1
|
Vasudevan J, Vijayakumar R, Reales-Calderon JA, Lam MSY, Ow JR, Aw J, Tan D, Tan AT, Bertoletti A, Adriani G, Pavesi A. In vitro integration of a functional vasculature to model endothelial regulation of chemotherapy and T-cell immunotherapy in liver cancer. Biomaterials 2025; 320:123175. [PMID: 40043483 DOI: 10.1016/j.biomaterials.2025.123175] [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: 09/17/2024] [Revised: 01/31/2025] [Accepted: 02/05/2025] [Indexed: 04/06/2025]
Abstract
The complex tumor microenvironment (TME) presents significant challenges to the development of effective therapies against solid tumors, highlighting the need for advanced in vitro models that better recapitulate TME biology. To address this, we developed a vascularized human liver tumor model using a microfluidic platform, designed to test both drug and cell-based therapies. This model mimics critical tumorigenic features such as hypoxia, extracellular matrix (ECM), and perfusable vascular networks. Intravascular administration of Sorafenib demonstrated its ability to disrupt vascular structures significantly, while eliciting heterogeneous responses in two distinct liver tumor cell lines, HepG2 and Hep3b. Furthermore, treatment with engineered T-cells revealed that the tumor vasculature impeded T-cell infiltration into the tumor core but preserved their cytotoxic capacity, albeit with reduced exhaustion levels. Cytokine analysis and spatial profiling of vascularized tumor samples identified proinflammatory factors that may enhance T-cell-mediated antitumor responses. By capturing key TME characteristics, this microfluidic platform provides a powerful tool enabling detailed investigation of tumor-immune and tumor-vascular interactions. Its versatility could serve as a promising bridge between preclinical studies and clinical testing, offering opportunities for developing and optimizing personalized therapeutic strategies for solid tumors.
Collapse
Affiliation(s)
- Jyothsna Vasudevan
- Mechanobiology Institute, National University of Singapore (NUS), 5A Engineering Drive 1, Singapore, 117411, Republic of Singapore
| | - Ragavi Vijayakumar
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), 61 Biopolis Drive, Singapore, 138673, Republic of Singapore
| | - Jose Antonio Reales-Calderon
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), 61 Biopolis Drive, Singapore, 138673, Republic of Singapore
| | - Maxine S Y Lam
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), 61 Biopolis Drive, Singapore, 138673, Republic of Singapore
| | - Jin Rong Ow
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), 61 Biopolis Drive, Singapore, 138673, Republic of Singapore
| | - Joey Aw
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), 61 Biopolis Drive, Singapore, 138673, Republic of Singapore
| | - Damien Tan
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), 61 Biopolis Drive, Singapore, 138673, Republic of Singapore
| | - Anthony Tanoto Tan
- Duke-NUS Medical School, 8 College Road, Singapore, 169857, Republic of Singapore
| | - Antonio Bertoletti
- Duke-NUS Medical School, 8 College Road, Singapore, 169857, Republic of Singapore
| | - Giulia Adriani
- Singapore Immunology Network (SIgN), Agency for Science, Technology, and Research (A∗STAR), 8A Biomedical Grove, Immunos, Singapore, 138648, Republic of Singapore; Department of Biomedical Engineering, National University of Singapore (NUS), 4 Engineering Drive 3, Singapore, 117583, Republic of Singapore
| | - Andrea Pavesi
- Mechanobiology Institute, National University of Singapore (NUS), 5A Engineering Drive 1, Singapore, 117411, Republic of Singapore; Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), 61 Biopolis Drive, Singapore, 138673, Republic of Singapore; Lee Kong Chian School of Medicine (LKCMedicine), Cancer Discovery and Regenerative Medicine Program, Nanyang Technological University, 308232, Republic of Singapore.
| |
Collapse
|
2
|
Meyer HJ, Potratz J, Jechorek D, Schramm KI, Borggrefe J, Surov A. Association between diffusion-weighted imaging and tumor matrix in liver cancer: a cross-sectional study. Transl Cancer Res 2025; 14:1764-1771. [PMID: 40224978 PMCID: PMC11985173 DOI: 10.21037/tcr-24-1516] [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: 08/25/2024] [Accepted: 12/16/2024] [Indexed: 04/15/2025]
Abstract
Background Imaging modalities can reflect the underlying histopathology of tumors. However, the precise interactions between histopathological microstructure and the resulting imaging phenotype remain elusive. Predicting histopathological features, including the extracellular matrix, in a non-invasive manner could improve clinical care of liver tumors. The present study used cross-sectional guided biopsy specimens to utilize accurate spatial biopsy localization to correlate magnetic resonance imaging (MRI) derived the apparent diffusion coefficient (ADC) values with collagen IV expression in various liver cancers. Methods A total of 127 patients (n=68 female; 45.6%) with a mean age of 65.3±12.3 years were included in the analysis. Inclusion criteria were an available cross-sectional biopsy, available biopsy specimens and a pre-interventional MRI with diffusion-weighed imaging (DWI) sequence. The tumors included 45 patients (35.4%) with hepatocellular carcinoma (HCC), 26 patients (20.5%) with cholangiocellular carcinoma and 56 patients (44.1%) with liver metastases of various primary tumors. Prebioptic liver MRI with diffusion-weighted imaging was used to correlate ADC values with collagen IV expression obtained from liver biopsy. The ADC values were measured in a co-registered way with cross-sectional biopsy imaging to ensure the spatial concordance between imaging and histopathology. The stained area and signal intensity of the immunohistochemical staining were examined. Results The mean average stained area of collagen IV was 32.6%±27.4% and the mean staining intensity was 2.03±1.01. HCC showed statistically less stained area compared to the other tumor types (analysis of variance P<0.0001). In the overall patient sample, there was no correlation between ADCmean and average stained area (r=0.05, P=0.55) and staining intensity (r=-0.04, P=0.60). In a subgroup analysis of HCC patients, there was a significant correlation between ADCmin and the staining intensity (r=-0.33, P=0.02). Conclusions ADC values are not associated with collagen IV expression in liver tumors. The complex extracellular matrix is not reflected by the DWI signal, which can be discussed as mainly be influenced by the cellularity of the tumors. Further research is needed to investigate the complex interactions between histopathology and the resulting imaging phenotype of MRI for clinical care.
Collapse
Affiliation(s)
- Hans-Jonas Meyer
- Department of Diagnostic and Interventional Radiology, University Hospital Leipzig, Leipzig, Germany
| | - Johann Potratz
- Department of Pathology, Otto von Guericke University, Magdeburg, Magdeburg, Germany
| | - Dörthe Jechorek
- Department of Pathology, Otto von Guericke University, Magdeburg, Magdeburg, Germany
| | - Kai Ina Schramm
- Department of Radiology and Nuclear Medicine, Otto von Guericke University, Magdeburg, Germany
| | - Jan Borggrefe
- Department of Radiology, Mühlenkreiskliniken Minden, Ruhr-University Bochum, Bochum, Germany
| | - Alexey Surov
- Department of Radiology, Mühlenkreiskliniken Minden, Ruhr-University Bochum, Bochum, Germany
| |
Collapse
|
3
|
Li Y, Wu R, Wang H, Zhong M, Qing Y, Lu S, Zhang Z, Ma T, Luo J, Xiao H, Qiu J, Li K. Non-epithelial Circulating Tumor Cells Enhance Disease Progression in High-risk Prostate Cancer through EMT and COL1A1 Expression. Int J Med Sci 2025; 22:1562-1573. [PMID: 40093812 PMCID: PMC11905264 DOI: 10.7150/ijms.107703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 02/06/2025] [Indexed: 03/19/2025] Open
Abstract
Introduction: Circulating tumor cells (CTCs) are important prognostic indicators for malignancies. However, a reliable positive/negative cutoff value of non-epithelial (NE+: hybrid and mesenchymal) CTCs phenotype in prostate cancer (PCa) patients has not been established. Here, we aimed to determine the cutoff value and the prognostic value of NE+ CTCs in high-risk prostate cancer (HRPC) patients after radical prostatectomy (RP). Methods: The cutoff value of NE+ CTCs was established in spiking experiments, and CTCs were detected in 208 HRPC patients using the CanPatrolTM platform. The expression and function of COL1A1 in PCa were examined via qRT-PCR, Western blot, wound healing assay, Transwell assay, and immunohistochemistry (IHC). Results: The cutoff value of NE+ CTCs was determined to be 45% by spiking experiments. In 208 HRPC patients, the NE+ CTCs positive group had higher prostate-specific antigen (PSA) levels, more advanced pathological tumor stage, and lymph node stage (P < 0.001, P = 0.002 and 0.002, respectively). Besides, patients with NE+ CTCs ≥ 45% had a shorter median progression-free survival (PFS) than those with NE+ CTCs < 45% (44.5 vs. 51.0 months, hazard ratio = 3.31, P < 0.05). Moreover, we identified that COL1A1 was associated with a high proportion of NE+ CTCs in HRPC patients via an EMT mechanism. Conclusion: Our findings suggest that NE+ CTCs represent a reliable prognostic indicator for HRPC patients and that targeting COL1A1 may prevent the formation of NE+ CTCs.
Collapse
Affiliation(s)
- Yiyuan Li
- Department of Urology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ruji Wu
- Department of Urology, SSL Central Hospital of Dongguan City, Dongguan, China
| | - Hua Wang
- Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Meinong Zhong
- Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yunhao Qing
- Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shuo Lu
- Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zixiao Zhang
- Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Tan Ma
- Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jieheng Luo
- Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hengjun Xiao
- Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jianguang Qiu
- Department of Urology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ke Li
- Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
4
|
Zhang J, Wang Y, Liu Y, Chen H, Chai N, Zhao Y, Li Y, Li X, He J, Zhang H. Proteomic profiling reveals biological processes and biomarkers involved in the pathogenesis of occult breast cancer. BMC Cancer 2025; 25:231. [PMID: 39930421 PMCID: PMC11812265 DOI: 10.1186/s12885-025-13657-4] [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: 10/13/2024] [Accepted: 02/05/2025] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND Occult breast cancer (OBC) is defined as axillary lymph node metastasis without any evidence of a primary tumor in the breast. Because of the limited number of clinical cases, the clinicopathological features and treatment recommendations of OBC are still controversial. In addition, its natural history is poorly understood and its proteomic signature remains unknown. MATERIALS AND METHODS We compared the clinicopathological features and prognosis of OBC patients and Non-OBC patients from the Surveillance, Epidemiology, and End Results (SEER) database and analyzed the effects of local treatment on the survival outcomes of OBC patients. Additionally, we performed a quantitative proteomic analysis for tissue samples of metastatic lymph nodes from OBC patients (OBC-LN), and paired tissue samples of metastatic lymph nodes (Non-OBC-LN) and primary tumors (Non-OBC-PT) from Non-OBC patients. We identified differentially expressed proteins in different comparable groups using Student's t test. Functional enrichment and protein-protein interaction network analyses were used to interpret the functions and interactions of the differentially expressed proteins in the comparison of OBC-LN vs Non-OBC-LN. Immunohistochemistry was used for the validation of the hub proteins. RESULTS Analysis of data from the SEER database demonstrated that OBC patients had a better prognosis than Non-OBC patients did and that either mastectomy or radiation therapy improved the outcomes of OBC patients. A total of 7208 comparable proteins were successfully quantified. Compared with those of the Non-OBC-LN samples, the OBC-LN protein profiles exhibited an active extracellular matrix and a thoroughly upregulated epithelial-mesenchymal transition phenotype. COL1A1, COL1A2, COL3A1, MMP2 and LUM were overexpressed in the OBC-LN samples and were identified as hub proteins. Immunohistochemical staining demonstrated that the five hub proteins were overexpressed in OBC-LN samples. CONCLUSION Our results provide insights for the clinical management of OBC and the proteomic signature of OBC offers molecular basis for further biological research.
Collapse
Affiliation(s)
- Jingyi Zhang
- Department of Breast Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, People's Republic of China
| | - Yidi Wang
- Department of Breast Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, People's Republic of China
| | - Yu Liu
- Department of Pathology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, People's Republic of China
| | - He Chen
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Na Chai
- Department of Breast Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, People's Republic of China
| | - Yuting Zhao
- Department of Gynecologic Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yijun Li
- Department of Breast Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, People's Republic of China
| | - Xiaoqian Li
- Department of Breast Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, People's Republic of China
| | - Jianjun He
- Department of Breast Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, People's Republic of China.
| | - Huimin Zhang
- Department of Breast Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, People's Republic of China.
| |
Collapse
|
5
|
Di H, Rong Z, Mao N, Li H, Chen J, Liu R, Wang A. Transcriptomic landscape of Hras12V oncogene-induced hepatocarcinogenesis with gender disparity. BMC Cancer 2025; 25:94. [PMID: 39819515 PMCID: PMC11737189 DOI: 10.1186/s12885-025-13476-7] [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: 08/11/2024] [Accepted: 01/08/2025] [Indexed: 01/19/2025] Open
Abstract
The genesis of hepatocellular carcinoma (HCC) is closely related to male factors and hyper-activated Ras signals. A transcriptomic database was established via RNA-Seq of HCC (T) and the adjacent precancerous liver tissue (P) of Hras12V transgenic mice (Ras-Tg, HCC model) and the normal liver tissue of wild-type mice (W) of both sexes. Comparative analysis within W, P, and T and correlation expression pattern analysis revealed common/unique cluster-enriched items towards HCC between the sexes. Specifically, the numbers of differentially expressed genes (DEGs) were much higher in females than in males, and tumor suppressor genes, such as p21Waf1/Cip1 and C6, were significantly higher in the female P. This finding denotes the higher sensitivity of female hepatocytes to the Ras oncogene and, therefore, the difficulty in developing HCC. Moreover, convergence in HCC between the sexes suggests the underlying mechanisms for the ineffectiveness of sex hormone therapies. Additionally, expression pattern analysis revealed that the DEGs and their relevant pathways were either positively or negatively associated with the HCC/Ras oncogene. Among them, the vital role of glutathione metabolism in HCC was established. This work provides a basis for future research on elucidating the underlying mechanisms, selecting the diagnostic biomarker, and planning the clinical therapy in HCC.
Collapse
Affiliation(s)
- Huaiyuan Di
- Department of Comparative Medicine, Laboratory Animal Center, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Zhuona Rong
- Department of Comparative Medicine, Laboratory Animal Center, Dalian Medical University, Dalian, Liaoning, 116044, China
- Department of Pharmacy, Peking University First Hospital, Xishiku Street, Xicheng District, Beijing, 100034, China
| | - Nan Mao
- Department of Comparative Medicine, Laboratory Animal Center, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Huiling Li
- Department of Comparative Medicine, Laboratory Animal Center, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Jun Chen
- Department of Comparative Medicine, Laboratory Animal Center, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Renwu Liu
- Central Hospital of Dalian, University of Technology, Dalian, Liaoning, 116044, China.
| | - Aiguo Wang
- Department of Comparative Medicine, Laboratory Animal Center, Dalian Medical University, Dalian, Liaoning, 116044, China.
| |
Collapse
|
6
|
Tang Z, Wei C, Deng X, Lin Q, Hu Q, Li S, Wang J, Wu Y, Liu D, Fang M, Zhan T. Serum proteomic and metabolomic profiling of hepatocellular carcinoma patients co-infected with Clonorchis sinensis. Front Immunol 2025; 15:1489077. [PMID: 39840062 PMCID: PMC11746118 DOI: 10.3389/fimmu.2024.1489077] [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: 08/31/2024] [Accepted: 12/10/2024] [Indexed: 01/23/2025] Open
Abstract
Background Clonorchis sinensis (C. sinensis) infection is a significant risk factor for hepatocellular carcinoma (HCC), yet its underlying mechanisms remain poorly understood. This study aimed to investigate the impact of C. sinensis infection on the serum proteomic and metabolomic profiling of HCC patients, focusing on the potential mechanisms. Method A retrospective clinical analysis was conducted on 1121 HCC patients, comparing those with and without C. sinensis infection. The influence of C. sinensis on serum proteome and metabolome in HCC was further assessed. Result C. sinensis infection correlated with a younger age at cancer onset, male predominance, advanced cancer stage, liver cirrhosis, and microvascular invasion in HCC patients. It also associated with shorter overall survival (OS) and recurrence-free survival (RFS). The levels of blood lipids (e.g., APO-A, HDL-C, and TG) were significantly altered after C. sinensis infection. Proteomic and metabolomic analyses revealed metabolic reprogramming caused by C. sinensis, with excessive depletion of argininosuccinate synthase (ASS) and D-glucose as potential factors in C. sinensis-associated HCC malignancy. Key molecules ILF2, CNN2, OLFM4, NOTCH3, and LysoPA were implicated in HCC progression. Furthermore, C. sinensis triggered inflammation, insulin resistance, and pro-tumor immune escape, and exacerbated the complication of degenerative diseases. Conclusion This study not only provides compelling evidence for elucidating the mechanisms underlying C. sinensis-mediated HCC development but also identifies potential therapeutic targets for HCC patients co-infected with C. sinensis.
Collapse
Affiliation(s)
- Zeli Tang
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Guangxi Medical University, Nanning, China
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, China
- Key Laboratory of Basic Research on Regional Diseases, Education Department of Guangxi Zhuang Autonomous Region, Guangxi Medical University, Nanning, China
| | - Caibiao Wei
- Department of Clinical Laboratory, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Xueling Deng
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Guangxi Medical University, Nanning, China
| | - Qiumei Lin
- Department of Clinical Laboratory, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Qiping Hu
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Guangxi Medical University, Nanning, China
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, China
- Key Laboratory of Basic Research on Regional Diseases, Education Department of Guangxi Zhuang Autonomous Region, Guangxi Medical University, Nanning, China
| | - Shitao Li
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Guangxi Medical University, Nanning, China
| | - Jilong Wang
- Department of Parasitology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, China
| | - Yuhong Wu
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Guangxi Medical University, Nanning, China
| | - Dengyu Liu
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, China
- Key Laboratory of Basic Research on Regional Diseases, Education Department of Guangxi Zhuang Autonomous Region, Guangxi Medical University, Nanning, China
- Department of Parasitology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, China
| | - Min Fang
- Department of Clinical Laboratory, Guangxi Medical University Cancer Hospital, Nanning, China
- Engineering Research Center for Tissue & Organ Injury and Repair Medicine, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Tingzheng Zhan
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, China
- Key Laboratory of Basic Research on Regional Diseases, Education Department of Guangxi Zhuang Autonomous Region, Guangxi Medical University, Nanning, China
- Department of Parasitology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, China
| |
Collapse
|
7
|
Lv R, Yao Y, Dong J, Chen Q. COL1A1, mediated by m6A methylation of METTL3, facilitates oral squamous cell carcinoma cell growth and metastasis. Odontology 2025; 113:191-200. [PMID: 38900231 DOI: 10.1007/s10266-024-00962-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 06/04/2024] [Indexed: 06/21/2024]
Abstract
Collagen type I alpha1 (COL1A1) has been found to be abnormal expressed in oral squamous cell carcinoma (OSCC) tissues, but its role and mechanism in OSCC need to be further elucidated. The expression levels of COL1A1 and methyltransferase-like 3 (METTL3) were measured by quantitative real-time PCR and western blot. Cell growth and metastasis were determined by CCK8, colony formation, EdU, flow cytometry and transwell assays. MeRIP, Co-IP and dual-luciferase reporter assays were performed to explore the interplay of COL1A1 and METTL3. COL1A1 mRNA stability was confirmed by Actinomycin D assay. Mice xenograft models were constructed to perform in vivo experiments. COL1A1 and METTL3 were upregulated in OSCC. COL1A1 knockdown suppressed OSCC cell growth and metastasis, while its overexpression had an opposite effect. The stability of COL1A1 mRNA was regulated by the m6A methylation of METTL3. METTL3 overexpression promoted OSCC cell growth and metastasis, and its knockdown-mediated OSCC cell function inhibition could be abolished by COL1A1 overexpression. Besides, silencing of METTL3 reduced OSCC tumor growth by reducing COL1A1 expression. METTL3-stabilized COL1A1 promoted OSCC progression, providing an exact molecular target for the treatment of OSCC.
Collapse
Affiliation(s)
- Ruya Lv
- Department of Stomatology, Jingzhou Central Hospital, No. 6 Jingzhong Road, Jingzhou District, Jingzhou, 434000, Hubei, China.
- Department of Stomatology, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, 434000, Hubei, China.
| | - Yao Yao
- Department of Stomatology, Jingzhou Central Hospital, No. 6 Jingzhong Road, Jingzhou District, Jingzhou, 434000, Hubei, China
- Department of Stomatology, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, 434000, Hubei, China
| | - Jingjing Dong
- Department of Stomatology, Jingzhou Central Hospital, No. 6 Jingzhong Road, Jingzhou District, Jingzhou, 434000, Hubei, China
- Department of Stomatology, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, 434000, Hubei, China
| | - Qian Chen
- Department of Stomatology, Jingzhou Central Hospital, No. 6 Jingzhong Road, Jingzhou District, Jingzhou, 434000, Hubei, China
- Department of Stomatology, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, 434000, Hubei, China
| |
Collapse
|
8
|
Gurgul A, Żurowski J, Szmatoła T, Kucharski M, Sawicki S, Semik-Gurgul E, Ocłoń E. Cannabidiol (CBD) modulates the transcriptional profile of ethanol-exposed human dermal fibroblast cells. J Appl Genet 2024; 65:773-796. [PMID: 39466591 PMCID: PMC11561130 DOI: 10.1007/s13353-024-00915-7] [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: 07/04/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 10/30/2024]
Abstract
Cannabidiol (CBD) is abundant in the Cannabis sativa plant and exhibits complex immunomodulatory, anxiolytic, antioxidant, and antiepileptic properties. Several studies suggest that CBD could be used for different purposes in alcohol use disorder (AUD) and alcohol-related injuries to the brain and the liver. In this study, we focused on analyzing transcriptional alterations in human dermal fibroblasts (HDFs) cell line challenged simultaneously with ethanol and CBD as an ethanol-protective agent. We aimed to expose the genes and pathways responsible for at least some of the CBD effects in those cells that can be related to the AUD. Transcriptome analysis was performed using HDFs cell line that expresses both cannabinoid receptors and can metabolize ethanol through alcohol dehydrogenase activity. Fibroblasts are also responsible for the progression of liver fibrosis, a common comorbidity in AUD. With the use of a cellular test, we found that CBD at the lowest applied concentration (0.75 μM) was able to stimulate depressed metabolism and reduce the level of apoptosis of cells treated with different concentrations of ethanol to the level observed in the control cells. Similar observations were made at the transcriptome level, in which cells treated with ethanol and CBD had similar expression profiles to the control cells. CBD also affects several genes connected with extracellular matrix formation (especially its collagen constituent), which can have potential implications for, e.g., fibrosis process.
Collapse
Affiliation(s)
- Artur Gurgul
- Faculty of Veterinary Medicine, Department of Basic Sciences, University of Agriculture in Kraków, Redzina 1C, 30-248, Krakow, Poland.
| | - Jakub Żurowski
- Faculty of Veterinary Medicine, Department of Basic Sciences, University of Agriculture in Kraków, Redzina 1C, 30-248, Krakow, Poland
| | - Tomasz Szmatoła
- Faculty of Veterinary Medicine, Department of Basic Sciences, University of Agriculture in Kraków, Redzina 1C, 30-248, Krakow, Poland
| | - Mirosław Kucharski
- Faculty of Animal Science, Department of Animal Physiology and Endocrinology, University of Agriculture in Kraków, Mickiewicza 24/28, 30‑059, Krakow, Poland
| | - Sebastian Sawicki
- Faculty of Animal Science, Department of Animal Reproduction, Anatomy and Genomics, University of Agriculture in Kraków, Mickiewicza 24/28, 30-059, Krakow, Poland
| | - Ewelina Semik-Gurgul
- Department of Animal Molecular Biology, National Research Institute of Animal Production, Krakowska 1, 32-083, Balice, Poland
| | - Ewa Ocłoń
- Faculty of Veterinary Medicine, Laboratory of Recombinant Proteins Production, University of Agriculture in Kraków, Rędzina 1C, 30-248, Kraków, Poland
| |
Collapse
|
9
|
Revert-Ros F, Ventura I, Prieto-Ruiz JA, Giner-Moreno E, Pérez-Cervera Á, Pérez-Rojas J, Revert F, Hernández-Andreu JM. Polyribonucleotide phosphorylase is overexpressed in hepatocellular cancer, promoting epithelial phenotype maintenance and tumor progression. Pathol Res Pract 2024; 264:155713. [PMID: 39522317 DOI: 10.1016/j.prp.2024.155713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 11/04/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Liver cancer, particularly hepatocellular carcinoma (HCC), is a major global health challenge, largely associated with cirrhosis caused by various factors. Prognosis is often guided by molecular and histological classifications. In this study, expression of Polyribonucleotide Phosphorylase (PNPT1) in HCC was investigated to better understand its role in tumor behavior and patient outcomes. The expression of the corresponding protein PNPase was assessed in HCC tissue samples using immunohistochemistry, while RNA-seq data from The Cancer Genome Atlas (TCGA) were analyzed via OncoDB. Additionally, PNPT1 silencing in HepG2 cells was followed by gene and protein expression analysis. The results revealed that PNPT1 is overexpressed in HCC tumors, particularly in those expressing E-cadherin. Notably, silencing PNPT1 in HepG2 cells triggered a shift towards a mesenchymal phenotype. In HCC tissues, PNPT1 expression was linked to markers of epithelial phenotype, oxidative stress, and poor prognosis, especially in non-viral HCC cases. These findings suggest that PNPase may play a crucial role in the progression of well-differentiated HCC tumors, serving as a poor prognostic biomarker.
Collapse
Affiliation(s)
- Francisco Revert-Ros
- Grupo de Medicina Molecular y Mitocondrial, Facultad de Medicina y Ciencias de la Salud, Universidad Católica de Valencia San Vicente Mártir, C/Quevedo 2, Valencia 46001, Spain
| | - Ignacio Ventura
- Grupo de Medicina Molecular y Mitocondrial, Facultad de Medicina y Ciencias de la Salud, Universidad Católica de Valencia San Vicente Mártir, C/Quevedo 2, Valencia 46001, Spain
| | - Jesús A Prieto-Ruiz
- Grupo de Medicina Molecular y Mitocondrial, Facultad de Medicina y Ciencias de la Salud, Universidad Católica de Valencia San Vicente Mártir, C/Quevedo 2, Valencia 46001, Spain
| | - Eduardo Giner-Moreno
- Department of Pathology, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Ángela Pérez-Cervera
- Grupo de Medicina Molecular y Mitocondrial, Facultad de Medicina y Ciencias de la Salud, Universidad Católica de Valencia San Vicente Mártir, C/Quevedo 2, Valencia 46001, Spain
| | - Judith Pérez-Rojas
- Department of Pathology, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Fernando Revert
- Grupo de Medicina Molecular y Mitocondrial, Facultad de Medicina y Ciencias de la Salud, Universidad Católica de Valencia San Vicente Mártir, C/Quevedo 2, Valencia 46001, Spain.
| | - José Miguel Hernández-Andreu
- Grupo de Medicina Molecular y Mitocondrial, Facultad de Medicina y Ciencias de la Salud, Universidad Católica de Valencia San Vicente Mártir, C/Quevedo 2, Valencia 46001, Spain
| |
Collapse
|
10
|
Tang L, Guo D, Jia D, Piao S, Fang C, Zhu Y, Wang Y, Pan Z. Exploring the therapeutic potential of "Tianyu" medicine pair in rheumatoid arthritis: an integrated study combining LC-MS/MS, bioinformatics, network pharmacology, and experimental validation. Front Med (Lausanne) 2024; 11:1475239. [PMID: 39430588 PMCID: PMC11488520 DOI: 10.3389/fmed.2024.1475239] [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: 08/03/2024] [Accepted: 09/23/2024] [Indexed: 10/22/2024] Open
Abstract
Background Rheumatoid arthritis (RA) is a widespread chronic autoimmune disease that primarily causes joint inflammation and damage. In advanced stages, RA can result in joint deformities and loss of function, severely impacting patients' quality of life. The "Tianyu" pair (TYP) is a traditional Chinese medicine formulation developed from clinical experience and has shown some effectiveness in treating RA. However, its role in the complex biological mechanisms underlying RA remains unclear and warrants further investigation. Methods We obtained gene sequencing data of synovial tissues from both RA patients and healthy individuals using two gene microarrays, GSE77298 and GSE55235, from the GEO database. Through an integrated approach involving bioinformatics, machine learning, and network pharmacology, we identified the core molecular targets of the "Tianyu" medicine pair (TYP) for RA treatment. Liquid chromatography-mass spectrometry was then employed to analyze the chemical components of TYP. To validate our findings, we conducted animal experiments with Wistar rats, comparing histopathological and key gene expression changes before and after TYP treatment. Results Our data analysis suggests that the onset of RA may be associated with inflammation-related immune cells involved in both adaptive and innate immune responses. Potential key targets for TYP treatment in RA include AKR1B10, MMP13, FABP4, NCF1, SPP1, COL1A1, and RASGRP1. Among the components of TYP, Kaempferol, Quercetin, and Salidroside were identified as key, with MMP13 and NCF1 showing the strongest binding affinity to these compounds. Animal experiments confirmed the findings from bioinformatics and network pharmacology, validating the key targets and therapeutic effects of TYP in treating RA. Conclusion Our study reveals that TYP has potential clinical value in the treatment of rheumatoid arthritis. This research enhances our understanding of RA's pathogenesis and provides insight into potential therapeutic mechanisms.
Collapse
Affiliation(s)
- Lu Tang
- Fangzheng Research Laboratory, Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, China
| | - Dingyuan Guo
- Department of Traditional Chinese Internal Medicine, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Dongye Jia
- Fangzheng Research Laboratory, Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, China
| | - Songlan Piao
- Department of Pathology Teaching and Research, Clinical Medical School, Changchun University of Chinese Medicine, Changchun, China
| | - Chunqiu Fang
- Fangzheng Research Laboratory, Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, China
| | - Yueya Zhu
- Fangzheng Research Laboratory, Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, China
| | - Yinghang Wang
- Department of Rheumatology and Immunology, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Zhi Pan
- Fangzheng Research Laboratory, Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
11
|
Nouri K, Piryaei A, Seydi H, Zarkesh I, Ghoytasi I, Shokouhian B, Najimi M, Vosough M. Fibrotic liver extracellular matrix induces cancerous phenotype in biomimetic micro-tissues of hepatocellular carcinoma model. Hepatobiliary Pancreat Dis Int 2024:S1499-3872(24)00115-2. [PMID: 39289044 DOI: 10.1016/j.hbpd.2024.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 07/25/2024] [Indexed: 09/19/2024]
Abstract
BACKGROUND Despite considerable advancements in identifying factors contributing to the development of hepatocellular carcinoma (HCC), the pathogenesis of HCC remains unclear. In many cases, HCC is a consequence of prolonged liver fibrosis, resulting in the formation of an intricate premalignant microenvironment. The accumulation of extracellular matrix (ECM) is a hallmark of premalignant microenvironment. Given the critical role of different matrix components in regulating cell phenotype and function, this study aimed to elucidate the interplay between the fibrotic matrix and malignant features in HCC. METHODS Liver tissues from both control (normal) and carbon tetrachloride (CCl4)-induced fibrotic rats were decellularized using sodium dodecyl sulfate (SDS) and Triton X-100. The resulting hydrogel from decellularized ECM was processed into micro-particles via the water-in-oil emulsion method. Micro-particles were subsequently incorporated into three-dimensional liver biomimetic micro-tissues (MTs) comprising Huh-7 cells, human umbilical vein endothelial cells (HUVECs), and LX-2 cells. The MTs were evaluated using the 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium (MTS) assay at day 11, immunofluorescence staining, immunoblotting, and spheroid migration assay at day 14 after co-culture. RESULTS Fibrotic matrix from CCl4-treated rat livers significantly enhanced the growth rate of the MTs and their expression of CCND1 as compared to the normal one. Fibrotic matrix, also induced the expression of epithelial-to-mesenchymal transition (EMT)-associated genes such as TWIST1, ACTA2, MMP9, CDH2, and VIMENTIN in the MTs as compared to the normal matrix. Conversely, the expression of CDH1 and hepatic maturation genes HNF4A, ALB, CYP3A4 was decreased in the MTs when the fibrotic matrix was used. Furthermore, the fibrotic matrix increased the migration of the MTs and their secretion of alpha-fetoprotein. CONCLUSIONS Our findings suggest a regulatory role for the fibrotic matrix in promoting cancerous phenotype, which could potentially accelerate the progression of malignancy in the liver.
Collapse
Affiliation(s)
- Kosar Nouri
- Department of Developmental Biology, University of Science and Culture, ACECR 14155-4364 Tehran, Iran; Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR 14155-4364 Tehran, Iran; Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR 14155-4364 Tehran, Iran
| | - Abbas Piryaei
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Homeyra Seydi
- Department of Developmental Biology, University of Science and Culture, ACECR 14155-4364 Tehran, Iran; Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR 14155-4364 Tehran, Iran; Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR 14155-4364 Tehran, Iran
| | - Ibrahim Zarkesh
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR 14155-4364 Tehran, Iran
| | - Ibrahim Ghoytasi
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR 14155-4364 Tehran, Iran
| | - Bahare Shokouhian
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR 14155-4364 Tehran, Iran; Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mustapha Najimi
- Laboratory of Pediatric Hepatology and Cell Therapy, Institute of Experimental and Clinical Research (IREC), UCLouvain, Brussels, Belgium
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR 14155-4364 Tehran, Iran; Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, Stockholm, Sweden.
| |
Collapse
|
12
|
Macdonald JK, Taylor HB, Wang M, Delacourt A, Edge C, Lewin DN, Kubota N, Fujiwara N, Rasha F, Marquez CA, Ono A, Oka S, Chayama K, Lewis S, Taouli B, Schwartz M, Fiel MI, Drake RR, Hoshida Y, Mehta AS, Angel PM. The Spatial Extracellular Proteomic Tumor Microenvironment Distinguishes Molecular Subtypes of Hepatocellular Carcinoma. J Proteome Res 2024; 23:3791-3805. [PMID: 38980715 PMCID: PMC11385377 DOI: 10.1021/acs.jproteome.4c00099] [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: 02/12/2024] [Revised: 05/31/2024] [Accepted: 06/15/2024] [Indexed: 07/10/2024]
Abstract
Hepatocellular carcinoma (HCC) mortality rates continue to increase faster than those of other cancer types due to high heterogeneity, which limits diagnosis and treatment. Pathological and molecular subtyping have identified that HCC tumors with poor outcomes are characterized by intratumoral collagenous accumulation. However, the translational and post-translational regulation of tumor collagen, which is critical to the outcome, remains largely unknown. Here, we investigate the spatial extracellular proteome to understand the differences associated with HCC tumors defined by Hoshida transcriptomic subtypes of poor outcome (Subtype 1; S1; n = 12) and better outcome (Subtype 3; S3; n = 24) that show differential stroma-regulated pathways. Collagen-targeted mass spectrometry imaging (MSI) with the same-tissue reference libraries, built from untargeted and targeted LC-MS/MS was used to spatially define the extracellular microenvironment from clinically-characterized, formalin-fixed, paraffin-embedded tissue sections. Collagen α-1(I) chain domains for discoidin-domain receptor and integrin binding showed distinctive spatial distribution within the tumor microenvironment. Hydroxylated proline (HYP)-containing peptides from the triple helical regions of fibrillar collagens distinguished S1 from S3 tumors. Exploratory machine learning on multiple peptides extracted from the tumor regions could distinguish S1 and S3 tumors (with an area under the receiver operating curve of ≥0.98; 95% confidence intervals between 0.976 and 1.00; and accuracies above 94%). An overall finding was that the extracellular microenvironment has a high potential to predict clinically relevant outcomes in HCC.
Collapse
Affiliation(s)
- Jade K. Macdonald
- Department
of Cell and Molecular Pharmacology, Medical
University of South Carolina, Charleston, South Carolina 29425, United States
| | - Harrison B. Taylor
- Department
of Cell and Molecular Pharmacology, Medical
University of South Carolina, Charleston, South Carolina 29425, United States
| | - Mengjun Wang
- Department
of Cell and Molecular Pharmacology, Medical
University of South Carolina, Charleston, South Carolina 29425, United States
| | - Andrew Delacourt
- Department
of Cell and Molecular Pharmacology, Medical
University of South Carolina, Charleston, South Carolina 29425, United States
| | - Christin Edge
- Department
of Cell and Molecular Pharmacology, Medical
University of South Carolina, Charleston, South Carolina 29425, United States
| | - David N. Lewin
- Department
of Cell and Molecular Pharmacology, Medical
University of South Carolina, Charleston, South Carolina 29425, United States
| | - Naoto Kubota
- Liver
Tumor Translational Research Program, Simmons Comprehensive Cancer
Center, Division of Digestive and Liver Diseases, Department of Internal
Medicine, University of Texas Southwestern
Medical Center, Dallas, Texas 75390, United States
| | - Naoto Fujiwara
- Liver
Tumor Translational Research Program, Simmons Comprehensive Cancer
Center, Division of Digestive and Liver Diseases, Department of Internal
Medicine, University of Texas Southwestern
Medical Center, Dallas, Texas 75390, United States
| | - Fahmida Rasha
- Liver
Tumor Translational Research Program, Simmons Comprehensive Cancer
Center, Division of Digestive and Liver Diseases, Department of Internal
Medicine, University of Texas Southwestern
Medical Center, Dallas, Texas 75390, United States
| | - Cesia A. Marquez
- Liver
Tumor Translational Research Program, Simmons Comprehensive Cancer
Center, Division of Digestive and Liver Diseases, Department of Internal
Medicine, University of Texas Southwestern
Medical Center, Dallas, Texas 75390, United States
| | - Atsushi Ono
- Department
of Gastroenterology, Graduate School of
Biomedical & Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| | - Shiro Oka
- Department
of Gastroenterology, Graduate School of
Biomedical & Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| | - Kazuaki Chayama
- Hiroshima
Institute of Life Sciences, Hiroshima 734-8553, Japan
- Collaborative
Research Laboratory of Medical Innovation, Research Center for Hepatology
and Gastroenterology, Hiroshima University, Hiroshima 734-8553, Japan
- RIKEN Center
for Integrative Medical Sciences, Yokohama 230-0045, Japan
| | - Sara Lewis
- Department
of Radiology, Icahn School of Medicine at
Mount Sinai, New York, New York 10029, United States
| | - Bachir Taouli
- Department
of Radiology, Icahn School of Medicine at
Mount Sinai, New York, New York 10029, United States
| | - Myron Schwartz
- Department
of Radiology, Icahn School of Medicine at
Mount Sinai, New York, New York 10029, United States
- Department
of Surgery, Icahn School of Medicine at
Mount Sinai, New York, New York 10029, United States
| | - M Isabel Fiel
- Department
of Radiology, Icahn School of Medicine at
Mount Sinai, New York, New York 10029, United States
- Department
of Pathology, Icahn School of Medicine at
Mount Sinai, New York, New York 10029, United States
| | - Richard R. Drake
- Department
of Cell and Molecular Pharmacology, Medical
University of South Carolina, Charleston, South Carolina 29425, United States
| | - Yujin Hoshida
- Liver
Tumor Translational Research Program, Simmons Comprehensive Cancer
Center, Division of Digestive and Liver Diseases, Department of Internal
Medicine, University of Texas Southwestern
Medical Center, Dallas, Texas 75390, United States
| | - Anand S. Mehta
- Department
of Cell and Molecular Pharmacology, Medical
University of South Carolina, Charleston, South Carolina 29425, United States
| | - Peggi M. Angel
- Department
of Cell and Molecular Pharmacology, Medical
University of South Carolina, Charleston, South Carolina 29425, United States
| |
Collapse
|
13
|
Chen ZY, Panga MJ, Zhang X, Qiao S, Chen S, Appiah C, Zhao Y. Estrogen alleviates liver fibrosis and restores metabolic homeostasis in ovariectomy-induced liver injury and carbon tetrachloride (CCl 4) exposure. Eur J Pharmacol 2024; 978:176774. [PMID: 38936452 DOI: 10.1016/j.ejphar.2024.176774] [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: 03/18/2024] [Revised: 06/17/2024] [Accepted: 06/23/2024] [Indexed: 06/29/2024]
Abstract
AIM Given estrogen's recognized regulatory influence on diverse metabolic and immune functions, this study sought to explore its potential impact on fibrosis and elucidate the underlying metabolic regulations. METHODS Female mice underwent ovary removal surgery, followed by carbon tetrachloride (CCl4) administration to induce liver injury. Biochemical index analysis and histopathological examination were then conducted. The expression levels of alpha-smooth muscle actin (α-SMA), transforming growth factor-β (TGF-β), and collagen type 1 alpha 1 chain (COL1A1) were assessed using western blotting to further elucidate the extent of liver injury. Finally, metabolite extraction and metabolomic analysis were performed to evaluate metabolic changes. RESULTS Ovary removal exacerbated CCl4-induced liver damage, while estrogen supplementation provided protection against hepatic changes resulting from OVX. Furthermore, estrogen mitigated liver injury induced by CCl4 treatment in vivo. Estrogen supplementation significantly restored liver damage induced by OVX and CCl4. Comparative analysis revealed significant alterations in pathways including aminoacyl-tRNA biosynthesis, glycine, serine, and threonine metabolism, lysine degradation, and taurine and hypotaurine metabolism in estrogen treatment. CONCLUSION Estrogen supplementation alleviates liver injury induced by OVX and CCl4, highlighting its protective effects against fibrosis and associated metabolic alterations.
Collapse
Affiliation(s)
- Zi Yi Chen
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211800, China
| | - Mogellah John Panga
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211800, China
| | - Xiangrui Zhang
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211800, China
| | - Shuai Qiao
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211800, China
| | - Shitian Chen
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211800, China
| | - Clara Appiah
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211800, China
| | - Ye Zhao
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211800, China.
| |
Collapse
|
14
|
Fan Y, Xiao Y, Zhang Y, Chen J, Huang S, Bai W. Transcriptomic and multi-cytokines profile analysis revealed new insights into the integrating mechanisms of cyanidin-3-O-glucoside on male reproductive damage amelioration. Food Res Int 2024; 192:114802. [PMID: 39147501 DOI: 10.1016/j.foodres.2024.114802] [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: 05/10/2024] [Revised: 06/28/2024] [Accepted: 07/17/2024] [Indexed: 08/17/2024]
Abstract
Ulcerative colitis is a public health issue with a rising worldwide incidence. It has been found that current medications for treating UC may cause varying degrees of damage to male fertility. Our previous study demonstrated that cyanidin-3-O-glucoside (C3G) treatment could effectively restore reproductive damage in a mouse model of DSS induced colitis. However, the underlying mechanism of C3G alleviates UC induced male reproductive disorders remain scarce. The aim of this study is to discover the molecular mechanisms of C3G on the amelioration of UC stimulated reproductive disorders. The targeted genes toward UC-induced reproductive injury upon C3G treatments were explored by transcriptomic analysis. Hematological analysis, histopathological examination, and real time transcription-polymerase chain reaction (RT-PCR) analysis were applied for conjoined identification. Results showed that C3G may effectively target for reducing pro-inflammatory cytokine IL-6 in testis through cytokine-cytokine receptor interaction pathway. Transcriptome sequencing found that a series of genetic pathways involved in the protective effects of C3G on male reproduction were identified by gene ontology and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis. Further results presented that C3G could effectively restore mRNA expression levels of Ly6a and Col1a1, closely linked with UC induced male reproductive damage pathways. Sufficient results implied that Ly6a and Col1a1 may be treated as the promising therapeutic targets for the mechanism of C3G in treating UC induced reproductive impairment. C3G administration might be an effective dietary supplementation strategy for male reproduction improvement.
Collapse
Affiliation(s)
- Yueyao Fan
- Department of Food Science and Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Yuhang Xiao
- Department of Food Science and Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Yingying Zhang
- Department of Food Science and Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Jiali Chen
- Department of Food Science and Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China.
| | - Sammi Huang
- Department of Food Science and Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Weibin Bai
- Department of Food Science and Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| |
Collapse
|
15
|
Shi S, Zhu C, Hu Y, Jiang P, Zhao J, Xu Q. ENG is a Biomarker of Prognosis and Angiogenesis in Liver Cancer, and Promotes the Differentiation of Tumor Cells into Vascular ECs. FRONT BIOSCI-LANDMRK 2024; 29:315. [PMID: 39344331 DOI: 10.31083/j.fbl2909315] [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: 06/28/2024] [Revised: 08/08/2024] [Accepted: 08/16/2024] [Indexed: 10/01/2024]
Abstract
BACKGROUND Liver cancer is a highly lethal malignancy with frequent recurrence, widespread metastasis, and low survival rates. The aim of this study was to explore the role of Endoglin (ENG) in liver cancer progression, as well as its impacts on angiogenesis, immune cell infiltration, and the therapeutic efficacy of sorafenib. METHODS A comprehensive evaluation was conducted using online databases Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA), 76 pairs of clinical specimens of tumor and adjacent non-tumor liver tissue, and tissue samples from 32 hepatocellular carcinoma (HCC) patients treated with sorafenib. ENG expression levels were evaluated using quantitative Reverse Transcription Polymerase Chain Reaction (qRT-PCR), Western blot, and immunohistochemical analysis. Cox regression analysis, Spearman rank correlation analysis, and survival analysis were used to assess the results. Functional experiments included Transwell migration assays and tube formation assays with Human Umbilical Vein Endothelial Cells (HUVECs). RESULTS Tumor cells exhibited retro-differentiation into endothelial-like cells, with a significant increase in ENG expression in these tumor-derived endothelial cells (TDECs). High expression of ENG was associated with more aggressive cancer characteristics and worse patient prognosis. Pathway enrichment and functional analyses identified ENG as a key regulator of immune responses and angiogenesis in liver cancer. Further studies confirmed that ENG increases the expression of Collagen type Iα1 (COL1A1), thereby promoting angiogenesis in liver cancer. Additionally, HCC patients with elevated ENG levels responded well to sorafenib treatment. CONCLUSIONS This study found that ENG is an important biomarker of prognosis in liver cancer. Moreover, ENG is associated with endothelial cell differentiation in liver cancer and plays a crucial role in formation of the tumor vasculature. The assessment of ENG expression could be a promising strategy to identify liver cancer patients who might benefit from targeted immunotherapies.
Collapse
MESH Headings
- Humans
- Liver Neoplasms/genetics
- Liver Neoplasms/metabolism
- Liver Neoplasms/pathology
- Liver Neoplasms/blood supply
- Liver Neoplasms/drug therapy
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/metabolism
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Prognosis
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/blood supply
- Carcinoma, Hepatocellular/drug therapy
- Sorafenib/pharmacology
- Sorafenib/therapeutic use
- Cell Differentiation
- Endoglin/metabolism
- Endoglin/genetics
- Male
- Female
- Middle Aged
- Cell Line, Tumor
- Phenylurea Compounds/pharmacology
- Human Umbilical Vein Endothelial Cells/metabolism
- Endothelial Cells/metabolism
- Endothelial Cells/pathology
- Niacinamide/analogs & derivatives
- Niacinamide/pharmacology
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Angiogenesis
Collapse
Affiliation(s)
- Shangheng Shi
- Organ Transplantation Center, The Affiliated Hospital of Qingdao University, 266003 Qingdao, Shandong, China
- The Institute of Transplantation Science, Qingdao University, 266003 Qingdao, Shandong, China
| | - Cunle Zhu
- Organ Transplantation Center, The Affiliated Hospital of Qingdao University, 266003 Qingdao, Shandong, China
- The Institute of Transplantation Science, Qingdao University, 266003 Qingdao, Shandong, China
| | - Yue Hu
- Hepatobiliary and Pancreatic Surgery Department, Affiliated First Hospital of Ningbo University, 315000 Ningbo, Zhejiang, China
| | - Peng Jiang
- Organ Transplantation Center, The Affiliated Hospital of Qingdao University, 266003 Qingdao, Shandong, China
- The Institute of Transplantation Science, Qingdao University, 266003 Qingdao, Shandong, China
| | - Jinxin Zhao
- Organ Transplantation Center, The Affiliated Hospital of Qingdao University, 266003 Qingdao, Shandong, China
- The Institute of Transplantation Science, Qingdao University, 266003 Qingdao, Shandong, China
| | - Qingguo Xu
- Organ Transplantation Center, The Affiliated Hospital of Qingdao University, 266003 Qingdao, Shandong, China
- The Institute of Transplantation Science, Qingdao University, 266003 Qingdao, Shandong, China
| |
Collapse
|
16
|
Sonkar R, Ma H, Waxman DJ. Steatotic liver disease induced by TCPOBOP-activated hepatic constitutive androstane receptor: primary and secondary gene responses with links to disease progression. Toxicol Sci 2024; 200:324-345. [PMID: 38710495 DOI: 10.1093/toxsci/kfae057] [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] [Indexed: 05/08/2024] Open
Abstract
Constitutive androstane receptor (CAR, Nr1i3), a liver nuclear receptor and xenobiotic sensor, induces drug, steroid, and lipid metabolizing enzymes, stimulates liver hypertrophy and hyperplasia, and ultimately, hepatocellular carcinogenesis. The mechanisms linking early CAR responses to later disease development are poorly understood. Here we show that exposure of CD-1 mice to TCPOBOP (1,4-bis[2-(3,5-dichloropyridyloxy)]benzene), a halogenated xenochemical and selective CAR agonist ligand, induces pericentral steatosis marked by hepatic accumulation of cholesterol and neutral lipid, and elevated circulating alanine aminotransferase, indicating hepatocyte damage. TCPOBOP-induced steatosis was weaker in the pericentral region but stronger in the periportal region in females compared with males. Early (1 day) TCPOBOP transcriptional responses were enriched for CAR-bound primary response genes, and for lipogenesis and xenobiotic metabolism and oxidative stress protection pathways; late (2 weeks) TCPOBOP responses included many CAR binding-independent secondary response genes, with enrichment for macrophage activation, immune response, and cytokine and reactive oxygen species production. Late upstream regulators specific to TCPOBOP-exposed male liver were linked to proinflammatory responses and hepatocellular carcinoma progression. TCPOBOP administered weekly to male mice using a high corn oil vehicle induced carbohydrate-responsive transcription factor (MLXIPL)-regulated target genes, dysregulated mitochondrial respiratory and translation regulatory pathways, and induced more advanced liver pathology. Overall, TCPOBOP exposure recapitulates histological and gene expression changes characteristic of emerging steatotic liver disease, including secondary gene responses in liver nonparenchymal cells indicative of transition to a more advanced disease state. Upstream regulators of both the early and late TCPOBOP response genes include novel biomarkers for foreign chemical-induced metabolic dysfunction-associated steatotic liver disease.
Collapse
Affiliation(s)
- Ravi Sonkar
- Department of Biology and Bioinformatics Program, Boston University, Boston, Massachusetts 02215, USA
| | - Hong Ma
- Department of Biology and Bioinformatics Program, Boston University, Boston, Massachusetts 02215, USA
| | - David J Waxman
- Department of Biology and Bioinformatics Program, Boston University, Boston, Massachusetts 02215, USA
| |
Collapse
|
17
|
Tai Y, Han D, Yang X, Cai G, Li H, Li J, Deng X. Endothelin-3 Suppresses Luteinizing Hormone Receptor Expression by Regulating the cAMP-PKA Pathway in Hen Granulosa Cells. Curr Issues Mol Biol 2024; 46:7832-7845. [PMID: 39194681 DOI: 10.3390/cimb46080464] [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: 05/28/2024] [Revised: 07/16/2024] [Accepted: 07/18/2024] [Indexed: 08/29/2024] Open
Abstract
Previous research identified the expression of EDN3 in granulosa cells of preovulatory follicles in chickens. Notably, the expression level of EDN3 in Silky Fowl with low egg-laying performance was significantly higher than that in high-yield laying breed White Leghorn. Given the crucial role of granulosa cells in follicular development and maturation, it is very important to study the effect of EDN3 on the biological function of granular cells. In this study, an EDN3 overexpression plasmid was constructed and transfected into granular cells. The viability of these cells was detected using quantiative (qPCR), Cell Counting Kit-8 (CCK8), and 5-Ethynyl-2'-deoxyuridine (EdU) assays. Gonadal hormone synthesis was detected using enzyme-linked immunosorbent assay (ELISA) techniques. Finally, transcriptome sequencing was employed to identify differentially expressed genes. Result showed thatoverexpression of EDN3 was observed to promote cell viability. In addition, it significantly inhibits the expressions of LHR and cAMP-PKA signaling pathways. Cell transcriptome sequencing data displayed that EDN3 can upregulate energy metabolism and immune-related signaling pathways, whereas follicle maturation and the GnRH signaling pathway were downregulated. In conclusion, this study demonstrates that EDN3 can enhance granulosa cell viability and inhibit the expression of LHCGR, a process likely mediated through the cAMP-PKA signaling pathway. However, further evidence is required to substantiate the regulatory relationship between EDN3 and the cAMP-PKA signaling pathway.
Collapse
Affiliation(s)
- Yurong Tai
- Sanya Institute, China Agricultural University, Sanya, 572000, China
- Hainan Seed Industry Laboratory, Yazhou 572024, China
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory For Animal Breeding, China Agricultural University, Beijing 100000, China
| | - Deping Han
- Sanya Institute, China Agricultural University, Sanya, 572000, China
| | - Xue Yang
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory For Animal Breeding, China Agricultural University, Beijing 100000, China
| | - Ganxian Cai
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory For Animal Breeding, China Agricultural University, Beijing 100000, China
| | - Huaiyu Li
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory For Animal Breeding, China Agricultural University, Beijing 100000, China
| | - Junying Li
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory For Animal Breeding, China Agricultural University, Beijing 100000, China
| | - Xuemei Deng
- Sanya Institute, China Agricultural University, Sanya, 572000, China
- Hainan Seed Industry Laboratory, Yazhou 572024, China
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory For Animal Breeding, China Agricultural University, Beijing 100000, China
| |
Collapse
|
18
|
Ding DY, Jiang SY, Zu YX, Yang Y, Gan XJ, Yuan SX, Zhou WP. Collagen in hepatocellular carcinoma: A novel biomarker and therapeutic target. Hepatol Commun 2024; 8:e0489. [PMID: 38967581 PMCID: PMC11227359 DOI: 10.1097/hc9.0000000000000489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 05/22/2024] [Indexed: 07/06/2024] Open
Abstract
HCC is globally recognized as a major health threat. Despite significant progress in the development of treatment strategies for liver cancer, recurrence, metastasis, and drug resistance remain key factors leading to a poor prognosis for the majority of liver cancer patients. Thus, there is an urgent need to develop effective biomarkers and therapeutic targets for HCC. Collagen, the most abundant and diverse protein in the tumor microenvironment, is highly expressed in various solid tumors and plays a crucial role in the initiation and progression of tumors. Recent studies have shown that abnormal expression of collagen in the tumor microenvironment is closely related to the occurrence, development, invasion, metastasis, drug resistance, and treatment of liver cancer, making it a potential therapeutic target and a possible diagnostic and prognostic biomarker for HCC. This article provides a comprehensive review of the structure, classification, and origin of collagen, as well as its role in the progression and treatment of HCC and its potential clinical value, offering new insights into the diagnosis, treatment, and prognosis assessment of liver cancer.
Collapse
Affiliation(s)
- Dong-yang Ding
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, P. R. China
| | - Shu-ya Jiang
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, P. R. China
| | - Yun-xi Zu
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, P. R. China
| | - Yuan Yang
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, P. R. China
| | - Xiao-jie Gan
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, P. R. China
| | - Sheng-xian Yuan
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, P. R. China
| | - Wei-ping Zhou
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, P. R. China
| |
Collapse
|
19
|
Giallongo S, Ferrigno J, Caltabiano R, Broggi G, Alanazi AM, Distefano A, Tropea E, Tramutola A, Perluigi M, Volti GL, Barone E, Barbagallo IA. Aging exacerbates oxidative stress and liver fibrosis in an animal model of Down Syndrome. Aging (Albany NY) 2024; 16:10203-10215. [PMID: 38942607 PMCID: PMC11236314 DOI: 10.18632/aging.205970] [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: 10/17/2023] [Accepted: 01/11/2024] [Indexed: 06/30/2024]
Abstract
Down Syndrome (DS) is a common genetic disorder characterized by an extra copy of chromosome 21, leading to dysregulation of various metabolic pathways. Oxidative stress in DS is associated with neurodevelopmental defects, neuronal dysfunction, and a dementia onset resembling Alzheimer's disease. Additionally, chronic oxidative stress contributes to cardiovascular diseases and certain cancers prevalent in DS individuals. This study investigates the impact of ageing on oxidative stress and liver fibrosis using a DS murine model (Ts2Cje mice). Our results show that DS mice show increased liver oxidative stress and impaired antioxidant defenses, as evidenced by reduced glutathione levels and increased lipid peroxidation. Therefore, DS liver exhibits an altered inflammatory response and mitochondrial fitness as we showed by assaying the expression of HMOX1, CLPP, and the heat shock proteins Hsp90 and Hsp60. DS liver also displays dysregulated lipid metabolism, indicated by altered expression of PPARα, PPARγ, FATP5, and CTP2. Consistently, these changes might contribute to non-alcoholic fatty liver disease development, a condition characterized by liver fat accumulation. Consistently, histological analysis of DS liver reveals increased fibrosis and steatosis, as showed by Col1a1 increased expression, indicative of potential progression to liver cirrhosis. Therefore, our findings suggest an increased risk of liver pathologies in DS individuals, particularly when combined with the higher prevalence of obesity and metabolic dysfunctions in DS patients. These results shed a light on the liver's role in DS-associated pathologies and suggest potential therapeutic strategies targeting oxidative stress and lipid metabolism to prevent or mitigate liver-related complications in DS individuals.
Collapse
Affiliation(s)
- Sebastiano Giallongo
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania 95124, Italy
| | - Jessica Ferrigno
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania 95124, Italy
| | - Rosario Caltabiano
- Department G.F. Ingrassia, Section of Anatomic Pathology, University of Catania, Catania 95124, Italy
| | - Giuseppe Broggi
- Department G.F. Ingrassia, Section of Anatomic Pathology, University of Catania, Catania 95124, Italy
| | - Amer M Alanazi
- Pharmaceutical Biotechnology Laboratory, Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Alfio Distefano
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania 95124, Italy
| | - Emanuela Tropea
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania 95124, Italy
| | - Antonella Tramutola
- Department of Biochemical Sciences “A. Rossi-Fanelli”, Sapienza University of Rome, Roma, RM 00185, Italy
| | - Marzia Perluigi
- Department of Biochemical Sciences “A. Rossi-Fanelli”, Sapienza University of Rome, Roma, RM 00185, Italy
| | - Giovanni Li Volti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania 95124, Italy
| | - Eugenio Barone
- Department G.F. Ingrassia, Section of Anatomic Pathology, University of Catania, Catania 95124, Italy
| | | |
Collapse
|
20
|
Shen XT, Chen ZC, Wang XY, Wang XF, Xie SZ, Zheng X, Yang LY, Lu L. Establishment of homotrimer collagen type I signature and its association with clinical manifestation and tertiary lymphoid structures formation in liver cancer. Heliyon 2024; 10:e31320. [PMID: 38841477 PMCID: PMC11152946 DOI: 10.1016/j.heliyon.2024.e31320] [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: 02/09/2024] [Revised: 04/08/2024] [Accepted: 05/14/2024] [Indexed: 06/07/2024] Open
Abstract
Background collagen type I is a fundamental composition of extracellular matrix. Typically it exists in the form of a heterotrimer, consisting of two α1 chains encoded by COL1A1 and one α2 chain encoded by COL1A2. However, in cancer a homotrimeric form of collagen type I comprises three α1 chains encoded by COL1A1 was founded. There is still a lack of transcriptional and histologic methods for detecting homotrimeric collagen type I. Furthermore, a comprehensive analysis of the pan-cancer distribution pattern and clinical relevance of homotrimeric collagen type I is conspicuously absent. Method Using transcriptional and immunoflourance method, we established homocol signature, which is able to transcriptionally and histologically detect homotrimeric collagen type I. We investigated the diagnostic and prognostic potential of homocol as a novel cancer biomarker in a pan-cancer cohort. Furthermore, we assessed its association with clinical manifestations in a liver cancer cohort undergoing treatment at our institute. Result Homotrimer Collagen Type I is predominantly expressed by cancer cells and is linked to several critical cancer hallmarks, particularly inflammatory response and proliferation. Survival analyses have indicated that a high Homocol expression is correlated with poor outcomes in most types of cancer studied. In terms of cancer detection, Homocol demonstrated strong performance in Receiver Operating Characteristic (ROC) analysis, with an Area Under Curve (AUC) of 0.83 for pan-cancer detection and between 0.72 and 0.99 for individual cancers.In cohorts undergoing PD1 treatment, we noted a higher presence of Homocol in the response group. In a Hepatocellular Carcinoma (HCC) clinical set, high Homocol expression was associated with an increased formation of intra-tumor tertiary lymphoid structures (TLS), larger tumor sizes, more advanced Barcelona Clinic Liver Cancer (BCLC) stages, higher microvascular invasion (MVI) grades, absence of a capsule, and an enriched para-tumor collagen presence. Conclusion our research has led to the development of a novel gene signature that facilitates the detection of Homotrimer Collagen Type I. This may greatly assist efforts in cancer detection, prognosis, treatment response prediction, and further research into Homotrimer Collagen Type I.
Collapse
Affiliation(s)
- Xiao-Tian Shen
- Hepatobiliary Surgery, Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai, 200040, China
- Cancer Metastasis Institute, Fudan University, Shanghai, China
| | - Zhen-Chao Chen
- Hepatobiliary Surgery, Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai, 200040, China
- Cancer Metastasis Institute, Fudan University, Shanghai, China
| | - Xiang-Yu Wang
- Hepatobiliary Surgery, Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai, 200040, China
- Cancer Metastasis Institute, Fudan University, Shanghai, China
| | - Xu-Feng Wang
- Hepatobiliary Surgery, Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai, 200040, China
- Cancer Metastasis Institute, Fudan University, Shanghai, China
| | - Sun-Zhe Xie
- Hepatobiliary Surgery, Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai, 200040, China
- Cancer Metastasis Institute, Fudan University, Shanghai, China
| | - Xin Zheng
- Hepatobiliary Surgery, Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai, 200040, China
- Cancer Metastasis Institute, Fudan University, Shanghai, China
| | - Lu-Yu Yang
- Hepatobiliary Surgery, Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai, 200040, China
- Cancer Metastasis Institute, Fudan University, Shanghai, China
| | - Lu Lu
- Hepatobiliary Surgery, Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai, 200040, China
- Cancer Metastasis Institute, Fudan University, Shanghai, China
| |
Collapse
|
21
|
Chacon-Millan P, Lama S, Del Gaudio N, Gravina AG, Federico A, Pellegrino R, Luce A, Altucci L, Facchiano A, Caraglia M, Stiuso P. A Combination of Microarray-Based Profiling and Biocomputational Analysis Identified miR331-3p and hsa-let-7d-5p as Potential Biomarkers of Ulcerative Colitis Progression to Colorectal Cancer. Int J Mol Sci 2024; 25:5699. [PMID: 38891888 PMCID: PMC11171846 DOI: 10.3390/ijms25115699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 05/06/2024] [Accepted: 05/15/2024] [Indexed: 06/21/2024] Open
Abstract
Ulcerative colitis (UC), an inflammatory bowel disease (IBD), may increase the risk of colorectal cancer (CRC) by activating chronic proinflammatory pathways. The goal of this study was to find serum prediction biomarkers in UC to CRC development by combining low-density miRNA microarray and biocomputational approaches. The UC and CRC miRNA expression profiles were compared by low-density miRNA microarray, finding five upregulated miRNAs specific to UC progression to CRC (hsa-let-7d-5p, hsa-miR-16-5p, hsa-miR-145-5p, hsa-miR-223-5p, and hsa-miR-331-3p). The circRNA/miRNA/mRNA competitive endogenous RNA (ceRNA) network analysis showed that the candidate miRNAs were connected to well-known colitis-associated CRC ACVR2A, SOCS1, IGF2BP1, FAM126A, and CCDC85C mRNAs, and circ-SHPRH circRNA. SST and SCARA5 genes regulated by hsa-let-7d-5p, hsa-miR-145-5p, and hsa-miR-331-3p were linked to a poor survival prognosis in a CRC patient dataset from The Cancer Genome Atlas (TCGA). Lastly, our mRNA and miRNA candidates were validated by comparing their expression to differentially expressed mRNAs and miRNAs from colitis-associated CRC tissue databases. A high level of hsa-miR-331-3p and a parallel reduction in SOCS1 mRNA were found in tissue and serum. We propose hsa-miR-331-3p and possibly hsa-let-7d-5p as novel serum biomarkers for predicting UC progression to CRC. More clinical sample analysis is required for further validation.
Collapse
Affiliation(s)
- Pilar Chacon-Millan
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (P.C.-M.); (S.L.); (N.D.G.); (A.G.G.); (A.F.); (R.P.); (A.L.); (L.A.); (M.C.)
| | - Stefania Lama
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (P.C.-M.); (S.L.); (N.D.G.); (A.G.G.); (A.F.); (R.P.); (A.L.); (L.A.); (M.C.)
| | - Nunzio Del Gaudio
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (P.C.-M.); (S.L.); (N.D.G.); (A.G.G.); (A.F.); (R.P.); (A.L.); (L.A.); (M.C.)
| | - Antonietta Gerarda Gravina
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (P.C.-M.); (S.L.); (N.D.G.); (A.G.G.); (A.F.); (R.P.); (A.L.); (L.A.); (M.C.)
| | - Alessandro Federico
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (P.C.-M.); (S.L.); (N.D.G.); (A.G.G.); (A.F.); (R.P.); (A.L.); (L.A.); (M.C.)
| | - Raffaele Pellegrino
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (P.C.-M.); (S.L.); (N.D.G.); (A.G.G.); (A.F.); (R.P.); (A.L.); (L.A.); (M.C.)
| | - Amalia Luce
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (P.C.-M.); (S.L.); (N.D.G.); (A.G.G.); (A.F.); (R.P.); (A.L.); (L.A.); (M.C.)
| | - Lucia Altucci
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (P.C.-M.); (S.L.); (N.D.G.); (A.G.G.); (A.F.); (R.P.); (A.L.); (L.A.); (M.C.)
- Biogem Scarl, Institute of Genetic Research, Laboratory of Molecular and Precision Oncology, 83031 Ariano Irpino, Italy
- Institute of Experimental Endocrinology and Oncology “Gaetano Salvatore” (IEOS)-National Research Council (CNR), Via Sergio Pansini, 80131 Naples, Italy
- Programma di Epigenetica Medica, A.O.U. “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Angelo Facchiano
- Institute of Food Sciences, National Research Council, 83100 Avellino, Italy;
| | - Michele Caraglia
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (P.C.-M.); (S.L.); (N.D.G.); (A.G.G.); (A.F.); (R.P.); (A.L.); (L.A.); (M.C.)
- Biogem Scarl, Institute of Genetic Research, Laboratory of Molecular and Precision Oncology, 83031 Ariano Irpino, Italy
- Programma di Epigenetica Medica, A.O.U. “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Paola Stiuso
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (P.C.-M.); (S.L.); (N.D.G.); (A.G.G.); (A.F.); (R.P.); (A.L.); (L.A.); (M.C.)
| |
Collapse
|
22
|
Jalil SMA, Henry JC, Cameron AJM. Targets in the Tumour Matrisome to Promote Cancer Therapy Response. Cancers (Basel) 2024; 16:1847. [PMID: 38791926 PMCID: PMC11119821 DOI: 10.3390/cancers16101847] [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: 03/13/2024] [Revised: 05/03/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
The extracellular matrix (ECM) is composed of complex fibrillar proteins, proteoglycans, and macromolecules, generated by stromal, immune, and cancer cells. The components and organisation of the matrix evolves as tumours progress to invasive disease and metastasis. In many solid tumours, dense fibrotic ECM has been hypothesised to impede therapy response by limiting drug and immune cell access. Interventions to target individual components of the ECM, collectively termed the matrisome, have, however, revealed complex tumour-suppressor, tumour-promoter, and immune-modulatory functions, which have complicated clinical translation. The degree to which distinct components of the matrisome can dictate tumour phenotypes and response to therapy is the subject of intense study. A primary aim is to identify therapeutic opportunities within the matrisome, which might support a better response to existing therapies. Many matrix signatures have been developed which can predict prognosis, immune cell content, and immunotherapy responses. In this review, we will examine key components of the matrisome which have been associated with advanced tumours and therapy resistance. We have primarily focussed here on targeting matrisome components, rather than specific cell types, although several examples are described where cells of origin can dramatically affect tumour roles for matrix components. As we unravel the complex biochemical, biophysical, and intracellular transduction mechanisms associated with the ECM, numerous therapeutic opportunities will be identified to modify tumour progression and therapy response.
Collapse
Affiliation(s)
| | | | - Angus J. M. Cameron
- Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK; (S.M.A.J.); (J.C.H.)
| |
Collapse
|
23
|
Li X, Jin Y, Xue J. Unveiling Collagen's Role in Breast Cancer: Insights into Expression Patterns, Functions and Clinical Implications. Int J Gen Med 2024; 17:1773-1787. [PMID: 38711825 PMCID: PMC11073151 DOI: 10.2147/ijgm.s463649] [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: 02/12/2024] [Accepted: 04/21/2024] [Indexed: 05/08/2024] Open
Abstract
Collagen, the predominant protein constituent of the mammalian extracellular matrix (ECM), comprises a diverse family of 28 members (I-XXVIII). Beyond its structural significance, collagen is implicated in various diseases or cancers, notably breast cancer, where it influences crucial cellular processes including proliferation, metastasis, apoptosis, and drug resistance, intricately shaping cancer progression and prognosis. In breast cancer, distinct collagens exhibit differential expression profiles, with some showing heightened or diminished levels in cancerous tissues or cells compared to normal counterparts, suggesting specific and pivotal biological functions. In this review, we meticulously analyze the expression of individual collagen members in breast cancer, utilizing Transcripts Per Million (TPM) data sourced from the GEPIA2 database. Through this analysis, we identify collagens that deviate from normal expression patterns in breast cancer, providing a comprehensive overview of their expression dynamics, functional roles, and underlying mechanisms. Our findings shed light on recent advancements in understanding the intricate interplay between these aberrantly expressed collagens and breast cancer. This exploration aims to offer valuable insights for the identification of potential biomarkers and therapeutic targets, thereby advancing the prospects of more effective interventions in breast cancer treatment.
Collapse
Affiliation(s)
- Xia Li
- Department of Molecular Diagnosis, Northern Jiangsu People’s Hospital, Yangzhou, People’s Republic of China
| | - Yue Jin
- Department of Molecular Diagnosis, Northern Jiangsu People’s Hospital, Yangzhou, People’s Republic of China
| | - Jian Xue
- Department of Emergency Medicine, Yizheng People’s Hospital, Yangzhou, People’s Republic of China
| |
Collapse
|
24
|
Liu J, Zhang X, Yang M, Zhang X. CircCOL1A1 promotes proliferation, migration, and invasion of colorectal cancer (CRC) cells and glutamine metabolism through GLS1 up-regulation by sponging miR-214-3p. J Cancer Res Clin Oncol 2024; 150:211. [PMID: 38662258 PMCID: PMC11045592 DOI: 10.1007/s00432-024-05736-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 03/31/2024] [Indexed: 04/26/2024]
Abstract
BACKGROUND Circular ribose nucleic acids (circRNAs), an abundant type of noncoding RNAs, are widely expressed in eukaryotic cells and exert a significant impact on the initiation and progression of various disorders, including different types of cancer. However, the specific role of various circRNAs in colorectal cancer (CRC) pathology is still not fully understood. METHODS The initial step involved the use of quantitative reverse transcription polymerase chain reaction (RT-qPCR) to assess the expression levels of circRNAs and messenger RNA (mRNA) in CRC cell lines and tissues. Subsequently, functional analyses of circCOL1A1 knockdown were conducted in vitro and in vivo through cell counting kit (CCK)-8, colony formation and transwell assays, as well as xenograft mouse model of tumor formation. Molecular expression and interactions were investigated using luciferase reporter assays, Western blot analysis, RNA immunoprecipitation (RIP), and immunohistochemical staining. RESULTS The RT-qPCR results revealed elevated levels of circCOL1A1 expressions in CRC tissues and cell lines as compared to the normal counterparts. In addition, circCOL1A1 expression level was found to be correlated with TNM stage, lymph node metastases, distant metastases, and invasion. Knockdown of circCOL1A1 resulted in impaired invasion, migration, and proliferation of CRC cells, and suppressed tumor generation in the animal model. We further demonstrated that circCOL1A1 could act as a sponge for miR-214-3p, suppressing miR-214-3p activity and leading to the upregulation of GLS1 protein to promote glutamine metabolism. CONCLUSION These findings suggest that circCOL1A1 functions as an oncogenic molecule to promote CRC progression via miR-214-3p/GLS1 axis, hinting on the potential of circCOL1A1 as a therapeutic target for CRC.
Collapse
Affiliation(s)
- Jia Liu
- Oncology Teaching and Research Office, Hebei Medical University, No. 361, Zhongshan East Road, Shijiazhuang City, 050017, Hebei, China
- Second Department of Oncology, Hebei General Hospital, No. 348 Heping West Road, Shijiazhuang City, 050051, Hebei Province, China
| | - Xianbo Zhang
- Second Department of Oncology, Hebei General Hospital, No. 348 Heping West Road, Shijiazhuang City, 050051, Hebei Province, China
| | - Meijian Yang
- Second Department of Oncology, Hebei General Hospital, No. 348 Heping West Road, Shijiazhuang City, 050051, Hebei Province, China
| | - Xianghong Zhang
- Department of Pathology, The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Shijiazhuang City, 050000, Hebei, China.
| |
Collapse
|
25
|
Sun Y, Chen Y, Zhao H, Wang J, Liu Y, Bai J, Hu C, Shang Z. Lactate-driven type I collagen deposition facilitates cancer stem cell-like phenotype of head and neck squamous cell carcinoma. iScience 2024; 27:109340. [PMID: 38500829 PMCID: PMC10945209 DOI: 10.1016/j.isci.2024.109340] [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: 11/05/2023] [Revised: 01/05/2024] [Accepted: 02/22/2024] [Indexed: 03/20/2024] Open
Abstract
Lactate is known to play a crucial role in the progression of malignancies. However, its mechanism in regulating the malignant phenotype of head and neck squamous cell carcinoma (HNSCC) remains unclear. This study found that lactate increases cancer stem cell (CSC) characteristics of HNSCC by influencing the deposition of type I collagen (Col I). Lactate promotes Col I deposition through two distinct pathways. One is to convert lactate to pyruvate, a substrate for Col I hydroxylation. The other is the activation of HIF1-α and P4HA1, the latter being a rate-limiting enzyme for Col I synthesis. Inhibition of these two pathways effectively counteracts lactate-induced enhanced cell stemness. Further studies revealed that Col I affects CSC properties by regulating cell cycle dynamics. In conclusion, our research proposes that lactate-driven Col I deposition is essential for the acquisition of CSC properties, and lactate-centric Col I deposition may be an effective target for CSCs.
Collapse
Affiliation(s)
- Yunqing Sun
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yang Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Oral and Maxillofacial Surgery, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Hui Zhao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Oral and Maxillofacial-Head and Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Jingjing Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yuantong Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Junqiang Bai
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Chuanyu Hu
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Zhengjun Shang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Oral and Maxillofacial-Head and Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| |
Collapse
|
26
|
Zhou L, Zeng Y, Liu Y, Du K, Luo Y, Dai Y, Pan W, Zhang L, Zhang L, Tian F, Gu C. Cellular senescence and metabolic reprogramming model based on bulk/single-cell RNA sequencing reveals PTGER4 as a therapeutic target for ccRCC. BMC Cancer 2024; 24:451. [PMID: 38605343 PMCID: PMC11007942 DOI: 10.1186/s12885-024-12234-5] [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: 02/16/2024] [Accepted: 04/08/2024] [Indexed: 04/13/2024] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is the prevailing histological subtype of renal cell carcinoma and has unique metabolic reprogramming during its occurrence and development. Cell senescence is one of the newly identified tumor characteristics. However, there is a dearth of methodical and all-encompassing investigations regarding the correlation between the broad-ranging alterations in metabolic processes associated with aging and ccRCC. We utilized a range of analytical methodologies, such as protein‒protein interaction network analysis and least absolute shrinkage and selection operator (LASSO) regression analysis, to form and validate a risk score model known as the senescence-metabolism-related risk model (SeMRM). Our study demonstrated that SeMRM could more precisely predict the OS of ccRCC patients than the clinical prognostic markers in use. By utilizing two distinct datasets of ccRCC, ICGC-KIRC (the International Cancer Genome Consortium) and GSE29609, as well as a single-cell dataset (GSE156632) and real patient clinical information, and further confirmed the relationship between the senescence-metabolism-related risk score (SeMRS) and ccRCC patient progression. It is worth noting that patients who were classified into different subgroups based on the SeMRS exhibited notable variations in metabolic activity, immune microenvironment, immune cell type transformation, mutant landscape, and drug responsiveness. We also demonstrated that PTGER4, a key gene in SeMRM, regulated ccRCC cell proliferation, lipid levels and the cell cycle in vivo and in vitro. Together, the utilization of SeMRM has the potential to function as a dependable clinical characteristic to increase the accuracy of prognostic assessment for patients diagnosed with ccRCC, thereby facilitating the selection of suitable treatment strategies.
Collapse
Affiliation(s)
- Lijie Zhou
- Department of Urology, First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan Province, China.
- Unit of Day Surgery Center, First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan Province, China.
| | - Youmiao Zeng
- Department of Urology, First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan Province, China
- Department of Urology, Zhengzhou Key Laboratory for Molecular Biology of Urological Tumor Research, Henan Institute of Urology, First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan Province, China
| | - Yuanhao Liu
- Department of Urology, First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan Province, China
- Unit of Day Surgery Center, First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan Province, China
| | - Kaixuan Du
- Department of Urology, First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan Province, China
- Unit of Day Surgery Center, First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan Province, China
| | - Yongbo Luo
- Department of Urology, First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan Province, China
- Unit of Day Surgery Center, First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan Province, China
| | - Yiheng Dai
- Department of Urology, First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan Province, China
- Department of Urology, Zhengzhou Key Laboratory for Molecular Biology of Urological Tumor Research, Henan Institute of Urology, First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan Province, China
| | - Wenbang Pan
- Department of Urology, First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan Province, China
- Unit of Day Surgery Center, First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan Province, China
| | - Lailai Zhang
- Department of Urology, First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan Province, China
- Unit of Day Surgery Center, First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan Province, China
| | - Lei Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan Province, China.
| | - Fengyan Tian
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan Province, China.
| | - Chaohui Gu
- Department of Urology, First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan Province, China.
- Unit of Day Surgery Center, First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan Province, China.
| |
Collapse
|
27
|
Liu Z, Zhao P. Integrative analysis unveils ECM signatures and pathways driving hepatocellular carcinoma progression: A multi-omics approach and prognostic model development. J Cell Mol Med 2024; 28:e18230. [PMID: 38568083 PMCID: PMC10989547 DOI: 10.1111/jcmm.18230] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 02/19/2024] [Accepted: 02/23/2024] [Indexed: 04/05/2024] Open
Abstract
Liver hepatocellular carcinoma (LIHC) is a highly lethal form of cancer that is among the deadliest cancer types globally. In terms of cancer-related mortality rates, liver cancer ranks among the top three, underscoring the severity of this disease. Insufficient analysis has been conducted to fully understand the potential value of the extracellular matrix (ECM) in immune infiltration and the prognostic stratification of LIHC, despite its recognised importance in the development of this disease. The scRNA-seq data of GSE149614 was used to conduct single-cell analysis on 10 LIHC samples. CellChat scores were calculated for seven cell populations in the descending cohort to investigate cellular communication, while PROGENy scores were calculated to determine tumour-associated pathway scores in different cell populations. The pathway analysis using GO and KEGG revealed the enrichment of ECM-associated genes in the pathway, highlighting the potential role of the ECM in LIHC development. By utilizing the TCGA-LIHC cohort, an ECM-based prognostic model for LIHC was developed using Lasso regression. Immune infiltration scores were calculated using two methods, and the performance of the ECM-related risk score was evaluated using an independent cohort from the CheckMate study. To determine the precise expression of ECM-associated risk genes in LIHC, we evaluated hepatocellular carcinoma cell lines using a range of assays, including Western blotting, invasion assays and Transwell assays. Using single-cell transcriptome analysis, we annotated the spatially-specific distribution of major immune cell types in single-cell samples of LIHC. The main cell types identified and annotated included hepatocytes, T cells, myeloid cells, epithelial cells, fibroblasts, endothelial cells and B cells. The utilisation of cellchat and PROGENy analyses enabled the investigation and unveiling of signalling interactions, protein functionalities and the prominent influential pathways facilitated by the primary immune cell types within the LIHC. Numerous tumour pathways, including PI2K, EGFR and TGFb, demonstrated a close correlation with the involvement of ECM in LIHC. Moreover, an evaluation was conducted to assess the primary ECM-related functional changes and biological pathway enrichment in LIHC. Differential genes associated with ECM were identified and utilised to create prognostic models. The prognostic stratification value of these models for LIHC patients was confirmed through validation in multiple databases. Furthermore, through immune infiltration analysis, it was discovered that ECM might be linked to the irregular expression and regulation of numerous immune cells. Additionally, histone acetylation was mapped against gene mutation frequencies and differential expression profiles. The prognostic stratification efficacy of the ECM prediction model constructed in the context of PD-1 inhibitor therapy was also examined, and it exhibited strong stratification performance. Cellular experiments, including Western blotting, invasion and Transwell assays, revealed that ECM-associated risk genes have a promoting effect on the development of LIHC. The creation of biomarkers for LIHC using ECM-related genes unveiled substantial correlations with immune microenvironmental infiltration and functional mutations in various tumour pathways. This enlightens us to the possibility that the influence of ECM on tumours may extend beyond simply promoting the fibrotic process and the stromal composition of tumours.
Collapse
Affiliation(s)
- Zhen Liu
- Department of RadiologyShengjing Hospital of China Medical UniversityShenyangChina
| | - Pengfei Zhao
- Department of RadiologyShengjing Hospital of China Medical UniversityShenyangChina
| |
Collapse
|
28
|
García-Carrillo R, Molina-Pelayo FA, Zarate-Lopez D, Cabrera-Aguilar A, Ortega-Domínguez B, Domínguez-López M, Chiquete-Félix N, Dagnino-Acosta A, Velasco-Loyden G, Chávez E, Castro-Sánchez L, de Sánchez VC. An adenosine derivative promotes mitochondrial supercomplexes reorganization and restoration of mitochondria structure and bioenergetics in a diethylnitrosamine-induced hepatocellular carcinoma model. Sci Rep 2024; 14:6348. [PMID: 38491051 PMCID: PMC10943223 DOI: 10.1038/s41598-024-56306-9] [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: 12/22/2023] [Accepted: 03/05/2024] [Indexed: 03/18/2024] Open
Abstract
Hepatocellular carcinoma (HCC) progression is associated with dysfunctional mitochondria and bioenergetics impairment. However, no data about the relationship between mitochondrial supercomplexes (hmwSC) formation and ATP production rates in HCC are available. Our group has developed an adenosine derivative, IFC-305, which improves mitochondrial function, and it has been proposed as a therapeutic candidate for HCC. We aimed to determine the role of IFC-305 on both mitochondrial structure and bioenergetics in a sequential cirrhosis-HCC model in rats. Our results showed that IFC-305 administration decreased the number and size of liver tumors, reduced the expression of tumoral markers, and reestablished the typical architecture of the hepatic parenchyma. The livers of treated rats showed a reduction of mitochondria number, recovery of the mtDNA/nDNA ratio, and mitochondrial length. Also, IFC-305 increased cardiolipin and phosphatidylcholine levels and promoted hmwSC reorganization with changes in the expression levels of hmwSC assembly-related genes. IFC-305 in HCC modified the expression of several genes encoding elements of electron transport chain complexes and increased the ATP levels by recovering the complex I, III, and V activity. We propose that IFC-305 restores the mitochondrial bioenergetics in HCC by normalizing the quantity, morphology, and function of mitochondria, possibly as part of its hepatic restorative effect.
Collapse
Grants
- Ciencia de Frontera-2019 project 501204 Consejo Nacional de Humanidades, Ciencias y Tecnologías (CONAHCYT)
- Ciencia de Frontera-2019 project 501204 Consejo Nacional de Humanidades, Ciencias y Tecnologías (CONAHCYT)
- Ciencia de Frontera-2019 project 501204 Consejo Nacional de Humanidades, Ciencias y Tecnologías (CONAHCYT)
- Ciencia de Frontera-2019 project 501204 Consejo Nacional de Humanidades, Ciencias y Tecnologías (CONAHCYT)
- Ciencia de Frontera-2019 project 501204 Consejo Nacional de Humanidades, Ciencias y Tecnologías (CONAHCYT)
- Ciencia de Frontera-2019 project 501204 Consejo Nacional de Humanidades, Ciencias y Tecnologías (CONAHCYT)
- FOP02-2022-02 project 321696 Consejo Nacional de Humanidades, Ciencias y Tecnologías (CONAHCYT)
- Ciencia de Frontera-2019 project 501204 Consejo Nacional de Humanidades, Ciencias y Tecnologías (CONAHCYT)
- Ciencia de Frontera-2019 project 501204 Consejo Nacional de Humanidades, Ciencias y Tecnologías (CONAHCYT)
- Ciencia de Frontera-2019 project 501204 Consejo Nacional de Humanidades, Ciencias y Tecnologías (CONAHCYT)
- Ciencia de Frontera-2019 project 501204 Consejo Nacional de Humanidades, Ciencias y Tecnologías (CONAHCYT)
- PAPIIT-UNAM project IN214419 Universidad Nacional Autónoma de México
- PAPIIT-UNAM project IN214419 Universidad Nacional Autónoma de México
- PAPIIT-UNAM project IN214419 Universidad Nacional Autónoma de México
- PAPIIT-UNAM project IN214419 Universidad Nacional Autónoma de México
- PAPIIT-UNAM project IN214419 Universidad Nacional Autónoma de México
- PAPIIT-UNAM project IN214419 Universidad Nacional Autónoma de México
Collapse
Affiliation(s)
- Rosendo García-Carrillo
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, 28045, Colima, México
| | | | - David Zarate-Lopez
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, 28045, Colima, México
| | - Alejandro Cabrera-Aguilar
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, Ciudad de México, México
| | - Bibiana Ortega-Domínguez
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, Ciudad de México, México
| | - Mariana Domínguez-López
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, Ciudad de México, México
| | - Natalia Chiquete-Félix
- Departamento de Genética Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, Ciudad de México, México
| | - Adan Dagnino-Acosta
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, 28045, Colima, México
- CONAHCYT-Universidad de Colima, 28045, Colima, México
| | - Gabriela Velasco-Loyden
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, Ciudad de México, México
| | - Enrique Chávez
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, Ciudad de México, México
| | - Luis Castro-Sánchez
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, 28045, Colima, México.
- CONAHCYT-Universidad de Colima, 28045, Colima, México.
| | - Victoria Chagoya de Sánchez
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, Ciudad de México, México.
| |
Collapse
|
29
|
Zein N, Yassin F, Ayoub HG, Elewa YHA, Mohamed SKA, Mahmoud MH, Elfeky M, Batiha GES, Zahran MH. In vivo investigation of the anti-liver fibrosis impact of Balanites aegyptiaca/ chitosan nanoparticles. Biomed Pharmacother 2024; 172:116193. [PMID: 38301419 DOI: 10.1016/j.biopha.2024.116193] [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: 10/06/2023] [Revised: 01/16/2024] [Accepted: 01/18/2024] [Indexed: 02/03/2024] Open
Abstract
Balanites aegyptiaca (B. aegyptiaca) is an African herb with traditional medical applications. Various pathogenic factors cause hepatic fibrosis and require novel treatment alternatives. Nanoformulation-based natural products can overcome the available drug problems by increasing the efficacy of natural products targeting disease markers. The current study investigated B. aegyptiaca methanolic extract using high-pressure liquid chromatography (HPLC), and B. aegyptiaca/chitosan nanoparticles were prepared. In vivo, evaluation tests were performed to assess the curative effect of the successfully prepared B. aegyptiaca/chitosan nanoparticles. For 30 days, the rats were divided into six groups, typical and fibrosis groups, where the liver fibrosis groups received B. aegyptiaca extract, silymarin, chitosan nanoparticles, and B. aegyptiaca/chitosan nanoparticles daily. In the current investigation, phenolic molecules are the major compounds detected in B. aegyptiaca extract. UV showed that the prepared B. aegyptiaca /chitosan nanoparticles had a single peak at 280 nm, a particle size of 35.0 ± 6.0 nm, and a negative charge at - 8.3 mV. The animal studies showed that the synthetic B. aegyptiaca/chitosan nanoparticles showed substantial anti-fibrotic protective effects against CCl4-induced hepatic fibrosis in rats when compared with other groups through optimization of biochemical and oxidative markers, improved histological changes, and modulated the expression of Col1a1, Acta2 and Cxcl9 genes, which manage liver fibrosis. In conclusion, the current research indicated that the prepared B. aegyptiaca/chitosan nanoparticles improved histological structure and significantly enhanced the biochemical and genetic markers of liver fibrosis in an animal model.
Collapse
Affiliation(s)
- Nabila Zein
- Biochemistry Department, Faculty of Science, Zagazig University, Zagazig 44519, Egypt
| | - Fathy Yassin
- Chemistry Department, Faculty of Science, Zagazig University, Zagazig 44519, Egypt
| | - Heba G Ayoub
- Biochemistry Department, Faculty of Science, Zagazig University, Zagazig 44519, Egypt
| | - Yaser Hosny Ali Elewa
- Department of Histology and Cytology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt; Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan.
| | - Sherif Kh A Mohamed
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Mohamed H Mahmoud
- Department of Biochemistry, College of Science, King Saud University, Kingdom of Saudi Arabia
| | - Mohamed Elfeky
- Department of Biochemistry, Faculty of Veterinary Medicine, Alexandria University, Alexandria 21526, Egypt
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhur University, Damanhur 22511, AlBeheira, Egypt
| | - Mahmoud Hosny Zahran
- Internal Medicine Department, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt
| |
Collapse
|
30
|
Tian X, Gu L, Zeng F, Liu X, Zhou Y, Dou Y, Han J, Zhao Y, Zhang Y, Luo Q, Wang F. Strophanthidin Induces Apoptosis of Human Lung Adenocarcinoma Cells by Promoting TRAIL-DR5 Signaling. Molecules 2024; 29:877. [PMID: 38398629 PMCID: PMC10892344 DOI: 10.3390/molecules29040877] [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: 01/17/2024] [Revised: 02/08/2024] [Accepted: 02/08/2024] [Indexed: 02/25/2024] Open
Abstract
Strophanthidin (SPTD), one of the cardiac glycosides, is refined from traditional Chinese medicines such as Semen Lepidii and Antiaris toxicaria, and was initially used for the treatment of heart failure disease in clinic. Recently, SPTD has been shown to be a potential anticancer agent, but the underlying mechanism of action is poorly understood. Herein, we explored the molecular mechanism by which SPTD exerts anticancer effects in A549 human lung adenocarcinoma cells by means of mass spectrometry-based quantitative proteomics in combination with bioinformatics analysis. We revealed that SPTD promoted the expression of tumor necrosis factor (TNF)-related apoptosis-inducing ligand receptor 2 (TRAIL-R2, or DR5) in A549 cells to activate caspase 3/6/8, in particular caspase 3. Consequently, the activated caspases elevated the expression level of apoptotic chromatin condensation inducer in the nucleus (ACIN1) and prelamin-A/C (LMNA), ultimately inducing apoptosis via cooperation with the SPTD-induced overexpressed barrier-to-autointegration factor 1 (Banf1). Moreover, the SPTD-induced DEPs interacted with each other to downregulate the p38 MAPK/ERK signaling, contributing to the SPTD inhibition of the growth of A549 cells. Additionally, the downregulation of collagen COL1A5 by SPTD was another anticancer benefit of SPTD through the modulation of the cell microenvironment.
Collapse
Affiliation(s)
- Xiao Tian
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China;
- Beijing National Laboratory for Molecular Sciences, CAS Research/Education Center for Excellence in Molecular Sciences, National Centre for Mass Spectrometry in Beijing, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China; (L.G.); (X.L.); (Y.Z.); (Y.D.); (J.H.); (Y.Z.); (Y.Z.)
| | - Liangzhen Gu
- Beijing National Laboratory for Molecular Sciences, CAS Research/Education Center for Excellence in Molecular Sciences, National Centre for Mass Spectrometry in Beijing, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China; (L.G.); (X.L.); (Y.Z.); (Y.D.); (J.H.); (Y.Z.); (Y.Z.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Fangang Zeng
- School of Environment of Natural Resources, Remin University of China, Beijing 100875, China;
| | - Xingkai Liu
- Beijing National Laboratory for Molecular Sciences, CAS Research/Education Center for Excellence in Molecular Sciences, National Centre for Mass Spectrometry in Beijing, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China; (L.G.); (X.L.); (Y.Z.); (Y.D.); (J.H.); (Y.Z.); (Y.Z.)
| | - Yang Zhou
- Beijing National Laboratory for Molecular Sciences, CAS Research/Education Center for Excellence in Molecular Sciences, National Centre for Mass Spectrometry in Beijing, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China; (L.G.); (X.L.); (Y.Z.); (Y.D.); (J.H.); (Y.Z.); (Y.Z.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yang Dou
- Beijing National Laboratory for Molecular Sciences, CAS Research/Education Center for Excellence in Molecular Sciences, National Centre for Mass Spectrometry in Beijing, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China; (L.G.); (X.L.); (Y.Z.); (Y.D.); (J.H.); (Y.Z.); (Y.Z.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Juanjuan Han
- Beijing National Laboratory for Molecular Sciences, CAS Research/Education Center for Excellence in Molecular Sciences, National Centre for Mass Spectrometry in Beijing, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China; (L.G.); (X.L.); (Y.Z.); (Y.D.); (J.H.); (Y.Z.); (Y.Z.)
| | - Yao Zhao
- Beijing National Laboratory for Molecular Sciences, CAS Research/Education Center for Excellence in Molecular Sciences, National Centre for Mass Spectrometry in Beijing, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China; (L.G.); (X.L.); (Y.Z.); (Y.D.); (J.H.); (Y.Z.); (Y.Z.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yanyan Zhang
- Beijing National Laboratory for Molecular Sciences, CAS Research/Education Center for Excellence in Molecular Sciences, National Centre for Mass Spectrometry in Beijing, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China; (L.G.); (X.L.); (Y.Z.); (Y.D.); (J.H.); (Y.Z.); (Y.Z.)
| | - Qun Luo
- Beijing National Laboratory for Molecular Sciences, CAS Research/Education Center for Excellence in Molecular Sciences, National Centre for Mass Spectrometry in Beijing, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China; (L.G.); (X.L.); (Y.Z.); (Y.D.); (J.H.); (Y.Z.); (Y.Z.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Fuyi Wang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China;
- Beijing National Laboratory for Molecular Sciences, CAS Research/Education Center for Excellence in Molecular Sciences, National Centre for Mass Spectrometry in Beijing, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China; (L.G.); (X.L.); (Y.Z.); (Y.D.); (J.H.); (Y.Z.); (Y.Z.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
31
|
Cai H, Liao Y, Zhu L, Wang Z, Song J. Improving Cancer Survival Prediction via Graph Convolutional Neural Network Learning on Protein-Protein Interaction Networks. IEEE J Biomed Health Inform 2024; 28:1134-1143. [PMID: 37963003 DOI: 10.1109/jbhi.2023.3332640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2023]
Abstract
Cancer is one of the most challenging health problems worldwide. Accurate cancer survival prediction is vital for clinical decision making. Many deep learning methods have been proposed to understand the association between patients' genomic features and survival time. In most cases, the gene expression matrix is fed directly to the deep learning model. However, this approach completely ignores the interactions between biomolecules, and the resulting models can only learn the expression levels of genes to predict patient survival. In essence, the interaction between biomolecules is the key to determining the direction and function of biological processes. Proteins are the building blocks and principal undertakings of life activities, and as such, their complex interaction network is potentially informative for deep learning methods. Therefore, a more reliable approach is to have the neural network learn both gene expression data and protein interaction networks. We propose a new computational approach, termed CRESCENT, which is a protein-protein interaction (PPI) prior knowledge graph-based convolutional neural network (GCN) to improve cancer survival prediction. CRESCENT relies on the gene expression networks rather than gene expression levels to predict patient survival. The performance of CRESCENT is evaluated on a large-scale pan-cancer dataset consisting of 5991 patients from 16 different types of cancers. Extensive benchmarking experiments demonstrate that our proposed method is competitive in terms of the evaluation metric of the time-dependent concordance index( Ctd) when compared with several existing state-of-the-art approaches. Experiments also show that incorporating the network structure between genomic features effectively improves cancer survival prediction.
Collapse
|
32
|
Oliveto S, Ritter P, Deroma G, Miluzio A, Cordiglieri C, Benvenuti MR, Mutti L, Raimondi MT, Biffo S. The Impact of 3D Nichoids and Matrix Stiffness on Primary Malignant Mesothelioma Cells. Genes (Basel) 2024; 15:199. [PMID: 38397189 PMCID: PMC10887956 DOI: 10.3390/genes15020199] [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: 12/24/2023] [Revised: 01/23/2024] [Accepted: 01/25/2024] [Indexed: 02/25/2024] Open
Abstract
Malignant mesothelioma is a type of cancer that affects the mesothelium. It is an aggressive and deadly form of cancer that is often caused by exposure to asbestos. At the molecular level, it is characterized by a low number of genetic mutations and high heterogeneity among patients. In this work, we analyzed the plasticity of gene expression of primary mesothelial cancer cells by comparing their properties on 2D versus 3D surfaces. First, we derived from primary human samples four independent primary cancer cells. Then, we used Nichoids, which are micro-engineered 3D substrates, as three-dimensional structures. Nichoids limit the dimension of adhering cells during expansion by counteracting cell migration between adjacent units of a substrate with their microarchitecture. Tumor cells grow effectively on Nichoids, where they show enhanced proliferation. We performed RNAseq analyses on all the samples and compared the gene expression pattern of Nichoid-grown tumor cells to that of cells grown in a 2D culture. The PCA analysis showed that 3D samples were more transcriptionally similar compared to the 2D ones. The 3D Nichoids induced a transcriptional remodeling that affected mainly genes involved in extracellular matrix assembly. Among these genes responsible for collagen formation, COL1A1 and COL5A1 exhibited elevated expression, suggesting changes in matrix stiffness. Overall, our data show that primary mesothelioma cells can be effectively expanded in Nichoids and that 3D growth affects the cells' tensegrity or the mechanical stability of their structure.
Collapse
Affiliation(s)
- Stefania Oliveto
- Department of Biosciences, University of Milan, 20133 Milan, Italy; (S.O.); (G.D.)
- National Institute of Molecular Genetics, Fondazione Romeo ed Enrica Invernizzi, INGM, 20122 Milan, Italy; (P.R.); (A.M.); (C.C.)
| | - Paolo Ritter
- National Institute of Molecular Genetics, Fondazione Romeo ed Enrica Invernizzi, INGM, 20122 Milan, Italy; (P.R.); (A.M.); (C.C.)
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, 20133 Milano, Italy;
| | - Giorgia Deroma
- Department of Biosciences, University of Milan, 20133 Milan, Italy; (S.O.); (G.D.)
- National Institute of Molecular Genetics, Fondazione Romeo ed Enrica Invernizzi, INGM, 20122 Milan, Italy; (P.R.); (A.M.); (C.C.)
| | - Annarita Miluzio
- National Institute of Molecular Genetics, Fondazione Romeo ed Enrica Invernizzi, INGM, 20122 Milan, Italy; (P.R.); (A.M.); (C.C.)
| | - Chiara Cordiglieri
- National Institute of Molecular Genetics, Fondazione Romeo ed Enrica Invernizzi, INGM, 20122 Milan, Italy; (P.R.); (A.M.); (C.C.)
| | - Mauro Roberto Benvenuti
- Thoracic Surgery Unit, Department of Medical and Surgical Specialties Radiological Sciences and Public Health, Medical Oncology, University of Brescia, ASST Spedali Civili of Brescia, 25123 Brescia, Italy;
| | - Luciano Mutti
- Department of Applied Clinical Sciences and Biotechnology, DISCAB, Aquila University, 67100 L’ Aquila, Italy;
- Department of Biotechnology, SHRO, Temple University, Philadelphia, PA 19122, USA
| | - Manuela Teresa Raimondi
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, 20133 Milano, Italy;
| | - Stefano Biffo
- Department of Biosciences, University of Milan, 20133 Milan, Italy; (S.O.); (G.D.)
- National Institute of Molecular Genetics, Fondazione Romeo ed Enrica Invernizzi, INGM, 20122 Milan, Italy; (P.R.); (A.M.); (C.C.)
| |
Collapse
|
33
|
Salimian N, Peymani M, Ghaedi K, Hashemi M, Rahimi E. Collagen 1A1 (COL1A1) and Collagen11A1(COL11A1) as diagnostic biomarkers in Breast, colorectal and gastric cancers. Gene 2024; 892:147867. [PMID: 37783295 DOI: 10.1016/j.gene.2023.147867] [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: 06/25/2023] [Revised: 09/28/2023] [Accepted: 09/29/2023] [Indexed: 10/04/2023]
Abstract
PURPOSE Collagen family genes (CFGs) play a significant role in the pathogenesis of cancers. This study aimed to evaluate changes in the expression levels (Els) of CFGs related to epithelial-mesenchymal transition (EMT) and metastasis in gastric (GC), breast (BC), and colorectal (CRC) cancers to introduce these genes as potential diagnostic biomarkers for these three types of cancer. METHODS The Cancer Genome Atlas (TCGA) examined ELS changes in CFGs associated with EMT and metastasis to determine their diagnostic value for GC, BC, and CRC. InteractiVenn was used to find genes shared by these three cancers. The biomarker role of CFGs was determined using the receiver operating characteristic (ROC) analysis. GC, BC, and CRC samples were analyzed using the RT-qPCR method to verify the bioinformatics results and evaluate the EL of the selected genes as biomarkers for these cancers. RESULTS The in-silico results showed a significant increase in the EL of several CFGs involved in EMT and metastasis in GC, BC, and CRC samples compared to healthy samples. Six common genes (COL11A1, COL12A1, COL1A1, COL1A2, COL5A1, and COL5A2) showed significantly increased in these three cancers, therebysupporting their oncogenic role. Furthermore, the biomarker-related analyses indicated that COL11A1 and COL1A1 were common diagnostic biomarkers for the three cancers. The RT-qPCR method confirmed that the ELs of COL11A1 and COL1A1 in the GC, BC, and CRC samples increased significantly compared to the adjacent normal samples. CONCLUSION CFGs in EMT and metastasis of GC, BC, and CRC are strong common diagnostic biomarkers for these cancers.
Collapse
Affiliation(s)
- Niloufar Salimian
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Maryam Peymani
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran.
| | - Kamran Ghaedi
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Mehrdad Hashemi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Ebrahim Rahimi
- Department of Food Hygiene, Faculty of Veterinary Medicine, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| |
Collapse
|
34
|
Deng X, Li Q, Yuan H, Hu H, Fan S. Galangin Promotes Tendon Repair Mediated by Tendon-Derived Stem Cells through Activating the TGF-β1/Smad3 Signaling Pathway. Chem Pharm Bull (Tokyo) 2024; 72:669-675. [PMID: 39010213 DOI: 10.1248/cpb.c24-00117] [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: 07/17/2024]
Abstract
Tendon injury is a prevalent orthopedic disease that currently lacks effective treatment. Galangin (GLN) is a vital flavonoid found abundantly in galangal and is known for its natural activity. This study aimed to investigate the GLN-mediated molecular mechanism of tendon-derived stem cells (TDSCs) in tendon repair. The TDSCs were characterized using alkaline phosphatase staining, alizarin red S staining, oil red O staining, and flow cytometry. The effect of GLN treatment on collagen deposition was evaluated using Sirius red staining and quantitative (q)PCR, while a Western bot was used to assess protein levels and analyze pathways. Results showed that GLN treatment not only increased the collagen deposition but also elevated the mRNA expression and protein levels of multiple tendon markers like collagen type I alpha 1 (COL1A1), decorin (DCN) and tenomodulin (TNMD) in TDSCs. Moreover, GLN was also found to upregulate the protein levels of transforming growth factor β1 (TGF-β1) and p-Smad3 to activate the TGF-β1/Smad3 signaling pathway, while GLN mediated collagen deposition in TDSCs was reversed by LY3200882, a TGF-β receptor inhibitor. The study concluded that GLN-mediated TDSCs enhanced tendon repair by activating the TGF-β1/Smad3 signaling pathway, suggesting a novel therapeutic option in treating tendon repair.
Collapse
Affiliation(s)
- Xiongwei Deng
- Department of Foot and Ankle Surgery, Nanchang Hongdu Hospital of Traditional Chinese Medicine
| | - Qiang Li
- Department of Foot and Ankle Surgery, Nanchang Hongdu Hospital of Traditional Chinese Medicine
| | - Haitao Yuan
- Department of Foot and Ankle Surgery, Nanchang Hongdu Hospital of Traditional Chinese Medicine
| | - Hejun Hu
- Department of Foot and Ankle Surgery, Nanchang Hongdu Hospital of Traditional Chinese Medicine
| | - Shaoyong Fan
- Department of Foot and Ankle Surgery, Nanchang Hongdu Hospital of Traditional Chinese Medicine
| |
Collapse
|
35
|
Liu ZT, Shen JT, Lei YJ, Huang YC, Zhao GQ, Zheng CH, Wang X, Wang YT, Chen L, Li ZX, Li SZ, Liao J, Yu TD. Molecular subtyping based on immune cell marker genes predicts prognosis and therapeutic response in patients with lung adenocarcinoma. BMC Cancer 2023; 23:1141. [PMID: 38001428 PMCID: PMC10668343 DOI: 10.1186/s12885-023-11579-7] [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: 02/06/2023] [Accepted: 10/27/2023] [Indexed: 11/26/2023] Open
Abstract
OBJECTIVE Lung adenocarcinoma (LA) is one of the most common malignancies and is responsible for the greatest number of tumor-related deaths. Our research aimed to explore the molecular subtype signatures of LA to clarify the correlation among the immune microenvironment, clinical outcomes, and therapeutic response. METHODS The LA immune cell marker genes (LICMGs) identified by single-cell RNA sequencing (scRNA-seq) analysis were used to discriminate the molecular subtypes and homologous immune and metabolic traits of GSE72094 LA cases. In addition, the model-building genes were identified from 1441 LICMGs by Cox-regression analysis, and a LA immune difference score (LIDscore) was developed to quantify individual differences in each patient, thereby predicting prognosis and susceptibility to immunotherapy and chemotherapy of LA patients. RESULTS Patients of the GSE72094 cohort were divided into two distinct molecular subtypes based on LICMGs: immune activating subtype (Cluster-C1) and metabolically activating subtype (cluster-C2). The two molecular subtypes have distinct characteristics regarding prognosis, clinicopathology, genomics, immune microenvironment, and response to immunotherapy. Among the LICMGs, LGR4, GOLM1, CYP24A1, SFTPB, COL1A1, HLA-DQA1, MS4A7, PPARG, and IL7R were enrolled to construct a LIDscore model. Low-LIDscore patients had a higher survival rate due to abundant immune cell infiltration, activated immunity, and lower genetic variation, but probably the higher levels of Treg cells in the immune microenvironment lead to immune cell dysfunction and promote tumor immune escape, thus decreasing the responsiveness to immunotherapy compared with that of the high-LIDscore patients. Overall, high-LIDscore patients had a higher responsiveness to immunotherapy and a higher sensitivity to chemotherapy than the low-LIDscore group. CONCLUSIONS Molecular subtypes based on LICMGs provided a promising strategy for predicting patient prognosis, biological characteristics, and immune microenvironment features. In addition, they helped identify the patients most likely to benefit from immunotherapy and chemotherapy.
Collapse
Affiliation(s)
- Zi-Tao Liu
- Department of Thoracic Surgery, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jun-Ting Shen
- Department of Thoracic Surgery, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yu-Jie Lei
- Department of Thoracic Surgery, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yun-Chao Huang
- Department of Thoracic Surgery, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Guang-Qiang Zhao
- Department of Thoracic Surgery, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Cheng-Hong Zheng
- Department of Ultrasound, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming, China
| | - Xi Wang
- Department of Thoracic Surgery, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yu-Tian Wang
- Department of Thoracic Surgery, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Long Chen
- Department of PET/CT Center, Cancer Center of Yunnan Province, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming, China
| | - Zi-Xuan Li
- Department of Thoracic Surgery, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Shou-Zhuo Li
- Department of Thoracic Surgery, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jun Liao
- Department of Thoracic Surgery, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Ting-Dong Yu
- Department of Thoracic Surgery, The Third Affiliated Hospital of Kunming Medical University, Kunming, China.
| |
Collapse
|
36
|
Patil A, Salvatori R, Smith L, Jenkins SM, Cannon A, Hartley CP, Graham RP, Moreira RK. Artificial intelligence-based reticulin proportionate area - a novel histological outcome predictor in hepatocellular carcinoma. Histopathology 2023; 83:512-525. [PMID: 37387193 DOI: 10.1111/his.15001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 05/18/2023] [Accepted: 06/17/2023] [Indexed: 07/01/2023]
Abstract
AIMS Reticulin stain is used routinely in the histological evaluation of hepatocellular carcinoma (HCC). The goal of this study was to assess whether the histological reticulin proportionate area (RPA) in HCCs predicts tumour-related outcomes. METHODS AND RESULTS We developed and validated a supervised artificial intelligence (AI) model that utilises a cloud-based, deep-learning AI platform (Aiforia Technologies, Helsinki, Finland) to specifically recognise and quantify the reticulin framework in normal livers and HCCs using routine reticulin staining. We applied this reticulin AI model to a cohort of consecutive HCC cases from patients undergoing curative resection between 2005 and 2015. A total of 101 HCC resections were included (median age = 68 years, 64 males, median follow-up time = 49.9 months). AI model RPA reduction of > 50% (compared to normal liver tissue) was predictive of metastasis [hazard ratio (HR) = 3.76, P = 0.004, disease-free survival (DFS, HR = 2.48, P < 0.001) and overall survival (OS), HR = 2.80, P = 0.001]. In a Cox regression model, which included clinical and pathological variables, RPA decrease was an independent predictor of DFS and OS and the only independent predictor of metastasis. Similar results were found in the moderately differentiated HCC subgroup (WHO grade 2), in which reticulin quantitative analysis was an independent predictor of metastasis, DFS and OS. CONCLUSION Our data indicate that decreased RPA is a strong predictor of various HCC-related outcomes, including within the moderately differentiated subgroup. Reticulin, therefore, may represent a novel and important prognostic HCC marker, to be further explored and validated.
Collapse
Affiliation(s)
- Ameya Patil
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Rebecca Salvatori
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | | | - Sarah M Jenkins
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Andrew Cannon
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | | | - Rondell P Graham
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Roger K Moreira
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| |
Collapse
|
37
|
Ezzo M, Hinz B. Novel approaches to target fibroblast mechanotransduction in fibroproliferative diseases. Pharmacol Ther 2023; 250:108528. [PMID: 37708995 DOI: 10.1016/j.pharmthera.2023.108528] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 08/09/2023] [Accepted: 09/07/2023] [Indexed: 09/16/2023]
Abstract
The ability of cells to sense and respond to changes in mechanical environment is vital in conditions of organ injury when the architecture of normal tissues is disturbed or lost. Among the various cellular players that respond to injury, fibroblasts take center stage in re-establishing tissue integrity by secreting and organizing extracellular matrix into stabilizing scar tissue. Activation, activity, survival, and death of scar-forming fibroblasts are tightly controlled by mechanical environment and proper mechanotransduction ensures that fibroblast activities cease after completion of the tissue repair process. Conversely, dysregulated mechanotransduction often results in fibroblast over-activation or persistence beyond the state of normal repair. The resulting pathological accumulation of extracellular matrix is called fibrosis, a condition that has been associated with over 40% of all deaths in the industrialized countries. Consequently, elements in fibroblast mechanotransduction are scrutinized for their suitability as anti-fibrotic therapeutic targets. We review the current knowledge on mechanically relevant factors in the fibroblast extracellular environment, cell-matrix and cell-cell adhesion structures, stretch-activated membrane channels, stress-regulated cytoskeletal structures, and co-transcription factors. We critically discuss the targetability of these elements in therapeutic approaches and their progress in pre-clinical and/or clinical trials to treat organ fibrosis.
Collapse
Affiliation(s)
- Maya Ezzo
- Keenan Research Institute for Biomedical Science of the St. Michael's Hospital, and Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Boris Hinz
- Keenan Research Institute for Biomedical Science of the St. Michael's Hospital, and Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
38
|
Ak A. Fibroblast Cell Responses to Vanadium and Niobium Titanium Alloys: A Biocompatibility Study. ACS OMEGA 2023; 8:33802-33808. [PMID: 37744787 PMCID: PMC10515373 DOI: 10.1021/acsomega.3c04252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 08/21/2023] [Indexed: 09/26/2023]
Abstract
The interactions of a biomaterial with tissues must be determined for the material to be fully compatible with the body for a long time. The tissue and environment where the material is implanted are highly affected by its content. Titanium-6Aluminum-4Vanadium is widely used in orthopedics and dentistry. Recently, Titanium-6Aluminum-7Niobium alloys have been studied because of Titanium-6Aluminum-4Vanadium toxicity, which may be caused by vanadium. The aim of this study was to determine whether Titanium-6Aluminum-4Vanadium and Titanium-6Aluminum-7Niobium affect fibroblast cell proliferation, mineralization, and collagen production and whether they change the expression of type 1 collagen and fibronectin genes. It was determined that the niobium-containing alloy increased cell proliferation and calcium mineralization compared with the vanadium-containing alloy (p < 0.05). However, the alloys did not cause changes in the expression of collagen type 1 or fibronectin in cells. The collagen content of the cells on the niobium-containing alloy was lower than that on both the vanadium-containing alloy and tissue culture plate surface (p < 0.05). The niobium-containing alloy was found to be superior to the vanadium-containing alloy in terms of cell proliferation and calcium mineralization. Furthermore, neither vanadium-containing alloy nor niobium-containing alloy implant materials altered gene expression. Although both alloys are considered compatible with bone tissue, it should be considered whether they are also biocompatible with fibroblast cells.
Collapse
Affiliation(s)
- Ayse Ak
- Kocaeli Vocational School
of Health Services, Department of Medical Services and Techniques,
Medical Imaging Techniques Program, Kocaeli
University, Kocaeli 41380, Turkey
| |
Collapse
|
39
|
Chen M, Zhu X, Zhang L, Zhao D. COL5A2 is a prognostic-related biomarker and correlated with immune infiltrates in gastric cancer based on transcriptomics and single-cell RNA sequencing. BMC Med Genomics 2023; 16:220. [PMID: 37723519 PMCID: PMC10506210 DOI: 10.1186/s12920-023-01659-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 09/09/2023] [Indexed: 09/20/2023] Open
Abstract
BACKGROUND There is still a therapeutic challenge in treating gastric cancer (GC) due to its high incidence and poor prognosis. Collagen type V alpha 2 (COL5A2) is increased in various cancers, yet it remains unclear how it contributes to the prognosis and immunity of GC. METHODS The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) datasets were used to download transcriptome profiling (TCGA-STAD; GSE84437), single-cell RNA sequencing (scRNA-seq) data (GSE167297) and clinical information. COL5A2 expression and its relationship with clinicopathological factors were analyzed. We conducted survival analysis and Cox regression analysis to evaluate the prognosis and independent factors of GC. Co-expressed analysis was also performed. To identify the underlying mechanism, we conducted analyses of differentially expressed genes (DEGs) and functional enrichment. The correlations between COL5A2 expression and immune cell infiltration levels and immune infiltrate gene marker sets were further explored. Additionally, we analyzed the association of COL5A2 expression with immunological checkpoint molecules. Furthermore, the relationship between COL5A2 expression and immunotherapy sensitivity was also investigated. RESULTS COL5A2 expression was elevated in GC. More than this, the scRNA-seq analysis revealed that COL5A2 expression had a spatial gradient. The upregulated COL5A2 was associated with worse overall survival. A significant correlation was found between COL5A2 overexpression and age, T classification and clinical stage in GC. COL5A2 was found to be an independent factor for the unfortunate outcome in Cox regression analysis. The co-expressed genes of COL5A2 were associated with tumor stage or poor survival. Enrichment analysis revealed that the DEGs were mainly associated with extracellular matrix (ECM)-related processes, PI3K-AKT signaling pathway, and focal adhesion. GSEA analyses revealed that COL5A2 was associated with tumor progression-related pathways. Meanwhile, COL5A2 expression was correlated with tumor-infiltrating immune cells. Moreover, immunophenoscore (IPS) analysis and PRJEB25780 cohorts showed that patients with low COL5A2 expression were highly sensitive to immunotherapy. CONCLUSIONS COL5A2 might act as a prognostic biomarker of GC prognosis and immune infiltration and may provide a therapeutic intervention strategy.
Collapse
Affiliation(s)
- Meiru Chen
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, 050000, China
- Department of Gastroenterology, Hengshui People's Hospital, Hengshui, Hebei Province, 053000, China
| | - Xinying Zhu
- Department of Gastroenterology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, 050000, China
| | - Lixian Zhang
- Department of Gastroenterology, Hengshui People's Hospital, Hengshui, Hebei Province, 053000, China
| | - Dongqiang Zhao
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, 050000, China.
| |
Collapse
|
40
|
Simon AG, Lyu SI, Laible M, Wöll S, Türeci Ö, Şahin U, Alakus H, Fahrig L, Zander T, Buettner R, Bruns CJ, Schroeder W, Gebauer F, Quaas A. The tight junction protein claudin 6 is a potential target for patient-individualized treatment in esophageal and gastric adenocarcinoma and is associated with poor prognosis. J Transl Med 2023; 21:552. [PMID: 37592303 PMCID: PMC10436499 DOI: 10.1186/s12967-023-04433-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 08/11/2023] [Indexed: 08/19/2023] Open
Abstract
BACKGROUND The prognosis of esophageal adenocarcinoma (EAC) and gastric adenocarcinoma (GAC) remains poor, and new therapeutic approaches are urgently needed. Claudin 6 (CLDN6) is an oncofetal antigen that is largely absent in healthy tissues and upregulated in several cancers, making it a promising therapeutical target. In this study, the expression of CLDN6 was assessed in an large Caucasian EAC and GAC cohort. METHODS RNA-Seq data from 89 EACs and 371 GACs were obtained from The Cancer Genome Atlas project and EAC/GAC cases were stratified by CLDN6 mRNA expression based on a survival-associated cutoff. For groups with CLDN6 expression above or below this cutoff, differential gene expression analyses were performed using DESeq, and dysregulated biological pathways were identified using the Enrichr tool. Additionally, CLDN6 protein expression was assessed in more than 800 EACs and almost 600 GACs using a CLDN6-specific immunohistochemical antibody (clone 58-4B-2) that is currently used in Phase I/II trials to identify patients with CLDN6-positive tumors (NCT05262530; NCT04503278). The expression of CLDN6 was also correlated with histopathological parameters and overall survival (OS). RESULTS EACs and GACs with high CLDN6 mRNA levels displayed an overexpression of pathways regulating the cell cycle, DNA replication, and receptor / extracellular matrix interactions. CLDN6 protein expression was associated with shorter OS in EAC and GAC, both in treatment-naïve subgroups and cohorts receiving neoadjuvant therapy. In multivariate analysis, CLDN6 protein expression was an independent adverse prognostic factor in EAC associated with a shorter OS (HR: 1.75; p = 0.01) and GAC (HR: 2.74; p = 0.028). CONCLUSIONS High expression of CLDN6 mRNA is associated with the dysregulation of distinct biological pathways regulating cell growth, proliferation, and cell-matrix interactions. Clinically, the expression of CLDN6 protein is a valuable adverse prognostic marker in EAC and GAC.
Collapse
Affiliation(s)
- Adrian Georg Simon
- Institute of Pathology, University Hospital Cologne, Medical Faculty, University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany.
| | - Su Ir Lyu
- Institute of Pathology, University Hospital Cologne, Medical Faculty, University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
| | | | | | | | | | - Hakan Alakus
- Department of General, Visceral and Cancer Surgery, University Hospital Cologne, Medical Faculty, University of Cologne, Cologne, Germany
| | - Luca Fahrig
- Department of General, Visceral and Cancer Surgery, University Hospital Cologne, Medical Faculty, University of Cologne, Cologne, Germany
| | - Thomas Zander
- Department of Internal Medicine I, University Hospital Cologne, Medical Faculty, University of Cologne, Cologne, Germany
| | - Reinhard Buettner
- Institute of Pathology, University Hospital Cologne, Medical Faculty, University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
| | - Christiane Josephine Bruns
- Department of General, Visceral and Cancer Surgery, University Hospital Cologne, Medical Faculty, University of Cologne, Cologne, Germany
| | - Wolfgang Schroeder
- Department of General, Visceral and Cancer Surgery, University Hospital Cologne, Medical Faculty, University of Cologne, Cologne, Germany
| | - Florian Gebauer
- Department of General, Visceral and Cancer Surgery, University Hospital Cologne, Medical Faculty, University of Cologne, Cologne, Germany
| | - Alexander Quaas
- Institute of Pathology, University Hospital Cologne, Medical Faculty, University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
| |
Collapse
|
41
|
Yang Y, Xiao B, Feng X, Chen Y, Wang Q, Fang J, Zhou P, Wei X, Cheng L. Identification of hub genes and key signaling pathways by weighted gene co-expression network analysis for human aortic stenosis and insufficiency. Front Cardiovasc Med 2023; 10:857578. [PMID: 37621558 PMCID: PMC10445149 DOI: 10.3389/fcvm.2023.857578] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 07/26/2023] [Indexed: 08/26/2023] Open
Abstract
Background Human aortic valve stenosis (AS) and insufficiency (AI) are common diseases in aging population. Identifying the molecular regulatory networks of AS and AI is expected to offer novel perspectives for AS and AI treatment. Methods Highly correlated modules with the progression of AS and AI were identified by weighted genes co-expression network analysis (WGCNA). Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were performed by the clusterProfiler program package. Differentially expressed genes (DEGs) were identified by the DESeqDataSetFromMatrix function of the DESeq2 program package. The protein-protein interaction (PPI) network analyses were implemented using the STRING online tool and visualized with Cytoscape software. The DEGs in AS and AI groups were overlapped with the top 30 genes with highest connectivity to screen out ten hub genes. The ten hub genes were verified by analyzing the data in high throughput RNA-sequencing dataset and real-time PCR assay using AS and AI aortic valve samples. Results By WGCNA algorithm, 302 highly correlated genes with the degree of AS, degree of AI, and heart failure were identified from highly correlated modules. GO analyses showed that highly correlated genes had close relationship with collagen fibril organization, extracellular matrix organization and extracellular structure organization. KEGG analyses also manifested that protein digestion and absorption, and glutathione metabolism were probably involved in AS and AI pathological courses. Moreover, DEGs were picked out for 302 highly correlated genes in AS and AI groups relative to the normal control group. The PPI network analyses indicated the connectivity among these highly correlated genes. Finally, ten hub genes (CD74, COL1A1, TXNRD1, CCND1, COL5A1, SERPINH1, BCL6, ITGA10, FOS, and JUNB) in AS and AI were found out and verified. Conclusion Our study may provide the underlying molecular targets for the mechanism research, diagnosis, and treatment of AS and AI in the future.
Collapse
Affiliation(s)
- Yang Yang
- Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of OrganTransplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, China
- NHC Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
- Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Bing Xiao
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Peking University, Beijing, China
| | - Xin Feng
- Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yue Chen
- Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qunhui Wang
- Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Fang
- Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ping Zhou
- Institute of OrganTransplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, China
- NHC Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
- Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Xiang Wei
- Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lin Cheng
- Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
42
|
Zhong NN, Li SR, Man QW, Liu B. Identification of Immune Infiltration in Odontogenic Keratocyst by Integrated Bioinformatics Analysis. BMC Oral Health 2023; 23:454. [PMID: 37415178 PMCID: PMC10324234 DOI: 10.1186/s12903-023-03175-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 06/26/2023] [Indexed: 07/08/2023] Open
Abstract
BACKGROUND Odontogenic keratocyst (OKC) is a relatively common odontogenic lesion characterized by local invasion in the maxillary and mandibular bones. In the pathological tissue slices of OKC, immune cell infiltrations are frequently observed. However, the immune cell profile and the molecular mechanism for immune cell infiltration of OKC are still unclear. We aimed to explore the immune cell profile of OKC and to explore the potential pathogenesis for immune cell infiltration in OKC. METHODS The microarray dataset GSE38494 including OKC and oral mucosa (OM) samples were obtained from the Gene Expression Omnibus (GEO) database. The differentially expressed genes (DEGs) in OKC were analyzed by R software. The hub genes of OKC were performed by protein-protein interaction (PPI) network. The differential immune cell infiltration and the potential relationship between immune cell infiltration and the hub genes were performed by single-sample gene set enrichment analysis (ssGSEA). The expression of COL1A1 and COL1A3 were confirmed by immunofluorescence and immunohistochemistry in 17 OKC and 8 OM samples. RESULTS We detected a total of 402 differentially expressed genes (DEGs), of which 247 were upregulated and 155 were downregulated. DEGs were mainly involved in collagen-containing extracellular matrix pathways, external encapsulating structure organization, and extracellular structure organization. We identified ten hub genes, namely FN1, COL1A1, COL3A1, COL1A2, BGN, POSTN, SPARC, FBN1, COL5A1, and COL5A2. A significant difference was observed in the abundances of eight types of infiltrating immune cells between the OM and OKC groups. Both COL1A1 and COL3A1 exhibited a significant positive correlation with natural killer T cells and memory B cells. Simultaneously, they demonstrated a significant negative correlation with CD56dim natural killer cells, neutrophils, immature dendritic cells, and activated dendritic cells. Immunohistochemistry analysis showed that COL1A1 (P = 0.0131) and COL1A3 (P < 0.001) were significantly elevated in OKC compared with OM. CONCLUSIONS Our findings provide insights into the pathogenesis of OKC and illuminate the immune microenvironment within these lesions. The key genes, including COL1A1 and COL1A3, may significantly impact the biological processes associated with OKC.
Collapse
Affiliation(s)
- Nian-Nian Zhong
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Su-Ran Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Qi-Wen Man
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China.
- Department of Oral & Maxillofacial - Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan, China.
| | - Bing Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China.
- Department of Oral & Maxillofacial - Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan, China.
| |
Collapse
|
43
|
Ding Y, Gong Y, Zeng H, Song G, Yu Z, Fu B, Liu Y, Huang D, Zhong Y. ZNF765 is a prognostic biomarker of hepatocellular carcinoma associated with cell cycle, immune infiltration, m 6A modification, and drug susceptibility. Aging (Albany NY) 2023; 15:6179-6211. [PMID: 37400985 PMCID: PMC10373972 DOI: 10.18632/aging.204827] [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: 09/21/2022] [Accepted: 06/05/2023] [Indexed: 07/05/2023]
Abstract
Hepatocellular carcinoma (HCC) is an ongoing challenge worldwide. Zinc finger protein 765 (ZNF765) is an important zinc finger protein that is related to the permeability of the blood-tumor barrier. However, the role of ZNF765 in HCC is unclear. This study evaluated the expression of ZNF765 in hepatocellular carcinoma and the impact of its expression on patient prognosis based on The Cancer Genome Atlas (TCGA). Immunohistochemical assays (IHC) were used to examine protein expression. Besides, a colony formation assay was used to examine cell viability. We also explored the relationship between ZNF765 and chemokines in the HCCLM3 cells by qRT-PCR. Moreover, we examined the effect of ZNF765 on cell resistance by measurement of the maximum half-inhibitory concentration. Our research revealed that ZNF765 expression in HCC samples was higher than that in normal samples, whose upregulation was not conducive to the prognosis. The results of GO, KEGG, and GSEA showed that ZNF765 was associated with the cell cycle and immune infiltration. Furthermore, we confirmed that the expression of ZNF765 had a strong connection with the infiltration level of various immune cells, such as B cells, CD4+ T cells, macrophages, and neutrophils. In addition, we found that ZNF765 was associated with m6A modification, which may affect the progression of HCC. Finally, drug sensitivity testing found that patients with HCC were sensitive to 20 drugs when they expressed high levels of ZNF765. In conclusion, ZNF765 may be a prognostic biomarker related to cell cycle, immune infiltration, m6A modification, and drug sensitivity for hepatocellular carcinoma.
Collapse
Affiliation(s)
- Yongqi Ding
- Second Affiliated Hospital of Nanchang University, Nanchang, China
- Second College of Clinical Medicine, Nanchang University, Nanchang, China
| | - Yiyang Gong
- Second College of Clinical Medicine, Nanchang University, Nanchang, China
| | - Hong Zeng
- Second College of Clinical Medicine, Nanchang University, Nanchang, China
| | - Gelin Song
- Second College of Clinical Medicine, Nanchang University, Nanchang, China
| | - Zichuan Yu
- Second College of Clinical Medicine, Nanchang University, Nanchang, China
| | - Bidong Fu
- Second College of Clinical Medicine, Nanchang University, Nanchang, China
| | - Yue Liu
- Second College of Clinical Medicine, Nanchang University, Nanchang, China
| | - Da Huang
- Department of Thyroid Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yanying Zhong
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
44
|
Xiong YX, Zhang XC, Zhu JH, Zhang YX, Pan YL, Wu Y, Zhao JP, Liu JJ, Lu YX, Liang HF, Zhang ZG, Zhang WG. Collagen I-DDR1 signaling promotes hepatocellular carcinoma cell stemness via Hippo signaling repression. Cell Death Differ 2023; 30:1648-1665. [PMID: 37117273 PMCID: PMC10307904 DOI: 10.1038/s41418-023-01166-5] [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: 06/18/2022] [Revised: 04/09/2023] [Accepted: 04/17/2023] [Indexed: 04/30/2023] Open
Abstract
Cancer stem cells (CSCs) are a minority population of cancer cells with stemness and multiple differentiation potentials, leading to cancer progression and therapeutic resistance. However, the concrete mechanism of CSCs in hepatocellular carcinoma (HCC) remains obscure. We found that in advanced HCC tissues, collagen I was upregulated, which is consistent with the expression of its receptor DDR1. Accordingly, high collagen I levels accompanied by high DDR1 expression are associated with poor prognoses in patients with HCC. Collagen I-induced DDR1 activation enhanced HCC cell stemness in vitro and in vivo. Mechanistically, DDR1 interacts with CD44, which acts as a co-receptor that amplifies collagen I-induced DDR1 signaling, and collagen I-DDR1 signaling antagonized Hippo signaling by facilitating the recruitment of PP2AA to MST1, leading to exaggerated YAP activation. The combined inhibition of DDR1 and YAP synergistically abrogated HCC cell stemness in vitro and tumorigenesis in vivo. A radiomic model based on T2 weighted images can noninvasively predict collagen I expression. These findings reveal the molecular basis of collagen I-DDR1 signaling inhibiting Hippo signaling and highlight the role of CD44/DDR1/YAP axis in promoting cancer cell stemness, suggesting that DDR1 and YAP may serve as novel prognostic biomarkers and therapeutic targets in HCC.
Collapse
Affiliation(s)
- Yi-Xiao Xiong
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, Hubei, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, China
| | - Xiao-Chao Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, Hubei, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, China
- Dermatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jing-Han Zhu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, Hubei, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, China
| | - Yu-Xin Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, Hubei, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, China
| | - Yong-Long Pan
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, Hubei, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, China
| | - Yu Wu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, Hubei, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, China
| | - Jian-Ping Zhao
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, Hubei, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, China
| | - Jun-Jie Liu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, Hubei, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, China
| | - Yuan-Xiang Lu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, Hubei, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, China
| | - Hui-Fang Liang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, Hubei, China.
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, China.
| | - Zhan-Guo Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, Hubei, China.
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, China.
| | - Wan-Guang Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, Hubei, China.
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, China.
| |
Collapse
|
45
|
Lee J, Kim H, Kang YW, Kim Y, Park MY, Song JH, Jo Y, Dao T, Ryu D, Lee J, Oh CM, Park S. LY6D is crucial for lipid accumulation and inflammation in nonalcoholic fatty liver disease. Exp Mol Med 2023; 55:1479-1491. [PMID: 37394588 PMCID: PMC10394021 DOI: 10.1038/s12276-023-01033-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: 01/08/2023] [Revised: 02/07/2023] [Accepted: 04/17/2023] [Indexed: 07/04/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a serious metabolic disorder characterized by excess fat accumulation in the liver. Over the past decade, NAFLD prevalence and incidence have risen globally. There are currently no effective licensed drugs for its treatment. Thus, further study is required to identify new targets for NAFLD prevention and treatment. In this study, we fed C57BL6/J mice one of three diets, a standard chow diet, high-sucrose diet, or high-fat diet, and then characterized them. The mice fed a high-sucrose diet had more severely compacted macrovesicular and microvesicular lipid droplets than those in the other groups. Mouse liver transcriptome analysis identified lymphocyte antigen 6 family member D (Ly6d) as a key regulator of hepatic steatosis and the inflammatory response. Data from the Genotype-Tissue Expression project database showed that individuals with high liver Ly6d expression had more severe NAFLD histology than those with low liver Ly6d expression. In AML12 mouse hepatocytes, Ly6d overexpression increased lipid accumulation, while Ly6d knockdown decreased lipid accumulation. Inhibition of Ly6d ameliorated hepatic steatosis in a diet-induced NAFLD mouse model. Western blot analysis showed that Ly6d phosphorylated and activated ATP citrate lyase, which is a key enzyme in de novo lipogenesis. In addition, RNA- and ATAC-sequencing analyses revealed that Ly6d drives NAFLD progression by causing genetic and epigenetic changes. In conclusion, Ly6d is responsible for the regulation of lipid metabolism, and inhibiting Ly6d can prevent diet-induced steatosis in the liver. These findings highlight Ly6d as a novel therapeutic target for NAFLD.
Collapse
Affiliation(s)
- Jibeom Lee
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Hyeonhui Kim
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
| | - Yun-Won Kang
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Yumin Kim
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Moon-Young Park
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Ji-Hong Song
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Yunju Jo
- Department of Molecular Cell Biology, Sungkyunkwan University (SKKU) School of Medicine, Suwon, Korea
| | - Tam Dao
- Department of Molecular Cell Biology, Sungkyunkwan University (SKKU) School of Medicine, Suwon, Korea
| | - Dongryeol Ryu
- Department of Molecular Cell Biology, Sungkyunkwan University (SKKU) School of Medicine, Suwon, Korea
| | - Junguee Lee
- Department of Pathology, St Mary's Hospital, the Catholic University of Korea, Daejeon, Korea
| | - Chang-Myung Oh
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Korea.
| | - Sangkyu Park
- Department of Precision Medicine, Yonsei University Wonju College of Medicine, Wonju, Korea.
- Mitohormesis Research Center, Yonsei University Wonju College of Medicine, Wonju, Gangwon-do, Korea.
| |
Collapse
|
46
|
Huber LT, Kraus JM, Ezić J, Wanli A, Groth M, Laban S, Hoffmann TK, Wollenberg B, Kestler HA, Brunner C. Liquid biopsy: an examination of platelet RNA obtained from head and neck squamous cell carcinoma patients for predictive molecular tumor markers. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2023; 4:422-446. [PMID: 37455825 PMCID: PMC10344902 DOI: 10.37349/etat.2023.00143] [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: 11/16/2022] [Accepted: 03/01/2023] [Indexed: 07/18/2023] Open
Abstract
Aim Recently, a tumor cell-platelet interaction was identified in different tumor entities, resulting in a transfer of tumor-derived RNA into platelets, named further "tumor-educated platelets (TEP)". The present pilot study aims to investigate whether such a tumor-platelet transfer of RNA occurs also in patients suffering from head and neck squamous cell carcinoma (HNSCC). Methods Sequencing analysis of RNA derived from platelets of tumor patients (TPs) and healthy donors (HDs) were performed. Subsequently, quantitative reverse transcription-polymerase chain reaction (qRT-PCR) was used for verification of differentially expressed genes in platelets from TPs and HDs in a second cohort of patients and HDs. Data were analyzed by applying bioinformatic tools. Results Sequencing of RNA derived from the tumor as well as from platelets of TPs and HDs revealed 426 significantly differentially existing RNA, at which 406 RNA were more and 20 RNA less abundant in platelets from TPs in comparison to that of HDs. In TPs' platelets, abundantly existing RNA coding for 49 genes were detected, characteristically expressed in epithelial cells and RNA, the products of which are involved in tumor progression. Applying bioinformatic tools and verification on a second TP/HD cohort, collagen type I alpha 1 chain (COL1A1) and zinc finger protein 750 (ZNF750) were identified as the strongest potentially platelet-RNA-sequencing (RNA-seq)-based biomarkers for HNSCC. Conclusions These results indicate a transfer of tumor-derived messenger RNA (mRNA) into platelets of HNSCC patients. Therefore, analyses of a patient's platelet RNA could be an efficient option for liquid biopsy in order to diagnose HNSCC or to monitor tumorigenesis as well as therapeutic responses at any time and in real time.
Collapse
Affiliation(s)
- Lisa T. Huber
- Department of Oto-Rhino-Laryngology, Head and Neck Surgery, Ulm University Medical Center, 89075 Ulm, Germany
| | - Johann M. Kraus
- Institute of Medical Systems Biology, Ulm University, 89081 Ulm, Germany
| | - Jasmin Ezić
- Department of Oto-Rhino-Laryngology, Head and Neck Surgery, Ulm University Medical Center, 89075 Ulm, Germany
| | - Amin Wanli
- Department of Oto-Rhino-Laryngology, Head and Neck Surgery, Ulm University Medical Center, 89075 Ulm, Germany
| | - Marco Groth
- Leibniz Institute of Aging – Fritz Lipmann Institute, CF DNA sequencing, 07745 Jena, Germany
| | - Simon Laban
- Department of Oto-Rhino-Laryngology, Head and Neck Surgery, Ulm University Medical Center, 89075 Ulm, Germany
| | - Thomas K. Hoffmann
- Department of Oto-Rhino-Laryngology, Head and Neck Surgery, Ulm University Medical Center, 89075 Ulm, Germany
| | - Barbara Wollenberg
- Clinic for Otorhinolaryngology, Head and Neck Surgery, Technical University of Munich, 80333 Munich, Germany
| | - Hans A. Kestler
- Institute of Medical Systems Biology, Ulm University, 89081 Ulm, Germany
| | - Cornelia Brunner
- Department of Oto-Rhino-Laryngology, Head and Neck Surgery, Ulm University Medical Center, 89075 Ulm, Germany
| |
Collapse
|
47
|
Lawal B, Kuo YC, Wu ATH, Huang HS. Therapeutic potential of EGFR/mTOR/Nf-kb targeting small molecule for the treatment of non-small cell lung cancer. Am J Cancer Res 2023; 13:2598-2616. [PMID: 37424807 PMCID: PMC10326574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 05/28/2023] [Indexed: 07/11/2023] Open
Abstract
Despite the therapeutic advancement with chemotherapy and targeted therapy against non-small-cell lung cancer (NSCLC), most patients ultimately develop resistance to these drugs, exhibiting disease progression, metastasis, and worse prognosis. There is, therefore, a need for the development of novel multi-targeted therapies that can offer a high therapeutic index with lesser chances of drug resistance against NSCLC. In the present study, we evaluated the therapeutic potential of a novel multi-target small molecule NLOC-015A for targeted treatment of NSCLC. Our in vitro studies revealed that NLOC-015A exhibited a broad spectrum of anticancer activities against lung cancer cell line. NLOC-015A decreased the viability of H1975 and H1299 cells with respective IC50 values of 2.07±0.19 and 1.90±0.23 µm. In addition, NLOC-015A attenuated the oncogenic properties (colony formation, migratory ability, and spheroid formation) with concomitant downregulation of expression levels of epidermal growth factor receptor (EGFR)/mammalian target of rapamycin (mTOR)/AKT, nuclear factor (NF)-κB, signaling network. In addition, the stemness inhibitory effect of NLOC0-15A was accompanied by decreased expression levels of aldehyde dehydrogenase (ALDH), MYC Proto-Oncogene (C-Myc), and (sex-determining region Y)-box 2 (SOX2) in both H1975 and H1299 cell lines. Furthermore, NLOC-015A suppressed the tumor burden and increased the body weight and survival of H1975 xenograft-bearing mice. Treatment with NLOC-015A also attenuated biochemical and hematological alterations in the tumor bearing mice. Interestingly, NLOC-015A synergistically enhanced the in vitro efficacy, and therapeutic outcome of osimertinib in vivo. In addition, the toxicity of osimertinib was significantly attenuated by combination with NLOC-015A. Altogether, our findings suggested that combining osimertinib with NLOC-015 appears to be a promising way to improve osimertinib's efficacy and achieve better therapeutic results against NSCLC. We therefore suggest that NLOC-015A might represent a new candidate for treating NSCLC via acting as a multitarget inhibitor of EGFR/mTOR/NF-Κb signaling networks and efficiently compromising the oncogenic phenotype of NSCLC.
Collapse
Affiliation(s)
- Bashir Lawal
- UPMC Hillman Cancer Center, University of PittsburghPittsburgh, PA 15260, USA
- Department of Pathology, University of PittsburghPittsburgh, PA 15260, USA
| | - Yu-Cheng Kuo
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical UniversityTaipei 11031, Taiwan
- School of Post-baccalaureate Chinese Medicine, College of Chinese Medicine, China Medical UniversityTaichung 40402, Taiwan
| | - Alexander TH Wu
- The PhD Program of Translational Medicine, College of Medical Science and Technology, Taipei Medical UniversityTaipei 11031, Taiwan
- Clinical Research Center, Taipei Medical University Hospital, Taipei Medical UniversityTaipei 11031, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei Medical UniversityTaipei 11031, Taiwan
- Graduate Institute of Medical Sciences, National Defense Medical CenterTaipei 11490, Taiwan
| | - Hsu-Shan Huang
- Graduate Institute of Medical Sciences, National Defense Medical CenterTaipei 11490, Taiwan
- PhD Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia SinicaTaipei 11031, Taiwan
- Graduate Institute for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical UniversityTaipei 11031, Taiwan
- School of Pharmacy, National Defense Medical CenterTaipei 11490, Taiwan
- PhD Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical UniversityTaipei 11031, Taiwan
| |
Collapse
|
48
|
Lange C, Brüggemann J, Thüner T, Jauckus J, Strowitzki T, Germeyer A. Changes in the expression of cancer- and metastasis-related genes and proteins after metformin treatment under different metabolic conditions in endometrial cancer cells. Heliyon 2023; 9:e16678. [PMID: 37313172 PMCID: PMC10258389 DOI: 10.1016/j.heliyon.2023.e16678] [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: 07/30/2022] [Revised: 05/20/2023] [Accepted: 05/24/2023] [Indexed: 06/15/2023] Open
Abstract
Research question Hyperinsulinemia and elevated estrogen levels are known risk factors for endometrial cancer (EC) development and are associated with obesity, type 2 diabetes mellitus (T2DM), insulin resistance, among others. Metformin, an insulin-sensitizing drug, displays anti-tumor effects in cancer patients, including EC, but the mechanism of action is still not completely understood. In the present study, the effects of metformin on gene and protein expression were investigated in pre- and postmenopausal EC in vitro models in order to identify candidates that are potentially involved in the drug's anti-cancer mechanism. Design After treating the cells with metformin (0.1 and 1.0 mmol/L), changes in the expression of >160 cancer- and metastasis-related gene transcripts were evaluated with RNA arrays. A total of 19 genes and 7 proteins were selected for a follow-up expression analysis, including further treatment conditions, in order to evaluate the influence of hyperinsulinemia and hyperglycemia on metformin-induced effects. Results Changes in the expression of BCL2L11, CDH1, CDKN1A, COL1A1, PTEN, MMP9 and TIMP2 were analyzed on gene and protein level. The consequences resulting from the detected expression changes as well as the influence of varying environmental influences are discussed in detail. With the presented data, we contribute to a better understanding of the direct anti-cancer activity of metformin as well as its underlying mechanism of action in EC cells. Conclusions Although further research will be necessary to confirm the data, the influence of different environmental settings on metformin-induced effects could be highlighted with the presented data. Additionally, gene and protein regulation were not similar in the pre- and postmenopausal in vitro models.
Collapse
|
49
|
Xia M, Jiao L, Wang XH, Tong M, Yao MD, Li XM, Yao J, Li D, Zhao PQ, Yan B. Single-cell RNA sequencing reveals a unique pericyte type associated with capillary dysfunction. Theranostics 2023; 13:2515-2530. [PMID: 37215579 PMCID: PMC10196835 DOI: 10.7150/thno.83532] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 04/10/2023] [Indexed: 05/24/2023] Open
Abstract
Background: Capillary dysfunction has been implicated in a series of life- threatening vascular diseases characterized by pericyte and endothelial cell (EC) degeneration. However, the molecular profiles that govern the heterogeneity of pericytes have not been fully elucidated. Methods: Single-cell RNA sequencing was conducted on oxygen-induced proliferative retinopathy (OIR) model. Bioinformatics analysis was conducted to identify specific pericytes involved in capillary dysfunction. qRT-PCRs and western blots were conducted to detect Col1a1 expression pattern during capillary dysfunction. Matrigel co-culture assays, PI staining, and JC-1 staining was conducted to determine the role of Col1a1 in pericyte biology. IB4 and NG2 staining was conducted to determine the role of Col1a1 in capillary dysfunction. Results: We constructed an atlas of > 76,000 single-cell transcriptomes from 4 mouse retinas, which could be annotated to 10 distinct retinal cell types. Using the sub-clustering analysis, we further characterized retinal pericytes into 3 different subpopulations. Notably, GO and KEGG pathway analysis demonstrated that pericyte sub-population 2 was identified to be vulnerable to retinal capillary dysfunction. Based on the single-cell sequencing results, Col1a1 was identified as a marker gene of pericyte sub-population 2 and a promising therapeutic target for capillary dysfunction. Col1a1 was abundantly expressed in pericytes and its expression was obviously upregulated in OIR retinas. Col1a1 silencing could retard the recruitment of pericytes toward endothelial cells and aggravated hypoxia-induced pericyte apoptosis in vitro. Col1a1 silencing could reduce the size of neovascular area and avascular area in OIR retinas and suppressed pericyte-myofibroblast transition and endothelial-mesenchymal transition. Moreover, Col1a1 expression was up-regulated in the aqueous humor of the patients with proliferative diabetic retinopathy (PDR) or retinopathy of prematurity (ROP) and up-regulated in the proliferative membranes of PDR patients. Conclusions: These findings enhance the understanding of the complexity and heterogeneity of retinal cells and have important implications for future treatment of capillary dysfunction.
Collapse
Affiliation(s)
- Min Xia
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing 210000, China
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing 210000, China
| | - Lyu Jiao
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Xiao-Han Wang
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Min Tong
- Eye Institute, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai 200030, China
| | - Mu-Di Yao
- Eye Institute, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai 200030, China
| | - Xiu-Miao Li
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing 210000, China
| | - Jin Yao
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing 210000, China
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing 210000, China
| | - Dan Li
- Eye Institute, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai 200030, China
| | - Pei-Quan Zhao
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Biao Yan
- Eye Institute, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai 200030, China
- NHC Key Laboratory of Myopia (Fudan University), Key Laboratory of Myopia, Chinese Academy of Medical Sciences, and Shanghai Key Laboratory of Visual Impairment and Restoration (Fudan University), Shanghai 200030, China
| |
Collapse
|
50
|
Ding Y, Gong Y, Zeng H, Zhou X, Yu Z, Pan J, Zhou M, Liu S, Lai W. Biological function analysis of ARHGAP39 as an independent prognostic biomarker in hepatocellular carcinoma. Aging (Albany NY) 2023; 15:2631-2666. [PMID: 37059586 PMCID: PMC10120899 DOI: 10.18632/aging.204635] [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: 01/01/2023] [Accepted: 03/11/2023] [Indexed: 04/16/2023]
Abstract
Hepatocellular carcinoma (HCC) is the most common subtype of liver cancer, with a high morbidity and low survival rate. Rho GTPase activating protein 39 (ARHGAP39) is a crucial activating protein of Rho GTPases, a novel target in cancer therapy, and it was identified as a hub gene for gastric cancer. However, the expression and role of ARHGAP39 in hepatocellular carcinoma remain unclear. Accordingly, the cancer genome atlas (TCGA) data were used to analyze the expression and clinical value of ARHGAP39 in hepatocellular carcinoma. Further, the LinkedOmics tool suggested functional enrichment pathways for ARHGAP39. To investigate in depth the possible role of ARHGAP39 on immune infiltration, we analyzed the relationship between ARHGAP39 and chemokines in HCCLM3 cells. Finally, the GSCA website was used to explore drug resistance in patients with high ARHGAP39 expression. Studies have shown that ARHGAP39 is highly expressed in hepatocellular carcinoma and relevant to clinicopathological features. In addition, the overexpression of ARHGAP39 leads to a poor prognosis. Besides, co-expressed genes and enrichment analysis showed a correlation with the cell cycle. Notably, ARHGAP39 may worsen the survival of hepatocellular carcinoma patients by increasing the level of immune infiltration through chemokines. Moreover, N6-methyladenosine (m6A) modification-related factors and drug sensitivity were also found to be associated with ARHGAP39. In brief, ARHGAP39 is a promising prognostic factor for hepatocellular carcinoma patients that is closely related to cell cycle, immune infiltration, m6A modification, and drug resistance.
Collapse
Affiliation(s)
- Yongqi Ding
- Department of Health Management Medical, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Second College of Clinical Medicine, Nanchang University, Nanchang, China
| | - Yiyang Gong
- Second College of Clinical Medicine, Nanchang University, Nanchang, China
| | - Hong Zeng
- Second College of Clinical Medicine, Nanchang University, Nanchang, China
| | - Xuanrui Zhou
- Second College of Clinical Medicine, Nanchang University, Nanchang, China
| | - Zichuan Yu
- Second College of Clinical Medicine, Nanchang University, Nanchang, China
| | - Jingying Pan
- Second College of Clinical Medicine, Nanchang University, Nanchang, China
| | - Minqin Zhou
- Second College of Clinical Medicine, Nanchang University, Nanchang, China
| | - Shiwen Liu
- Emergency Intensive Care Unit, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Wei Lai
- Department of Health Management Medical, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|