1
|
Zhang L, Wang K, Li F, Zhang L, Wu L, Tie R, Litifu K, Fu Y, Liu S, Ni J, Chang P, Xu J, Zhao H, Liu L. Ribosomal protein S3A (RPS3A), as a transcription regulator of colony-stimulating factor 1 (CSF1), promotes glioma progression through regulating the recruitment and autophagy-mediated M2 polarization of tumor-associated macrophages. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:5437-5452. [PMID: 39560749 DOI: 10.1007/s00210-024-03601-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 11/01/2024] [Indexed: 11/20/2024]
Abstract
Dysregulated expression of ribosomal protein S3A (RPS3A) is associated with the tissue infiltration of immune-related cells in a variety of cancers. However, the role of RPS3A in immune cell infiltration in glioma remains unclear. This study aimed to explore the role of RPS3A in the glioma immune microenvironment. RPS3A expression was detected in tumor tissues from patients with glioma. U251 cells were transfected with RPS3A shRNA (sh-RPS3A) and overexpression vector (pcDNA-RPS3A) and then co-cultured with PMA-induced THP-1 cells. Cell viability, invasion, and apoptosis were detected by Edu staining, Transwell, and flow cytometry, respectively. The expression of tumor-associated macrophage (TAM) M1 and M2 markers was detected with RT-qPCR. Next, the interaction between RPS3A and E4 transcription factor 1 (E4F1) was verified by Co-IP analysis, and the binding of E4F1 to colony-stimulating factor 1 (CSF1) promoter was verified by ChIP analysis. Overexpression vectors of CSF1 and E4F1 were used to treat sh-RPS3A-transfected U251 cells for reversal experiments. Finally, U251 cells transfected with sh-RPS3A adenovirus vectors were subcutaneously injected into nude mice to construct a xenograft tumor model, and the growth and metastasis of glioma in vivo were monitored. RPS3A was significantly upregulated in glioma tissues. Overexpression of RPS3A promoted glioma cell proliferation and invasion and inhibited apoptosis. Moreover, overexpression of RPS3A promoted TAM proliferation, invasion, and M2 polarization. Silencing RPS3A had the opposite effect. Silencing RPS3A inhibited autophagy in U251 cells, whereas rapamycin, an activator of autophagy, reversed the inhibitory effect of RPS3A silencing on TAM M2 polarization. Meanwhile, RPS3A promoted its expression by interacting with E4F1, and E4F1 promoted CSF1 transcriptional activation. Overexpression of CSF1 promoted the proliferation and invasion of U251 cells and reversed the inhibitory effect of RPS3A silencing on TAM proliferation and invasion, but had no effect on TAM M2 polarization. The results of in vivo experiments showed that knockdown of RPS3A significantly inhibited glioma tumor growth and metastasis in vivo. This study revealed that RPS3A recruited TAMs by upregulating E4F1-mediated transcription activation of CSF1, and promoted the M2 polarization of TAMs through autophagy, promoting glioma cell malignant growth and tumor progression.
Collapse
Affiliation(s)
- Liang Zhang
- Northwest University, Guodu Education and Technology Industrial Zone, No. 1 Xuefu Street, Chang'an District, Xi'an, 710127, China
- Xi'an Daxing Hospital, No. 353 Laodong North Road, Lianhu District, Xi'an, 710016, China
- Second Affiliated Hospital of Xi'an Medical University, No. 167 Fangdong Street, Xi'an, 710038, China
| | - Kun Wang
- Second Affiliated Hospital of Xi'an Medical University, No. 167 Fangdong Street, Xi'an, 710038, China
| | - Fei Li
- Second Affiliated Hospital of Xi'an Medical University, No. 167 Fangdong Street, Xi'an, 710038, China
| | - Lingxue Zhang
- Second Affiliated Hospital of Xi'an Medical University, No. 167 Fangdong Street, Xi'an, 710038, China
| | - Lin Wu
- Second Affiliated Hospital of Xi'an Medical University, No. 167 Fangdong Street, Xi'an, 710038, China
| | - Ru Tie
- Second Affiliated Hospital of Xi'an Medical University, No. 167 Fangdong Street, Xi'an, 710038, China
| | - Kamulan Litifu
- Second Affiliated Hospital of Xi'an Medical University, No. 167 Fangdong Street, Xi'an, 710038, China
| | - Yujie Fu
- Second Affiliated Hospital of Xi'an Medical University, No. 167 Fangdong Street, Xi'an, 710038, China
| | - Simeng Liu
- Second Affiliated Hospital of Xi'an Medical University, No. 167 Fangdong Street, Xi'an, 710038, China
| | - Jiaxin Ni
- Second Affiliated Hospital of Xi'an Medical University, No. 167 Fangdong Street, Xi'an, 710038, China
| | - Pan Chang
- Second Affiliated Hospital of Xi'an Medical University, No. 167 Fangdong Street, Xi'an, 710038, China
| | - Jun Xu
- Xi'an Daxing Hospital, No. 353 Laodong North Road, Lianhu District, Xi'an, 710016, China
| | - Haikang Zhao
- Second Affiliated Hospital of Xi'an Medical University, No. 167 Fangdong Street, Xi'an, 710038, China.
| | - Lingtong Liu
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, No.32 West Second Section First Ring Road, Chengdu, 610072, China.
| |
Collapse
|
2
|
Zhan X, Wang W, Lian J, Li Y, Gu J, Guo D, Xu D, Ju G. Identification of prognostic hub genes and functional role of BAIAP2L2 in prostate cancer progression: a transcriptomic and experimental study. Front Immunol 2025; 16:1543476. [PMID: 40356901 PMCID: PMC12066489 DOI: 10.3389/fimmu.2025.1543476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 04/08/2025] [Indexed: 05/15/2025] Open
Abstract
Background Prostate cancer (PCa) is a prevalent malignancy in men, and understanding its molecular mechanisms is crucial for identifying therapeutic targets. Methods Transcriptomic data from prostate tumors and matched healthy tissues were obtained from The Cancer Genome Atlas (TCGA). Differential expression analysis using the DESeq2 algorithm identified differentially expressed genes (DEGs). Cox proportional hazards regression was used to evaluate prognostic significance. Clinical validation involved comparing tumor specimens with normal tissues, focusing on BAIAP2L2, which showed significant differential expression and was further examined via immunohistochemical analysis. In vitro knockdown experiments were conducted in PC3 and DU145 cell lines to assess BAIAP2L2's functional role through assays for migration, colony formation, and proliferation. Results A total of 1,449 DEGs were identified, including 775 upregulated and 674 downregulated genes. Prognostic analysis revealed 748 genes linked to clinical outcomes, with 19 hub genes identified. QPCR confirmed significant upregulation of four candidates, including BAIAP2L2, which was also elevated in malignant tissues. BAIAP2L2 knockdown significantly impaired migration, proliferation, and viability in PCa cells. Conclusion This study highlights crucial molecular mechanisms in PCa progression, particularly the significance of BAIAP2L2 as a potential therapeutic target, warranting further investigation into additional hub genes for effective targeted strategies.
Collapse
Affiliation(s)
- Xiangyang Zhan
- Urology Center, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Surgical Institute of Integrative Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Surgical Institute, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, China
| | - Wenkai Wang
- Department of Oncology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jie Lian
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People‘s Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Yichun Li
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, China
| | - Jianyi Gu
- Urology Center, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Surgical Institute of Integrative Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Surgical Institute, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, China
| | - Dongdong Guo
- Urology Center, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Surgical Institute of Integrative Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Surgical Institute, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, China
| | - Dongliang Xu
- Urology Center, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Surgical Institute of Integrative Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Surgical Institute, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, China
| | - Guanqun Ju
- Urology Center, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Surgical Institute of Integrative Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Surgical Institute, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, China
| |
Collapse
|
3
|
Li P, Cui Y, Hu K, Wang X, Yu Y. Silencing APLNR enhances the radiosensitivity of prostate cancer by modulating the PI3K/AKT/mTOR signaling pathway. Clin Transl Oncol 2025; 27:1728-1737. [PMID: 39251496 DOI: 10.1007/s12094-024-03692-1] [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: 06/17/2024] [Accepted: 08/21/2024] [Indexed: 09/11/2024]
Abstract
BACKGROUND Aberrant expression of apelin receptor (APLNR) has been found to be involved in various cancers' development, however, its function in prostate cancer (PCa) remains unclear. The research aimed to investigate the role and potential mechanism of APLNR in PCa. METHODS The mRNA expression of APLNR was detected via qRT-PCR assay. PCa cell proliferation and apoptosis were determined through plate cloning and flow cytometry. In addition, the expression of apoptosis-related proteins (Bax, Bcl-2, and cleaved caspase-3) was evaluated using western blot. DNA damage marker (γ-H2AX) was analyzed by immunofluorescence and western blot. GSEA analysis was performed for seeking enrichment pathways of APLNR in PCa, and the protein levels of PI3K, p-PI3K, AKT, p-AKT, mTOR, and p-mTOR were tested using western blot. RESULTS APLNR expression was up-regulated in PCa tissues and cells. Silencing APLNR enhanced the sensitivity of PCa cells to radiotherapy, which was manifested by inhibiting cell proliferation, promoting cell apoptosis, and promoting DNA damage. Next, silencing APLNR inhibited the PI3K/AKT/mTOR pathway. Specifically, 740Y-P (the PI3K/AKT/mTOR pathway activator) reversed the effects of silencing APLNR on PCa cell proliferation, apoptosis and DNA damage. CONCLUSION Silencing APLNR inhibited cell proliferation, promoted cell apoptosis, and enhanced the radiosensitivity of PCa cells, which was involved in the PI3K/AKT/mTOR signaling pathway. This study is conducive to the deeper understanding of PCa and further provides a new perspective for the treatment of PCa.
Collapse
Affiliation(s)
- Peng Li
- Department of Urology, Yantaishan Hospital, Laishan District Science and Technology Avenue 10087, Yantai, 264003, Shandong, People's Republic of China
| | - Yanfang Cui
- Department of Ultrasonography, Yantaishan Hospital, Yantai, 264003, Shandong, People's Republic of China
| | - Keyao Hu
- Department of Urology, Yantaishan Hospital, Laishan District Science and Technology Avenue 10087, Yantai, 264003, Shandong, People's Republic of China
| | - Xiaofei Wang
- Department of Urology, Yantaishan Hospital, Laishan District Science and Technology Avenue 10087, Yantai, 264003, Shandong, People's Republic of China
| | - Yizhi Yu
- Department of Urology, Yantaishan Hospital, Laishan District Science and Technology Avenue 10087, Yantai, 264003, Shandong, People's Republic of China.
| |
Collapse
|
4
|
Zhang D, Zhao M, Jiang P, Zhou Y, Yan X, Zhou C, Mu Y, Xiao S, Ji G, Wu N, Sun D, Cui X, Ning S, Meng H, Xiao S, Jin Y. RPL22L1 fosters malignant features of cervical cancer via the modulation of DUSP6-ERK axis. J Transl Med 2025; 23:244. [PMID: 40022129 PMCID: PMC11871735 DOI: 10.1186/s12967-025-06249-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Accepted: 02/11/2025] [Indexed: 03/03/2025] Open
Abstract
BACKGROUND Cervical cancer remains one of the leading causes of cancer-related deaths among women globally, and there is still a need to research molecular targets that can be used for prognosis assessment and personalized molecular therapies. Here, we investigate the role of potential molecular target ribosomal L22-like 1 (RPL22L1) on cervical cancer, identify its potential mechanisms, and explore its related applications in prognosis and molecular therapies. METHODS Multiple cervical cancer cohorts online, tissue microarrays and clinical tissue specimens were analyzed for the association between RPL22L1 expression and patient outcomes. Functional and molecular biology studies of cell and mice models were used to clarify the effects and potential mechanisms of RPL22L1 on cervical cancer. RESULTS RPL22L1 is highly expressed in both cervical adenocarcinoma and squamous cell carcinoma, and its expression is significantly associated with histology grade, clinical stage, recurrence, vascular space involvement, tumor sizes and poor prognosis. In vitro and in vivo experiment revealed that RPL22L1 overexpression significantly promoted cervical cancer cell proliferation, migration, invasion, tumorigenicity and Sorafenib resistance, which were attenuated by RPL22L1 knockdown. Mechanistically, RPL22L1 competitively binds to ERK phosphatase DUSP6, leading to excessive activation of ERK. The combined application of ERK inhibitors can effectively inhibit RPL22L1 overexpressing cervical cancer cells both in vivo and in vitro. CONCLUSION RPL22L1 promotes malignant biological behavior of cervical cancer cells by competitively binding with DUSP6, thereby activating the ERK pathway. The combined use of Sorafenib and an ERK inhibitor is a potentially effective molecular targeted therapy for RPL22L1-high cervical cancer.
Collapse
Affiliation(s)
- Dongmei Zhang
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, 150081, China
- Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Harbin Medical University, Harbin, 150081, China
- Key Laboratory for Molecular Targeted Drug of Heilongjiang Province, College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Meiqi Zhao
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, 150081, China
- Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Harbin Medical University, Harbin, 150081, China
| | - Ping Jiang
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, 150081, China
- Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Harbin Medical University, Harbin, 150081, China
| | - Yunzhen Zhou
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, 150081, China
- Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Harbin Medical University, Harbin, 150081, China
| | - Xu Yan
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, 150081, China
- Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Harbin Medical University, Harbin, 150081, China
| | - Chong Zhou
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, 150081, China
- Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Harbin Medical University, Harbin, 150081, China
| | - Yu Mu
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, 150081, China
- Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Harbin Medical University, Harbin, 150081, China
| | - Shan Xiao
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, 150081, China
- Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Harbin Medical University, Harbin, 150081, China
| | - Guohua Ji
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, 150081, China
- Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Harbin Medical University, Harbin, 150081, China
| | - Nan Wu
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, 150081, China
- Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Harbin Medical University, Harbin, 150081, China
| | - Donglin Sun
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, 150081, China
- Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Harbin Medical University, Harbin, 150081, China
| | - Xiaobo Cui
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, 150081, China
- Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Harbin Medical University, Harbin, 150081, China
| | - Shangwei Ning
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Hongxue Meng
- Department of Pathology, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Sheng Xiao
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Yan Jin
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, 150081, China.
- Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Harbin Medical University, Harbin, 150081, China.
- State Key Laboratory of Zone Cardiovascular Diseases in China, Harbin Medical University, Harbin, 150081, China.
| |
Collapse
|
5
|
Liu B, Lu Y, Lin R, Xu J, Shang Z, Hou X, Shao X, Pan Z, Yu T, Feng W. Preclinical studies of the falnidamol as a highly potent and specific active ABCB1 transporter inhibitor. BMC Cancer 2025; 25:24. [PMID: 39773145 PMCID: PMC11707883 DOI: 10.1186/s12885-024-13371-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 12/19/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND ABCB1 overexpression is a key factor in causing multidrug resistance (MDR). As a result, it is crucial to discover effective medications against ABCB1 to overcome MDR. Falnidamol, a tyrosine kinase inhibitor (TKI) targeting the epidermal growth factor receptor (EGFR), is currently in phase 1 clinical trials for the treatment of solid tumors. In this work, we assessed whether falnidamol could act as an inhibitor of ABCB1 to reverse ABCB1-mediated MDR. METHODS The reversal effect of falnidamol on MDR was assessed by MTT, colony formation, 3D microsphere, and xenograft model assays. The protein expression or cellular localization was tested by western blot and immunofluorescence analysis. The intracellular doxorubicin accumulation and efflux were assessed by flow cytometry. The ATPase activity of ABCB1 was detected by a microplate reader. The interaction between falnidamol and ABCB1 was evaluated by docking analysis and cellular thermal shift assay. RESULTS Our data showed that falnidamol specifically reversed ABCB1-mediated MDR but not ABCG2-mediated MDR in vitro and in vivo. Mechanistic studies suggested falnidamol had no effect on ABCB1 expression or cellular localization, nor on the AKT or ERK pathways. Further studies found that falnidamol reduced ABCB1's efflux function, resulting in enhanced intracellular agent accumulation and thus overcoming MDR. ATPase assay showed that falnidamol suppressed the ABCB1 ATPase activity. Furthermore, docking analysis and cellular thermal shift assay indicated that falnidamol bound directly to the drug-binding site of ABCB1 transporter. CONCLUSION The present study proves that falnidamol acts as a highly potent and specific active ABCB1 transporter inhibitor, and can reverse ABCB1-mediated MDR, implying that combining falnidamol with ABCB1 substrate chemotherapeutic agents has the potential to overcome ABCB1-mediated MDR.
Collapse
Affiliation(s)
- Baojie Liu
- School of Life Science and Technology, Shandong Second Medical University, Weifang, Shandong, 261053, P.R. China
| | - Yongzheng Lu
- School of Life Science and Technology, Shandong Second Medical University, Weifang, Shandong, 261053, P.R. China
| | - Ruihui Lin
- School of Life Science and Technology, Shandong Second Medical University, Weifang, Shandong, 261053, P.R. China
| | - Junbao Xu
- Shandong Public Health Clinical Center, Shandong University, Jinan, Shandong, 250102, P.R. China
| | - Zilin Shang
- School of Life Science and Technology, Shandong Second Medical University, Weifang, Shandong, 261053, P.R. China
| | - Xinyu Hou
- School of Life Science and Technology, Shandong Second Medical University, Weifang, Shandong, 261053, P.R. China
| | - Xulong Shao
- School of Life Science and Technology, Shandong Second Medical University, Weifang, Shandong, 261053, P.R. China
| | - Zhifang Pan
- School of Life Science and Technology, Shandong Second Medical University, Weifang, Shandong, 261053, P.R. China.
| | - Tao Yu
- School of Life Science and Technology, Shandong Second Medical University, Weifang, Shandong, 261053, P.R. China.
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong, 266003, China.
| | - Weiguo Feng
- School of Life Science and Technology, Shandong Second Medical University, Weifang, Shandong, 261053, P.R. China.
| |
Collapse
|
6
|
Hong Y, Lin Q, Zhang Y, Liu J, Zheng Z. Research Progress of Ribosomal Proteins in Reproductive Development. Int J Mol Sci 2024; 25:13151. [PMID: 39684863 DOI: 10.3390/ijms252313151] [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: 10/09/2024] [Revised: 11/28/2024] [Accepted: 12/03/2024] [Indexed: 12/18/2024] Open
Abstract
Ribosomal proteins constitute the principal components of ribosomes, and their functions span a wide spectrum. Recent investigations have unveiled their involvement in oocyte and embryo development, playing a pivotal role in reproductive development. Numerous pieces of evidence indicate that ribosomal proteins participate in the regulation of various cellular activities, including nucleolar stress, oxidative stress, cell proliferation and autophagy. Despite these findings, the precise mechanisms through which ribosomal proteins influence reproductive development via these cellular activities remain elusive. Therefore, elucidating the mechanisms of action is essential for a comprehensive understanding of the role and function of ribosomal proteins in reproductive development. This paper systematically reviews the progress in research on nucleolar stress, oxidative stress, cell proliferation and autophagy concerning ribosomal proteins during reproductive development. Furthermore, we explore the potential of ribosomal proteins as diagnostic markers for various diseases. Additionally, we propose the development of drugs and therapies targeting ribosomal proteins, underscoring the potential for novel medical interventions in the context of reproductive health.
Collapse
Affiliation(s)
- Yuqi Hong
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Qisheng Lin
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Yuan Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Jilong Liu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Zhanhong Zheng
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
7
|
Li C, Du X, Zhang H, Liu S. Knockdown of ribosomal protein L22-like 1 arrests the cell cycle and promotes apoptosis in colorectal cancer. Cytojournal 2024; 21:45. [PMID: 39737125 PMCID: PMC11683392 DOI: 10.25259/cytojournal_29_2024] [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: 03/11/2024] [Accepted: 09/27/2024] [Indexed: 01/01/2025] Open
Abstract
Objective Colorectal cancer (CRC) remains a remarkable challenge despite considerable advancements in its treatment, due to its high recurrence rate, metastasis, drug resistance, and heterogeneity. Molecular targets that can effectively inhibit CRC growth must be identified to address these challenges. Therefore, we aim to reveal the regulatory effect of ribosomal protein L22-like 1 (RPL22L1) on the proliferation and apoptosis of CRC cells and its potential mechanism. Material and Methods We detected the expression of RPL22L1 from the Cancer Genome Atlas, Gene Expression Omnibus and UALCAN databases. The effects of RPL22L1 on CRC growth and migration were determined by knocking down RPL22L1 in human CRC cell lines and those on the cell cycle and apoptosis using flow cytometry. The influence of RPL22L1 knockdown on xenograft tumor growth was verified in vivo. The potential RPL22L1 mechanisms in promoting cancer were predicted with RNA sequencing (RNAseq). The molecular mechanism of enhanced apoptosis and cell cycle arrest in RPL22L1 knockdown was revealed using real-time reverse transcriptase-quantitative polymerase chain reaction (RT-qPCR) and Western blotting. Results The present study reveals a considerable upregulation of RPL22L1 expression in CRC as well as in diverse tumor tissues, and most cells within the CRC tumor microenvironment (TME) demonstrate RPL22L1 expression. Notably, this elevated expression level of RPL22L1 exhibits a strong association with an unfavorable prognosis among patients diagnosed with CRC (P < 0.05). Furthermore, the association between RPL22L1 expression and the CRC TME index did not exhibit statistical significance (P > 0.05). However, RPL22L1 knockdown experiments revealed a substantial suppression of growth and migratory capacities in CRC cells RKO and HCT116 (P < 0.05). Flow cytometry analysis exhibited that on RPL22L1 knockdown, a remarkable arrest of the G1 and S phases of the cell cycle (P < 0.05) occurred. In addition, a remarkable elevation in the level of cell apoptosis was observed (P < 0.001). RNAseq exhibited that cell cycle, DNA replication, and mechanistic target of rapamycin (mTOR) complex 1pathway were inhibited after RPL22L1 knockdown, whereas the apoptosis pathway was activated (P < 0.05). Validation through RT-qPCR and western blot analysis also corroborated the downregulation of P70S6K, MCM3, MCM7, GADD45B, WEE1, and MKI67 expression levels, following RPL22L1 knockdown (P < 0.05). Consequent rescue experiments offered supportive evidence, indicating the involvement of the mTOR pathway in mediating the influence of RPL22L1 on the promotion of cell cycle progression. Moreover, in vivo assays involving tumor-bearing mice exhibited that diminished RPL22L1 levels led to arrested CRC growth (P < 0.05). Conclusion These findings support RPL22L1 as a possible prognostic and therapeutic target in CRC, providing novel insights into the development of anticancer medications.
Collapse
Affiliation(s)
- Chunming Li
- Key Laboratory of Microecology-Immune Regulatory Network and Related Diseases, College of Basic Medicine, Jiamusi University, Jiamusi, China
| | - Xinna Du
- Key Laboratory of Microecology-Immune Regulatory Network and Related Diseases, College of Basic Medicine, Jiamusi University, Jiamusi, China
- Department of Physiology and Biochemistry, Jiangsu Vocational College of Medicine, Yancheng, China
| | - Hu Zhang
- Key Laboratory of Microecology-Immune Regulatory Network and Related Diseases, College of Basic Medicine, Jiamusi University, Jiamusi, China
- Department of Physiology and Biochemistry, Jiangsu Vocational College of Medicine, Yancheng, China
| | - Shuang Liu
- Key Laboratory of Microecology-Immune Regulatory Network and Related Diseases, College of Basic Medicine, Jiamusi University, Jiamusi, China
| |
Collapse
|
8
|
Zhang Z, Gao Z, Fang H, Zhao Y, Xing R. Therapeutic importance and diagnostic function of circRNAs in urological cancers: from metastasis to drug resistance. Cancer Metastasis Rev 2024; 43:867-888. [PMID: 38252399 DOI: 10.1007/s10555-023-10152-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 10/31/2023] [Indexed: 01/23/2024]
Abstract
Circular RNAs (circRNAs) are a member of non-coding RNAs with no ability in encoding proteins and their aberrant dysregulation is observed in cancers. Their closed-loop structure has increased their stability, and they are reliable biomarkers for cancer diagnosis. Urological cancers have been responsible for high mortality and morbidity worldwide, and developing new strategies in their treatment, especially based on gene therapy, is of importance since these malignant diseases do not respond to conventional therapies. In the current review, three important aims are followed. At the first step, the role of circRNAs in increasing or decreasing the progression of urological cancers is discussed, and the double-edged sword function of them is also highlighted. At the second step, the interaction of circRNAs with molecular targets responsible for urological cancer progression is discussed, and their impact on molecular processes such as apoptosis, autophagy, EMT, and MMPs is highlighted. Finally, the use of circRNAs as biomarkers in the diagnosis and prognosis of urological cancer patients is discussed to translate current findings in the clinic for better treatment of patients. Furthermore, since circRNAs can be transferred to tumor via exosomes and the interactions in tumor microenvironment provided by exosomes such as between macrophages and cancer cells is of importance in cancer progression, a separate section has been devoted to the role of exosomal circRNAs in urological tumors.
Collapse
Affiliation(s)
- Zhibin Zhang
- College of Traditional Chinese Medicine, Chengde Medical College, Chengde, 067000, Hebei, China.
| | - Zhixu Gao
- Chengde Medical College, Chengde, 067000, Hebei, China
| | - Huimin Fang
- Chengde Medical College, Chengde, 067000, Hebei, China
| | - Yutang Zhao
- Chengde Medical College, Chengde, 067000, Hebei, China
| | - Rong Xing
- Chengde Medical College, Chengde, 067000, Hebei, China
| |
Collapse
|
9
|
Yang J, Wu Y, Zhu Q, Qu X, Ou H, Liu H, Wei Y, Ge D, Lu C, Jiang B, Song X. Discovery of a first-in-class protein degrader for the c-ros oncogene 1 (ROS1). Bioorg Chem 2024; 150:107590. [PMID: 38955003 DOI: 10.1016/j.bioorg.2024.107590] [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: 05/12/2024] [Revised: 06/16/2024] [Accepted: 06/22/2024] [Indexed: 07/04/2024]
Abstract
The c-ros oncogene 1 (ROS1), an oncogenic driver, is known to induce non-small cell lung cancer (NSCLC) when overactivated, particularly through the formation of fusion proteins. Traditional targeted therapies focus on inhibiting ROS1 activity with ROS 1 inhibitors to manage cancer progression. However, a new strategy involving the design of protein degraders offers a more potent approach by completely degrading ROS1 fusion oncoproteins, thereby effectively blocking their kinase activity and enhancing anti-tumour potential. Utilizing PROteolysis-TArgeting Chimera (PROTAC) technology and informed by molecular docking and rational design, we report the first ROS1-specific PROTAC, SIAIS039. This degrader effectively targets multiple ROS1 fusion oncoproteins (CD74-ROS1, SDC4-ROS1 and SLC34A2-ROS1) in engineered Ba/F3 cells and HCC78 cells, demonstrating anti-tumour effects against ROS1 fusion-driven cancer cells. It suppresses cell proliferation, induces cell cycle arrest, and apoptosis, and inhibits clonogenicity. The anti-tumour efficacy of SIAIS039 surpasses two approved drugs, crizotinib and entrectinib, and matches that of the top inhibitors, including lorlatinib and taletrectinib. Mechanistic studies confirm that the degradation induced by 039 requires the participation of ROS1 ligands and E3 ubiquitin ligases, and involves the proteasome and ubiquitination. In addition, 039 exhibited excellent oral bioavailability in a mouse xenograft model, highlighting its potential for clinical application. In conclusion, our study presents a promising and novel therapeutic strategy for ROS1 fusion-positive NSCLC by targeting ROS1 fusion oncoproteins for degradation, laying the foundation for the development of further PROTAC and offering hope for patients with ROS1 fusion-positive NSCLC.
Collapse
Affiliation(s)
- Jiawen Yang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China; Shanghai Clinical Research and Trial Center, Shanghai 201210, China
| | - Yifan Wu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Qiaoliang Zhu
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xiaojuan Qu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Hongyue Ou
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Haixia Liu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China; School of Physical Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yongqi Wei
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Di Ge
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Chunlai Lu
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| | - Biao Jiang
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China; CAS Key Laboratory of Synthetic Chemistry of Natural Substances, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, China.
| | - Xiaoling Song
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China.
| |
Collapse
|
10
|
Xing S, Li D, Zhao Q. RPL22L1 is a novel biomarker for prognosis and immune infiltration in lung adenocarcinoma, promoting the growth and metastasis of LUAD cells by inhibiting the MDM2/P53 signaling pathway. Aging (Albany NY) 2024; 16:12392-12413. [PMID: 39207452 PMCID: PMC11424578 DOI: 10.18632/aging.206096] [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: 11/20/2023] [Accepted: 06/13/2024] [Indexed: 09/04/2024]
Abstract
The ribosomal protein L22-like1 (RPL22L1) is a constituent of the 60 S ribosomal subunit whose function in lung adenocarcinoma (LUAD) remains ambiguous. This study aims to elucidate the role of RPL22L1 in LUAD through a thorough analysis and experimental validation. Our findings indicate that RPL22L1 exhibits abnormal expression patterns in various cancer types, including LUAD. Moreover, a statistically significant association was observed between elevated levels of RPL22L1 expression in LUAD patients and several clinical parameters, such as pathological stage (p = 0.0083) and gender (p = 0.0038). The high expression of RPL22L1 in LUAD demonstrated a significant association with poorer overall survival (OS) (p = 0.005), progression-free survival (PFS) (p = 0.027), and disease-specific survival (p = 0.015). The expression of RPL22L1 in LUAD (p = 0.005) was identified as an independent prognostic factor. Additionally, RPL22L1 expression in LUAD was found to be correlated with immune infiltration, immune checkpoint genes, TMB/MSI, and mRNAsi. Notably, the expression of RPL22L1 exhibited significant negative correlations with 1-BET-762, Trametinib, and WZ3105 in LUAD. The RPL22L1 gene exhibited up-regulation in multiple individual cells of LUAD, leading to a comparatively shorter PFS in the RPL22L1 variant group as opposed to the RPL22L1 variant-free group in LUAD. Significantly increased expression of RPL22L1 was noted in LUAD cell lines, where it was found to enhance the growth and metastasis of LUAD cells by suppressing the MDM2/P53 signaling pathway. Therefore, RPL22L1 may serve as a promising prognostic biomarker and therapeutic target for patients with LUAD.
Collapse
Affiliation(s)
- Shigui Xing
- Department of Thoracic Surgery, Nanjing Gaochun People’s Hospital, Nanjing 211300, Jiangsu, China
| | - Dongbing Li
- Scientific Research Center, Beijing ChosenMed Clinical Laboratory Co., Ltd., Beijing 100176, China
| | - Qi Zhao
- Department of Pulmonary and Critical Care Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, Jiangsu, China
| |
Collapse
|
11
|
Wu Y, Yao N, Du B, Zhu Y, Ji X, Lv C, Lai J. Ribosomal protein L22 like 1: a promising biomarker for lung adenocarcinoma. J Cancer 2024; 15:2549-2560. [PMID: 38577587 PMCID: PMC10988297 DOI: 10.7150/jca.91759] [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: 11/01/2023] [Accepted: 01/15/2024] [Indexed: 04/06/2024] Open
Abstract
No studies have reported the effect of ribosomal protein L22 like 1 (RPL22L1) in lung adenocarcinoma (LUAD). Therefore, we aimed to systematically investigate the role of RPL22L1 in LUAD. The expression of RPL22L1 was analyzed using TCGA, GEO, TIMER, UALCAN databases, and validated by immunohistochemistry (IHC). Gene methylation analysis was performed using the UALCAN, GSCA and MethSurv databases. The immune infiltrates were investigated using the Single Sample Gene Set Enrichment Analysis (ssGSEA), TIMER database, and TISCH database. The results demonstrated that RPL22L1 was up-regulated in LUAD, and verified by IHC. Kaplan-Meier analysis suggested that patients with high RPL22L1 expression had poor prognosis. Multivariate analysis confirmed that RPL22L1 was an independent prognostic factor. Furthermore, RPL22L1 overexpression was associated with hypomethylation, and two CpGs of RPL22L1 were significantly associated with prognosis. Up-regulated RPL22L1 was enriched in MYC targets, E2F targets, G2M checkpoint, mTORC1 signaling, cell cycle, and so on. Moreover, RPL22L1 expression was negatively correlated with immune cell infiltration, and patients with high RPL22L1 expression had lower immune, stromal, and estimate scores. Single-cell analysis suggested that RPL22L1 might have a potential function in the tumor microenvironment (TME) of LUAD. In conclusion, RPL22L1 may be a promising biomarker for LUAD.
Collapse
Affiliation(s)
- Yahua Wu
- Department of Medical Oncology, Fujian Medical University Union Hospital, No. 29 Xinquan Street, Fuzhou, 350000, Fujian, China
| | - Na Yao
- Department of Medical Oncology, Fujian Medical University Union Hospital, No. 29 Xinquan Street, Fuzhou, 350000, Fujian, China
| | - Bin Du
- Department of Medical Oncology, Fujian Medical University Union Hospital, No. 29 Xinquan Street, Fuzhou, 350000, Fujian, China
| | - Yingjiao Zhu
- Department of Medical Oncology, Fujian Medical University Union Hospital, No. 29 Xinquan Street, Fuzhou, 350000, Fujian, China
| | - Xiaohui Ji
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Chengliu Lv
- Department of Medical Oncology, Fujian Medical University Union Hospital, No. 29 Xinquan Street, Fuzhou, 350000, Fujian, China
| | - Jinhuo Lai
- Department of Medical Oncology, Fujian Medical University Union Hospital, No. 29 Xinquan Street, Fuzhou, 350000, Fujian, China
| |
Collapse
|
12
|
Molinaro M, Torrente Y, Villa C, Farini A. Advancing Biomarker Discovery and Therapeutic Targets in Duchenne Muscular Dystrophy: A Comprehensive Review. Int J Mol Sci 2024; 25:631. [PMID: 38203802 PMCID: PMC10778889 DOI: 10.3390/ijms25010631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 12/25/2023] [Accepted: 12/28/2023] [Indexed: 01/12/2024] Open
Abstract
Mounting evidence underscores the intricate interplay between the immune system and skeletal muscles in Duchenne muscular dystrophy (DMD), as well as during regular muscle regeneration. While immune cell infiltration into skeletal muscles stands out as a prominent feature in the disease pathophysiology, a myriad of secondary defects involving metabolic and inflammatory pathways persist, with the key players yet to be fully elucidated. Steroids, currently the sole effective therapy for delaying onset and symptom control, come with adverse side effects, limiting their widespread use. Preliminary evidence spotlighting the distinctive features of T cell profiling in DMD prompts the immuno-characterization of circulating cells. A molecular analysis of their transcriptome and secretome holds the promise of identifying a subpopulation of cells suitable as disease biomarkers. Furthermore, it provides a gateway to unraveling new pathological pathways and pinpointing potential therapeutic targets. Simultaneously, the last decade has witnessed the emergence of novel approaches. The development and equilibrium of both innate and adaptive immune systems are intricately linked to the gut microbiota. Modulating microbiota-derived metabolites could potentially exacerbate muscle damage through immune system activation. Concurrently, genome sequencing has conferred clinical utility for rare disease diagnosis since innovative methodologies have been deployed to interpret the functional consequences of genomic variations. Despite numerous genes falling short as clinical targets for MD, the exploration of Tdark genes holds promise for unearthing novel and uncharted therapeutic insights. In the quest to expedite the translation of fundamental knowledge into clinical applications, the identification of novel biomarkers and disease targets is paramount. This initiative not only advances our understanding but also paves the way for the design of innovative therapeutic strategies, contributing to enhanced care for individuals grappling with these incapacitating diseases.
Collapse
Affiliation(s)
- Monica Molinaro
- Neurology Unit, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122 Milan, Italy; (M.M.); (Y.T.)
| | - Yvan Torrente
- Neurology Unit, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122 Milan, Italy; (M.M.); (Y.T.)
- Stem Cell Laboratory, Dino Ferrari Center, Department of Pathophysiology and Transplantation, University of Milan, 20100 Milan, Italy;
| | - Chiara Villa
- Stem Cell Laboratory, Dino Ferrari Center, Department of Pathophysiology and Transplantation, University of Milan, 20100 Milan, Italy;
| | - Andrea Farini
- Neurology Unit, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122 Milan, Italy; (M.M.); (Y.T.)
| |
Collapse
|
13
|
Zhang X, Yu T, Gao G, Xu J, Lin R, Pan Z, Liu J, Feng W. Cell division cycle 42 effector protein 4 inhibits prostate cancer progression by suppressing ERK signaling pathway. BIOMOLECULES & BIOMEDICINE 2023; 24:840-847. [PMID: 38153517 PMCID: PMC11293231 DOI: 10.17305/bb.2023.9986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 12/09/2023] [Accepted: 12/27/2023] [Indexed: 12/29/2023]
Abstract
Prostate cancer (PCa) is the most common malignancy among men worldwide. The cell division cycle 42 effector protein 4 (CDC42EP4) functions downstream of CDC42, yet its role and molecular mechanisms in PCa remain unexplored. This study aimed to elucidate the role of CDC42EP4 in the progression of PCa and its underlying mechanisms. Bioinformatical analysis indicated that CDC42EP4 expression was significantly lower in PCa tissue compared to normal prostate tissue. Cellular phenotyping analysis suggested that CDC42EP4 markedly inhibited the proliferation, migration, and invasion of PCa cells. Xenograft tumor assays further demonstrated that CDC42EP4 suppressed the growth of PCa cells in vivo. Mechanistically, the study established that CDC42EP4 inhibited the ERK pathway in PCa cells. Additionally, the ERK pathway inhibitor PD0325901 was employed, revealing that PD0325901 significantly nullified the effects of CDC42EP4 on PCa cell proliferation, migration, and invasion. Collectively, our findings demonstrate that CDC42EP4 acts as a critical tumor suppressor gene, inhibiting PCa cell proliferation, migration, and invasion through the ERK pathway, thereby presenting potential targets for PCa therapy.
Collapse
Affiliation(s)
- Xiaowen Zhang
- School of Life Science and Technology, Weifang Medical University, Weifang, China
| | - Tao Yu
- School of Life Science and Technology, Weifang Medical University, Weifang, China
| | - Guojun Gao
- Department of Urology Surgery, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Junbao Xu
- Cancer Center, Shandong Public Health Clinical Center, Shandong, China
| | - Ruihui Lin
- School of Life Science and Technology, Weifang Medical University, Weifang, China
| | - Zhifang Pan
- School of Life Science and Technology, Weifang Medical University, Weifang, China
| | - Jianying Liu
- Department of Nuclear Medicine, The Third Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Weiguo Feng
- School of Life Science and Technology, Weifang Medical University, Weifang, China
| |
Collapse
|
14
|
Jia W, Yuan J, Li S, Cheng B. The role of dysregulated mRNA translation machinery in cancer pathogenesis and therapeutic value of ribosome-inactivating proteins. Biochim Biophys Acta Rev Cancer 2023; 1878:189018. [PMID: 37944831 DOI: 10.1016/j.bbcan.2023.189018] [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: 09/14/2023] [Revised: 10/17/2023] [Accepted: 11/02/2023] [Indexed: 11/12/2023]
Abstract
Dysregulated protein synthesis is a hallmark of tumors. mRNA translation reprogramming contributes to tumorigenesis, which is fueled by abnormalities in ribosome formation, tRNA abundance and modification, and translation factors. Not only malignant cells but also stromal cells within tumor microenvironment can undergo transformation toward tumorigenic phenotypes during translational reprogramming. Ribosome-inactivating proteins (RIPs) have garnered interests for their ability to selectively inhibit protein synthesis and suppress tumor growth. This review summarizes the role of dysregulated translation machinery in tumor development and explores the potential of RIPs in cancer treatment.
Collapse
Affiliation(s)
- Wentao Jia
- Oncology Department of Traditional Chinese Medicine, the First Affiliated Hospital of Naval Medical University, Shanghai 200433, China; Faculty of Traditional Chinese Medicine, Naval Medical University, Shanghai 200043, China
| | - Jiaying Yuan
- Department of Pulmonary and Critical Care Medicine, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Shu Li
- Department of Gastroenterology, Baoshan Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201900, China.
| | - Binbin Cheng
- Oncology Department of Traditional Chinese Medicine, the First Affiliated Hospital of Naval Medical University, Shanghai 200433, China; Faculty of Traditional Chinese Medicine, Naval Medical University, Shanghai 200043, China.
| |
Collapse
|
15
|
Naik A, Dalpatraj N, Thakur N. Comparative analysis of the occupancy of Histone H3 Lysine 4 methylation in the cells treated with TGFβ and Interferonγ. Gene 2023:147601. [PMID: 37394048 DOI: 10.1016/j.gene.2023.147601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 06/21/2023] [Accepted: 06/29/2023] [Indexed: 07/04/2023]
Abstract
In this current study, we have compared our H3K4me3 Chip-Sequencing data in PC3 cells in response to 6h and 24h TGFβ stimulation with the IFNγ stimulated/unstimulated HeLa S3 cells Since both TGFβ and IFNγ play an essential role in tumorigenesis both as a tumor promoter and tumor suppressor and known to antagonize each other's signalling, it would be of utmost importance to find out the regions undergoing histone modification changes in response to TGFβ and IFNγ and compare them to explore the genes common to both as well as the specific for each ligand. Our study has compared the genes showing H3K4me3 occupancy in response to both TGFβ and IFNγ. Several genes were found to be shared between the TGFβ and IFNγ. DAVID Functional enrichment analysis in the TGFβ and IFNγ dataset revealed association of genes with different biological processes such as miRNA-mediated gene silencing, positive regulation of ERK cascade, hypoxia-induced apoptosis repression, translational regulation and molecular functions such as TGFβR activity, GPCR activity, TGFβ binding activity. Further analysis of these genes can reveal fascinating insights into epigenetic regulation by growth factor stimulation.
Collapse
Affiliation(s)
- Ankit Naik
- Biological and Life Sciences, School of Arts and Sciences, Ahmedabad University, Navrangpura, Ahmedabad-380009, Gujarat, India
| | - Nidhi Dalpatraj
- Biological and Life Sciences, School of Arts and Sciences, Ahmedabad University, Navrangpura, Ahmedabad-380009, Gujarat, India
| | - Noopur Thakur
- Biological and Life Sciences, School of Arts and Sciences, Ahmedabad University, Navrangpura, Ahmedabad-380009, Gujarat, India.
| |
Collapse
|