1
|
Li H, Zhang C, Zhu N, Shi Y, Qin L. Sensitivity of renal cell carcinoma to cuproptosis: a bioinformatics analysis and experimental verification. J Cancer 2025; 16:952-968. [PMID: 39781354 PMCID: PMC11705067 DOI: 10.7150/jca.103708] [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: 09/15/2024] [Accepted: 12/10/2024] [Indexed: 01/12/2025] Open
Abstract
Purpose: Targeting cuproptosis is considered as a promising therapeutic strategy for the prevention of tumors. However, the potential role of cuproptosis and its related genes in clear cell renal cell carcinoma (ccRCC) remains elusive. The present study aims to explore the sensitivity of ccRCC to cuproptosis and its underlying mechanism. Methods: Cuproptosis differential genes (CDGs) were extracted using the GSE53757 and GSE66272 datasets. A comprehensive analysis of the role of CDGs was conducted through multiple public databases and experiments. Results: It was found that cuproptosis inducer elesclomol significantly induced cell death in 786-O and A498 cells. FDX and DLAT exhibited significantly low expression, which were independent prognostic factors for poor survival, and had a strong positive correlation in ccRCC patients. Functional analysis of differentially expressed genes positively or negatively correlated with both FDX1 and DLAT indicated that acetyl-CoA biosynthetic process and acetyl-CoA metabolic process were remarkably affected. In ccRCC patients, the methylation levels and sites of FDX1 and DLAT genes were dramatically correlated with overall survival (OS). The expressions of FDX1 and DLAT were closely related to immune infiltration and immune checkpoints. Docking results indicated that mitotane, adicicol and dihydrolipoic acid might be potential drug targets for FDX1 and DLAT. Conclusions: Overall, the present study demonstrates the sensitivity of ccRCC to cuproptosis, and targeting the combination of FDX1 and DLAT may be a novel therapeutic strategy to induce cuproptosis in ccRCC.
Collapse
Affiliation(s)
- Hongfang Li
- Laboratory of Stem Cell Regulation with Chinese Medicine and Its Application, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Chanjuan Zhang
- Laboratory of Stem Cell Regulation with Chinese Medicine and Its Application, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Neng Zhu
- Department of Urology, The First Hospital of Hunan University of Chinese Medicine, Changsha 410007, China
| | - Yaning Shi
- Laboratory of Stem Cell Regulation with Chinese Medicine and Its Application, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Li Qin
- Laboratory of Stem Cell Regulation with Chinese Medicine and Its Application, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
- Institutional Key Laboratory of Vascular Biology and Translational Medicine in Hunan Province, Changsha, China
- Hunan Province Engineering Research Center of Bioactive Substance Discovery of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
2
|
Xu Y, Shen Y, Zhang C, Zheng L, Ji F, Chen J, Cheng S, Zheng Y. Exploring the Effect of Fidgetin-Like 1 on Colorectal Cancer Through Tissue Chip and In Vitro Experiments. Balkan Med J 2024; 41:491-498. [PMID: 39319820 PMCID: PMC11589218 DOI: 10.4274/balkanmedj.galenos.2024.2024-7-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 08/22/2024] [Indexed: 09/26/2024] Open
Abstract
Background Fidgetin-like 1 (FIGNL1) is extensively overexpressed in a variety of cancers. It facilitates non‑small cell lung cancer tumor cell proliferation and hepatocellular carcinoma formation due to abnormal DNA repair. Clinically relevant data indicates that its high expression is linked with the poor prognosis of patients with renal clear-cell carcinoma, low-grade gliomas, and hepatocellular carcinoma. Nevertheless, the scope of FIGNL1’s involvement in cancer, particularly colorectal cancer (CRC), remains unclear. Aims To investigate the function of FIGNL1 in CRC. Study Design Cell culture study. Methods The TCGA database and immunohistochemistry analysis were employed to investigate FIGNL1 expression in CRC tissue. A cell viability assay was performed using the Cell Counting Kit-8. The cell migration and invasion were evaluated using the transwell assay. Small interfering RNA (siRNA) transfection was conducted to knockdown FIGNL1 expression. Infection with FIGNL1 overexpression lentivirus was performed to promote FIGNL1 overexpression. The STRING database was employed for predicting protein interaction. Results FIGNL1 was substantially upregulated in human CRC tissues and was associated with TNM stages and lymph node metastasis in patients. The inhibition of CRC cell proliferation, migration, and invasion in Caco-2 cells was achieved by silencing FIGNL1 using siRNA. Additional investigations suggested that FIGNL1 overexpression could promote CRC cell proliferation, migration, and invasion via P38 signaling pathway activation in Colo-205 cells. Subsequent experiments demonstrated that FIGNL1-mediated P38 phosphorylation was contingent upon SPIDR interaction. Conclusion These results implied that FIGNL1 was a potential anticancer drug target, which also offered a novel strategy for future CRC treatment.
Collapse
Affiliation(s)
- Yunxing Xu
- Clinic of Central Laboratory, Hai’an City People’s Hospital of Jiangsu Province, Hai’an Hospital Affiliated to Nantong University, Nantong, China
| | - Yucheng Shen
- Clinic of Central Laboratory, Hai’an City People’s Hospital of Jiangsu Province, Hai’an Hospital Affiliated to Nantong University, Nantong, China
| | - Chen Zhang
- Clinic of Central Laboratory, Hai’an City People’s Hospital of Jiangsu Province, Hai’an Hospital Affiliated to Nantong University, Nantong, China
| | - Liangfeng Zheng
- Clinic of Central Laboratory, Hai’an City People’s Hospital of Jiangsu Province, Hai’an Hospital Affiliated to Nantong University, Nantong, China
| | - Feiyue Ji
- Clinic of Central Laboratory, Hai’an City People’s Hospital of Jiangsu Province, Hai’an Hospital Affiliated to Nantong University, Nantong, China
| | - Jin Chen
- Clinic of Central Laboratory, Hai’an City People’s Hospital of Jiangsu Province, Hai’an Hospital Affiliated to Nantong University, Nantong, China
| | - Shouliang Cheng
- Clinic of Central Laboratory, Hai’an City People’s Hospital of Jiangsu Province, Hai’an Hospital Affiliated to Nantong University, Nantong, China
| | - Yu Zheng
- Clinic of Central Laboratory, Hai’an City People’s Hospital of Jiangsu Province, Hai’an Hospital Affiliated to Nantong University, Nantong, China
| |
Collapse
|
3
|
Smart K, Sharp DJ. The fidgetin family: Shaking things up among the microtubule-severing enzymes. Cytoskeleton (Hoboken) 2024; 81:151-166. [PMID: 37823563 DOI: 10.1002/cm.21799] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 09/23/2023] [Accepted: 09/25/2023] [Indexed: 10/13/2023]
Abstract
The microtubule cytoskeleton is required for several crucial cellular processes, including chromosome segregation, cell polarity and orientation, and intracellular transport. These functions rely on microtubule stability and dynamics, which are regulated by microtubule-binding proteins (MTBPs). One such type of regulator is the microtubule-severing enzymes (MSEs), which are ATPases Associated with Diverse Cellular Activities (AAA+ ATPases). The most recently identified family are the fidgetins, which contain three members: fidgetin, fidgetin-like 1 (FL1), and fidgetin-like 2 (FL2). Of the three known MSE families, the fidgetins have the most diverse range of functions in the cell, spanning mitosis/meiosis, development, cell migration, DNA repair, and neuronal function. Furthermore, they offer intriguing novel therapeutic targets for cancer, cardiovascular disease, and wound healing. In the two decades since their first report, there has been great progress in our understanding of the fidgetins; however, there is still much left unknown about this unusual family. This review aims to consolidate the present body of knowledge of the fidgetin family of MSEs and to inspire deeper exploration into the fidgetins and the MSEs as a whole.
Collapse
Affiliation(s)
- Karishma Smart
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - David J Sharp
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, USA
- Microcures, Inc., Bronx, New York, USA
| |
Collapse
|
4
|
Mannar D, Ahmed S, Subramaniam S. AAA ATPase protein-protein interactions as therapeutic targets in cancer. Curr Opin Cell Biol 2024; 86:102291. [PMID: 38056141 DOI: 10.1016/j.ceb.2023.102291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 10/30/2023] [Accepted: 11/09/2023] [Indexed: 12/08/2023]
Abstract
AAA ATPases are a conserved group of enzymes that couple ATP hydrolysis to diverse activities critical for cellular homeostasis by targeted protein-protein interactions. Some of these interactions are potential therapeutic targets because of their role in cancers which rely on increased AAA ATPase activities for maintenance of genomic stability. Two well-characterized members of this family are p97/VCP and RUVBL ATPases where there is a growing understanding of their structure and function, as well as an emerging landscape of selective inhibitors. Here we highlight recent progress in this field, with particular emphasis on structural advances enabled by cryo-electron microscopy (cryo-EM).
Collapse
Affiliation(s)
- Dhiraj Mannar
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Sana Ahmed
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Sriram Subramaniam
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; Gandeeva Therapeutics, Inc., Burnaby, BC V5C 6N5, Canada.
| |
Collapse
|
5
|
Wang J, Sun L, Liu Y, Zhang Y. FIGNL1 Promotes Hepatocellular Carcinoma Formation via Remodeling ECM-receptor Interaction Pathway Mediated by HMMR. Curr Gene Ther 2024; 24:249-263. [PMID: 37929733 PMCID: PMC11071652 DOI: 10.2174/0115665232274223231017052707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 09/02/2023] [Accepted: 09/03/2023] [Indexed: 11/07/2023]
Abstract
BACKGROUND The development of novel biomarkers is crucial for the treatment of HCC. In this study, we investigated a new molecular therapeutic target for HCC. Fidgetin-like 1 (FIGNL1) has been reported to play a vital role in lung adenocarcinoma. However, the potential function of FIGNL1 in HCC is still unknown. OBJECTIVE This study aims to investigate the key regulatory mechanisms of FIGNL1 in the formation of HCC. METHODS The regulatory effect of FIGNL1 on HCC was studied by lentivirus infection. In vitro, the effects of FIGNL1 on the proliferation, migration and apoptosis of cells were investigated by CCK8, colony formation assay, transwell and flow cytometry. Meanwhile, the regulation of FIGNL1 on HCC formation in vivo was studied by subcutaneous transplanted tumors. In addition, using transcriptome sequencing technology, we further explored the specific molecular mechanism of FIGNL1 regulating the formation of HCC. RESULTS Functionally, we demonstrated that FIGNL1 knockdown significantly inhibited HCC cell proliferation, migration and promoted cell apoptosis in vitro. Similarly, the knockdown of FIGNL1 meaningfully weakened hepatocarcinogenesis in nude mice. Transcriptome sequencing revealed that FIGNL1 affected the expression of genes involved in extracellular matrix-receptor (ECM-receptor) interaction pathway, such as hyaluronan mediated motility receptor (HMMR). Further validation found that overexpression of HMMR based on knockdown FIGNL1 can rescue the expression abundance of related genes involved in the ECM-receptor interaction pathway. CONCLUSION Our study revealed that FIGNL1 could modulate the ECM-receptor interaction pathway through the regulation of HMMR, thus regulating the formation of HCC.
Collapse
Affiliation(s)
- Jiabei Wang
- Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Department of Hepatobiliary Surgery, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230001, China
| | - Linmao Sun
- Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Department of Hepatobiliary Surgery, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230001, China
| | - Yao Liu
- Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Department of Hepatobiliary Surgery, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230001, China
| | - Yunguang Zhang
- Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Department of Hepatobiliary Surgery, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230001, China
| |
Collapse
|
6
|
Kwon JT, Kim Y, Choi S, Yoon BL, Kim HS, Shim I, Sul D. Pulmonary Toxicity and Proteomic Analysis in Bronchoalveolar Lavage Fluids and Lungs of Rats Exposed to Copper Oxide Nanoparticles. Int J Mol Sci 2022; 23:13265. [PMID: 36362054 PMCID: PMC9655042 DOI: 10.3390/ijms232113265] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 10/14/2022] [Accepted: 10/25/2022] [Indexed: 07/21/2023] Open
Abstract
Copper oxide nanoparticles (CuO NPs) were intratracheally instilled into lungs at concentrations of 0, 0.15, and 1.5 mg/kg bodyweight to 7-week-old Sprague-Dawley rats. The cytotoxicity, immunotoxicity, and oxidative stress were evaluated, followed by proteomic analysis of bronchoalveolar lavage fluid (BALF) and lungs of rats. The CuO NPs-exposed groups revealed dose-dependent increases in total cells, polymorphonuclear leukocytes, lactate dyhydrogenase, and total protein levels in BALF. Inflammatory cytokines, including macrophage inflammatory protein-2 and tumor necrosis factor-α, were increased in the CuO NPs-treated groups. The expression levels of catalase, glutathione peroxidase-1, and peroxiredoxin-2 were downregulated, whereas that of superoxide dismutase-2 was upregulated in the CuO NPs-exposed groups. Five heat shock proteins were downregulated in rats exposed to high concentrations of CuO NPs. In proteomic analysis, 17 proteins were upregulated or downregulated, and 6 proteins were validated via Western blot analysis. Significant upregulation of 3-hydroxy-3-methylglutaryl-CoA synthase and fidgetin-like 1 and downregulation of annexin II, HSP 47 and proteasome α1 occurred in the CuO NPs exposed groups. Taken together, this study provides additional insight into pulmonary cytotoxicity and immunotoxicity as well as oxidative stress in rats exposed to CuO NPs. Proteomic analysis revealed potential toxicological biomarkers of CuO NPs, which also reveals the toxicity mechanisms of CuO NPs.
Collapse
Affiliation(s)
- Jung-Taek Kwon
- Environmental Health Research Department, National Institute of Environmental Research, Incheon 22689, Korea
| | - Yoonjin Kim
- Environmental Health Research Department, National Institute of Environmental Research, Incheon 22689, Korea
| | - Seonyoung Choi
- Graduate School of Medicine, Korea University, 73 Inchon-ro, Sungbuk-ku, Seoul 136-705, Korea
| | - Byung-ll Yoon
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon 24341, Korea
| | - Hyun-Sook Kim
- Department of Biomedical Laboratory Science, College of Health Science, Cheongju University, Cheongju 28503, Korea
| | - Ilseob Shim
- Environmental Health Research Department, National Institute of Environmental Research, Incheon 22689, Korea
| | - Donggeun Sul
- Graduate School of Medicine, Korea University, 73 Inchon-ro, Sungbuk-ku, Seoul 136-705, Korea
| |
Collapse
|