1
|
Paraskar G, Bhattacharya S, Kuttiappan A. The Role of Proteomics and Genomics in the Development of Colorectal Cancer Diagnostic Tools and Potential New Treatments. ACS Pharmacol Transl Sci 2025; 8:1227-1250. [PMID: 40370990 PMCID: PMC12070319 DOI: 10.1021/acsptsci.4c00686] [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: 11/26/2024] [Revised: 03/14/2025] [Accepted: 04/03/2025] [Indexed: 05/16/2025]
Abstract
The complex molecular mechanisms involving genetic and epigenetic modifications contribute to colorectal cancer (CRC), which remains a significant threat to world health. This review elucidates the role of proteomics and genomics in the progression, diagnosis, and treatment of colorectal cancers. All potential key pathways involved in CRC, including WNT, MAPK, PI3K, and TGF-β pathways, are reviewed with a systematic analysis, concluding with their involvement in tumorigenesis and therapeutic resistance. Emerging next-generation sequencing technologies revealed critical mutations that are relevant to CRC development. Proteomics has contributed to identifying biomarkers and post-translational modifications that hold promise for targeted therapies. Recent technological advances have provided functional insights into protein signaling networks and pathways through mass spectrometry and integrated proteogenomic approaches. This work emphasizes biomarker-driven translational efforts that integrate genomic insights with protein expression profiles to refine personalized treatments. The application of innovations in liquid biopsy and computational biology advocates for precision medicine paths to improve the outcomes for CRC. Now, pharmacoproteomics offers novel domains for drug discovery and resistance management and serves as a foundation for comprehensive CRC treatment paradigms.
Collapse
Affiliation(s)
- Gaurav Paraskar
- School of Pharmacy &
Technology Management, SVKM’S NMIMS Deemed-to-be University, Shirpur, Maharashtra 425405, India
| | - Sankha Bhattacharya
- School of Pharmacy &
Technology Management, SVKM’S NMIMS Deemed-to-be University, Shirpur, Maharashtra 425405, India
| | - Anitha Kuttiappan
- School of Pharmacy &
Technology Management, SVKM’S NMIMS Deemed-to-be University, Shirpur, Maharashtra 425405, India
| |
Collapse
|
2
|
Fatfat Z, Hussein M, Fatfat M, Gali-Muhtasib H. Omics technologies as powerful approaches to unravel colorectal cancer complexity and improve its management. Mol Cells 2025; 48:100200. [PMID: 40024318 PMCID: PMC11976254 DOI: 10.1016/j.mocell.2025.100200] [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: 10/25/2024] [Revised: 01/31/2025] [Accepted: 02/22/2025] [Indexed: 03/04/2025] Open
Abstract
Colorectal cancer (CRC) continues to rank among the deadliest and most prevalent cancers worldwide, necessitating an innovative and comprehensive approach that addresses this serious health challenge at various stages, from screening and diagnosis to treatment and prognosis. As CRC research progresses, the adoption of an omics-centered approach holds transformative potential to revolutionize the management of this disease. Advances in omics technologies encompassing genomics, transcriptomics, proteomics, metabolomics, and epigenomics allow to unravel the oncogenic alterations at these levels, elucidating the intricacies and the heterogeneous nature of CRC. By providing a comprehensive molecular landscape of CRC, omics technologies enable the discovery of potential biomarkers for early non-invasive detection of CRC, definition of CRC subtypes, prediction of its staging, prognosis, and overall survival of CRC patients. They also allow the identification of potential therapeutic targets, prediction of drug response, tracking treatment efficacy, detection of residual disease and cancer relapse, and deciphering the mechanisms of drug resistance. Moreover, they allow the distinction of non-metastatic CRC patients from metastatic ones as well as the stratification of metastatic risk. Importantly, omics technologies open up new opportunities to establish molecular-based criteria to guide the selection of effective treatment paving the way for the personalization of therapy for CRC patients. This review consolidates current knowledge on the omics-based preclinical discoveries in CRC research emphasizing the significant potential of these technologies to improve CRC screening, diagnosis, and prognosis and promote the implementation of personalized medicine to ultimately reduce CRC prevalence and mortality.
Collapse
Affiliation(s)
- Zaynab Fatfat
- Department of Biology, American University of Beirut, Beirut, Lebanon
| | - Marwa Hussein
- Department of Biological Sciences, Faculty of Science, Beirut Arab University, Beirut, Lebanon
| | - Maamoun Fatfat
- Department of Pharmacology and Toxicology, American University of Beirut, Beirut, Lebanon
| | | |
Collapse
|
3
|
Xu Y, Yu J. TMC5 as a Marker of Tumor-Associated Telocytes in Hepatocellular Carcinoma. FRONT BIOSCI-LANDMRK 2025; 30:36583. [PMID: 40302342 DOI: 10.31083/fbl36583] [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: 12/25/2024] [Revised: 03/07/2025] [Accepted: 03/19/2025] [Indexed: 05/02/2025]
Abstract
BACKGROUND Tumor-associated telocytes (TATCs) perform a pivotal role in hepatocellular carcinoma (HCC) progression and correlate with poor patient outcomes. This study aims to identify specific markers of TATCs in HCC using single-nucleus RNA sequencing (snRNA-seq) and transcriptomic analyses. METHODS Comprehensive snRNA-seq and transcriptomic profiling were performed on HCC and adjacent non-cancerous tissues to detect differential expressed genes (DEGs) in TATCs. Bioinformatics tools, including STING and Cytoscape software, were employed to analyze protein-protein interactions and hub genes. Immune cell interactions were assessed via ligand-receptor network analysis. RESULT TATCs constituted 0.35% of cells in HCC tissues, with reduced proportions compared to para-cancerous tissues (0.35% vs 8.19%). Hub genes, including TOP2A (DNA topoisomerase Ⅱ alpha), BUB1B (BUB1 mitotic checkpoint serine/threonine kinase B), KIF11 (kinesin family member 11), and CENPF (centromere protein F) were identified in telocytes (TCs). Transcriptomics revealed 622 upregulated and 758 downregulated genes in TATCs versus TCs. TMC5 (transmembrane channel like 5) and SLC35F3 (solute carrier family 35 member F3) emerged as unique TATCs biomarkers, revealing significant associations with poor overall survival (OS) in HCC patients (HR = 1.499 for TMC5; HR = 1.562 for SLC35F3). CONCLUSION TMC5 and SLC35F3 are promising biomarkers for TATCs in HCC, warranting further validation to explore their clinical and therapeutic implications.
Collapse
Affiliation(s)
- Ying Xu
- Department of General Surgery, Shandong Provincial Third Hospital, Shandong University, 250031 Jinan, Shandong, China
| | - Jing Yu
- Department of Pathology, Shandong Provincial Third Hospital, Shandong University, 250031 Jinan, Shandong, China
| |
Collapse
|
4
|
Montero-Calle A, Garranzo-Asensio M, Poves C, Sanz R, Dziakova J, Peláez-García A, de Los Ríos V, Martinez-Useros J, Fernández-Aceñero MJ, Barderas R. In-Depth Proteomic Analysis of Paraffin-Embedded Tissue Samples from Colorectal Cancer Patients Revealed TXNDC17 and SLC8A1 as Key Proteins Associated with the Disease. J Proteome Res 2024; 23:4802-4820. [PMID: 39441737 DOI: 10.1021/acs.jproteome.3c00749] [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: 10/25/2024]
Abstract
A deeper understanding of colorectal cancer (CRC) biology would help to identify specific early diagnostic markers. Here, we conducted quantitative proteomics on FFPE healthy, adenoma, and adenocarcinoma tissue samples from six stage I sporadic CRC patients to identify dysregulated proteins during early CRC development. Two independent quantitative 10-plex TMT experiments were separately performed. After protein extraction, trypsin digestion, and labeling, proteins were identified and quantified by using a Q Exactive mass spectrometer. A total of 2681 proteins were identified and quantified after data analysis and bioinformatics with MaxQuant and the R program. Among them, 284 and 280 proteins showed significant upregulation and downregulation (expression ratio ≥1.5 or ≤0.67, p-value ≤0.05), respectively, in adenoma and/or adenocarcinoma compared to healthy tissue. Ten dysregulated proteins were selected to study their role in CRC by WB, IHC, TMA, and ELISA using tissue and plasma samples from CRC patients, individuals with premalignant colorectal lesions (adenomas), and healthy individuals. In vitro loss-of-function cell-based assays and in vivo experiments using three CRC cell lines with different metastatic properties assessed the important roles of SLC8A1 and TXNDC17 in CRC and liver metastasis. Additionally, SLC8A1 and TXNDC17 protein levels in plasma possessed the diagnostic ability of early CRC stages.
Collapse
Affiliation(s)
- Ana Montero-Calle
- Chronic Disease Programme (UFIEC), Instituto de Salud Carlos III, Majadahonda, E-28220 Madrid, Spain
- Proteomics Core UCCTs, Instituto de Salud Carlos III, Majadahonda, E-28220 Madrid, Spain
| | - María Garranzo-Asensio
- Chronic Disease Programme (UFIEC), Instituto de Salud Carlos III, Majadahonda, E-28220 Madrid, Spain
| | - Carmen Poves
- Gastroenterology Unit, Hospital Universitario Clínico San Carlos, E-28040 Madrid, Spain
| | - Rodrigo Sanz
- Surgical Digestive Department, Hospital Universitario Clínico San Carlos, E-28040 Madrid, Spain
| | - Jana Dziakova
- Surgical Digestive Department, Hospital Universitario Clínico San Carlos, E-28040 Madrid, Spain
| | - Alberto Peláez-García
- Molecular Pathology and Therapeutic Targets Group, Hospital La Paz Institute for Health Research (IdiPAZ), E-28046 Madrid, Spain
| | | | - Javier Martinez-Useros
- Translational Oncology Division, OncoHealth Institute, Health Research Institute-Fundación Jiménez Díaz, Fundación Jiménez Díaz University Hospital/Universidad Autónoma de Madrid (IIS-FJD/UAM), E-28040 Madrid, Spain
- Area of Physiology, Department of Basic Health Sciences, Faculty of Health Sciences, Rey Juan Carlos University, Alcorcón, E-28922 Madrid, Spain
| | | | - Rodrigo Barderas
- Chronic Disease Programme (UFIEC), Instituto de Salud Carlos III, Majadahonda, E-28220 Madrid, Spain
- Proteomics Core UCCTs, Instituto de Salud Carlos III, Majadahonda, E-28220 Madrid, Spain
- CIBER of Frailty and Healthy Aging (CIBERFES), E-28029 Madrid, Spain
| |
Collapse
|
5
|
Altieri B, Secener AK, Sai S, Fischer C, Sbiera S, Arampatzi P, Kircher S, Herterich S, Landwehr L, Vitcetz SN, Braeuning C, Fassnacht M, Ronchi CL, Sauer S. Single-nucleus and spatial transcriptome reveal adrenal homeostasis in normal and tumoural adrenal glands. Clin Transl Med 2024; 14:e1798. [PMID: 39167619 PMCID: PMC11338279 DOI: 10.1002/ctm2.1798] [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: 11/21/2023] [Revised: 07/11/2024] [Accepted: 07/26/2024] [Indexed: 08/23/2024] Open
Abstract
The human adrenal gland is a complex endocrine tissue. Studies on adrenal renewal have been limited to animal models or human foetuses. Enhancing our understanding of adult human adrenal homeostasis is crucial for gaining insights into the pathogenesis of adrenal diseases, such as adrenocortical tumours. Here, we present a comprehensive cellular genomics analysis of the adult human normal adrenal gland, combining single-nuclei RNA sequencing and spatial transcriptome data to reconstruct adrenal gland homeostasis. As expected, we identified primary cells of the various zones of the adrenal cortex and medulla, but we also uncovered additional cell types. They constitute the adrenal microenvironment, including immune cells, mostly composed of a large population of M2 macrophages, and new cell populations, including different subpopulations of vascular-endothelial cells and cortical-neuroendocrine cells. Utilizing spatial transcriptome and pseudotime trajectory analysis, we support evidence of the centripetal dynamics of adrenocortical cell maintenance and the essential role played by Wnt/β-catenin, sonic hedgehog, and fibroblast growth factor pathways in the adult adrenocortical homeostasis. Furthermore, we compared single-nuclei transcriptional profiles obtained from six healthy adrenal glands and twelve adrenocortical adenomas. This analysis unveiled a notable heterogeneity in cell populations within the adenoma samples. In addition, we identified six distinct adenoma-specific clusters, each with varying distributions based on steroid profiles and tumour mutational status. Overall, our results provide novel insights into adrenal homeostasis and molecular mechanisms potentially underlying early adrenocortical tumorigenesis and/or autonomous steroid secretion. Our cell atlas represents a powerful resource to investigate other adrenal-related pathologies.
Collapse
Affiliation(s)
- Barbara Altieri
- Division of Endocrinology and DiabetesDepartment of Internal Medicine IUniversity HospitalUniversity of WürzburgWürzburgGermany
| | - A. Kerim Secener
- Max Delbrück Center for Molecular MedicineBerlinGermany
- Berlin Institute of HealthBerlinGermany
- Department of BiologyChemistry and PharmacyInstitute of BiochemistryFree University BerlinBerlinGermany
| | - Somesh Sai
- Max Delbrück Center for Molecular MedicineBerlinGermany
- Berlin Institute of HealthBerlinGermany
- Department of BiologyChemistry and PharmacyInstitute of BiochemistryFree University BerlinBerlinGermany
| | - Cornelius Fischer
- Max Delbrück Center for Molecular MedicineBerlinGermany
- Berlin Institute of HealthBerlinGermany
| | - Silviu Sbiera
- Division of Endocrinology and DiabetesDepartment of Internal Medicine IUniversity HospitalUniversity of WürzburgWürzburgGermany
| | | | - Stefan Kircher
- Institute of PathologyUniversity of WürzburgWürzburgGermany
| | | | - Laura‐Sophie Landwehr
- Division of Endocrinology and DiabetesDepartment of Internal Medicine IUniversity HospitalUniversity of WürzburgWürzburgGermany
| | - Sarah N. Vitcetz
- Max Delbrück Center for Molecular MedicineBerlinGermany
- Berlin Institute of HealthBerlinGermany
| | | | - Martin Fassnacht
- Division of Endocrinology and DiabetesDepartment of Internal Medicine IUniversity HospitalUniversity of WürzburgWürzburgGermany
- Central Laboratory University Hospital WürzburgWürzburgGermany
| | - Cristina L. Ronchi
- Division of Endocrinology and DiabetesDepartment of Internal Medicine IUniversity HospitalUniversity of WürzburgWürzburgGermany
- Institute of Metabolism and System ResearchUniversity of BirminghamEdgabston, BirminghamUK
| | - Sascha Sauer
- Max Delbrück Center for Molecular MedicineBerlinGermany
- Berlin Institute of HealthBerlinGermany
- Core Unit SysMedUniversity of WürzburgWürzburgGermany
| |
Collapse
|
6
|
Kiran N, Yashaswini C, Maheshwari R, Bhattacharya S, Prajapati BG. Advances in Precision Medicine Approaches for Colorectal Cancer: From Molecular Profiling to Targeted Therapies. ACS Pharmacol Transl Sci 2024; 7:967-990. [PMID: 38633600 PMCID: PMC11019743 DOI: 10.1021/acsptsci.4c00008] [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: 01/10/2024] [Revised: 03/06/2024] [Accepted: 03/07/2024] [Indexed: 04/19/2024]
Abstract
Precision medicine is transforming colorectal cancer treatment through the integration of advanced technologies and biomarkers, enhancing personalized and effective disease management. Identification of key driver mutations and molecular profiling have deepened our comprehension of the genetic alterations in colorectal cancer, facilitating targeted therapy and immunotherapy selection. Biomarkers such as microsatellite instability (MSI) and DNA mismatch repair deficiency (dMMR) guide treatment decisions, opening avenues for immunotherapy. Emerging technologies such as liquid biopsies, artificial intelligence, and machine learning promise to revolutionize early detection, monitoring, and treatment selection in precision medicine. Despite these advancements, ethical and regulatory challenges, including equitable access and data privacy, emphasize the importance of responsible implementation. The dynamic nature of colorectal cancer, with its tumor heterogeneity and clonal evolution, underscores the necessity for adaptive and personalized treatment strategies. The future of precision medicine in colorectal cancer lies in its potential to enhance patient care, clinical outcomes, and our understanding of this intricate disease, marked by ongoing evolution in the field. The current reviews focus on providing in-depth knowledge on the various and diverse approaches utilized for precision medicine against colorectal cancer, at both molecular and biochemical levels.
Collapse
Affiliation(s)
- Neelakanta
Sarvashiva Kiran
- Department
of Biotechnology, School of Applied Sciences, REVA University, Bengaluru, Karnataka 560064, India
| | - Chandrashekar Yashaswini
- Department
of Biotechnology, School of Applied Sciences, REVA University, Bengaluru, Karnataka 560064, India
| | - Rahul Maheshwari
- School
of Pharmacy and Technology Management, SVKM’s
Narsee Monjee Institute of Management Studies (NMIMS) Deemed-to-University, Green Industrial Park, TSIIC,, Jadcherla, Hyderabad 509301, India
| | - Sankha Bhattacharya
- School
of Pharmacy and Technology Management, SVKM’S
NMIMS Deemed-to-be University, Shirpur, Maharashtra 425405, India
| | - Bhupendra G. Prajapati
- Shree.
S. K. Patel College of Pharmaceutical Education and Research, Ganpat University, Kherva, Gujarat 384012, India
| |
Collapse
|
7
|
Bi B, Qiu M, Liu P, Wang Q, Wen Y, Li Y, Li B, Li Y, He Y, Zhao J. Protein post-translational modifications: A key factor in colorectal cancer resistance mechanisms. BIOCHIMICA ET BIOPHYSICA ACTA. GENE REGULATORY MECHANISMS 2023; 1866:194977. [PMID: 37625568 DOI: 10.1016/j.bbagrm.2023.194977] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 07/16/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023]
Abstract
Colorectal cancer (CRC) is one of the leading causes of cancer-related death. Despite advances in treatment, drug resistance remains a critical impediment. Post-translational modifications (PTMs) regulate protein stability, localization, and activity, impacting vital cellular processes. Recent research has highlighted the essential role of PTMs in the development of CRC resistance. This review summarizes recent advancements in understanding PTMs' roles in CRC resistance, focusing on the latest discoveries. We discuss the functional impact of PTMs on signaling pathways and molecules involved in CRC resistance, progress in drug development, and potential therapeutic targets. We also summarize the primary enrichment methods for PTMs. Finally, we discuss current challenges and future directions, including the need for more comprehensive PTM analysis methods and PTM-targeted therapies. This review identifies potential therapeutic interventions for addressing medication resistance in CRC, proposes prospective therapeutic options, and gives an overview of the function of PTMs in CRC resistance.
Collapse
Affiliation(s)
- Bo Bi
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong, China; Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Miaojuan Qiu
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong, China; Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Sun Yat-Sen University, Shenzhen, Guangdong, China; Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Peng Liu
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong, China; Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Qiang Wang
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong, China; Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Yingfei Wen
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - You Li
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Binbin Li
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong, China; Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Sun Yat-Sen University, Shenzhen, Guangdong, China; Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Yongshu Li
- Hubei Normal University, College of Life Sciences Huangshi, Hubei, China.
| | - Yulong He
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong, China; Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Sun Yat-Sen University, Shenzhen, Guangdong, China.
| | - Jing Zhao
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong, China; Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong, China.
| |
Collapse
|
8
|
Xu J, Quan G, Huang W, Jiang J. VSIG2 promotes malignant progression of pancreatic ductal adenocarcinoma by enhancing LAMTOR2-mediated mTOR activation. Cell Commun Signal 2023; 21:223. [PMID: 37626304 PMCID: PMC10463957 DOI: 10.1186/s12964-023-01209-x] [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: 04/05/2023] [Accepted: 06/30/2023] [Indexed: 08/27/2023] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is one of the most intractable malignancies to overcome clinically due to its insidious onset as well as rapid progression. It is urgent to seek new diagnostic markers and therapeutic targets in order to furthest ameliorate the prognosis of patients with PDAC. V-set and immunoglobulin domain containing 2 (VSIG2) belongs to immunoglobulin superfamily (IgSF), which function as coinhibitory molecule to mediate immune evasion of tumors. Nevertheless, the role of VSIG2 in PDAC and related mechanism still keep unclear. METHODS Different expression of VSIG2 in PDAC tissues and cells were detected by bioinformatic analysis, immunohistochemistry, real-time quantitative PCR as well as western blotting. CCK-8, colony formation, Transwell assay, and scratch experiment were utilized to assess proliferation, invasion and migration properties of PDAC cells. The relationship of VSIG2 with late endosomal/lysosomal adaptor, MAPK and MTOR activator 2 (LAMTOR2) and mechanistic target of rapamycin (mTOR) was identified using mass spectrometry, co-immunoprecipitation and immunofluorescence. GO and KEGG enrichment analysis were performed for further pathway verification using western blotting. Additionally, subcutaneous xenograft tumor model and clinical samples analysis were implemented to further elucidate the oncogenic effect of VSIG2 on PDAC in vivo and clinically. RESULTS VSIG2 was highly expressed in PDAC tissues and cells. Overexpression of VSIG2 facilitated the proliferation, invasion and migration abilities of PDAC cells, while VSIG2-inhibition exerted opposite effects. Mechanistically, VSIG2 could simultaneously bind to LAMTOR2 and mTOR, thereby enhancing interaction between two molecules, which resulted in elevated phosphorylation-modificatory activation of mTOR and downstream key molecules. Clinically, up-regulation of VSIG2 was positively associated with advanced stage, overall survival and disease-free survival of PDAC patients. CONCLUSIONS Our study disclosed that VSIG2 was overexpressed in PDAC, which promoted the proliferation, invasion and metastasis. Mechanically, VSIG2 acted as a scaffold to recruit LAMTOR2 and mTOR simultaneously, stabilize the interaction between them, thus enhancing LAMTOR2-mediated mTOR phosphorylated activation. Collectively, VSIG2 could be exploited as a biomarker for diagnosis and prognosis monitor of PDAC in the future, meanwhile, targeting VSIG2 in PDAC management is expected to be a novel strategy. Video Abstract. Video Abstract.
Collapse
Affiliation(s)
- Jichuan Xu
- Department of Hepatobiliary, Pancreatic and Splenic Surgery, Affiliated Dongguan Hospital, Southern Medical University (Dongguan People's Hospital), 78 Wandao Road, Wanjiang Street, Dongguan City, Guangdong Province, 523058, People's Republic of China
| | - Gang Quan
- Department of Hepatobiliary, Pancreatic and Splenic Surgery, Affiliated Dongguan Hospital, Southern Medical University (Dongguan People's Hospital), 78 Wandao Road, Wanjiang Street, Dongguan City, Guangdong Province, 523058, People's Republic of China
- Department of Hepatobiliary Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Wei Huang
- Department of Hepatobiliary, Pancreatic and Splenic Surgery, Affiliated Dongguan Hospital, Southern Medical University (Dongguan People's Hospital), 78 Wandao Road, Wanjiang Street, Dongguan City, Guangdong Province, 523058, People's Republic of China
| | - Jianxin Jiang
- Department of Hepatobiliary, Pancreatic and Splenic Surgery, Affiliated Dongguan Hospital, Southern Medical University (Dongguan People's Hospital), 78 Wandao Road, Wanjiang Street, Dongguan City, Guangdong Province, 523058, People's Republic of China.
| |
Collapse
|