attenuation.
Collapse
Affiliation(s)
- Guodong Liang
- a Department of Gastrointestinal Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, P.R. China
| | - Xuedong Fang
- a Department of Gastrointestinal Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, P.R. China
| | - Yubo Yang
- b Department of Gastrointestinal Surgery, Jilin Provincial Cancer Hospital, Changchun 130012, P.R. China
| | - Yan Song
- a Department of Gastrointestinal Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, P.R. China
| |
Collapse
|
29
|
Peng Y, Cao J, Yao XY, Wang JX, Zhong MZ, Gan PP, Li JH. TUSC3 induces autophagy in human non-small cell lung cancer cells through Wnt/β-catenin signaling. Oncotarget 2017; 8:52960-52974. [PMID: 28881786 PMCID: PMC5581085 DOI: 10.18632/oncotarget.17674] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 03/19/2017] [Indexed: 02/06/2023] Open
Abstract
We investigated the effects of tumor suppressor candidate 3 (TUSC3) on autophagy in human non-small cell lung cancer (NSCLC) cells. A total of 118 NSCLC patients (88 males and 30 females) who underwent surgery at our institute were enrolled in the study. Immunohistochemical analysis revealed that TUSC3 protein expression was lower in NSCLC specimens than adjacent normal tissue. Correspondingly, there was greater methylation of TUSC3 in NSCLC than adjacent normal tissue. After transient transfection of A549 NSCLC cells with constructs designed to up-regulate or down-regulate TUSC3 expression, we analyzed the effects of inhibiting the Wnt pathway (XAV939) and autophagy (chloroquine, CQ) on the behavior of NSCLC cells. We also performed TOP/FOP-Flash reporter assays, MTT assays, Annexin V-FITC/propidium iodide staining, and acridine orange staining to evaluate Wnt/β-catenin signaling, cell proliferation, apoptosis, and autophagy, respectively. Expression of Wnt/β-catenin pathway components and autophagy-related proteins was analyzed using qRT-PCR and Western blotting. We found that TUSC3 inhibited cell proliferation and promoted both apoptosis and autophagy in A549 cells. In addition, TUSC3 increased expression of autophagy-related proteins. It also increased expression of Wnt/β-catenin signaling pathway components and promoted nuclear transfer of β-catenin, resulting in activation of Wnt/β-catenin signaling. TUSC3 thus induces autophagy in human NSCLC cells through activation of the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Yun Peng
- 1 International Medical Center, Xiangya Hospital, Central South University, Changsha 410008, P.R. China
| | - Jun Cao
- 2 Department of Medical Oncology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410008, P.R. China
| | - Xiao-Yi Yao
- 3 Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, P.R. China
| | - Jian-Xin Wang
- 4 School of Information Science and Engineering, Central South University, Changsha 410008, P.R. China
| | - Mei-Zuo Zhong
- 3 Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, P.R. China
| | - Ping-Ping Gan
- 3 Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, P.R. China
| | - Jian-Huang Li
- 3 Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, P.R. China
| |
Collapse
|
30
|
Vitamin D receptor-binding site variants affect prostate cancer progression. Oncotarget 2017; 8:74119-74128. [PMID: 29088772 PMCID: PMC5650327 DOI: 10.18632/oncotarget.18271] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 05/15/2017] [Indexed: 11/25/2022] Open
Abstract
Vitamin D is an important modulator of cellular proliferation through the vitamin D receptor (VDR) that binds to DNA in the regulatory sequences of target genes. We hypothesized that single nucleotide polymorphisms (SNPs) in VDR-binding sites might affect target gene expression and influence the progression of prostate cancer. Using a genome-wide prediction database, 62 SNPs in VDR-binding sites were selected for genotyping in 515 prostate cancer patients and the findings were replicated in an independent cohort of 411 patients. Prognostic significance on prostate cancer progression was assessed by Kaplan-Meier analysis and the Cox regression model. According to multivariate analyses adjusted for known predictors, HFE rs9393682 was found to be associated with disease progression for localized prostate cancer, and TUSC3 rs1378033 was associated with progression for advanced prostate cancer in both cohorts. Vitamin D treatment inhibited HFE mRNA expression, and down-regulation of HFE by transfecting small interfering RNA suppressed PC-3 human prostate cancer cell proliferation and wound healing ability. In contrast, vitamin D treatment induced TUSC3 expression, and silencing TUSC3 promoted prostate cancer cell growth and migration. Further analysis of an independent microarray dataset confirmed that low TUSC3 expression correlated with poor patient prognosis. Our results warrant further studies using larger cohorts. This study identifies common variants in VDR-binding sites as prognostic markers of prostate cancer progression and HFE and TUSC3 as plausible susceptibility genes.
Collapse
|
31
|
Gu Y, Pei X, Ren Y, Cai K, Guo K, Chen J, Qin W, Lin M, Wang Q, Tang N, Cheng Z, Ding Y, Lin J. Oncogenic function of TUSC3 in non-small cell lung cancer is associated with Hedgehog signalling pathway. Biochim Biophys Acta Mol Basis Dis 2017; 1863:1749-1760. [PMID: 28487226 DOI: 10.1016/j.bbadis.2017.05.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Revised: 05/03/2017] [Accepted: 05/05/2017] [Indexed: 12/14/2022]
Abstract
Non-small cell lung cancer (NSCLC) represents 75-80% of all lung carcinomas, which is the most common cause of death from cancer. Tumour suppressor candidate 3 (TUSC3) is pivotal in many biochemical functions and cytological processes. Dis-regulation of TUSC3 is frequently observed in epithelial cancers. In this study, we observed up-regulated TUSC3 expression at the mRNA and protein levels in clinical NSCLC samples compared with adjacent non-tumorous lung tissues. The expression level of TUSC3 is significantly correlated with tumour metastasis and patient survival. Overexpression of TUSC3 in NSCLC cells led to increased proliferation, migration, and invasion in vitro and accelerated xenograft tumour growth in vivo, while the opposite effects were achieved in TUSC3-silenced cells. Increased GLI1, SMO, PTCH1, and PTCH2 abundance were observed in TUSC3 overexpressed cells using western blotting. Co-immunoprecipitation and immunofluorescence analyses further revealed interaction between TUSC3 and GLI1. In conclusion, our study demonstrated an oncogenic role of TUSC3 in NSCLC and showed that dis-regulation of TUSC3 may affect tumour cell invasion and migration through possible involvement in the Hedgehog (Hh) signalling pathway.
Collapse
Affiliation(s)
- Ye Gu
- Department of Pathology, Nanfang Hospital & School of Basic Medicine, Southern Medical University, 1838 Guangzhou Avenue, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, 1838 Guangzhou Avenue, Guangzhou 510515, PR China
| | - Xiaojuan Pei
- Department of Pathology, Nanfang Hospital & School of Basic Medicine, Southern Medical University, 1838 Guangzhou Avenue, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, 1838 Guangzhou Avenue, Guangzhou 510515, PR China; Department of Pathology, Huizhou Central People's Hospital, Huizhou, Guangdong 516001, PR China
| | - Yansong Ren
- Department of Pathology, Nanfang Hospital & School of Basic Medicine, Southern Medical University, 1838 Guangzhou Avenue, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, 1838 Guangzhou Avenue, Guangzhou 510515, PR China
| | - Kaican Cai
- Department of Thoracic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China
| | - Kang Guo
- Department of Pathology, Nanfang Hospital & School of Basic Medicine, Southern Medical University, 1838 Guangzhou Avenue, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, 1838 Guangzhou Avenue, Guangzhou 510515, PR China
| | - Jiaye Chen
- Department of Pathology, Nanfang Hospital & School of Basic Medicine, Southern Medical University, 1838 Guangzhou Avenue, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, 1838 Guangzhou Avenue, Guangzhou 510515, PR China
| | - Weizhao Qin
- Department of Pathology, Nanfang Hospital & School of Basic Medicine, Southern Medical University, 1838 Guangzhou Avenue, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, 1838 Guangzhou Avenue, Guangzhou 510515, PR China
| | - Mingdao Lin
- Department of Pathology, Nanfang Hospital & School of Basic Medicine, Southern Medical University, 1838 Guangzhou Avenue, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, 1838 Guangzhou Avenue, Guangzhou 510515, PR China
| | - Qian Wang
- Department of Pathology, Nanfang Hospital & School of Basic Medicine, Southern Medical University, 1838 Guangzhou Avenue, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, 1838 Guangzhou Avenue, Guangzhou 510515, PR China
| | - Na Tang
- Department of Pathology, Shenzhen People's Hospital, Shenzhen 510820, PR China
| | - Zhiqiang Cheng
- Department of Pathology, Shenzhen People's Hospital, Shenzhen 510820, PR China
| | - Yanqing Ding
- Department of Pathology, Nanfang Hospital & School of Basic Medicine, Southern Medical University, 1838 Guangzhou Avenue, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, 1838 Guangzhou Avenue, Guangzhou 510515, PR China
| | - Jie Lin
- Department of Pathology, Nanfang Hospital & School of Basic Medicine, Southern Medical University, 1838 Guangzhou Avenue, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, 1838 Guangzhou Avenue, Guangzhou 510515, PR China.
| |
Collapse
|
32
|
Liang Q, Wang K, Wang B, Cai Q. HCMV-encoded miR-UL112-3p promotes glioblastoma progression via tumour suppressor candidate 3. Sci Rep 2017; 7:44705. [PMID: 28303930 PMCID: PMC5356197 DOI: 10.1038/srep44705] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 02/13/2017] [Indexed: 01/19/2023] Open
Abstract
Glioblastoma (GBM) is the most prevalent and lethal type of primary malignant brain tumour. Recent studies suggest that the discovery of human cytomegalovirus (HCMV)-encoded microRNAs (miRNAs) might play a role in the pathogenesis of diseases, including GBM. In this study, we aimed to analyse the expression and function of HCMV-encoded miRNAs in GBM. We found that miR-UL112-3p expression was significantly elevated in GBM, and its expression levels were highly associated with glioma size, differentiation, WHO stage and the overall and disease-free survival of patients. The overexpression of miR-UL112-3p in the GBM cells promoted cell proliferation, clone formation, migration and invasion. In contrast, the down-regulation of miR-UL112-3p exerted an inverse effects. Tumour suppressor candidate 3 (TUSC3), a potential target gene of miR-UL112-3p, was inversely correlated with miR-UL112-3p expression in GBM tissues and cell lines. Furthermore, we demonstrated that TUSC3 was directly regulated by miR-UL112-3p, and the ectopic expression of TUSC3 reversed the effects of miR-UL112-3p on GBM progression via the AKT signalling pathway. Taken together, these findings collectively demonstrate that miR-UL112-3p exerts its oncogene function by directly targeting TUSC3 in GBM, indicating a potential novel therapeutic target for GBM.
Collapse
Affiliation(s)
- Qing Liang
- MOE&MOH Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Kejia Wang
- Department of Special Medicine, Qingdao University, Qingdao 266071, Shandong, China
| | - Bin Wang
- Department of Special Medicine, Qingdao University, Qingdao 266071, Shandong, China
| | - Qiliang Cai
- MOE&MOH Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| |
Collapse
|
33
|
Liu K, Xie F, Gao A, Zhang R, Zhang L, Xiao Z, Hu Q, Huang W, Huang Q, Lin B, Zhu J, Wang H, Que J, Lan X. SOX2 regulates multiple malignant processes of breast cancer development through the SOX2/miR-181a-5p, miR-30e-5p/TUSC3 axis. Mol Cancer 2017; 16:62. [PMID: 28288641 PMCID: PMC5348847 DOI: 10.1186/s12943-017-0632-9] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 03/06/2017] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND High levels of SOX2 protein are correlated with increased dissemination of breast cancer. However, the underlying molecular mechanisms are not fully understood. METHODS In this study we investigate the role of SOX2 in breast cancer metastasis using multiple in vitro and in vivo assays including cell culture, shRNA-mediated knockdown, wound healing, colony formation, transwell chamber, xenograft and tail vein injection. Moreover, western blot, immunostaining, microarray and real-time PCR were used to determine the change of protein and miRNA levels. Luciferase assays were also used to evaluate activity which TUSC3 is a target of miR-181a-5p and miR-30e-5p, and the clinical survival relevance was analyzed by Kaplan-Meier analysis. RESULTS We identified a novel pathway involving SOX2 regulation of microRNAs to control the proliferation and migration of breast cancer cells. shRNA-mediated knockdown of SOX2 inhibits breast cancer cell expansion and migration. More importantly, we found that these changes are accompanied by significant reduction in the levels of two microRNAs, miR-181a-5p and miR-30e-5p. Overexpression of these two microRNAs leads to reduced protein levels of Tumor Suppressor Candidate 3 (TUSC3) in breast cancer cells; mutations of the potential binding sites in the 3'-UTR of TUSC3 abrogate the inhibitory effects of the microRNAs. We further found that upregulation of TUSC3 expression leads to reduced proliferation and migration of breast cancer cells. In human breast cancer samples the levels of TUSC3 protein are inversely correlated with those of SOX2 protein. CONCLUSIONS Taken together, our work reveals a novel SOX2-mediated regulatory axis that plays critical roles in the proliferation, migration and invasiveness of breast cancer cells. Targeting this axis may provide beneficial effect in the treatment of breast cancer.
Collapse
Affiliation(s)
- Kuancan Liu
- Institute for Laboratory Medicine, Fuzhou General Hospital, PLA, Fuzhou, 350025, Fujian, People's Republic of China. .,Department of Medicine, Columbia University Medical Center, New York, 10032, NY, USA. .,Dong fang Hospital, Xiamen University, Fuzhou, 350025, Fujian, People's Republic of China. .,Fuzhou General Hospital Clinical Medical School, Fujian Medical University, Fuzhou, 350025, People's Republic of China.
| | - Fuan Xie
- Institute for Laboratory Medicine, Fuzhou General Hospital, PLA, Fuzhou, 350025, Fujian, People's Republic of China.,Dong fang Hospital, Xiamen University, Fuzhou, 350025, Fujian, People's Republic of China
| | - Anding Gao
- Institute for Laboratory Medicine, Fuzhou General Hospital, PLA, Fuzhou, 350025, Fujian, People's Republic of China.,Dong fang Hospital, Xiamen University, Fuzhou, 350025, Fujian, People's Republic of China
| | - Rui Zhang
- Institute for Laboratory Medicine, Fuzhou General Hospital, PLA, Fuzhou, 350025, Fujian, People's Republic of China.,Fuzhou General Hospital Clinical Medical School, Fujian Medical University, Fuzhou, 350025, People's Republic of China
| | - Long Zhang
- Life Science Institute, Zhejiang University, Hangzhou, 310058, Zhejiang, People's Republic of China
| | - Zhangwu Xiao
- Emergency Department of the 476 Hospital, Fuzhou General Hospital, PLA, Fuzhou, 350002, Fujian, People's Republic of China
| | - Qiong Hu
- Dong fang Hospital, Xiamen University, Fuzhou, 350025, Fujian, People's Republic of China
| | - Weifeng Huang
- Medical College, China Three Gorges University, Yichang, 443002, Hubei, People's Republic of China
| | - Qiaojia Huang
- Institute for Laboratory Medicine, Fuzhou General Hospital, PLA, Fuzhou, 350025, Fujian, People's Republic of China
| | - Baoshun Lin
- Institute for Laboratory Medicine, Fuzhou General Hospital, PLA, Fuzhou, 350025, Fujian, People's Republic of China.,Dong fang Hospital, Xiamen University, Fuzhou, 350025, Fujian, People's Republic of China
| | - Jian Zhu
- Department of Microbiology and Immunology, University of Rochester, Rochester, 14642, NY, USA
| | - Haikun Wang
- Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, 200031, People's Republic of China
| | - Jianwen Que
- Department of Medicine, Columbia University Medical Center, New York, 10032, NY, USA.
| | - Xiaopeng Lan
- Institute for Laboratory Medicine, Fuzhou General Hospital, PLA, Fuzhou, 350025, Fujian, People's Republic of China. .,Fuzhou General Hospital Clinical Medical School, Fujian Medical University, Fuzhou, 350025, People's Republic of China.
| |
Collapse
|
34
|
Yu X, Zhai C, Fan Y, Zhang J, Liang N, Liu F, Cao L, Wang J, Du J. TUSC3: a novel tumour suppressor gene and its functional implications. J Cell Mol Med 2017; 21:1711-1718. [PMID: 28272772 PMCID: PMC5571513 DOI: 10.1111/jcmm.13128] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 01/13/2017] [Indexed: 12/31/2022] Open
Abstract
The tumour suppressor candidate 3 (TUSC3) gene is located on chromosome region 8p22 and encodes the 34 kD TUSC3 protein, which is a subunit of the oligosaccharyl transferase responsible for the N‐glycosylation of nascent proteins. Known to be related to autosomal recessive mental retardation for several years, TUSC3 has only recently been identified as a potential tumour suppressor gene. Based on the structure and function of TUSC3, specific mechanisms in various diseases have been investigated. Several studies have demonstrated that TUSC3 is an Mg2+‐transporter involved in magnesium transport and homeostasis, which is important for learning and memory, embryonic development and testis maturation. Moreover, dysfunction or deletion of TUSC3 exerts its oncological effects as a modulator by inhibiting glycosylation efficiency and consequently inducing endoplasmic reticulum stress and malignant cell transformation. In this study, we summarize the advances in the studies of TUSC3 and comment on the potential roles of TUSC3 in diagnosis and treatment of TUSC3‐related diseases, especially cancer.
Collapse
Affiliation(s)
- Xinshuang Yu
- Department of Radiation Oncology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Chunjuan Zhai
- Department of Cardiology, Shandong Provincial Hospital affiliated to Shandong University, Shandong University, Jinan, China
| | - Yujun Fan
- Medical Management Service Center of Shandong Provincial Health and Family Planning Commission, Jinan, China
| | - Jiandong Zhang
- Department of Radiation Oncology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Ning Liang
- Department of Radiation Oncology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Fengjun Liu
- Department of Radiation Oncology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Lili Cao
- Medical Research Center, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Jia Wang
- China Institute of Veterinary Drugs Control, Beijing, China
| | - Juan Du
- Department of Radiation Oncology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China.,Medical Research Center, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| |
Collapse
|
35
|
Munkley J. Glycosylation is a global target for androgen control in prostate cancer cells. Endocr Relat Cancer 2017; 24:R49-R64. [PMID: 28159857 DOI: 10.1530/erc-16-0569] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 02/03/2017] [Indexed: 12/17/2022]
Abstract
Changes in glycan composition are common in cancer and can play important roles in all of the recognised hallmarks of cancer. We recently identified glycosylation as a global target for androgen control in prostate cancer cells and further defined a set of 8 glycosylation enzymes (GALNT7, ST6GalNAc1, GCNT1, UAP1, PGM3, CSGALNACT1, ST6GAL1 and EDEM3), which are also significantly upregulated in prostate cancer tissue. These 8 enzymes are under direct control of the androgen receptor (AR) and are linked to the synthesis of important cancer-associated glycans such as sialyl-Tn (sTn), sialyl LewisX (SLeX), O-GlcNAc and chondroitin sulfate. Glycosylation has a key role in many important biological processes in cancer including cell adhesion, migration, interactions with the cell matrix, immune surveillance, cell signalling and cellular metabolism. Our results suggest that alterations in patterns of glycosylation via androgen control might modify some or all of these processes in prostate cancer. The prostate is an abundant secretor of glycoproteins of all types, and alterations in glycans are, therefore, attractive as potential biomarkers and therapeutic targets. Emerging data on these often overlooked glycan modifications have the potential to improve risk stratification and therapeutic strategies in patients with prostate cancer.
Collapse
Affiliation(s)
- Jennifer Munkley
- Institute of Genetic MedicineNewcastle University, Newcastle-upon-Tyne, UK
| |
Collapse
|
36
|
Li YG, Liang NX, Qin YZ, Ma DJ, Huang CJ, Liu L, Li SQ. Effects of RNAi-mediated TUSC3 silencing on radiation-induced autophagy and radiation sensitivity of human lung adenocarcinoma cell line A549 under hypoxic condition. Tumour Biol 2016; 37:16357–16365. [PMID: 27900564 DOI: 10.1007/s13277-016-5458-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2016] [Accepted: 09/23/2016] [Indexed: 12/13/2022] Open
Abstract
This study examined the effects of RNAi-mediated TUSC3 silencing on radiation-induced autophagy and radiation sensitivity of human lung adenocarcinoma cell line A549 under hypoxic condition. Different CoCl2 concentrations were used to treat A549 cells and establish a CoCl2-induced hypoxic model of A549 cells. MTT and clone formation assays were used to determine the effects of different concentrations of CoCl2 on the growth and proliferation of A549 cells treated by different doses of X-ray irradiation. The siRNA-expressing vector was transfected by liposomes and for silencing of TUSC3. Flow cytometry was used to measure cell cycle changes and apoptosis rate. Real-time quantitative polymerase chain reaction (qRT-PCR) assay was performed to detect the expression of TUSC3 mRNA. Western blotting was applied to detect the changes of TUSC3, LC3, and p62 proteins under different CoCl2 concentrations and after siRNA silencing of TUSC3. The TUSC3 levels in A549 cells increased under hypoxic conditions in a dose-dependent manner (P < 0.05). Hypoxia inhibited the growth and proliferation of A549 cells and promoted apoptosis (P < 0.05). With an increasing dose of X-ray irradiation, A549 cells showed significantly increased growth and proliferation and decreased apoptosis (P < 0.05). After siRNA-TUSC3 was transfected by liposome, the TUSC3 level was substantially inhibited (P < 0.05). Silencing TUSC3 inhibited A549 cell growth and proliferation after radiotherapy under hypoxic condition, promoted apoptosis, increased G0/G1 phase cells, and reduced S phase cells (all P < 0.05). Hypoxia and radiation along with different CoCl2 concentrations could induce cell autophagy, which increased with concentration and dose, while silencing the TUSC3 gene inhibited autophagy (all P < 0.05). RNAi silencing of TUSC3 inhibited growth and proliferation, while enhanced apoptosis and radiation sensitivity of hypoxic A549 lung adenocarcinoma cells.
Collapse
Affiliation(s)
- Ya-Guang Li
- Department of Thoracic Surgery, Peking Union Medical College Hospital, No. 1 Shuaifuyuan, Wangfujing Street, Dongcheng District, Beijing, 100730, People's Republic of China
| | - Nai-Xin Liang
- Department of Thoracic Surgery, Peking Union Medical College Hospital, No. 1 Shuaifuyuan, Wangfujing Street, Dongcheng District, Beijing, 100730, People's Republic of China
| | - Ying-Zhi Qin
- Department of Thoracic Surgery, Peking Union Medical College Hospital, No. 1 Shuaifuyuan, Wangfujing Street, Dongcheng District, Beijing, 100730, People's Republic of China
| | - Dong-Jie Ma
- Department of Thoracic Surgery, Peking Union Medical College Hospital, No. 1 Shuaifuyuan, Wangfujing Street, Dongcheng District, Beijing, 100730, People's Republic of China
| | - Chang-Jin Huang
- Plastic Surgery Hospital, CAMS, PUMC, Beijing, 100144, People's Republic of China
| | - Lei Liu
- Department of Thoracic Surgery, Peking Union Medical College Hospital, No. 1 Shuaifuyuan, Wangfujing Street, Dongcheng District, Beijing, 100730, People's Republic of China
| | - Shan-Qing Li
- Department of Thoracic Surgery, Peking Union Medical College Hospital, No. 1 Shuaifuyuan, Wangfujing Street, Dongcheng District, Beijing, 100730, People's Republic of China.
| |
Collapse
|
37
|
Yu X, Zhang J, Zhong H, Liu F, Liang N, Wang Y, Meng X, Du J. Decreased Tumor Suppressor Candidate 3 Predicts Poor Prognosis of Patients with Esophageal Squamous Cell Carcinoma. Int J Med Sci 2016; 13:963-969. [PMID: 27994502 PMCID: PMC5165690 DOI: 10.7150/ijms.16381] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Accepted: 10/07/2016] [Indexed: 12/01/2022] Open
Abstract
TUSC3 was recently identified as a potential tumor suppressor gene in a variety of human malignancies. However, no data are currently available regarding the expressions of TUSC3 in esophageal cancer (ESCC).The purposes of this study was to investigated the expressions of TUSC3 in ESCC tissues and assess the relationship between TUSC3 levels and clinico-pathological characteristics of ESCC patients. TUSC3 protein expressions were evaluated by immunohistochemistry (IHC) on tissue microarray slides in esophageal cancer, which included 95 esophageal squamous carcinoma specimens (ESCC), and 75 normal esophageal mucosa (NEM). We found that TUSC3 in ESCC was significant lower than that in NEM (P=0.000). According to multi-clinical classifications, TUSC3 level varied significantly with TNM stage, T stage, and N stage (p<0.001, p=0.0368, p<0.0001, respectively). Univariate analysis showed that gender, TNM stage, T stage, N stage, TUSC3 expression were prognostic factors for survival. Multivariate analysis showed that in our study, only TUSC3 expression was independent prognostic factors for ESCC. Our results indicated for the first time, a combined analysis of TUSC3 expressions as well as the clinical variables will help predict the prognosis of ESCC patients. Further large-sample validation and functional analysis should be performed to evaluate its potential prognostic and therapeutic values for ESCC patients.
Collapse
Affiliation(s)
- Xinshuang Yu
- Department of Radiation Oncology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250014, P. R.China
| | - Jiandong Zhang
- Department of Radiation Oncology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250014, P. R.China
| | - Hua Zhong
- Department of Traditional Chinese Medicine, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, 250014, P. R.China
- Department of Oncology, Shandong University of Traditional Chinese Medicine Jinan, 250355, P. R.China
| | - Fengjun Liu
- Department of Radiation Oncology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250014, P. R.China
| | - Ning Liang
- Department of Radiation Oncology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250014, P. R.China
| | - Yao Wang
- Medical Research Center, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, 250014, P. R.China
| | - Xiangjiao Meng
- Department of Radiation Oncology, Shandong Cancer Hospital, Jinan, 250117, P. R.China
| | - Juan Du
- Department of Radiation Oncology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250014, P. R.China
- Medical Research Center, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, 250014, P. R.China
| |
Collapse
|
38
|
Yu X, Zhang K, Liu F, Zhang J, Zhai C, Cao L, Song X, Wang Y, Li B, Sun H, Du J. Tumor suppressor candidate 3 as a novel predictor for lymph node metastasis in lung cancer patients. Oncol Lett 2016; 12:5099-5105. [PMID: 28105217 PMCID: PMC5228539 DOI: 10.3892/ol.2016.5333] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 10/05/2016] [Indexed: 11/06/2022] Open
Abstract
Tumor suppressor candidate 3 (TUSC3) was recently identified as a potential tumor suppressor gene in several cancer types. However, no data are currently available regarding the expression of TUSC3 in lung cancer. The present study investigated the expression of TUSC3 in patients with lung cancer and determined its association with the clinicopathological parameters of the disease. Cytoplasmic TUSC3 expression was evaluated by immunohistochemistry on tissue microarray slides, which included 35 small cell lung cancer (SCLC) specimens, 80 squamous cell lung cancer specimens (SCC), 80 adenocarcinoma lung cancer (ADC) specimens and 37 normal lung tissue specimens. Analysis showed significantly reduced TUSC3 expression in the SCLC patients, but not in the ADC and SCC patients, as compared with the normal controls. Additionally, TUSC3 expression in the patients with a degree of differentiation of 1–2 (well-moderately differentiated) was significantly higher than that in patients with a differentiation degree of 3–4 (poorly differentiated-undifferentiated). Further analysis showed that TUSC3 expression levels were negatively correlated with the degree of differentiation in the ADC and SCC patients. Notably, a marked decrease in TUSC3 expression was identified in the patients who were lymph node metastasis-positive (LNM+) compared with patients who were LNM−. Further analysis showed that significant differences in TUSC3 expression were identified among the different N stages (LNM status) in the SCLC, ADC and SCC patients. Correlation analysis also identified a negative correlation between TUSC3 expression and LNM in all three pathological types of lung cancer tested. Overall, these results indicated that a reduction in TUSC3 may be associated with a poorly-differentiated grade of lung cancer. Importantly, TUSC3 expression may be a useful predictor of LNM in lung cancer patients. A combined analysis of TUSC3 expression and the clinical variables will aid in predicting the incidence of LNM.
Collapse
Affiliation(s)
- Xinshuang Yu
- Department of Radiation Oncology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Kaixian Zhang
- Department of Radiation Oncology, Tengzhou Central People Hospital, Tengzhou, Shandong 277500, P.R. China
| | - Fengjun Liu
- Department of Radiation Oncology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Jiandong Zhang
- Department of Radiation Oncology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Chunjuan Zhai
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Lili Cao
- Medical Research Center, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250014, P.R. China
| | - Xingye Song
- Health Center of the Third Sanatorium, Jinan Military Area Command, Jinan, Shandong 250002, P.R. China
| | - Yao Wang
- Medical Research Center, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250014, P.R. China
| | - Baosheng Li
- Sixth Department of Radiation Oncology, Shandong Cancer Hospital, Jinan, Shandong 250117, P.R. China
| | - Hongjun Sun
- Medical Research Center, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250014, P.R. China
| | - Juan Du
- Department of Radiation Oncology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China; Medical Research Center, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250014, P.R. China
| |
Collapse
|
39
|
Storm M, Sheng X, Arnoldussen YJ, Saatcioglu F. Prostate cancer and the unfolded protein response. Oncotarget 2016; 7:54051-54066. [PMID: 27303918 PMCID: PMC5288241 DOI: 10.18632/oncotarget.9912] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 05/23/2016] [Indexed: 01/01/2023] Open
Abstract
The endoplasmic reticulum (ER) is an essential organelle that contributes to several key cellular functions, including lipogenesis, gluconeogenesis, calcium storage, and organelle biogenesis. The ER also serves as the major site for protein folding and trafficking, especially in specialized secretory cells. Accumulation of misfolded proteins and failure of ER adaptive capacity activates the unfolded protein response (UPR) which has been implicated in several chronic diseases, including cancer. A number of recent studies have implicated UPR in prostate cancer (PCa) and greatly expanded our understanding of this key stress signaling pathway and its regulation in PCa. Here we summarize these developments and discuss their potential therapeutic implications.
Collapse
Affiliation(s)
| | - Xia Sheng
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Yke Jildouw Arnoldussen
- Department of Biological and Chemical Work Environment, National Institute of Occupational Health, Oslo, Norway
| | - Fahri Saatcioglu
- Department of Biosciences, University of Oslo, Oslo, Norway
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
40
|
Jiang Z, Guo M, Zhang X, Yao L, Shen J, Ma G, Liu L, Zhao L, Xie C, Liang H, Wang H, Zhu M, Hu L, Song Y, Shen H, Lin Z. TUSC3 suppresses glioblastoma development by inhibiting Akt signaling. Tumour Biol 2016; 37:12039-12047. [PMID: 27177902 DOI: 10.1007/s13277-016-5072-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 05/05/2016] [Indexed: 11/29/2022] Open
Abstract
Glioblastoma multiform is one of the most common and most aggressive brain tumors in humans. The molecular and cellular mechanisms responsible for the onset and progression of GBM are elusive and controversial. The function of tumor suppressor candidate 3 (TUSC3) has not been previously characterized in GBM. TUSC3 was originally identified as part of an enzyme complex involved in N-glycosylation of proteins, but was recently implicated as a potential tumor suppressor gene in a variety of cancer types. In this study, we demonstrated that the expression levels of TUSC3 were downregulated in both GBM tissues and cells, and also found that overexpression of TUSC3 inhibits GBM cell proliferation and invasion. In addition, the effects of increased levels of methylation on the TUSC3 promoter were responsible for decreased expression of TUSC3 in GBM. Finally, we determined that TUSC3 regulates proliferation and invasion of GBM cells by inhibiting the activity of the Akt signaling pathway.
Collapse
Affiliation(s)
- Zhenfeng Jiang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Harbin, Heilongjiang Province, 150001, China
| | - Mian Guo
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiangtong Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Harbin, Heilongjiang Province, 150001, China
| | - Lifen Yao
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jia Shen
- Orthopaedic Hospital Research Center, University of California, Los Angeles, CA, USA
| | - Guizhen Ma
- Department of Operating Rooms, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Li Liu
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Harbin, Heilongjiang Province, 150001, China
| | - Liwei Zhao
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Harbin, Heilongjiang Province, 150001, China
| | - Chuncheng Xie
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Harbin, Heilongjiang Province, 150001, China
| | - Hongsheng Liang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Harbin, Heilongjiang Province, 150001, China
| | - Haiyang Wang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Harbin, Heilongjiang Province, 150001, China
| | - Minwei Zhu
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Harbin, Heilongjiang Province, 150001, China
| | - Li Hu
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Harbin, Heilongjiang Province, 150001, China
| | - Yuanyuan Song
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Harbin, Heilongjiang Province, 150001, China
| | - Hong Shen
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Harbin, Heilongjiang Province, 150001, China
| | - Zhiguo Lin
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Harbin, Heilongjiang Province, 150001, China.
| |
Collapse
|
41
|
He DX, Gu F, Gao F, Hao JJ, Gong D, Gu XT, Mao AQ, Jin J, Fu L, Ma X. Genome-wide profiles of methylation, microRNAs, and gene expression in chemoresistant breast cancer. Sci Rep 2016; 6:24706. [PMID: 27094684 PMCID: PMC4837395 DOI: 10.1038/srep24706] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 03/30/2016] [Indexed: 12/17/2022] Open
Abstract
Cancer chemoresistance is regulated by complex genetic and epigenetic networks. In this study, the features of gene expression, methylation, and microRNA (miRNA) expression were investigated with high-throughput sequencing in human breast cancer MCF-7 cells resistant to adriamycin (MCF-7/ADM) and paclitaxel (MCF-7/PTX). We found that: ① both of the chemoresistant cell lines had similar, massive changes in gene expression, methylation, and miRNA expression versus chemosensitive controls. ② Pairwise integration of the data highlighted sets of genes that were regulated by either methylation or miRNAs, and sets of miRNAs whose expression was controlled by DNA methylation in chemoresistant cells. ③ By combining the three sets of high-throughput data, we obtained a list of genes whose expression was regulated by both methylation and miRNAs in chemoresistant cells; ④ Expression of these genes was then validated in clinical breast cancer samples to generate a 17-gene signature that showed good predictive and prognostic power in triple-negative breast cancer patients receiving anthracycline-taxane-based neoadjuvant chemotherapy. In conclusion, our results have generated a new workflow for the integrated analysis of the effects of miRNAs and methylation on gene expression during the development of chemoresistance.
Collapse
Affiliation(s)
- Dong-Xu He
- National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi 214122, China
| | - Feng Gu
- Department of Breast Cancer Pathology and Research Laboratory, State Key Laboratory of Breast Cancer Research, Cancer Institute and Hospital, Tianjin Medical University, Tianjin 300060, PR China
| | - Fei Gao
- Agricultural Genomes Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518120, China
| | - Jun-jun Hao
- State Key Lab of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
| | - Desheng Gong
- Agricultural Genomes Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518120, China
| | - Xiao-Ting Gu
- Department of Cellular and Molecular Pharmacology, School of Pharmaceutical Sciences, Jiangnan University, Wuxi 214122, China
| | - Ai-Qin Mao
- Department of Cellular and Molecular Pharmacology, School of Pharmaceutical Sciences, Jiangnan University, Wuxi 214122, China
| | - Jian Jin
- Department of Cellular and Molecular Pharmacology, School of Pharmaceutical Sciences, Jiangnan University, Wuxi 214122, China
| | - Li Fu
- Department of Breast Cancer Pathology and Research Laboratory, State Key Laboratory of Breast Cancer Research, Cancer Institute and Hospital, Tianjin Medical University, Tianjin 300060, PR China
| | - Xin Ma
- Department of Cellular and Molecular Pharmacology, School of Pharmaceutical Sciences, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
42
|
Cherepanova N, Shrimal S, Gilmore R. N-linked glycosylation and homeostasis of the endoplasmic reticulum. Curr Opin Cell Biol 2016; 41:57-65. [PMID: 27085638 DOI: 10.1016/j.ceb.2016.03.021] [Citation(s) in RCA: 182] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 03/24/2016] [Accepted: 03/30/2016] [Indexed: 01/17/2023]
Abstract
As a major site of protein biosynthesis, homeostasis of the endoplasmic reticulum is critical for cell viability. Asparagine linked glycosylation of newly synthesized proteins by the oligosaccharyltransferase plays a central role in ER homeostasis due to the use of protein-linked oligosaccharides as recognition and timing markers for glycoprotein quality control pathways that discriminate between correctly folded proteins and terminally malfolded proteins destined for ER associated degradation. Recent findings indicate how the oligosaccharyltransferase achieves efficient and accurate glycosylation of the diverse proteins that enter the endoplasmic reticulum. In metazoan organisms two distinct OST complexes cooperate to maximize the glycosylation of nascent proteins. The STT3B complex glycosylates acceptor sites that have been skipped by the translocation channel associated STT3A complex.
Collapse
Affiliation(s)
- Natalia Cherepanova
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA 01605, United States
| | - Shiteshu Shrimal
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA 01605, United States
| | - Reid Gilmore
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA 01605, United States.
| |
Collapse
|
43
|
Gu Y, Wang Q, Guo K, Qin W, Liao W, Wang S, Ding Y, Lin J. TUSC3 promotes colorectal cancer progression and epithelial-mesenchymal transition (EMT) through WNT/β-catenin and MAPK signalling. J Pathol 2016; 239:60-71. [PMID: 27071482 DOI: 10.1002/path.4697] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 11/25/2015] [Accepted: 01/15/2016] [Indexed: 01/05/2023]
Abstract
Colorectal cancer (CRC) is one of the most common malignancies and is the second leading cause of cancer death in humans. Tumour suppressor candidate 3 (TUSC3) plays an important role in embryogenesis and metabolism. Deletion of TUSC3 often causes non-syndromic mental retardation. Even though TUSC3 deregulation is frequently observed in epithelial cancers, the function of TUSC3 in CRC has remained unknown. In this study, we observed greater expression of TUSC3 at the mRNA and protein level in clinical colorectal tumour samples compared with paired normal tissues. Gain- and loss-of-function analyses were performed to evaluate the functional significance of TUSC3 in CRC initiation and progression. Immunoblotting, immunofluorescence, and co-immunoprecipitation analyses were used to identify potential pathways with which TUSC3 might be involved. Overexpression of TUSC3 in CRC cells induced epithelial-mesenchymal transition (EMT) in CRC cells, accompanied by down-regulation of the epithelial marker, E-cadherin, and up-regulation of the mesenchymal marker, vimentin. Increased proliferation, migration, and invasion, as well as accelerated xenograft tumour growth, were observed in TUSC3-overexpressing CRC cells, while opposite effects were achieved in TUSC3-silenced cells. In conclusion, our study demonstrated the oncogenic role of TUSC3 in CRC and showed that TUSC3 may be responsible for alternations in the proliferation ability, aggressiveness, and invasive/metastatic potential of CRC through regulating the MAPK, PI3K/Akt, and Wnt/β-catenin signalling pathways.
Collapse
Affiliation(s)
- Ye Gu
- Department of Pathology, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue, Guangzhou, PR China.,Department of Pathology, School of Basic Medicine, Southern Medical University, 1838 Guangzhou Avenue, Guangzhou, PR China.,Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, 1838 Guangzhou Avenue, Guangzhou, PR China
| | - Qian Wang
- Department of Pathology, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue, Guangzhou, PR China.,Department of Pathology, School of Basic Medicine, Southern Medical University, 1838 Guangzhou Avenue, Guangzhou, PR China.,Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, 1838 Guangzhou Avenue, Guangzhou, PR China
| | - Kang Guo
- Department of Pathology, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue, Guangzhou, PR China.,Department of Pathology, School of Basic Medicine, Southern Medical University, 1838 Guangzhou Avenue, Guangzhou, PR China.,Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, 1838 Guangzhou Avenue, Guangzhou, PR China
| | - Weizhao Qin
- Department of Pathology, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue, Guangzhou, PR China.,Department of Pathology, School of Basic Medicine, Southern Medical University, 1838 Guangzhou Avenue, Guangzhou, PR China.,Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, 1838 Guangzhou Avenue, Guangzhou, PR China
| | - Wenting Liao
- Department of Pathology, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue, Guangzhou, PR China.,Department of Pathology, School of Basic Medicine, Southern Medical University, 1838 Guangzhou Avenue, Guangzhou, PR China.,Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, 1838 Guangzhou Avenue, Guangzhou, PR China
| | - Shuang Wang
- Department of Pathology, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue, Guangzhou, PR China.,Department of Pathology, School of Basic Medicine, Southern Medical University, 1838 Guangzhou Avenue, Guangzhou, PR China.,Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, 1838 Guangzhou Avenue, Guangzhou, PR China
| | - Yanqing Ding
- Department of Pathology, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue, Guangzhou, PR China.,Department of Pathology, School of Basic Medicine, Southern Medical University, 1838 Guangzhou Avenue, Guangzhou, PR China.,Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, 1838 Guangzhou Avenue, Guangzhou, PR China
| | - Jie Lin
- Department of Pathology, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue, Guangzhou, PR China.,Department of Pathology, School of Basic Medicine, Southern Medical University, 1838 Guangzhou Avenue, Guangzhou, PR China.,Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, 1838 Guangzhou Avenue, Guangzhou, PR China
| |
Collapse
|
44
|
Fan X, Zhang X, Shen J, Zhao H, Yu X, Chen Y, Zhuang Z, Deng X, Feng H, Wang Y, Peng L. Decreased TUSC3 Promotes Pancreatic Cancer Proliferation, Invasion and Metastasis. PLoS One 2016; 11:e0149028. [PMID: 26871953 PMCID: PMC4752499 DOI: 10.1371/journal.pone.0149028] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2015] [Accepted: 01/26/2016] [Indexed: 12/24/2022] Open
Abstract
Pancreatic cancer is an aggressive disease with dismal prognosis. It is of paramount importance to understand the underlying etiological mechanisms and identify novel, consistent, and easy-to-apply prognostic factors for precision therapy. TUSC3 (tumor suppressor candidate 3) was identified as a potential tumor suppressor gene and previous study showed TUSC3 is decreased in pancreatic cancer at mRNA level, but its putative tumor suppressor function remains to be verified. In this study, TUSC3 expression was found to be suppressed both at mRNA and protein levels in cell line models as well as in clinical samples; decreased TUSC3 expression was associated with higher pathological TNM staging and poorer outcome. In three pairs of cell lines with different NF-κB activity, TUSC3 expression was found to be reversely correlated with NF-κB activity. TUSC3-silenced pancreatic cancer cell line exhibited enhanced potential of proliferation, migration and invasion. In an orthotopic implanted mice model, TUSC3 silenced cells exhibited more aggressive phenotype with more liver metastasis. In conclusion, the current study shows that decreased immunological TUSC3 staining is a factor prognostic of poor survival in pancreatic cancer patients and decreased TUSC3 promotes pancreatic cancer cell proliferation, invasion and metastasis. The reverse correlation between NF-κB activity and TUSC3 expression may suggest a novel regulation pattern for this molecule.
Collapse
Affiliation(s)
- Xiaoqiang Fan
- Department of Oncology, Jimin Hospital, Shanghai, China
| | - Xiu Zhang
- Department of Oncology, Jimin Hospital, Shanghai, China
| | - Jie Shen
- Department of Oncology, Jimin Hospital, Shanghai, China
- * E-mail:
| | - Haibin Zhao
- Department of Pathology, Taihu Hospital, Wuxi, Jiangsu Province, China
| | - Xuetao Yu
- Department of Oncology, Jimin Hospital, Shanghai, China
| | - Yong’an Chen
- Department of Oncology, Jimin Hospital, Shanghai, China
| | - Zhuonan Zhuang
- Department of General Surgery, Shandong University Affiliated Qilu Hospital, Ji’nan, Shandong Province, China
| | - Xiaolong Deng
- Department of Oncology, Jimin Hospital, Shanghai, China
| | - Hua Feng
- Department of Oncology, Jimin Hospital, Shanghai, China
| | - Yunfei Wang
- Department of Oncology, Jimin Hospital, Shanghai, China
| | - Long Peng
- Department of Oncology, Jimin Hospital, Shanghai, China
| |
Collapse
|
45
|
The role of the endoplasmic reticulum stress in stemness, pluripotency and development. Eur J Cell Biol 2016; 95:115-23. [PMID: 26905505 DOI: 10.1016/j.ejcb.2016.02.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 02/01/2016] [Accepted: 02/05/2016] [Indexed: 01/15/2023] Open
Abstract
The molecular machinery of endoplasmic reticulum (ER) integrates various intracellular and extracellular cues to maintain homeostasis in diverse physiological or pathological scenarios. ER stress and the unfolded protein response (UPR) have been found to mediate molecular and biochemical mechanisms that affect cell proliferation, differentiation, and apoptosis. Although a number of reviews on the ER stress response have been published, comprehensive reviews that broadly summarize ER physiology in the context of pluripotency, embryonic development, and tissue homeostasis are lacking. This review complements the current ER literature and provides a summary of the important findings on the role of the ER stress and UPR in embryonic development and pluripotent stem cells.
Collapse
|
46
|
Zhang TW, Xing L, Tang JL, Lu JX, Liu CX. Marchantin M Induces Apoptosis of Prostate Cancer Cells Through Endoplasmic Reticulum Stress. Med Sci Monit 2015; 21:3570-6. [PMID: 26581488 PMCID: PMC4657764 DOI: 10.12659/msm.894476] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 07/22/2015] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Apoptosis is mediated by the endoplasmic reticulum (ER) stress pathway, mitochondrial pathway, and death receptor. Data herein suggested an inhibitory effect of marchantin M on tumor formation in nude mice as well as the impact on CHOP and GRP78 expression. MATERIAL AND METHODS The role of marchantin M on proliferation and apoptosis of DU145 cells were measured by MTT and flow cytometry, respectively. Western blot was applied to detect the expression of GRP78 and CHOP. The mice received abdominal injection at 1 time/2 d and 2 ml/time. Tumor volume was measured every 6 days. The mice were euthanatized 30 days after marchantin injection and tumor weight was measured. Cell apoptosis was determined by TUNEL. The expressions of CHOP and GRP78 were detected by immunohistochemistry. RESULTS Tumor size and weight in marchantin groups were significantly lower than in the control group (A, B) (P<0.05), and the inhibitory rate presented a dose-dependent increase. Compared with controls, the levels of CHOP and GRP78 expression elevated obviously following the treatment with marchantin (P<0.05). It showed statistically significant difference among groups C, D, E, with different levels of apoptosis indexes incremented in groups of marchantin H, M, L, compared with groups A and B (P<0.05). CONCLUSIONS Overall, this study shows that marchantin M circumvents the growth of prostate cancer PC-3 tumor and up-regulates expressions of CHOP and GRP78. Our data also indicate that marchantin M limits the proliferation and favors apoptosis of DU145 cells in a time- and dose-dependent manner.
Collapse
Affiliation(s)
- Tian-Wei Zhang
- Department of Urology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, P.R. China
- The First Clinical Medical College, Southern Medical University, Guangzhou, Guangdong, P.R. China
| | - Li Xing
- The First Clinical Medical College, Southern Medical University, Guangzhou, Guangdong, P.R. China
| | - Jun-Long Tang
- Shenzhen Tumor Immuno-Gene Therapy Clinical Application Engineering Lab, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, P.R. China
- Department of Urology, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, P.R. China
| | - Jing-Xiao Lu
- Shenzhen Tumor Immuno-Gene Therapy Clinical Application Engineering Lab, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, P.R. China
- Department of Urology, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, P.R. China
| | - Chun-Xiao Liu
- Department of Urology, Zhujiang Hospital, Second Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, P.R. China
| |
Collapse
|
47
|
Feng R, Ye J, Zhou C, Qi L, Fu Z, Yan B, Liang Z, Li R, Zhai W. Calreticulin down-regulation inhibits the cell growth, invasion and cell cycle progression of human hepatocellular carcinoma cells. Diagn Pathol 2015; 10:149. [PMID: 26307067 PMCID: PMC4549864 DOI: 10.1186/s13000-015-0382-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 08/19/2015] [Indexed: 01/08/2023] Open
Abstract
Background Hepatocellular carcinoma (HCC) is one of the most frequent cancers in the world. Calreticulin(CRT) is aberrantly overexpressed in many human cancer cells. The function of CRT in HCC cells remains unclear. We attempted to investigate the effects and the underlying mechanisms of CRT down-regulation on HCC cell growth, apoptosis, cell cycle progression and invasion. Methods To investigate the function of CRT in HCC cells, small interfering RNA (siRNA) was used to knock down the expression of CRT in SMMC7721 and HepG2 HCC cells. CRT expression was examined by Western blot and immunofluorescence. Cell proliferation was detected by CCK-8 assay. Cell cycle and apoptosis were measured by the flow cytometry. The invasion capability was assessed by transwell assay. The phosphorylation level of Akt was evaluated by Western blot. Results Compared with human hepatic cells L02, CRT was apparently up-regulated in SMMC7721, HepG2 and Huh7 HCC cells. Down-regulation of CRT expression effectively inhibited HCC cell growth and invasion. CRT knockdown induced cell cycle arrest and the apoptosis in SMMC7721 and HepG2 cells. Furthermore, down-regulation of CRT expression significantly decreased the Akt phosphorylation. Conclusions CRT was aberrantly over-expressed in HCC cell lines. CRT over-expression contributes greatly to HCC malignant behavior, likely via PI3K/Akt pathway. CRT could serve as a potential biomarker and therapeutic target for hepatocellular carcinoma.
Collapse
Affiliation(s)
- Ruo Feng
- Department of Histology and Embryology, Medical College of Zhengzhou University, Zhengzhou, People's Republic of China.
| | - Jianwen Ye
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China. .,Key Laboratory of Hepatobiliary and Pancreatic Surgery & Digestive Organ Transplantation of Henan Province, Zhengzhou, People's Republic of China.
| | - Chuang Zhou
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China. .,Key Laboratory of Hepatobiliary and Pancreatic Surgery & Digestive Organ Transplantation of Henan Province, Zhengzhou, People's Republic of China.
| | - Lei Qi
- Department of Endocrine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Zhe Fu
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China.,Key Laboratory of Hepatobiliary and Pancreatic Surgery & Digestive Organ Transplantation of Henan Province, Zhengzhou, People's Republic of China
| | - Bing Yan
- Key Laboratory of Hepatobiliary and Pancreatic Surgery & Digestive Organ Transplantation of Henan Province, Zhengzhou, People's Republic of China.
| | - Zhiwei Liang
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China. .,Key Laboratory of Hepatobiliary and Pancreatic Surgery & Digestive Organ Transplantation of Henan Province, Zhengzhou, People's Republic of China.
| | - Renfeng Li
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China.,Key Laboratory of Hepatobiliary and Pancreatic Surgery & Digestive Organ Transplantation of Henan Province, Zhengzhou, People's Republic of China
| | - Wenlong Zhai
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China. .,Key Laboratory of Hepatobiliary and Pancreatic Surgery & Digestive Organ Transplantation of Henan Province, Zhengzhou, People's Republic of China.
| |
Collapse
|
48
|
Vasseur S, Manié SN. ER stress and hexosamine pathway during tumourigenesis: A pas de deux? Semin Cancer Biol 2015; 33:34-9. [PMID: 25931390 DOI: 10.1016/j.semcancer.2015.04.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 04/16/2015] [Indexed: 12/28/2022]
Abstract
Both the hexosamine biosynthetic pathway (HBP) and the endoplasmic reticulum (ER) are considered sensors for the nutritional state of the cell. The former is a branch of the glucose metabolic pathway that provides donor molecules for glycosylation processes, whereas the second requires co-translational N-glycosylation to ensure proper protein folding. It has become clear that the microenvironment of solid tumours, characterised by poor oxygen and nutrient supply, challenges optimal functions of the ER and the HBP. Here, we review recent advances demonstrating that the ER stress (ERS) response and HBP pathways are interconnected to promote cell viability. We then develop the idea that communication between ER and HBP is a survival feature of neoplastic cells that plays a prominent role during tumourigenesis.
Collapse
Affiliation(s)
- Sophie Vasseur
- INSERM U1068, Centre de Recherche en Cancérologie de Marseille, France; Institut Paoli-Calmettes, France; CNRS, UMR7258, F-13009 Marseille, France; Université Aix-Marseille, F-13284 Marseille, France
| | - Serge N Manié
- INSERM U1052, CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France; Université de Lyon, Université Lyon 1, F-69000 Lyon, France; Centre Léon Bérard, F-69008 Lyon, France.
| |
Collapse
|
49
|
Kratochvílová K, Horak P, Ešner M, Souček K, Pils D, Anees M, Tomasich E, Dráfi F, Jurtíková V, Hampl A, Krainer M, Vaňhara P. Tumor suppressor candidate 3 (TUSC3) prevents the epithelial-to-mesenchymal transition and inhibits tumor growth by modulating the endoplasmic reticulum stress response in ovarian cancer cells. Int J Cancer 2015; 137:1330-40. [PMID: 25735931 DOI: 10.1002/ijc.29502] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2014] [Revised: 01/28/2015] [Accepted: 02/18/2015] [Indexed: 01/17/2023]
Abstract
Ovarian cancer is one of the most common malignancies in women and contributes greatly to cancer-related deaths. Tumor suppressor candidate 3 (TUSC3) is a putative tumor suppressor gene located at chromosomal region 8p22, which is often lost in epithelial cancers. Epigenetic silencing of TUSC3 has been associated with poor prognosis, and hypermethylation of its promoter provides an independent biomarker of overall and disease-free survival in ovarian cancer patients. TUSC3 is localized to the endoplasmic reticulum in an oligosaccharyl tranferase complex responsible for the N-glycosylation of proteins. However, the precise molecular role of TUSC3 in ovarian cancer remains unclear. In this study, we establish TUSC3 as a novel ovarian cancer tumor suppressor using a xenograft mouse model and demonstrate that loss of TUSC3 alters the molecular response to endoplasmic reticulum stress and induces hallmarks of the epithelial-to-mesenchymal transition in ovarian cancer cells. In summary, we have confirmed the tumor-suppressive function of TUSC3 and identified the possible mechanism driving TUSC3-deficient ovarian cancer cells toward a malignant phenotype.
Collapse
Affiliation(s)
- Kateřina Kratochvílová
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic.,Center of Biomolecular and Cellular Engineering, International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
| | - Peter Horak
- Department of Internal Medicine I and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Milan Ešner
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Karel Souček
- Center of Biomolecular and Cellular Engineering, International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic.,Department of Cytokinetics, Institute of Biophysics, Academy of Sciences of the Czech Republic, v.v.i., Brno, Czech Republic.,Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Dietmar Pils
- Department of Obstetrics and Gynecology, Molecular Oncology Group, Medical University of Vienna, Vienna, Austria
| | - Mariam Anees
- Department of Internal Medicine I and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria.,Department of Biochemistry, Quaid-i-Azam University, Islamabad, Pakistan
| | - Erwin Tomasich
- Department of Internal Medicine I and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - František Dráfi
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Veronika Jurtíková
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Aleš Hampl
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Michael Krainer
- Department of Internal Medicine I and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Petr Vaňhara
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| |
Collapse
|
50
|
Drake RR, Jones EE, Powers TW, Nyalwidhe JO. Altered glycosylation in prostate cancer. Adv Cancer Res 2015; 126:345-82. [PMID: 25727153 DOI: 10.1016/bs.acr.2014.12.001] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Prostate cancer is annually the most common newly diagnosed cancer in men. The prostate functions as a major secretory gland for the production of glycoproteins critical to sperm activation and reproduction. Prostate-specific antigen (PSA), produced by the prostate, is one of the most commonly assayed glycoproteins in blood, serving as a biomarker for early detection and progression of prostate cancer. The single site of N-glycosylation on PSA has been the target of multiple glycan characterization studies. In this review, the extensive number of studies that have characterized the changes in O-linked and N-linked glycosylations associated with prostate cancer development and progression will be summarized. This includes analysis of the glycosylation of PSA, and other prostate glycoproteins, in tissues, clinical biofluids, and cell line models. Other studies are summarized in the context of understanding the complexities of these glycan changes in order to address the many confounding questions associated with prostate cancer, as well as efforts to improve prostate cancer biomarker assays using targeted glycomic-based strategies.
Collapse
Affiliation(s)
- Richard R Drake
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, South Carolina, USA.
| | - E Ellen Jones
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Thomas W Powers
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Julius O Nyalwidhe
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, Virginia, USA
| |
Collapse
|