1
|
Si Y, Ou H, Jin X, Gu M, Sheng S, Peng W, Yang D, Zhan X, Zhang L, Yu Q, Liu X, Liu Y. G protein pathway suppressor 2 suppresses aerobic glycolysis through RACK1-mediated HIF-1α degradation in breast cancer. Free Radic Biol Med 2024; 222:478-492. [PMID: 38942092 DOI: 10.1016/j.freeradbiomed.2024.06.021] [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: 03/25/2024] [Revised: 06/16/2024] [Accepted: 06/25/2024] [Indexed: 06/30/2024]
Abstract
Aerobic glycolysis has been recognized as a hallmark of human cancer. G protein pathway suppressor 2 (GPS2) is a negative regulator of the G protein-MAPK pathway and a core subunit of the NCoR/SMRT transcriptional co-repressor complex. However, how its biological properties intersect with cellular metabolism in breast cancer (BC) development remains poorly elucidated. Here, we report that GPS2 is low expressed in BC tissues and negatively correlated with poor prognosis. Both in vitro and in vivo studies demonstrate that GPS2 suppresses malignant progression of BC. Moreover, GPS2 suppresses aerobic glycolysis in BC cells. Mechanistically, GPS2 destabilizes HIF-1α to reduce the transcription of its downstream glycolytic regulators (PGK1, PGAM1, ENO1, PKM2, LDHA, PDK1, PDK2, and PDK4), and then suppresses cellular aerobic glycolysis. Notably, receptor for activated C kinase 1 (RACK1) is identified as a key ubiquitin ligase for GPS2 to promote HIF-1α degradation. GPS2 stabilizes the binding of HIF-1α to RACK1 by directly binding to RACK1, resulting in polyubiquitination and instability of HIF-1α. Amino acid residues 70-92 aa of the GPS2 N-terminus bind RACK1. A 23-amino-acid-long GPS2-derived peptide was developed based on this N-terminal region, which promotes the interaction of RACK1 with HIF-1α, downregulates HIF-1α expression and significantly suppresses BC tumorigenesis in vitro and in vivo. In conclusion, our findings indicate that GPS2 decreases the stability of HIF-1α, which in turn suppresses aerobic glycolysis and tumorigenesis in BC, suggesting that targeting HIF-1α degradation and treating with peptides may be a promising approach to treat BC.
Collapse
Affiliation(s)
- Yuan Si
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, Hubei, China.
| | - Hongling Ou
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, Hubei, China
| | - Xin Jin
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Institute of Modern Biology, Nanjing University, Nanjing, Jiangsu, China
| | - Manxiang Gu
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, Hubei, China
| | - Songran Sheng
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, Hubei, China
| | - Wenkang Peng
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, Hubei, China
| | - Dan Yang
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, Hubei, China
| | - Xiangrong Zhan
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, Hubei, China
| | - Liang Zhang
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, Hubei, China
| | - Qingqing Yu
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, Hubei, China
| | - Xuewen Liu
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, Hubei, China; Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, Hubei, China.
| | - Ying Liu
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, Hubei, China; Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, Hubei, China.
| |
Collapse
|
2
|
Kluth M, Hitzschke M, Furlano K, Plage H, Hofbauer S, Weinberger S, Ralla B, Fendler A, de Martino M, Roßner F, Schallenberg S, Elezkurtaj S, Lennartz M, Marx AH, Samtleben H, Fisch M, Rink M, Slojewski M, Kaczmarek K, Ecke T, Koch S, Adamini N, Weischenfeldt J, Klatte T, Minner S, Simon R, Sauter G, Schlomm T, Horst D, Zecha H. 17p13 (TP53) Deletions Are Associated With an Aggressive Phenotype but Unrelated to Patient Prognosis in Urothelial Bladder Carcinomas. Genes Chromosomes Cancer 2024; 63:e23271. [PMID: 39324446 DOI: 10.1002/gcc.23271] [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: 04/29/2024] [Revised: 08/24/2024] [Accepted: 09/05/2024] [Indexed: 09/27/2024] Open
Abstract
17p13 deletions including TP53 and other genes represent a common cause for reduced/lost p53 function in tumor cells. In this study, we analyzed the impact of 17p13 (TP53) deletions and p53 expression on tumor aggressiveness and patient prognosis in urothelial carcinoma. The 17p13 copy number status was analyzed by fluorescence in situ hybridization (FISH) on more than 2700 urothelial bladder carcinomas in a tissue microarray format. 17p13 deletion data were compared to p53 expression data measured by immunohistochemistry (IHC) in a previous study. Different types of p53 alterations were compared with tumor phenotype and clinical outcome data. Deletions of 17p13 occurred in 23% of 2185 analyzable carcinomas. The fraction of tumors with 17p13 deletions increased from pTa G2 low (9%) to pTa G3 (24%, p < 0.0001). In muscle-invasive carcinomas, 17p13 deletions were associated with advanced pT stage (p = 0.0246), but unrelated to patient prognosis (p > 0.5). 17p13 deletions were significantly related to p53 immunostaining (p = 0.0375). 17p13 deletions were most common in tumors with complete lack of p53 staining (31%), which supports the concept that many of these tumors have a complete loss of p53 function (p53 null phenotype). 17p13 deletions were also increased in tumors with high p53 staining (25%). In conclusion, 17p13 deletions were most commonly seen in p53 negative cancers, supporting their role as a cause for the p53 null phenotype in urothelial cancer. The association of 17p13 deletions with high grade and advanced pT stage may reflect increasing genomic instability going along with stage and grade progression.
Collapse
Affiliation(s)
- Martina Kluth
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Melanie Hitzschke
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kira Furlano
- Department of Urology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Henning Plage
- Department of Urology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Sebastian Hofbauer
- Department of Urology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Sarah Weinberger
- Department of Urology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Bernhard Ralla
- Department of Urology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Annika Fendler
- Department of Urology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Michela de Martino
- Department of Urology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Florian Roßner
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Simon Schallenberg
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Sefer Elezkurtaj
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Maximilian Lennartz
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Andreas H Marx
- Department of Pathology, Academic Hospital Fuerth, Fuerth, Germany
| | - Henrik Samtleben
- Department of Pathology, Academic Hospital Fuerth, Fuerth, Germany
| | - Margit Fisch
- Department of Urology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Michael Rink
- Department of Urology, Marienhospital Hamburg, Hamburg, Germany
| | - Marcin Slojewski
- Department of Urology and Urological Oncology, Pomeranian Medical University, Szczecin, Poland
| | - Krystian Kaczmarek
- Department of Urology and Urological Oncology, Pomeranian Medical University, Szczecin, Poland
| | - Thorsten Ecke
- Department of Urology, Helios Hospital Bad Saarow, Bad Saarow, Germany
| | - Stefan Koch
- Department of Pathology, Helios Hospital Bad Saarow, Bad Saarow, Germany
| | - Nico Adamini
- Department of Urology, Albertinen Hospital, Hamburg, Germany
| | - Joachim Weischenfeldt
- Department of Urology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
- Biotech Research & Innovation Center (BRIC), University of Copenhagen, Copenhagen, Denmark
- Finsen Laboratory, Rigshospitalet, Copenhagen, Denmark
| | - Tobias Klatte
- Department of Urology, Helios Hospital Bad Saarow, Bad Saarow, Germany
| | - Sarah Minner
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ronald Simon
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Guido Sauter
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thorsten Schlomm
- Department of Urology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - David Horst
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Henrik Zecha
- Department of Urology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
- Department of Urology, Albertinen Hospital, Hamburg, Germany
| |
Collapse
|
3
|
Hu T, Mu C, Li Y, Hao W, Yu X, Wang Y, Han W, Li Q. GPS2 ameliorates cigarette smoking-induced pulmonary vascular remodeling by modulating the ras-Raf-ERK axis. Respir Res 2024; 25:210. [PMID: 38755610 PMCID: PMC11100185 DOI: 10.1186/s12931-024-02831-0] [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: 01/10/2024] [Accepted: 05/01/2024] [Indexed: 05/18/2024] Open
Abstract
BACKGROUND Mitogen-activated protein kinase (MAPK)signaling-mediated smoking-associated pulmonary vascular remodeling (PVR) plays an important role in the pathogenesis of group 3 pulmonary hypertension (PH). And G protein pathway suppressor 2 (GPS2) could suppress G-protein signaling such as Ras and MAPK, but its role in cigarette smoking -induced PVR (CS-PVR) is unclear. METHODS An in vivo model of smoke-exposed rats was constructed to assess the role of GPS2 in smoking-induced PH and PVR. In vitro, the effects of GPS2 overexpression and silencing on the function of human pulmonary arterial smooth cells (HPASMCs) and the underlying mechanisms were explored. RESULTS GPS2 expression was downregulated in rat pulmonary arteries (PAs) and HPASMCs after CS exposure. More importantly, CS-exposed rats with GPS2 overexpression had lower right ventricular systolic pressure (RVSP), right ventricular hypertrophy index (RVHI), and wall thickness (WT%) than those without. And enhanced proliferation and migration of HPASMCs induced by cigarette smoking extract (CSE) can be evidently inhibited by overexpressed GPS2. Besides, GPS2siRNA significantly enhanced the proliferation, and migration of HPASMCs as well as activated Ras and Raf/ERK signaling, while these effects were inhibited by zoledronic acid (ZOL). In addition, GPS2 promoter methylation level in rat PAs and HPASMCs was increased after CS exposure, and 5-aza-2-deoxycytidine (5-aza) inhibited CSE-induced GPS2 hypermethylation and downregulation in vitro. CONCLUSIONS GPS2 overexpression could improve the CS-PVR, suggesting that GPS2 might serve as a novel therapeutic target for PH-COPD in the future.
Collapse
Affiliation(s)
- Ting Hu
- Department of Respiratory and Critical Care Medicine, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
- Qingdao Key Lab of Common Diseases, Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, 5 Donghai Middle Road, Qingdao, 266071, China
| | - Chaohui Mu
- Department of Respiratory and Critical Care Medicine, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
- Qingdao Key Lab of Common Diseases, Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, 5 Donghai Middle Road, Qingdao, 266071, China
| | - Yanmiao Li
- Department of Respiratory and Critical Care Medicine, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
- Qingdao Key Lab of Common Diseases, Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, 5 Donghai Middle Road, Qingdao, 266071, China
| | - Wanming Hao
- Qingdao Key Lab of Common Diseases, Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, 5 Donghai Middle Road, Qingdao, 266071, China
- Department of Respiratory and Critical Care Medicine, Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, 5 Donghai Middle Road, Qingdao, 266071, China
| | - Xinjuan Yu
- Qingdao Key Lab of Common Diseases, Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, 5 Donghai Middle Road, Qingdao, 266071, China
- Department of Respiratory and Critical Care Medicine, Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, 5 Donghai Middle Road, Qingdao, 266071, China
| | - Yixuan Wang
- Qingdao Key Lab of Common Diseases, Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, 5 Donghai Middle Road, Qingdao, 266071, China
- Department of Respiratory and Critical Care Medicine, Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, 5 Donghai Middle Road, Qingdao, 266071, China
| | - Wei Han
- Qingdao Key Lab of Common Diseases, Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, 5 Donghai Middle Road, Qingdao, 266071, China.
- Department of Respiratory and Critical Care Medicine, Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, 5 Donghai Middle Road, Qingdao, 266071, China.
| | - Qinghai Li
- Qingdao Key Lab of Common Diseases, Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, 5 Donghai Middle Road, Qingdao, 266071, China.
- Department of Respiratory and Critical Care Medicine, Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, 5 Donghai Middle Road, Qingdao, 266071, China.
| |
Collapse
|
4
|
Genome-wide CRISPR knockout screening identified G protein pathway suppressor 2 as a novel tumor suppressor for uveal melanoma metastasis. J Cancer Res Clin Oncol 2022:10.1007/s00432-022-04160-5. [PMID: 35941228 DOI: 10.1007/s00432-022-04160-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 06/16/2022] [Indexed: 10/15/2022]
Abstract
PURPOSE Uveal melanoma (UM) is the most common intraocular malignant tumor in adults. Due to the lack of effective treatments for metastatic UM, the survival of UM has not changed over the past 3 decades. Therefore, it is important to identify essential genes regulating the metastasis of UM. METHODS In this study, a genome-wide CRISPR knockout screen in an orthotopic mouse model of UM was performed to identify the regulatory genes conferring the metastatic phenotype. Loss-of-function analyses were performed to explore the function of G protein pathway suppressor 2 (GPS2) in UM metastasis in vitro and in vivo. RNA sequencing was performed to investigate the molecular mechanism underlying the function of GPS2 as a tumor suppressor in UM. RESULTS Among the highest-ranking genes, we found several validated tumor suppressors, such as SHPRH, GPS2, PRPH2, and hsa-mir-1229; GPS2 was chosen as the candidate gene for further studies. GPS2 was lower expressed in the tumor tissues of UM patients. Furthermore, knocking-down GPS2 promoted the proliferation and metastatic abilities of UM cells both in vivo and in vitro. Finally, analysis of the transcriptome data revealed that silencing GPS2 upregulates oncogenic signaling pathways MAPK and PI3K-Akt, and in the meantime downregulates tumor suppressor signaling pathway Slit/Robo in UM cells. CONCLUSION Altogether, our study proved that the GPS2 gene functions as a tumor suppressor and might be a novel potential therapeutic target for UM treatment.
Collapse
|
5
|
Si Y, Zhang H, Peng P, Zhu C, Shen J, Xiong Y, Liu X, Xiang Y, Li W, Ren Y, Wan F, Zhang L, Liu Y. G protein pathway suppressor 2 suppresses gastric cancer by destabilizing epidermal growth factor receptor. Cancer Sci 2021; 112:4867-4882. [PMID: 34609770 PMCID: PMC8645722 DOI: 10.1111/cas.15151] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 09/13/2021] [Accepted: 09/18/2021] [Indexed: 11/29/2022] Open
Abstract
G protein pathway suppressor 2 (GPS2) is expressed in most human tissues, including the stomach. However, the biological functions of GPS2 in cancer, as well as the underlying molecular mechanisms, remain poorly understood. Here, we report that GPS2 expression was aberrantly downregulated in gastric cancer (GC) tissues compared with control tissues. Clinicopathologic analysis showed that low GPS2 expression was significantly correlated with pathological grade, lymph node stage, and invasive depth. Kaplan-Meier analysis indicated that patients with low GPS2 expression showed poorer overall survival rates than those with high GPS2 expression. Moreover, GPS2 overexpression decreased GC cell proliferation, colony formation, tumorigenesis, and invasion. Overexpression of GPS2 reduced the protein expression of epidermal growth factor receptor (EGFR) and inhibited its downstream signaling in GC cells. Interestingly, GPS2 decreased EGFR protein expression, which was reversed by a lysosome inhibitor. Furthermore, GPS2 reduced EGFR protein stability by enhancing the binding of EGFR and an E3 ligase, c-Cbl, which promoted the ubiquitination of EGFR, ultimately leading to its degradation through the lysosomal pathway. Further analysis indicated that GPS2 activated autophagy and promoted the autophagic flux by destabilizing EGFR. Taken together, these results suggest that low GPS2 expression is associated with GC progression and provide insights into the applicability of the GPS2-EGFR axis as a potential therapeutic target in GC.
Collapse
Affiliation(s)
- Yuan Si
- Laboratory of Molecular Targeted Therapy of CancerInstitute of Basic Medical SciencesHubei University of MedicineShiyanChina
- Hubei Key Laboratory of Wudang Local Chinese Medicine ResearchHubei University of MedicineShiyanChina
- Hubei Key Laboratory of Embryonic Stem Cell ResearchHubei University of MedicineShiyanChina
| | - Haitao Zhang
- Sir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhouChina
| | - Peng Peng
- Laboratory of Molecular Targeted Therapy of CancerInstitute of Basic Medical SciencesHubei University of MedicineShiyanChina
- Hubei Key Laboratory of Embryonic Stem Cell ResearchHubei University of MedicineShiyanChina
- Laboratory of Molecular Targeted Therapy of CancerBiomedical Research InstituteHubei University of MedicineShiyanChina
| | - Chu Zhu
- Sir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhouChina
| | - Jie Shen
- Laboratory of Molecular Targeted Therapy of CancerInstitute of Basic Medical SciencesHubei University of MedicineShiyanChina
- Hubei Key Laboratory of Embryonic Stem Cell ResearchHubei University of MedicineShiyanChina
| | - Yilian Xiong
- Laboratory of Molecular Targeted Therapy of CancerInstitute of Basic Medical SciencesHubei University of MedicineShiyanChina
- Hubei Key Laboratory of Embryonic Stem Cell ResearchHubei University of MedicineShiyanChina
| | - Xuewen Liu
- Laboratory of Molecular Targeted Therapy of CancerInstitute of Basic Medical SciencesHubei University of MedicineShiyanChina
- Laboratory of Molecular Targeted Therapy of CancerBiomedical Research InstituteHubei University of MedicineShiyanChina
| | - Yuchen Xiang
- Laboratory of Molecular Targeted Therapy of CancerInstitute of Basic Medical SciencesHubei University of MedicineShiyanChina
- Hubei Key Laboratory of Embryonic Stem Cell ResearchHubei University of MedicineShiyanChina
- Laboratory of Molecular Targeted Therapy of CancerBiomedical Research InstituteHubei University of MedicineShiyanChina
| | - Wenjuan Li
- Laboratory of Molecular Targeted Therapy of CancerInstitute of Basic Medical SciencesHubei University of MedicineShiyanChina
- Hubei Key Laboratory of Embryonic Stem Cell ResearchHubei University of MedicineShiyanChina
| | - Yuliang Ren
- Laboratory of Molecular Targeted Therapy of CancerInstitute of Basic Medical SciencesHubei University of MedicineShiyanChina
- Hubei Key Laboratory of Embryonic Stem Cell ResearchHubei University of MedicineShiyanChina
| | - Fang Wan
- Laboratory of Molecular Targeted Therapy of CancerInstitute of Basic Medical SciencesHubei University of MedicineShiyanChina
- Hubei Key Laboratory of Embryonic Stem Cell ResearchHubei University of MedicineShiyanChina
| | - Liang Zhang
- Laboratory of Molecular Targeted Therapy of CancerInstitute of Basic Medical SciencesHubei University of MedicineShiyanChina
- Hubei Key Laboratory of Embryonic Stem Cell ResearchHubei University of MedicineShiyanChina
- Laboratory of Molecular Targeted Therapy of CancerBiomedical Research InstituteHubei University of MedicineShiyanChina
| | - Ying Liu
- Laboratory of Molecular Targeted Therapy of CancerInstitute of Basic Medical SciencesHubei University of MedicineShiyanChina
- Hubei Key Laboratory of Wudang Local Chinese Medicine ResearchHubei University of MedicineShiyanChina
- Hubei Key Laboratory of Embryonic Stem Cell ResearchHubei University of MedicineShiyanChina
- Laboratory of Molecular Targeted Therapy of CancerBiomedical Research InstituteHubei University of MedicineShiyanChina
| |
Collapse
|
6
|
Hadjimichael AC, Pergaris A, Kaspiris A, Foukas AF, Theocharis SE. Liquid Biopsy: A New Translational Diagnostic and Monitoring Tool for Musculoskeletal Tumors. Int J Mol Sci 2021; 22:11526. [PMID: 34768955 PMCID: PMC8583711 DOI: 10.3390/ijms222111526] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 10/21/2021] [Accepted: 10/23/2021] [Indexed: 12/17/2022] Open
Abstract
Soft tissue and bone sarcomas represent a group of aggressive neoplasms often accompanied by dismal patient prognosis, especially when distant metastases are present. Moreover, effective treatment can pose a challenge, as recurrences are frequent and almost half of patients present with advanced disease. Researchers have unveiled the molecular abnormalities implicated in sarcomas' carcinogenesis, paving the way for novel treatment strategies based on each individual tumor's characteristics. Therefore, the development of new techniques aiding in early disease detection and tumor molecular profiling is imperative. Liquid biopsy refers to the sampling and analysis of patients' fluids, such as blood, to identify tumor biomarkers, through a variety of methods, including qRT-PCR, qPCR, droplet digital PCR, magnetic microbeads and digital PCR. Assessment of circulating tumor cells (CTCs), circulating free DNA (ctDNA), micro RNAs (miRs), long non-coding RNAs (lncRNAs), exosomes and exosome-associated proteins can yield a plethora of information on tumor molecular signature, histologic type and disease stage. In addition, the minimal invasiveness of the procedure renders possible its wide application in the clinical setting, and, therefore, the early detection of the presence of tumors. In this review of the literature, we gathered information on biomarkers assessed through liquid biopsy in soft tissue and bone sarcoma patients and we present the information they can yield for each individual tumor type.
Collapse
Affiliation(s)
- Argyris C. Hadjimichael
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 75, Mikras Asias Street, Bld 10, Goudi, 11527 Athens, Greece; (A.C.H.); (A.P.)
- Third Department of Orthopaedic Surgery, “KAT” General Hospital of Athens, Nikis 2, 14561 Kifissia, Greece;
| | - Alexandros Pergaris
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 75, Mikras Asias Street, Bld 10, Goudi, 11527 Athens, Greece; (A.C.H.); (A.P.)
| | - Angelos Kaspiris
- Division for Orthopaedic Research, Laboratory of Molecular Pharmacology, School of Health Sciences, University of Patras, 26504 Patras, Greece;
| | - Athanasios F. Foukas
- Third Department of Orthopaedic Surgery, “KAT” General Hospital of Athens, Nikis 2, 14561 Kifissia, Greece;
| | - Stamatios E. Theocharis
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 75, Mikras Asias Street, Bld 10, Goudi, 11527 Athens, Greece; (A.C.H.); (A.P.)
| |
Collapse
|
7
|
Liu M, Katsevich E, Janson L, Ramdas A. Fast and powerful conditional randomization testing via distillation. Biometrika 2021. [DOI: 10.1093/biomet/asab039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Summary
We consider the problem of conditional independence testing: given a response $Y$ and covariates $(X,Z)$, we test the null hypothesis that $Y {\perp\!\!\!\perp} X \mid Z$. The conditional randomization test was recently proposed as a way to use distributional information about $X\mid Z$ to exactly and nonasymptotically control Type-I error using any test statistic in any dimensionality without assuming anything about $Y\mid (X,Z)$. This flexibility, in principle, allows one to derive powerful test statistics from complex prediction algorithms while maintaining statistical validity. Yet the direct use of such advanced test statistics in the conditional randomization test is prohibitively computationally expensive, especially with multiple testing, due to the requirement to recompute the test statistic many times on resampled data. We propose the distilled conditional randomization test, a novel approach to using state-of-the-art machine learning algorithms in the conditional randomization test while drastically reducing the number of times those algorithms need to be run, thereby taking advantage of their power and the conditional randomization test’s statistical guarantees without suffering the usual computational expense. In addition to distillation, we propose a number of other tricks, like screening and recycling computations, to further speed up the conditional randomization test without sacrificing its high power and exact validity. Indeed, we show in simulations that all our proposals combined lead to a test that has similar power to most powerful existing conditional randomization test implementations, but requires orders of magnitude less computation, making it a practical tool even for large datasets. We demonstrate these benefits on a breast cancer dataset by identifying biomarkers related to cancer stage.
Collapse
Affiliation(s)
- Molei Liu
- Department of Biostatistics, Harvard Chan School of Public Health, 677 Huntington Avenue, Boston, Massachusetts 02115, U.S.A
| | - Eugene Katsevich
- Department of Statistics and Data Science, Wharton School of the University of Pennsylvania, 265 South 37th Street, Philadelphia, Pennsylvania 19104, U.S.A
| | - Lucas Janson
- Department of Statistics, Harvard University, One Oxford Street, Cambridge, Massachusetts 02138, U.S.A
| | - Aaditya Ramdas
- Department of Statistics & Data Science, Carnegie Mellon University, 132H Baker Hall, Pittsburgh, Pennsylvania 15213, U.S.A
| |
Collapse
|
8
|
Ye Z, Zou S, Niu Z, Xu Z, Hu Y. A Novel Risk Model Based on Lipid Metabolism-Associated Genes Predicts Prognosis and Indicates Immune Microenvironment in Breast Cancer. Front Cell Dev Biol 2021; 9:691676. [PMID: 34195202 PMCID: PMC8236894 DOI: 10.3389/fcell.2021.691676] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 05/07/2021] [Indexed: 02/05/2023] Open
Abstract
Background Breast cancer (BRCA) is the most common tumor in women, and lipid metabolism involvement has been demonstrated in its tumorigenesis and development. However, the role of lipid metabolism-associated genes (LMAGs) in the immune microenvironment and prognosis of BRCA remains unclear. Methods A total of 1076 patients with BRCA were extracted from The Cancer Genome Atlas database and randomly assigned to the training cohort (n = 760) or validation cohort (n = 316). Kaplan–Meier analysis was used to assess differences in survival. Consensus clustering was performed to categorize the patients with BRCA into subtypes. Using multivariate Cox regression analysis, an LMAG-based prognostic risk model was constructed from the training cohort and validated using the validation cohort. The immune microenvironment was evaluated using the ESTIMATE and tumor immune estimation resource algorithms, CIBERSORT, and single sample gene set enrichment analyses. Results Consensus clustering classified the patients with BRCA into two subgroups with significantly different overall survival rates and immune microenvironments. Better prognosis was associated with high immune infiltration. The prognostic risk model, based on four LMAGs (MED10, PLA2G2D, CYP4F11, and GPS2), successfully stratified the patients into high- and low-risk groups in both the training and validation sets. High risk scores predicted poor prognosis and indicated low immune status. Subgroup analysis suggested that the risk model was an independent predictor of prognosis in BRCA. Conclusion This study demonstrated, for the first time, that LMAG expression plays a crucial role in BRCA. The LMAG-based risk model successfully predicted the prognosis and indicated the immune microenvironment of patients with BRCA. Our study may provide inspiration for further research on BRCA pathomechanisms.
Collapse
Affiliation(s)
- Zhimin Ye
- Department of Pathology, School of Basic Medical Sciences, Central South University, Changsha, China.,Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Shengmei Zou
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhiyuan Niu
- Department of Pathology, School of Basic Medical Sciences, Central South University, Changsha, China.,Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Zhijie Xu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Yongbin Hu
- Department of Pathology, School of Basic Medical Sciences, Central South University, Changsha, China.,Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
9
|
Interaction of NEP with G Protein Pathway Suppressor 2 Facilitates Influenza A Virus Replication by Weakening the Inhibition of GPS2 to RNA Synthesis and Ribonucleoprotein Assembly. J Virol 2021; 95:JVI.00008-21. [PMID: 33658351 PMCID: PMC8139649 DOI: 10.1128/jvi.00008-21] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The nuclear export protein (NEP) serves multiple functions in the life cycle of influenza A virus (IAV). Identifying novel host proteins that interact with NEP and understanding their functions in IAV replication are of great interest. In this study, we screened and confirmed the direct interaction of G protein pathway suppressor 2 (GPS2) with NEP through a yeast two-hybrid screening assay and glutathione S-transferase-pulldown and co-immunoprecipitation assays. Knockdown or knockout of GPS2 enhanced IAV titers, whereas overexpression of GPS2 impaired IAV replication, demonstrating that GPS2 acted as a negative host factor in IAV replication. Meanwhile, GPS2 inhibited viral RNA synthesis by reducing the assembly of IAV polymerase. Interestingly, IAV NEP interacted with GPS2 and mediated its nuclear export, thereby activated the degradation of GPS2. Thus, NEP-GPS2 interaction weakened the inhibition of GPS2 to viral polymerase activity and benefited virus replication. Overall, this study identified the novel NEP-binding host partner GPS2 as a critical host factor to participate in IAV replication. These findings provided novel insights into the interactions between IAV and host cells, revealing a new function for GPS2 during IAV replication.Importance: NEP is proposed to play multiple biologically important roles in the life cycle of IAV, which largely relies on host factors by interaction. Our study demonstrated that GPS2 could reduce the interaction between PB1 and PB2 and interfere with vRNP assembly. Thus, GPS2 inhibited the RNA synthesis of IAV and negatively regulated its replication. Importantly, IAV NEP interacted with GPS2 and mediated the nuclear export of GPS2, thereby activated the degradation of GPS2. Thus, NEP-GPS2 interaction weakened the inhibition of GPS2 to viral polymerase activity and benefited virus replication.
Collapse
|
10
|
Chan S, Smith E, Gao Y, Kwan J, Blum BC, Tilston-Lunel AM, Turcinovic I, Varelas X, Cardamone MD, Monti S, Emili A, Perissi V. Loss of G-Protein Pathway Suppressor 2 Promotes Tumor Growth Through Activation of AKT Signaling. Front Cell Dev Biol 2021; 8:608044. [PMID: 33490071 PMCID: PMC7817781 DOI: 10.3389/fcell.2020.608044] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 12/04/2020] [Indexed: 02/06/2023] Open
Abstract
G Protein Suppressor 2 (GPS2) is a multifunctional protein that exerts important roles in inflammation and metabolism in adipose, liver, and immune cells. GPS2 has recently been identified as a significantly mutated gene in breast cancer and other malignancies and proposed to work as a putative tumor suppressor. However, molecular mechanisms by which GPS2 prevents cancer development and/or progression are largely unknown. Here, we have profiled the phenotypic changes induced by GPS2 depletion in MDA-MB-231 triple negative breast cancer cells and investigated the underlying molecular mechanisms. We found that GPS2-deleted MDA-MB-231 cells exhibited increased proliferative, migratory, and invasive properties in vitro, and conferred greater tumor burden in vivo in an orthotopic xenograft mouse model. Transcriptomic, proteomic and phospho-proteomic profiling of GPS2-deleted MBA-MB-231 revealed a network of altered signals that relate to cell growth and PI3K/AKT signaling. Overlay of GPS2-regulated gene expression with MDA-MB-231 cells modified to express constitutively active AKT showed significant overlap, suggesting that sustained AKT activation is associated with loss of GPS2. Accordingly, we demonstrate that the pro-oncogenic phenotypes associated with GPS2 deletion are rescued by pharmacological inhibition of AKT with MK2206. Collectively, these observations confirm a tumor suppressor role for GPS2 and reveal that loss of GPS2 promotes breast cancer cell proliferation and tumor growth through uncontrolled activation of AKT signaling. Moreover, our study points to GPS2 as a potential biomarker for a subclass of breast cancers that would be responsive to PI3K-class inhibitor drugs.
Collapse
Affiliation(s)
- Stefanie Chan
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, United States
| | - Emma Smith
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, United States
| | - Yuan Gao
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, United States
| | - Julian Kwan
- Center for Network Systems Biology, Boston University, Boston, MA, United States
| | - Benjamin C. Blum
- Center for Network Systems Biology, Boston University, Boston, MA, United States
| | | | - Isabella Turcinovic
- Center for Network Systems Biology, Boston University, Boston, MA, United States
| | - Xaralabos Varelas
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, United States
| | - Maria Dafne Cardamone
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, United States
| | - Stefano Monti
- Division of Computational Biology, Department of Medicine, Boston University School of Medicine, Boston, MA, United States
| | - Andrew Emili
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, United States
- Center for Network Systems Biology, Boston University, Boston, MA, United States
| | - Valentina Perissi
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, United States
| |
Collapse
|
11
|
Lerebours A, Robson S, Sharpe C, Nagorskaya L, Gudkov D, Haynes-Lovatt C, Smith JT. Transcriptional Changes in the Ovaries of Perch from Chernobyl. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2020; 54:10078-10087. [PMID: 32686935 DOI: 10.1021/acs.est.0c02575] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Fish have been highly exposed to radiation in freshwater systems after the Chernobyl Nuclear Power Plant (NPP) accident in 1986 and in freshwater and marine systems after the more recent Fukushima NPP accident in 2011. In the years after the accident, the radioactivity levels rapidly declined due to radioactive decay and environmental processes, but chronic lower dose exposures persisted. To gain insights into the long-term effects of environmental low dose radiation on fish ovaries development, a high-throughput transcriptomic approach including a de novo assembly was applied to different gonad phenotypes of female perch: developed gonads from reference lakes, developed/irradiated from medium contaminated lake, and both developed/irradiated and undeveloped from more highly contaminated lakes. This is the most comprehensive analysis to date of the gene responses in wildlife reproductive system to radiation. Some gene responses that were modulated in irradiated gonads were found to be involved in biological processes including cell differentiation and proliferation (ggnb2, mod5, rergl), cytoskeleton organization (k1C18, mtpn), gonad development (nell2, tcp4), lipid metabolism (ldah, at11b, nltp), reproduction (cyb5, cyp17A, ovos), DNA damage repair (wdhd1, rad51, hus1), and epigenetic mechanisms (dmap1). Identification of these genes provides a better understanding of the underlying molecular mechanisms underpinning the development of the gonad phenotypes of wild perch and how fish may respond to chronic exposure to radiation in their natural environment, though causal attribution of gene responses remains unclear in the undeveloped gonads.
Collapse
Affiliation(s)
- Adélaïde Lerebours
- School of the Environment, Geography and Geosciences, University of Portsmouth, Portsmouth PO1 3QL, United Kingdom
- School of Biological Sciences, University of Portsmouth, Portsmouth PO1 2DY, United Kingdom
| | - Samuel Robson
- Centre for Enzyme Innovation, University of Portsmouth, Portsmouth PO1 2DT, United Kingdom
| | - Colin Sharpe
- School of Biological Sciences, University of Portsmouth, Portsmouth PO1 2DY, United Kingdom
| | - Liubov Nagorskaya
- Applied Science Center for Bioresources of the National Academy of Sciences of Belarus, Minsk 220072, Belarus
| | - Dmitri Gudkov
- Institute of Hydrobiology of the National Academy of Sciences of Ukraine, Kiev UA-04210, Ukraine
| | | | - Jim T Smith
- School of the Environment, Geography and Geosciences, University of Portsmouth, Portsmouth PO1 3QL, United Kingdom
| |
Collapse
|
12
|
Anjitha R, Antony A, Shilpa O, Anupama KP, Mallikarjunaiah S, Gurushankara HP. Malathion induced cancer-linked gene expression in human lymphocytes. ENVIRONMENTAL RESEARCH 2020; 182:109131. [PMID: 32069766 DOI: 10.1016/j.envres.2020.109131] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 01/08/2020] [Accepted: 01/09/2020] [Indexed: 06/10/2023]
Abstract
BACKGROUND Malathion is the most widely used organophosphate pesticide in agriculture. Increasing cancer incidence in agricultural workers and their children links to the exposure of malathion. Identification of genes involved in the process of carcinogenesis is essential for exploring the role of malathion. The alteration in gene expression by malathion in human lymphocytes has not been explored yet, although hematological malignancies are rampant in humans. OBJECTIVE This study investigates the malathion induced expression of cancer associated genes in human lymphocytes. METHODS Human lymphocyte viability and colony-forming ability were analyzed in malathion treated and control groups. Gene expression profile in control and malathion treated human lymphocytes were performed using a microarray platform. The genes which have significant functions and those involved in different pathways were analyzed using the DAVID database. Differential gene expression upon malathion exposure was validated by quantitative real-time (qRT)-PCR. RESULTS Malathion caused a concentration-dependent reduction in human lymphocyte viability. At low concentration (50 μg/mL) of malathion treatment, human lymphocytes were viable indicating that low concentration of malathion is not cytotoxic and induces the colony formation. Total of 659 genes (15%) were up regulated and 3729 genes (85%) were down regulated in malathion treated human lymphocytes. About 57 cancer associated genes related to the growth and differentiation of B and T cells, immunoglobulin production, haematopoiesis, tumor suppression, oncogenes and signal transduction pathways like MAPK and RAS were induced by malathion. CONCLUSION This study evidences the carcinogenic nature of malathion. Low concentration of this pesticide is not cytotoxic and induces differentially regulated genes in human lymphocytes, which are involved in the initiation, progression, and pathogenesis of cancer.
Collapse
Affiliation(s)
- Ramakrishnan Anjitha
- Department of Zoology, School of Biological Sciences, Central University of Kerala, Periya, 671 320, Kasaragod, Kerala, India
| | - Anet Antony
- Department of Zoology, School of Biological Sciences, Central University of Kerala, Periya, 671 320, Kasaragod, Kerala, India
| | - Olakkaran Shilpa
- Department of Zoology, School of Biological Sciences, Central University of Kerala, Periya, 671 320, Kasaragod, Kerala, India
| | - Kizhakke P Anupama
- Department of Zoology, School of Biological Sciences, Central University of Kerala, Periya, 671 320, Kasaragod, Kerala, India
| | - Shanthala Mallikarjunaiah
- Center for Applied Genetics, Department of Studies in Zoology, Bangalore University, Jnanabharathi, Bengaluru, 560 056, Karnataka, India
| | - Hunasanahally P Gurushankara
- Department of Zoology, School of Biological Sciences, Central University of Kerala, Periya, 671 320, Kasaragod, Kerala, India.
| |
Collapse
|
13
|
Yang L, Chen S, Luo P, Yan W, Wang C. Liposarcoma: Advances in Cellular and Molecular Genetics Alterations and Corresponding Clinical Treatment. J Cancer 2020; 11:100-107. [PMID: 31892977 PMCID: PMC6930414 DOI: 10.7150/jca.36380] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 09/12/2019] [Indexed: 12/12/2022] Open
Abstract
Liposarcoma is a malignant tumor of mesenchymal origin with significant tissue diversity. It is composed of adipocytes with different degrees of differentiation and different degrees of heteromorphosis. It is not sensitive to traditional radiotherapy and chemotherapy and has a poor prognosis. In recent years, with the rapid development of basic immunology, molecular genetics and tumor molecular biology, the histological classification of liposarcoma has become increasingly clear. More and more new methods and technologies, such as gene expression profile analysis, the whole genome sequencing, miRNA expression profile analysis and RNA sequencing, have been successfully applied to liposarcoma, bringing about a deeper understanding of gene expression changes and molecular pathogenic mechanisms in the occurrence and development of liposarcoma. This study reviews the present research status and progress of cellular and molecular alterations of liposarcoma and corresponding clinical treatment progress.
Collapse
Affiliation(s)
- Lingge Yang
- Department of Musculoskeletal Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shiqi Chen
- Department of Musculoskeletal Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Peng Luo
- Department of Musculoskeletal Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wangjun Yan
- Department of Musculoskeletal Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Chunmeng Wang
- Department of Musculoskeletal Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
14
|
Koyama T, Rhrissorrakrai K, Parida L. Analysis on GENIE reveals novel recurrent variants that affect molecular diagnosis of sizable number of cancer patients. BMC Cancer 2019; 19:114. [PMID: 30709382 PMCID: PMC6359859 DOI: 10.1186/s12885-019-5313-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 01/22/2019] [Indexed: 12/18/2022] Open
Abstract
Background Significant numbers of variants detected in cancer patients are often left labeled only as variants of unknown significance (VUS). In order to expand precision medicine to a wider population, we need to extend our knowledge of pathogenicity and drug response in the context of VUS’s. Methods In this study, we analyzed variants from AACR Project GENIE Consortium APG (Cancer Discov 7:818-831, 2017) and compared them to the COSMIC database Forbes et al. (Nucleic Acids Res 43:D805-811, 2015) to identify recurrent variants that would merit further study. We filtered out known hotspot variants, inactivating variants in tumor suppressors, and likely benign variants by comparing with COSMIC and ExAC Lee et al. (Science 337:967-971, 2012). Results We have identified 45,933 novel variants with unknown significance unique to GENIE. In our analysis, we found on average six variants per patient where two could be considered as pathogenic or likely pathogenic and the majority are VUS’s. More importantly, we have discovered 730 recurrent variants that appear more than 3 times in GENIE but less than 3 in COSMIC. If we combine the recurrences of GENIE and COSMIC for all variants, 2586 are newly identified as occurring more than 3 times than when using COSMIC alone. Conclusions Although it would be inappropriate to blindly accept these recurrent variants as pathogenic, they may warrant higher priority than other observed VUS’s. These newly identified recurrent variants might affect the molecular profiles of approximately 1 in 6 patients. Further analysis and characterization of these variants in both research and clinical contexts will improve patient treatments and the development of new therapeutics. Electronic supplementary material The online version of this article (10.1186/s12885-019-5313-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Takahiko Koyama
- IBM TJ Watson Research Center, Yorktown Heights, NY, 10598, USA.
| | | | - Laxmi Parida
- IBM TJ Watson Research Center, Yorktown Heights, NY, 10598, USA
| |
Collapse
|