1
|
Massah S, Pinette N, Foo J, Datta S, Guo M, Bell R, Haegert A, Tekoglu TE, Terrado M, Volik S, Bihan SL, Bui JM, Lack NA, Gleave ME, Rhie SK, Collins CC, Gsponer J, Lallous N. AR-V7 condensates drive androgen-independent transcription in castration resistant prostate cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.08.631986. [PMID: 39868336 PMCID: PMC11760419 DOI: 10.1101/2025.01.08.631986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Biomolecular condensates organize cellular environments and regulate key processes such as transcription. We previously showed that full-length androgen receptor (AR-FL), a major oncogenic driver in prostate cancer (PCa), forms nuclear condensates upon androgen stimulation in androgen-sensitive PCa cells. Disrupting these condensates impairs AR-FL transcriptional activity, highlighting their functional importance. However, resistance to androgen deprivation therapy often leads to castration-resistant prostate cancer (CRPC), driven by constitutively active splice variants like AR variant 7 (AR-V7). The mechanisms underlying AR-V7's role in CRPC remain unclear. In this study, we characterized the condensate-forming ability of AR-V7 and compared its phase behavior with AR-FL across a spectrum of PCa models and in vitro conditions. Our findings indicate that cellular context can influence AR-V7's condensate-forming capacity. Unlike AR-FL, AR-V7 spontaneously forms condensates in the absence of androgen stimulation and functions independently of AR-FL in CRPC models. However, AR-V7 requires a higher concentration to form condensates, both in cellular contexts and in vitro . We further reveal that AR-V7 drives transcription via both condensate-dependent and condensate-independent mechanisms. Using an AR-V7 mutant incapable of forming condensates, while retaining nuclear localization and DNA-binding ability, we reveal that the condensate-dependent regime activates part of the oncogenic KRAS pathway in CRPC models. Genes under this condensate-dependent regime were found to harbor significantly higher numbers of AR-binding sites and exhibited boosted expression in response to AR-V7. These findings uncover a previously unrecognized role of AR-V7 condensate formation in driving oncogenic transcriptional programs and shed light on its unique contribution to CRPC progression. Highlights AR-V7 condensates form independently of both androgens and AR-FL in CRPC models.AR-V7 mediates condensate-dependent and independent transcriptionCondensate-dependent transcription enables boosted expression of oncogenic KRAS genesCondensate-dependent genes exhibit an exponential increase in expression, with a higher number of AR binding sites potentially playing a key role in their reliance on condensate formation.
Collapse
|
2
|
Meng X, Du Y, Liu C, Zhai Z, Pan J. GTO: a comprehensive gene therapy omnibus. Nucleic Acids Res 2025; 53:D1393-D1403. [PMID: 39540438 PMCID: PMC11701634 DOI: 10.1093/nar/gkae1051] [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: 08/07/2024] [Revised: 10/17/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
Gene therapy, which involves the delivery of genetic material into cells to correct an underlying genetic problem, has emerged as a promising approach for treating various conditions. To promote research in this rapidly evolving field, we developed the Gene Therapy Omnibus (GTO) (http://www.inbirg.com/gto/), a comprehensive resource containing detailed clinical trial data and molecular information related to gene therapy. The GTO includes 6333 clinical trial records and 3466 transcriptome profiles, with information on 614 altered genes and 22 types of gene therapy, including DNA therapies, RNA therapies and genetically-modified cell therapies. For each gene therapy product in a clinical trial, detailed information, such as altered gene name, structural components, indication, vector information, phase of the clinical trial, clinical outcomes and adverse effects, is provided when available. Additionally, 345 comparison datasets, including 29 single-cell RNA-sequencing datasets comprising information on both gene therapy and control samples, were established. Differential gene expression and downstream functional enrichment analyses were performed through standardized pipelines to elucidate the molecular alterations induced by gene therapy. The user-friendly interface of the GTO supports efficient data retrieval, visualization and analysis, making it an invaluable resource for researchers and clinicians performing clinical research on gene therapy and the underlying mechanisms.
Collapse
Affiliation(s)
- Xuehang Meng
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, College of Pharmacy, Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Yujia Du
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, College of Pharmacy, Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Chang Liu
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, College of Pharmacy, Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Zhaoyu Zhai
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, College of Pharmacy, Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Jianbo Pan
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, College of Pharmacy, Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
- Precision Medicine Center, The Second Affiliated Hospital of Chongqing Medical University, No. 76 Linjiang Road, Yuzhong District, Chongqing, 400010, China
| |
Collapse
|
3
|
Elmanfi S, Onyedibe KI, Aryal UK, Könönen E, Sintim HO, Gürsoy UK. Activation of cellular responses by cyclic dinucleotides and porphyromonas gingivalis lipopolysaccharide: a proteomic study on gingival fibroblasts. J Oral Microbiol 2024; 17:2431453. [PMID: 39669221 PMCID: PMC11632945 DOI: 10.1080/20002297.2024.2431453] [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: 04/01/2024] [Revised: 09/02/2024] [Accepted: 09/17/2024] [Indexed: 12/14/2024] Open
Abstract
Background Bacterial cyclic dinucleotides (CDNs), cyclic di-guanosine monophosphate (c-di-GMP), and cyclic di-adenosine monophosphate (c-di-AMP) upregulate interferon signaling proteins of human gingival fibroblasts (HGFs). However, the simultaneous effect of bacterial CDNs and lipopolysaccharides (LPS) on the HGF proteome is unknown. Aim The aim was to apply an unbiased proteomics approach to evaluate how simultaneous exposure to CDNs and Porphyromonas gingivalis (Pg) LPS affect the global proteome of HGFs. Methods The proteomic responses of HGFs were examined under three different treatment conditions (c-di-AMP+Pg LPS, c-di-GMP+Pg LPS, and Pg LPS alone) by label-free quantitative mass spectrometry analysis. Results Simultaneous exposure to CDNs and Pg LPS significantly upregulated innate immunity-related and interferon signaling-related proteins, such as ubiquitin-like protein ISG15 (ISG15), deoxynucleoside triphosphate triphosphohydrolase (SAMHD1), interferon regulatory factor 9 (IRF-9), interferon-induced GTP-binding protein Mx (MX)1, and MX2. Interferon signaling pathway was the most significantly regulated canonical pathway in both CDN treatment groups. Conclusion Simultaneous exposure to CDNs and Pg LPS stimulates the periodontal immune response by activating the anti-microbial cellular responses of HGFs with some notable differences from individual exposures.
Collapse
Affiliation(s)
- Samira Elmanfi
- Department of Periodontology, Institute of Dentistry, University of Turku, Turku, Finland
| | - Kenneth I. Onyedibe
- Department of Chemistry, Purdue University, West Lafayette, USA
- Purdue Institute for Inflammation, Immunology and Infectious Disease and Purdue Institute for Drug Discovery, Purdue University, West Lafayette, USA
- Department of Biomedical Sciences, Mercer University School of Medicine, MaconGA, USA
| | - Uma K. Aryal
- Purdue Proteomics Facility, Bindley Bioscience Center, Purdue University, West Lafayette, USA
- Department of Comparative Pathobiology, Purdue University, West Lafayette, USA
| | - Eija Könönen
- Department of Periodontology, Institute of Dentistry, University of Turku, Turku, Finland
| | - Herman O. Sintim
- Department of Chemistry, Purdue University, West Lafayette, USA
- Purdue Institute for Inflammation, Immunology and Infectious Disease and Purdue Institute for Drug Discovery, Purdue University, West Lafayette, USA
| | - Ulvi Kahraman Gürsoy
- Department of Periodontology, Institute of Dentistry, University of Turku, Turku, Finland
| |
Collapse
|
4
|
Lun Y, Sun J, Wei L, Liu B, Li Z, Dong W, Zhao W. SPINK13 acts as a tumor suppressor in hepatocellular carcinoma by inhibiting Akt phosphorylation. Cell Death Dis 2024; 15:822. [PMID: 39537605 PMCID: PMC11561306 DOI: 10.1038/s41419-024-07214-3] [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: 04/04/2024] [Revised: 10/29/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024]
Abstract
The PI3K/Akt pathway is overexpressed in nearly 50% of hepatocellular carcinomas and inhibits apoptosis by promoting the expression of antiapoptotic genes. Serine protease inhibitors have been shown to induce apoptosis in hepatoma cells by downregulating SPINK13 in the PI3K/Akt pathway. In this study, SPINK13 was expressed in lentiviral vectors. Changes in signaling pathway adapter proteins, apoptosis regulatory proteins, cell cycle regulatory proteins, and the biological behavior of hepatocellular carcinoma were observed in cell and nude mouse xenograft models. The underlying mechanism of endogenous SPINK13-induced apoptosis in hepatocellular carcinoma cells was explored via transcriptomics. As a result, endogenous SPINK13 might inhibit the activity of Furin protease, downregulate the Notch1/Hes1 pathway in a binding manner, activate the direct effector PTEN, inhibit Akt phosphorylation, inactivate the downstream PI3K/Akt pathway, and ultimately lead to mitochondrial apoptosis and cell cycle arrest in hepatoma cells. Therefore, the Notch1/Hes1/PTEN pathway may act upstream of SPINK13 to downregulate the PI3K/Akt signaling pathway. Our study helps elucidate the underlying mechanism of SPINK13 in anti-hepatocellular carcinoma and lays a theoretical foundation for the development of novel therapeutic serine protease inhibitors.
Collapse
MESH Headings
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/metabolism
- Liver Neoplasms/pathology
- Liver Neoplasms/genetics
- Liver Neoplasms/metabolism
- Humans
- Proto-Oncogene Proteins c-akt/metabolism
- Animals
- Mice, Nude
- Phosphorylation
- Apoptosis/genetics
- Mice
- Signal Transduction
- Cell Line, Tumor
- Phosphatidylinositol 3-Kinases/metabolism
- PTEN Phosphohydrolase/metabolism
- PTEN Phosphohydrolase/genetics
- Serine Peptidase Inhibitors, Kazal Type/metabolism
- Serine Peptidase Inhibitors, Kazal Type/genetics
- Receptor, Notch1/metabolism
- Receptor, Notch1/genetics
- Trypsin Inhibitor, Kazal Pancreatic/metabolism
- Trypsin Inhibitor, Kazal Pancreatic/genetics
- Transcription Factor HES-1/metabolism
- Transcription Factor HES-1/genetics
- Hep G2 Cells
- Mice, Inbred BALB C
- Cell Proliferation
- Gene Expression Regulation, Neoplastic
Collapse
Affiliation(s)
- Yongzhi Lun
- Key Laboratory of Screening and Control of Infectious Diseases, Fujian Provincial University, Quanzhou Medical College, Quanzhou, 362011, Fujian, China.
- Department of Laboratory Medicine, Putian University, Putian, 351100, Fujian, China.
| | - Jie Sun
- Key Laboratory of Screening and Control of Infectious Diseases, Fujian Provincial University, Quanzhou Medical College, Quanzhou, 362011, Fujian, China
- Department of Laboratory Medicine, Putian University, Putian, 351100, Fujian, China
| | - Ling Wei
- Beijing Centre for Physical and Chemical Analysis, 100089, Beijing, China
| | - Ben Liu
- Key Laboratory of Screening and Control of Infectious Diseases, Fujian Provincial University, Quanzhou Medical College, Quanzhou, 362011, Fujian, China
- Department of Laboratory Medicine, Putian University, Putian, 351100, Fujian, China
| | - Zhixue Li
- Key Laboratory of Screening and Control of Infectious Diseases, Fujian Provincial University, Quanzhou Medical College, Quanzhou, 362011, Fujian, China
- College of Chemistry, Fuzhou University, Fuzhou, 350108, Fujian, China
| | - Wen Dong
- Department of Laboratory Medicine, Putian University, Putian, 351100, Fujian, China
| | - Wenqi Zhao
- Key Laboratory of Screening and Control of Infectious Diseases, Fujian Provincial University, Quanzhou Medical College, Quanzhou, 362011, Fujian, China
| |
Collapse
|
5
|
Yang X, Jiang S, Yuan Z, Jiang J, Yang M, Luo J, Ye T. SPINK4 modulates inhibition of glycolysis against colorectal cancer progression. BIOMOLECULES & BIOMEDICINE 2024; 24:1571-1585. [PMID: 38747892 PMCID: PMC11496861 DOI: 10.17305/bb.2024.10338] [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: 02/05/2024] [Revised: 05/08/2024] [Accepted: 05/08/2024] [Indexed: 10/18/2024]
Abstract
Dysregulation of glycolysis is frequently linked to aggressive tumor activity in colorectal cancer (CRC). Although serine peptidase inhibitor, Kazal type 4 (SPINK4) has been linked to CRC, its exact linkage to glycolytic processes and gene expression remains unclear. Differentially expressed genes (DEGs) were screened from two CRC-related datasets (GSE32323 and GSE141174), followed by expression and prognostic analysis of SPINK4. In vitro techniques such as flow cytometry, western blotting, transwell assay, and quantitative real-time polymerase chain reaction (qRT-PCR) were used to assess SPINK4 expression in CRC cells. Its effects on apoptosis, glycolysis, and the cell cycle were also investigated. Finally, the impact of SPINK4 overexpression on tumor development was assessed using a xenograft model, while histological and immunohistochemical analyses characterized SPINK4 expression patterns in CRC tissues. SPINK4 expression was downregulated in CRC, correlating with poor patient prognosis. In vitro assays confirmed that overexpression of SPINK4 reduced CRC cell proliferation, invasion, and migration, while its knockdown promoted these processes and caused G1 arrest. SPINK4 also regulated apoptosis by altering caspase activation and Bcl-2 expression. Besides, SPINK4 overexpression altered glycolytic activity, reduced 2-Deoxy-D-glucose (2-DG) absorption, and controlled critical glycolytic enzymes, resulting in alterations in metabolic pathways, whereas SPINK4 knockdown reversed this effect. SPINK4 overexpression significantly reduced tumor volume in vivo, indicating its inhibitory role in carcinogenesis. Moreover, high expression of SPINK4, hexokinase 2 (HK2), glucose transporter 1 (GLUT1), lactate dehydrogenase A (LDHA), and pyruvate kinase M2 (PKM2) was observed in CRC tissues. As a key inhibitor of glycolytic metabolism in CRC, SPINK4 promises metabolic intervention in CRC therapy due to its impact on tumor growth and cell proliferation.
Collapse
Affiliation(s)
- Xiaodi Yang
- Department of Oncology, Minhang Branch, Zhongshan Hospital, Fudan University, Shanghai, China
- Key laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital and AHS, Fudan University, Shanghai, China
| | - Sen Jiang
- Department of Internal Emergency Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
- Shanghai East Clinical Medical College, Nanjing Medical University, Shanghai, China
| | - Zhen Yuan
- Department of Oncology, Minhang Branch, Zhongshan Hospital, Fudan University, Shanghai, China
- Key laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital and AHS, Fudan University, Shanghai, China
| | - Jun Jiang
- State Key Laboratory of Genetic Engineering, Shanghai Engineering Research Center of Industrial Microorganisms, School of Life Sciences, Fudan University, Shanghai, China
| | - Mengxuan Yang
- Department of Oncology, Minhang Branch, Zhongshan Hospital, Fudan University, Shanghai, China
- Key laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital and AHS, Fudan University, Shanghai, China
| | - Jing Luo
- Department of Oncology, Minhang Branch, Zhongshan Hospital, Fudan University, Shanghai, China
- Key laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital and AHS, Fudan University, Shanghai, China
| | - Tao Ye
- Department of Oncology, Minhang Branch, Zhongshan Hospital, Fudan University, Shanghai, China
- Key laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital and AHS, Fudan University, Shanghai, China
| |
Collapse
|
6
|
Chen Y, Li Q, Yu X, Lu L, Zhou Z, Li M, Xia R, Gan X, Hu Y, Guo G, Guo J, Li H, Li Q, Liu Y, Liu X, Sun M. The microprotein HDSP promotes gastric cancer progression through activating the MECOM-SPINK1-EGFR signaling axis. Nat Commun 2024; 15:8381. [PMID: 39333095 PMCID: PMC11437185 DOI: 10.1038/s41467-024-50986-7] [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: 07/07/2023] [Accepted: 07/27/2024] [Indexed: 09/29/2024] Open
Abstract
The presence of noncanonical open reading frames within lncRNAs (long non-coding RNAs) suggests their potential for translation, yielding various functional peptides or proteins. However, the existence and specific roles of these products in gastric cancer remain largely unclear. Here we identify the HOXA10-HOXA9-derived small protein (HDSP) in gastric cancer through comprehensive analysis and experimental validation, including mass spectrometry and western blotting. HDSP exhibits high expression and oncogenic roles in gastric cancer. Mechanistically, HDSP blocks TRIM25-mediated ubiquitination and degradation by interacting with MECOM, leading to MECOM accumulation and enhanced SPINK1 transcription-a gene promoting cancer via the EGFR signaling pathway. Furthermore, MECOM fosters HOXA10-HOXA9 transcription, establishing a feedback loop activating SPINK1-EGFR signaling. HDSP knockdown inhibits tumor growth in a PDX (patient-derived xenograft) model, and infusion of an artificially synthesized HDSP peptide as a neoantigen enhances immune cell-mediated anti-tumor efficacy against gastric cancer in vitro and in vivo. These findings propose HDSP as a potential therapeutic target or neoantigen candidate for gastric cancer treatment.
Collapse
Affiliation(s)
- Yuli Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, China
- Suzhou Cancer Center Core Laboratory, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, 215001, China
| | - Qiuhui Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, China
| | - Xiang Yu
- Department of General Surgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, China
| | - Lu Lu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, China
| | - Zihan Zhou
- The First Clinical Medical College of Nanjing Medical University, Nanjing, 211166, China
| | - Mingjie Li
- Asset Management Company, Nanjing Medical University, Nanjing, 211166, China
| | - Rui Xia
- Department of Laboratory, Nanjing Chest Hospital, Nanjing, 210029, China
| | - Xiongkang Gan
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Yanming Hu
- Suzhou Cancer Center Core Laboratory, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, 215001, China
| | - Guoqing Guo
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, China
| | - Jiahao Guo
- Suzhou Cancer Center Core Laboratory, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, 215001, China
| | - Hanyang Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, China
| | - Qiunuo Li
- The First Clinical Medical College of Nanjing Medical University, Nanjing, 211166, China
| | - Yanwen Liu
- Department of Oncology, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, China
| | - Xianghua Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, China.
| | - Ming Sun
- Suzhou Cancer Center Core Laboratory, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, 215001, China.
| |
Collapse
|
7
|
Kawabata-Iwakawa R, Iwasa N, Satoh K, Colinge J, Shimada M, Takeuchi S, Fujiwara H, Eguchi H, Oishi T, Sugiyama T, Suzuki M, Hasegawa K, Fujiwara K, Nishiyama M. Prediction of response to promising first-line chemotherapy in ovarian cancer patients with residual peritoneal tumors: practical biomarkers and robust multiplex models. Int J Clin Oncol 2024; 29:1334-1346. [PMID: 38767719 DOI: 10.1007/s10147-024-02552-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: 12/27/2023] [Accepted: 05/14/2024] [Indexed: 05/22/2024]
Abstract
BACKGROUND Platinum/taxane (TC) chemotherapy with debulking surgery stays the mainstay of the treatment in ovarian cancer patients with peritoneal metastasis, and recently its novel modality, intraperitoneal carboplatin with dose-dense paclitaxel (ddTCip), was shown to have greater therapeutic impact. Nevertheless, the response varies among patients and consequent recurrence, or relapse often occurs. Discovery of therapeutic response predictor to ddTCip and/or TC therapy is eagerly awaited to improve the treatment outcome. METHODS Using datasets in 76 participants in our ddTCip study and published databases on patients received TC therapy, we first validated a total of 75 previously suggested markers, sought out more active biomarkers through the association analyses of genome-wide transcriptome and genotyping data with progression-free survival (PFS) and adverse events, and then developed multiplex statistical prediction models for PFS and toxicity by mainly using multiple regression analysis and the classification and regression tree (CART) algorithm. RESULTS The association analyses revealed that SPINK1 could be a possible biomarker of ddTCip efficacy, while ABCB1 rs1045642 and ERCC1 rs11615 would be a predictor of hematologic toxicity and peripheral neuropathy, respectively. Multiple regression analyses and CART algorithm finally provided a potent efficacy prediction model using 5 gene expression data and robust multiplex toxicity prediction models-CART models using a total of 4 genotype combinations and multiple regression models using 15 polymorphisms on 12 genes. CONCLUSION Biomarkers and multiplex models composed here could work well in the response prediction of ddTCip and/or TC therapy, which might contribute to realize optimal selection of the key therapy.
Collapse
Grants
- DOFMET-08 Development Organization for Frontier Medical Education and Therapeutics in Japan
- H21-3rd Comprehensive 10-year Strategy for Cancer Control-010 Ministry of Health, Labour and Welfare
- University Reform Action Plan "Gunma University Initiative for Advanced Research (GIAR) Ministry of Education, Culture, Sports, Science, and Technology (JP)
- University Reform Action Plan "Gunma University Initiative for Advanced Research (GIAR)" Ministry of Education, Culture, Sports, Science, and Technology (JP)
- Promotion Plan for the Platform of Human Resource Development for Cancer Ministry of Education, Culture, Sports, Science, and Technology (JP)
- the Fostering Health Professionals for Changing Needs of Cancer Ministry of Education, Culture, Sports, Science, and Technology (JP)
- New Paradigms - Establishing Center for Fostering Medical Researchers of the Future Programs Ministry of Education, Culture, Sports, Science, and Technology (JP)
Collapse
Affiliation(s)
- Reika Kawabata-Iwakawa
- Division of Integrated Oncology Research, Gunma University Initiative for Advanced Research, Gunma University, Maebashi, Gunma, 371-8511, Japan
- Research Unit and Immunology and Inflammation, Department of Translational Research, Division of Sohyaku Innovative Research, Tanabe Mitsubishi Pharma, Osaka, Japan
| | - Norihiro Iwasa
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Hidaka, Saitama, 350-1298, Japan
| | - Kenichi Satoh
- Faculty of Data Science, Shiga University, Hikone, Shiga, 522-8522, Japan
| | - Jacques Colinge
- Cancer Bioinformatics and System Biology, Institute of Cancer Research of Montpellier (IRCM), Inserm, University of Montpellier, ICM, 34298, Montpellier, France
| | - Muneaki Shimada
- Department of Gynecology and Obstetrics, Tohoku University Graduate School of Medicine, Sendai, Miyagi, 980-8574, Japan
- Department of Obstetrics and Gynecology, Tottori University School of Medicine, Yonago, Tottori, 683-8504, Japan
| | - Satoshi Takeuchi
- Department of Gynecology, Kobe Tokushukai Hospital, Kobe, Hyogo, 655-0017, Japan
- Department of Obstetrics and Gynecology, Iwate Medical University, Morioka, Iwate, 020-8505, Japan
| | - Hiroyuki Fujiwara
- Department of Obstetrics and Gynecology, Jichi Medical University, Shimotsuke, Tochigi, 329-0498, Japan
| | - Hidetaka Eguchi
- Division of Translational Research, Research Center for Genomic Medicine, Saitama Medical University, Hidaka, Saitama, 350-1241, Japan
- Diagnosis and Therapeutics of Intractable Diseases, Intractable Disease Research Center, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
| | - Tetsuro Oishi
- Department of Obstetrics and Gynecology, Tottori University School of Medicine, Yonago, Tottori, 683-8504, Japan
- Department of Obstetrics and Gynecology, Matsue City Hospital, Matsue, Shimane, 690-8509, Japan
| | - Toru Sugiyama
- Department of Obstetrics and Gynecology, Iwate Medical University, Morioka, Iwate, 020-8505, Japan
- Department of Obstetrics and Gynecology, St. Mary's Hospital, Kurume, Fukuoka, 830-8543, Japan
| | - Mitsuaki Suzuki
- Department of Obstetrics and Gynecology, Tottori University School of Medicine, Yonago, Tottori, 683-8504, Japan
| | - Kosei Hasegawa
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Hidaka, Saitama, 350-1298, Japan
- Project Research Division, Research Center for Genomic Medicine, Saitama Medical University, Hidaka, Saitama, 350-1241, Japan
| | - Keiichi Fujiwara
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Hidaka, Saitama, 350-1298, Japan
- Project Research Division, Research Center for Genomic Medicine, Saitama Medical University, Hidaka, Saitama, 350-1241, Japan
| | - Masahiko Nishiyama
- Division of Integrated Oncology Research, Gunma University Initiative for Advanced Research, Gunma University, Maebashi, Gunma, 371-8511, Japan.
- Division of Translational Research, Research Center for Genomic Medicine, Saitama Medical University, Hidaka, Saitama, 350-1241, Japan.
- Project Research Division, Research Center for Genomic Medicine, Saitama Medical University, Hidaka, Saitama, 350-1241, Japan.
- Laboratory for Analytical Instruments, Education and Research Support Center, Gunma University Graduate School of Medicine, 3-39-22 Showa-Machi, Maebashi, Gunma, 371-8511, Japan.
| |
Collapse
|
8
|
Wang Y, Han J, Yang G, Zheng S, Zhou G, Liu X, Cao X, Li G, Zhang B, Xie Z, Li L, Zhang M, Li X, Chen M, Zhang S. Therapeutic potential of the secreted Kazal-type serine protease inhibitor SPINK4 in colitis. Nat Commun 2024; 15:5874. [PMID: 38997284 PMCID: PMC11245600 DOI: 10.1038/s41467-024-50048-y] [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: 10/16/2023] [Accepted: 06/28/2024] [Indexed: 07/14/2024] Open
Abstract
Mucus injury associated with goblet cell (GC) depletion constitutes an early event in inflammatory bowel disease (IBD). Using single-cell sequencing to detect critical events in mucus dysfunction, we discover that the Kazal-type serine protease inhibitor SPINK4 is dynamically regulated in colitic intestine in parallel with disease activities. Under chemically induced colitic conditions, the grim status in Spink4-conditional knockout mice is successfully rescued by recombinant murine SPINK4. Notably, its therapeutic potential is synergistic with existing TNF-α inhibitor infliximab in colitis treatment. Mechanistically, SPINK4 promotes GC differentiation using a Kazal-like motif to modulate EGFR-Wnt/β-catenin and -Hippo pathways. Microbiota-derived diacylated lipoprotein Pam2CSK4 triggers SPINK4 production. We also show that monitoring SPINK4 in circulation is a reliable noninvasive technique to distinguish IBD patients from healthy controls and assess disease activity. Thus, SPINK4 serves as a serologic biomarker of IBD and has therapeutic potential for colitis via intrinsic EGFR activation in intestinal homeostasis.
Collapse
Affiliation(s)
- Ying Wang
- Division of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P. R. China
| | - Jing Han
- Division of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P. R. China
- Division of Gastroenterology, Guangxi Hospital Division of The First Affiliated Hospital, Sun Yat-sen University, Nanning, P. R. China
| | - Guang Yang
- Department of Minimally Invasive Intervention, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Shuhui Zheng
- Research Center for Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P. R. China
| | - Gaoshi Zhou
- Division of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P. R. China
| | - Xinjuan Liu
- Department of Gastroenterology, Beijing Chaoyang Hospital, Capital Medical University, Chaoyang District, Beijing, P. R. China
| | - Xiaocang Cao
- Department of Hepato-Gastroenterology, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, P. R. China
| | - Guang Li
- Department of Gastroenterology, Beijing Chaoyang Hospital, Capital Medical University, Chaoyang District, Beijing, P. R. China
| | - Bowen Zhang
- College of Life Sciences, Beijing Normal University, Beijing, P. R. China
| | - Zhuo Xie
- Division of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P. R. China
| | - Li Li
- Division of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P. R. China
| | - Mudan Zhang
- Division of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P. R. China
| | - Xiaoling Li
- Division of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P. R. China
| | - Minhu Chen
- Division of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P. R. China
| | - Shenghong Zhang
- Division of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P. R. China.
- Division of Gastroenterology, Guangxi Hospital Division of The First Affiliated Hospital, Sun Yat-sen University, Nanning, P. R. China.
| |
Collapse
|
9
|
Kawka E, Herzog R, Ruciński M, Malińska A, Unterwurzacher M, Sacnun JM, Wagner A, Kowalska K, Jopek K, Kucz-Chrostowska A, Kratochwill K, Witowski J. Effect of cellular senescence on the response of human peritoneal mesothelial cells to TGF-β. Sci Rep 2024; 14:12744. [PMID: 38830931 PMCID: PMC11148043 DOI: 10.1038/s41598-024-63250-1] [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: 02/05/2024] [Accepted: 05/27/2024] [Indexed: 06/05/2024] Open
Abstract
Transforming growth factor β (TGF-β) is implicated in both mesothelial-to-mesenchymal transition (MMT) and cellular senescence of human peritoneal mesothelial cells (HPMCs). We previously showed that senescent HPMCs could spontaneously acquire some phenotypic features of MMT, which in young HPMCs were induced by TGF-β. Here, we used electron microscopy, as well as global gene and protein profiling to assess in detail how exposure to TGF-β impacts on young and senescent HPMCs in vitro. We found that TGF-β induced structural changes consistent with MMT in young, but not in senescent HPMCs. Of all genes and proteins identified reliably in HPMCs across all treatments and states, 4,656 targets represented overlapping genes and proteins. Following exposure to TGF-β, 137 proteins and 46 transcripts were significantly changed in young cells, compared to 225 proteins and only 2 transcripts in senescent cells. Identified differences between young and senescent HPMCs were related predominantly to wound healing, integrin-mediated signalling, production of proteases and extracellular matrix components, and cytoskeleton structure. Thus, the response of senescent HPMCs to TGF-β differs or is less pronounced compared to young cells. As a result, the character and magnitude of the postulated contribution of HPMCs to TGF-β-induced peritoneal remodelling may change with cell senescence.
Collapse
Affiliation(s)
- Edyta Kawka
- Department of Pathophysiology, Poznan University of Medical Sciences, Poznan, Poland.
| | - Rebecca Herzog
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Medical University of Vienna, Vienna, Austria
- Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Marcin Ruciński
- Department of Histology and Embryology, Poznan University of Medical Sciences, Poznan, Poland
| | - Agnieszka Malińska
- Department of Histology and Embryology, Poznan University of Medical Sciences, Poznan, Poland
| | - Markus Unterwurzacher
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Medical University of Vienna, Vienna, Austria
- Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Juan Manuel Sacnun
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Medical University of Vienna, Vienna, Austria
- Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Anja Wagner
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Medical University of Vienna, Vienna, Austria
- Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Katarzyna Kowalska
- Department of Histology and Embryology, Poznan University of Medical Sciences, Poznan, Poland
| | - Karol Jopek
- Department of Histology and Embryology, Poznan University of Medical Sciences, Poznan, Poland
| | | | - Klaus Kratochwill
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Medical University of Vienna, Vienna, Austria
- Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Janusz Witowski
- Department of Pathophysiology, Poznan University of Medical Sciences, Poznan, Poland.
| |
Collapse
|
10
|
Chen D, Shi Z, Gao X, Yang Y, Lei X, Hu Y. SPINK1 is a Potential Diagnostic and Prognostic Biomarker for Sepsis. Infect Drug Resist 2024; 17:875-884. [PMID: 38476769 PMCID: PMC10929552 DOI: 10.2147/idr.s440117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 02/19/2024] [Indexed: 03/14/2024] Open
Abstract
Purpose There are no satisfactory diagnostic biomarkers for sepsis. Accordingly, this study screened biomarkers valuable for sepsis diagnosis and prognosis using data-independent acquisition (DIA) combined with clinical data analysis. Patients and Methods Serine protease inhibitor Kazal-type 1 (SPINK1) is a differentially expressed protein that was screened using DIA and bioinformatics in sepsis patients (n = 22) and healthy controls (n = 10). The plasma SPINK1 levels were detected using an enzyme-linked immunosorbent assay (ELISA) in an expanded population (sepsis patients, n = 52; healthy controls, n = 10). The diagnostic value of SPINK1 in sepsis was evaluated using receiver operating characteristic (ROC) curve analysis based on clinical data. The prognostic value of SPINK1 for sepsis was evaluated using correlation and survival analyses. Results DIA quality control identified 78 differential proteins (72 upregulated and six downregulated), among which SPINK1 was highly expressed in sepsis. The ELISA results suggested that SPINK1 expression was significantly elevated in the sepsis group (P < 0.05). ROC analysis of SPINK1 yielded an area under the curve (AUC) of 0.9096. Combining SPINK1 with procalcitonin (PCT) for ROC analysis yielded an AUC of 1. SPINK1 expression was positively correlated with the Sequential Organ Failure Assessment (SOFA) score (r = 3497, P = 0.0053) and APACHE II score (r = 3223, P = 0.0106). High plasma SPINK1 protein expression was negatively correlated with the 28-day survival rate of patients with sepsis (P = 0.0149). Conclusion The plasma of sepsis patients contained increased SPINK1 protein expression. Combining SPINK1 with PCT might have a high diagnostic value for sepsis. SPINK1 was associated with the SOFA score, APACHE II score, and the 28-day survival rate in patients with sepsis.
Collapse
Affiliation(s)
- Dexiu Chen
- Department of Critical Care Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, People’s Republic of China
| | - Zhangjing Shi
- Department of Critical Care Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, People’s Republic of China
| | - Xiaolan Gao
- Department of Critical Care Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, People’s Republic of China
| | - Yuxiang Yang
- Department of Critical Care Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, People’s Republic of China
| | - Xianying Lei
- Department of Critical Care Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, People’s Republic of China
| | - Yingchun Hu
- Department of Emergency Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, People’s Republic of China
| |
Collapse
|
11
|
Guo Y, Chai B, Zhang H, Chai X, Chen Y, Xu J, Qin L, Chai Y. RARRES1 inhibits hepatocellular carcinoma progression and increases its sensitivity to lenvatinib through interaction with SPINK2. Biol Direct 2024; 19:15. [PMID: 38388961 PMCID: PMC10885466 DOI: 10.1186/s13062-024-00459-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 02/07/2024] [Indexed: 02/24/2024] Open
Abstract
BACKGROUND Lenvatinib is an oral small molecule inhibitor approved for treating patients with unresectable hepatocellular carcinoma (HCC) worldwide. Increasing cell sensitivity to lenvatinib would be an effective method of improving therapeutic efficacy. METHODS High throughput methods was used to scan the differentially expressed genes (DEGs) related to lenvatinib sensitivity in HCC cells. Gain- and loss-function experiments were used to explore the functions of these DEGs in HCC and lenvatinib sensitivity. CO-IP assay and rescue experiments were utilized to investigate the mechanism. RESULTS We identified that RAR responder protein 1 (RARRES1), a podocyte-specific growth arrest gene, was among significantly upregulated DEGs in HCC cells following lenvatinib treatment. Functional analysis showed that ectopic RARRES1 expression decreased HCC progression in vitro and in vivo, as well as improving tumor sensitivity to lenvatinib, while RARRES1 silencing increased HCC cell proliferation and migration. Mechanistically, co-immunoprecipitation assays demonstrated that RARRES1 interacted with serine protease inhibitor Kazal-type 2 (SPINK2) in HCC cells. Further, SPINK2 overexpression suppressed HCC cell proliferation and migration, as well as increasing sensitivity to lenvatinib whereas SPINK2 knockdown promoted cell progression and decreased lenvatinib sensitivity. The mRNA and protein levels of RARRES1 and SPINK2 were low in HCC tissue samples, relative to those in normal liver tissue. CONCLUSIONS Our findings highlighted that RARRES1 can inhibit HCC progression and regulate HCC sensitivity to lenvatinib by interacting SPINK2, representing a new tumor suppressor RARRES1/SPINK2 axis in HCC that modulates sensitivity to lenvatinib.
Collapse
Affiliation(s)
- Yarong Guo
- Department of Digestive System Oncology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, 030032, Taiyuan, Shanxi, China
- Department of Oncology, The First Affiliated Hospital of Shanxi Medical University, 030001, Taiyuan, Shanxi, China
| | - Bao Chai
- Department of Gastroenterology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, 030032, Taiyuan, Shanxi, China
| | - Hezhao Zhang
- Department of Surgery, The First Affiliated Hospital of Shanxi Medical University, 030001, Taiyuan, Shanxi, China
| | - Xinhao Chai
- Department of Oncology, The First Affiliated Hospital of Shanxi Medical University, 030001, Taiyuan, Shanxi, China
| | - Yan Chen
- Department of Oncology, The First Affiliated Hospital of Shanxi Medical University, 030001, Taiyuan, Shanxi, China
| | - Jun Xu
- Department of Surgery, The First Affiliated Hospital of Shanxi Medical University, 030001, Taiyuan, Shanxi, China.
| | - Liwei Qin
- Shanxi Medical University, 030001, Taiyuan, Shanxi, China
| | - Yuting Chai
- Shanxi Medical University, 030001, Taiyuan, Shanxi, China
| |
Collapse
|
12
|
Yang C, Guo L, Du J, Zhang Q, Zhang L. SPINK1 Overexpression Correlates with Hepatocellular Carcinoma Treatment Resistance Revealed by Single Cell RNA-Sequencing and Spatial Transcriptomics. Biomolecules 2024; 14:265. [PMID: 38540686 PMCID: PMC10968071 DOI: 10.3390/biom14030265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 02/09/2024] [Accepted: 02/10/2024] [Indexed: 01/04/2025] Open
Abstract
Low efficacy of treatments and chemoresistance are challenges in addressing refractory hepatocellular carcinoma (HCC). SPINK1, an oncogenic protein, is frequently overexpressed in many HCC cases. However, the impact of SPINK1 on HCC treatment resistance remains poorly understood. Here, we elucidate the functions of SPINK1 on HCC therapy resistance. Analysis of SPINK1 protein level reveals a correlation between elevated SPINK1 expression and unfavorable prognosis. Furthermore, intercellular variations in SPINK1 expression levels are observed. Subsequent examination of single cell RNA-sequencing data from two HCC cohorts further suggest that SPINK1-high cells exhibit heightened activity in drug metabolic pathways compared to SPINK1-low HCC cells. High SPINK1 expression is associated with reduced sensitivities to both chemotherapy drugs and targeted therapies. Moreover, spatial transcriptomics data indicate that elevated SPINK1 expression correlates with non-responsive phenotype during treatment with targeted therapy and immune checkpoint inhibitors. This is attributed to increased levels of drug metabolic regulators, especially CES2 and CYP3A5, in SPINK1-high cells. Experimental evidence further demonstrates that SPINK1 overexpression induces the expression of CES2 and CYP3A5, consequently promoting chemoresistance to sorafenib and oxaliplatin. In summary, our study unveils the predictive role of SPINK1 on HCC treatment resistance, identifying it as a potential therapeutic target for refractory HCC.
Collapse
Affiliation(s)
- Chunyuan Yang
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Peking University Third Hospital, Peking University Health Science Center, Beijing 100191, China; (C.Y.); (J.D.); (Q.Z.)
| | - Limei Guo
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Peking University Third Hospital, Peking University Health Science Center, Beijing 100191, China; (C.Y.); (J.D.); (Q.Z.)
| | - Juan Du
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Peking University Third Hospital, Peking University Health Science Center, Beijing 100191, China; (C.Y.); (J.D.); (Q.Z.)
| | - Qiulu Zhang
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Peking University Third Hospital, Peking University Health Science Center, Beijing 100191, China; (C.Y.); (J.D.); (Q.Z.)
| | - Lingfu Zhang
- Department of General Surgery, Peking University Third Hospital, Beijing 100191, China;
| |
Collapse
|
13
|
Arora R, Cao C, Kumar M, Sinha S, Chanda A, McNeil R, Samuel D, Arora RK, Matthews TW, Chandarana S, Hart R, Dort JC, Biernaskie J, Neri P, Hyrcza MD, Bose P. Spatial transcriptomics reveals distinct and conserved tumor core and edge architectures that predict survival and targeted therapy response. Nat Commun 2023; 14:5029. [PMID: 37596273 PMCID: PMC10439131 DOI: 10.1038/s41467-023-40271-4] [Citation(s) in RCA: 79] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 07/19/2023] [Indexed: 08/20/2023] Open
Abstract
The spatial organization of the tumor microenvironment has a profound impact on biology and therapy response. Here, we perform an integrative single-cell and spatial transcriptomic analysis on HPV-negative oral squamous cell carcinoma (OSCC) to comprehensively characterize malignant cells in tumor core (TC) and leading edge (LE) transcriptional architectures. We show that the TC and LE are characterized by unique transcriptional profiles, neighboring cellular compositions, and ligand-receptor interactions. We demonstrate that the gene expression profile associated with the LE is conserved across different cancers while the TC is tissue specific, highlighting common mechanisms underlying tumor progression and invasion. Additionally, we find our LE gene signature is associated with worse clinical outcomes while TC gene signature is associated with improved prognosis across multiple cancer types. Finally, using an in silico modeling approach, we describe spatially-regulated patterns of cell development in OSCC that are predictably associated with drug response. Our work provides pan-cancer insights into TC and LE biology and interactive spatial atlases ( http://www.pboselab.ca/spatial_OSCC/ ; http://www.pboselab.ca/dynamo_OSCC/ ) that can be foundational for developing novel targeted therapies.
Collapse
Affiliation(s)
- Rohit Arora
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Christian Cao
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Mehul Kumar
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Sarthak Sinha
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Ayan Chanda
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Reid McNeil
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Divya Samuel
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Rahul K Arora
- Center for Health Informatics, University of Calgary, Calgary, AB, Canada
- Institute of Biomedical Engineering, University of Oxford, Oxford, United Kingdom
| | - T Wayne Matthews
- Ohlson Research Initiative, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Section of Otolaryngology Head & Neck Surgery, Department of Surgery, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Shamir Chandarana
- Ohlson Research Initiative, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Section of Otolaryngology Head & Neck Surgery, Department of Surgery, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Robert Hart
- Ohlson Research Initiative, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Section of Otolaryngology Head & Neck Surgery, Department of Surgery, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Joseph C Dort
- Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Ohlson Research Initiative, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Section of Otolaryngology Head & Neck Surgery, Department of Surgery, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Community Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Jeff Biernaskie
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
- Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Surgery, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Paola Neri
- Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Division of Hematology, Department of Oncology, University of Calgary, Calgary, AB, Canada
| | - Martin D Hyrcza
- Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, AB, Canada
| | - Pinaki Bose
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
- Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
- Institute of Biomedical Engineering, University of Oxford, Oxford, United Kingdom.
- Department of Oncology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
14
|
Ravindran F, Jain A, Desai S, Menon N, Srivastava K, Bawa PS, Sateesh K, Srivatsa N, Raghunath SK, Srinivasan S, Choudhary B. Whole-exome sequencing of Indian prostate cancer reveals a novel therapeutic target: POLQ. J Cancer Res Clin Oncol 2023; 149:2451-2462. [PMID: 35737091 DOI: 10.1007/s00432-022-04111-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 06/03/2022] [Indexed: 11/25/2022]
Abstract
PURPOSE Prostate cancer is the second most common cancer diagnosed worldwide and the third most common cancer among men in India. This study's objective was to characterise the mutational landscape of Indian prostate cancer using whole-exome sequencing to identify population-specific polymorphisms. METHODS Whole-exome sequencing was performed of 58 treatment-naive primary prostate tumors of Indian origin. Multiple computational and statistical analyses were used to profile the known common mutations, other deleterious mutations, driver genes, prognostic biomarkers, and gene signatures unique to each clinical parameter. Cox analysis was performed to validate survival-associated genes. McNemar test identified genes significant to recurrence and receiver-operating characteristic (ROC) analysis was conducted to determine its accuracy. OncodriveCLUSTL algorithm was used to deduce driver genes. The druggable target identified was modeled with its known inhibitor using Autodock. RESULTS TP53 was the most commonly mutated gene in our cohort. Three novel deleterious variants unique to the Indian prostate cancer subtype were identified: POLQ, FTHL17, and OR8G1. COX regression analysis identified ACSM5, a mitochondrial gene responsible for survival. CYLC1 gene, which encodes for sperm head cytoskeletal protein, was identified as an unfavorable prognostic biomarker indicative of recurrence. The novel POLQ mutant, also identified as a driver gene, was evaluated as the druggable target in this study. POLQ, a DNA repair enzyme implicated in various cancer types, is overexpressed and is associated with a poor prognosis. The mutant POLQ was subjected to structural analysis and modeled with its known inhibitor novobiocin resulting in decreased binding efficiency necessitating the development of a better drug. CONCLUSION In this pilot study, the molecular profiling using multiple computational and statistical analyses revealed distinct polymorphisms in the Indian prostate cancer cohort. The mutational signatures identified provide a valuable resource for prognostic stratification and targeted treatment strategies for Indian prostate cancer patients. The DNA repair enzyme, POLQ, was identified as the druggable target in this study.
Collapse
Affiliation(s)
- Febina Ravindran
- Institute of Bioinformatics and Applied Biotechnology, Electronic City Phase 1, Bangalore, Karnataka, India
| | - Anika Jain
- Institute of Bioinformatics and Applied Biotechnology, Electronic City Phase 1, Bangalore, Karnataka, India
| | - Sagar Desai
- Institute of Bioinformatics and Applied Biotechnology, Electronic City Phase 1, Bangalore, Karnataka, India
- Manipal Academy of Higher Education, Manipal, India
| | - Navjoth Menon
- Institute of Bioinformatics and Applied Biotechnology, Electronic City Phase 1, Bangalore, Karnataka, India
| | - Kriti Srivastava
- Institute of Bioinformatics and Applied Biotechnology, Electronic City Phase 1, Bangalore, Karnataka, India
| | - Pushpinder Singh Bawa
- Institute of Bioinformatics and Applied Biotechnology, Electronic City Phase 1, Bangalore, Karnataka, India
| | - K Sateesh
- Healthcare Global Enterprises Ltd, Cancer Centre, Bangalore, India
| | - N Srivatsa
- Healthcare Global Enterprises Ltd, Cancer Centre, Bangalore, India
| | - S K Raghunath
- Healthcare Global Enterprises Ltd, Cancer Centre, Bangalore, India
| | - Subhashini Srinivasan
- Institute of Bioinformatics and Applied Biotechnology, Electronic City Phase 1, Bangalore, Karnataka, India
| | - Bibha Choudhary
- Institute of Bioinformatics and Applied Biotechnology, Electronic City Phase 1, Bangalore, Karnataka, India.
| |
Collapse
|