1
|
Zhu X, Wu J, Chen X, Shi D, Hui P, Wang H, Wu Z, Wu S, Bao W, Fan H. DNA ligase III mediates deoxynivalenol exposure-induced DNA damage in intestinal epithelial cells by regulating oxidative stress and interaction with PCNA. Int J Biol Macromol 2024; 282:137137. [PMID: 39505167 DOI: 10.1016/j.ijbiomac.2024.137137] [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: 08/25/2024] [Revised: 10/23/2024] [Accepted: 10/29/2024] [Indexed: 11/08/2024]
Abstract
Deoxynivalenol (DON) is a widely distributed mycotoxin that is severely cytotoxic and genotoxic to animals and humans. The gut is the initial site of DON exposure and absorption, which can cause severe intestinal damage. However, the underlying mechanisms and effective therapeutic approaches remain unknown. Here, the study indicated that DON exposure caused significant DNA damage in intestinal porcine epithelial cells (IPEC-J2), enhanced significantly the expression of γ-H2AX and 8-hydroxy-2'-deoxyguanosine, and altered the mRNA expression of key genes in the DNA repair pathway. Among them, ligases3 (LIG3) is the key DNA damage/repair gene and the only ligase responsible for the replication and maintenance of mitochondrial DNA. The expression of LIG3 was significantly decreased after DON exposure and showed a dose-dependent effect, decreased expression of LIG3 exacerbates DON-induced cytotoxicity and genotoxicity, decreased cell viability, induced apoptosis and cell cycle arrest, activation of inflammatory factors and MAPK pathway. Furthermore, LIG3 directly binds and regulates PCNA and play a positive regulatory role in the cellular cytotoxicity and genotoxicity upon DON exposure. Collectively, the findings elucidate the regulatory function of LIG3 in DON-induced DNA damage, providing valuable insights into identifying molecular targets for the comprehensive prevention and control of DON contamination.
Collapse
Affiliation(s)
- Xiaoyang Zhu
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design, college of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Jiayun Wu
- Jiangsu Agri-animal Husbandry Vocational College, Taizhou 225300, China
| | - Xiaolei Chen
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design, college of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Dongfeng Shi
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design, college of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Peng Hui
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design, college of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Haifei Wang
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design, college of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; Joint international Research Laboratory of Agriculture & Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Jiangsu, Yangzhou 225009, China
| | - Zhengchang Wu
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design, college of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Shenglong Wu
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design, college of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; Joint international Research Laboratory of Agriculture & Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Jiangsu, Yangzhou 225009, China
| | - Wenbin Bao
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design, college of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; Joint international Research Laboratory of Agriculture & Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Jiangsu, Yangzhou 225009, China
| | - Hairui Fan
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design, college of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China.
| |
Collapse
|
2
|
Sugiyanto RN, Metzger C, Inal A, Truckenmueller F, Gür K, Eiteneuer E, Huth T, Fraas A, Heinze I, Kirkpatrick J, Sticht C, Albrecht T, Goeppert B, Poth T, Pusch S, Mehrabi A, Schirmacher P, Ji J, Ori A, Roessler S. Proteomic profiling reveals CEACAM6 function in driving gallbladder cancer aggressiveness through integrin receptor, PRKCD and AKT/ERK signaling. Cell Death Dis 2024; 15:780. [PMID: 39468006 PMCID: PMC11519453 DOI: 10.1038/s41419-024-07171-x] [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: 04/09/2024] [Revised: 10/08/2024] [Accepted: 10/18/2024] [Indexed: 10/30/2024]
Abstract
Gallbladder cancer (GBC) presents as an aggressive malignancy with poor patient outcome. Like other epithelial cancers, the mechanisms of GBC cancer progression remain vague and efforts in finding targeted therapies fall below expectations. This study combined proteomic analysis of formalin-fixed paraffin-embedded (FFPE) GBC samples, functional and molecular characterization of potential oncogenes and identification of potential therapeutic strategies for GBC. We identified Carcinoembryonic Antigen-related Cell Adhesion Molecule 6 (CEACAM6) as one of the significantly most upregulated proteins in GBC. CEACAM6 overexpression has been observed in other cancer entities but the molecular function remains unclear. Our functional analyses in vitro and in vivo mouse models revealed that CEACAM6 supported the initial steps of cancer progression and metastasis by decreasing cell adhesion and promoting migration and invasion of GBC cells. Conversely, CEACAM6 knockdown abolished GBC aggressiveness by increasing cell adhesion while reducing cell migration, cell proliferation, and colony formation. BirA-BioID followed by mass-spectrometry revealed Integrin Beta-1 (ITGB1) and Protein Kinase C Delta (PRKCD) as direct molecular and functional partners of CEACAM6 supporting GBC cell migration. ERK and AKT signaling and their downstream target genes were regulated by CEACAM6 and thus the treatment with AKT inhibitor capivasertib or ERK inhibitor ulixertinib mitigated the CEACAM6-induced migration. These findings demonstrate that CEACAM6 is crucially involved in gallbladder cancer progression by promoting migration and inhibiting cell adhesion through ERK and AKT signaling providing specific options for treatment of CEACAM6-positive cancers.
Collapse
Affiliation(s)
- Raisatun Nisa Sugiyanto
- Institute of Pathology, University Hospital Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Carmen Metzger
- Institute of Pathology, University Hospital Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Aslihan Inal
- Institute of Pathology, University Hospital Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Felicia Truckenmueller
- Institute of Pathology, University Hospital Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Kira Gür
- Institute of Pathology, University Hospital Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Eva Eiteneuer
- Institute of Pathology, University Hospital Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Thorben Huth
- Institute of Pathology, University Hospital Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Angelika Fraas
- Institute of Pathology, University Hospital Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Ivonne Heinze
- Leibniz Institute on Aging-Fritz Lipmann Institute (FLI), Jena, Germany
| | | | - Carsten Sticht
- NGS Core Facility, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Thomas Albrecht
- Institute of Pathology, University Hospital Heidelberg, Heidelberg University, Heidelberg, Germany
- Liver Cancer Centre Heidelberg (LCCH), Heidelberg, Germany
| | - Benjamin Goeppert
- Institute of Pathology, University Hospital Heidelberg, Heidelberg University, Heidelberg, Germany
- Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
- Institute of Pathology and Neuropathology, RKH Hospital Ludwigsburg, Ludwigsburg, Germany
| | - Tanja Poth
- Center for Model System and Comparative Pathology, Institute of Pathology, University Hospital Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Stefan Pusch
- Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neuropathology, Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Arianeb Mehrabi
- Liver Cancer Centre Heidelberg (LCCH), Heidelberg, Germany
- Department of General Visceral and Transplantation Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Peter Schirmacher
- Institute of Pathology, University Hospital Heidelberg, Heidelberg University, Heidelberg, Germany
- Liver Cancer Centre Heidelberg (LCCH), Heidelberg, Germany
| | - Junfang Ji
- The MOE Key Laboratory of Biosystems Homeostasis & Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, China
| | - Alessandro Ori
- Leibniz Institute on Aging-Fritz Lipmann Institute (FLI), Jena, Germany
| | - Stephanie Roessler
- Institute of Pathology, University Hospital Heidelberg, Heidelberg University, Heidelberg, Germany.
- Liver Cancer Centre Heidelberg (LCCH), Heidelberg, Germany.
| |
Collapse
|
3
|
Wang J, Liu H, Yu Z, Zhou Q, Sun F, Han J, Gao L, Dou B, Zhang H, Fu J, Jia W, Chen W, Hu J, Han B. Reciprocal regulation between RACGAP1 and AR contributes to endocrine therapy resistance in prostate cancer. Cell Commun Signal 2024; 22:339. [PMID: 38898473 PMCID: PMC11186203 DOI: 10.1186/s12964-024-01703-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 06/06/2024] [Indexed: 06/21/2024] Open
Abstract
BACKGROUND Endocrine resistance driven by sustained activation of androgen receptor (AR) signaling pathway in advanced prostate cancer (PCa) is fatal. Characterization of mechanisms underlying aberrant AR pathway activation to search for potential therapeutic strategy is particularly important. Rac GTPase-activating protein 1 (RACGAP1) is one of the specific GTPase-activating proteins. As a novel tumor proto-oncogene, overexpression of RACGAP1 was related to the occurrence of various tumors. METHODS Bioinformatics methods were used to analyze the relationship of expression level between RACGAP1 and AR as well as AR pathway activation. qRT-PCR and western blotting assays were performed to assess the expression of AR/AR-V7 and RACGAP1 in PCa cells. Immunoprecipitation and immunofluorescence experiments were conducted to detect the interaction and co-localization between RACGAP1 and AR/AR-V7. Gain- and loss-of-function analyses were conducted to investigate the biological roles of RACGAP1 in PCa cells, using MTS and colony formation assays. In vivo experiments were conducted to evaluate the effect of RACGAP1 inhibition on the tumor growth. RESULTS RACGAP1 was a gene activated by AR, which was markedly upregulated in PCa patients with CRPC and enzalutamide resistance. AR transcriptionally activated RACGAP1 expression by binding to its promoter region. Reciprocally, nuclear RACGAP1 bound to the N-terminal domain (NTD) of both AR and AR-V7, blocking their interaction with the E3 ubiquitin ligase MDM2. Consequently, this prevented the degradation of AR/AR-V7 in a ubiquitin-proteasome-dependent pathway. Notably, the positive feedback loop between RACGAP1 and AR/AR-V7 contributed to endocrine therapy resistance of CRPC. Combination of enzalutamide and in vivo cholesterol-conjugated RIG-I siRNA drugs targeting RACGAP1 induced potent inhibition of xenograft tumor growth of PCa. CONCLUSION In summary, our results reveal that reciprocal regulation between RACGAP1 and AR/AR-V7 contributes to the endocrine resistance in PCa. These findings highlight the therapeutic potential of combined RACGAP1 inhibition and enzalutamide in treatment of advanced PCa.
Collapse
Affiliation(s)
- Jiajia Wang
- The Key Laboratory of Experimental Teratology, Department of Pathology, School of Basic Medical Sciences, Ministry of Education, Shandong University, Jinan, 250012, Shandong, China
| | - Hui Liu
- Department of Pathology, Qilu Hospital, Shandong University, Jinan, 250012, China
| | - Zeyuan Yu
- The Key Laboratory of Experimental Teratology, Department of Pathology, School of Basic Medical Sciences, Ministry of Education, Shandong University, Jinan, 250012, Shandong, China
| | - Qianqian Zhou
- The Key Laboratory of Experimental Teratology, Department of Pathology, School of Basic Medical Sciences, Ministry of Education, Shandong University, Jinan, 250012, Shandong, China
| | - Feifei Sun
- Department of Pathology, Qilu Hospital, Shandong University, Jinan, 250012, China
| | - Jingying Han
- The Key Laboratory of Experimental Teratology, Department of Pathology, School of Basic Medical Sciences, Ministry of Education, Shandong University, Jinan, 250012, Shandong, China
| | - Lin Gao
- The Key Laboratory of Experimental Teratology, Department of Pathology, School of Basic Medical Sciences, Ministry of Education, Shandong University, Jinan, 250012, Shandong, China
| | - Baokai Dou
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Hanwen Zhang
- The Key Laboratory of Experimental Teratology, Department of Pathology, School of Basic Medical Sciences, Ministry of Education, Shandong University, Jinan, 250012, Shandong, China
| | - Jiawei Fu
- The Key Laboratory of Experimental Teratology, Department of Pathology, School of Basic Medical Sciences, Ministry of Education, Shandong University, Jinan, 250012, Shandong, China
| | - Wenqiao Jia
- Department of Pathology, Qilu Hospital, Shandong University, Jinan, 250012, China
| | - Weiwen Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shandong University, Jinan, 250012, Shandong, China
| | - Jing Hu
- Department of Pathology, Qilu Hospital, Shandong University, Jinan, 250012, China.
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA.
| | - Bo Han
- The Key Laboratory of Experimental Teratology, Department of Pathology, School of Basic Medical Sciences, Ministry of Education, Shandong University, Jinan, 250012, Shandong, China.
- Department of Pathology, Qilu Hospital, Shandong University, Jinan, 250012, China.
| |
Collapse
|
4
|
Ye J, Zhang X, Xie JX, Hou Y, Fan WM, Wang XQ, Zhang LW, Yang XM, Li J, Fei H. RACGAP1 knockdown synergizes and enhances the effects of chemotherapeutics on ovarian cancer. Am J Transl Res 2024; 16:2132-2146. [PMID: 38883382 PMCID: PMC11170603 DOI: 10.62347/qnzu1402] [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: 11/18/2023] [Accepted: 05/06/2024] [Indexed: 06/18/2024]
Abstract
Among the three most prevalent cancers affecting the female reproductive system, ovarian cancer (OV) ranks as the second most frequently diagnosed. It is important to investigate the genomic complexity of OV to develop diagnostic and therapeutic strategies. Through the utilization of bioinformatics analysis, it was determined that RacGTPase Activating Protein 1 (RACGAP1) holds significant significance in the field of OV chemotherapeutics, an aspect that has not been thoroughly explored in prior investigations. In our study, a notable increase in RACGAP1 expression was detected in ovarian cancer, demonstrating a robust association with clinicopathological features and patient prognosis. In vivo and in vitro testing revealed that RACGAP1 acts synergistically with chemotherapeutics to enhance their effects on ovarian cancer. Furthermore, an interaction between RACGAP1 and the subunit G2 of the condensin II complex, known as non-SMC condensin II complex subunit G2 (NCAPG2), has been identified. Our findings may provide new insight for improving therapeutic strategies for OV.
Collapse
Affiliation(s)
- Jun Ye
- Department of Obstetrics and Gynecology, The Fifth People's Hospital of Shanghai, Fudan University Shanghai, China
| | - Xiang Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai, China
| | - Jia-Xuan Xie
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai, China
| | - Yue Hou
- Department of Obstetrics and Gynecology, The Fifth People's Hospital of Shanghai, Fudan University Shanghai, China
| | - Wei-Min Fan
- Department of Obstetrics and Gynecology, The Fifth People's Hospital of Shanghai, Fudan University Shanghai, China
| | - Xiao-Qin Wang
- Department of Obstetrics and Gynecology, The Fifth People's Hospital of Shanghai, Fudan University Shanghai, China
| | - Li-Wen Zhang
- Department of Obstetrics and Gynecology, The Fifth People's Hospital of Shanghai, Fudan University Shanghai, China
| | - Xiao-Mei Yang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai, China
| | - Jun Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai, China
| | - He Fei
- Department of Obstetrics and Gynecology, The Fifth People's Hospital of Shanghai, Fudan University Shanghai, China
| |
Collapse
|
5
|
Wang X, Sun J, Liu Y, Lin Z, Jiang X, Ye Y, Lv C, Lian X, Xu W, Luo S, Liao S, Chen Z, Wang S. Trps1 predicts poor prognosis in advanced high grade serous ovarian carcinoma. Int J Cancer 2024; 154:1639-1651. [PMID: 38212905 DOI: 10.1002/ijc.34844] [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: 08/17/2023] [Revised: 12/17/2023] [Accepted: 12/22/2023] [Indexed: 01/13/2024]
Abstract
TRPS1 is aberrantly expressed in a variety of tumors, including breast, prostate, and gastric cancers, and is strongly associated with tumorigenesis or prognosis. However, the role of TRPS1 in high grade serous ovarian carcinoma (HGSC) is unknown. We investigated the relationship between TRPS1 expression and clinicopathology in HGSC patients. The tumor-related regulatory mechanisms of TRPS1 was explored through in vivo and vitro experiments. The results showed that TRPS1 was highly expressed in HGSC compared to normal tissues. It was also linked to the cell proliferation index Ki67 and poor prognosis. In vivo experiments showed that knockdown of TRPS1 could inhibit tumor growth. In vitro experiments, knockdown of TRPS1 inhibited the proliferation of ovarian cancer cells. TRPS1 exerted its regulatory role as a transcription factor, binding to the PSAT1 promoter and promoting the expression of PSAT1 gene. Meanwhile, PSAT1 was positively correlated with CCND1 expression. These results suggest that TRPS1 affects HGSC proliferation and cell cycle by regulating PSAT1 and thus CCND1 expression.
Collapse
Affiliation(s)
- Xiaojiang Wang
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine of Fujian Province University, Fujian Medical University, Fuzhou, China
- Department of Molecular Pathology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China
- Department of Histology and Embryology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Jiandong Sun
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine of Fujian Province University, Fujian Medical University, Fuzhou, China
- Department of Histology and Embryology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Yue Liu
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine of Fujian Province University, Fujian Medical University, Fuzhou, China
- Department of Histology and Embryology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Zihang Lin
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine of Fujian Province University, Fujian Medical University, Fuzhou, China
- Department of Histology and Embryology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Xia Jiang
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine of Fujian Province University, Fujian Medical University, Fuzhou, China
- Department of Histology and Embryology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Yuhong Ye
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine of Fujian Province University, Fujian Medical University, Fuzhou, China
- Department of Histology and Embryology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
- Department of Pathology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Chengyu Lv
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine of Fujian Province University, Fujian Medical University, Fuzhou, China
- Department of Histology and Embryology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
- Department of Obstetrics and Gynecology, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Xiuli Lian
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine of Fujian Province University, Fujian Medical University, Fuzhou, China
- Department of Histology and Embryology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Weiwei Xu
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine of Fujian Province University, Fujian Medical University, Fuzhou, China
- Department of Histology and Embryology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Shanshan Luo
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine of Fujian Province University, Fujian Medical University, Fuzhou, China
- Department of Histology and Embryology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Shumin Liao
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine of Fujian Province University, Fujian Medical University, Fuzhou, China
- Department of Histology and Embryology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Zhangting Chen
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine of Fujian Province University, Fujian Medical University, Fuzhou, China
- Department of Histology and Embryology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Shie Wang
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine of Fujian Province University, Fujian Medical University, Fuzhou, China
- Department of Histology and Embryology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| |
Collapse
|
6
|
Yao H, Li J, Zhou D, Pan X, Chu Y, Yin J. FOXM1 transcriptional regulation of RacGAP1 activates the PI3K/AKT signaling pathway to promote the proliferation, migration, and invasion of cervical cancer cells. Int J Clin Oncol 2024; 29:333-344. [PMID: 38172354 DOI: 10.1007/s10147-023-02452-5] [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: 07/10/2023] [Accepted: 12/05/2023] [Indexed: 01/05/2024]
Abstract
BACKGROUND Cervical cancer (CC) is the most common gynecological tumor disease in women, which occurs at the junction of cervical squamous columnar epithelium. We investigated the effect and mechanism of transcription factor FOXM1 synergizing RacGAP1 in the proliferation, migration, and invasion of CC cells. METHODS Here, we analyzed the correlation between FOXM1 and RacGAP1 and the clinicopathological features of 68 CC patients. RT-qPCR was used to assess FOXM1 and RacGAP1 mRNA expression in CC tissues and cells. Cell proliferation was assessed by CCK-8 and EDU assays. Transwell assay was applied to test migration and invasion. Cell apoptosis was evaluated utilizing flow cytometry. ChIP and dual-luciferase reporter assays confirmed the interaction of FOXM1 and RacGAP1. Protein levels of FOXM1 and RacGAP1, as well as PI3K/AKT, were analyzed by Western blot. RESULTS FOXM1 expression was correlated with FIGO stage and histological grade, and RacGAP1 expression was correlated with histological grade. FOXM1 and RacGAP1 levels were increased in CC tissues, and higher expressed in human CC cell lines than that in an immortalized HPV-negative skin keratinocyte line (HaCaT). Depleted RacGAP1 suppressed CC cell proliferation, migration and invasion, and promoted apoptosis. RacGAP1 was a target gene of FOXM1, and FOXM1 positively regulated RacGAP1 expression. FOXM1 had a synergistic effect with RacGAP1 to exert oncogenic function in CC by activating the PI3K/AKT signaling. CONCLUSION FOXM1 cooperates with RacGAP1 to induce CC cell proliferation, migration and invasion, inhibit apoptosis, and regulate PI3K/AKT signaling to promote CC progression.
Collapse
Affiliation(s)
- Hongye Yao
- Department of Gynecology, The First People's Hospital of Jiashan County, Jiaxing, Zhejiang, 314100, People's Republic of China
| | - Juan Li
- Department of Gynaecology and Obstetrics, Maternal and Child Health Hospital of Ninghai City, Ningbo, Zhejiang, 315600, People's Republic of China
| | - Dan Zhou
- Department of Gynecology, The First People's Hospital of Jiashan County, Jiaxing, Zhejiang, 314100, People's Republic of China
| | - Xiaotian Pan
- Department of Gynecology, The First People's Hospital of Jiashan County, Jiaxing, Zhejiang, 314100, People's Republic of China
| | - Yaying Chu
- Department of Gynecology, The First People's Hospital of Jiashan County, Jiaxing, Zhejiang, 314100, People's Republic of China
| | - Jun Yin
- Department of Gynecology, Maternal and Child Health Hospital of Jiashan County, No. 20, Luoxing Road, Luoxing Street, Jiaxing, Zhejiang, 314100, People's Republic of China.
| |
Collapse
|
7
|
Zheng CS, Huang WM, Xia HM, Mi JL, Li YQ, Liang HQ, Zhou L, Lu ZX, Wu F. Oncogenic and immunological roles of RACGAP1 in pan-cancer and its potential value in nasopharyngeal carcinoma. Apoptosis 2024; 29:243-266. [PMID: 37670104 DOI: 10.1007/s10495-023-01884-8] [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] [Accepted: 08/17/2023] [Indexed: 09/07/2023]
Abstract
A particular GTPase-activating protein called RACGAP1 is involved in apoptosis, proliferation, invasion, metastasis, and drug resistance in a variety of malignancies. Nevertheless, the role of RACGAP1 in pan-cancer was less studied, and its value of the expression and prognostic of nasopharyngeal carcinoma (NPC) has not been explored. Hence, the goal of this study was to investigate the oncogenic and immunological roles of RACGAP1 in various cancers and its potential value in NPC. We comprehensively analyzed RACGAP1 expression, prognostic value, function, methylation levels, relationship with immune cells, immune infiltration, and immunotherapy response in pan-cancer utilizing multiple databases. The results discovered that RACGAP1 expression was elevated in most cancers and suggested poor prognosis, which could be related to the involvement of RACGAP1 in various cancer-related pathways such as the cell cycle and correlated with RACGAP1 methylation levels, immune cell infiltration and reaction to immunotherapy, and chemoresistance. RACGAP1 could inhibit anti-tumor immunity and immunotherapy responses by fostering immune cell infiltration and cytotoxic T lymphocyte dysfunction. Significantly, we validated that RACGAP1 mRNA and protein were highly expressed in NPC. The Gene Expression Omnibus database revealed that elevated RACGAP1 expression was associated with shorter PFS in patients with NPC, and RACGAP1 potentially influenced cell cycle progression, DNA replication, metabolism, and immune-related pathways, resulting in the recurrence and metastasis of NPC. This study indicated that RACGAP1 could be a potential biomarker in pan-cancer and NPC.
Collapse
Affiliation(s)
- Cheng-Shan Zheng
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, 530021, Guangxi, China
| | - Wei-Mei Huang
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Hong-Mei Xia
- Department of Gynecology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, China
| | - Jing-Lin Mi
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, 530021, Guangxi, China
| | - Yuan-Qing Li
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, 530021, Guangxi, China
| | - Hui-Qing Liang
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, 530021, Guangxi, China
| | - Li Zhou
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, 530021, Guangxi, China
| | - Zhou-Xue Lu
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, 530021, Guangxi, China
| | - Fang Wu
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China.
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, 530021, Guangxi, China.
| |
Collapse
|
8
|
Zhu L, Cao P, Yang S, Lin F, Wang J. Prolonged exposure to environmental levels of microcystin-LR triggers ferroptosis in brain via the activation of Erk/MAPK signaling pathway. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 267:115651. [PMID: 37913581 DOI: 10.1016/j.ecoenv.2023.115651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 10/16/2023] [Accepted: 10/27/2023] [Indexed: 11/03/2023]
Abstract
While existing research has illuminated the environmental dangers and neurotoxic effects of MC-LR exposure, the molecular underpinnings of brain damage from environmentally-relevant MC-LR exposure remain elusive. Employing a comprehensive approach involving RNA sequencing, histopathological examination, and biochemical analyses, we discovered genes differentially expressed and enriched in the ferroptosis pathway. This finding was associated with mitochondrial structural impairment and downregulation of Gpx4 and Slc7a11 in mice brains subjected to low-dose MC-LR over 180 days. Mirroring these findings, we noted reduced cell viability and GSH/GSSH ratio, along with an increased ROS level, in HT-22, BV-2, and bEnd.3 cells following MC-LR exposure. Intriguingly, MC-LR also amplified phospho-Erk levels in both in vivo and in vitro settings, and the effects were mitigated by treatment with PD98059, an Erk inhibitor. Taken together, our findings implicate the activation of the Erk/MAPK signaling pathway in MC-LR-induced ferroptosis, shedding valuable light on the neurotoxic mechanisms of MC-LR. These insights could guide future strategies to prevent MC-induced neurodegenerative diseases.
Collapse
Affiliation(s)
- Lingyun Zhu
- Department of Cell Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Pingping Cao
- Department of Cell Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Suisui Yang
- Department of Cell Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Fan Lin
- Department of Cell Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Jing Wang
- Department of Cell Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
9
|
Wu X, Xu Z, Li W, Lu Y, Pu J. HIF‑1α and RACGAP1 promote the progression of hepatocellular carcinoma in a mutually regulatory way. Mol Med Rep 2023; 28:218. [PMID: 37772389 PMCID: PMC10568255 DOI: 10.3892/mmr.2023.13105] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 09/12/2023] [Indexed: 09/30/2023] Open
Abstract
Hypoxia, a condition characterized by low oxygen levels, serves an important role in the progression of hepatocellular carcinoma (HCC). However, the precise molecular mechanisms underlying hypoxia‑induced HCC progression are yet to be fully elucidated. The present study assessed the involvement of two key factors, hypoxia‑inducible factor‑1α (HIF‑1α) and Rac GTPase activating protein 1 (RACGAP1), in HCC development under hypoxic conditions. HIF‑1α and RACGAP1 genes were overexpressed and knocked down in Hep3B and Huh7 cells using lentiviral transduction and the levels of HIF‑1α and RACGAP1 in the cells were assessed using quantitative PCR, western blotting and immunofluorescence. Co‑immunoprecipitation experiments were performed to evaluate the interaction between HIF‑1α and RACGAP1. Subsequently, the proliferation, apoptosis, migration and invasion of Hep3B and Huh7 cells were assessed using the Cell Counting Kit‑8 assay, flow cytometry, Transwell assay and migration experiments. The expression levels of HIF‑1α and RACGAP1 in normal and HCC tumor samples were analyzed utilizing the Gene Expression Profiling Interactive Analysis database. Furthermore, correlations between HIF‑1α/RACGAP1 gene expression levels and patient survival outcomes were evaluated using the Kaplan‑Meier plotter. Knockdown of HIF‑1α resulted in a significant decrease in RACGAP1 expression, whilst overexpression of HIF‑1α resulted in a significant increase in RACGAP1 expression. Moreover, overexpression and knockdown of RACGAP1 had the same effect on HIF‑1α expression. Additionally, it was demonstrated that HIF‑1α and RACGAP1 interacted directly within a complex. Overexpression of HIF‑1α or RACGAP1 significantly increased proliferation, invasion and migration, and significantly decreased the proportion of apoptotic Hep3B and Huh7 cells. Conversely, knockdown of HIF‑1α or RACGAP1 significantly decreased proliferation, invasion and migration, and significantly increased the proportion of apoptotic Hep3B and Huh7 cells. In addition, the combined knockdown or overexpression of HIF‑1α and RACGAP1 had a more pronounced effect on HCC cell migration compared with knockdown of HIF‑1α alone. Furthermore, there was a significant positive correlation between the expression levels of HIF‑1α and RACGAP1 in HCC tissues and patients with HCC and upregulation of both HIF‑1α and RACGAP1 demonstrated a lower overall survival probability. In conclusion, HIF‑1α and RACGAP1 may synergistically contribute to the development of HCC, highlighting their potential as valuable targets for HCC therapy.
Collapse
Affiliation(s)
- Xianjian Wu
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi 533000, P.R. China
| | - Zuoming Xu
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi 533000, P.R. China
| | - Wenchuan Li
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi 533000, P.R. China
| | - Yuan Lu
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi 533000, P.R. China
| | - Jian Pu
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi 533000, P.R. China
| |
Collapse
|
10
|
Anjali K, Kumar T, Kar AG, Kumar P, Narayan G, Singh S. Association of haplotype and linkage disequilibrium of PARP1 polymorphisms rs1136410, rs1805405 and rs3219088 with gallbladder cancer. Dig Liver Dis 2022; 55:807-814. [PMID: 36581511 DOI: 10.1016/j.dld.2022.12.006] [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: 07/08/2022] [Revised: 10/05/2022] [Accepted: 12/07/2022] [Indexed: 12/31/2022]
Abstract
BACKGROUND Previously, we have reported that PARP1 rs1136410 is significantly associated with increased the risk of gallbladder cancer. AIM We aimed to investigate the association of PARP1 rs1805405 and rs3219088 polymorphisms with risk of GBC and also association of the haplotype and combined effect of PARP1 SNPs (rs1805405 G/A, rs3219088 G/T and rs1136410 A/G). We have also investigated the expression profile of PARP1 and its correlation with polymorphisms, clinical parameters and overall survival. METHODS PARP1 polymorphisms were genotyped by PCR-RFLP and the expression profile of PARP1 at mRNA level was analyzed by semi-quantitative PCR. Overall survival was analyzed using Kaplan-Meier plot and Cox-regression analysis. RESULTS Haplotype analysis of the PARP1 polymorphisms revealed that AGG, AAG and GGT haplotypes are significantly associated with decreased risk of GBC, while AAT, AGT, GGG and GAG haplotypes are significantly associated with increased risk of GBC. Patients with T1+T2 and treated with chemotherapy having risk genotypes of rs1805405 have decreased overall survival. Upregulation of PARP1 is significantly associated with longer overall survival in patients with GBC with different clinical parameters. SNPs rs1136410 and rs1805405 genotypes are significantly associated with PARP1 expression. CONCLUSION Haplotype analysis suggests that PARP1 may have a potential role in gallbladder carcinogenesis.
Collapse
Affiliation(s)
- Kumari Anjali
- Department of Zoology, Mahila Mahavidyalaya, Banaras Hindu University, Varanasi 221005, Uttar Pradesh
| | - Tarun Kumar
- Department of Surgical Oncology, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, Uttar Pradesh
| | - Amrita Ghosh Kar
- Department of Pathology, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, Uttar Pradesh
| | - Puneet Kumar
- Department of General Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, Uttar Pradesh
| | - Gopeshwar Narayan
- Cancer Genetics Laboratory, Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi 221005, Uttar Pradesh
| | - Sunita Singh
- Department of Zoology, Mahila Mahavidyalaya, Banaras Hindu University, Varanasi 221005, Uttar Pradesh.
| |
Collapse
|
11
|
XPO1-Mediated EIF1AX Cytoplasmic Relocation Promotes Tumor Migration and Invasion in Endometrial Carcinoma. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:1361135. [PMID: 36589683 PMCID: PMC9800903 DOI: 10.1155/2022/1361135] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 10/30/2022] [Accepted: 12/01/2022] [Indexed: 12/24/2022]
Abstract
Dysregulation of eukaryotic translation initiation factor 1A, X-linked (EIF1AX), has been implicated in the pathogenesis of some cancers. However, the role of EIF1AX in endometrial carcinoma (EC) remains unknown. We investigated the EIF1AX expression in EC patients and assessed its tumorigenesis-associated function and nucleocytoplasmic transport mechanism in vitro and in vivo. The results indicated that the cytoplasmic EIF1AX expression showed a gradual increase when going from endometrium normal tissue, simple endometrial hyperplasia, complex endometrial hyperplasia, and endometrial atypical hyperplasia to EC, while vice versa for the nuclear EIF1AX expression. In addition, the cytoplasmic EIF1AX expression was positively correlated with histologic type, high International Federation of Gynecology and Obstetrics (FIGO) grade, advanced FIGO stage, deeper infiltration, high Ki67 index, and shorter recurrence-free survival in EC patients. In vitro, short hairpin RNA-mediated EIF1AX depletion or SV40NLS-mediated EIF1AX import into the nucleus in multiple human EC cells potently suppressed cell migration and invasion, epithelial-mesenchymal transition, and lung metastasis. Moreover, exportin 1 induced the transport of EIF1AX from the nucleus to the cytoplasm that could be inhibited by leptomycin B treatment or the mutation in the EIF1AX location sequence. These results demonstrate that cytoplasmic EIF1AX may play a key role in the incidence and promotion of EC, and thus, targeting EIF1AX or its nucleocytoplasmic transport process may offer an effective new therapeutic approach to EC.
Collapse
|
12
|
Zou L, Yang Y, Zhou B, Li W, Liu K, Li G, Miao H, Song X, Yang J, Geng Y, Li M, Bao R, Liu Y. tRF-3013b inhibits gallbladder cancer proliferation by targeting TPRG1L. Cell Mol Biol Lett 2022; 27:99. [PMID: 36401185 PMCID: PMC9673407 DOI: 10.1186/s11658-022-00398-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 10/25/2022] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND tRNA-derived fragments (tRFs) are newly discovered noncoding RNAs and regulate tumor progression via diverse molecular mechanisms. However, the expression and biofunction of tRFs in gallbladder cancer (GBC) have not been reported yet. METHODS The expression of tRFs in GBC was detected by tRF and tiRNA sequencing in GBC tissues and adjacent tissues. The biological function of tRFs was investigated by cell proliferation assay, clonal formation assay, cell cycle assay, and xenotransplantation model in GBC cell lines. The molecular mechanism was discovered and verified by transcriptome sequencing, fluorescence in situ hybridization (FISH), target gene site prediction, and RNA binding protein immunoprecipitation (RIP). RESULTS tRF-3013b was significantly downregulated in GBC compared with para-cancer tissues. Decreased expression of tRF-3013b in GBC patients was correlated with poor overall survival. Dicer regulated the production of tRF-3013b, and its expression was positively correlated with tRF-3013b in GBC tissues. Functional experiments demonstrated that tRF-3013b inhibited GBC cell proliferation and induced cell-cycle arrest. Mechanically, tRF-3013b exerted RNA silencing effect on TPRG1L by binding to AGO3, and then inhibited NF-κB. TPRG1L overexpression could rescue the effects of tRF-3013b on GBC cell proliferation. CONCLUSIONS This study indicated that Dicer-induced tRF-3013b inhibited GBC proliferation by targeting TPRG1L and repressed NF-κB, pointing to tRF-3013b as a novel potential therapeutic target of GBC.
Collapse
Affiliation(s)
- Lu Zou
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China
- Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai, 200092, China
- Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200032, China
| | - Yang Yang
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China
- Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai, 200092, China
- Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200032, China
| | - Biyu Zhou
- Department of Plastic and Reconstructive Surgery, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200127, China
| | - Weijian Li
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China
- Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai, 200092, China
- Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200032, China
| | - Ke Liu
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China
- Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai, 200092, China
- Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200032, China
| | - Guoqiang Li
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China
- Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai, 200092, China
- Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200032, China
| | - Huijie Miao
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China
- Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai, 200092, China
- Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200032, China
| | - Xiaoling Song
- Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai, 200092, China
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665, Kongjiang Road, Shanghai, 200092, China
| | - Jiahua Yang
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China
- Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai, 200092, China
- Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200032, China
| | - Yajun Geng
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China
- Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai, 200092, China
- Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200032, China
| | - Maolan Li
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China.
- Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai, 200092, China.
- Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200032, China.
| | - Runfa Bao
- Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai, 200092, China.
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665, Kongjiang Road, Shanghai, 200092, China.
| | - Yingbin Liu
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China.
- Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai, 200092, China.
- Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200032, China.
| |
Collapse
|
13
|
Xiong J, Wu R, He A, Hou P, Wang J, Zhang R, Liao W, Wu L, Li E. Comprehensive analysis of the effects of KIF2C on prognosis, biological functions and immune infiltration in PAAD. Tissue Cell 2022; 78:101900. [DOI: 10.1016/j.tice.2022.101900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 08/14/2022] [Accepted: 08/17/2022] [Indexed: 10/15/2022]
|
14
|
Up-Regulation of RACGAP1 Promotes Progressions of Hepatocellular Carcinoma Regulated by GABPA via PI3K/AKT Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3034150. [PMID: 35958019 PMCID: PMC9363186 DOI: 10.1155/2022/3034150] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 06/08/2022] [Accepted: 07/21/2022] [Indexed: 11/18/2022]
Abstract
Hepatocellular carcinoma (HCC) is one of the dominating tumors causing death due to lack of timely discovery and valid treatment. Abnormal increase of Rac GTPase activating protein 1 (RACGAP1) has been verified to be an oncogene in plenty tumors. The profound mechanism of RACGAP1 was rarely reported in HCC. In this study, we explored the function and mechanism of RACGAP1 in HCC through multiple analysis and experiments. RACGAP1 expression was up-regulated in HCC samples and the high expression of RACGAP1 was an independent prognostic risk factor for HCC patients. Meanwhile, RACGAP1 promoted developments of HCC both in vitro and in vivo. We verified that RACGAP1 promoted proliferation of HCC via PI3K/AKT/CDK2 and PI3K/AKT/GSK3β/Cyclin D1 signaling pathway. RACGAP1 accelerated the invasion and metastasis of HCC via phosphorylation of GSK3β and nuclear translocation of β-catenin. Furthermore, by luciferase reporter assay and Chromatin immunoprecipitation (ChIP) assay, we confirmed Recombinant GA Binding Protein Transcription Factor Alpha (GABPA) regulated the transcription of RACGAP1. All these findings revealed that RACGAP1 promotes the progression of HCC through a novel mechanism, which might be a new therapeutic target for HCC patients.
Collapse
|
15
|
Liu R, Huang Y. CDC7 as a novel biomarker and druggable target in cancer. Clin Transl Oncol 2022; 24:1856-1864. [PMID: 35657477 DOI: 10.1007/s12094-022-02853-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 05/05/2022] [Indexed: 11/25/2022]
Abstract
Due to the bottlenecks encountered in traditional treatment for tumor, more effective drug targets need to be developed. Cell division cycle 7 kinase plays an important role in DNA replication, DNA repair and recombination signaling pathways. In this review, we first describe recent studies on the role of CDC7 in DNA replication in normal human tissues, and then we integrate new evidence focusing on the important role of CDC7 in replication stress tolerance of tumor cells and its impact on the prognosis of clinical oncology patients. Finally, we comb through the CDC7 inhibitors identified in recent studies as a reference for further research in clinical practice.
Collapse
Affiliation(s)
- Runze Liu
- National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-Targeting Theranostics, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Yong Huang
- National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-Targeting Theranostics, Guangxi Medical University, Nanning, 530021, Guangxi, China.
| |
Collapse
|
16
|
EGF-induced nuclear translocation of SHCBP1 promotes bladder cancer progression through inhibiting RACGAP1-mediated RAC1 inactivation. Cell Death Dis 2022; 13:39. [PMID: 35013128 PMCID: PMC8748695 DOI: 10.1038/s41419-021-04479-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 12/01/2021] [Accepted: 12/14/2021] [Indexed: 12/12/2022]
Abstract
Bladder cancer is a highly heterogeneous and aggressive malignancy with a poor prognosis. EGF/EGFR activation causes the detachment of SHC-binding protein 1 (SHCBP1) from SHC adapter protein 1 (SHC1), which subsequently translocates into the nucleus and promotes cancer development via multiple signaling pathways. However, the role of the EGF-SHCBP1 axis in bladder cancer progression remains unexplored. Herein, we report that SHCBP1 is upregulated in bladder cancer tissues and cells, with cytoplasmic or nuclear localization. Released SHCBP1 responds to EGF stimulation by translocating into the nucleus following Ser273 phosphorylation. Depletion of SHCBP1 reduces EGF-induced cell migration and invasiveness of bladder cancer cells. Mechanistically, SHCBP1 binds to RACGAP1 via its N-terminal domain of amino acids 1 ~ 428, and this interaction is enhanced following EGF treatment. Furthermore, SHCBP1 facilitates cell migration by inhibiting RACGAP-mediated GTP-RAC1 inactivation, whose activity is indispensable for cell movement. Collectively, we demonstrate that the EGF-SHCBP1-RACGAP1-RAC1 axis acts as a novel regulatory mechanism of bladder cancer progression, which offers a new clinical therapeutic strategy to combat bladder cancer.
Collapse
|