1
|
Nadukkandy AS, Blaize B, Kumar CD, Mori G, Cordani M, Kumar LD. Non-coding RNAs as mediators of epithelial to mesenchymal transition in metastatic colorectal cancers. Cell Signal 2025; 127:111605. [PMID: 39842529 DOI: 10.1016/j.cellsig.2025.111605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 01/06/2025] [Accepted: 01/12/2025] [Indexed: 01/24/2025]
Abstract
Colorectal cancer (CRC) remains a leading cause of cancer-related mortality globally, necessitating the development of innovative treatment strategies. Recent research has underscored the significant role of non-coding RNAs (ncRNAs) in CRC pathogenesis, offering new avenues for diagnosis and therapy. In this review, we delve into the intricate roles of various ncRNAs, including microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs), in CRC progression, epithelial-mesenchymal transition (EMT), metastasis, and drug resistance. We highlight the interaction of these ncRNAs with and regulation of key signaling pathways, such as Wnt/β-catenin, Notch, JAK-STAT, EGFR, and TGF-β, and the functional relevance of these interactions in CRC progression. Additionally, the review highlights the emerging applications of nanotechnology in enhancing the delivery and efficacy of ncRNA-based therapeutics, which could address existing challenges related to specificity and side effects. Future research directions, including advanced diagnostic tools, targeted therapeutics, strategies to overcome drug resistance, and the integration of personalized medicine approaches are discussed. Integrating nanotechnology with a deeper understanding of CRC biology offers the potential for more effective, targeted, and personalized strategies, though further research is essential to validate these approaches.
Collapse
Affiliation(s)
- Aisha Shigna Nadukkandy
- Cancer Biology, CSIR-Centre for Cellular and Molecular Biology (CCMB), Hyderabad 500007, India
| | - Britny Blaize
- Cancer Biology, CSIR-Centre for Cellular and Molecular Biology (CCMB), Hyderabad 500007, India
| | - Chethana D Kumar
- Department of Surgical ICU, Christian Medical College, IDA Scudder Road, Vellore 632004, Tamil Nadu, India
| | - Giulia Mori
- Department Of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| | - Marco Cordani
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, Complutense University of Madrid, 28040 Madrid, Spain; Instituto de Investigaciones Sanitarias San Carlos (IdISSC), 28040 Madrid, Spain.
| | - Lekha Dinesh Kumar
- Cancer Biology, CSIR-Centre for Cellular and Molecular Biology (CCMB), Hyderabad 500007, India.
| |
Collapse
|
2
|
Huang H, Jiang NN, Lu GW, Xu F, Sun LL, Zhu J, Dong Z, Zhang ZJ, Liu S. CircMETTL9 targets CCAR2 to induce neuronal oxidative stress and apoptosis via mitochondria-mediated pathways following traumatic brain injury. Free Radic Biol Med 2025; 228:44-61. [PMID: 39709098 DOI: 10.1016/j.freeradbiomed.2024.12.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 11/18/2024] [Accepted: 12/19/2024] [Indexed: 12/23/2024]
Abstract
Traumatic brain injury (TBI) remains a principal factor in neurological disorders, often resulting in significant morbidity due to secondary neuroinflammatory and oxidative stress responses. While circular RNAs are recognized for their high expression levels in the nervous system and play crucial roles in various neurological processes, their specific contributions to the pathophysiology of TBI remain underexplored. In this study, the possible molecular mechanisms through which circMETTL9 modulated oxidative stress and neurological outcomes following TBI were investigated. In vitro model of oxidative stress utilizing SH-SY5Y cells revealed that circMETTL9 knockdown significantly attenuated H₂O₂-induced reactive oxygen species (ROS) production, reduced apoptosis, and preserved mitochondrial function. Additionally, CCAR2 has been identified as a circMETTL9-binding protein by mass spectrometry and RNA immunoprecipitation, with circMETTL9 positively regulating CCAR2 expression. Meanwhile, on the basis of silencing CCAR2, it was verified that the regulation of oxidative stress in SH-SY5Y cells by circMETTL9 was mediated by CCAR2. In vivo experiments using a TBI rat model further confirmed that CCAR2 knockdown alleviated central nervous system (CNS) injury, reduced oxidative stress and apoptosis, and protected mitochondrial integrity following TBI. These findings suggest a novel mechanism by which circMETTL9 targets CCAR2 via mitochondria-mediated Bax/Bcl-2/caspase-3 signaling to regulate apoptosis. CircMETTL9 may provide a viable therapeutic target for mitigating neurological dysfunction following TBI, offering new insights into potential interventions aimed at reducing secondary brain injury.
Collapse
Affiliation(s)
- Hao Huang
- Department of Rehabilitation Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu Province, China; School of Nursing and Rehabilitation, Nantong University, Nantong, 226001, Jiangsu Province, China
| | - Nan-Nan Jiang
- Department of Rehabilitation Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu Province, China; School of Nursing and Rehabilitation, Nantong University, Nantong, 226001, Jiangsu Province, China
| | - Gui-Wei Lu
- Department of Rehabilitation Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu Province, China; School of Nursing and Rehabilitation, Nantong University, Nantong, 226001, Jiangsu Province, China; Department of Rehabilitation Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, 225300, Jiangsu Province, China
| | - Feng Xu
- The Second People's Hospital of Nantong, Nantong, 226002, Jiangsu Province, China
| | - Lu-Lu Sun
- Department of Rehabilitation Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu Province, China
| | - Jing Zhu
- Department of Rehabilitation Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu Province, China
| | - Zhao Dong
- Nanjing Vocational Health College, Nanjing, 210038, Jiangsu Province, China.
| | - Zhi-Jun Zhang
- Department of Human Anatomy, School of Medicine, Nantong University, Nantong, 226001, Jiangsu Province, China.
| | - Su Liu
- Department of Rehabilitation Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu Province, China.
| |
Collapse
|
3
|
ZHOU ZHENGYANG, QIAO LEI, WANG TONGTONG, PAN WEN, DUAN JINGJING, ZHANG HAIYANG, DENG TING, BA YI, HE YI. Exosomal miR-224-3p promotes lymphangiogenesis and lymph node metastasis by targeting GSK3B in gastric cancer. Oncol Res 2025; 33:327-345. [PMID: 39866224 PMCID: PMC11753999 DOI: 10.32604/or.2024.050431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 05/27/2024] [Indexed: 01/28/2025] Open
Abstract
Background Patients with gastric cancer (GC) are prone to lymph node metastasis (LNM), which is an important factor for recurrence and poor prognosis of GC. Nowadays, more and more studies have confirmed that exosomes can participate in tumor lymphangiogenesis. An in-depth exploration of the pathological mechanism in the process of LNM in GC may provide effective targets and improve the diagnosis and treatment effect. Materials and Methods We used sequencing analysis of collected serum to screen out exo-miRNA related to LNM in GC. ELISA, qRT-PCR, Western Blot, RNA pull-down assay, Transwell assay, animal experiments, and other experiments were used to verify the results. Results In this study, we screened out miR-224-3p related to GC progression and LNM in a vascular endothelial growth Factor C (VEGFC)-independent manner. We found that exo-miR-224-3p derived from GC cells could enter human lymphatic endothelial cells (HLECs) and promote the tube formation and migration of HLECs. In addition, it was revealed that miR-224-3p could bind to the 3'UTR region of GSK3B mRNA. Then, we proved that inhibiting the expression of GSK3B could suppress the phosphorylation of β-catenin and promote the transcription of PROX1, thus leading to tumor lymphangiogenesis. Furthermore, it was also found that hnRNPA1 mediated the sorting of miR-224-3p into exosomes, and the high expression of PKM2 promoted the secretion of exo-miR-224-3p. Conclusions Our discovery of the exo-miR-224-3p/GSK3B/β-catenin/PROX1 axis may provide a new direction for the clinical treatment of GC.
Collapse
Affiliation(s)
- ZHENGYANG ZHOU
- Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, 300060, China
| | - LEI QIAO
- Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, 300060, China
| | - TONGTONG WANG
- Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, 300060, China
| | - WEN PAN
- Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, 300060, China
| | - JINGJING DUAN
- Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, 300060, China
| | - HAIYANG ZHANG
- Tianjin Union Medical Center of Nankai University, Nankai University, Tianjin, 300060, China
| | - TING DENG
- Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, 300060, China
| | - YI BA
- Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, 300060, China
| | - YI HE
- Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, 300060, China
| |
Collapse
|
4
|
Gan X, Feng Y, Liu Y, Lin X, Yu X, Rong X, Han Q. Identification of zinc finger MIZ-type containing 2 as an oncoprotein enhancing NAD-dependent protein deacetylase sirtuin-1 deacetylase activity to regulate Wnt and Hippo pathways in non-small-cell lung cancer. Cell Mol Biol Lett 2024; 29:122. [PMID: 39266996 PMCID: PMC11391738 DOI: 10.1186/s11658-024-00636-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 08/19/2024] [Indexed: 09/14/2024] Open
Abstract
BACKGROUND Zinc finger MIZ-type containing 2 (ZMIZ2) can function as a coactivator and participate in the progression of certain malignant tumors; however, its expression and underlying molecular mechanism in non-small-cell lung cancer (NSCLC) remains unknown. In this study, we aim to analyze the expression of ZMIZ2 and its tumorigenic function in NSCLC, identifying its related factors. METHODS ZMIZ2 expression in NSCLC tissue samples and cell lines was examined using immunohistochemistry and western blotting; its biological role was investigated using in vivo and in vitro assays. The association between ZMIZ2 and NAD-dependent protein deacetylase sirtuin-1 (SIRT1) was demonstrated using mass spectrometry and immunoprecipitation experiments. Kyoto Encyclopedia of Genes and Genomes Pathway (KEGG)-based enrichment analysis, luciferase reporter assay, and real-time quantitative polymerase chain reaction (RT-qPCR) were conducted to verify the impact of ZMIZ2-SIRT1 combination on Hippo/Wnt pathways. RESULTS ZMIZ2 was highly expressed in NSCLC and positively associated with advanced pTNM staging, lymph node metastasis, and poor overall survival. Functional experiments revealed that ZMIZ2 promotes the proliferation, migration, and invasiveness of lung cancer cells-establishing its role as a promoter of oncogenes. Molecular mechanism studies identified SIRT1 as an assisted key factor interacting with ZMIZ2. KEGG enrichment analysis revealed that ZMIZ2 is closely related to Wnt/Hippo pathways; ZMIZ2-SIRT1 interaction enhanced SIRT1 deacetylase activity. Direct downregulation of intranuclear β-catenin and yes-associated protein (YAP) acetylation levels occurred independently of upstream proteins in Wnt/Hippo pathways; transcriptional activities of β-catenin-transcription factor 4 (TCF4) and YAP-TEA domain family transcription factors (TEADs) were amplified. CONCLUSIONS ZMIZ2 promotes the malignant phenotype of lung cancer by regulating Wnt/Hippo pathways through SIRT1, providing an experimental basis for discovering novel biomarkers and developing tumor-targeted drugs.
Collapse
Affiliation(s)
- Xueting Gan
- Department of Pathology, Shenbei New Area, College of Basic Medical Sciences and the First Hospital of China Medical University. No, 77 Puhe Road, Shenyang, 110122, Liaoning Province, People's Republic of China
| | - Yuheng Feng
- Department of Pathology, Shenbei New Area, College of Basic Medical Sciences and the First Hospital of China Medical University. No, 77 Puhe Road, Shenyang, 110122, Liaoning Province, People's Republic of China
| | - Yang Liu
- Department of Pathology, Shenbei New Area, College of Basic Medical Sciences and the First Hospital of China Medical University. No, 77 Puhe Road, Shenyang, 110122, Liaoning Province, People's Republic of China
| | - Xuyong Lin
- Department of Pathology, Shenbei New Area, College of Basic Medical Sciences and the First Hospital of China Medical University. No, 77 Puhe Road, Shenyang, 110122, Liaoning Province, People's Republic of China
| | - Xinmiao Yu
- Department of Surgical Oncology and Breast Surgery, the First Hospital of China Medical University, Shenyang. No. 155 Nanjing North Street, Heping Area, Shenyang, 110001, Liaoning Province, People's Republic of China.
| | - Xuezhu Rong
- Department of Pathology, the First Hospital of China Medical University. No, 155 Nanjing North Street, Heping Area, Shenyang, 110001, Liaoning Province, People's Republic of China.
| | - Qiang Han
- Department of Pathology, Shenbei New Area, College of Basic Medical Sciences and the First Hospital of China Medical University. No, 77 Puhe Road, Shenyang, 110122, Liaoning Province, People's Republic of China.
| |
Collapse
|
5
|
Mohan N, Johnson GS, Tovar Perez JE, Dashwood WM, Rajendran P, Dashwood RH. Alternative splicing of BAZ1A in colorectal cancer disrupts the DNA damage response and increases chemosensitization. Cell Death Dis 2024; 15:570. [PMID: 39112459 PMCID: PMC11306231 DOI: 10.1038/s41419-024-06954-6] [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: 03/14/2024] [Revised: 07/23/2024] [Accepted: 07/25/2024] [Indexed: 08/10/2024]
Abstract
Bromodomain Adjacent to Zinc Finger Domain 1A (BAZ1A) is a critical regulator of chromatin remodeling. We sought to clarify the roles of BAZ1A in the etiology of colorectal cancer, including the mechanisms of its alternatively spliced variants. Public databases were examined and revealed high BAZ1A expression in the majority of colorectal cancer patients, which was corroborated in a panel of human colon cancer cell lines. BAZ1A silencing reduced cell viability and increased markers of DNA damage, apoptosis, and senescence, along with the downregulation of Wnt/β-catenin signaling. The corresponding molecular changes resulted in tumor growth inhibition when BAZ1A-knockout cells were implanted into nude mice. In rescue experiments, a short isoform of BAZ1A that was associated with alternative splicing by the DBIRD complex failed to restore DNA repair activity in colon cancer cells and maintained chemosensitivity to phleomycin treatment, unlike the full-length BAZ1A. A working model proposes that a buried domain in the N-terminus of the BAZ1A short isoform lacks the ability to access linker DNA, thereby disrupting the activity of the associated chromatin remodeling complexes. Given the current interest in RNA splicing deregulation and cancer etiology, additional mechanistic studies are warranted with new lead compounds targeting BAZ1A, and other members of the BAZ family, with a view to improved therapeutic interventions.
Collapse
Affiliation(s)
- Nivedhitha Mohan
- Center for Epigenetics & Disease Prevention, Texas A&M Health, Houston, TX, USA
| | - Gavin S Johnson
- Center for Epigenetics & Disease Prevention, Texas A&M Health, Houston, TX, USA
- CRISPR Therapeutics, South Boston, MA, USA
| | | | | | - Praveen Rajendran
- Center for Epigenetics & Disease Prevention, Texas A&M Health, Houston, TX, USA.
- Department of Translational Medical Sciences, Texas A&M College of Medicine, Houston, TX, USA.
| | - Roderick H Dashwood
- Center for Epigenetics & Disease Prevention, Texas A&M Health, Houston, TX, USA.
- Department of Translational Medical Sciences, Texas A&M College of Medicine, Houston, TX, USA.
| |
Collapse
|
6
|
Liu J, Zhu P. A Novel Gene Signature Associated with Protein Post-translational Modification to Predict Clinical Outcomes and Therapeutic Responses of Colorectal Cancer. Mol Biotechnol 2024; 66:2106-2122. [PMID: 37592152 DOI: 10.1007/s12033-023-00852-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 08/03/2023] [Indexed: 08/19/2023]
Abstract
Accumulated evidence highlights the biological significance of diverse protein post-translational modifications (PTMs) in tumorigenicity and progression of colorectal cancer (CRC). In this study, ten PTM patterns (ubiquitination, methylation, phosphorylation, glycosylation, acetylation, SUMOylation, citrullination, neddylation, palmitoylation, and ADP-ribosylation) were analyzed for model construction. A post-translational modification index (PTMI) with a 14-gene signature was established. CRC patients with high PTMI had a worse prognosis after validating in nine independent datasets. By incorporating PTMI with clinical features, a nomogram with excellent predictive performance was constructed. Two molecular subtypes of CRC with obvious difference in survival time were identified by unsupervised clustering. Furthermore, PTMI was related to known immunoregulators and key tumor microenvironment components. Low-PTMI patients responded better to fluorouracil-based chemotherapy and immune checkpoint blockade therapy compared to high-PTMI patients, which was validated in multiple independent datasets. However, patients with high PTMI might be sensitive to bevacizumab. In short, we established a novel PTMI model by comprehensively analyzing diverse post-translational modification patterns, which can accurately predict clinical prognosis and treatment response of CRC patients.
Collapse
Affiliation(s)
- Jun Liu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Road, Yuzhong District, Chongqing, 400010, China
| | - Peng Zhu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Road, Yuzhong District, Chongqing, 400010, China.
| |
Collapse
|
7
|
Wang Y, Wang JM, Xiao Y, Hu XB, Zheng SY, Fu JL, Zhang L, Gan YW, Liang XM, Li DWC. SUMO1-regulated DBC1 promotes p53-dependent stress-induced apoptosis of lens epithelial cells. Aging (Albany NY) 2023; 15:8812-8832. [PMID: 37683133 PMCID: PMC10522365 DOI: 10.18632/aging.205001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 08/20/2023] [Indexed: 09/10/2023]
Abstract
Deleted in breast cancer 1 (DBC1) was initially identified from a homozygously deleted region in human chromosome 8p21. It has been well established that DBC1 plays a dual role during cancer development. Depending on the physiological context, it can promote or inhibit tumorigenesis. Whether it plays a role in lens pathogenesis remains elusive. In the present study, we demonstrated that DBC1 is highly expressed in lens epithelial cells from different vertebrates and in retina pigment epithelial cells as well. Moreover, DBC1 is SUMOylated through SUMO1 conjugation at K591 residue in human and mouse lens epithelial cells. The SUMOylated DBC1 is localized in the nucleus and plays an essential role in promoting stress-induced apoptosis. Silence of DBC1 attenuates oxidative stress-induced apoptosis. In contrast, overexpression of DBC1 enhances oxidative stress-induced apoptosis, and this process depends on p53. Mechanistically, DBC1 interacts with p53 to regulate its phosphorylation status at multiple sites and the SUMOylation of DBC1 enhances its interaction with p53. Together, our results identify that DBC1 is an important regulator mediating stress-induced apoptosis in lens, and thus participates in control of lens cataractogenesis.
Collapse
Affiliation(s)
- Yan Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong 510060, China
| | - Jing-Miao Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong 510060, China
| | - Yuan Xiao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong 510060, China
| | - Xue-Bin Hu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong 510060, China
| | - Shu-Yu Zheng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong 510060, China
| | - Jia-Ling Fu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong 510060, China
| | - Lan Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong 510060, China
| | - Yu-Wen Gan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong 510060, China
| | - Xing-Miao Liang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong 510060, China
| | - David Wan-Cheng Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong 510060, China
| |
Collapse
|
8
|
Arai M, Ochi H, Sunamura S, Ito N, Nangaku M, Takeda S, Sato S. A Novel Long Noncoding RNA in Osteocytes Regulates Bone Formation through the Wnt/β-Catenin Signaling Pathway. Int J Mol Sci 2023; 24:13633. [PMID: 37686441 PMCID: PMC10488071 DOI: 10.3390/ijms241713633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 08/30/2023] [Accepted: 08/31/2023] [Indexed: 09/10/2023] Open
Abstract
The vast majority of transcribed RNAs are noncoding RNAs. Among noncoding RNAs, long noncoding RNAs (lncRNAs), which contain hundreds to thousands of bases, have received attention in many fields. The vast majority of the constituent cells in bone tissue are osteocytes, but their regulatory mechanisms are incompletely understood. Considering the wide range of potential contributions of lncRNAs to physiological processes and pathological conditions, we hypothesized that lncRNAs in osteocytes, which have not been reported, could be involved in bone metabolism. Here, we first isolated osteocytes from femurs of mice with osteocyte-specific GFP expression. Then, through RNA-sequencing, we identified osteocyte-specific lncRNAs and focused on a novel lncRNA, 9530026P05Rik (lncRNA953Rik), which strongly suppressed osteogenic differentiation. In the IDG-SW3 osteocyte line with lncRNA953Rik overexpression, the expression of Osterix and its downstream genes was reduced. RNA pull-down and subsequent LC-MS/MS analysis revealed that lncRNA953Rik bound the nuclear protein CCAR2. We demonstrated that CCAR2 promoted Wnt/β-catenin signaling and that lncRNA953Rik inhibited this pathway. lncRNA953Rik sequestered CCAR2 from HDAC1, leading to deacetylation of H3K27 in the Osterix promoter and consequent transcriptional downregulation of Osterix. This research is the first to clarify the role of a lncRNA in osteocytes. Our findings can pave the way for novel therapeutic options targeting lncRNAs in osteocytes to treat bone metabolic diseases such as osteoporosis.
Collapse
Affiliation(s)
- Makoto Arai
- Division of Nephrology and Endocrinology, The University of Tokyo Hospital, Tokyo 113-8655, Japan
| | - Hiroki Ochi
- Department of Rehabilitation for Motor Functions, Research Institute, National Rehabilitation Center for Persons with Disabilities, Tokorozawa 359-8555, Japan
| | - Satoko Sunamura
- Department of Orthopaedic Surgery, Graduate School, Tokyo Medical and Dental University (TMDU), Tokyo 113-8519, Japan
| | - Nobuaki Ito
- Division of Nephrology and Endocrinology, The University of Tokyo Hospital, Tokyo 113-8655, Japan
- Osteoporosis Center, The University of Tokyo Hospital, Tokyo 113-8655, Japan
| | - Masaomi Nangaku
- Division of Nephrology and Endocrinology, The University of Tokyo Hospital, Tokyo 113-8655, Japan
| | - Shu Takeda
- Division of Endocrinology, Toranomon Hospital Endocrine Center, Tokyo 105-8470, Japan
| | - Shingo Sato
- Center for Innovative Cancer Treatment, Tokyo Medical and Dental University (TMDU), Tokyo 113-8519, Japan
| |
Collapse
|
9
|
Kim HJ, Moon SJ, Kim JH. Mechanistic insights into the dual role of CCAR2/DBC1 in cancer. Exp Mol Med 2023; 55:1691-1701. [PMID: 37524873 PMCID: PMC10474295 DOI: 10.1038/s12276-023-01058-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/21/2023] [Accepted: 05/17/2023] [Indexed: 08/02/2023] Open
Abstract
Cell cycle and apoptosis regulator 2 (CCAR2), also known as deleted in breast cancer 1 (DBC1), has been recently identified as a master regulator of transcriptional processes and plays diverse roles in physiology and pathophysiology, including as a regulator of apoptosis, DNA repair, metabolism, and tumorigenesis. CCAR2 functions as a coregulator of various transcription factors and a critical regulator of numerous epigenetic modifiers. Based on its ability to stimulate apoptosis by activating and stabilizing p53, CCAR2 was initially considered to be a tumor suppressor. However, an increasing number of studies have shown that CCAR2 also functions as a tumor-promoting coregulator by activating oncogenic transcription factors and regulating the enzymatic activity of epigenetic modifiers, indicating that CCAR2 may play a dual role in cancer progression by acting as a tumor suppressor and tumor promoter. Here, we review recent progress in understanding the dual tumor-suppressing and oncogenic roles of CCAR2 in cancer. We discuss CCAR2 domain structures, its interaction partners, and the molecular mechanisms by which it regulates the activities of transcription factors and epigenetic modifiers.
Collapse
Affiliation(s)
- Hwa Jin Kim
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, 06351, South Korea
- Research Institute for Future Medicine, Samsung Medical Center, Seoul, 06351, South Korea
| | - Sue Jin Moon
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, 06351, South Korea
- Research Institute for Future Medicine, Samsung Medical Center, Seoul, 06351, South Korea
| | - Jeong Hoon Kim
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, 06351, South Korea.
- Research Institute for Future Medicine, Samsung Medical Center, Seoul, 06351, South Korea.
| |
Collapse
|
10
|
Gan L, Li Q, Nie W, Zhang Y, Jiang H, Tan C, Zhang L, Zhang J, Li Q, Hou P, Yuan Y, Sun X, Liu D, Sheng W, Liu T, Xu M, Guo W. PROX1-mediated epigenetic silencing of SIRT3 contributes to proliferation and glucose metabolism in colorectal cancer. Int J Biol Sci 2023; 19:50-65. [PMID: 36594098 PMCID: PMC9760442 DOI: 10.7150/ijbs.73530] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 10/12/2022] [Indexed: 11/24/2022] Open
Abstract
Prospero-related homeobox 1 (PROX1) is a homeobox transcription factor known to promote malignant transformation and stemness in human colorectal cancer (CRC). However, the biological function of PROX1 in metabolic rearrangement in CRC remains unclear. Here, we aimed to uncover the relationship between the expression profile and role of PROX1 and CRC cell glucose metabolism and to elucidate the underlying molecular mechanism. PROX1 expression was significantly upregulated in human CRC tissues and positively associated with the maximum standardized uptake value (SUVmax), a measure of tissue 18-fluoro-2-deoxy-D-glucose uptake and an indicator of glycolysis and tumor cell activity, in patients with CRC. Knockdown of PROX1 suppressed CRC cell proliferation and glucose metabolism in vitro and in vivo. Mechanistically, through a physical interaction, PROX1 recruited EZH2 to the SIRT3 promoter and inhibited SIRT3 promoter activity. Moreover, PROX1 or EZH2 knockdown decreased cell glycolysis by targeting SIRT3. Clinically, high PROX1 expression combined with low SIRT3 expression predicted poor prognosis in patients with CRC. Thus, our study suggests that the PROX1-EZH2 complex positively regulates cell proliferation and glucose metabolism by engaging SIRT3 in CRC, which may serve as a promising therapeutic strategy for CRC.
Collapse
Affiliation(s)
- Lu Gan
- Department of Gastrointestinal Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Qingguo Li
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Wei Nie
- Department of Pulmonary Medicine, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Yi Zhang
- Department of Gastroenterology & Clinical Nutrition, The 452nd Hospital of PLA, Chengdu 610000, Sichuan, China
| | - Hesheng Jiang
- Department of Surgery, United Health Services Southern California Medical Education Consortium, Temecula Valley Hospital, Temecula, CA 92592, USA
| | - Cong Tan
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Institute of Pathology, Fudan University, Shanghai, 200032, China
| | - Long Zhang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Jieyun Zhang
- Department of Gastrointestinal Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Qian Li
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Pengcong Hou
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yitao Yuan
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xun Sun
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Dongmei Liu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Weiqi Sheng
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Institute of Pathology, Fudan University, Shanghai, 200032, China
| | - Tianshu Liu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Midie Xu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Institute of Pathology, Fudan University, Shanghai, 200032, China.,✉ Corresponding authors: Weijian Guo, PhD, Department of Gastrointestinal Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China. E-mail: ; Midie Xu, PhD, Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China. E-mail:
| | - Weijian Guo
- Department of Gastrointestinal Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,✉ Corresponding authors: Weijian Guo, PhD, Department of Gastrointestinal Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China. E-mail: ; Midie Xu, PhD, Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China. E-mail:
| |
Collapse
|
11
|
Benítez-Rosendo A, Lagos P, Cal K, Colman L, Escande C, Calliari A. Impaired hippocampal neurogenesis and cognitive performance in adult DBC1-knock out mice. Mol Cell Neurosci 2022; 123:103781. [PMID: 36122891 DOI: 10.1016/j.mcn.2022.103781] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 08/10/2022] [Accepted: 09/13/2022] [Indexed: 12/30/2022] Open
Abstract
The protein DBC1 is the main SIRT1 regulator known so far, and by doing so, it is involved in the regulation of energy metabolism, especially in liver and fat adipose tissue. DBC1 also has an important function in cell cycle progression and regulation in cancer cells, affecting tumorigenesis. We recently showed that during quiescence, non-transformed cells need DBC1 in order to re-enter and progress through the cell cycle. Moreover, we showed that deletion of DBC1 affects cell cycle progression during liver regeneration. This novel concept prompted us to evaluate the role of DBC1 during adult neurogenesis, where transition from quiescence to proliferation in neuronal progenitors is key and tightly regulated. Herein, we analyzed several markers of cell cycle expressed in the dentate gyrus of the hippocampus of controls and DBC1 KO adult mice. Our results suggest a reduced number of neuroblasts therein present, probably due to a decline of neuroblast generation or an impairment in neural differentiation. In agreement with this, we also found that adult DBC1 KO mice had a reduction in the volume of the granule cell layer of the dentate gyrus. Interestingly, behavioral analysis of KO and control mice revealed that deletion of DBC1 parallels to specific cognitive impairments, concerning learning and possibly memory formation. Our results show, for the first time, that DBC1 plays an active role in the nervous system. In particular, specific anatomical and behavioral changes are observed when is absent.
Collapse
Affiliation(s)
- Andrés Benítez-Rosendo
- Department of Biosciences, Facultad de Veterinaria, Universidad de la República (UdelaR), Montevideo, Uruguay; Laboratory of Metabolic Diseases and Aging, INDICyO Program, Institut Pasteur de Montevideo, Uruguay.
| | - Patricia Lagos
- Department of Physiology, Facultad de Medicina, Universidad de la República (UdelaR), Montevideo, Uruguay.
| | - Karina Cal
- Department of Biosciences, Facultad de Veterinaria, Universidad de la República (UdelaR), Montevideo, Uruguay; Laboratory of Metabolic Diseases and Aging, INDICyO Program, Institut Pasteur de Montevideo, Uruguay
| | - Laura Colman
- Laboratory of Metabolic Diseases and Aging, INDICyO Program, Institut Pasteur de Montevideo, Uruguay; Department of Organic Chemistry, Facultad de Química, Universidad de la República, (UdelaR), Montevideo, Uruguay.
| | - Carlos Escande
- Laboratory of Metabolic Diseases and Aging, INDICyO Program, Institut Pasteur de Montevideo, Uruguay.
| | - Aldo Calliari
- Department of Biosciences, Facultad de Veterinaria, Universidad de la República (UdelaR), Montevideo, Uruguay; Laboratory of Metabolic Diseases and Aging, INDICyO Program, Institut Pasteur de Montevideo, Uruguay.
| |
Collapse
|
12
|
Identification of sitagliptin binding proteins by affinity purification mass spectrometry. Acta Biochim Biophys Sin (Shanghai) 2022; 54:1453-1463. [PMID: 36239351 PMCID: PMC9827809 DOI: 10.3724/abbs.2022142] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) is recognized as a serious public health concern with increasing incidence. The dipeptidyl peptidase-4 (DPP-4) inhibitor sitagliptin has been used for the treatment of T2DM worldwide. Although sitagliptin has excellent therapeutic outcome, adverse effects are observed. In addition, previous studies have suggested that sitagliptin may have pleiotropic effects other than treating T2DM. These pieces of evidence point to the importance of further investigation of the molecular mechanisms of sitagliptin, starting from the identification of sitagliptin-binding proteins. In this study, by combining affinity purification mass spectrometry (AP-MS) and stable isotope labeling by amino acids in cell culture (SILAC), we discover seven high-confidence targets that can interact with sitagliptin. Surface plasmon resonance (SPR) assay confirms the binding of sitagliptin to three proteins, i. e., LYPLAL1, TCP1, and CCAR2, with binding affinities (K D) ranging from 50.1 μM to 1490 μM. Molecular docking followed by molecular dynamic (MD) simulation reveals hydrogen binding between sitagliptin and the catalytic triad of LYPLAL1, and also between sitagliptin and the P-loop of ATP-binding pocket of TCP1. Molecular mechanics Poisson-Boltzmann Surface Area (MMPBSA) analysis indicates that sitagliptin can stably bind to LYPLAL1 and TCP1 in active sites, which may have an impact on the functions of these proteins. SPR analysis validates the binding affinity of sitagliptin to TCP1 mutant D88A is ~10 times lower than that to the wild-type TCP1. Our findings provide insights into the sitagliptin-targets interplay and demonstrate the potential of sitagliptin in regulating gluconeogenesis and in anti-tumor drug development.
Collapse
|
13
|
Kim HJ, Moon SJ, Hong S, Won HH, Kim JH. DBC1 is a key positive regulator of enhancer epigenomic writers KMT2D and p300. Nucleic Acids Res 2022; 50:7873-7888. [PMID: 35801925 PMCID: PMC9371912 DOI: 10.1093/nar/gkac585] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 06/14/2022] [Accepted: 06/24/2022] [Indexed: 11/13/2022] Open
Abstract
Histone modification is a key epigenetic mechanism for regulation of chromatin dynamics and gene expression. Deleted in breast cancer 1 (DBC1) has been shown to act as a negative regulator of epigenetic modifiers and as a co-activator for nuclear receptors and other transcription factors. However, little is known about the role of DBC1 in the regulation of histone modifications and chromatin landscapes. Here, we analyzed genome-wide profiles of active enhancer and promoter marks in colorectal cancer cells and report DBC1 as a critical positive regulator of histone epigenetic writers KMT2D (H3K4 methyltransferase) and p300 (histone acetyltransferase). DBC1 is required for establishing the landscape of active enhancers, for genome-wide chromatin binding and enhancer recruitment of KMT2D and p300, and for gene activation involved in colorectal cancer progression. DBC1 interacts directly with KMT2D and p300, and enhances KMT2D-mediated histone H3K4 methylation (H3K4me1/2/3) and p300-mediated H3 acetylation. Importantly, DBC1 contributes to super-enhancer formation and function by facilitating the recruitment of KMT2D and p300 and by enhancing their functional interaction and cooperative cross-talk. Our results highlight the critical role of DBC1 as a key positive regulator of KMT2D and p300, and provide insights into regulatory mechanisms underlying the interplay between the enhancer epigenomic writers in enhancer activation.
Collapse
Affiliation(s)
- Hwa Jin Kim
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, 06351, South Korea.,Research Institute for Future Medicine, Samsung Medical Center, Seoul, 06351, South Korea
| | - Sue Jin Moon
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, 06351, South Korea.,Research Institute for Future Medicine, Samsung Medical Center, Seoul, 06351, South Korea
| | - Sanghoon Hong
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, 06351, South Korea
| | - Hong-Hee Won
- Research Institute for Future Medicine, Samsung Medical Center, Seoul, 06351, South Korea.,Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, 06351, South Korea
| | - Jeong Hoon Kim
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, 06351, South Korea.,Research Institute for Future Medicine, Samsung Medical Center, Seoul, 06351, South Korea
| |
Collapse
|
14
|
Sarwar Z, Nabi N, Bhat SA, Gillani SQ, Reshi I, Un Nisa M, Adelmant G, Marto J, Andrabi S. Interaction of DBC1 with polyoma small T antigen promotes its degradation and negatively regulates tumorigenesis. J Biol Chem 2021; 298:101496. [PMID: 34921839 PMCID: PMC8784333 DOI: 10.1016/j.jbc.2021.101496] [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: 05/15/2021] [Revised: 11/09/2021] [Accepted: 12/10/2021] [Indexed: 12/05/2022] Open
Abstract
Deleted in Breast Cancer 1 (DBC1) is an important metabolic sensor. Previous studies have implicated DBC1 in various cellular functions, notably cell proliferation, apoptosis, histone modification, and adipogenesis. However, current reports about the role of DBC1 in tumorigenesis are controversial and designate DBC1 alternatively as a tumor suppressor or a tumor promoter. In the present study, we report that polyoma small T antigen (PyST) associates with DBC1 in mammalian cells, and this interaction leads to the posttranslational downregulation of DBC1 protein levels. When coexpressed, DBC1 overcomes PyST-induced mitotic arrest and promotes the exit of cells from mitosis. Using both transient and stable modes of PyST expression, we also show that cellular DBC1 is subjected to degradation by LKB1, a tumor suppressor and cellular energy sensor kinase, in an AMP kinase-independent manner. Moreover, LKB1 negatively regulates the phosphorylation as well as activity of the prosurvival kinase AKT1 through DBC1 and its downstream pseudokinase substrate, Tribbles 3 (TRB3). Using both transient transfection and stable cell line approaches as well as soft agar assay, we demonstrate that DBC1 has oncogenic potential. In conclusion, our study provides insight into a novel signaling axis that connects LKB1, DBC1, TRB3, and AKT1. We propose that the LKB1–DBC1–AKT1 signaling paradigm may have an important role in the regulation of cell cycle and apoptosis and consequently tumorigenesis.
Collapse
Affiliation(s)
- Zarka Sarwar
- Department of Biochemistry, University of Kashmir, Srinagar, India, 190006
| | - Nusrat Nabi
- Department of Biochemistry, University of Kashmir, Srinagar, India, 190006
| | - Sameer Ahmed Bhat
- Department of Biochemistry, University of Kashmir, Srinagar, India, 190006
| | | | - Irfana Reshi
- Department of Biochemistry, University of Kashmir, Srinagar, India, 190006
| | - Misbah Un Nisa
- Department of Biochemistry, University of Kashmir, Srinagar, India, 190006
| | - Guillaume Adelmant
- Blais Proteomics Centre, Dana Farber Cancer Institute, Harvard University, Boston, USA
| | - Jarrod Marto
- Blais Proteomics Centre, Dana Farber Cancer Institute, Harvard University, Boston, USA
| | - Shaida Andrabi
- Department of Biochemistry, University of Kashmir, Srinagar, India, 190006.
| |
Collapse
|
15
|
He Q, Li Z, Lei X, Zou Q, Yu H, Ding Y, Xu G, Zhu W. The underlying molecular mechanisms and prognostic factors of RNA binding protein in colorectal cancer: a study based on multiple online databases. Cancer Cell Int 2021; 21:325. [PMID: 34193169 PMCID: PMC8244213 DOI: 10.1186/s12935-021-02031-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 06/19/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND RNA binding protein (RBP) is an active factor involved in the occurrence and development of colorectal cancer (CRC). Therefore, the potential mechanism of RBP in CRC needs to be clarified by dry-lab analyses or wet-lab experiments. METHODS The differential RBP gene obtained from the GEPIA 2 (Gene Expression Profiling Interactive Analysis 2) were performed functional enrichment analysis. Then, the alternative splicing (AS) events related to survival were acquired by univariate regression analysis, and the correlation between RBP and AS was analyzed by R software. The online databases were conducted to analyze the mutation and methylation of RBPs in CRC. Moreover, 5 key RBP signatures were obtained through univariate and multivariate Cox regression analysis and established as RBP prognosis model. Subsequently, the above model was verified through another randomized group of TCGA CRC cohorts. Finally, multiple online databases and qRT-PCR analysis were carried to further confirm the expression of the above 5 RBP signatures in CRC. RESULTS Through a comprehensive bioinformatics analysis, it was revealed that RBPs had genetic and epigenetic changes in CRC. We obtained 300 differentially expressed RBPs in CRC samples. The functional analysis suggested that they mainly participated in spliceosome. Then, a regulatory network for RBP was established to participate in AS and DDX39B was detected to act as a potentially essential factor in the regulation of AS in CRC. Our analysis discovered that 11 differentially expressed RBPs with a mutation frequency higher than 5%. Furthermore, we found that 10 differentially expressed RBPs had methylation sites related to the prognosis of CRC, and a prognostic model was constructed by the 5 RBP signatures. In another randomized group of TCGA CRC cohorts, the prognostic performance of the 5 RBP signatures was verified. CONCLUSION The potential mechanisms that regulate the aberrant expression of RBPs in the development of CRC was explored, a network that regulated AS was established, and the RBP-related prognosis model was constructed and verified, which could improve the individualized prognosis prediction of CRC.
Collapse
Affiliation(s)
- Qinglian He
- Department of Pathology, Guangdong Medical University, No.1 Xincheng Road, Dongguan, 523808, Guangdong Province, China
| | - Ziqi Li
- Department of Pathology, Guangdong Medical University, No.1 Xincheng Road, Dongguan, 523808, Guangdong Province, China
| | - Xue Lei
- Department of Pathology, Guangdong Medical University, No.1 Xincheng Road, Dongguan, 523808, Guangdong Province, China
| | - Qian Zou
- Department of Pathology, Guangdong Medical University, No.1 Xincheng Road, Dongguan, 523808, Guangdong Province, China
| | - Haibing Yu
- School of Public Health, Guangdong Medical University, Dongguan, 523808, Guangdong Province, China
| | - Yuanlin Ding
- School of Public Health, Guangdong Medical University, Dongguan, 523808, Guangdong Province, China
| | - Guangxian Xu
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, School of Medical Technology, Institute of Clinical Laboratory, Guangdong Medical University, Dongguan, 523808, Guangdong Province, China
| | - Wei Zhu
- Department of Pathology, Guangdong Medical University, No.1 Xincheng Road, Dongguan, 523808, Guangdong Province, China.
| |
Collapse
|
16
|
Johnson GS, Rajendran P, Dashwood RH. CCAR1 and CCAR2 as gene chameleons with antagonistic duality: Preclinical, human translational, and mechanistic basis. Cancer Sci 2020; 111:3416-3425. [PMID: 33403784 PMCID: PMC7540973 DOI: 10.1111/cas.14579] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 07/04/2020] [Accepted: 07/14/2020] [Indexed: 12/12/2022] Open
Abstract
Cell Cycle and Apoptosis Regulator 1 (CCAR1) and Cell Cycle and Apoptosis Regulator 2 (CCAR2) have emerged as key players in physiology and pathophysiology, with critical roles in the DNA damage response, nuclear receptor function, and Wnt signaling, among other activities. Contradictory reports exist on the functional duality of CCAR1 and CCAR2 as either tumor promoters or suppressors, suggesting that CCAR1 and CCAR2 have the hallmarks of gene chameleons. We review herein the mechanistic, preclinical, and human translational findings for CCAR1 and CCAR2, based on available RNA and protein expression data from human studies, The Cancer Genome Atlas (TCGA) data mining, gene knockout mouse models, and cell-based assays. Multiple factors contribute to the divergent activities of CCAR1 and CCAR2, including tissue type, mutation/genetic background, protein-protein interactions, dynamic regulation via posttranslational modifications, and alternative RNA splicing. An array of protein partners interact with CCAR1 and CCAR2 in the context of tumor promotion and suppression, including β-catenin, androgen receptor, p21Cip1/Waf1, tumor protein p53 (p53), sirtuin 1, and histone deacetylase 3. Genetic changes frequently found in cancer, such as TP53 mutation, also serve as critical determinants of survival outcomes in cancer patients. This review seeks to provide the impetus for further investigation into CCAR1 and CCAR2 as potential master regulators of metabolism, aging, and cancer.
Collapse
Affiliation(s)
- Gavin S. Johnson
- Center for Epigenetics & Disease PreventionTexas A&M Health Science CenterHoustonTXUSA
| | - Praveen Rajendran
- Center for Epigenetics & Disease PreventionTexas A&M Health Science CenterHoustonTXUSA
| | - Roderick H. Dashwood
- Center for Epigenetics & Disease PreventionTexas A&M Health Science CenterHoustonTXUSA
- Department of Translational Medical Sciences, Texas A&M College of MedicineTexas A&M UniversityHouston CampusTXUSA
- Department of Clinical Cancer PreventionThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| |
Collapse
|
17
|
Fang Q, Bellanti JA, Zheng SG. Advances on the role of the deleted in breast cancer (DBC1) in cancer and autoimmune diseases. J Leukoc Biol 2020; 109:449-454. [PMID: 32337788 DOI: 10.1002/jlb.6mr0320-086r] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/07/2020] [Accepted: 03/21/2020] [Indexed: 12/24/2022] Open
Abstract
DBC1 (deleted in breast cancer 1) is a human nuclear protein that modulates the activities of various proteins. Most of the research on DBC1 has focused on metabolism and epigenetics because it is a crucial endogenic inhibitor of deacetylase Sirtuin1 (SIRT1). In this review, we have discussed and summarized the new advances in DBC1 research, mostly focusing on its structure, regulatory function, and significance in cancer and autoimmune diseases.
Collapse
Affiliation(s)
- Qiannan Fang
- Department of Clinical Immunology, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.,Department of Internal Medicine, Ohio State University College of Medicine and Wexner Medical Center, Columbus, Ohio, USA
| | - Joseph A Bellanti
- Department of Pediatrics and Microbiology-Immunology, Georgetown University Medical Center, Washington, District of Columbia, USA
| | - Song Guo Zheng
- Department of Internal Medicine, Ohio State University College of Medicine and Wexner Medical Center, Columbus, Ohio, USA
| |
Collapse
|
18
|
Liu J, Meng C, Li C, Tang K, Tang H, Liao J. Deleted in Breast Cancer 1 as a Novel Prognostic Biomarker for Digestive System Cancers: A Meta-Analysis. J Cancer 2019; 10:1633-1641. [PMID: 31205519 PMCID: PMC6548013 DOI: 10.7150/jca.26935] [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: 04/27/2018] [Accepted: 01/19/2019] [Indexed: 11/06/2022] Open
Abstract
Deleted in Breast Cancer 1 (DBC1/CCAR2) is a regulatory protein involved in cell survival and cancer progression. Herein, we focused on summarizing the overall prognostic value of DBC1 for digestive system cancers. Therefore, we conducted a meta-analysis based on 9 studies with 2391 patients to generated combined hazard ratios (HR) or odds ratio (OR) with its 95% confidence intervals (CI) for overall survival (OS) and clinicopathological features. Positive DBC1 expression was significantly associated with poor OS of digestive system cancers (pooled HR=1.650, 95% CI=1.087-2.504, P<0.019). Stratified analysis also verified the potential prognostic prediction of DBC1 in some subgroups, such as digestive tract cancers (pooled HR=1.685, 95% CI=1.013-2.802, P=0.044), univariate analysis method (pooled HR=2.077, 95%CI=1.221-3.533, P=0.007), publication date within five years (pooled HR=1.609, 95%CI=1.097-2.358, P =0.015), study sample size smaller than 200 (pooled HR=2.304, 95%CI=1.716-3.093, P<0.001) and cutoff value for positive tumor cells more than 50% (pooled HR=1.944, 95% CI=1.479-2.556, P<0.001). Additionally, in terms of the association between DBC1 expression and clinicopathological characteristics, DBC1 expression was correlated to age (pooled OR=0.596, 95%CI =0.467-0.761, P<0.001), WHO classification (pooled OR =3.780, 95% CI=2.303-6.205, P <0.001), Lauren classification (pooled OR=2.000, 95%CI =1.492-2.680, P<0.001), and lymph node metastasis (pooled OR=0.405, 95%CI=0.203-0.806, P=0.010). In conclusion, DBC1 could not only be an independent prognostic factor for survival of patients with digestive system cancer, but might also be a novel target for cancer therapy.
Collapse
Affiliation(s)
- Jingting Liu
- Department of Emergency, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 Qingchun East Road, Hangzhou, Zhejiang, 310020, China
| | - Chunyan Meng
- Department of General Surgery, Zhejiang Hospital, 12 Lingyin Road, Zhejiang 310013, China
| | - Changcan Li
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine , 100 Haining Road, Shanghai 200080, China
| | - Kaifeng Tang
- Department of General Surgery, Zhejiang Hospital, 12 Lingyin Road, Zhejiang 310013, China
| | - Hongchao Tang
- Department of General Surgery, Zhejiang Hospital, 12 Lingyin Road, Zhejiang 310013, China
| | - Jianhua Liao
- Department of General Surgery, Zhejiang Hospital, 12 Lingyin Road, Zhejiang 310013, China
| |
Collapse
|
19
|
Gfi1b regulates the level of Wnt/β-catenin signaling in hematopoietic stem cells and megakaryocytes. Nat Commun 2019; 10:1270. [PMID: 30894540 PMCID: PMC6426870 DOI: 10.1038/s41467-019-09273-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 02/25/2019] [Indexed: 01/06/2023] Open
Abstract
Gfi1b is a transcriptional repressor expressed in hematopoietic stem cells (HSCs) and megakaryocytes (MKs). Gfi1b deficiency leads to expansion of both cell types and abrogates the ability of MKs to respond to integrin. Here we show that Gfi1b forms complexes with β-catenin, its co-factors Pontin52, CHD8, TLE3 and CtBP1 and regulates Wnt/β-catenin-dependent gene expression. In reporter assays, Gfi1b can activate TCF-dependent transcription and Wnt3a treatment enhances this activation. This requires interaction between Gfi1b and LSD1 and suggests that a tripartite β-catenin/Gfi1b/LSD1 complex exists, which regulates Wnt/β-catenin target genes. Consistently, numerous canonical Wnt/β-catenin target genes, co-occupied by Gfi1b, β-catenin and LSD1, have their expression deregulated in Gfi1b-deficient cells. When Gfi1b-deficient cells are treated with Wnt3a, their normal cellularity is restored and Gfi1b-deficient MKs regained their ability to spread on integrin substrates. This indicates that Gfi1b controls both the cellularity and functional integrity of HSCs and MKs by regulating Wnt/β-catenin signaling pathway. Gfi1b regulates cellularity of haematopoietic stem cells (HSCs) and megakaryocytes (MKs) as well as spreading of MKs on matrix. Here the authors show that Gfi1b regulates this behaviour by recruiting LSD1 and β-catenin to Wnt/β-catenin signalling targets.
Collapse
|
20
|
Liu G, Wu Q, Wang Y, Xiong Q, Fu F. Deleted in breast cancer 1 as a potential prognostic biomarker in human cancers: a pooled analysis of 2,254 patients. Onco Targets Ther 2019; 12:1563-1574. [PMID: 30863120 PMCID: PMC6390861 DOI: 10.2147/ott.s189618] [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] [Indexed: 11/23/2022] Open
Abstract
Background Deleted in breast cancer 1 (DBC1) is believed to be involved in human cancers. However, it is still uncertain whether DBC1 expression can be regarded as a prognostic factor in patients with various cancers. This meta-analysis aimed to evaluate the relationship between high levels of DBC1 and prognosis in tumor patients. Methods Electronic databases were searched and 14 studies meeting the selection criteria were included. Overall survival (OS), relapse-free survival (RFS), and 95% CIs were extracted and analyzed. HRs from individual studies were pooled using fixed-or random-effects models, depending on the heterogeneity of the included studies, and publication bias analyses were also performed to increase the reliability of the results. Results A total of 2,254 patients with tumors from 14 published studies were included in the meta-analysis. DBC1 overexpression was associated with worse OS (univariate analysis: HR=2.94; 95% CI: [2.38–3.63]; multivariate analysis: HR=1.98, 95% CI: [1.21–3.25]) and RFS (univariate analysis: HR=2.83, 95% CI: [2.30–3.49]; multivariate analysis: HR=2.71, 95% CI: [2.07–3.53]) for various tumors. No publication bias was observed according to test of funnel plot asymmetry and Egger’s test. Conclusion Current evidence supports the conclusion that the upregulation of DBC1 is correlated with poor survival among tumor patients, suggesting that DBC1 represents an independent prognostic factor significantly associated with OS and RFS, and could serve as a novel therapeutic target in patients with tumors. Nevertheless, further large-scale prospective trials and well-designed studies are warranted to confirm this finding.
Collapse
Affiliation(s)
- Gang Liu
- Department of Breast Surgery, The Third Hospital of Nanchang City, Key Laboratory of Breast Diseases, Nanchang, Jiangxi 330009, China, ;
| | - Qiaosheng Wu
- Department of Breast Surgery, The Third Hospital of Nanchang City, Key Laboratory of Breast Diseases, Nanchang, Jiangxi 330009, China, ;
| | - Yili Wang
- Department of Breast Surgery, The Third Hospital of Nanchang City, Key Laboratory of Breast Diseases, Nanchang, Jiangxi 330009, China, ;
| | - Qiuyun Xiong
- Department of Breast Surgery, The Third Hospital of Nanchang City, Key Laboratory of Breast Diseases, Nanchang, Jiangxi 330009, China, ;
| | - Feiguo Fu
- Department of Breast Surgery, The Third Hospital of Nanchang City, Key Laboratory of Breast Diseases, Nanchang, Jiangxi 330009, China, ;
| |
Collapse
|
21
|
Rajendran P, Johnson G, Li L, Chen YS, Dashwood M, Nguyen N, Ulusan A, Ertem F, Zhang M, Li J, Sun D, Huang Y, Wang S, Leung HC, Lieberman D, Beaver L, Ho E, Bedford M, Chang K, Vilar E, Dashwood R. Acetylation of CCAR2 Establishes a BET/BRD9 Acetyl Switch in Response to Combined Deacetylase and Bromodomain Inhibition. Cancer Res 2019; 79:918-927. [PMID: 30643017 DOI: 10.1158/0008-5472.can-18-2003] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 10/19/2018] [Accepted: 01/09/2019] [Indexed: 12/15/2022]
Abstract
There continues to be interest in targeting epigenetic "readers, writers, and erasers" for the treatment of cancer and other pathologies. However, a mechanistic understanding is frequently lacking for the synergy observed when combining deacetylase and bromodomain inhibitors. Here we identify cell cycle and apoptosis regulator 2 (CCAR2) as an early target for acetylation in colon cancer cells treated with sulforaphane. N-terminal acetylation of CCAR2 diminished its interactions with histone deacetylase 3 and β-catenin, interfering with Wnt coactivator functions of CCAR2, including in cells harboring genetically encoded CCAR2 acetylation. Protein domain arrays and pull-down assays identified acetyl "reader" proteins that recognized CCAR2 acetylation sites, including BRD9 and members of the bromodomain and extraterminal domain (BET) family. Treatment with the BET inhibitor JQ1 synergized with sulforaphane in colon cancer cells and suppressed tumor development effectively in a preclinical model of colorectal cancer. Studies with sulforaphane+JQ1 in combination implicated a BET/BRD9 acetyl switch and a shift in the pool of acetyl "reader" proteins in favor of BRD9-regulated target genes. SIGNIFICANCE: These results highlight the competition that exists among the "readers" of acetylated histone and nonhistone proteins and provide a mechanistic basis for potential new therapeutic avenues involving epigenetic combination treatments.
Collapse
Affiliation(s)
- Praveen Rajendran
- Center for Epigenetics & Disease Prevention, Texas A&M College of Medicine, Houston, Texas.
| | - Gavin Johnson
- Center for Epigenetics & Disease Prevention, Texas A&M College of Medicine, Houston, Texas
| | - Li Li
- Center for Epigenetics & Disease Prevention, Texas A&M College of Medicine, Houston, Texas
| | - Ying-Shiuan Chen
- Center for Epigenetics & Disease Prevention, Texas A&M College of Medicine, Houston, Texas
| | - Mohaiza Dashwood
- Center for Epigenetics & Disease Prevention, Texas A&M College of Medicine, Houston, Texas
| | - Nhung Nguyen
- Center for Epigenetics & Disease Prevention, Texas A&M College of Medicine, Houston, Texas
| | - Ahmet Ulusan
- Center for Epigenetics & Disease Prevention, Texas A&M College of Medicine, Houston, Texas
| | - Furkan Ertem
- Center for Epigenetics & Disease Prevention, Texas A&M College of Medicine, Houston, Texas
| | - Mutian Zhang
- Center for Epigenetics & Disease Prevention, Texas A&M College of Medicine, Houston, Texas
| | - Jia Li
- Center for Epigenetics & Disease Prevention, Texas A&M College of Medicine, Houston, Texas
| | - Deqiang Sun
- Center for Epigenetics & Disease Prevention, Texas A&M College of Medicine, Houston, Texas
| | - Yun Huang
- Center for Epigenetics & Disease Prevention, Texas A&M College of Medicine, Houston, Texas
| | - Shan Wang
- Center for Epigenetics & Disease Prevention, Texas A&M College of Medicine, Houston, Texas
| | - Hon-Chiu Leung
- Mass Spectrometry-Proteomics Core, Baylor College of Medicine, Houston, Texas
| | - David Lieberman
- Division of Gastroenterology and Hepatology, Oregon Health & Science University, Portland, Oregon
| | - Laura Beaver
- College of Public Health and Human Sciences, Oregon State University, Corvallis, Oregon
| | - Emily Ho
- College of Public Health and Human Sciences, Oregon State University, Corvallis, Oregon
| | - Mark Bedford
- The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kyle Chang
- The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Eduardo Vilar
- The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Roderick Dashwood
- Center for Epigenetics & Disease Prevention, Texas A&M College of Medicine, Houston, Texas. .,The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
22
|
Yu X, Wang M, Han Q, Zhang X, Mao X, Wang X, Li X, Ma W, Jin F. ZNF326 promotes a malignant phenotype of breast cancer by interacting with DBC1. Mol Carcinog 2018; 57:1803-1815. [PMID: 30175866 DOI: 10.1002/mc.22898] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2018] [Accepted: 08/28/2018] [Indexed: 12/30/2022]
Affiliation(s)
- Xinmiao Yu
- Department of Breast Surgery; The First Hospital of China Medical University; Shenyang China
| | - Minghao Wang
- Department of Neurosurgery; The First Hospital of China Medical University; Shenyang China
| | - Qiang Han
- Department of Pathology; College of Basic Medical Sciences and The First Hospital; China Medical University; Shenyang China
| | - Xiupeng Zhang
- Department of Pathology; College of Basic Medical Sciences and The First Hospital; China Medical University; Shenyang China
| | - Xiaoyun Mao
- Department of Breast Surgery; The First Hospital of China Medical University; Shenyang China
| | - Xu Wang
- Department of Breast Surgery; The First Hospital of China Medical University; Shenyang China
| | - Xiaoying Li
- Department of Breast Surgery; The First Hospital of China Medical University; Shenyang China
| | - Wei Ma
- Department of Breast Surgery; The First Hospital of China Medical University; Shenyang China
| | - Feng Jin
- Department of Breast Surgery; The First Hospital of China Medical University; Shenyang China
| |
Collapse
|
23
|
Kim HJ, Moon SJ, Kim SH, Heo K, Kim JH. DBC1 regulates Wnt/β-catenin-mediated expression of MACC1, a key regulator of cancer progression, in colon cancer. Cell Death Dis 2018; 9:831. [PMID: 30082743 PMCID: PMC6079074 DOI: 10.1038/s41419-018-0899-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 07/19/2018] [Accepted: 07/20/2018] [Indexed: 12/22/2022]
Abstract
Metastasis-associated in colon cancer 1 (MACC1) has been reported to be overexpressed in multiple cancers and promote proliferation, metastasis, cancer stem cell-like properties, and drug resistance of cancer cells. Despite its significance and the considerable knowledge accumulated on the function of MACC1 in various types of human malignancies, regulatory mechanisms underlying MACC1 expression remain unclear. Here we report that MACC1 is a direct target of Wnt/β-catenin signaling pathway in colon cancer cells and that DBC1 functions as a coactivator for Wnt-mediated MACC1 expression by promoting the activity of a LEF1/β-catenin-dependent enhancer located in intron 1 of MACC1 gene. DBC1 is required for LEF1/β-catenin complex formation on the MACC1 enhancer and for long-distance enhancer-promoter interaction of the MACC1 locus. MACC1 expression was increased in colonosphere cells compared to adherent colon cancer cells, and DBC1 overexpression further increased MACC1 expression in colonospheres and promoted sphere-forming abilities of colon cancer cells and drug resistance of colonospheres. Importantly, expressions of MACC1 and DBC1 are positively correlated with each other, upregulated in high-risk groups of colorectal cancer patients, and associated with poor survival. Our results establish MACC1 as a transcriptional target of Wnt/β-catenin signaling and suggest that DBC1 plays a key role in colorectal cancer progression through Wnt/β-catenin-MACC1 signaling axis.
Collapse
Affiliation(s)
- Hwa Jin Kim
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, 06351, Korea.,Department of Biomedical Sciences, Samsung Biomedical Research Institute, Samsung Medical Center, Seoul, 06351, Korea
| | - Sue Jin Moon
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, 06351, Korea.,Department of Biomedical Sciences, Samsung Biomedical Research Institute, Samsung Medical Center, Seoul, 06351, Korea
| | - Seok-Hyung Kim
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, 06351, Korea.,Department of Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Korea
| | - Kyu Heo
- Department of Clinical Research, Dongnam Institute of Radiological and Medical Sciences, Busan, 46033, Korea
| | - Jeong Hoon Kim
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, 06351, Korea. .,Department of Biomedical Sciences, Samsung Biomedical Research Institute, Samsung Medical Center, Seoul, 06351, Korea.
| |
Collapse
|
24
|
Magni M, Buscemi G, Zannini L. Cell cycle and apoptosis regulator 2 at the interface between DNA damage response and cell physiology. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2018; 776:1-9. [DOI: 10.1016/j.mrrev.2018.03.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 03/16/2018] [Accepted: 03/17/2018] [Indexed: 01/06/2023]
|
25
|
Wang ZP, Lu H. Clinical significance of expression of deleted in breast cancer-1 in human gastric cancer. Shijie Huaren Xiaohua Zazhi 2018; 26:150-158. [DOI: 10.11569/wcjd.v26.i3.150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM To detect the expression of deleted in breast cancer-1 (DBC1) in gastric cancer (GC) and analyze its potential clinical significance.
METHODS The expression of DBC-1 in normal gastric mucosa cells (GSE-1) and GC cells (SGC-7901 and BGC-823) was detected by RT-PCR. Immunohistochemistry was performed to detect the expression of DBC1 in 201 cases of GC and matched tumor-adjacent non-tumor tissues. The correlation between DBC1 expression and clinicopathologic features was analyzed.
RESULTS The relative expression level of DBC1 in GC cells was significantly higher than that in normal gastric mucosa cells (P < 0.05). A higher positive rate of DBC1 was detected in GC tissues than in non-cancer tissues (75.12% vs 20.90%, P < 0.05). The expression level of DBC1 was correlated with Bormann type (P < 0.05), tumor size (P < 0.05), Lauren type (P < 0.05), tumor differentiation (P < 0.05), vessel invasion (P < 0.05), perinerural invasion (P < 0.05), and TNM stage (P < 0.05), but there was no difference between DBC1 expression and gender (P > 0.05) or age (P > 0.05). Kaplan-Meier and Cox analyses showed that DBC1 expression was associated with shorter disease-free survival (P < 0.05) and overall survival (P < 0.05).
CONCLUSION DBC1 is overexpressed in GC and can act as a prognostic factor in GC. These findings indicate that DBC1 may play an important role in the development of GC.
Collapse
Affiliation(s)
- Zhao-Ping Wang
- Department of General Surgery, the First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121000, Liaoning Province, China
| | - Hang Lu
- Department of General Surgery, the First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121000, Liaoning Province, China
| |
Collapse
|
26
|
Abstract
Mammalian silent information regulator 1 (SIRT1) is reported to play a role in cancers of the secretory organs, including thyroid, pancreatic endocrine, and ovarian tumors [1, 2, 3, 4]. A recent meta-analysis conducted on 37 selected studies of human cancers analyzed the correlations of overall survival (OS), disease-free survival (DFS) and relapse-free survival (RFS) with SIRT1 expression [5]. This study reported that SIRT1 overexpression was associated with a worse OS in liver and lung cancers, while it was not correlated with OS in breast cancer, colorectal cancer, or gastric carcinoma. Collectively, the meta-analysis revealed that an unfavorable OS was associated with SIRT1 expression for solid malignancies. Given the growing importance of this class of lysine/histone deacetylases in human endocrine malignancies, a rational and focused literature assessment is desirable in light of future clinical translations.
Collapse
|
27
|
Moon SJ, Jeong BC, Kim HJ, Lim JE, Kwon GY, Kim JH. DBC1 promotes castration-resistant prostate cancer by positively regulating DNA binding and stability of AR-V7. Oncogene 2017; 37:1326-1339. [PMID: 29249800 DOI: 10.1038/s41388-017-0047-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 09/20/2017] [Accepted: 10/09/2017] [Indexed: 12/21/2022]
Abstract
Constitutively active AR-V7, one of the major androgen receptor (AR) splice variants lacking the ligand-binding domain, plays a key role in the development of castration-resistant prostate cancer (CRPC) and anti-androgen resistance. However, our understanding of the regulatory mechanisms of AR-V7-driven transcription is limited. Here we report DBC1 as a key regulator of AR-V7 transcriptional activity and stability in CRPC cells. DBC1 functions as a coactivator for AR-V7 and is required for the expression of AR-V7 target genes including CDH2, a mesenchymal marker linked to CRPC progression. DBC1 is required for recruitment of AR-V7 to its target enhancers and for long-range chromatin looping between the CDH2 enhancer and promoter. Mechanistically, DBC1 enhances DNA-binding activity of AR-V7 by direct interaction and inhibits CHIP E3 ligase-mediated ubiquitination and degradation of AR-V7 by competing with CHIP for AR-V7 binding, thereby stabilizing and activating AR-V7. Importantly, DBC1 depletion suppresses the tumorigenic and metastatic properties of CRPC cells. Our results firmly establish DBC1 as a critical AR-V7 coactivator that plays a key role in the regulation of DNA binding and stability of AR-V7 and has an important physiological role in CRPC progression.
Collapse
Affiliation(s)
- Sue Jin Moon
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, Korea.,Department of Biomedical Sciences, Samsung Biomedical Research Institute, Samsung Medical Center, Seoul, Korea
| | - Byong Chang Jeong
- Department of Urology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hwa Jin Kim
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, Korea.,Department of Biomedical Sciences, Samsung Biomedical Research Institute, Samsung Medical Center, Seoul, Korea
| | - Joung Eun Lim
- Department of Urology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Ghee Young Kwon
- Department of Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jeong Hoon Kim
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, Korea. .,Department of Biomedical Sciences, Samsung Biomedical Research Institute, Samsung Medical Center, Seoul, Korea.
| |
Collapse
|
28
|
Kim W, Pyo J, Noh BJ, Jeong JW, Lee J, Kim JE. CCAR2 negatively regulates IL-8 production in cervical cancer cells. Oncotarget 2017; 9:1143-1155. [PMID: 29416683 PMCID: PMC5787426 DOI: 10.18632/oncotarget.23199] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 10/27/2017] [Indexed: 01/10/2023] Open
Abstract
Cell cycle and apoptosis regulator 2 (CCAR2) is a multifaceted protein that controls diverse cellular functions; however, its function in cancer is unclear. To better understand its potential role in cancer, we examined gene expression patterns regulated by CCAR2 in cervical cancer cells. Cytokine and chemokine production by CCAR2-deficient cells increased under oxidative conditions. In particular, H2O2-treated CCAR2-depleted cells showed a significant increase in interleukin-8 (IL-8) production, indicating a negative regulation of IL-8 by CCAR2. Upregulation of IL-8 expression in CCAR2-deficient cells occurred via activation of transcription factor AP-1. The negative correlation between CCAR2 and IL-8 expression was confirmed by examining mRNA and protein levels in tissues from cervical cancer patients. Furthermore, CCAR2-regulated IL-8 expression is associated with a shorter survival of cervical cancer patients. Overall, the data suggest that CCAR2 plays a critical role in controlling both the cancer secretome and cancer progression.
Collapse
Affiliation(s)
- Wootae Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Jaehyuk Pyo
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Byeong-Joo Noh
- Department of Pathology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Joo-Won Jeong
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea.,Department of Anatomy and Neurobiology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Juhie Lee
- Department of Pathology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Ja-Eun Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea.,Department of Pharmacology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
29
|
Li C, Liao J, Wu S, Fan J, Peng Z, Wang Z. Overexpression of DBC1, correlated with poor prognosis, is a potential therapeutic target for hepatocellular carcinoma. Biochem Biophys Res Commun 2017; 494:511-517. [PMID: 29106957 DOI: 10.1016/j.bbrc.2017.10.134] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 10/25/2017] [Indexed: 11/26/2022]
Abstract
Deleted in Breast Cancer 1 (DBC1) is a regulatory protein involved in cell metabolism and cancer progression. Nevertheless, the expression and prognostic values of DBC1 in hepatocellular carcinoma (HCC) are still not well understood. The following study investigated the clinical significance and biological function of DBC1 in HCC. Briefly, overexpression of DBC1 at transcriptional and translational levels in human HCC tissues compared to adjacent normal tissues was observed using quantitative real-time polymerase chain reaction (qRT-PCR), western blot (WB) and immunohistochemistry (IHC) approach. Furthermore, upregulated DBC1 was significantly correlated with tumor size (p = 0.005), N stage (p = 0.016), M stage (p = 0.011), tumor differentiation (p < 0.001), and American Joint Committee on Cancer (AJCC) stage (p = 0.001). Moreover, Kaplan-Meier survival analysis revealed that DBC1 was an independent prognosis predictor for disease-free survival (DFS) (p < 0.001) and overall survival (OS) (p < 0.001). In addition, by using Cell Counting Kit-8 (CCK8) assays and colony formation assays, we found that the knockdown of DBC1 significantly suppressed the proliferation of HCC cells in vitro. To conclude, these findings demonstrated that DBC1 was essential in tumorigenesis and proliferation. Moreover, it was identified as a potential therapeutic target for HCC.
Collapse
Affiliation(s)
- Changcan Li
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China.
| | - Jianhua Liao
- Department of General Surgery, Zhejiang Hospital, Hangzhou 310013, China.
| | - Shaohan Wu
- Department of General Surgery, The Second Affiliated Hospital of Jiaxing College, Jiaxing 314000, China
| | - Junwei Fan
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Zhihai Peng
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Zhaowen Wang
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China.
| |
Collapse
|
30
|
Johnson GS, Li J, Beaver LM, Dashwood WM, Sun D, Rajendran P, Williams DE, Ho E, Dashwood RH. A functional pseudogene, NMRAL2P, is regulated by Nrf2 and serves as a coactivator of NQO1 in sulforaphane-treated colon cancer cells. Mol Nutr Food Res 2017; 61. [PMID: 27860235 DOI: 10.1002/mnfr.201600769] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 11/11/2016] [Accepted: 11/11/2016] [Indexed: 12/13/2022]
Abstract
SCOPE The anticancer agent sulforaphane (SFN) acts via multiple mechanisms to modulate gene expression, including the induction of nuclear factor (erythroid-derived 2)-like 2 (Nrf2)-dependent signaling and the inhibition of histone deacetylase activity. Transcriptomics studies were performed in SFN-treated human colon cancer cells and in nontransformed colonic epithelial cells in order to pursue new mechanistic leads. METHODS AND RESULTS RNA-sequencing corroborated the expected changes in cancer-related pathways after SFN treatment. In addition to NAD(P)H quinone dehydrogenase 1 (NQO1) and other well-known Nrf2-dependent targets, SFN strongly induced the expression of Loc344887. This noncoding RNA was confirmed as a novel functional pseudogene for NmrA-like redox sensor 1, and was given the name NmrA-like redox sensor 2 pseudogene (NMRAL2P). Chromatin immunoprecipitation experiments corroborated the presence of Nrf2 interactions on the NMRAL2P genomic region, and interestingly, NMRAL2P also served as a coregulator of NQO1 in human colon cancer cells. Silencing of NMRAL2P via CRISPR/Cas9 genome-editing protected against SFN-mediated inhibition of cancer cell growth, colony formation, and migration. CONCLUSION NMRAL2P is the first functional pseudogene to be identified both as a direct transcriptional target of Nrf2, and as a downstream regulator of Nrf2-dependent NQO1 induction. Further studies are warranted on NMRAL2P-Nrf2 crosstalk and the associated mechanisms of gene regulation.
Collapse
Affiliation(s)
- Gavin S Johnson
- Center for Epigenetics & Disease Prevention, Institute of Biosciences & Technology, Texas A&M Health Science Center, Houston, TX, USA
| | - Jia Li
- Center for Epigenetics & Disease Prevention, Institute of Biosciences & Technology, Texas A&M Health Science Center, Houston, TX, USA
| | - Laura M Beaver
- Linus Pauling Institute, Oregon State University, Corvallis, OR, USA.,School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR, USA
| | - W Mohaiza Dashwood
- Center for Epigenetics & Disease Prevention, Institute of Biosciences & Technology, Texas A&M Health Science Center, Houston, TX, USA
| | - Deqiang Sun
- Center for Epigenetics & Disease Prevention, Institute of Biosciences & Technology, Texas A&M Health Science Center, Houston, TX, USA
| | - Praveen Rajendran
- Center for Epigenetics & Disease Prevention, Institute of Biosciences & Technology, Texas A&M Health Science Center, Houston, TX, USA
| | - David E Williams
- Linus Pauling Institute, Oregon State University, Corvallis, OR, USA.,Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR, USA
| | - Emily Ho
- Linus Pauling Institute, Oregon State University, Corvallis, OR, USA.,School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR, USA
| | - Roderick H Dashwood
- Center for Epigenetics & Disease Prevention, Institute of Biosciences & Technology, Texas A&M Health Science Center, Houston, TX, USA.,Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of Nutrition and Food Science, Texas A&M University, College Station, TX, USA.,Department of Molecular and Cellular Medicine, Texas A&M College of Medicine, College Station, TX, USA
| |
Collapse
|
31
|
Restelli M, Magni M, Ruscica V, Pinciroli P, De Cecco L, Buscemi G, Delia D, Zannini L. A novel crosstalk between CCAR2 and AKT pathway in the regulation of cancer cell proliferation. Cell Death Dis 2016; 7:e2453. [PMID: 27809307 PMCID: PMC5260903 DOI: 10.1038/cddis.2016.359] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 09/29/2016] [Accepted: 10/04/2016] [Indexed: 02/06/2023]
Abstract
Human CCAR2 has recently emerged as having a pivotal role in the DNA damage response, promoting apoptosis and repair of heterochromatic DNA breaks. However, less is known about the function of CCAR2 in tumor formation and cancer progression. Here, we demonstrate, for the first time, that CCAR2 loss inhibits the proliferation of cancer cells, but preserves the growth of normal cells. Investigating the mechanisms responsible for this differential effect, we found that CCAR2 depletion specifically impairs the activation of AKT pathway in cancer cells, but not in normal cells, by reducing AKT phosphorylation on Ser473. This effect is achieved through the transcriptional upregulation of TRB3 gene and accumulation of TRB3 protein, which then binds to and inhibits the phosphorylation and activation of AKT. The defective activation of AKT finally results in reduced GSK3β phosphorylation, prevention of G1/S transition and inhibition of cancer cell growth. These results establish an important role for CCAR2 in cancer cells proliferation and could shed new light on novel therapeutic strategies against cancer, devoid of detrimental side effects.
Collapse
Affiliation(s)
- Michela Restelli
- Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, Milan 20133, Italy
| | - Martina Magni
- Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, Milan 20133, Italy
| | - Vincenzo Ruscica
- Department of Biochemistry, Max Planck Institute for Developmental Biology, Tubingen, Germany
| | - Patrizia Pinciroli
- Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, Milan 20133, Italy
| | - Loris De Cecco
- Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, Milan 20133, Italy
| | - Giacomo Buscemi
- Department of Biosciences, University of Milan, via Celoria 26, Milan 20133, Italy
| | - Domenico Delia
- Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, Milan 20133, Italy
| | - Laura Zannini
- Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, Milan 20133, Italy
| |
Collapse
|
32
|
Best SA, Nwaobasi AN, Schmults CD, Ramsey MR. CCAR2 Is Required for Proliferation and Tumor Maintenance in Human Squamous Cell Carcinoma. J Invest Dermatol 2016; 137:506-512. [PMID: 27725203 DOI: 10.1016/j.jid.2016.09.027] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 09/21/2016] [Accepted: 09/26/2016] [Indexed: 02/09/2023]
Abstract
CCAR2 is a widely expressed protein involved in the regulation of a variety of transcriptional complexes. High expression of CCAR2 correlates with poor outcomes in many human tumor types such as squamous cell carcinoma (SCC). Paradoxically, loss of Ccar2 in the mouse results in an increased tumor burden, suggesting that CCAR2 may in fact function as a tumor suppressor. This tumor suppressor function is dependent on p53, a protein that is inactivated in the vast majority of SCC tumors, leaving the role of CCAR2 in p53-null tumors unclear. We sought to identify p53-independent CCAR2 functions in SCC and to examine its role in tumorigenesis. We found that CCAR2 is highly overexpressed in p53-deficient SCC cell lines compared with normal primary keratinocytes due to increased protein stability. We identify a role for CCAR2 in promoting the stability of the transcription factors RFX1 and CREB1, which are both required for proliferation. Finally, we show that CCAR2 is required for proliferation in vitro and in established SCC tumors in vivo. Our data suggest an important role for CCAR2 in maintaining cell cycle progression and promoting SCC tumorigenesis.
Collapse
Affiliation(s)
- Sarah A Best
- Brigham and Women's Hospital, Boston, Massachusetts, USA; Harvard Medical School, Department of Dermatology, Boston, Massachusetts, USA
| | - Amy N Nwaobasi
- Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Chrysalyne D Schmults
- Brigham and Women's Hospital, Boston, Massachusetts, USA; Harvard Medical School, Department of Dermatology, Boston, Massachusetts, USA
| | - Matthew R Ramsey
- Brigham and Women's Hospital, Boston, Massachusetts, USA; Harvard Medical School, Department of Dermatology, Boston, Massachusetts, USA.
| |
Collapse
|
33
|
Saukkonen K, Hagström J, Mustonen H, Juuti A, Nordling S, Kallio P, Alitalo K, Seppänen H, Haglund C. PROX1 and β-catenin are prognostic markers in pancreatic ductal adenocarcinoma. BMC Cancer 2016; 16:472. [PMID: 27411302 PMCID: PMC4944261 DOI: 10.1186/s12885-016-2497-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 06/28/2016] [Indexed: 12/27/2022] Open
Abstract
Background The Wnt/β-catenin pathway has a key role in regulating cellular processes and its aberrant signaling can lead to cancer development. The role of β-catenin expression in pancreatic ductal adenocarcinoma is somewhat controversial. Transcription factor PROX1 is a target of Wnt/β-catenin signaling and it is involved in carcinogenesis through alterations in its expression. The actions can be either oncogenic or tumor suppressive depending on the tissue. The aim of this study was to investigate PROX1 and β-catenin expression in pancreatic ductal adenocarcinoma (PDAC). Methods Expression of PROX1 and β-catenin were evaluated in 156 patients by immunohistochemistry of tissue microarrays. Associations between tumor marker expression and clinicopathological parameters were assessed by the Fischer’s exact-test or the linear-by-linear association test. The Kaplan-Meier method and log-rank test were used for survival analysis. Uni- and multivariate survival analyses were carried out by the Cox regression proportional hazard model. Results High PROX1 expression was seen in 74 (48 %) tumors, and high β-catenin expression in 100 (65 %). High β-catenin expression was associated with lower tumor grade (p = 0.025). High PROX1 and β-catenin expression associated significantly with lower risk of death from PDAC in multivariate analysis (HR = 0.63; 95 % CI 0.42–0.95, p = 0.026; and HR = 0.54; 95 % CI 0.35–0.82, p = 0.004; respectively). The combined high expression of PROX1 and β-catenin also predicted lower risk of death from PDAC (HR = 0.46; 95 % CI 0.28–0.76, p = 0.002). Conclusion In conclusion, high PROX1 and β-catenin expression were independent factors for better prognosis in pancreatic ductal adenocarcinoma. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2497-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kapo Saukkonen
- Department of Surgery, University of Helsinki and Helsinki University Hospital, P.O. Box 440, FIN-00029 HUS, Helsinki, Finland. .,Research Programs Unit, Translational Cancer Biology, University of Helsinki, P.O. Box 63, Helsinki, FIN-00014, Finland.
| | - Jaana Hagström
- Research Programs Unit, Translational Cancer Biology, University of Helsinki, P.O. Box 63, Helsinki, FIN-00014, Finland.,Department of Pathology, Haartman Institute and HUSLAB, University of Helsinki and Helsinki University Hospital, Helsinki, FIN-00014, Finland
| | - Harri Mustonen
- Department of Surgery, University of Helsinki and Helsinki University Hospital, P.O. Box 440, FIN-00029 HUS, Helsinki, Finland
| | - Anne Juuti
- Department of Surgery, University of Helsinki and Helsinki University Hospital, P.O. Box 440, FIN-00029 HUS, Helsinki, Finland
| | - Stig Nordling
- Department of Pathology, Haartman Institute and HUSLAB, University of Helsinki and Helsinki University Hospital, Helsinki, FIN-00014, Finland
| | - Pauliina Kallio
- Research Programs Unit, Translational Cancer Biology, University of Helsinki, P.O. Box 63, Helsinki, FIN-00014, Finland
| | - Kari Alitalo
- Research Programs Unit, Translational Cancer Biology, University of Helsinki, P.O. Box 63, Helsinki, FIN-00014, Finland
| | - Hanna Seppänen
- Department of Surgery, University of Helsinki and Helsinki University Hospital, P.O. Box 440, FIN-00029 HUS, Helsinki, Finland
| | - Caj Haglund
- Department of Surgery, University of Helsinki and Helsinki University Hospital, P.O. Box 440, FIN-00029 HUS, Helsinki, Finland.,Research Programs Unit, Translational Cancer Biology, University of Helsinki, P.O. Box 63, Helsinki, FIN-00014, Finland
| |
Collapse
|