1
|
Bao L, Wu Y, Ren Z, Huang Y, Jiang Y, Li K, Xu X, Ye Y, Gui Z. Comprehensive pan-cancer analysis indicates UCHL5 as a novel cancer biomarker and promotes cervical cancer progression through the Wnt signaling pathway. Biol Direct 2024; 19:139. [PMID: 39702250 DOI: 10.1186/s13062-024-00588-6] [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: 10/25/2024] [Accepted: 12/11/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUND UCHL5 was initially recognized as a multifunctional molecule. While recent research has highlighted its involvement in tumor malignant biological behaviors, its specific role in promoting tumor cell apoptosis has drawn particular attention. However, the precise relationship between UCHL5 and various tumor types, as well as its influence within the immune microenvironment, remains unclear. METHODS The transcriptomic data and clinicopathological parameters across 33 cancer types were obtained from TCGA. Clinical pathological parameters of tumor patients, including gender, age, survival time, and staging, are utilized to evaluate the association between UCHL5 and pan-cancer characteristics. The prognostic significance of UCHL5 was evaluated through Cox analysis and Kaplan-Meier (K-M) methods. Protein expression data for UCHL5 were obtained from The Human Protein Atlas database, and its subcellular localization was further investigated. Additionally, potential correlations between UCHL5 and factors such as tumor-infiltrating immune cells, immunomodulators, microsatellite instability (MSI), and tumor mutation burden (TMB) were explored. The relationship between UCHL5 and immunotherapy efficacy was also assessed in independent cohorts, including IMvigor210, GSE78220, GSE67501, and GSE168204. Finally, the impact of UCHL5 on the malignant biological behavior of cervical cancer cells was investigated through in vitro experiments, along with an exploration of the underlying mechanisms. RESULTS We observed that UCHL5 expression levels were elevated in 11 types of cancer tissues compared to their corresponding normal tissues, while levels were lower in five tumor types. Additionally, UCHL5 expression displayed a significant correlation with tumor stage in BRCA, KIRC, LUAD, and TGCT. Cox and K-M analysis indicated that UCHL5 expression was significantly associated with prognosis in KIRC, KICH, CESC, ACC, and UVM. UCHL5 expression was negatively associated with stromal and immune scores in certain cancers. In terms of immune cell infiltration, UCHL5 expression in UCEC, SKCM, and COAD showed a negative correlation with regulatory T cells (Tregs). Furthermore, UCHL5 was widely associated with three types of immunomodulators. It also demonstrated a significant relationship with MSI and TMB in certain cancers and was connected to the immunotherapy efficacy. Finally, in vitro experiments confirmed that UCHL5 knockout enhances apoptosis in cervical cancer cells and disrupts Wnt/β-catenin signaling. CONCLUSIONS Pan-cancer analysis indicates that UCHL5 is dysregulated in various tumor tissues and is closely associated with survival prognosis, the tumor immune microenvironment, and the efficacy of immunotherapy in certain cancer types. UCHL5 shows promise as a predictive biomarker, and its specific regulatory mechanisms across different cancers warrant further investigation.
Collapse
Affiliation(s)
- Lingling Bao
- Department of Hematology and Oncology, Beilun Branch of the First Affiliated Hospital, College of Medicine, Zhejiang University, Ningbo, China
- Department of Hematology and Oncology, Beilun People's Hospital, Ningbo, China
| | - Yuefei Wu
- Radiotherapy Department, The Second People's Hospital of Wuhu, Wuhu, China
| | - Zhengting Ren
- Department of Radiation Oncology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yi Huang
- Oncology Department of Integrated Traditional Chinese and Western Medicine, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yue Jiang
- Oncology Department of Integrated Traditional Chinese and Western Medicine, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Kailang Li
- Department of Hematology and Oncology, Beilun Branch of the First Affiliated Hospital, College of Medicine, Zhejiang University, Ningbo, China
- Department of Hematology and Oncology, Beilun People's Hospital, Ningbo, China
| | - Xin Xu
- Department of Hematology and Oncology, Beilun Branch of the First Affiliated Hospital, College of Medicine, Zhejiang University, Ningbo, China.
- Department of Hematology and Oncology, Beilun People's Hospital, Ningbo, China.
| | - Yingquan Ye
- Oncology Department of Integrated Traditional Chinese and Western Medicine, First Affiliated Hospital of Anhui Medical University, Hefei, China.
| | - Zhongxuan Gui
- Oncology Department of Integrated Traditional Chinese and Western Medicine, First Affiliated Hospital of Anhui Medical University, Hefei, China.
| |
Collapse
|
2
|
Gong K, Zheng Y, Liu Y, Zhang T, Song Y, Chen W, Guo L, Zhou J, Liu W, Fang T, Chen Y, Wang J, Pan F, Shi K. Phosphocholine inhibits proliferation and reduces stemness of endometrial cancer cells by downregulating mTOR-c-Myc signaling. Cell Mol Life Sci 2024; 82:3. [PMID: 39680126 DOI: 10.1007/s00018-024-05517-4] [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: 08/03/2024] [Revised: 11/10/2024] [Accepted: 11/18/2024] [Indexed: 12/17/2024]
Abstract
BACKGROUND Endometrial cancer (EC) represents a serious health concern among women globally. Excessive activation of the protooncogene c-Myc (c-Myc) is associated with the proliferation and stemness of EC cells. Phosphocholine (PC), which is synthesized by choline kinase alpha (CHKA) catalysis, is upregulated in EC tumor tissues. The present study aimed to investigate the effect of PC accumulation on EC cells and clarify the relationship between PC accumulation and c-Myc activity in EC. METHODS The c-Myc and CHKA expression in EC tumor tissues were examined using immunohistochemistry. Cell Counting Kit-8 assay, colony formation assay, flow cytometry, western blotting, BrdU staining, and tumorsphere formation assay were used to assess the effect of PC accumulation on EC cells. The mechanism by which PC accumulation inhibits c-Myc was evaluated using RNA-sequencing. Patient-derived organoid (PDO) models were utilised to explore the preclinical efficacy of PC against EC cells. RESULTS PC accumulation suppressed EC cell proliferation and stemness by inhibiting the activation of the mammalian target of rapamycin (mTOR)-c-Myc signaling. PC accumulation promoted excessive reactive oxygen species production, which reduced the expression of GTPase HRAS. This, in turn, inhibited the mTOR-c-Myc axis and induced EC cell apoptosis. Finally, PC impeded proliferation and downregulated the expression of the mTOR-MYC signaling in EC PDO models. CONCLUSIONS PC accumulation impairs the proliferation ability and stem cell characteristics of EC cells by inhibiting the activated mTOR-c-Myc axis, potentially offering a promising strategy to enhance the efficacy of EC clinical therapy through the promotion of PC accumulation in tumor cells.
Collapse
Affiliation(s)
- Kunxiang Gong
- Department of Gynecology and Obstetrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
- Institute of Reproductive Health and Perinatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Yanqin Zheng
- Department of Gynecology and Obstetrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
- Institute of Reproductive Health and Perinatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Yaqiong Liu
- Department of Gynecology and Obstetrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
| | - Tiansong Zhang
- Department of Gynecology and Obstetrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
| | - Yiming Song
- Department of Gynecology and Obstetrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
- Institute of Reproductive Health and Perinatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Weiwei Chen
- Department of Gynecology and Obstetrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
| | - Lirong Guo
- Department of Gynecology and Obstetrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
- Institute of Reproductive Health and Perinatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Jie Zhou
- Department of Gynecology and Obstetrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
| | - Wenjie Liu
- Department of Gynecology and Obstetrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
| | - Tianlin Fang
- Department of Gynecology and Obstetrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
| | - Yun Chen
- Department of Gynecology and Obstetrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
- Institute of Reproductive Health and Perinatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Jingyao Wang
- Department of Gynecology and Obstetrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
- Institute of Reproductive Health and Perinatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Feifei Pan
- Department of Gynecology and Obstetrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
- Institute of Reproductive Health and Perinatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Kun Shi
- Department of Gynecology and Obstetrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China.
- Institute of Reproductive Health and Perinatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China.
| |
Collapse
|
3
|
Wu L, Wang J, Chai L, Chen J, Jin X. Roles of deubiquitinases in urologic cancers (Review). Oncol Lett 2024; 28:609. [PMID: 39525605 PMCID: PMC11544529 DOI: 10.3892/ol.2024.14743] [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: 07/23/2024] [Accepted: 09/23/2024] [Indexed: 11/16/2024] Open
Abstract
Human health is endangered by the occurrence and progression of urological cancers, including renal cell carcinoma, prostate cancer and bladder cancer, which are usually associated with the activation of oncogenic factors and inhibition of cancer suppressors. The primary mechanism for protein breakdown in cells is the ubiquitin-proteasome system, whilst deubiquitinases contribute to the reversal of this process. However, both are important for protein homeostasis. Deubiquitination may also be involved in the control of the cell cycle, proliferation and apoptosis, and dysregulated deubiquitination is associated with the malignant transformation, invasion and metastasis of urologic malignancies. Therefore, a comprehensive summary of the mechanisms underlying deubiquitination in urological cancers may provide novel strategies and insights for diagnosis and treatment. The present review aimed to methodically clarify the role of deubiquitinating enzymes in urinary system cancers as well as their prospective application prospects for clinical treatment.
Collapse
Affiliation(s)
- Liangpei Wu
- Department of Chemoradiotherapy, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang 315040, P.R. China
- Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Jiahui Wang
- Department of Chemoradiotherapy, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang 315040, P.R. China
- Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Lin Chai
- Department of Chemoradiotherapy, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang 315040, P.R. China
- Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Jun Chen
- Department of Chemoradiotherapy, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang 315040, P.R. China
| | - Xiaofeng Jin
- Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| |
Collapse
|
4
|
Hashemi M, Rezaei M, Rezaeiaghdam H, Jamali B, Koohpar ZK, Tanha M, Bizhanpour A, Asadi S, Jafari AM, Khosroshahi EM, Eslami M, Salimimoghadam S, Nabavi N, Rashidi M, Fattah E, Taheriazam A, Entezari M. Highlighting function of Wnt signalling in urological cancers: Molecular interactions, therapeutic strategies, and (nano)strategies. Transl Oncol 2024; 50:102145. [PMID: 39357465 PMCID: PMC11474201 DOI: 10.1016/j.tranon.2024.102145] [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: 10/25/2023] [Revised: 05/06/2024] [Accepted: 09/27/2024] [Indexed: 10/04/2024] Open
Abstract
Cancer is a complex, multistep process characterized by abnormal cell growth and metastasis as well as the capacity of the tumor cells in therapy resistance development. The urological system is particularly susceptible to a group of malignancies known as urological cancers, where an accumulation of genetic alterations drives carcinogenesis. In various human cancers, Wnt singalling is dysregulated; following nuclear transfer of β-catenin, it promotes tumor progression and affects genes expression. Elevated levels of Wnt have been documented in urological cancers, where its overexpression enhances growth and metastasis. Additionally, increased Wnt singalling contributes to chemoresistance in urological cancers, leading to reduced sensitivity to chemotherapy agents like cisplatin, doxorubicin, and paclitaxel. Wnt upregulation can change radiotherapy response of urological cancers. The regulation of Wnt involves various molecular pathways, including Akt, miRNAs, lncRNAs, and circRNAs, all of which play roles in carcinogenesis. Targeting and silencing Wnt or its associated pathways can mitigate tumorigenesis in urological cancers. Anti-cancer compounds such as curcumin and thymoquinone have shown efficacy in suppressing tumorigenesis through the downregulation of Wnt singalling. Notably, nanoparticles have proven effective in treating urological cancers, with several studies in prostate cancer (PCa) using nanoparticles to downregulate Wnt and suppress tumor growth. Future research should focus on developing small molecules that inhibit Wnt singalling to further suppress tumorigenesis and advance the treatment of urological cancers. Moreover, Wnt can be used as reliable biomarker for the diagnosis and prognosis of urological cancers.
Collapse
Affiliation(s)
- 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
| | - Mahdi Rezaei
- Health Research Center, Chamran Hospital, Tehran, Iran
| | - Hadi Rezaeiaghdam
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Behdokht Jamali
- Department of Microbiology and Genetics, Kherad Institute of Higher Education, Bushehr, Iran
| | - Zeinab Khazaei Koohpar
- Department Of Cell and Molecular Biology, Faculty of Biological Sciences,Tonekabon Branch, Islamic Azad University, Tonekabon, Iran
| | - Mahsa Tanha
- Department Of Biological Sciences, University Of Alabama, Tuscaloosa, Al, United States
| | - Anahita Bizhanpour
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Saba Asadi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Ali Moghadas Jafari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Elaheh Mohandesi Khosroshahi
- 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
| | - Maedeh Eslami
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Shokooh Salimimoghadam
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Noushin Nabavi
- Independent Researcher, Victoria, British Columbia, V8V 1P7, Canada
| | - Mohsen Rashidi
- The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari, Iran; Department Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Eisa Fattah
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Maliheh Entezari
- 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.
| |
Collapse
|
5
|
Ma XF, Liu AJ, Zheng Z, Hu BX, Zhi YX, Liu C, Tian SJ. Resolving and functional analysis of RNA editing sites in sheep ovaries and associations with litter size. Animal 2024; 18:101342. [PMID: 39471744 DOI: 10.1016/j.animal.2024.101342] [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: 12/05/2023] [Revised: 09/14/2024] [Accepted: 09/16/2024] [Indexed: 11/01/2024] Open
Abstract
Sheep litter size is a critical trait in mutton production. While litter size regulation in relation to DNA transcription have been rigorously investigated, the function of RNA editing remains less explored. To elucidate the mechanisms controlling sheep fecundity at the RNA editing level and identify pivotal RNA editing sites, this study scrutinised RNA editing sites (RESs) in follicular and luteal phases of ovaries from sheep with high and low fecundity, and the functions of population-specific RESs were subsequently analysed. A total of 2 182 475 RESs, 74.61% of which were A-to-I and C-to-U sites, were identified. These RESs were fairly evenly dispersed over the chromosomes, with 46.8% showing close clustering (inter-site distance < 300 bp). Notably, 93% were primarily situated in intronic and intergenic regions. In the follicular phase, pivotal RESs were found in the introns of genes including LPS responsive beige-like anchor, MCC regulator of Wnt signalling, and RWD domain containing 3, among others, and in the exon region of EvC ciliary complex subunit 2. In the luteal phase, RESs were observed in the introns of genes such as H/ACA ribonucleoprotein assembly factor and SDA1 domain-containing 1, and the exon and 3'UTR regions of polypeptide N-acetylgalactosaminyltransferase 15 and ilvB acetolactate synthase-like, respectively. High-fecundity sheep showed RESs in the follicular phase in genes such as fibrillin 1, cyclin-dependent kinase 6, and roundabout 1, and in genes such as autophagy-related 2B and versican in the luteal phase. Thirteen RESs specific to the follicular phase and eight specific to the luteal phase were identified in high-fecundity sheep ovaries. These RESs offer promising molecular targets and enhance understanding of multiple births in sheep from the perspective of posttranscriptional alterations.
Collapse
Affiliation(s)
- X F Ma
- College of Animal Science and Technology, Hebei Agricultural University, Hebei, Baoding 071000, China
| | - A J Liu
- Department of Agricultural and Animal Husbandry Engineering, Cangzhou Technical College, Hebei, Cangzhou, China
| | - Z Zheng
- College of Animal Science and Technology, Hebei Agricultural University, Hebei, Baoding 071000, China
| | - B X Hu
- College of Animal Science and Technology, Hebei Agricultural University, Hebei, Baoding 071000, China
| | - Y X Zhi
- College of Animal Science and Technology, Hebei Agricultural University, Hebei, Baoding 071000, China
| | - C Liu
- College of Animal Science and Technology, Hebei Agricultural University, Hebei, Baoding 071000, China
| | - S J Tian
- College of Animal Science and Technology, Hebei Agricultural University, Hebei, Baoding 071000, China; The Research Center of Cattle and Sheep Embryonic Technique of Hebei Province, Hebei, Baoding, 071000 Baoding, China.
| |
Collapse
|
6
|
Liu B, Zheng H, Ma G, Shen H, Pang Z, Huang G, Song Q, Wang G, Du J. Involvement of ICAM5 in Carcinostasis Effects on LUAD Based on the ROS1-Related Prognostic Model. J Inflamm Res 2024; 17:6583-6602. [PMID: 39318995 PMCID: PMC11421455 DOI: 10.2147/jir.s475088] [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: 06/25/2024] [Accepted: 09/07/2024] [Indexed: 09/26/2024] Open
Abstract
Background Lung cancer is the most common type of cancer in the world. In lung adenocarcinoma (LUAD), studies on receptor tyrosine kinase ROS proto-oncogene 1 (ROS1) have mainly focused on the oncogenic effects of its fusion mutations, whereas ROS1 has been reported to be aberrantly expressed in a variety of cancers and can extensively regulate the growth, survival, and proliferation of tumor cells through multiple signaling pathways. The comprehensive analysis of ROS1 expression has not been fully investigated regarding its predictive value for LUAD patients. Methods Gene expression profiles collected from The Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus (GEO) databases were used to build and validate prognostic risk models. The association of ROS1 with overall survival and the immune landscape was obtained from the Tumor Immune Estimation Resource (TIMER) database. The following analyses were performed using the R package to determine the model's validity: pathway dysregulation analysis, gene set enrichment analysis, Gene Oncology analysis, immune invasion analysis, chemotherapy, radiotherapy, and immunotherapy sensitivity analysis. Finally, we conducted a pan-cancer analysis and performed in vitro experiments to explore the regulatory role of intercellular adhesion molecule 5 (ICAM5) in the progression of LUAD. Results We constructed a 17-gene model that categorized patients into two risk groups. The model had predictive accuracy for tumor prognosis and was specific for patients with high ROS1 expression. Comprehensive analysis showed that patients in the high-risk group were characterized by marked dysregulation of multiple pathways (eg, unfolded protein response), immune suppression of the tumor microenvironment, and poor benefit from immunotherapy and radiotherapy compared with patients in the low-risk group. PLX4720 may be a suitable treatment for the high-risk patient population. The ICAM5 gene has been demonstrated to inhibit the proliferation, cell cycle, invasion, and migration of LUAD cells. Conclusion We constructed a 17-gene prognostic risk model and found differences in immune-related cells, biological processes, and prognosis among patients in different risk groups based on the correlation between ROS1 and immunity. Personalized therapy may play an essential role in treatment. We further investigated the role of ICAM5 in inhibiting the malignant bioactivity of LUAD cells.
Collapse
Affiliation(s)
- Baoliang Liu
- Department of Thoracic Surgery, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, People's Republic of China
| | - Haotian Zheng
- Department of Thoracic Surgery, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, People's Republic of China
| | - Guoyuan Ma
- Department of Thoracic Surgery, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, People's Republic of China
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People's Republic of China
| | - Hongchang Shen
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People's Republic of China
| | - Zhaofei Pang
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People's Republic of China
| | - Gemu Huang
- Research and Development Department, Amoy Diagnostics Co., LTD., Xiamen, Fujian, People's Republic of China
| | - Qingtao Song
- Research and Development Department, Amoy Diagnostics Co., LTD., Xiamen, Fujian, People's Republic of China
| | - Guanghui Wang
- Department of Thoracic Surgery, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, People's Republic of China
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People's Republic of China
| | - Jiajun Du
- Department of Thoracic Surgery, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, People's Republic of China
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People's Republic of China
| |
Collapse
|
7
|
Jiang J, Li X, Wang J, Chen S, Chen L. SLC25A19 drives colorectal cancer progression by regulating p53. Cancer Med 2024; 13:e70253. [PMID: 39344563 PMCID: PMC11440145 DOI: 10.1002/cam4.70253] [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: 01/24/2024] [Revised: 09/02/2024] [Accepted: 09/10/2024] [Indexed: 10/01/2024] Open
Abstract
BACKGROUND Investigating the molecular mechanism of colorectal cancer (CRC), a common lethal malignancies worldwide, is of great clinical significance. Solute carrier family 25 member 19 (SLC25A19) is a member of the solute carrier family that contribute to cellular functions, including tumor biology. Recently, many studies have attention on uncovering the relationship of SLC25A19 with malignant cancers, but its precise involvement in the regulation of CRC has not been thoroughly understood. This study sought to uncover the role and mechanism of SLC25A19 in CRC development. METHODS The GEPIA database and immunohistochemical staining were utilized to detect the expression of SLC25A19 in CRC tissues. The functional influences of SLC25A19 on CRC cell phenotypes were evaluated through a series of assays including celigo cell count, colony formation, CCK-8, flow cytometry, wound healing, and transwell assays following knocking down SLC25A19. Subsequently, the xenograft tumor model was constructed to evaluate the effect of SLC25A19 on tumor growth in vivo. The underlying mechanisms of SLC25A19 silencing were investigated using the human phospho-kinase array. RESULTS This study demonstrated the upregulation of SLC25A19 in CRC and its significant correlation with unfavorable prognosis in CRC patients. Suppression of SLC25A19 resulted in significant inhibition of cell proliferation, colony formation, and cell migration, alongside a boost in cell apoptosis. In vivo experiments revealed that silenced SLC25A19 displayed reduced growth rates and formed smaller xenografts. Mechanistically, the p53 pathway was found to be upregulated by SLC25A19 knockdown and mediated the function of SLC25A19. CONCLUSIONS Consequently, SLC25A19 was identified as a novel molecule with key regulatory ability in CRC development.
Collapse
Affiliation(s)
- Jinbo Jiang
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Xuemei Li
- Advanced Medical Research Institute, School of Medicine, Shandong University, Jinan, Shandong, China
| | - Jiayong Wang
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Shaofei Chen
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lingjuan Chen
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
8
|
Zhang M, Li J, Liu S, Zhou F, Zhang L. UCHL5 is a putative prognostic marker in renal cell carcinoma: a study of UCHL family. MOLECULAR BIOMEDICINE 2024; 5:28. [PMID: 39034372 PMCID: PMC11265068 DOI: 10.1186/s43556-024-00192-0] [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: 04/19/2024] [Accepted: 06/18/2024] [Indexed: 07/23/2024] Open
Abstract
A macroscopic perspective is indispensable for understanding the intricate relationship between deubiquitinases and tumorigenesis. Proteomics has been proposed as a viable approach for elucidating the complex role of deubiquitylation in cellular progression. Instead of studying the function of a single ubiquitinase, research on a deubiquitinase family with similar catalytic core(s) may provide a new perspective for the pathological understanding of cancer. The Ubiquitin C-terminal hydrolase L (UCHL) family consists of four members: UCHL1, UCHL3, UCHL5, and BRAC1 associated protein-1 (BAP1), and they have been implicated in tumorigenesis and metastasis. Some members are considered hallmarks of intracranial lesions, colon cancer, chromatin remodeling, and histone stability. The present study uncovered an unknown correlation between the UCHL family and renal cancer. We discovered that UCHLs exhibit diverse regulatory effects in renal cancer, establishing connections between the renal cancer and truncated gene mutations, mitochondrial energetic metastasis, immune cell infiltration, and chromosomal stability of UCHLs family. Notably, we found that the increase of UCHL5 expression in renal cancer cells decreases the antigen processing and presentation of RCC tumor-infiltrating B cells. Further research identified that the expression of UCHL5 in RCC tumors is correlated with transport proteins, which led us to find that the abundance of UCHL5 in the blood of late-stage renal cell cancer patients is upregulated from 18 ng/L to 500 ng/L. Therefore, we propose that the abundance of UCHL5 in patients' blood can be a possible indicator of poor prognosis for renal cell cancer.
Collapse
Affiliation(s)
- Mengdi Zhang
- Life Sciences Institute, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310058, PR China
| | - Jingxian Li
- Life Sciences Institute, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310058, PR China
| | - Sijia Liu
- International Biomed-X Research Center, Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310058, PR China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, 310058, PR China
| | - Fangfang Zhou
- The First Affiliated Hospital, the Institutes of Biology and Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, 215123, PR China
| | - Long Zhang
- Life Sciences Institute, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310058, PR China.
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China.
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310058, PR China.
| |
Collapse
|
9
|
Bogale DE. The roles of FGFR3 and c-MYC in urothelial bladder cancer. Discov Oncol 2024; 15:295. [PMID: 39031286 PMCID: PMC11264706 DOI: 10.1007/s12672-024-01173-z] [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: 04/24/2024] [Accepted: 07/16/2024] [Indexed: 07/22/2024] Open
Abstract
Bladder cancer is one of the most frequently occurring cancers worldwide. At diagnosis, 75% of urothelial bladder cancer cases have non-muscle invasive bladder cancer while 25% have muscle invasive or metastatic disease. Aberrantly activated fibroblast growth factor receptor (FGFR)-3 has been implicated in the pathogenesis of bladder cancer. Activating mutations of FGFR3 are observed in around 70% of NMIBC cases and ~ 15% of MIBCs. Activated FGFR3 leads to ligand-independent receptor dimerization and activation of downstream signaling pathways that promote cell proliferation and survival. FGFR3 is an important therapeutic target in bladder cancer, and clinical studies have shown the benefit of FGFR inhibitors in a subset of bladder cancer patients. c-MYC is a well-known major driver of carcinogenesis and is one of the most commonly deregulated oncogenes identified in human cancers. Studies have shown that the antitumor effects of FGFR inhibition in FGFR3 dependent bladder cancer cells and other FGFR dependent cancers may be mediated through c-MYC, a key downstream effector of activated FGFR that is involved tumorigenesis. This review will summarize the current general understanding of FGFR signaling and MYC alterations in cancer, and the role of FGFR3 and MYC dysregulation in the pathogenesis of urothelial bladder cancer with the possible therapeutic implications.
Collapse
Affiliation(s)
- Dereje E Bogale
- School of Medicine, Department of Oncology, Addis Ababa University, Addis Ababa, Ethiopia.
| |
Collapse
|
10
|
Hushmandi K, Saadat SH, Raei M, Daneshi S, Aref AR, Nabavi N, Taheriazam A, Hashemi M. Implications of c-Myc in the pathogenesis and treatment efficacy of urological cancers. Pathol Res Pract 2024; 259:155381. [PMID: 38833803 DOI: 10.1016/j.prp.2024.155381] [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: 02/20/2024] [Revised: 05/08/2024] [Accepted: 05/28/2024] [Indexed: 06/06/2024]
Abstract
Urological cancers, including prostate, bladder, and renal cancers, are significant causes of death and negatively impact the quality of life for patients. The development and progression of these cancers are linked to the dysregulation of molecular pathways. c-Myc, recognized as an oncogene, exhibits abnormal levels in various types of tumors, and current evidence supports the therapeutic targeting of c-Myc in cancer treatment. This review aims to elucidate the role of c-Myc in driving the progression of urological cancers. c-Myc functions to enhance tumorigenesis and has been documented to increase growth and metastasis in prostate, bladder, and renal cancers. Furthermore, the dysregulation of c-Myc can result in a diminished response to therapy in these cancers. Non-coding RNAs, β-catenin, and XIAP are among the regulators of c-Myc in urological cancers. Targeting and suppressing c-Myc therapeutically for the treatment of these cancers has been explored. Additionally, the expression level of c-Myc may serve as a prognostic factor in clinical settings.
Collapse
Affiliation(s)
- Kiavash Hushmandi
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Seyed Hassan Saadat
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mehdi Raei
- Health Research Center, Life Style Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran; Department of Epidemiology and Biostatistics, School of Health, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Salman Daneshi
- Department of Public Health,School of Health,Jiroft University Of Medical Sciences, Jiroft, Iran
| | - Amir Reza Aref
- Department of Translational Sciences, Xsphera Biosciences Inc. Boston, MA, USA; Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Noushin Nabavi
- Department of Urologic Sciences and Vancouver Prostate Centre, University of British Columbia, V6H3Z6, Vancouver, BC, Canada
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
11
|
Fu Q, Zheng H, Wang X, Tang F, Yu H, Wang H, Wan Z, Zheng Z, Yang Z, Liu T, Peng J. GINS1 promotes the initiation and progression of bladder cancer by activating the AKT/mTOR/c-Myc signaling pathway. Exp Cell Res 2024; 440:114125. [PMID: 38880324 DOI: 10.1016/j.yexcr.2024.114125] [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: 03/22/2024] [Revised: 05/21/2024] [Accepted: 06/07/2024] [Indexed: 06/18/2024]
Abstract
Bladder cancer(BC) is one of the most prevalent cancers in the urinary tract, with high recurrence and fatality rates. Research indicates that go-ichi-ni-san complex subunit 1 (GINS1) crucially influences cancer progression by regulating DNA replication through cell cycle modulation. Thus, suppressing the active proliferation of cells in tumor tissues may require silencing GINS1. However, the consequences of GINS1 in bladder cancer aren't to be determined. In this paper, we examine the role and mechanism of GINS1 in the development of bladder cancer. GINS1 expression levels and prognostic relevance in bladder cancer were validated using Western blotting, immunohistochemistry, and Kaplan-Meier survival analysis. The influence of GINS1 on bladder cancer was investigated using a variety of approaches, including cell transfection, cell counts, transwell migrations, colony formation, and flow cytometry. Immunohistochemistry studies demonstrate that GINS1 expression is increased in bladder cancer tissues. GINS1 silencing resulted in an arrest of the cell cycle at the phase of G0/G1, which inhibited BC cell growth both in vitro and in vivo. GINS1 knockdown also hindered the AKT/mTOR pathway. Furthermore, increased GINS1 expression affects the cell cycle and stimulates the AKT/mTOR pathway, allowing BC to develop more quickly. Consequently, GINS1 occurs as a latent therapeutic target, particularly for individuals with BC.
Collapse
Affiliation(s)
- Qiqi Fu
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China.
| | - Hang Zheng
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China.
| | - Xia Wang
- Department of Public Health, Wuhan University Hospital, Wuhan University, Wuhan, China.
| | - Feng Tang
- Department of Urology, Jingzhou Central Hospital, Jingzhou, China.
| | - Hua Yu
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| | - Hao Wang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China.
| | - Ziyu Wan
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China.
| | - Zhangjie Zheng
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China.
| | - Zhonghua Yang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China.
| | - Tao Liu
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China.
| | - Jianping Peng
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
12
|
Xu Z, Zhang N, Shi L. Potential roles of UCH family deubiquitinases in tumorigenesis and chemical inhibitors developed against them. Am J Cancer Res 2024; 14:2666-2694. [PMID: 39005671 PMCID: PMC11236784 DOI: 10.62347/oege2648] [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: 02/18/2024] [Accepted: 05/30/2024] [Indexed: 07/16/2024] Open
Abstract
Deubiquitinating enzymes (DUBs) are a large group of proteases that reverse ubiquitination process and maintain protein homeostasis. The DUBs have been classified into seven subfamilies according to their primary sequence and structural similarity. As a small subfamily of DUBs, the ubiquitin C-terminal hydrolases (UCHs) subfamily only contains four members including UCHL1, UCHL3, UCHL5, and BRCA1-associated protein-1 (BAP1). Despite sharing the deubiquitinase activity with a similar catalysis mechanism, the UCHs exhibit distinctive biological functions which are mainly determined by their specific subcellular localization and partner substrates. Besides, growing evidence indicates that the UCH enzymes are involved in human malignancies. In this review, the structural information and biological functions of the UCHs are briefly described. Meanwhile, the roles of these enzymes in tumorigenesis and the discovered inhibitors against them are also summarized to give an insight into the cancer therapy with the potential alternative strategy.
Collapse
Affiliation(s)
- Zhuo Xu
- State Key Laboratory of Chemical Biology, Analytical Research Center for Organic and Biological Molecules, Shanghai Institute of Materia Medica, Chinese Academy of Sciences555 Zu Chong Zhi Road, Shanghai 201203, China
- University of The Chinese Academy of Sciences19A Yuquan Road, Beijing 100049, China
| | - Naixia Zhang
- State Key Laboratory of Chemical Biology, Analytical Research Center for Organic and Biological Molecules, Shanghai Institute of Materia Medica, Chinese Academy of Sciences555 Zu Chong Zhi Road, Shanghai 201203, China
- University of The Chinese Academy of Sciences19A Yuquan Road, Beijing 100049, China
| | - Li Shi
- State Key Laboratory of Chemical Biology, Analytical Research Center for Organic and Biological Molecules, Shanghai Institute of Materia Medica, Chinese Academy of Sciences555 Zu Chong Zhi Road, Shanghai 201203, China
| |
Collapse
|
13
|
Hao S, Shen L, Liu P, Yong Q, Wang Y, Zheng X. Development of a prognostic model for muscle-invasive bladder cancer using glutamine metabolism. Comput Biol Med 2024; 171:108223. [PMID: 38430744 DOI: 10.1016/j.compbiomed.2024.108223] [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: 11/07/2023] [Revised: 01/31/2024] [Accepted: 02/25/2024] [Indexed: 03/05/2024]
Abstract
BACKGROUND Muscle-invasive bladder cancer (MIBC) is distinguished by its pronounced invasiveness and unfavorable prognosis. Immunotherapy and targeted therapy have emerged as key treatment options for various types of cancer. Altered metabolism is a defining characteristic of cancer cells, and there is mounting evidence suggesting the important role of glutamine metabolism (GM) in tumor metabolism. Nevertheless, the relationship between GM and clinical outcomes, immune microenvironment, and immunotherapy in MIBC remains unknown. METHODS This study employed Mendelian randomization to explore the causal relationship between blood metabolites and bladder tumors. We systematically evaluated 373 glutamine metabolism-related genes and identified prognostic-related genes, leading to the construction of a glutamine-associated prognostic model. Further analysis confirmed the correlation between high and low-risk groups with the tumor microenvironment, immune cell infiltration, and tumor mutation burden. Subsequently, we assessed the relationship between the risk score and the sensitivity to various immunotherapies and anticancer drugs. RESULTS We identified 14 blood metabolites at the molecular level that have a causal relationship with bladder tumors. At the gene level, the study discussed differentially expressed GM genes in MIBC. First, we established a risk model predicting overall survival (OS) based on GM genes, confirming its reliable predictive ability in MIBC patients and validated it in a GEO cohort. Additionally, a reliable column line chart was created. Secondly, two distinct molecular subtypes were identified, and the associations between different risk groups and tumor microenvironment and immune infiltration were observed. In addition, the predicted risk values correlated with responses to a broad range of pharmaceutical agents. CONCLUSION In summary, we confirmed the causal relationship between blood metabolites and bladder tumors. Furthermore, a risk scoring model related to glutamine metabolism consisting of 9 genes was developed. This model could potentially serve as a useful tool for predicting prognosis and guiding the treatment of MIBC patients.
Collapse
Affiliation(s)
- Sida Hao
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, Zhejiang, China; Department of Urology, Zhejiang Integrated Traditional Chinese and Western Medicine Hospital, Hangzhou, 310003, Zhejiang, China.
| | - Lin Shen
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, Zhejiang, China.
| | - Pengju Liu
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, Zhejiang, China.
| | - Qin Yong
- Department of Urology, Zhejiang Integrated Traditional Chinese and Western Medicine Hospital, Hangzhou, 310003, Zhejiang, China.
| | - Yeqiang Wang
- Department of Urology, Zhejiang Integrated Traditional Chinese and Western Medicine Hospital, Hangzhou, 310003, Zhejiang, China.
| | - Xiangyi Zheng
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, Zhejiang, China.
| |
Collapse
|
14
|
Li Z, Ying Y, Zeng X, Liu J, Xie Y, Deng Z, Hu Z, Yang J. DNMT1/DNMT3a-mediated promoter hypermethylation and transcription activation of ICAM5 augments thyroid carcinoma progression. Funct Integr Genomics 2024; 24:12. [PMID: 38228798 DOI: 10.1007/s10142-024-01293-3] [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: 09/08/2023] [Revised: 01/03/2024] [Accepted: 01/05/2024] [Indexed: 01/18/2024]
Abstract
Promoter methylation is one of the most studied epigenetic modifications and it is highly relevant to the onset and progression of thyroid carcinoma (THCA). This study investigates the promoter methylation and expression pattern of intercellular adhesion molecule 5 (ICAM5) in THCA. CpG islands with aberrant methylation pattern in THCA, and the expression profiles of the corresponding genes in THCA, were analyzed using bioinformatics. ICAM5 was suggested to have a hypermethylation status, and it was highly expressed in THCA tissues and cells. Its overexpression promoted proliferation, mobility, and tumorigenic activity of THCA cells. As for the downstream signaling, ICAM5 was found to activate the MAPK/ERK and MAPK/JNK signaling pathways. Either inhibition of ERK or JNK blocked the oncogenic effects of ICAM5. DNA methyltransferases 1 (DNMT1) and DNMT3a were found to induce promoter hypermethylation of ICAM5 in THCA cells. Knockdown of DNMT1 or DNMT3a decreased the ICAM5 expression and suppressed malignant properties of THCA cells in vitro and in vivo, which were, however, restored by further artificial ICAM5 overexpression. Collectively, this study reveals that DNMT1 and DNMT3a mediates promoter hypermethylation and transcription activation of ICAM5 in THCA, which promotes malignant progression of THCA through the MAPK signaling pathway.
Collapse
Affiliation(s)
- Zanbin Li
- Department of Thyroid and Hernia Surgery, First Affiliated Hospital of Gannan Medical College, No. 128, Jinling West Road, Ganzhou, 341000, Jiangxi, People's Republic of China
| | - Yong Ying
- Department of Thyroid and Hernia Surgery, First Affiliated Hospital of Gannan Medical College, No. 128, Jinling West Road, Ganzhou, 341000, Jiangxi, People's Republic of China
| | - Xiangtai Zeng
- Department of Thyroid and Hernia Surgery, First Affiliated Hospital of Gannan Medical College, No. 128, Jinling West Road, Ganzhou, 341000, Jiangxi, People's Republic of China
| | - Jiafeng Liu
- Department of Thyroid and Hernia Surgery, First Affiliated Hospital of Gannan Medical College, No. 128, Jinling West Road, Ganzhou, 341000, Jiangxi, People's Republic of China
| | - Yang Xie
- Department of Thyroid and Hernia Surgery, First Affiliated Hospital of Gannan Medical College, No. 128, Jinling West Road, Ganzhou, 341000, Jiangxi, People's Republic of China
| | - Zefu Deng
- Department of Thyroid and Hernia Surgery, First Affiliated Hospital of Gannan Medical College, No. 128, Jinling West Road, Ganzhou, 341000, Jiangxi, People's Republic of China
| | - Zhiqiang Hu
- Department of Thyroid and Hernia Surgery, First Affiliated Hospital of Gannan Medical College, No. 128, Jinling West Road, Ganzhou, 341000, Jiangxi, People's Republic of China
| | - Junjie Yang
- Department of Thyroid and Hernia Surgery, First Affiliated Hospital of Gannan Medical College, No. 128, Jinling West Road, Ganzhou, 341000, Jiangxi, People's Republic of China.
| |
Collapse
|
15
|
Wang M, Zhang Z, Li Z, Zhu Y, Xu C. E3 ubiquitin ligases and deubiquitinases in bladder cancer tumorigenesis and implications for immunotherapies. Front Immunol 2023; 14:1226057. [PMID: 37497216 PMCID: PMC10366618 DOI: 10.3389/fimmu.2023.1226057] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Accepted: 06/23/2023] [Indexed: 07/28/2023] Open
Abstract
With the rapidly increasing incidence of bladder cancer in China and worldwide, great efforts have been made to understand the detailed mechanism of bladder cancer tumorigenesis. Recently, the introduction of immune checkpoint inhibitor-based immunotherapy has changed the treatment strategy for bladder cancer, especially for advanced bladder cancer, and has improved the survival of patients. The ubiquitin-proteasome system, which affects many biological processes, plays an important role in bladder cancer. Several E3 ubiquitin ligases and deubiquitinases target immune checkpoints, either directly or indirectly. In this review, we summarize the recent progress in E3 ubiquitin ligases and deubiquitinases in bladder cancer tumorigenesis and further highlight the implications for bladder cancer immunotherapies.
Collapse
Affiliation(s)
- Maoyu Wang
- Department of Urology, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Zhensheng Zhang
- Department of Urology, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Zhizhou Li
- Department of Urology, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Yasheng Zhu
- Department of Urology, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
- Department of Urology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Chuanliang Xu
- Department of Urology, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
16
|
Kim YJ, Lee Y, Shin H, Hwang S, Park J, Song EJ. Ubiquitin-proteasome system as a target for anticancer treatment-an update. Arch Pharm Res 2023; 46:573-597. [PMID: 37541992 DOI: 10.1007/s12272-023-01455-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 07/05/2023] [Indexed: 08/06/2023]
Abstract
As the ubiquitin-proteasome system (UPS) regulates almost every biological process, the dysregulation or aberrant expression of the UPS components causes many pathological disorders, including cancers. To find a novel target for anticancer therapy, the UPS has been an active area of research since the FDA's first approval of a proteasome inhibitor bortezomib in 2003 for treating multiple myeloma (MM). Here, we summarize newly described UPS components, including E3 ubiquitin ligases, deubiquitinases (DUBs), and immunoproteasome, whose malfunction leads to tumorigenesis and whose inhibitors have been investigated in clinical trials as anticancer therapy since 2020. We explain the mechanism and effects of several inhibitors in depth to better comprehend the advantages of targeting UPS components for cancer treatment. In addition, we describe attempts to overcome resistance and limited efficacy of some launched proteasome inhibitors, as well as an emerging PROTAC-based tool targeting UPS components for anticancer therapy.
Collapse
Affiliation(s)
- Yeon Jung Kim
- College of Pharmacy, Ewha Womans University, 52, Ewhayeodae-gil, Seodaemun-gu, Seoul, 03760, Republic of Korea
| | - Yeonjoo Lee
- College of Pharmacy, Ewha Womans University, 52, Ewhayeodae-gil, Seodaemun-gu, Seoul, 03760, Republic of Korea
| | - Hyungkyung Shin
- College of Pharmacy, Ewha Womans University, 52, Ewhayeodae-gil, Seodaemun-gu, Seoul, 03760, Republic of Korea
| | - SuA Hwang
- College of Pharmacy, Ewha Womans University, 52, Ewhayeodae-gil, Seodaemun-gu, Seoul, 03760, Republic of Korea
| | - Jinyoung Park
- Center for Advanced Biomolecular Recognition, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Division of Bio‑Medical Science and Technology, KIST‑School, University of Science and Technology (UST), Seoul, 02792, Republic of Korea
| | - Eun Joo Song
- College of Pharmacy, Ewha Womans University, 52, Ewhayeodae-gil, Seodaemun-gu, Seoul, 03760, Republic of Korea.
| |
Collapse
|
17
|
Nakamura K, Asanuma K, Okamoto T, Iino T, Hagi T, Nakamura T, Sudo A. Combination of Everolimus and Bortezomib Inhibits the Growth and Metastasis of Bone and Soft Tissue Sarcomas via JNK/p38/ERK MAPK and AKT Pathways. Cancers (Basel) 2023; 15:cancers15092468. [PMID: 37173935 PMCID: PMC10177427 DOI: 10.3390/cancers15092468] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 04/20/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
The combination of the mammalian target of rapamycin and proteasome inhibitors is a new treatment strategy for various tumors. Herein, we investigated the synergistic effect of everolimus and bortezomib on tumor growth and metastasis in bone and soft tissue sarcomas. The antitumor effects of everolimus and bortezomib were assessed in a human fibrosarcoma (FS) cell line (HT1080) and mouse osteosarcoma (OS) cell line (LM8) by MTS assays and Western blotting. The effects of everolimus and bortezomib on HT1080 and LM8 tumor growth in xenograft mouse models were evaluated using tumor volume and the number of metastatic nodes of the resected lungs. Immunohistochemistry was used to evaluate cleaved PARP expression. The combination therapy decreased FS and OS cell proliferation compared with either drug alone. This combination induced more intense p-p38, p-JNK, and p-ERK and activated apoptosis signals, such as caspase-3, compared with single-agent treatment. The combination treatment reduced p-AKT and MYC expression, decreased FS and OS tumor volumes, and suppressed lung metastases of OS. The combination therapy inhibited tumor growth in FS and OS and metastatic progression of OS via the JNK/p38/ERK MAPK and AKT pathways. These results could aid in the development of new therapeutic strategies for sarcomas.
Collapse
Affiliation(s)
- Koichi Nakamura
- Department of Orthopaedic Surgery, Mie University Graduate School of Medicine, Tsu 514-0001, Japan
| | - Kunihiro Asanuma
- Department of Orthopaedic Surgery, Mie University Graduate School of Medicine, Tsu 514-0001, Japan
| | - Takayuki Okamoto
- Department of Pharmacology, Faculty of Medicine, Shimane University, Izumo 693-8501, Japan
| | - Takahiro Iino
- Department of Orthopaedic Surgery, Mie University Graduate School of Medicine, Tsu 514-0001, Japan
| | - Tomohito Hagi
- Department of Orthopaedic Surgery, Mie University Graduate School of Medicine, Tsu 514-0001, Japan
| | - Tomoki Nakamura
- Department of Orthopaedic Surgery, Mie University Graduate School of Medicine, Tsu 514-0001, Japan
| | - Akihiro Sudo
- Department of Orthopaedic Surgery, Mie University Graduate School of Medicine, Tsu 514-0001, Japan
| |
Collapse
|