1
|
Martinez-Val A, Van der Hoeven L, Bekker-Jensen DB, Jørgensen MM, Nors J, Franciosa G, Andersen CL, Bramsen JB, Olsen JV. Proteomics of colorectal tumors identifies the role of CAVIN1 in tumor relapse. Mol Syst Biol 2025:10.1038/s44320-025-00102-8. [PMID: 40269326 DOI: 10.1038/s44320-025-00102-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 03/28/2025] [Accepted: 04/02/2025] [Indexed: 04/25/2025] Open
Abstract
Colorectal cancer molecular signatures derived from omics data can be employed to stratify CRC patients and aid decisions about therapies or evaluate prognostic outcome. However, molecular biomarkers for identification of patients at increased risk of disease relapse are currently lacking. Here, we present a comprehensive multi-omics analysis of a Danish colorectal cancer tumor cohort composed of 412 biopsies from tumors of 371 patients diagnosed at TNM stage II or III. From mass spectrometry-based patient proteome profiles, we classified the tumors into four molecular subtypes, including a mesenchymal-like subtype. As the mesenchymal-rich tumors are known to represent the most invasive and metastatic phenotype, we focused on the protein signature defining this subtype to evaluate their potential as relapse risk markers. Among signature-specific proteins, we followed-up Caveolae-Associated Protein-1 (CAVIN1) and demonstrated its role in tumor progression in a 3D in vitro model of colorectal cancer. Compared to previous omics analyses of CRC, our multi-omics classification provided deeper insights into EMT in cancer cells with stronger correlations with risk of relapse.
Collapse
Affiliation(s)
- Ana Martinez-Val
- Novo Nordisk Foundation Center for Protein Research, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain.
- Cardiovascular Proteomics Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain.
| | - Leander Van der Hoeven
- Novo Nordisk Foundation Center for Protein Research, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Dorte B Bekker-Jensen
- Novo Nordisk Foundation Center for Protein Research, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Evosep Biosystems, Odense, Denmark
| | - Margarita Melnikova Jørgensen
- Institute of Pathology, Randers Regional Hospital, Randers, Denmark
- Department of Pathology, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Jesper Nors
- Department of Clinical Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Giulia Franciosa
- Novo Nordisk Foundation Center for Protein Research, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Claus L Andersen
- Department of Clinical Medicine, Aarhus University Hospital, Aarhus, Denmark.
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark.
| | - Jesper B Bramsen
- Department of Clinical Medicine, Aarhus University Hospital, Aarhus, Denmark.
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark.
| | - Jesper V Olsen
- Novo Nordisk Foundation Center for Protein Research, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
2
|
Pan Z, Tan Z, Xu N, Yao Z, Zheng C, Shang J, Xie L, Xu J, Wang J, Jiang L, Zhu X, Yu D, Li Y, Che Y, Gong Y, Qin Z, Zhang Y, Zou X, Xu T, Guo Z, Jin T, Guo T, Wang W, Chen W, Sun Y, Wang W, Peng X, Yin C, Ding C, Huang P, Ge M. Integrative proteogenomic characterization reveals therapeutic targets in poorly differentiated and anaplastic thyroid cancers. Nat Commun 2025; 16:3601. [PMID: 40234451 PMCID: PMC12000556 DOI: 10.1038/s41467-025-58910-3] [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: 08/19/2024] [Accepted: 04/03/2025] [Indexed: 04/17/2025] Open
Abstract
Poorly differentiated thyroid cancer (PDTC) and anaplastic thyroid cancer (ATC) present major challenges in treatment owing to extreme aggressiveness and high heterogeneity. In this study, deep-scale analyses spanning genomic, proteomic, and phosphoproteomic data are performed on 348 thyroid-cancer and 119 tumor-adjacent samples. TP53 (48%), TERT promoter (36.5%), and BRAF (23%) are most frequently mutated in PDTC and ATC. Ribosome biogenesis is identified as a common hallmark of ATC, and RRP9 silencing dramatically inhibits tumor growth. Proteomic clustering identified three ATC/PDTC subtypes. Pro-I subtype is characterized with aberrant insulin signaling and low immune cell infiltration, and Pro-II is featured with DNA repair signaling, while Pro-III harbors high frequency of TP53 and BRAF mutation and intensive C5AR1+ myeloid infiltration. Targeting C5AR1 synergistically improves antitumor effect of PD-1 blockade against ATC cell-derived tumors. These findings provide systematic insights into tumor biology and opportunities for drug discovery, accelerating precision therapy for virulent thyroid cancers.
Collapse
Affiliation(s)
- Zongfu Pan
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
- Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Zhuo Tan
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
- Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Ning Xu
- Clinical Research Center for Cell-based Immunotherapy of Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetics and Development of Complex Phenotypes, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China
| | - Zhenmei Yao
- Clinical Research Center for Cell-based Immunotherapy of Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetics and Development of Complex Phenotypes, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China
| | - Chuanming Zheng
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
- Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Jinbiao Shang
- Department of Thyroid Surgery, Zhejiang Cancer Hospital, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Malignant Tumor, Hangzhou, China
| | - Lei Xie
- Department of Head and Neck Surgery, The Affiliated Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jiajie Xu
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
- Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Jiafeng Wang
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
- Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Liehao Jiang
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
- Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Xuhang Zhu
- Department of Thyroid Surgery, Zhejiang Cancer Hospital, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Malignant Tumor, Hangzhou, China
| | - Dingyi Yu
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
- Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Ying Li
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
| | - Yulu Che
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
| | - Yingying Gong
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
| | - Zhaoyu Qin
- Clinical Research Center for Cell-based Immunotherapy of Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetics and Development of Complex Phenotypes, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China
| | - Yiwen Zhang
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
- Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Xiaozhou Zou
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
- Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Tong Xu
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
- Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Zhenying Guo
- Department of Pathology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
| | - Tiefeng Jin
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
| | - Tiannan Guo
- School of Medicine, Westlake University, Hangzhou, China
| | - Wei Wang
- Department of Pathology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
| | - Wanyuan Chen
- Department of Pathology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
| | - Yaoting Sun
- School of Medicine, Westlake University, Hangzhou, China
| | - Weixin Wang
- Hangzhou Cosmos Wisdom Biotechnology Co. Ltd, Hangzhou, China
| | - Xiaojun Peng
- Hangzhou Cosmos Wisdom Biotechnology Co. Ltd, Hangzhou, China
| | - Changtian Yin
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
- Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Chen Ding
- Clinical Research Center for Cell-based Immunotherapy of Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetics and Development of Complex Phenotypes, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China.
| | - Ping Huang
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China.
- Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, China.
- Zhejiang Provincial Clinical Research Center for Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, China.
| | - Minghua Ge
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China.
- Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, China.
- Zhejiang Provincial Clinical Research Center for Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, China.
| |
Collapse
|
3
|
Wang H, Xu JY, Wang T, Xu G, Luo G, Zhang M, Tang G, Wang C, Wang L, Fu W, Ni X, Zhai L, Xu R, Li J, Ye Y, Qiu X, Wu Z, Li J, Zhou Y, Yang J, Tan M, Li J. Integrative proteogenomic and pharmacological landscape of acute myeloid leukaemia. Sci Bull (Beijing) 2025; 70:1051-1056. [PMID: 39609214 DOI: 10.1016/j.scib.2024.11.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 08/23/2024] [Accepted: 11/07/2024] [Indexed: 11/30/2024]
Affiliation(s)
- Hanlin Wang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China; Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264117, China; College of Pharmacy, Fudan University, Shanghai 210023, China; University of Chinese Academy of Sciences, Beijing 100049, China; The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jun-Yu Xu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Tao Wang
- Department of Hematology, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Gaoya Xu
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Guanghao Luo
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China; School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310000, China
| | - Mingya Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China; School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Gusheng Tang
- Department of Hematology, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Chang Wang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Libing Wang
- Department of Hematology, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Weijia Fu
- Department of Hematology, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Xiong Ni
- Department of Hematology, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Linhui Zhai
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
| | - Ran Xu
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jianan Li
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yunfei Ye
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaohui Qiu
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
| | - Zhiqi Wu
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
| | - Jing Li
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yubo Zhou
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China; University of Chinese Academy of Sciences, Beijing 100049, China; School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China; The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.
| | - Jianmin Yang
- Department of Hematology, Changhai Hospital, Naval Medical University, Shanghai 200433, China.
| | - Minjia Tan
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China; University of Chinese Academy of Sciences, Beijing 100049, China; School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Jia Li
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China; Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264117, China; College of Pharmacy, Fudan University, Shanghai 210023, China; University of Chinese Academy of Sciences, Beijing 100049, China; School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China; School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310000, China; The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.
| |
Collapse
|
4
|
Oskolas H, Nogueira FCN, Domont GB, Yu KH, Semenov YR, Sorger P, Steinfelder E, Corps L, Schulz L, Wieslander E, Fenyö D, Kárpáti S, Holló P, Kemény LV, Döme B, Megyesfalvi Z, Pawłowski K, Nishimura T, Kwon H, Encarnación-Guevara S, Szasz AM, Veréb Z, Gyulai R, Németh IB, Appelqvist R, Rezeli M, Baldetorp B, Horvatovich P, Malmström J, Pla I, Sanchez A, Knudsen B, Kiss A, Malm J, Marko-Varga G, Gil J. Comprehensive biobanking strategy with clinical impact at the European Cancer Moonshot Lund Center. J Proteomics 2025; 316:105442. [PMID: 40246065 DOI: 10.1016/j.jprot.2025.105442] [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: 01/17/2025] [Revised: 03/26/2025] [Accepted: 04/12/2025] [Indexed: 04/19/2025]
Abstract
This white paper presents a comprehensive biobanking framework developed at the European Cancer Moonshot Lund Center that merges rigorous sample handling, advanced automation, and multi-omic analyses to accelerate precision oncology. Tumor and blood-based workflows, supported by automated fractionation systems and standardized protocols, ensure the collection of high-quality biospecimens suitable for proteomic, genomic, and metabolic studies. A robust informatics infrastructure, integrating LIMS, barcoding, and REDCap, supports end-to-end traceability and realtime data synchronization, thereby enriching each sample with critical clinical metadata. Proteogenomic integration lies at the core of this initiative, uncovering tumor- and blood-based molecular profiles that inform cancer heterogeneity, metastasis, and therapeutic resistance. Machine learning and AI-driven models further enhance these datasets by stratifying patient populations, predicting therapeutic responses, and expediting the discovery of actionable targets and companion biomarkers. This synergy between technology, automation, and high-dimensional data analytics enables individualized treatment strategies in melanoma, lung, and other cancer types. Aligned with international programs such as the Cancer Moonshot and the ICPC, the Lund Center's approach fosters open collaboration and data sharing on a global scale. This scalable, patient-centric biobanking paradigm provides an adaptable model for institutions aiming to unify clinical, molecular, and computational resources for transformative cancer research.
Collapse
Affiliation(s)
- Henriett Oskolas
- Section for Clinical Chemistry, Department of Translational Medicine, Lund University, Sweden
| | - Fábio C N Nogueira
- Research Center for Precision Medicine, IBCCF & Chemistry Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gilberto B Domont
- Research Center for Precision Medicine, IBCCF & Chemistry Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Kun-Hsing Yu
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Yevgeniy R Semenov
- Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Peter Sorger
- Department of Systems Biology, Harvrad Medical School, Boston, MA, USA
| | | | - Les Corps
- Alderley Park, Macclesfield, Cheshire, England, United Kingdom
| | | | - Elisabet Wieslander
- Section for Clinical Chemistry, Department of Translational Medicine, Lund University, Sweden
| | - David Fenyö
- Department of Biochemistry and Molecular Pharmacology, Institute for Systems Genetics, New York University Grossman School of Medicine, New York, USA
| | - Sarolta Kárpáti
- Department of Dermatology, Venerology and Dermato oncology, Semmelweis University, Budapest, Hungary
| | - Péter Holló
- Department of Dermatology, Venerology and Dermato oncology, Semmelweis University, Budapest, Hungary
| | - Lajos V Kemény
- Department of Dermatology, Venerology and Dermato oncology, Semmelweis University, Budapest, Hungary; Department of Physiology, HCEMM-SU Translational Dermatology Research Group, Semmelweis University, Budapest, Hungary
| | - Balazs Döme
- Section for Clinical Chemistry, Department of Translational Medicine, Lund University, Sweden; Department of Thoracic Surgery, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria; Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology, Budapest, Hungary; National Koranyi Institute of Pulmonology, Budapest, Hungary
| | - Zsolt Megyesfalvi
- Department of Thoracic Surgery, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria; Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology, Budapest, Hungary; National Koranyi Institute of Pulmonology, Budapest, Hungary
| | - Krzysztof Pawłowski
- Department of Molecular Biology, University of Texas Southwestern Medical Center, TX, USA
| | | | - HoJeong Kwon
- Chemical Genomics Global Research Lab, Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | | | - A Marcell Szasz
- Department of Internal Medicine and Oncology, Semmelweis University, Budapest, Hungary
| | - Zoltán Veréb
- University Hospital Szeged Biobank, Szeged, Hungary
| | - Rolland Gyulai
- Department of Dermatology and Allergology, University of Szeged, Szeged, Hungary
| | - István Balázs Németh
- Department of Dermatology and Allergology, University of Szeged, Szeged, Hungary
| | - Roger Appelqvist
- Clinical Protein Science & Imaging, Biomedical Centre, Department of Biomedical Engineering, Lund University, Sweden
| | - Melinda Rezeli
- Clinical Protein Science & Imaging, Biomedical Centre, Department of Biomedical Engineering, Lund University, Sweden
| | - Bo Baldetorp
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Peter Horvatovich
- Department of Analytical Biochemistry, Faculty of Science and Engineering, University of Groningen, Groningen, the Netherlands
| | - Johan Malmström
- Division of Infection Medicine, Department of Clinical Sciences, Lund University, Lund, Sweden; BioMS, Department of Mass Spectrometry, Lund University, Lund, Sweden
| | - Indira Pla
- Proteomics Center of Excellence, Northwestern University, Evanston, IL, USA
| | - Aniel Sanchez
- Proteomics Center of Excellence, Northwestern University, Evanston, IL, USA
| | - Beatrice Knudsen
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - András Kiss
- 2nd Department of Pathology, Semmelweis University, Budapest, Hungary
| | - Johan Malm
- Section for Clinical Chemistry, Department of Translational Medicine, Lund University, Sweden
| | - György Marko-Varga
- Board of Directors, Japan Society of Clinical Proteogenomics, Tokyo, Japan; Department of Dermatology and Allergology, University of Szeged, Szeged, Hungary; 1st Department of Surgery, Tokyo Medical University, Tokyo, Japan
| | - Jeovanis Gil
- Section for Clinical Chemistry, Department of Translational Medicine, Lund University, Sweden.
| |
Collapse
|
5
|
Ji S, Cao L, Gao J, Du Y, Ye Z, Lou X, Liu F, Zhang Y, Xu J, Shi X, Wang H, Li P, Li Y, Chen H, Yang Z, Gao S, Zhang W, Huang D, Ni S, Wei M, Wang F, Wang Y, Ding T, Jing D, Fan G, Gong Z, Lu R, Qin Y, Chen J, Xu X, Wang P, Zhang B, Ding L, Robles AI, Rodriguez H, Chang DK, Hruban RH, Gao D, Gao D, Jin G, Zhou H, Wu J, Yu X. Proteogenomic characterization of non-functional pancreatic neuroendocrine tumors unravels clinically relevant subgroups. Cancer Cell 2025; 43:776-796.e14. [PMID: 40185092 DOI: 10.1016/j.ccell.2025.03.016] [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: 06/11/2024] [Revised: 01/27/2025] [Accepted: 03/12/2025] [Indexed: 04/07/2025]
Abstract
The majority of neuroendocrine neoplasms in pancreas are non-functional pancreatic neuroendocrine tumors (NF-PanNETs), which exhibit a high occurrence of distant metastases with limited therapeutic options. Here, we perform a comprehensive molecular characterization of 108 NF-PanNETs through integrative analysis of genomic, transcriptomic, proteomic, and phosphoproteomic profiles. Proteogenomic analysis provides functional insights into the genomic driver alterations of NF-PanNETs, revealing a potential mediator of MEN1 alterations using Men1-conditional knockout mice. Machine-learning-based modeling uncovers a three-protein signature as an independent prognostic factor, which is validated by an independent external cohort. Proteomic and phosphoproteomic-based stratification identifies four subtypes with distinct molecular characteristics, immune microenvironments, and clinicopathological features. Drug screening using patient-derived tumor organoids identifies cyclin-dependent kinase (CDK) 5 and Calcium Voltage-Gated Channel Subunit Alpha1 D (CACNA1D) as ubiquitous and subtype-specific targets, respectively, with in vivo validation using xenograft models. Together, our proteogenomic analyses illustrate a comprehensive molecular landscape of NF-PanNETs, revealing biological insights and therapeutic vulnerabilities.
Collapse
Affiliation(s)
- Shunrong Ji
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Lihua Cao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Center for Cancer Bioinformatics, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Jing Gao
- Department of Analytical Chemistry, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yang Du
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Center for Cancer Bioinformatics, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Zeng Ye
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Xin Lou
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Fen Liu
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yehan Zhang
- Key Laboratory of Multi-Cell Systems, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Junfeng Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai 200032, China
| | - Xiaohan Shi
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai 200433, China
| | - Huan Wang
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai 200433, China
| | - Penghao Li
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai 200433, China
| | - Yikai Li
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai 200433, China
| | - Hongxu Chen
- Department of Analytical Chemistry, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Zhicheng Yang
- Department of Analytical Chemistry, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Number 19A Yuquan Road, Beijing 100049, China
| | - Suizhi Gao
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai 200433, China
| | - Wuhu Zhang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai 200032, China
| | - Dan Huang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Shujuan Ni
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Miaoyan Wei
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai 200032, China
| | - Fei Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai 200032, China
| | - Yan Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai 200032, China
| | - Tian Ding
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai 200032, China
| | - Desheng Jing
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai 200032, China
| | - Guixiong Fan
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai 200032, China
| | - Zhiyun Gong
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Renquan Lu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Yi Qin
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai 200032, China
| | - Jie Chen
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Center for Neuroendocrine Tumors, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Xiaowu Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai 200032, China
| | - Pei Wang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NewYork, NY 10029, USA
| | - Bing Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Li Ding
- Department of Medicine, McDonnell Genome Institute, Washington University, St. Louis, MO 63108, USA
| | - Ana I Robles
- Office of Cancer Clinical Proteomics Research, National Cancer Institute, Rockville, MD 20850, USA
| | - Henry Rodriguez
- Office of Cancer Clinical Proteomics Research, National Cancer Institute, Rockville, MD 20850, USA
| | - David K Chang
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1BD, UK
| | - Ralph H Hruban
- The Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Dong Gao
- Key Laboratory of Multi-Cell Systems, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Daming Gao
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Number 19A Yuquan Road, Beijing 100049, China; Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China.
| | - Gang Jin
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai 200433, China.
| | - Hu Zhou
- Department of Analytical Chemistry, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Number 19A Yuquan Road, Beijing 100049, China; School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China; Shanghai Institute of Materia Medica-University of Ottawa Joint Research Center in Systems and Personalized Pharmacology, 555 Zuchongzhi Road, Shanghai 201203, China.
| | - Jianmin Wu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Center for Cancer Bioinformatics, Peking University Cancer Hospital & Institute, Beijing 100142, China; Peking University International Cancer Institute, Peking University, Beijing 100191, China.
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai 200032, China.
| |
Collapse
|
6
|
Lukyanov DK, Kriukova VV, Ladell K, Shagina IA, Staroverov DB, Minasian BE, Fedosova AS, Shelyakin P, Suchalko ON, Komkov AY, Blagodatskikh KA, Miners KL, Britanova OV, Franke A, Price DA, Chudakov DM. Repertoire-based mapping and time-tracking of T helper cell subsets in scRNA-Seq. Front Immunol 2025; 16:1536302. [PMID: 40255395 PMCID: PMC12006041 DOI: 10.3389/fimmu.2025.1536302] [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: 11/28/2024] [Accepted: 02/21/2025] [Indexed: 04/22/2025] Open
Abstract
Introduction The functional programs of CD4+ T helper (Th) cell clones play a central role in shaping immune responses to different challenges. While advances in single-cell RNA sequencing (scRNA-Seq) have significantly improved our understanding of the diversity of Th cells, the relationship between scRNA-Seq clusters and the traditionally characterized Th subsets remains ambiguous. Methods In this study, we introduce TCR-Track, a method leveraging immune repertoire data to map phenotypically sorted Th subsets onto scRNA-Seq profiles. Results and discussion This approach accurately positions the Th1, Th1-17, Th17, Th22, Th2a, Th2, T follicular helper (Tfh), and regulatory T-cell (Treg) subsets, outperforming mapping based on CITE-Seq. Remarkably, the mapping is tightly focused on specific scRNA-Seq clusters, despite 4-year interval between subset sorting and the effector CD4+ scRNA-Seq experiment. These findings highlight the intrinsic program stability of Th clones circulating in peripheral blood. Repertoire overlap analysis at the scRNA-Seq level confirms that the circulating Th1, Th2, Th2a, Th17, Th22, and Treg subsets are clonally independent. However, a significant clonal overlap between the Th1 and cytotoxic CD4+ T-cell clusters suggests that cytotoxic CD4+ T cells differentiate from Th1 clones. In addition, this study resolves a longstanding ambiguity: we demonstrate that, while CCR10+ Th cells align with a specific Th22 scRNA-Seq cluster, CCR10-CCR6+CXCR3-CCR4+ cells, typically classified as Th17, represent a mixture of bona fide Th17 cells and clonally unrelated CCR10low Th22 cells. The clear distinction between the Th17 and Th22 subsets should influence the development of vaccine- and T-cell-based therapies. Furthermore, we show that severe acute SARS-CoV-2 infection induces systemic type 1 interferon (IFN) activation of naive Th cells. An increased proportion of effector IFN-induced Th cells is associated with a moderate course of the disease but remains low in critical COVID-19 cases. Using integrated scRNA-Seq, TCR-Track, and CITE-Seq data from 122 donors, we provide a comprehensive Th scRNA-Seq reference that should facilitate further investigation of Th subsets in fundamental and clinical studies.
Collapse
Affiliation(s)
- Daniil K. Lukyanov
- Center for Molecular and Cellular Biology, Moscow, Russia
- Genomics of Adaptive Immunity Department, Institute of Bioorganic Chemistry, Moscow, Russia
| | | | - Kristin Ladell
- Division of Infection and Immunity, Cardiff University School of Medicine, University Hospital of Wales, Cardiff, United Kingdom
| | - Irina A. Shagina
- Genomics of Adaptive Immunity Department, Institute of Bioorganic Chemistry, Moscow, Russia
- Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Dmitry B. Staroverov
- Genomics of Adaptive Immunity Department, Institute of Bioorganic Chemistry, Moscow, Russia
- Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia
| | | | | | - Pavel Shelyakin
- Abu Dhabi Stem Cell Center, Al Muntazah, United Arab Emirates
| | | | | | | | - Kelly L. Miners
- Division of Infection and Immunity, Cardiff University School of Medicine, University Hospital of Wales, Cardiff, United Kingdom
| | - Olga V. Britanova
- Genomics of Adaptive Immunity Department, Institute of Bioorganic Chemistry, Moscow, Russia
- Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia
- Abu Dhabi Stem Cell Center, Al Muntazah, United Arab Emirates
| | - Andre Franke
- Institute of Clinical Molecular Biology, Kiel University, Kiel, Germany
| | - David A. Price
- Division of Infection and Immunity, Cardiff University School of Medicine, University Hospital of Wales, Cardiff, United Kingdom
- Systems Immunity Research Institute, Cardiff University School of Medicine, University Hospital of Wales, Cardiff, United Kingdom
| | - Dmitry M. Chudakov
- Center for Molecular and Cellular Biology, Moscow, Russia
- Genomics of Adaptive Immunity Department, Institute of Bioorganic Chemistry, Moscow, Russia
- Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia
- Abu Dhabi Stem Cell Center, Al Muntazah, United Arab Emirates
- Department of Molecular Medicine, Central European Institute of Technology, Brno, Czechia
| |
Collapse
|
7
|
Zhu Q, Balasubramanian A, Asirvatham JR, Chatterjee M, Piyarathna B, Kaur J, Mohamed N, Wu L, Wang S, Pourfarrokh N, Binsol PD, Bhargava M, Rasaily U, Xu Y, Zheng J, Jebakumar D, Rao A, Gutierrez C, Omilian A, Morrison C, Das GM, Ambrosone C, Seeley EH, Chen SH, Li Y, Chang E, Li X, Baker E, Aneja R, Zhang XHF, Sreekumar A. Integrative spatial omics reveals distinct tumor-promoting multicellular niches and immunosuppressive mechanisms in Black American and White American patients with TNBC. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.03.17.585428. [PMID: 38562769 PMCID: PMC10983891 DOI: 10.1101/2024.03.17.585428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Racial disparities in the clinical outcomes of triple-negative breast cancer (TNBC) have been well-documented, but the underlying biological mechanisms remain poorly understood. To investigate these disparities, we employed a multi-omic approach integrating imaging mass cytometry and spatial transcriptomics to characterize the tumor microenvironment (TME) in self-identified Black American (BA) and White American (WA) TNBC patients. Our analysis revealed that the TME in BA patients is marked by a network of endothelial cells, macrophages, and mesenchymal-like cells, which correlates with reduced patient survival. In contrast, the WA TNBC microenvironment is enriched in T-cells and neutrophils, indicative of T-cell exhaustion and suppressed immune responses. Ligand-receptor and pathway analyses further demonstrated that BA TNBC tumors exhibit a relatively "immune-cold" profile, while WA TNBC tumors display features of an "inflamed" TME, suggesting the evolution of a unique immunosuppressive mechanism. These findings provide insight into racially distinct tumor-promoting and immunosuppressive microenvironments, which may contribute to the observed differences in clinical outcomes among BA and WA TNBC patients. Statement of Significance This study identifies distinct tumor microenvironment (TME) profiles in Black and White American TNBC patients, providing new insights into the biological mechanisms driving outcome disparities. Our findings highlight the role of the tumor-endothelial-macrophage niche in these disparities, offering a potential therapeutic target for race-inclusive strategies aimed at improving clinical outcomes. By revealing racial differences in treatment response profiles, this work underscores the necessity for tailored therapies in TNBC. These insights lay the groundwork for the development of inclusive, precision-driven treatment approaches that may help mitigate racial disparities and enhance patient outcomes.
Collapse
|
8
|
Zeltser N, Zhu C, Oh J, Li CH, Boutros PC. Sex Differences in Cancer Functional Genomics: Gene Dependency and Drug Sensitivity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.05.636540. [PMID: 39975298 PMCID: PMC11838570 DOI: 10.1101/2025.02.05.636540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Patient sex influences a wide range of cancer phenotypes, including prevalence, response to therapy and survival endpoints. Molecular sex differences have been identified at all levels of the central dogma. It is hypothesized that these molecular differences may drive the observed clinical sex differences. Yet despite a growing catalog of molecular sex differences in a range of cancer types, their specific functional consequences remain unclear. To directly assess how patient sex impacts cancer cell function, we evaluated 1,209 cell lines subjected to CRISPR knockout, RNAi knockdown or drug exposures. Despite limited statistical power, we identified pan- and per-cancer sex differences in gene essentiality in six sex-linked and fourteen autosomal genes, and in drug sensitivity for two compounds. These data fill a gap in our understanding of the link between sex-differential molecular effects and patient phenotypes. They call for much more careful and systematic consideration of sex-specific effects in mechanistic and functional studies.
Collapse
Affiliation(s)
- Nicole Zeltser
- Department of Human Genetics, University of California, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA
- Department of Urology, University of California, Los Angeles, CA, USA
- Institute for Precision Health, University of California, Los Angeles, CA, USA
- Broad Stem Cell Research Center, University of California, Los Angeles, CA, USA
| | - Chenghao Zhu
- Department of Human Genetics, University of California, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA
- Department of Urology, University of California, Los Angeles, CA, USA
- Institute for Precision Health, University of California, Los Angeles, CA, USA
- Broad Stem Cell Research Center, University of California, Los Angeles, CA, USA
| | - Jieun Oh
- Department of Human Genetics, University of California, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA
- Department of Urology, University of California, Los Angeles, CA, USA
- Institute for Precision Health, University of California, Los Angeles, CA, USA
- Broad Stem Cell Research Center, University of California, Los Angeles, CA, USA
| | - Constance H. Li
- Department of Human Genetics, University of California, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA
- Department of Urology, University of California, Los Angeles, CA, USA
- Institute for Precision Health, University of California, Los Angeles, CA, USA
- Broad Stem Cell Research Center, University of California, Los Angeles, CA, USA
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Paul C. Boutros
- Department of Human Genetics, University of California, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA
- Department of Urology, University of California, Los Angeles, CA, USA
- Institute for Precision Health, University of California, Los Angeles, CA, USA
- Broad Stem Cell Research Center, University of California, Los Angeles, CA, USA
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
9
|
Grothey B, Lyu SI, Quaas A, Simon AG, Jung JO, Schröder W, Bruns CJ, Schiffmann LM, Popp FC, Schmidt T, Knipper K. Proteomic characterization of MET-amplified esophageal adenocarcinomas reveals enrichment of alternative splicing- and androgen signaling-related proteins. Cell Mol Life Sci 2025; 82:112. [PMID: 40074836 PMCID: PMC11904063 DOI: 10.1007/s00018-025-05635-7] [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: 09/10/2024] [Revised: 02/11/2025] [Accepted: 02/20/2025] [Indexed: 03/14/2025]
Abstract
BACKGROUND Esophageal adenocarcinomas (EACs) represent an evolving tumor entity with high mortality rates. MET amplification is a recurrent driver in EACs and is associated with decreased patient survival. However, the response to MET inhibitors is limited. Recent studies have identified several mechanisms that lead to resistance against MET inhibitors in different tumor entities. Nonetheless, a characterization of additional vulnerable targets beyond MET has not been conducted in MET-amplified EACs. METHODS In this study, we determined the MET amplification status in a cohort of more than 900 EACs using fluorescence in situ hybridization (FISH) and compared the proteomes of MET-amplified (n = 20) versus non-amplified tumors (n = 39) by mass spectrometry. RESULTS We identified a phenotype, present in almost all MET-amplified tumors, which shows an enrichment of alternative RNA splicing, and androgen receptor signaling proteins, as well as decreased patient survival. Additionally, our analyses revealed a negative correlation between MET expression and patient survival in MET-amplified EACs, indicating biological heterogeneity with clinical relevance despite the presence of MET amplification as the predominant oncogenic driver. Furthermore, quantitative immunohistochemical analysis of the inflammatory tumor microenvironment showed that an increased percentage of M2 macrophages is associated with lower overall survival in MET-amplified EACs. CONCLUSIONS Our results provide valuable insights into possible new therapeutic approaches for MET-amplified EACs for further research.
Collapse
Affiliation(s)
- Bastian Grothey
- Faculty of Medicine, Institute of Pathology, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Su Ir Lyu
- Faculty of Medicine, Institute of Pathology, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Alexander Quaas
- Faculty of Medicine, Institute of Pathology, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Adrian Georg Simon
- Faculty of Medicine, Institute of Pathology, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Jin-On Jung
- Faculty of Medicine, Department of General, Visceral and Cancer Surgery, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Wolfgang Schröder
- Faculty of Medicine, Department of General, Visceral and Cancer Surgery, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Christiane J Bruns
- Faculty of Medicine, Department of General, Visceral and Cancer Surgery, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Lars M Schiffmann
- Faculty of Medicine, Department of General, Visceral and Cancer Surgery, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Felix C Popp
- Faculty of Medicine, Department of General, Visceral and Cancer Surgery, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Thomas Schmidt
- Faculty of Medicine, Department of General, Visceral and Cancer Surgery, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Karl Knipper
- Faculty of Medicine, Department of General, Visceral and Cancer Surgery, University Hospital of Cologne, University of Cologne, Cologne, Germany.
| |
Collapse
|
10
|
Zhang J, Xiong A, Yang Y, Cao Y, Yang M, Su C, Lei M, Chen Y, Shen X, Wang P, Shi C, Zhou R, Ren N, Zhu H, Yuan C, Liu S, Teng F. In-Depth Proteomic Analysis of Tissue Interstitial Fluid Reveals Biomarker Candidates Related to Varying Differentiation Statuses in Gastric Adenocarcinoma. J Proteome Res 2025; 24:1386-1401. [PMID: 39912886 DOI: 10.1021/acs.jproteome.4c01067] [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] [Indexed: 02/07/2025]
Abstract
The proteomic heterogeneity of gastric adenocarcinoma (GC) has been extensively investigated at the bulk tissue level, which can only provide an average molecular state. In this study, we collected an in-depth quantitative proteomic dataset of tissues and interstitial fluids (ISFs) from both poorly and non-poorly differentiated GC and presented a comprehensive analysis from several perspectives. Comparison of proteomes between ISFs and tissues revealed that ISF exhibited higher abundances of proteins associated with blood microparticles, protein-lipid complexes, immunoglobulin complexes, and high-density lipoprotein particles. Also, consistent and inconsistent protein abundance changes between them were revealed by a correlation analysis. Interestingly, a more pronounced difference between tumors and normal adjacent tissues was found at the ISF level, which accurately reflected tissue properties compared to those of bulk tissue. Two ISF-derived biomarker candidates, calsyntenin-1 (CLSTN1) and prosaposin (PSAP), were identified by distinguishing patients with different differentiation statuses and were further validated in serum samples. Additionally, the silencing of CLSTN1 and PSAP was demonstrated to suppress cell proliferation, migration, and invasion in poorly differentiated gastric cancer cell lines. In summary, the ISF proteome offers a new perspective on tumor biology. This study provides a valuable resource that significantly enhances the understanding of GC and may ultimately benefit clinical practice.
Collapse
Affiliation(s)
- Juxiang Zhang
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering, Institute of Medical Robotics and Shanghai Academy of Experimental Medicine, Shanghai Jiao Tong University, Shanghai 200030, P. R. China
| | - An Xiong
- Department of Gastrointestinal Surgery, Minhang Hospital, Fudan University, Shanghai 201199, P. R. China
- Key Laboratory of Whole-period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital & AHS, Fudan University, Shanghai 201199, P. R. China
| | - Yuanyuan Yang
- Key Laboratory of Whole-period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital & AHS, Fudan University, Shanghai 201199, P. R. China
- Department of Pathology, Minhang Hospital & School of Pharmacy, Fudan University, Shanghai 201199, P. R. China
| | - Yiou Cao
- Department of Gastrointestinal Surgery, Minhang Hospital, Fudan University, Shanghai 201199, P. R. China
- Key Laboratory of Whole-period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital & AHS, Fudan University, Shanghai 201199, P. R. China
| | - Mengxuan Yang
- Department of Gastrointestinal Surgery, Minhang Hospital, Fudan University, Shanghai 201199, P. R. China
- Key Laboratory of Whole-period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital & AHS, Fudan University, Shanghai 201199, P. R. China
| | - Chang Su
- Department of Gastrointestinal Surgery, Minhang Hospital, Fudan University, Shanghai 201199, P. R. China
- Key Laboratory of Whole-period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital & AHS, Fudan University, Shanghai 201199, P. R. China
| | - Ming Lei
- Department of Gastrointestinal Surgery, Minhang Hospital, Fudan University, Shanghai 201199, P. R. China
- Key Laboratory of Whole-period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital & AHS, Fudan University, Shanghai 201199, P. R. China
| | - Yi Chen
- Department of Gastrointestinal Surgery, Minhang Hospital, Fudan University, Shanghai 201199, P. R. China
- Key Laboratory of Whole-period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital & AHS, Fudan University, Shanghai 201199, P. R. China
| | - Xiaodong Shen
- Department of Gastrointestinal Surgery, Minhang Hospital, Fudan University, Shanghai 201199, P. R. China
- Key Laboratory of Whole-period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital & AHS, Fudan University, Shanghai 201199, P. R. China
| | - Puhua Wang
- Department of Gastrointestinal Surgery, Minhang Hospital, Fudan University, Shanghai 201199, P. R. China
- Key Laboratory of Whole-period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital & AHS, Fudan University, Shanghai 201199, P. R. China
| | - Chencheng Shi
- Department of Gastrointestinal Surgery, Minhang Hospital, Fudan University, Shanghai 201199, P. R. China
- Key Laboratory of Whole-period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital & AHS, Fudan University, Shanghai 201199, P. R. China
| | - Rongjian Zhou
- Department of Gastrointestinal Surgery, Minhang Hospital, Fudan University, Shanghai 201199, P. R. China
- Key Laboratory of Whole-period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital & AHS, Fudan University, Shanghai 201199, P. R. China
| | - Ning Ren
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, P. R. China
| | - Hongwen Zhu
- Precise Genome Engineering Center, School of Life Sciences, Guangzhou University, 510006 Guangzhou, China
| | - Chunyan Yuan
- Key Laboratory of Whole-period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital & AHS, Fudan University, Shanghai 201199, P. R. China
| | - Shaoqun Liu
- Department of Gastrointestinal Surgery, Minhang Hospital, Fudan University, Shanghai 201199, P. R. China
- Key Laboratory of Whole-period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital & AHS, Fudan University, Shanghai 201199, P. R. China
| | - Fei Teng
- Department of Gastrointestinal Surgery, Minhang Hospital, Fudan University, Shanghai 201199, P. R. China
- Key Laboratory of Whole-period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital & AHS, Fudan University, Shanghai 201199, P. R. China
| |
Collapse
|
11
|
Tran D, Nguyen H, Pham VD, Nguyen P, Nguyen Luu H, Minh Phan L, Blair DeStefano C, Jim Yeung SC, Nguyen T. A comprehensive review of cancer survival prediction using multi-omics integration and clinical variables. Brief Bioinform 2025; 26:bbaf150. [PMID: 40221959 PMCID: PMC11994034 DOI: 10.1093/bib/bbaf150] [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: 10/27/2024] [Revised: 01/29/2025] [Accepted: 03/19/2025] [Indexed: 04/15/2025] Open
Abstract
Cancer is an umbrella term that includes a wide spectrum of disease severity, from those that are malignant, metastatic, and aggressive to benign lesions with very low potential for progression or death. The ability to prognosticate patient outcomes would facilitate management of various malignancies: patients whose cancer is likely to advance quickly would receive necessary treatment that is commensurate with the predicted biology of the disease. Former prognostic models based on clinical variables (age, gender, cancer stage, tumor grade, etc.), though helpful, cannot account for genetic differences, molecular etiology, tumor heterogeneity, and important host biological mechanisms. Therefore, recent prognostic models have shifted toward the integration of complementary information available in both molecular data and clinical variables to better predict patient outcomes: vital status (overall survival), metastasis (metastasis-free survival), and recurrence (progression-free survival). In this article, we review 20 survival prediction approaches that integrate multi-omics and clinical data to predict patient outcomes. We discuss their strategies for modeling survival time (continuous and discrete), the incorporation of molecular measurements and clinical variables into risk models (clinical and multi-omics data), how to cope with censored patient records, the effectiveness of data integration techniques, prediction methodologies, model validation, and assessment metrics. The goal is to inform life scientists of available resources, and to provide a complete review of important building blocks in survival prediction. At the same time, we thoroughly describe the pros and cons of each methodology, and discuss in depth the outstanding challenges that need to be addressed in future method development.
Collapse
Affiliation(s)
- Dao Tran
- Department of Computer Science and Software Engineering, Auburn University, 345 W Magnolia Avenue, Auburn, AL 36849, United States
| | - Ha Nguyen
- Department of Computer Science and Software Engineering, Auburn University, 345 W Magnolia Avenue, Auburn, AL 36849, United States
| | - Van-Dung Pham
- Department of Computer Science and Software Engineering, Auburn University, 345 W Magnolia Avenue, Auburn, AL 36849, United States
| | - Phuong Nguyen
- Department of Computer Science and Software Engineering, Auburn University, 345 W Magnolia Avenue, Auburn, AL 36849, United States
| | - Hung Nguyen Luu
- UPMC Hillman Cancer Center, University of Pittsburgh Medical Center, 5150 Centre Avenue, Pittsburgh, PA 15232, United States
- Department of Epidemiology, School of Public Health, University of Pittsburgh, 130 De Soto Street, Pittsburgh, PA 15261, United States
| | - Liem Minh Phan
- David Grant USAF Medical Center—Clinical Investigation Facility, 60 Medical Group, Defense Health Agency, 101 Bodin Circle, Travis Air Force Base, CA 94535, United States
| | - Christin Blair DeStefano
- Walter Reed National Military Medical Center, Defense Health Agency, 8901 Rockville Pike, Bethesda, MD 20889, United States
| | - Sai-Ching Jim Yeung
- Department of Emergency Medicine, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, Houston, TX 77030, United States
| | - Tin Nguyen
- Department of Computer Science and Software Engineering, Auburn University, 345 W Magnolia Avenue, Auburn, AL 36849, United States
| |
Collapse
|
12
|
Xu X, Su J, Zhu R, Li K, Zhao X, Fan J, Mao F. From morphology to single-cell molecules: high-resolution 3D histology in biomedicine. Mol Cancer 2025; 24:63. [PMID: 40033282 DOI: 10.1186/s12943-025-02240-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 01/18/2025] [Indexed: 03/05/2025] Open
Abstract
High-resolution three-dimensional (3D) tissue analysis has emerged as a transformative innovation in the life sciences, providing detailed insights into the spatial organization and molecular composition of biological tissues. This review begins by tracing the historical milestones that have shaped the development of high-resolution 3D histology, highlighting key breakthroughs that have facilitated the advancement of current technologies. We then systematically categorize the various families of high-resolution 3D histology techniques, discussing their core principles, capabilities, and inherent limitations. These 3D histology techniques include microscopy imaging, tomographic approaches, single-cell and spatial omics, computational methods and 3D tissue reconstruction (e.g. 3D cultures and spheroids). Additionally, we explore a wide range of applications for single-cell 3D histology, demonstrating how single-cell and spatial technologies are being utilized in the fields such as oncology, cardiology, neuroscience, immunology, developmental biology and regenerative medicine. Despite the remarkable progress made in recent years, the field still faces significant challenges, including high barriers to entry, issues with data robustness, ambiguous best practices for experimental design, and a lack of standardization across methodologies. This review offers a thorough analysis of these challenges and presents recommendations to surmount them, with the overarching goal of nurturing ongoing innovation and broader integration of cellular 3D tissue analysis in both biology research and clinical practice.
Collapse
Affiliation(s)
- Xintian Xu
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
- Cancer Center, Peking University Third Hospital, Beijing, China
- Department of Biochemistry and Molecular Biology, Beijing, Key Laboratory of Protein Posttranslational Modifications and Cell Function, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Jimeng Su
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
- Cancer Center, Peking University Third Hospital, Beijing, China
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, China
| | - Rongyi Zhu
- Department of Biochemistry and Molecular Biology, Beijing, Key Laboratory of Protein Posttranslational Modifications and Cell Function, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Kailong Li
- Department of Biochemistry and Molecular Biology, Beijing, Key Laboratory of Protein Posttranslational Modifications and Cell Function, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Xiaolu Zhao
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and GynecologyNational Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital)Key Laboratory of Assisted Reproduction (Peking University), Ministry of EducationBeijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Peking University Third Hospital, Beijing, China.
| | - Jibiao Fan
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, China.
| | - Fengbiao Mao
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China.
- Cancer Center, Peking University Third Hospital, Beijing, China.
- Beijing Key Laboratory for Interdisciplinary Research in Gastrointestinal Oncology (BLGO), Beijing, China.
| |
Collapse
|
13
|
Papaccio F, Cabeza-Segura M, García-Micó B, Gimeno-Valiente F, Zúñiga-Trejos S, Gambardella V, Gutiérrez-Bravo MF, Martinez-Ciarpaglini C, Rentero-Garrido P, Fleitas T, Roselló S, Carbonell-Asins JA, Huerta M, Moro-Valdezate D, Roda D, Tarazona N, Sánchez Del Pino MM, Cervantes A, Castillo J. Decoding chromosomal instability insights in CRC by integrating omics and patient-derived organoids. J Exp Clin Cancer Res 2025; 44:77. [PMID: 40022181 PMCID: PMC11869439 DOI: 10.1186/s13046-025-03308-8] [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: 10/08/2024] [Accepted: 01/28/2025] [Indexed: 03/03/2025] Open
Abstract
BACKGROUND Chromosomal instability (CIN) is involved in about 70% of colorectal cancers (CRCs) and is associated with poor prognosis and drug resistance. From a clinical perspective, a better knowledge of these tumour's biology will help to guide therapeutic strategies more effectively. METHODS We used high-density chromosomal microarray analysis to evaluate CIN level of patient-derived organoids (PDOs) and their original mCRC tissues. We integrated the RNA-seq and mass spectrometry-based proteomics data from PDOs in a functional interaction network to identify the significantly dysregulated processes in CIN. This was followed by a proteome-wGII Pearson correlation analysis and an in silico validation of main findings using functional genomic databases and patient-tissues datasets to prioritize the high-confidence CIN features. RESULTS By applying the weighted Genome Instability Index (wGII) to identify CIN, we classified PDOs and demonstrated a good correlation with tissues. Multi-omics analysis showed that our organoids recapitulated genomic, transcriptomic and proteomic CIN features of independent tissues cohorts. Thanks to proteotranscriptomics, we uncovered significant associations between mitochondrial metabolism and epithelial-mesenchymal transition in CIN CRC PDOs. Correlating PDOs wGII with protein abundance, we identified a subset of proteins significantly correlated with CIN. Co-localisation analysis in PDOs strengthened the putative role of IPO7 and YAP, and, through in silico analysis, we found that some of the targets give significant dependencies in cell lines with CIN compatible status. CONCLUSIONS We first demonstrated that PDO models are a faithful reflection of CIN tissues at the genetic and phenotypic level. Our new findings prioritize a subset of genes and molecular processes putatively required to cope with the burden on cellular fitness imposed by CIN and associated with disease aggressiveness.
Collapse
Affiliation(s)
- Federica Papaccio
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Via S. Allende, 84081, Baronissi, Italy.
- Department of Medical Oncology, Hospital Clínico Universitario de Valencia, INCLIVA Biomedical Research Institute, University of Valencia, Avda. Blasco Ibañez 17, 46010, Valencia, Spain.
| | - Manuel Cabeza-Segura
- Department of Medical Oncology, Hospital Clínico Universitario de Valencia, INCLIVA Biomedical Research Institute, University of Valencia, Avda. Blasco Ibañez 17, 46010, Valencia, Spain
| | - Blanca García-Micó
- Department of Medical Oncology, Hospital Clínico Universitario de Valencia, INCLIVA Biomedical Research Institute, University of Valencia, Avda. Blasco Ibañez 17, 46010, Valencia, Spain
- Centro de Investigación Biomédica en Red (CIBERONC), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Francisco Gimeno-Valiente
- Cancer Evolution and Genome Instability Laboratory, University College London Cancer Institute, London, UK
| | - Sheila Zúñiga-Trejos
- Bioinformatic Unit, INCLIVA Biomedical Research Institute, Avda. Menéndez y Pelayo 3, 46010, Valencia, Spain
| | - Valentina Gambardella
- Department of Medical Oncology, Hospital Clínico Universitario de Valencia, INCLIVA Biomedical Research Institute, University of Valencia, Avda. Blasco Ibañez 17, 46010, Valencia, Spain
- Centro de Investigación Biomédica en Red (CIBERONC), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - María Fernanda Gutiérrez-Bravo
- Experimental and Applied Biomedicine Research Group, Health Sciences Faculty, Universidad Particular Internacional SEK (UISEK), Quito, 170302, Ecuador
| | - Carolina Martinez-Ciarpaglini
- Centro de Investigación Biomédica en Red (CIBERONC), Instituto de Salud Carlos III, 28029, Madrid, Spain
- Department of Pathology, Hospital Clínico Universitario, INCLIVA Biomedical Research Institute, University of Valencia, Avda. Blasco Ibañez 17, 46010, Valencia, Spain
| | - Pilar Rentero-Garrido
- Precision Medicine Unit, INCLIVA Biomedical Research Institute, Avda. Menéndez y Pelayo 4, 46010, Valencia, Spain
| | - Tania Fleitas
- Department of Medical Oncology, Hospital Clínico Universitario de Valencia, INCLIVA Biomedical Research Institute, University of Valencia, Avda. Blasco Ibañez 17, 46010, Valencia, Spain
- Centro de Investigación Biomédica en Red (CIBERONC), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Susana Roselló
- Department of Medical Oncology, Hospital Clínico Universitario de Valencia, INCLIVA Biomedical Research Institute, University of Valencia, Avda. Blasco Ibañez 17, 46010, Valencia, Spain
- Centro de Investigación Biomédica en Red (CIBERONC), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | | | - Marisol Huerta
- Department of Medical Oncology, Hospital Clínico Universitario de Valencia, INCLIVA Biomedical Research Institute, University of Valencia, Avda. Blasco Ibañez 17, 46010, Valencia, Spain
| | - David Moro-Valdezate
- Department of General Surgery, INCLIVA Biomedical Research Institute, Hospital Clínico Universitario de Valencia, University of Valencia, Valencia, Spain
| | - Desamparados Roda
- Department of Medical Oncology, Hospital Clínico Universitario de Valencia, INCLIVA Biomedical Research Institute, University of Valencia, Avda. Blasco Ibañez 17, 46010, Valencia, Spain
- Centro de Investigación Biomédica en Red (CIBERONC), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Noelia Tarazona
- Department of Medical Oncology, Hospital Clínico Universitario de Valencia, INCLIVA Biomedical Research Institute, University of Valencia, Avda. Blasco Ibañez 17, 46010, Valencia, Spain
- Centro de Investigación Biomédica en Red (CIBERONC), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Manuel M Sánchez Del Pino
- Institute of Biotechnology and Biomedicine (BIOTECMED), University of Valencia, 46100, Burjassot, Spain.
- Department of Biochemistry and Molecular Biology, University of Valencia, 46100, Burjassot, Spain.
| | - Andrés Cervantes
- Department of Medical Oncology, Hospital Clínico Universitario de Valencia, INCLIVA Biomedical Research Institute, University of Valencia, Avda. Blasco Ibañez 17, 46010, Valencia, Spain.
- Centro de Investigación Biomédica en Red (CIBERONC), Instituto de Salud Carlos III, 28029, Madrid, Spain.
| | - Josefa Castillo
- Department of Medical Oncology, Hospital Clínico Universitario de Valencia, INCLIVA Biomedical Research Institute, University of Valencia, Avda. Blasco Ibañez 17, 46010, Valencia, Spain.
- Centro de Investigación Biomédica en Red (CIBERONC), Instituto de Salud Carlos III, 28029, Madrid, Spain.
- Department of Biochemistry and Molecular Biology, University of Valencia, 46100, Burjassot, Spain.
| |
Collapse
|
14
|
Liu H, Ma Z, Lih TM, Chen L, Hu Y, Wang Y, Sun Z, Huang Y, Xu Y, Zhang H. Machine Learning-Enhanced Extraction of Protein Signatures of Renal Cell Carcinoma from Proteomics Data. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.17.638651. [PMID: 40027663 PMCID: PMC11870591 DOI: 10.1101/2025.02.17.638651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
In this study, we generated label-free data-independent acquisition (DIA)-based liquid chromatography (LC)-mass spectrometry (MS) proteomics data from 261 renal cell carcinomas (RCC) and 195 normal adjacent tissues (NAT). The RCC tumors included 48 non-clear cell renal cell carcinomas (non-ccRCC) and 213 ccRCC. A total of 219,740 peptides and 11,943 protein groups were identified with 9,787 protein groups per sample on average. We adopted a comprehensive approach to select representative samples with different mutation sites, considering histopathological, immune, methylation, and non-negative matrix factorization (NMF)-based subtypes, along with clinical characteristics (gender, grade, and stage) to capture the complexity and diversity of ccRCC tumors. We used machine learning identified 55 protein signatures that distinguish RCC tumors from NATs. Furthermore, 39 protein signatures that differentiate different RCC tumor subtypes were also identified. Our findings offer an extensive perspective of the proteomic landscape in RCC, illuminating specific proteins that serve to distinguish RCC tumors from NATs and among various RCC tumor subtypes.
Collapse
Affiliation(s)
- Hongyi Liu
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Zhuo Ma
- Krieger school of Arts and Sciences, Johns Hopkins University, MD 21218, USA
| | - T. Mamie Lih
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Lijun Chen
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Yingwei Hu
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Yuefan Wang
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Zhenyu Sun
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Yuanyu Huang
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Yuanwei Xu
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Hui Zhang
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| |
Collapse
|
15
|
Li QK, Lih TM, Clark DJ, Chen L, Schnaubelt M, Zhang H. Sonication-assisted protein extraction improves proteomic detection of membrane-bound and DNA-binding proteins from tumor tissues. Nat Protoc 2025:10.1038/s41596-024-01113-9. [PMID: 39962197 DOI: 10.1038/s41596-024-01113-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 11/15/2024] [Indexed: 03/21/2025]
Abstract
Deep-scale, mass spectrometry-based proteomic studies by the Clinical Proteomic Tumor Analysis Consortium (CPTAC) program involves tissue lysis using urea buffer before data acquisition via mass spectrometry for quantitative global proteomic and phosphoproteomic analysis. This is described in a 2018 protocol1. Here we report an update to this initial protocol by implementing a sonication step into urea-based tissue lysis. Similar to the initial CPTAC protocol, we identified >12,000 proteins and >25,000 phosphopeptides in a tandem mass tag (TMT) set containing both nonsonicated and sonicated tumor tissues from patient-derived xenograft mouse models. An improvement in the detection of membrane-bound and DNA-binding proteins was observed by including the sonication. We also offer recommendations for optimal sonication conditions such as the buffer composition, timing of sonication cycle, instrumentation settings and a troubleshooting section for potential users. Additionally, the protocol is equally applicable to other biological specimens.
Collapse
Affiliation(s)
- Qing Kay Li
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, USA.
| | - T Mamie Lih
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - David J Clark
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Lijun Chen
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Michael Schnaubelt
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Hui Zhang
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, USA.
| |
Collapse
|
16
|
Madoz-Gúrpide J, Serrano-López J, Sanz-Álvarez M, Morales-Gallego M, Rodríguez-Pinilla SM, Rovira A, Albanell J, Rojo F. Adaptive Proteomic Changes in Protein Metabolism and Mitochondrial Alterations Associated with Resistance to Trastuzumab and Pertuzumab Therapy in HER2-Positive Breast Cancer. Int J Mol Sci 2025; 26:1559. [PMID: 40004024 PMCID: PMC11855744 DOI: 10.3390/ijms26041559] [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: 12/16/2024] [Revised: 01/30/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
HER2 (human epidermal growth factor receptor 2) is overexpressed in approximately 15-20% of breast cancers, leading to aggressive tumour growth and poor prognosis. Anti-HER2 therapies, such as trastuzumab and pertuzumab, have significantly improved the outcomes for patients with HER2-positive breast cancer by blocking HER2 signalling. However, intrinsic and acquired resistance remains a major clinical challenge, limiting the long-term effectiveness of these therapies. Understanding the mechanisms of resistance is essential for developing strategies to overcome it and improve the therapeutic outcomes. We generated multiple HER2-positive breast cancer cell line models resistant to trastuzumab and pertuzumab combination therapy. Using mass spectrometry-based proteomics, we conducted a comprehensive analysis to identify the mechanisms underlying resistance. Proteomic analysis identified 618 differentially expressed proteins, with a core of 83 overexpressed and 118 downregulated proteins. Through a series of advanced bioinformatics analyses, we identified significant protein alterations and signalling pathways potentially responsible for the development of resistance, revealing key alterations in the protein metabolism, mitochondrial function, and signalling pathways, such as MAPK, TNF, and TGFβ. These findings identify mitochondrial activity and detoxification processes as pivotal mechanisms underlying the resistance to anti-HER2 therapy. Additionally, we identified key proteins, including ANXA1, SLC2A1, and PPIG, which contribute to the tumour progression and resistance phenotype. Our study suggests that targeting these pathways and proteins could form the basis of novel therapeutic strategies to overcome resistance in HER2-positive breast cancer.
Collapse
MESH Headings
- Humans
- Trastuzumab/therapeutic use
- Trastuzumab/pharmacology
- Breast Neoplasms/drug therapy
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Breast Neoplasms/genetics
- Drug Resistance, Neoplasm
- Female
- Receptor, ErbB-2/metabolism
- Mitochondria/metabolism
- Mitochondria/drug effects
- Proteomics/methods
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/pharmacology
- Cell Line, Tumor
- Signal Transduction/drug effects
- Gene Expression Regulation, Neoplastic/drug effects
- Antineoplastic Agents, Immunological/therapeutic use
- Antineoplastic Agents, Immunological/pharmacology
- Proteome/metabolism
Collapse
Affiliation(s)
- Juan Madoz-Gúrpide
- Department of Pathology, Fundación Jiménez Díaz University Hospital Health Research Institute (IIS—FJD, UAM)—CIBERONC, 28040 Madrid, Spain (M.M.-G.); (S.M.R.-P.)
| | - Juana Serrano-López
- Department of Haematology, Fundación Jiménez Díaz University Hospital Health Research Institute (IIS—FJD, UAM)—CIBERONC, 28040 Madrid, Spain;
| | - Marta Sanz-Álvarez
- Department of Pathology, Fundación Jiménez Díaz University Hospital Health Research Institute (IIS—FJD, UAM)—CIBERONC, 28040 Madrid, Spain (M.M.-G.); (S.M.R.-P.)
| | - Miriam Morales-Gallego
- Department of Pathology, Fundación Jiménez Díaz University Hospital Health Research Institute (IIS—FJD, UAM)—CIBERONC, 28040 Madrid, Spain (M.M.-G.); (S.M.R.-P.)
| | - Socorro María Rodríguez-Pinilla
- Department of Pathology, Fundación Jiménez Díaz University Hospital Health Research Institute (IIS—FJD, UAM)—CIBERONC, 28040 Madrid, Spain (M.M.-G.); (S.M.R.-P.)
| | - Ana Rovira
- Cancer Research Program, IMIM (Hospital del Mar Research Institute), 08003 Barcelona, Spain;
| | - Joan Albanell
- Department of Medical Oncology, Hospital del Mar—CIBERONC, 08003 Barcelona, Spain;
| | - Federico Rojo
- Department of Pathology, Fundación Jiménez Díaz University Hospital Health Research Institute (IIS—FJD, UAM)—CIBERONC, 28040 Madrid, Spain (M.M.-G.); (S.M.R.-P.)
| |
Collapse
|
17
|
Wen M, Qiu Y, Wang M, Tang F, Hu W, Zhu Y, Zhao W, Hu W, Chen Z, Duan Y, Geng A, Tan F, Li Y, Pei Q, Pei H, Mao Z, Wu N, Sun L, Tan R. Enhancing low-dose radiotherapy efficacy with PARP inhibitors via FBL-mediated oxidative stress response in colorectal cancer. Oncogene 2025; 44:228-240. [PMID: 39516657 PMCID: PMC11746129 DOI: 10.1038/s41388-024-03207-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 10/17/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024]
Abstract
The effectiveness of radiotherapy in colorectal cancer (CRC) relies on its ability to induce cell death via the generation of reactive oxygen species (ROS). However, genes responsible for mitigating oxidative stress can impede radiotherapy's efficacy. In this study, we elucidate a significant association between the nucleolar protein Fibrillarin (FBL) and the oxidative stress response in CRC tumors. Our findings reveal elevated expression of FBL in colorectal cancer, which positively correlates with oxidative stress levels. Mechanistically, FBL demonstrates direct accumulation at DNA damage sites under the regulation of PARP1. Specifically, the N-terminal GAR domain of FBL is susceptible to PARylation by PARP1, enabling FBL to recognize PARylated proteins. The accumulation of damaged FBL plays a pivotal role in facilitating short-patched base excision repair by recruiting Ligase III and disassociating PCNA and FEN1. Moreover, tumors with heightened FBL expression exhibit reduced DNA damage levels but increased sensitivity to combined low-dose radiotherapy and olaparib treatment. This underscores the potential of leveraging PARP inhibitors to augment radiotherapy sensitivity in CRC cases characterized by elevated FBL expression, offering a promising therapeutic avenue.
Collapse
Affiliation(s)
- Ming Wen
- Department of Oncology, Xiangya Cancer Center, Xiangya Hospital, Central South University, Changsha, 410008, China
- Key Laboratory of Molecular Radiation Oncology Hunan Province, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Huan, 410008, China
- Hunan International Science and Technology Collaboration Base of Precision Medicine for Cancer, Changsha, 410008, China
- Center for Molecular Imaging of Central South University, Xiangya Hospital, Changsha, 410008, China
| | - Yanfang Qiu
- Department of Oncology, Xiangya Cancer Center, Xiangya Hospital, Central South University, Changsha, 410008, China
- Key Laboratory of Molecular Radiation Oncology Hunan Province, Changsha, 410008, China
- Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Meng Wang
- Department of Oncology, Xiangya Cancer Center, Xiangya Hospital, Central South University, Changsha, 410008, China
- Key Laboratory of Molecular Radiation Oncology Hunan Province, Changsha, 410008, China
| | - Feiyu Tang
- Department of Oncology, Xiangya Cancer Center, Xiangya Hospital, Central South University, Changsha, 410008, China
- Key Laboratory of Molecular Radiation Oncology Hunan Province, Changsha, 410008, China
| | - Wenfeng Hu
- Department of Oncology, Xiangya Cancer Center, Xiangya Hospital, Central South University, Changsha, 410008, China
- Key Laboratory of Molecular Radiation Oncology Hunan Province, Changsha, 410008, China
| | - Yongwei Zhu
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Wenchao Zhao
- Department of Oncology, Xiangya Cancer Center, Xiangya Hospital, Central South University, Changsha, 410008, China
- Key Laboratory of Molecular Radiation Oncology Hunan Province, Changsha, 410008, China
| | - Wenzhen Hu
- Department of Oncology, Xiangya Cancer Center, Xiangya Hospital, Central South University, Changsha, 410008, China
- Key Laboratory of Molecular Radiation Oncology Hunan Province, Changsha, 410008, China
| | - Zhuohang Chen
- Department of Oncology, Xiangya Cancer Center, Xiangya Hospital, Central South University, Changsha, 410008, China
- Key Laboratory of Molecular Radiation Oncology Hunan Province, Changsha, 410008, China
| | - Yumei Duan
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Anke Geng
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Fengbo Tan
- General Surgery Department, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Yuqiang Li
- General Surgery Department, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Qian Pei
- General Surgery Department, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Haiping Pei
- General Surgery Department, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Zhiyong Mao
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Ningbo Wu
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Lunquan Sun
- Department of Oncology, Xiangya Cancer Center, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Key Laboratory of Molecular Radiation Oncology Hunan Province, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Huan, 410008, China.
- Hunan International Science and Technology Collaboration Base of Precision Medicine for Cancer, Changsha, 410008, China.
- Center for Molecular Imaging of Central South University, Xiangya Hospital, Changsha, 410008, China.
| | - Rong Tan
- Department of Oncology, Xiangya Cancer Center, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Key Laboratory of Molecular Radiation Oncology Hunan Province, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Huan, 410008, China.
- Hunan International Science and Technology Collaboration Base of Precision Medicine for Cancer, Changsha, 410008, China.
- Center for Molecular Imaging of Central South University, Xiangya Hospital, Changsha, 410008, China.
- Hunan key laboratory of aging biology, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
18
|
Vlachavas EI, Voutetakis K, Kosmidou V, Tsikalakis S, Roditis S, Pateas K, Kim R, Pagel K, Wolf S, Warsow G, Dimitrakopoulou-Strauss A, Zografos GN, Pintzas A, Betge J, Papadodima O, Wiemann S. Molecular and functional profiling unravels targetable vulnerabilities in colorectal cancer. Mol Oncol 2025. [PMID: 39876058 DOI: 10.1002/1878-0261.13814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 11/11/2024] [Accepted: 01/20/2025] [Indexed: 01/30/2025] Open
Abstract
Colorectal cancer (CRC) patients with microsatellite-stable (MSS) tumors are mostly treated with chemotherapy. Clinical benefits of targeted therapies depend on mutational states and tumor location. Many tumors carry mutations in KRAS proto-oncogene, GTPase (KRAS) or B-Raf proto-oncogene, serine/threonine kinase (BRAF), rendering them more resistant to therapies. We performed whole-exome sequencing and RNA-Sequencing of 28 tumors of the Athens Comprehensive Cancer Center CRC cohort, and molecularly characterized CRC patients based on their microsatellite instability (MSI) status, single-nucleotide variations (SNVs)/copy number alterations (CNAs), and pathway/transcription factor activities at the individual patient level. Variants were classified using a computational score for integrative cancer variant annotation and prioritization. Complementing this with public multi-omics datasets, we identified activation of transforming growth factor beta (TGFβ) signaling to be more strongly activated in MSS patients, whereas Janus kinase (JAK)-signal transducer and activator of transcription (STAT) and mitogen-activated protein kinase (MAPK) molecular cascades were activated specifically in MSI tumors. We unraveled mechanisms consistently perturbed in the transcriptional and mutational circuits and identified Runt-related transcription factors (RUNX transcription factors) as putative biomarkers in CRC, given their role in the regulation of pathways involved in tumor progression and immune evasion. Assessing the immunogenicity of CRC tumors in the context of RAS/RAF mutations and MSI/MSS status revealed a critical impact that KRAS mutations have on immunogenicity, particularly in the MSS patient subgroup, with implications for diagnosis and treatment.
Collapse
Affiliation(s)
| | | | - Vivian Kosmidou
- Institute of Chemical Biology, National Hellenic Research Foundation, Athens, Greece
| | - Spyridon Tsikalakis
- Division of Molecular Genome Analysis, German Cancer Research Center, Heidelberg, Germany
| | - Spyridon Roditis
- 3rd Surgical Department G.Gennimatas Hospital, Athens, Greece
- Surgical Department, University Hospital of North Midlands, Stoke-on-Trent, UK
| | | | | | | | - Stephan Wolf
- High-Throughput Sequencing Core Facility, German Cancer Research Center, Heidelberg, Germany
| | - Gregor Warsow
- Omics IT and Data Management Core Facility, German Cancer Research Center, Heidelberg, Germany
| | | | | | - Alexander Pintzas
- Institute of Chemical Biology, National Hellenic Research Foundation, Athens, Greece
| | - Johannes Betge
- Junior Clinical Cooperation Unit Translational Gastrointestinal Oncology and Preclinical Models, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Germany
- DKFZ-Hector Cancer Institute at University Medical Center Mannheim, Germany
| | - Olga Papadodima
- Institute of Chemical Biology, National Hellenic Research Foundation, Athens, Greece
| | - Stefan Wiemann
- Division of Molecular Genome Analysis, German Cancer Research Center, Heidelberg, Germany
| |
Collapse
|
19
|
Yang X, Yang D, Qi X, Luo X, Zhang G. Endocrine treatment mechanisms in triple-positive breast cancer: from targeted therapies to advances in precision medicine. Front Oncol 2025; 14:1467033. [PMID: 39845328 PMCID: PMC11753220 DOI: 10.3389/fonc.2024.1467033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 12/09/2024] [Indexed: 01/24/2025] Open
Abstract
Triple-positive breast cancer (TPBC), defined by the co-expression of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2), poses unique therapeutic challenges due to complex signaling interactions and resulting treatment resistance. This review summarizes key findings on the molecular mechanisms and cross-talk among ER, PR, and HER2 pathways, which drive tumor proliferation and resistance to conventional therapies. Current strategies in TPBC treatment, including endocrine and HER2-targeted therapies, are explored alongside emerging approaches such as immunotherapy and CRISPR/Cas9 gene editing. Additionally, we discuss the tumor microenvironment (TME) and its role in treatment resistance, highlighting promising avenues for intervention through combination therapies and predictive biomarkers. By addressing these interdependent pathways and optimizing therapeutic strategies, precision medicine holds significant potential for improving TPBC patient outcomes and advancing individualized cancer care.
Collapse
Affiliation(s)
| | | | | | | | - Guangmei Zhang
- Department of Medical Oncology, Third Division, Jilin City Second People’s Hospital, Jilin, China
| |
Collapse
|
20
|
Zhang Q, Xu X, Jiang D, Wang Y, Wang H, Zhu J, Tang S, Wang R, Zhao S, Li K, Feng J, Xiang H, Yao Z, Xu N, Fang R, Guo W, Liu Y, Hou Y, Ding C. Integrated proteogenomic characterization of ampullary adenocarcinoma. Cell Discov 2025; 11:2. [PMID: 39762212 PMCID: PMC11704194 DOI: 10.1038/s41421-024-00742-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 09/29/2024] [Indexed: 01/11/2025] Open
Abstract
Ampullary adenocarcinoma (AMPAC) is a rare and heterogeneous malignancy. Here we performed a comprehensive proteogenomic analysis of 198 samples from Chinese AMPAC patients and duodenum patients. Genomic data illustrate that 4q loss causes fatty acid accumulation and cell proliferation. Proteomic analysis has revealed three distinct clusters (C-FAM, C-AD, C-CC), among which the most aggressive cluster, C-AD, is associated with the poorest prognosis and is characterized by focal adhesion. Immune clustering identifies three immune clusters and reveals that immune cluster M1 (macrophage infiltration cluster) and M3 (DC cell infiltration cluster), which exhibit a higher immune score compared to cluster M2 (CD4+ T-cell infiltration cluster), are associated with a poor prognosis due to the potential secretion of IL-6 by tumor cells and its consequential influence. This study provides a comprehensive proteogenomic analysis for seeking for better understanding and potential treatment of AMPAC.
Collapse
Affiliation(s)
- Qiao Zhang
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, 200433, China
| | - Xiaomeng Xu
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, 200433, China
| | - Dongxian Jiang
- Department of Pathology, Zhongshan Hospital Fudan University, Shanghai, China
| | - Yunzhi Wang
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, 200433, China
| | - Haixing Wang
- Department of Pathology, Zhongshan Hospital Fudan University, Shanghai, China
| | - Jiajun Zhu
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, 200433, China
| | - Shaoshuai Tang
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, 200433, China
| | - Ronghua Wang
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Key Laboratory of Pediatric Hematology & Oncology Ministry of Health, Department of Hematology & Oncology, Shanghai Jiao Tong University, Shanghai, China
| | - Shuang Zhao
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Key Laboratory of Pediatric Hematology & Oncology Ministry of Health, Department of Hematology & Oncology, Shanghai Jiao Tong University, Shanghai, China
| | - Kai Li
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, 200433, China
| | - Jinwen Feng
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, 200433, China
| | - Hang Xiang
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, 200433, China
| | - Zhenmei Yao
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, 200433, China
| | - Ning Xu
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, 200433, China
| | - Rundong Fang
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, 200433, China
| | - Wenjia Guo
- Departments of Cancer Research Institute, Affiliated Cancer Hospital of Xinjiang Medical University, Xinjiang Key Laboratory of Translational Biomedical Engineering, Urumqi, Xinjiang, China
| | - Yu Liu
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Key Laboratory of Pediatric Hematology & Oncology Ministry of Health, Department of Hematology & Oncology, Shanghai Jiao Tong University, Shanghai, China.
| | - Yingyong Hou
- Department of Pathology, Zhongshan Hospital Fudan University, Shanghai, China.
| | - Chen Ding
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, 200433, China.
- Departments of Cancer Research Institute, Affiliated Cancer Hospital of Xinjiang Medical University, Xinjiang Key Laboratory of Translational Biomedical Engineering, Urumqi, Xinjiang, China.
| |
Collapse
|
21
|
Radhakrishnan SK, Nath D, Russ D, Merodio LB, Lad P, Daisi FK, Acharjee A. Machine learning-based identification of proteomic markers in colorectal cancer using UK Biobank data. Front Oncol 2025; 14:1505675. [PMID: 39839775 PMCID: PMC11746037 DOI: 10.3389/fonc.2024.1505675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 12/02/2024] [Indexed: 01/23/2025] Open
Abstract
Colorectal cancer is one of the leading causes of cancer-related mortality in the world. Incidence and mortality are predicted to rise globally during the next several decades. When detected early, colorectal cancer is treatable with surgery and medications. This leads to the requirement for prognostic and diagnostic biomarker development. Our study integrates machine learning models and protein network analysis to identify protein biomarkers for colorectal cancer. Our methodology leverages an extensive collection of proteome profiles from both healthy and colorectal cancer individuals. To identify a potential biomarker with high predictive ability, we used three machine learning models. To enhance the interpretability of our models, we quantify each protein's contribution to the model's predictions using SHapley Additive exPlanations values. Three classifiers-LASSO, XGBoost, and LightGBM were evaluated for predictive performance along with hyperparameter tuning of each model using grid search, with LASSO achieving the highest AUC of 75% in the UK Biobank dataset and the AUCs for LightGBM and XGBoost are 69.61% and 71.42%, respectively. Using SHapley Additive exPlanations values, TFF3, LCN2, and CEACAM5 were found to be key biomarkers associated with cell adhesion and inflammation. Protein quantitative trait loci analyze studies provided further evidence for the involvement of TFF1, CEACAM5, and SELE in colorectal cancer, with possible connections to the PI3K/Akt and MAPK signaling pathways. By offering insights into colorectal cancer diagnostics and targeted therapeutics, our findings set the stage for further biomarker validation.
Collapse
Affiliation(s)
| | - Dipanwita Nath
- College of Medicine and Health, School of Medical Sciences, Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Dominic Russ
- College of Medicine and Health, School of Medical Sciences, Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
- Institute of Translational Medicine, University Hospitals Birmingham National Health Service (NHS) Foundation Trust, Birmingham, United Kingdom
- Centre for Health Data Research, University of Birmingham, Birmingham, United Kingdom
| | - Laura Bravo Merodio
- College of Medicine and Health, School of Medical Sciences, Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
- Institute of Translational Medicine, University Hospitals Birmingham National Health Service (NHS) Foundation Trust, Birmingham, United Kingdom
- Centre for Health Data Research, University of Birmingham, Birmingham, United Kingdom
| | - Priyani Lad
- College of Medicine and Health, School of Medical Sciences, Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Folakemi Kola Daisi
- College of Medicine and Health, School of Medical Sciences, Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Animesh Acharjee
- College of Medicine and Health, School of Medical Sciences, Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
- Institute of Translational Medicine, University Hospitals Birmingham National Health Service (NHS) Foundation Trust, Birmingham, United Kingdom
- Centre for Health Data Research, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
22
|
Liu Y, Elmas A, Huang KL. Mutation impact on mRNA versus protein expression across human cancers. Gigascience 2025; 14:giae113. [PMID: 39775839 PMCID: PMC11702362 DOI: 10.1093/gigascience/giae113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 09/13/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Cancer mutations are often assumed to alter proteins, thus promoting tumorigenesis. However, how mutations affect protein expression-in addition to gene expression-has rarely been systematically investigated. This is significant as mRNA and protein levels frequently show only moderate correlation, driven by factors such as translation efficiency and protein degradation. Proteogenomic datasets from large tumor cohorts provide an opportunity to systematically analyze the effects of somatic mutations on mRNA and protein abundance and identify mutations with distinct impacts on these molecular levels. RESULTS We conduct a comprehensive analysis of mutation impacts on mRNA- and protein-level expressions of 953 cancer cases with paired genomics and global proteomic profiling across 6 cancer types. Protein-level impacts are validated for 47.2% of the somatic expression quantitative trait loci (seQTLs), including CDH1 and MSH3 truncations, as well as other mutations from likely "long-tail" driver genes. Devising a statistical pipeline for identifying somatic protein-specific QTLs (spsQTLs), we reveal several gene mutations, including NF1 and MAP2K4 truncations and TP53 missenses showing disproportional influence on protein abundance not readily explained by transcriptomics. Cross-validating with data from massively parallel assays of variant effects (MAVE), TP53 missenses associated with high tumor TP53 proteins are more likely to be experimentally confirmed as functional. CONCLUSION This study reveals that somatic mutations can exhibit distinct impacts on mRNA and protein levels, underscoring the necessity of integrating proteogenomic data to comprehensively identify functionally significant cancer mutations. These insights provide a framework for prioritizing mutations for further functional validation and therapeutic targeting.
Collapse
Affiliation(s)
- Yuqi Liu
- Department of Genetics and Genomic Sciences, Department of Artificial Intelligence and Human Health, Center for Transformative Disease Modeling, Tisch Cancer Institute, Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Abdulkadir Elmas
- Department of Genetics and Genomic Sciences, Department of Artificial Intelligence and Human Health, Center for Transformative Disease Modeling, Tisch Cancer Institute, Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kuan-lin Huang
- Department of Genetics and Genomic Sciences, Department of Artificial Intelligence and Human Health, Center for Transformative Disease Modeling, Tisch Cancer Institute, Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
23
|
Shi Z, Lei JT, Elizarraras JM, Zhang B. Mapping the functional network of human cancer through machine learning and pan-cancer proteogenomics. NATURE CANCER 2025; 6:205-222. [PMID: 39663389 DOI: 10.1038/s43018-024-00869-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 10/25/2024] [Indexed: 12/13/2024]
Abstract
Large-scale omics profiling has uncovered a vast array of somatic mutations and cancer-associated proteins, posing substantial challenges for their functional interpretation. Here we present a network-based approach centered on FunMap, a pan-cancer functional network constructed using supervised machine learning on extensive proteomics and RNA sequencing data from 1,194 individuals spanning 11 cancer types. Comprising 10,525 protein-coding genes, FunMap connects functionally associated genes with unprecedented precision, surpassing traditional protein-protein interaction maps. Network analysis identifies functional protein modules, reveals a hierarchical structure linked to cancer hallmarks and clinical phenotypes, provides deeper insights into established cancer drivers and predicts functions for understudied cancer-associated proteins. Additionally, applying graph-neural-network-based deep learning to FunMap uncovers drivers with low mutation frequency. This study establishes FunMap as a powerful and unbiased tool for interpreting somatic mutations and understudied proteins, with broad implications for advancing cancer biology and informing therapeutic strategies.
Collapse
Affiliation(s)
- Zhiao Shi
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Jonathan T Lei
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - John M Elizarraras
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Bing Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA.
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
24
|
Rao R, Gulfishan M, Kim MS, Kashyap MK. Deciphering Cancer Complexity: Integrative Proteogenomics and Proteomics Approaches for Biomarker Discovery. Methods Mol Biol 2025; 2859:211-237. [PMID: 39436604 DOI: 10.1007/978-1-0716-4152-1_12] [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] [Indexed: 10/23/2024]
Abstract
Proteomics has revolutionized the field of cancer biology because the use of a large number of in vivo (SILAC), in vitro (iTRAQ, ICAT, TMT, stable-isotope Dimethyl, and 18O) labeling techniques or label-free methods (spectral counting or peak intensities) coupled with mass spectrometry enables us to profile and identify dysregulated proteins in diseases such as cancer. These proteome and genome studies have led to many challenges, such as the lack of consistency or correlation between copy numbers, RNA, and protein-level data. This review covers solely mass spectrometry-based approaches used for cancer biomarker discovery. It also touches on the emerging role of oncoproteogenomics or proteogenomics in cancer biomarker discovery and how this new area is attracting the integration of genomics and proteomics areas to address some of the important questions to help impinge on the biology and pathophysiology of different malignancies to make these mass spectrometry-based studies more realistic and relevant to clinical settings.
Collapse
Affiliation(s)
- Rashmi Rao
- School of Life and Allied Health Sciences, Glocal University, Saharanpur, UP, India
| | - Mohd Gulfishan
- School of Life and Allied Health Sciences, Glocal University, Saharanpur, UP, India
| | - Min-Sik Kim
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu-42988, Republic of Korea
| | - Manoj Kumar Kashyap
- Amity Stem Cell Institute (ASCI), Amity Medical School (AMS), Amity University Haryana, Panchgaon (Manesar), Gurugram, Haryana, India.
| |
Collapse
|
25
|
Zhao W, Luo Q, Zhan H, Du Z, Deng T, Duan H. DDX18 influences chemotherapy sensitivity in colorectal cancer by regulating genomic stability. Exp Cell Res 2025; 444:114344. [PMID: 39577603 DOI: 10.1016/j.yexcr.2024.114344] [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: 09/29/2024] [Revised: 11/18/2024] [Accepted: 11/18/2024] [Indexed: 11/24/2024]
Abstract
Chromosomal Instability (CIN) encompasses approximately 65 %-70 % of colorectal cancer (CRC) patients, playing a pivotal role in tumor progression. However, controversies persist regarding the molecular characteristics and treatment strategies associated with these patients. Integrative colorectal cancer proteogenomic analysis identified DDX18 in colorectal cancer. We investigated the molecular mechanisms underlying the regulation of colorectal cancer by the R-loop binding protein DDX18 using colon cancer tissues, cell lines and patient-derived organoids. Our findings revealed that DDX18 expression positively correlates with the expression of genomic instability marker R-loops. Moreover, heightened DDX18 expression delays the completion of DNA damage repair, leading to an increase in double-strand DNA breaks, thereby promoting genomic instability. Notably, the upregulation of DDX18 enhances sensitivity to DNA-damaging. This study elucidated DDX18 beyond participating in fundamental physiological functions, may play a crucial role in the regulation of genomic stability, and also provides a powerful resource for further functional exploration of DDX18 in cancer progression and therapeutic application.
Collapse
Affiliation(s)
- Wenchao Zhao
- Department of Oncology, Hunan Provincial People's Hospital, the First Affiliated Hospital of Hunan Normal University, Changsha, 410000, China; Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, China; Hunan Hepatobiliary and Pancreatic Cancer Clinical Medical Research Center, China.
| | - Qingqing Luo
- Department of Oncology, Hunan Provincial People's Hospital, the First Affiliated Hospital of Hunan Normal University, Changsha, 410000, China; Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, China; Hunan Hepatobiliary and Pancreatic Cancer Clinical Medical Research Center, China
| | - Han Zhan
- 921 Hospital of the Chinese People's Liberation Army Joint Logistic Support Force, China
| | - Zhen Du
- Hunan Provincial People's Hospital, the First Affiliated Hospital of Hunan Normal University, Changsha, 410000, China
| | - Tan Deng
- Department of Oncology, Hunan Provincial People's Hospital, the First Affiliated Hospital of Hunan Normal University, Changsha, 410000, China; Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, China; Hunan Hepatobiliary and Pancreatic Cancer Clinical Medical Research Center, China.
| | - Huaxin Duan
- Department of Oncology, Hunan Provincial People's Hospital, the First Affiliated Hospital of Hunan Normal University, Changsha, 410000, China; Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, China; Hunan Hepatobiliary and Pancreatic Cancer Clinical Medical Research Center, China.
| |
Collapse
|
26
|
Zou J, Wang D, Yin G, Lu K, Chang K, Li H. Prognostic significance of p27 in colorectal cancer: a meta-analysis and bioinformatics analysis. Front Oncol 2024; 14:1495476. [PMID: 39845325 PMCID: PMC11751620 DOI: 10.3389/fonc.2024.1495476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 11/29/2024] [Indexed: 01/24/2025] Open
Abstract
Background In the past, numerous investigations have delved into the influence of p27 (p27kip) on the prognosis and clinicopathological characteristics of colorectal cancer (CRC), yielding conclusions that are not universally statistically significant, thus rendering the discourse rather contentious. Methods We collected available articles published before August 2024 and extracted data to analyze the association between the expression of p27 and the prognosis and clinicopathological features of CRC. In addition, we used Gene Expression Profiling Interactive Analysis (GEPIA), University of Alabama at Birmingham's Cancer Data Analysis Portal (UALCAN), and the Human Protein Atlas (HPA) to validate our results. Results Through an extensive examination of four prominent databases, a total of 21 original articles encompassing a cohort of 3,378 patients were identified. The findings indicated that a low expression of p27 could lead to shorter overall survival (OS) [hazard ratio (HR) = 0.44, 95% confidence interval (95%CI) = 0.31-0.61, Z = 4.89, p = 0.000] and disease-free survival (DFS) (HR = 0.40, 95%CI = 0.28-0.59, Z = 4.75, p = 0.000). In addition, a low expression of p27 predisposed tumors to the right colon [odds ratio (OR) = 0.61, 95%CI = 0.46-0.82, Z = 3.32, p = 0.001] and limited tumor differentiation (OR = 0.56, 95%CI = 0.41-0.77, Z = 3.62, p = 0.000), but had no effect on TNM staging (OR = 0.80, 95%CI = 0.52-1.22, Z = 1.05, p = 0.295), lymph node metastasis (OR = 0.90, 95%CI = 0.25-3.28, Z = 0.16, p = 0.876), and tumor size (OR = 0.94, 95%CI = 0.54-1.65, Z = 0.21, p = 0.835). The results from GEPIA and UALCAN showed that p27 had no effect on TNM staging, lymph node metastasis, DFS, and OS; moreover, there was no expression difference between tumor tissues and normal tissues. The findings from the HPA indicated that there was lower expression of p27 in tumor tissues compared with normal tissues. Conclusion Although inconsistent results were reached with the bioinformatics analysis from this meta-analysis, it was confirmed that a low expression of p27 can adversely affect the prognosis of patients with CRC and make a meaningful impact on a part of the clinicopathological features in the meta-analysis with abundant data. In the future, predicting the prognosis of patients with CRC and guiding treatment might emerge as a significant objective.
Collapse
Affiliation(s)
- Jing Zou
- Department of Radiology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong, China
| | - Dong Wang
- Department of Stomach and Intestine, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong, China
| | - Gaoping Yin
- Department of Stomach and Intestine, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong, China
| | - Kexiang Lu
- Department of Stomach and Intestine, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong, China
| | - Kaibin Chang
- Department of Stomach and Intestine, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong, China
| | - He Li
- Department of Stomach and Intestine, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong, China
| |
Collapse
|
27
|
Ramirez-Falcon M, Suarez-Pajes E, Flores C. Defining the Differential Corticosteroid Response Basis from Multiple Omics Approaches. Int J Mol Sci 2024; 25:13611. [PMID: 39769372 PMCID: PMC11679800 DOI: 10.3390/ijms252413611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
Since their discovery, corticosteroids have been widely used in the treatment of several diseases, including asthma, acute lymphoblastic leukemia, chronic obstructive pulmonary disease, and many other conditions. However, it has been noted that some patients develop undesired side effects or even fail to respond to treatment. The reasons behind this have not yet been fully elucidated. This poses a significant challenge to effective treatment that needs to be addressed urgently. Recent genomic, transcriptomic, and other omics-based approximations have begun to shed light into the genetic factors influencing interindividual variability in corticosteroid efficacy and its side effects. Here, we comprehensively revise the recent literature on corticosteroid response in various critical and chronic diseases, with a focus on omics approaches, and highlight existing knowledge gaps where further investigation is urgently needed.
Collapse
Affiliation(s)
- Melody Ramirez-Falcon
- Research Unit, Hospital Universitario Ntra. Sra. de Candelaria, Instituto de Investigación Sanitaria de Canarias, 38010 Santa Cruz de Tenerife, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Eva Suarez-Pajes
- Research Unit, Hospital Universitario Ntra. Sra. de Candelaria, Instituto de Investigación Sanitaria de Canarias, 38010 Santa Cruz de Tenerife, Spain
| | - Carlos Flores
- Research Unit, Hospital Universitario Ntra. Sra. de Candelaria, Instituto de Investigación Sanitaria de Canarias, 38010 Santa Cruz de Tenerife, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Genomics Division, Instituto Tecnológico y de Energías Renovables, Granadilla de Abona, 38600 Santa Cruz de Tenerife, Spain
- Facultad de Ciencias de la Salud, Universidad Fernando Pessoa Canarias, 35450 Las Palmas de Gran Canaria, Spain
| |
Collapse
|
28
|
Ren Y, Yue Y, Li X, Weng S, Xu H, Liu L, Cheng Q, Luo P, Zhang T, Liu Z, Han X. Proteogenomics offers a novel avenue in neoantigen identification for cancer immunotherapy. Int Immunopharmacol 2024; 142:113147. [PMID: 39270345 DOI: 10.1016/j.intimp.2024.113147] [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: 05/11/2024] [Revised: 08/11/2024] [Accepted: 09/08/2024] [Indexed: 09/15/2024]
Abstract
Cancer neoantigens are tumor-specific non-synonymous mutant peptides that activate the immune system to produce an anti-tumor response. Personalized cancer vaccines based on neoantigens are currently one of the most promising therapeutic approaches for cancer treatment. By utilizing the unique mutations within each patient's tumor, these vaccines aim to elicit a strong and specific immune response against cancer cells. However, the identification of neoantigens remains challenging due to the low accuracy of current prediction tools and the high false-positive rate of candidate neoantigens. Since the concept of "proteogenomics" emerged in 2004, it has evolved rapidly with the increased sequencing depth of next-generation sequencing technologies and the maturation of mass spectrometry-based proteomics technologies to become a more comprehensive approach to neoantigen identification, allowing the discovery of high-confidence candidate neoantigens. In this review, we summarize the reason why cancer neoantigens have become attractive targets for immunotherapy, the mechanism of cancer vaccines and the advances in cancer immunotherapy. Considerations relevant to the application emerging of proteogenomics technologies for neoantigen identification and challenges in this field are described.
Collapse
Affiliation(s)
- Yuqing Ren
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China; Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Yi Yue
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Xinyang Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Siyuan Weng
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Hui Xu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Long Liu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Tengfei Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.
| | - Zaoqu Liu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China; Interventional Institute of Zhengzhou University, Zhengzhou, Henan 450052, China; Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan 450052, China.
| |
Collapse
|
29
|
Zhou L, Zhu Z, Gao H, Wang C, Khan MA, Ullah M, Khan SU. Multi‐omics graph convolutional networks for digestive system tumour classification and early‐late stage diagnosis. CAAI TRANSACTIONS ON INTELLIGENCE TECHNOLOGY 2024; 9:1572-1586. [DOI: 10.1049/cit2.12395] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 06/05/2024] [Indexed: 01/12/2025] Open
Abstract
AbstractThe prevalence of digestive system tumours (DST) poses a significant challenge in the global crusade against cancer. These neoplasms constitute 20% of all documented cancer diagnoses and contribute to 22.5% of cancer‐related fatalities. The accurate diagnosis of DST is paramount for vigilant patient monitoring and the judicious selection of optimal treatments. Addressing this challenge, the authors introduce a novel methodology, denominated as the Multi‐omics Graph Transformer Convolutional Network (MGTCN). This innovative approach aims to discern various DST tumour types and proficiently discern between early‐late stage tumours, ensuring a high degree of accuracy. The MGTCN model incorporates the Graph Transformer Layer framework to meticulously transform the multi‐omics adjacency matrix, thereby illuminating potential associations among diverse samples. A rigorous experimental evaluation was undertaken on the DST dataset from The Cancer Genome Atlas to scrutinise the efficacy of the MGTCN model. The outcomes unequivocally underscore the efficiency and precision of MGTCN in diagnosing diverse DST tumour types and successfully discriminating between early‐late stage DST cases. The source code for this groundbreaking study is readily accessible for download at https://github.com/bigone1/MGTCN.
Collapse
Affiliation(s)
- Lin Zhou
- School of Information Science and Technology University of Science and Technology of China Hefei Anhui China
- Anhui Engineering Research Center on Information Fusion and Control of Intelligent Robot Wuhu Anhui China
| | - Zhengzhi Zhu
- Department of Breast Center West District of The Affiliated Hospital of University of Science and Technology of China Division of Life Sciences and Medicine University of Science and Technology of China Hefei Anhui China
| | - Hongbo Gao
- School of Information Science and Technology University of Science and Technology of China Hefei Anhui China
- Institute of Advanced Technology University of Science and Technology of China Hefei Anhui China
- School of Electrical and Electronic Engineering Nanyang Technological University Singapore Singapore
| | - Chunyu Wang
- School of Biological and Environmental Engineering Chaohu University Chaohu Regional Collaborative Technology Service Center for Rural Revitalization Hefei China
| | - Muhammad Attique Khan
- Department of Artificial Intelligence College of Computer Engineering and Science Prince Mohammad Bin Fahd University Al‐Khobar Saudi Arabia
| | - Mati Ullah
- School of Information Science and Technology University of Science and Technology of China Hefei Anhui China
- School of Automation Northwestern Polytechnical University Xi'an Shaanxi China
| | - Siffat Ullah Khan
- School of Information Science and Technology University of Science and Technology of China Hefei Anhui China
- Institute of Engineering and Computing Science University of Science and Technology of Bannu KPK Bannu Pakistan
| |
Collapse
|
30
|
Xu G, Yu J, Lyu J, Zhan M, Xu J, Huang M, Zhao R, Li Y, Zhu J, Feng J, Tan S, Ran P, Su Z, Liu X, Zhao J, Zhang H, Xu C, Chang J, Hou Y, Ding C. Proteogenomic Landscape of Breast Ductal Carcinoma Reveals Tumor Progression Characteristics and Therapeutic Targets. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401041. [PMID: 39418072 PMCID: PMC11633542 DOI: 10.1002/advs.202401041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 09/04/2024] [Indexed: 10/19/2024]
Abstract
Multi-omics studies of breast ductal carcinoma (BRDC) have advanced the understanding of the disease's biology and accelerated targeted therapies. However, the temporal order of a series of biological events in the progression of BRDC is still poorly understood. A comprehensive proteogenomic analysis of 224 samples from 168 patients with malignant and benign breast diseases is carried out. Proteogenomic analysis reveals the characteristics of linear multi-step progression of BRDC, such as tumor protein P53 (TP53) mutation-associated estrogen receptor 1 (ESR1) overexpression is involved in the transition from ductal hyperplasia (DH) to ductal carcinoma in situ (DCIS). 6q21 amplification-associated nuclear receptor subfamily 3 group C member 1 (NR3C1) overexpression helps DCIS_Pure (pure DCIS, no histologic evidence of invasion) cells avoid immune destruction. The T-cell lymphoma invasion and metastasis 1, androgen receptor, and aldo-keto reductase family 1 member C1 (TIAM1-AR-AKR1C1) axis promotes cell invasion and migration in DCIS_adjIDC (DCIS regions of invasive cancers). In addition, AKR1C1 is identified as a potential therapeutic target and demonstrated the inhibitory effect of aspirin and dydrogesterone as its inhibitors on tumor cells. The integrative multi-omics analysis helps to understand the progression of BRDC and provides an opportunity to treat BRDC in different stages.
Collapse
Affiliation(s)
- Ganfei Xu
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Juan Yu
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Jiacheng Lyu
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Mengna Zhan
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Jie Xu
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Minjing Huang
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Rui Zhao
- Institute for Developmental and Regenerative Cardiovascular MedicineMOE‐Shanghai Key Laboratory of Children's Environmental HealthXinhua HospitalShanghai Jiao Tong University School of MedicineShanghai200092China
| | - Yan Li
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Jiajun Zhu
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Jinwen Feng
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Subei Tan
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Peng Ran
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Zhenghua Su
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Xinhua Liu
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Jianyuan Zhao
- Institute for Developmental and Regenerative Cardiovascular MedicineMOE‐Shanghai Key Laboratory of Children's Environmental HealthXinhua HospitalShanghai Jiao Tong University School of MedicineShanghai200092China
| | - Hongwei Zhang
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Chen Xu
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Jun Chang
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Yingyong Hou
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Chen Ding
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
- Departments of Cancer Research InstituteAffiliated Cancer Hospital of Xinjiang Medical UniversityXinjiang Key Laboratory of Translational Biomedical EngineeringUrumqi830000P. R. China
| |
Collapse
|
31
|
Wang J, Wang Y, Li S, Fang X, Zhang C, Wang Z, Zheng Y, Deng H, Xu S, Mi Y. Exploring acetylation-related gene markers in polycystic ovary syndrome: insights into pathogenesis and diagnostic potential using machine learning. Gynecol Endocrinol 2024; 40:2427202. [PMID: 39585802 DOI: 10.1080/09513590.2024.2427202] [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: 07/12/2024] [Revised: 10/20/2024] [Accepted: 11/04/2024] [Indexed: 11/27/2024] Open
Abstract
OBJECTIVE Polycystic ovary syndrome (PCOS) is a prevalent cause of menstrual irregularities and infertility in women, impacting quality of life. Despite advancements, current understanding of PCOS pathogenesis and treatment remains limited. This study uses machine learning-based data mining to identify acetylation-related genetic markers associated with PCOS, aiming to enhance diagnostic precision and therapeutic efficacy. METHODS Advanced machine learning techniques were used to improve the precision of key gene identification and reveal their biological mechanisms. Validation on an independent dataset (GSE48301) confirmed their diagnostic value, assessed through ROC curves and nomograms for PCOS risk prediction. Molecular mechanisms of acetylation-related gene regulation in PCOS were further examined through clustering, immune-environmental, and gene network analyses. RESULTS Our analysis identified 15 key acetylation-regulated genes differentially expressed in PCOS, including SGF29, NOL6, KLF15, and INO80D, which are relevant to PCOS pathogenesis. ROC curve analyses on training and validation datasets confirmed the model's high diagnostic accuracy. Additionally, these genes were associated with immune cell infiltration, offering insights into the inflammatory aspect of PCOS. CONCLUSION The identified acetylation gene markers offer novel insights into the molecular mechanisms underlying PCOS and hold promise for enhancing the development of precise diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Jiqing Wang
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuqing Wang
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shanshan Li
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaoqin Fang
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chaoyue Zhang
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zuqing Wang
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yi Zheng
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hanzhi Deng
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shifen Xu
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yiqun Mi
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
32
|
Bhargava M, Crouser ED. Application of laboratory models for sarcoidosis research. J Autoimmun 2024; 149:103184. [PMID: 38443221 DOI: 10.1016/j.jaut.2024.103184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 02/12/2024] [Accepted: 02/15/2024] [Indexed: 03/07/2024]
Abstract
This manuscript will review the implications and applications of sarcoidosis models towards advancing our understanding of sarcoidosis disease mechanisms, identification of biomarkers, and preclinical testing of novel therapies. Emerging disease models and innovative research tools will also be considered.
Collapse
Affiliation(s)
- Maneesh Bhargava
- University of Minnesota Medical Center, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, 420 Delaware Street SE, MMC 276. Minneapolis, MN 55455, USA
| | - Elliott D Crouser
- Ohio State University Wexner Medicine Center, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, 241 W. 11th Street, Suite 5000, Columbus, OH 43201, USA.
| |
Collapse
|
33
|
Ma W, Tang W, Kwok JS, Tong AH, Lo CW, Chu AT, Chung BH. A review on trends in development and translation of omics signatures in cancer. Comput Struct Biotechnol J 2024; 23:954-971. [PMID: 38385061 PMCID: PMC10879706 DOI: 10.1016/j.csbj.2024.01.024] [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: 10/27/2023] [Revised: 01/31/2024] [Accepted: 01/31/2024] [Indexed: 02/23/2024] Open
Abstract
The field of cancer genomics and transcriptomics has evolved from targeted profiling to swift sequencing of individual tumor genome and transcriptome. The steady growth in genome, epigenome, and transcriptome datasets on a genome-wide scale has significantly increased our capability in capturing signatures that represent both the intrinsic and extrinsic biological features of tumors. These biological differences can help in precise molecular subtyping of cancer, predicting tumor progression, metastatic potential, and resistance to therapeutic agents. In this review, we summarized the current development of genomic, methylomic, transcriptomic, proteomic and metabolic signatures in the field of cancer research and highlighted their potentials in clinical applications to improve diagnosis, prognosis, and treatment decision in cancer patients.
Collapse
Affiliation(s)
- Wei Ma
- Hong Kong Genome Institute, Hong Kong, China
| | - Wenshu Tang
- Hong Kong Genome Institute, Hong Kong, China
| | | | | | | | | | - Brian H.Y. Chung
- Hong Kong Genome Institute, Hong Kong, China
- Department of Pediatrics and Adolescent Medicine, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Hong Kong Genome Project
- Hong Kong Genome Institute, Hong Kong, China
- Department of Pediatrics and Adolescent Medicine, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
34
|
Zhu R, Gao Z, Wu S, Ma S, Zhu Y, Zhang S, Zhang Y, Zeng H, Ma C, Zhao J, Ye J, Zhang W. Multi-omics and network pharmacology approaches reveal Gui-Ling-Ji alleviates oligoasthenoteratozoospermia by regulating arachidonic acid pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156184. [PMID: 39488872 DOI: 10.1016/j.phymed.2024.156184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 10/15/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND Gui-Ling-Ji (GLJ) described in the ancient medical book 'Yunji Qijian' is a traditional Chinese medicine formula used to improve male fertility. It is now available for the treatment of oligoasthenoteratozoospermia (OAT). However, the active ingredients and mechanism of GLJ are not clear. PURPOSE The aim of this study was to clarify the active ingredients and mechanism of GLJ in OAT. METHODS Firstly, the cyclophosphamide-induced OAT rat model was established to evaluate the efficacy of GLJ. Secondly, serum/urine-based metabolomics and lipidomics and tissue-based transcriptomics were performed to discover the differential metabolites and genes in rats. Furthermore, network pharmacology was constructed to explore the associated mechanisms based on the results of multi-omics analysis. Finally, cellular experiment on testicular mesenchymal stromal cells (TM3) was used to validate the active ingredients and the key metabolic pathway. RESULTS Rats were administered GLJ by gavage every day for 3 weeks. Testicular damage and weight loss caused by cyclophosphamide were restored in rats, the sperm count and motility were improved, and levels of luteinizing hormone (LH), follicle-stimulating hormone (FSH) and testosterone (T) secretion were also elevated. Compared to the metabolites of OAT rats, 51 and 37 differential metabolites regulated by GLJ were identified from serum and urine respectively, 54 lipid differential metabolites regulated by GLJ were identified by lipidomics. At the same time, 23 of the 258 differential genes were found to be regulated by OAT rats and then reverse-regulated by GLJ. Network pharmacology has identified 13 pathways (Steroid hormone biosynthesis, Taurine and hypotaurine metabolism, Primary bile acid biosynthesis, Linoleic acid metabolism, Retinol metabolism, Glycerophospholipid metabolism, Ether lipid metabolism, Sphingolipid metabolism, Arachidonic acid metabolism, Glutathione metabolism, Arginine biosynthesis, Arginine and proline metabolism, D-Arginine and D-ornithine metabolism), four metabolites (arachidonic acid, oestrone sulphate, phosphatidylglycerol choline and sphingomyelin) and 15 targets (ABCB11, ALDH18A1, CCL3, CD244, CIITA, CYP2C8, DLL1, ITGA4, ESR1, AR, ABCB1, ABCC1, ALB, PLA2G1B and NOS2). GLJ, psoralen, isopsoralen, liquiritin, isoliquiritin, liquiritigenin, and ginsenoside Ro could significantly promote T secretion from TM3 cells. Additionally, arachidonic acid metabolism particularly the cyclooxygenase pathway, is closely related to the promotion of testosterone secretion by GLJ in TM3. CONCLUSION GLJ has a therapeutic efficacy in cyclophosphamide-induced OAT rats, which can modulate the disorders of lipid metabolism and amino acid metabolism. Arachidonic acid metabolism may be a key pathway, and six prototype compounds are potential key active ingredients for GLJ.
Collapse
Affiliation(s)
- Renwen Zhu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China..
| | - Ziqing Gao
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Shiyu Wu
- School of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
| | - Siyi Ma
- School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Yiqing Zhu
- School of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
| | - Shiyu Zhang
- School of Pharmacy, Henan University, Kaifeng, 475004, China
| | - Yuhao Zhang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Huawu Zeng
- School of Pharmacy, Naval Medical University, Shanghai 200433, China
| | - Chi Ma
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Jing Zhao
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China..
| | - Ji Ye
- School of Pharmacy, Naval Medical University, Shanghai 200433, China..
| | - Weidong Zhang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.; School of Pharmacy, Naval Medical University, Shanghai 200433, China.; School of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China.; School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China..
| |
Collapse
|
35
|
Vigdorovits A, Olteanu GE, Pascalau AV, Pirlog R, Berindan-Neagoe I, Pop OL. Novel Immunohistochemical Profiling of Small-Cell Lung Cancer: Correlations Between Tumor Subtypes and Immune Microenvironment. Diagnostics (Basel) 2024; 14:2660. [PMID: 39682568 DOI: 10.3390/diagnostics14232660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/20/2024] [Accepted: 11/21/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND/OBJECTIVES Small-cell lung cancer (SCLC) is a highly aggressive malignancy with an emerging molecular classification based on the expression of the transcription factors ASCL1, NEUROD1, and POU2F3. This study aimed to explore the relationship between these novel subtypes and the tumor immune microenvironment (TIME), particularly CD8+ and CD4+ tumor-infiltrating lymphocytes (TILs). METHODS In 51 cases of patients with SCLC, immunohistochemical (IHC) stains for ASCL1, NEUROD1, POU2F3, CD56, Ki67, CD8, and CD4 were performed. H-scores for the novel transcription factors were calculated to determine tumor subtype. CD8+ and CD4+ TIL counts were averaged across 10 high-power fields. The Kruskal-Wallis test and subsequent post hoc Dunn tests were used to determine the differences in transcription factor expression and TILs across subtypes. RESULTS In our cohort, 68.62% of our cases were SCLC-A, 9.80% were SCLC-N, 7.84% were SCLC-P, and 13.72% were SCLC-I. Significant differences were observed in the expression of ASCL1, NEUROD1, and POU2F3 across subtypes. CD8+ TILs were more abundant in SCLC-P and SCLC-I. CD8+ TILs were negatively correlated with ASCL1 expression (p < 0.05) and positively correlated with POU2F3 expression (p < 0.005). CONCLUSIONS This study highlights the need to integrate the novel SCLC classification with data regarding the TIME to better inform patient prognosis and treatment.
Collapse
Affiliation(s)
- Alon Vigdorovits
- Department of Morphological Disciplines, University of Oradea, 410087 Oradea, Romania
| | - Gheorghe-Emilian Olteanu
- British Columbia Cancer, Department of Pathology, Vancouver, BC V5Z 4E6, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z7, Canada
| | | | - Radu Pirlog
- Département de Pathologie, Hôpitaux Universitaires Henri Mondor, AP-HP, 94010 Créteil, France
- INSERM U955, Université Paris Est Créteil, 94010 Créteil, France
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
| | - Ioana Berindan-Neagoe
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
- Doctoral School, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
| | - Ovidiu-Laurean Pop
- Department of Morphological Disciplines, University of Oradea, 410087 Oradea, Romania
| |
Collapse
|
36
|
Su Z, Fang M, Smolnikov A, Vafaee F, Dinger ME, Oates EC. Post-transcriptional regulation supports the homeostatic expression of mature RNA. Brief Bioinform 2024; 26:bbaf027. [PMID: 39913622 PMCID: PMC11801271 DOI: 10.1093/bib/bbaf027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 10/31/2024] [Accepted: 02/05/2025] [Indexed: 02/09/2025] Open
Abstract
Gene expression regulation is a sophisticated, multi-stage process, and its robustness is critical to normal cell function and the survival of an organism. Previous studies indicate that differential gene expression at the RNA level is typically attenuated at the protein level through translational regulation. However, how post-transcriptional regulation (PTR) influences expression change during the RNA maturation process remains unclear. In this study, we investigated this by quantifying the magnitude of expression change in precursor RNA and mature RNA across a vast range of different biological conditions. We analyzed bulk tissue RNA sequencing data from 4689 samples, including healthy and diseased tissues from human, chimpanzee, rhesus macaque, and murine sources. We demonstrated that PTR tends to support homeostatic expression of mature RNA by amplifying normal tissue-specific expression of precursor RNA, while reducing expression change of precursor RNA in disease contexts. Our study provides insight into the general influence of PTR on gene expression homeostasis. Our analysis also suggests that intronic reads in RNA-seq studies may contain under-utilized information about disease associations. Additionally, our findings may assist in identifying new disease biomarkers and more effective ways of altering gene expression as a therapeutic strategy.
Collapse
Affiliation(s)
- Zheng Su
- School of Biotechnology and Biomolecular Sciences, Faculty of Science, The University of New South Wales, Biological Sciences North Building (D26), Upper Kensington Campus, Sydney, New South Wales 2052, Australia
| | - Mingyan Fang
- BGI Research, Building 1, Future Science and Technology Innovation Mansion, No. 59, Science and Technology 3rd Road, East Lake High-tech Development Zone, Wuhan City, Hubei Province, 430074, China
- BGI Australia, L6, CBCRC, QIMR Medical Research Institute, 300 Herston Road, Herston, QLD 4006, Australia
| | - Andrei Smolnikov
- School of Biotechnology and Biomolecular Sciences, Faculty of Science, The University of New South Wales, Biological Sciences North Building (D26), Upper Kensington Campus, Sydney, New South Wales 2052, Australia
| | - Fatemeh Vafaee
- School of Biotechnology and Biomolecular Sciences, Faculty of Science, The University of New South Wales, Biological Sciences North Building (D26), Upper Kensington Campus, Sydney, New South Wales 2052, Australia
| | - Marcel E Dinger
- School of Biotechnology and Biomolecular Sciences, Faculty of Science, The University of New South Wales, Biological Sciences North Building (D26), Upper Kensington Campus, Sydney, New South Wales 2052, Australia
- School of Life and Environmental Sciences, Faculty of Science, University of Sydney, F22 Life, Earth and Environmental Sciences (LEES) Building, Camperdown NSW 2050, Australia
| | - Emily C Oates
- School of Biotechnology and Biomolecular Sciences, Faculty of Science, The University of New South Wales, Biological Sciences North Building (D26), Upper Kensington Campus, Sydney, New South Wales 2052, Australia
- Department of Neurology, Sydney Children’s Hospital, High St, Randwick NSW 2031, Australia
| |
Collapse
|
37
|
Zhang J, Sun R, Lyu Y, Liu C, Liu Y, Feng Y, Fu M, Wong PJC, Du Z, Qiu T, Zhang Y, Zhuang D, Qin Z, Yao Y, Zhu W, Guo T, Hua W, Yang H, Mao Y. Proteomic profiling of gliomas unveils immune and metabolism-driven subtypes with implications for anti-nucleotide metabolism therapy. Nat Commun 2024; 15:10005. [PMID: 39562821 PMCID: PMC11577044 DOI: 10.1038/s41467-024-54352-5] [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: 12/26/2023] [Accepted: 11/08/2024] [Indexed: 11/21/2024] Open
Abstract
Gliomas exhibit high heterogeneity and poor prognosis. Despite substantial progress has been made at the genomic and transcriptomic levels, comprehensive proteomic characterization and its implications remain largely unexplored. In this study, we perform proteomic profiling of gliomas using 343 formalin-fixed and paraffin-embedded tumor samples and 53 normal-appearing brain samples from 188 patients, integrating these data with genomic panel information and clinical outcomes. The proteomic analysis uncovers two distinct subgroups: Subgroup 1, the metabolic neural subgroup, enriched in metabolic enzymes and neurotransmitter receptor proteins, and Subgroup 2, the immune subgroup, marked by upregulation of immune and inflammatory proteins. These proteomic subgroups show significant differences in prognosis, tumorigenesis, microenvironment dysregulation, and potential therapeutics, highlighting the critical roles of metabolic and immune processes in glioma biology and patient outcomes. Through a detailed investigation of metabolic pathways guided by our proteomic findings, dihydropyrimidine dehydrogenase (DPYD) and thymidine phosphorylase (TYMP) emerge as potential prognostic biomarkers linked to the reprogramming of nucleotide metabolism. Functional validation in patient-derived glioma stem cells and animal models highlights nucleotide metabolism as a promising therapy target for gliomas. This integrated multi-omics analysis introduces a proteomic classification for gliomas and identifies DPYD and TYMP as key metabolic biomarkers, offering insights into glioma pathogenesis and potential treatment strategies.
Collapse
Affiliation(s)
- Jinsen Zhang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
| | - Rui Sun
- Affiliated Hangzhou First People's Hospital, State Key Laboratory of Medical Proteomics, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
- Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Research Center for Industries of the Future, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Yingying Lyu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
| | - Chaxian Liu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
| | - Ying Liu
- Department of Pathology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yuan Feng
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
| | - Minjie Fu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
| | - Peter Jih Cheng Wong
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
| | - Zunguo Du
- Department of Pathology, Huashan Hospital, Fudan University, Shanghai, China
| | - Tianming Qiu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
| | - Yi Zhang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
| | - Dongxiao Zhuang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
| | - Zhiyong Qin
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
| | - Yu Yao
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
| | - Wei Zhu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
| | - Tiannan Guo
- Affiliated Hangzhou First People's Hospital, State Key Laboratory of Medical Proteomics, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
- Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Research Center for Industries of the Future, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Wei Hua
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China.
- National Center for Neurological Disorders, Shanghai, China.
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China.
- Neurosurgical Institute of Fudan University, Shanghai, China.
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China.
| | - Hui Yang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China.
- National Center for Neurological Disorders, Shanghai, China.
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China.
- Neurosurgical Institute of Fudan University, Shanghai, China.
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China.
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institute for Translational Brain Research, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Ying Mao
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China.
- National Center for Neurological Disorders, Shanghai, China.
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China.
- Neurosurgical Institute of Fudan University, Shanghai, China.
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China.
| |
Collapse
|
38
|
Liu J, Chang Y, Ou Q, Chen L, Yan H, Guo D, Wang C, Zhang S. Advances in research on the relationship between mitochondrial function and colorectal cancer: a bibliometric study from 2013 to 2023. Front Immunol 2024; 15:1480596. [PMID: 39611141 PMCID: PMC11602704 DOI: 10.3389/fimmu.2024.1480596] [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: 08/14/2024] [Accepted: 10/28/2024] [Indexed: 11/30/2024] Open
Abstract
The study provides a thorough examination of literature from 2013 to 2023, delving into the intricate relationship between mitochondrial function and colorectal cancer (CRC). It offers a concise overview of the current landscape and emerging trends in this rapidly evolving research area. The findings indicate a consistent rise in annual publications, reflecting growing interest and significant potential in the field. China emerges as the leading contributor, followed by the United States and India. However, despite China's dominance in output, its average citation rate is lower than that of the US, which leads in citations per publication, highlighting a noticeable disparity. In the realm of research institutions, Shanghai Jiao Tong University and China Medical University are identified as major contributors, yet the potential for inter-institutional collaboration remains largely untapped, suggesting avenues for future synergy. Internationally, China-US collaborations are particularly robust, fostering cross-border knowledge exchange. Hyun Jin Won and Li Wei are recognized as prolific authors, while Ahmedin Jemal is an influential co-cited scholar, noted for his seminal contributions. Keyword analysis reveals research focus areas, such as the complex CRC tumor microenvironment, molecular mechanisms of oxidative stress, and key multidrug resistance pathways. It also highlights the promising potential of mitochondria-targeted therapies and nanomolecular technologies in clinical practice, signaling their growing significance in addressing complex health challenges. The study underscores the imperative to validate complex mitochondrial mechanisms and signaling pathways in CRC, with a particular emphasis on translating these insights into drug targets for clinical trials. Advancing this research is expected to refine and enhance CRC treatment strategies. Additionally, it highlights the urgency of validating mitochondrial complexities in CRC, advocating for collaborative efforts to link these mechanisms with tailored therapeutic interventions for clinical testing. This integrated approach promises significant advancements in developing effective, targeted CRC treatments, ultimately improving patient outcomes.
Collapse
Affiliation(s)
- Jinhui Liu
- College of Integrated Traditional Chinese & Western Medicine, Hunan University of Traditional Chinese Medicine, Changsha, Hunan, China
| | - Yonglong Chang
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qinling Ou
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Addiction Medicine, Hunan Institute of Mental Health, Brain Hospital of Hunan Province (The Second People’s Hospital of Hunan Province), Changsha, Hunan, China
| | - Linzi Chen
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Haixia Yan
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Duanyang Guo
- College of Integrated Traditional Chinese & Western Medicine, Hunan University of Traditional Chinese Medicine, Changsha, Hunan, China
| | - Chongjie Wang
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Sifang Zhang
- College of Integrated Traditional Chinese & Western Medicine, Hunan University of Traditional Chinese Medicine, Changsha, Hunan, China
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
39
|
Hamilton AK, Radaoui AB, Tsang M, Martinez D, Conkrite KL, Patel K, Sidoli S, Delaidelli A, Modi A, Rokita JL, Lane MV, Hartnett N, Lopez RD, Zhang B, Zhong C, Ennis B, Miller DP, Brown MA, Rathi KS, Raman P, Pogoriler J, Bhatti T, Pawel B, Glisovic-Aplenc T, Teicher B, Erickson SW, Earley EJ, Bosse KR, Sorensen PH, Krytska K, Mosse YP, Havenith KE, Zammarchi F, van Berkel PH, Smith MA, Garcia BA, Maris JM, Diskin SJ. A proteogenomic surfaceome study identifies DLK1 as an immunotherapeutic target in neuroblastoma. Cancer Cell 2024; 42:1970-1982.e7. [PMID: 39454577 PMCID: PMC11560519 DOI: 10.1016/j.ccell.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 08/14/2024] [Accepted: 10/03/2024] [Indexed: 10/28/2024]
Abstract
Cancer immunotherapies produce remarkable results in B cell malignancies; however, optimal cell surface targets for many solid cancers remain elusive. Here, we present an integrative proteomic, transcriptomic, and epigenomic analysis of tumor and normal tissues to identify biologically relevant cell surface immunotherapeutic targets for neuroblastoma, an often-fatal childhood cancer. Proteogenomic analyses reveal sixty high-confidence candidate immunotherapeutic targets, and we prioritize delta-like canonical notch ligand 1 (DLK1) for further study. High expression of DLK1 directly correlates with a super-enhancer. Immunofluorescence, flow cytometry, and immunohistochemistry show robust cell surface expression of DLK1. Short hairpin RNA mediated silencing of DLK1 in neuroblastoma cells results in increased cellular differentiation. ADCT-701, a DLK1-targeting antibody-drug conjugate (ADC), shows potent and specific cytotoxicity in DLK1-expressing neuroblastoma xenograft models. Since high DLK1 expression is found in several adult and pediatric cancers, our study demonstrates the utility of a proteogenomic approach and credentials DLK1 as an immunotherapeutic target.
Collapse
MESH Headings
- Neuroblastoma/drug therapy
- Neuroblastoma/immunology
- Neuroblastoma/mortality
- Neuroblastoma/pathology
- Immunotherapy/methods
- Proteogenomics
- Calcium-Binding Proteins/analysis
- Calcium-Binding Proteins/antagonists & inhibitors
- Calcium-Binding Proteins/immunology
- Calcium-Binding Proteins/metabolism
- Membrane Proteins/analysis
- Membrane Proteins/antagonists & inhibitors
- Membrane Proteins/immunology
- Membrane Proteins/metabolism
- Cell Line, Tumor
- Xenograft Model Antitumor Assays
- Mice, SCID
- Humans
- Female
- Animals
- Mice
- Kaplan-Meier Estimate
- Biomarkers, Tumor/analysis
- Biomarkers, Tumor/antagonists & inhibitors
- Biomarkers, Tumor/immunology
- Biomarkers, Tumor/metabolism
- Gene Expression Regulation, Neoplastic/drug effects
- Gene Expression Regulation, Neoplastic/immunology
- Immunoconjugates/pharmacology
- Immunoconjugates/therapeutic use
- Antineoplastic Agents, Immunological/pharmacology
- Antineoplastic Agents, Immunological/therapeutic use
- RNA-Seq
- Child
- Molecular Targeted Therapy/methods
Collapse
Affiliation(s)
- Amber K Hamilton
- Center for Childhood Cancer Research and Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Alexander B Radaoui
- Center for Childhood Cancer Research and Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Matthew Tsang
- Center for Childhood Cancer Research and Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Daniel Martinez
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Karina L Conkrite
- Center for Childhood Cancer Research and Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Khushbu Patel
- Center for Childhood Cancer Research and Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Simone Sidoli
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Alberto Delaidelli
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada; Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, BC, Canada
| | - Apexa Modi
- Center for Childhood Cancer Research and Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Jo Lynne Rokita
- Center for Data-Driven Discovery in Biomedicine and Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Maria V Lane
- Center for Childhood Cancer Research and Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Nicholas Hartnett
- Center for Childhood Cancer Research and Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Raphael D Lopez
- Center for Childhood Cancer Research and Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Bo Zhang
- Center for Data-Driven Discovery in Biomedicine and Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Chuwei Zhong
- Center for Data-Driven Discovery in Biomedicine and Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Brian Ennis
- Center for Data-Driven Discovery in Biomedicine and Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Daniel P Miller
- Center for Data-Driven Discovery in Biomedicine and Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Miguel A Brown
- Center for Data-Driven Discovery in Biomedicine and Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Komal S Rathi
- Center for Childhood Cancer Research and Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Center for Data-Driven Discovery in Biomedicine and Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Pichai Raman
- Center for Childhood Cancer Research and Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Center for Data-Driven Discovery in Biomedicine and Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Jennifer Pogoriler
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Tricia Bhatti
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Bruce Pawel
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Tina Glisovic-Aplenc
- Center for Childhood Cancer Research and Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | | | | | - Eric J Earley
- RTI International, Research Triangle Park, Durham, NC 27709, USA
| | - Kristopher R Bosse
- Center for Childhood Cancer Research and Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Poul H Sorensen
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada; Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, BC, Canada
| | - Kateryna Krytska
- Center for Childhood Cancer Research and Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Yael P Mosse
- Center for Childhood Cancer Research and Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | | | | - Benjamin A Garcia
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - John M Maris
- Center for Childhood Cancer Research and Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Sharon J Diskin
- Center for Childhood Cancer Research and Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
40
|
Zhu C, Liu LY, Ha A, Yamaguchi TN, Zhu H, Hugh-White R, Livingstone J, Patel Y, Kislinger T, Boutros PC. moPepGen: Rapid and Comprehensive Identification of Non-canonical Peptides. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.28.587261. [PMID: 38585946 PMCID: PMC10996593 DOI: 10.1101/2024.03.28.587261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Gene expression is a multi-step transformation of biological information from its storage form (DNA) into functional forms (protein and some RNAs). Regulatory activities at each step of this transformation multiply a single gene into a myriad of proteoforms. Proteogenomics is the study of how genomic and transcriptomic variation creates this proteomic diversity, and is limited by the challenges of modeling the complexities of gene-expression. We therefore created moPepGen, a graph-based algorithm that comprehensively generates non-canonical peptides in linear time. moPepGen works with multiple technologies, in multiple species and on all types of genetic and transcriptomic data. In human cancer proteomes, it enumerates previously unobservable noncanonical peptides arising from germline and somatic genomic variants, noncoding open reading frames, RNA fusions and RNA circularization. By enabling efficient detection and quantitation of previously hidden proteins in both existing and new proteomic data, moPepGen facilitates all proteogenomics applications. It is available at: https://github.com/uclahs-cds/package-moPepGen.
Collapse
Affiliation(s)
- Chenghao Zhu
- Department of Human Genetics, University of California, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA
- Institute for Precision Health, University of California, Los Angeles, CA, USA
- Department of Urology, University of California, Los Angeles, CA, USA
| | - Lydia Y. Liu
- Department of Human Genetics, University of California, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
- Vector Institute for Artificial Intelligence, Toronto, Canada
| | - Annie Ha
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Takafumi N. Yamaguchi
- Department of Human Genetics, University of California, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA
- Institute for Precision Health, University of California, Los Angeles, CA, USA
| | - Helen Zhu
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
- Vector Institute for Artificial Intelligence, Toronto, Canada
| | - Rupert Hugh-White
- Department of Human Genetics, University of California, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA
- Institute for Precision Health, University of California, Los Angeles, CA, USA
| | - Julie Livingstone
- Department of Human Genetics, University of California, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA
- Institute for Precision Health, University of California, Los Angeles, CA, USA
| | - Yash Patel
- Department of Human Genetics, University of California, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA
- Institute for Precision Health, University of California, Los Angeles, CA, USA
| | - Thomas Kislinger
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Paul C. Boutros
- Department of Human Genetics, University of California, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA
- Institute for Precision Health, University of California, Los Angeles, CA, USA
- Department of Urology, University of California, Los Angeles, CA, USA
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
| |
Collapse
|
41
|
Stroggilos R, Tserga A, Zoidakis J, Vlahou A, Makridakis M. Tissue proteomics repositories for data reanalysis. MASS SPECTROMETRY REVIEWS 2024; 43:1270-1284. [PMID: 37534389 DOI: 10.1002/mas.21860] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 07/17/2023] [Accepted: 07/18/2023] [Indexed: 08/04/2023]
Abstract
We are approaching the third decade since the establishment of the very first proteomics repositories back in the mid-'00s. New experimental approaches and technologies continuously enrich the field while producing vast amounts of mass spectrometry data. Together with initiatives to establish standard terminology and file formats, proteomics is rapidly transforming into a mature component of systems biology. Here we describe the ProteomeXchange consortium repositories. We specifically search, collect and evaluate public human tissue datasets (categorized as "complete" by the repository) submitted in 2015-2022, to both map the existing information and assess the data set reusability. Human tissue data are variably represented in the repositories reviewed, ranging between 10% and 25% of the total data submitted, with cancers being the most represented, followed by neuronal and cardiovascular diseases. About half of the retrieved data sets were found to lack annotations or metadata necessary to directly replicate the analysis. This poses a rough challenge to data reusability and highlights the need to increase awareness of the mage-tab file format for metadata in the community. Overall, proteomics repositories have evolved greatly over the past 7 years, as they have grown in size and become equipped with various powerful applications and tools that enable data searching and analytical tasks. However, to make the most of this potential, priority must be given to finding ways to secure detailed metadata for each submission, which is likely the next major milestone for proteomics repositories.
Collapse
Affiliation(s)
- Rafael Stroggilos
- Biomedical Research Foundation, Academy of Athens, Department of Biotechnology, Athens, Greece
| | - Aggeliki Tserga
- Biomedical Research Foundation, Academy of Athens, Department of Biotechnology, Athens, Greece
| | - Jerome Zoidakis
- Biomedical Research Foundation, Academy of Athens, Department of Biotechnology, Athens, Greece
| | - Antonia Vlahou
- Biomedical Research Foundation, Academy of Athens, Department of Biotechnology, Athens, Greece
| | - Manousos Makridakis
- Biomedical Research Foundation, Academy of Athens, Department of Biotechnology, Athens, Greece
| |
Collapse
|
42
|
Sweatt AJ, Griffiths CD, Groves SM, Paudel BB, Wang L, Kashatus DF, Janes KA. Proteome-wide copy-number estimation from transcriptomics. Mol Syst Biol 2024; 20:1230-1256. [PMID: 39333715 PMCID: PMC11535397 DOI: 10.1038/s44320-024-00064-3] [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: 08/02/2023] [Revised: 08/22/2024] [Accepted: 09/02/2024] [Indexed: 09/29/2024] Open
Abstract
Protein copy numbers constrain systems-level properties of regulatory networks, but proportional proteomic data remain scarce compared to RNA-seq. We related mRNA to protein statistically using best-available data from quantitative proteomics and transcriptomics for 4366 genes in 369 cell lines. The approach starts with a protein's median copy number and hierarchically appends mRNA-protein and mRNA-mRNA dependencies to define an optimal gene-specific model linking mRNAs to protein. For dozens of cell lines and primary samples, these protein inferences from mRNA outmatch stringent null models, a count-based protein-abundance repository, empirical mRNA-to-protein ratios, and a proteogenomic DREAM challenge winner. The optimal mRNA-to-protein relationships capture biological processes along with hundreds of known protein-protein complexes, suggesting mechanistic relationships. We use the method to identify a viral-receptor abundance threshold for coxsackievirus B3 susceptibility from 1489 systems-biology infection models parameterized by protein inference. When applied to 796 RNA-seq profiles of breast cancer, inferred copy-number estimates collectively re-classify 26-29% of luminal tumors. By adopting a gene-centered perspective of mRNA-protein covariation across different biological contexts, we achieve accuracies comparable to the technical reproducibility of contemporary proteomics.
Collapse
Affiliation(s)
- Andrew J Sweatt
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, 22908, USA
| | - Cameron D Griffiths
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, 22908, USA
| | - Sarah M Groves
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, 22908, USA
| | - B Bishal Paudel
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, 22908, USA
| | - Lixin Wang
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, 22908, USA
| | - David F Kashatus
- Department of Microbiology, Immunology & Cancer Biology, University of Virginia, Charlottesville, VA, 22908, USA
| | - Kevin A Janes
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, 22908, USA.
- Department of Biochemistry & Molecular Genetics, University of Virginia, Charlottesville, VA, 22908, USA.
| |
Collapse
|
43
|
Creighton CJ. Clinical proteomics towards multiomics in cancer. MASS SPECTROMETRY REVIEWS 2024; 43:1255-1269. [PMID: 36495097 DOI: 10.1002/mas.21827] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Recent technological advancements in mass spectrometry (MS)-based proteomics technologies have accelerated its application to study greater and greater numbers of human tumor specimens. Over the last several years, the Clinical Proteomic Tumor Analysis Consortium, the International Cancer Proteogenome Consortium, and others have generated MS-based proteomic profiling data combined with corresponding multiomics data on thousands of human tumors to date. Proteomic data sets in the public domain can be re-examined by other researchers with different questions in mind from what the original studies explored. In this review, we examine the increasing role of proteomics in studying cancer, along with the potential for previous studies and their associated data sets to contribute to improving the diagnosis and treatment of cancer in the clinical setting. We also explore publicly available proteomics and multi-omics data from cancer cell line models to show how such data may aid in identifying therapeutic strategies for cancer subsets.
Collapse
Affiliation(s)
- Chad J Creighton
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, USA
- Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
44
|
Zhang K, Xu Y, Zheng Y, Zhang T, Wu Y, Yan Y, Lei Y, Cao X, Wang X, Yan F, Lei Z, Brugger D, Chen Y, Deng L, Yang Y. Bifidobacterium pseudolongum-Derived Bile Acid from Dietary Carvacrol and Thymol Supplementation Attenuates Colitis via cGMP-PKG-mTORC1 Pathway. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2406917. [PMID: 39308187 DOI: 10.1002/advs.202406917] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 09/09/2024] [Indexed: 11/22/2024]
Abstract
Carvacrol and thymol (CAT) have been widely recognized for their antimicrobial and anti-inflammatory properties, yet their specific effects on colitis and the mechanisms involved remain insufficiently understood. This study establishes a causative link between CAT administration and colitis mitigation, primarily through the enhancement of Bifidobacterium pseudolongum abundance in the colon. This increase promotes the production of secondary bile acids, particularly hyodeoxycholic acid (HDCA) and 12-ketodeoxycholic acid (12-KCAC), which exert anti-inflammatory effects. Notably, CAT does not alleviate colitis symptoms in germ-free mice, indicating the necessity of gut microbiota. This research uncovers a novel regulatory mechanism where HDCA and 12-KCAC inhibit colonic inflammation by reducing the expression of transmembrane guanylate cyclase 1A in the colonic epithelium. This downregulation elevates intracellular Ca2+ and cGMP levels, activating protein kinase G (PKG). Activated PKG subsequently suppresses the mTOR signaling pathway, thereby ameliorating dextran sulfate sodium (DSS)-induced colonic damage. These findings highlight potential metabolites and therapeutic targets for preventing and treating colitis. Bifidobacterium pseudolongum, HDCA, and 12-KCAC emerge as promising candidates for therapeutic interventions in colitis and related disorders characterized by impaired tight junction function.
Collapse
Affiliation(s)
- Ke Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Yangbin Xu
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Yining Zheng
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Ting Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Yujiang Wu
- Institute of Animal Sciences, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa, 850009, China
| | - Yiting Yan
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Yu Lei
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Xi Cao
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Xiaolong Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Frances Yan
- Novus International Inc, Research Park Drive, Saint Charles, MO, 63304, USA
| | - Zhaomin Lei
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China
| | - Daniel Brugger
- Institute of Animal Nutrition and Dietetics, Vetsuisse-Faculty, University of Zurich, Zurich, 8057, Switzerland
| | - Yulin Chen
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Lu Deng
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Yuxin Yang
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| |
Collapse
|
45
|
Junior SM, Levander F. Automated multiplexed affinity-based enrichment of peptides for LC-MS/MS plasma proteomics. Proteomics 2024; 24:e2400049. [PMID: 39192483 DOI: 10.1002/pmic.202400049] [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: 02/06/2024] [Revised: 08/05/2024] [Accepted: 08/19/2024] [Indexed: 08/29/2024]
Abstract
Plasma proteomics offers high potential for biomarker discovery, as plasma is collected through a minimally invasive procedure and constitutes the most complex human-derived proteome. However, the wide dynamic range poses a significant challenge. Here, we propose a semi-automated method based on the use of multiple single chain variable fragment antibodies, each enriching for peptides found in up to a few hundred proteins. This approach allows for the analysis of a complementary fraction compared to full proteome analysis. Proteins from pooled plasma were extracted and digested before testing the performance of 29 different antibodies with the aim of reproducibly maximizing peptide enrichment. Our results demonstrate the enrichment of 3662 peptides not detected in neat plasma or negative controls. Moreover, most antibodies were able to enrich for at least 155 peptides across different levels of abundance in plasma. To further reduce analysis time, a combination of antibodies was used in a multiplexed setting. Repeated sample analyses showed low coefficients of variation, and the method is flexible in terms of affinity binders. It does not impose drastic increases in instrument time, thus showing excellent potential for usage in large scale discovery projects.
Collapse
Affiliation(s)
| | - Fredrik Levander
- Department of Immunotechnology, Lund University, Lund, Sweden
- National Bioinformatics Infrastructure Sweden (NBIS), Science for Life Laboratory, Lund University, Lund, Sweden
| |
Collapse
|
46
|
Choi S, An JY. Multiomics in cancer biomarker discovery and cancer subtyping. Adv Clin Chem 2024; 124:161-195. [PMID: 39818436 DOI: 10.1016/bs.acc.2024.10.004] [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] [Indexed: 01/18/2025]
Abstract
The advent of multiomics has ushered in a new era of cancer research characterized by integrated genomic, transcriptomic and proteomic analysis to unravel the complexities of cancer biology and facilitate the discovery of novel biomarkers. This chapter provides a comprehensive overview of the concept of multiomics, detailing the significant advances in the underlying technologies and their contributions to our understanding of cancer. It delves into the evolution of genomics and transcriptomics, breakthroughs in proteomics, and overarching progress in multiomic methodologies, highlighting their collective impact on cancer biomarker discovery. Furthermore, this chapter explores the computational methods essential for multiomic studies, including clustering techniques for delineating cancer subtypes, strategies for estimating molecular features and activities, and utility of pathway enrichment analyses for interpreting multiomic datasets. Particular focus has been placed on the application of these methods for identifying distinct cancer subtypes, thereby enabling a more personalized approach to cancer treatment. Through a detailed discussion of the scientific principles, technological advancements, and practical applications of multiomics, this chapter aims to underscore the pivotal role of multiomics in advancing cancer research and paving the way for personalized medicine. The insights provided herein not only illuminate the current landscape of cancer biomarker discovery, but also forecast future directions of multiomics research in oncology, advocating for a more integrated and nuanced approach to understanding and combating cancer.
Collapse
Affiliation(s)
- Seunghwan Choi
- School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul, Republic of Korea
| | - Joon-Yong An
- School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul, Republic of Korea; Department of Integrated Biomedical and Life Science, Korea University, Seoul, Republic of Korea; BK21FOUR R&E Center for Learning Health Systems, Korea University, Seoul, Republic of Korea; L-HOPE Program for Community-Based Total Learning Health Systems, Korea University, Seoul, Republic of Korea.
| |
Collapse
|
47
|
Smitha Pillai K, Laxton O, Li G, Lin J, Karginova O, Nanda R, Olopade OI, Tay S, Moellering RE. Single-cell chemoproteomics identifies metastatic activity signatures in breast cancer. SCIENCE ADVANCES 2024; 10:eadp2622. [PMID: 39441940 PMCID: PMC11498211 DOI: 10.1126/sciadv.adp2622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 09/18/2024] [Indexed: 10/25/2024]
Abstract
Protein activity state, rather than protein or mRNA abundance, is a biologically regulated and relevant input to many processes in signaling, differentiation, development, and diseases such as cancer. While there are numerous methods to detect and quantify mRNA and protein abundance in biological samples, there are no general approaches to detect and quantify endogenous protein activity with single-cell resolution. Here, we report the development of a chemoproteomic platform, single-cell activity-dependent proximity ligation, which uses automated, microfluidics-based single-cell capture and nanoliter volume manipulations to convert the interactions of family-wide chemical activity probes with native protein targets into multiplexed, amplifiable oligonucleotide barcodes. We demonstrate accurate, reproducible, and multiplexed quantitation of a six-enzyme (Ag-6) panel with known ties to cancer cell aggressiveness directly in single cells. We further identified increased Ag-6 enzyme activity across breast cancer cell lines of increasing metastatic potential, as well as in primary patient-derived tumor cells and organoids from patients with breast cancer.
Collapse
Affiliation(s)
- Kavya Smitha Pillai
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
- Institute for Genomics and Systems Biology, The University of Chicago, Chicago, IL 60637, USA
| | - Olivia Laxton
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
- Institute for Genomics and Systems Biology, The University of Chicago, Chicago, IL 60637, USA
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL 60637, USA
| | - Gang Li
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
- Institute for Genomics and Systems Biology, The University of Chicago, Chicago, IL 60637, USA
| | - Jing Lin
- Institute for Genomics and Systems Biology, The University of Chicago, Chicago, IL 60637, USA
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL 60637, USA
| | - Olga Karginova
- Center for Clinical Cancer Genetics and Global Health, Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Rita Nanda
- Center for Clinical Cancer Genetics and Global Health, Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Olufunmilayo I. Olopade
- Center for Clinical Cancer Genetics and Global Health, Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Savaş Tay
- Institute for Genomics and Systems Biology, The University of Chicago, Chicago, IL 60637, USA
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL 60637, USA
| | - Raymond E. Moellering
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
- Institute for Genomics and Systems Biology, The University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
48
|
Wong GYM, Li J, McKay M, Castaneda M, Bhimani N, Diakos C, Hugh TJ, Molloy MP. Proteogenomic Characterization of Early Intrahepatic Recurrence after Curative-Intent Treatment of Colorectal Liver Metastases. J Proteome Res 2024; 23:4523-4537. [PMID: 39264718 DOI: 10.1021/acs.jproteome.4c00440] [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] [Indexed: 09/14/2024]
Abstract
Clinical and pathological factors are insufficient to accurately identify patients at risk of early recurrence after curative-intent treatment of colorectal liver metastases (CRLM). This study aimed to identify candidate prognostic proteogenomic biomarkers for early intrahepatic recurrence after curative-intent resection of CRLM. Patients diagnosed with intrahepatic recurrence within 6 months of liver resection were categorized as the "early recurrence" group, while those who achieved a recurrence-free status for 10 years were designated as "durable remission". Comprehensive genomic and proteomic profiling of fresh frozen samples from these prognostically distinct groups was performed using the TruSight Oncology 500 assay and label-free data-dependent acquisition liquid chromatography-mass spectrometry. Genetic alterations were identified in 117 of the 523 profiled genes in patients with early recurrence. The most common somatic mutations linked to early recurrence were TP53 (88%), APC (71%), KRAS (38%), and SMAD4 (21%). SMAD4 alterations were absent in samples from patients with a durable remission. Calponin-2, versican core protein, glutathione peroxidase 3, fibulin-5, and amyloid-β precursor protein were upregulated more than 2-fold in early recurrence. Exploratory analysis of these proteogenomic biomarkers suggests that SMAD4, calponin-2, and glutathione peroxidase 3 may have the potential to predict early recurrence, enabling improved prognostication and precision oncology in CRLM.
Collapse
Affiliation(s)
- Geoffrey Yuet Mun Wong
- Department of Upper Gastrointestinal Surgery, Royal North Shore Hospital, Sydney, New South Wales 2065, Australia
- Northern Clinical School, The University of Sydney, Sydney, New South Wales 2065, Australia
- Bowel Cancer and Biomarker Research Laboratory, Kolling Institute, St Leonards, New South Wales 2065, Australia
| | - Jun Li
- Bowel Cancer and Biomarker Research Laboratory, Kolling Institute, St Leonards, New South Wales 2065, Australia
| | - Matthew McKay
- Bowel Cancer and Biomarker Research Laboratory, Kolling Institute, St Leonards, New South Wales 2065, Australia
| | - Miguel Castaneda
- Bowel Cancer and Biomarker Research Laboratory, Kolling Institute, St Leonards, New South Wales 2065, Australia
| | - Nazim Bhimani
- Department of Upper Gastrointestinal Surgery, Royal North Shore Hospital, Sydney, New South Wales 2065, Australia
- Faculty of Medicine and Health, University of Sydney, Camperdown, New South Wales 2050, Australia
| | - Connie Diakos
- Northern Clinical School, The University of Sydney, Sydney, New South Wales 2065, Australia
- Department of Medical Oncology, Royal North Shore Hospital, Sydney, New South Wales 2065, Australia
| | - Thomas J Hugh
- Department of Upper Gastrointestinal Surgery, Royal North Shore Hospital, Sydney, New South Wales 2065, Australia
- Northern Clinical School, The University of Sydney, Sydney, New South Wales 2065, Australia
| | - Mark P Molloy
- Bowel Cancer and Biomarker Research Laboratory, Kolling Institute, St Leonards, New South Wales 2065, Australia
| |
Collapse
|
49
|
Shi Q, Zhang P, Hu Q, Zhang T, Hou R, Yin S, Zou Y, Chen F, Jiao S, Si L, Zheng B, Chen Y, Zhan T, Liu Y, Zhu W, Qi N. Role of TOMM34 on NF-κB activation-related hyperinflammation in severely ill patients with COVID-19 and influenza. EBioMedicine 2024; 108:105343. [PMID: 39276680 PMCID: PMC11418153 DOI: 10.1016/j.ebiom.2024.105343] [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: 03/07/2024] [Revised: 08/21/2024] [Accepted: 09/02/2024] [Indexed: 09/17/2024] Open
Abstract
BACKGROUND Highly pathogenic respiratory RNA viruses such as SARS-CoV-2 and its associated syndrome COVID-19 pose a tremendous threat to the global public health. Innate immune responses to SARS-CoV-2 depend mainly upon the NF-κB-mediated inflammation. Identifying unknown host factors driving the NF-κB activation and inflammation is crucial for the development of immune intervention strategies. METHODS Published single-cell RNA sequencing (scRNA-seq) data was used to analyze the differential transcriptome profiles of bronchoalveolar lavage (BAL) cells between healthy individuals (n = 27) and patients with severe COVID-19 (n = 21), as well as the differential transcriptome profiles of peripheral blood mononuclear cells (PBMCs) between healthy individuals (n = 22) and severely ill patients with COVID-19 (n = 45) or influenza (n = 16). Loss-of-function and gain-of-function assays were performed in diverse viruses-infected cells and male mice models to identify the role of TOMM34 in antiviral innate immunity. FINDINGS TOMM34, together with a list of genes encoding pro-inflammatory cytokines and antiviral immune proteins, was transcriptionally upregulated in circulating monocytes, lung epithelium and innate immune cells from individuals with severe COVID-19 or influenza. Deficiency of TOMM34/Tomm34 significantly impaired the type I interferon responses and NF-κB-mediated inflammation in various human/murine cell lines, murine bone marrow-derived macrophages (BMDMs) and in vivo. Mechanistically, TOMM34 recruits TRAF6 to facilitate the K63-linked polyubiquitination of NEMO upon viral infection, thus promoting the downstream NF-κB activation. INTERPRETATION In this study, viral induction of TOMM34 is positively correlated with the hyperinflammation in severely ill patients with COVID-19 and influenza. Our findings also highlight the physiological role of TOMM34 in the innate antiviral signallings. FUNDING A full list of funding sources can be found in the acknowledgements section.
Collapse
Affiliation(s)
- Qiwen Shi
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Pengfei Zhang
- Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou, 510005, China
| | - Qingtao Hu
- State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Tianxin Zhang
- Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou, 510005, China
| | - Ruixia Hou
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Shengxiang Yin
- Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou, 510005, China
| | - Yilin Zou
- Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou, 510005, China
| | - Fenghua Chen
- State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Shuang Jiao
- Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou, 510005, China
| | - Lanlan Si
- State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Bangjin Zheng
- Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou, 510005, China
| | - Yichao Chen
- Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou, 510005, China
| | - Tingzhu Zhan
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Yongxiang Liu
- Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou, 510005, China.
| | - Wenting Zhu
- Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou, 510005, China.
| | - Nan Qi
- State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China; Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou, 510005, China.
| |
Collapse
|
50
|
Wang Z, Yu H, Bao W, Qu M, Wang Y, Zhang L, Liu X, Liu C, He M, Li J, Dong Z, Zhang Y, Yang B, Hou J, Xu C, Wang L, Li X, Gao X, Yang C. Proteomic and phosphoproteomic landscape of localized prostate cancer unveils distinct molecular subtypes and insights into precision therapeutics. Proc Natl Acad Sci U S A 2024; 121:e2402741121. [PMID: 39320917 PMCID: PMC11459144 DOI: 10.1073/pnas.2402741121] [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: 08/27/2024] [Indexed: 09/26/2024] Open
Abstract
Building upon our previous investigation of genomic, epigenomic, and transcriptomic profiles of prostate cancer in China, we conducted a comprehensive analysis of proteomic and phosphoproteomic profiles of 82 tumor tissues and matched adjacent normal tissues from 41 Chinese patients with localized prostate cancer. We identified three distinct proteomic subtypes with significant difference in both molecular features and clinical prognosis. Notably, these proteomic subtypes exhibited a parallel degree of heterogeneity in the phosphoproteome, featuring unique metabolism, proliferation, and immune infiltration characteristics. We further demonstrated that a combination of proteins and phosphosites serves as the most effective biomarkers in prostate cancer to predict biochemical recurrence. Through an integrated multiomics analysis, we revealed mechanistic differences underlying different proteomic subtypes and highlighted the potential significance of Serine/arginine-rich splicing factor 1 (SRSF1) phosphorylation in promoting the malignant characteristics of prostate cancer cells. Our multiomics data provide valuable resources for understanding the molecular mechanisms of prostate cancer within the Chinese population, which have the potential to inform the development of personalized treatment strategies and enhance prognostic analyses for prostate cancer patients.
Collapse
Affiliation(s)
- Zengming Wang
- Department of Urology, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai200433, China
- Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai200031, China
| | - Haolan Yu
- Department of Urology, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai200433, China
- Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai200031, China
| | - Wei Bao
- Department of Urology, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai200433, China
- Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai200031, China
| | - Min Qu
- Department of Urology, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai200433, China
- Shanghai Key Laboratory of Cell Engineering, Shanghai200433, China
| | - Yan Wang
- Department of Urology, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai200433, China
- Shanghai Key Laboratory of Cell Engineering, Shanghai200433, China
| | - Liandong Zhang
- Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai200031, China
| | - Xubing Liu
- Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai200031, China
| | - Chen Liu
- Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai200031, China
| | - Miaoxia He
- Department of Pathology, Changhai Hospital, Second Military Medical University, Shanghai200433, China
| | - Jing Li
- Center for Translational Medicine, Second Military Medical University (Naval Medical University), Shanghai200433, China
| | - Zhenyang Dong
- Department of Urology, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai200433, China
| | - Yun Zhang
- Department of Urology, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai200433, China
| | - Bo Yang
- Department of Urology, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai200433, China
- Shanghai Key Laboratory of Cell Engineering, Shanghai200433, China
| | - Jianguo Hou
- Department of Urology, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai200433, China
- Shanghai Key Laboratory of Cell Engineering, Shanghai200433, China
| | - Chuanliang Xu
- Department of Urology, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai200433, China
- Shanghai Key Laboratory of Cell Engineering, Shanghai200433, China
| | - Linhui Wang
- Department of Urology, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai200433, China
- Shanghai Key Laboratory of Cell Engineering, Shanghai200433, China
| | - Xin Li
- Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai200031, China
| | - Xu Gao
- Department of Urology, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai200433, China
- Shanghai Key Laboratory of Cell Engineering, Shanghai200433, China
| | - Chenghua Yang
- Department of Urology, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai200433, China
- Shanghai Key Laboratory of Cell Engineering, Shanghai200433, China
| |
Collapse
|