1
|
Cheng C, Cui L, Cui X, Zhan Q, Ju J, Hong B, Huang Y, Ding Y, Xu H, Qiu T, Kang C, Liu X, Wang Q, Zeng L. ADAM12 promotes temozolomide resistance in glioblastoma by activating the TNF-α - NF-κB pathway. Cancer Lett 2025; 620:217684. [PMID: 40180114 DOI: 10.1016/j.canlet.2025.217684] [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: 10/05/2024] [Revised: 03/27/2025] [Accepted: 03/28/2025] [Indexed: 04/05/2025]
Abstract
Development of temozolomide (TMZ) resistance is a critical factor contributing to a poor prognosis in glioma patients. TMZ resistance is also closely associated with the phosphorylation level of NF-κB, yet targeted inhibition of NF-κB activity in glioma can be leveraged to overcome TMZ resistance. ADAM12, a protein significantly overexpressed in glioma cells, is implicated in the pathogenesis and progression of glioma, yet its role in the development of TMZ resistance is completely understood. We found that knockdown of ADAM12 was shown to arrest the glioma cell cycle, enhance apoptosis, inhibit DNA damage repair mechanisms, and sensitize glioma cells to TMZ. Targeting ADAM12 in vivo was found to increase the sensitivity of glioma cells to TMZ. Survival analysis indicated that ADAM12 serves as a prognostic marker for TMZ treatment. Using ELISA and protein interaction predictions via docking simulation, we identified the TNF-α shedding function of ADAM12 as a critical regulator of glioma progression. Furthermore, in glioma cell lines with unmethylated MGMT, the knockdown of ADAM12 enhanced sensitivity to TMZ by inhibiting the TNF-α/NF-κB pathway and reducing MGMT expression. In all, these results demonstrated that ADAM12 aids in shedding of membrane-bound TNF-a to drive TMZ resistance in glioma.
Collapse
Affiliation(s)
- Chunchao Cheng
- Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Department of Neurosurgery, Tianjin Medical University General Hospital and Key Laboratory of Neurotrauma, Variation, and Regeneration, Ministry of Education and Tianjin Municipal Government, Tianjin, 300052, China; Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Longtao Cui
- Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Department of Neurosurgery, Tianjin Medical University General Hospital and Key Laboratory of Neurotrauma, Variation, and Regeneration, Ministry of Education and Tianjin Municipal Government, Tianjin, 300052, China; Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Xiaoteng Cui
- Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Department of Neurosurgery, Tianjin Medical University General Hospital and Key Laboratory of Neurotrauma, Variation, and Regeneration, Ministry of Education and Tianjin Municipal Government, Tianjin, 300052, China; Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Qi Zhan
- Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Department of Neurosurgery, Tianjin Medical University General Hospital and Key Laboratory of Neurotrauma, Variation, and Regeneration, Ministry of Education and Tianjin Municipal Government, Tianjin, 300052, China; Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Jiasheng Ju
- Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Department of Neurosurgery, Tianjin Medical University General Hospital and Key Laboratory of Neurotrauma, Variation, and Regeneration, Ministry of Education and Tianjin Municipal Government, Tianjin, 300052, China; Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Biao Hong
- Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Department of Neurosurgery, Tianjin Medical University General Hospital and Key Laboratory of Neurotrauma, Variation, and Regeneration, Ministry of Education and Tianjin Municipal Government, Tianjin, 300052, China; Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Yanping Huang
- Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Department of Neurosurgery, Tianjin Medical University General Hospital and Key Laboratory of Neurotrauma, Variation, and Regeneration, Ministry of Education and Tianjin Municipal Government, Tianjin, 300052, China; Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Yaqing Ding
- Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Department of Neurosurgery, Tianjin Medical University General Hospital and Key Laboratory of Neurotrauma, Variation, and Regeneration, Ministry of Education and Tianjin Municipal Government, Tianjin, 300052, China; Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Hanyi Xu
- Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Department of Neurosurgery, Tianjin Medical University General Hospital and Key Laboratory of Neurotrauma, Variation, and Regeneration, Ministry of Education and Tianjin Municipal Government, Tianjin, 300052, China; Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Tian Qiu
- Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Department of Neurosurgery, Tianjin Medical University General Hospital and Key Laboratory of Neurotrauma, Variation, and Regeneration, Ministry of Education and Tianjin Municipal Government, Tianjin, 300052, China; Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Chunsheng Kang
- Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Department of Neurosurgery, Tianjin Medical University General Hospital and Key Laboratory of Neurotrauma, Variation, and Regeneration, Ministry of Education and Tianjin Municipal Government, Tianjin, 300052, China; Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Xiaomin Liu
- Neuro-Oncology Center, Tianjin Huanhu Hospital, Tianjin, 300350, China.
| | - Qixue Wang
- Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Department of Neurosurgery, Tianjin Medical University General Hospital and Key Laboratory of Neurotrauma, Variation, and Regeneration, Ministry of Education and Tianjin Municipal Government, Tianjin, 300052, China; Tianjin Medical University General Hospital, Tianjin, 300052, China.
| | - Liang Zeng
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| |
Collapse
|
2
|
Torabinejad S, Miro C, Nappi A, Del Giudice F, Cicatiello AG, Sagliocchi S, Acampora L, Restolfer F, Murolo M, Di Cicco E, Capone F, Imbimbo C, Dentice M, Crocetto F. Obesity alters the fitness of peritumoral adipose tissue, exacerbating tumor invasiveness in renal cancer through the induction of ADAM12 and CYP1B1. Mol Oncol 2025. [PMID: 39806854 DOI: 10.1002/1878-0261.13782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 10/21/2024] [Accepted: 11/28/2024] [Indexed: 01/16/2025] Open
Abstract
Obesity exacerbates the risk and aggressiveness of many types of cancer. Adipose tissue (AT) represents a prevalent component of the tumor microenvironment (TME) and contributes to cancer development and progression. Reciprocal communication between cancer and adipose cells leads to the generation of cancer-associated adipocytes (CAAs), which in turn foster tumor invasiveness by producing paracrine metabolites, adipocytokines, and growth factors. Interfering with the crosstalk between CAAs and cancer cells is of key relevance in the prevention of tumor progression. The present study aimed to analyze the contribution of peritumoral AT in renal cell carcinoma (RCC) progression in lean versus overweight or obese patients. By isolating human adipose-derived stromal/stem cells from the three groups of patients and performing conditioned medium studies with RCC cells along with in vivo xenograft experiments, we found that peritumoral adipocytes from the three groups show a distinct expression profile of genes. In particular, ADAM metallopeptidase domain 12 (ADAM12) and cytochrome P450 family 1 subfamily B member 1 (CYP1B1) were found to be upregulated in obesity and their silencing reduced RCC cell invasiveness. In conclusion, high ADAM12 and CYP1B1 expressions in the peritumoral adipocytes boost tumor invasiveness and may serve as an indicator of poor prognosis in RCC.
Collapse
Affiliation(s)
- Sepehr Torabinejad
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Italy
| | - Caterina Miro
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Italy
| | - Annarita Nappi
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Italy
| | - Francesco Del Giudice
- Department of Neurosciences, Reproductive Sciences and Odontostomatology, University of Naples "Federico II", Italy
| | | | - Serena Sagliocchi
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Italy
| | - Lucia Acampora
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Italy
| | - Federica Restolfer
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Italy
| | - Melania Murolo
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Italy
| | - Emery Di Cicco
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Italy
| | - Federico Capone
- Department of Neurosciences, Reproductive Sciences and Odontostomatology, University of Naples "Federico II", Italy
| | - Ciro Imbimbo
- Department of Neurosciences, Reproductive Sciences and Odontostomatology, University of Naples "Federico II", Italy
| | - Monica Dentice
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Italy
- CEINGE - Biotecnologie Avanzate S.c.a.r.l., Naples, Italy
| | - Felice Crocetto
- Department of Neurosciences, Reproductive Sciences and Odontostomatology, University of Naples "Federico II", Italy
| |
Collapse
|
3
|
Zhang H, Yang B. ADAM12 Silencing Mediated by FOXC2 Represses Meningioma Progression Through Inactivating the JAK1/STAT3/VEGFA Pathway. Biochem Genet 2024:10.1007/s10528-024-10893-4. [PMID: 39066954 DOI: 10.1007/s10528-024-10893-4] [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: 05/18/2024] [Accepted: 07/18/2024] [Indexed: 07/30/2024]
Abstract
Meningioma is a prevalently intracranial tumor, and the malignant type is aggressive with high recurrence. A Disintegrin and Metalloprotease 12 (ADAM12) is a common oncogene and differentially expressed in meningioma. However, its roles and mechanisms in meningioma development remain obscure. The differentially expressed genes in meningioma were analyzed by GEO (GSE77259 and GSE43290) datasets and weighted gene co-expression network analysis (WGCNA) based on GSE16581. ADAM12 expression was measured via qRT-PCR and western blot. The correlation between ADAM12 and FOXC2 was predicted through JASPER tool and identified via luciferase reporter analysis. Cell proliferation, migration and invasion were investigated using CCK-8, EdU, transwell assays. The JAK1/STAT3/VEGFA signaling was activated by IL-6, and analyzed via western blot. The differentially expressed ADAM12 in meningioma was screened by WGCNA and GEO analyses. ADAM12 silencing repressed meningioma cell proliferation, and decreased migration and invasion. The transcription factor FOXC2 expression was enhanced in meningioma based on GSE77259 and GSE43290 datasets, and positively induced ADAM12 transcription. The JAK1/STAT3/VEGFA signaling was inactivated due to ADAM12 silencing and activated via IL-6. Upregulation of FOXC2 promoted cell proliferation, migration and invasion, and these effects were reversed by silencing ADAM12. ADAM12 knockdown mediated via FOXC2 silencing restrained proliferation, migration and invasion of meningioma cells through inactivating the JAK1/STAT3/VEGFA pathway.
Collapse
Affiliation(s)
- Huaming Zhang
- Department of Neurosurgery, China Resources Wisco General Hospital, Wuhan University of Science and Technology, No. 209 Yejin Avenue, Qingshan District, Wuhan, 430080, Hubei, China.
| | - Bing Yang
- Department of Neurology, Wuhan Eighth Hospital, Wuhan, 430014, Hubei, China
| |
Collapse
|
4
|
Dong X, Yang Y, Hou J, Chen W, Yuan Q, Xu G, Liu J, Li C, Wu G. Weighted gene co-expression network reveals driver genes contributing to phenotypes of anaplastic thyroid carcinoma and immune checkpoint identification for therapeutic targets. Front Oncol 2022; 12:1018479. [PMID: 36530988 PMCID: PMC9751455 DOI: 10.3389/fonc.2022.1018479] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Accepted: 11/14/2022] [Indexed: 12/02/2022] Open
Abstract
Background Anaplastic thyroid carcinoma (ATC) is a rare but extremely malignant tumor, with a rapid growth rate and early metastasis thus leading to poor survival of patients. The molecular mechanisms underlying these aggressive traits of ATC remain unknown, which impedes the substantial progress in treatment to prolong ATC patient survival. Methods We applied weighted gene co-expression network analysis (WGCNA) to identify ATC-specific modules. The Metascape web and R package clusterProfiler were employed to perform enrichment analysis. Combined with differentially expressed gene analysis, we screened out the most potential driver genes and validated them using receiver operator characteristic (ROC) analysis, quantitative reverse transcription polymerase chain reaction (qRT-PCR), western blotting, immunohistochemistry (IHC), and triple immunofluorescence staining. Results A gene expression matrix covering 75 normal samples, 83 papillary thyroid carcinoma (PTC), 26 follicular thyroid carcinoma (FTC), 19 poor-differentiated thyroid carcinoma (PDTC), and 41 ATC tissue samples were integrated, based on which we detected three most potential ATC-specific modules and found that hub genes of these modules were enriched in distinct biological signals. Hub genes in the turquoise module were mainly enriched in mitotic cell cycle, tube morphogenesis, and cell differentiation, hub genes in the magenta module were mainly clustered in the extracellular matrix organization, positive regulation of cell motility, and regulation of Wnt signaling pathway, while hub genes in the blue module primarily participated in the inflammatory response, innate immune response, and adaptive immune response. We showed that 9 top genes, 8 transcription factors (TFs), and 4 immune checkpoint genes (ICGs) were differentially expressed in ATC compared to other thyroid samples and had high diagnostic values for ATC, among which, 9 novel ATC-specific genes (ADAM12, RNASE2, CASP5, KIAA1524, E2F7, MYBL1, SRPX2, HAVCR2, and TDO2) were validated with our clinical samples. Furthermore, we illustrated that ADAM12, RNASE2, and HAVCR2 were predominantly present in the cytoplasm. Conclusion Our study identified a set of novel ATC-specific genes that were mainly related to cell proliferation, invasion, metastasis, and immunosuppression, which might throw light on molecular mechanisms underlying aggressive phenotypes of ATC and provide promisingly diagnostic biomarkers and therapeutic targets.
Collapse
|
5
|
Graziosi A, Sita G, Corrieri C, Angelini S, d’Emmanuele di Villa Bianca R, Mitidieri E, Sorrentino R, Hrelia P, Morroni F. Effects of Subtoxic Concentrations of Atrazine, Cypermethrin, and Vinclozolin on microRNA-Mediated PI3K/Akt/mTOR Signaling in SH-SY5Y Cells. Int J Mol Sci 2022; 23:ijms232314538. [PMID: 36498866 PMCID: PMC9737829 DOI: 10.3390/ijms232314538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 11/18/2022] [Accepted: 11/19/2022] [Indexed: 11/24/2022] Open
Abstract
Endocrine-disrupting chemicals (EDCs) are different natural and synthetic chemicals that may interfere with several mechanisms of the endocrine system producing adverse developmental, metabolic, reproductive, and neurological effects in both human beings and wildlife. Among pesticides, numerous chemicals have been identified as EDCs. MicroRNAs (miRNAs) can regulate gene expression, making fine adjustments in mRNA abundance and regulating proteostasis. We hypothesized that exposure to low doses of atrazine, cypermethrin, and vinclozolin may lead to effects on miRNA expression in SH-SY5Y cells. In particular, the exposure of SH-SY5Y cells to subtoxic concentrations of vinclozolin is able to downregulate miR-29b-3p expression leading to the increase in the related gene expression of ADAM12 and CDK6, which may promote a pro-oncogenic response through the activation of the PI3K/Akt/mTOR pathway and counteracting p53 activity. A better understanding of the molecular mechanisms of EDCs could provide important insight into their role in human disease.
Collapse
Affiliation(s)
- Agnese Graziosi
- Department of Pharmacy and BioTechnology—FaBiT, Alma Mater Studiorum—University of Bologna, via Irnerio 48, 40126 Bologna, Italy
| | - Giulia Sita
- Department of Pharmacy and BioTechnology—FaBiT, Alma Mater Studiorum—University of Bologna, via Irnerio 48, 40126 Bologna, Italy
| | - Camilla Corrieri
- Department of Pharmacy and BioTechnology—FaBiT, Alma Mater Studiorum—University of Bologna, via Irnerio 48, 40126 Bologna, Italy
| | - Sabrina Angelini
- Department of Pharmacy and BioTechnology—FaBiT, Alma Mater Studiorum—University of Bologna, via Irnerio 48, 40126 Bologna, Italy
| | | | - Emma Mitidieri
- Department of Pharmacy, School of Medicine and Surgery, University of Study of Naples—Federico II, via Montesano 49, 80131 Naples, Italy
| | - Raffaella Sorrentino
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine and Surgery, University of Study of Naples—Federico II, via Pansini 5, 80131 Naples, Italy
| | - Patrizia Hrelia
- Department of Pharmacy and BioTechnology—FaBiT, Alma Mater Studiorum—University of Bologna, via Irnerio 48, 40126 Bologna, Italy
- Correspondence: ; Tel.: +39-051-209-1798
| | - Fabiana Morroni
- Department of Pharmacy and BioTechnology—FaBiT, Alma Mater Studiorum—University of Bologna, via Irnerio 48, 40126 Bologna, Italy
| |
Collapse
|
6
|
Zhang Y, Zhang H, Zhang X, Liu B. CBR3-AS1 Accelerates the Malignant Proliferation of Gestational Choriocarcinoma Cells by Stabilizing SETD4. DISEASE MARKERS 2022; 2022:7155525. [PMID: 35655916 PMCID: PMC9155919 DOI: 10.1155/2022/7155525] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 04/28/2022] [Indexed: 12/15/2022]
Abstract
Background Gestational choriocarcinoma (GC) is a rare malignant gestational trophoblastic tumor. Long noncoding RNA (lncRNA) CBR3 antisense RNA 1 (CBR3-AS1) has been reported to serve as a critical oncogene and facilitate tumor progression. Besides, we found that CBR3-AS1 is implicated in GC progression. Materials and Methods Gene and protein expression was detected via quantitative reverse transcription PCR (RT-qPCR) and western blot analyses, respectively. CCK-8 assay and colony formation assay were performed to assess cell proliferative abilities while flow cytometry analysis was applied for cell cycle and apoptosis. To analyze the specific mechanism among CBR3-AS1, SET domain containing 4 (SETD4), and polypyrimidine tract binding protein 1 (PTBP1), RNA binding protein immunoprecipitation (RIP), RNA pulldown, and mRNA stability assays were conducted. Results CBR3-AS1 was markedly upregulated in GC cells, and its downregulation suppressed cell proliferation, induced cell cycle arrest, but promoted cell apoptosis in GC. SETD4 was determined as the downstream mRNA of CBR3-AS1 and positively regulated by CBR3-AS1 in GC cells. Furthermore, CBR3-AS1 could interact with its RNA binding protein (RBP) PTBP1, thereby stabilizing SETD4 mRNA. Rescue assays verified that CBR3-AS1 facilitates GC cell malignant proliferation via SETD4. Conclusion CBR3-AS1 accelerates the malignant proliferation of GC cells via stabilizing SETD4.
Collapse
Affiliation(s)
- Yajuan Zhang
- Department of Reproductive Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hongxiu Zhang
- Department of Reproductive Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiaolei Zhang
- Department of Reproductive Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Bin Liu
- Department of Reproductive Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|