1
|
Li J, Zhang A, Li W, Duan Z, Li S, Fan Y, Hao H. Mitotic spindle positioning protein serves as prognostic biomarker in patients with colorectal cancer. Scand J Gastroenterol 2024; 59:1240-1248. [PMID: 39369263 DOI: 10.1080/00365521.2024.2411405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 09/18/2024] [Accepted: 09/27/2024] [Indexed: 10/07/2024]
Abstract
BACKGROUND Colorectal cancer (CRC) ranks among the most aggressive types of cancer globally. Currently, clinical tumor prognostic biomarkers still lack accuracy. Mitotic spindle positioning (MISP) protein connects microtubules to the actin cytoskeleton and adhesive plaques, playing a critical role in spindle positioning, orientation, and the process of cell division. MISP can regulate the malignant biological functions of pancreatic cancer and intrahepatic cholangiocarcinoma and it acts as biomarker for prognosis, but its role in CRC remains unclear. METHODS This study has collected 37 CRC tissue samples and 37 corresponding adjacent nontumor tissue samples, and 57 additional CRC tissues samples. Clinical data were obtained from the patients with CRC. MISP mRNA and protein expression levels were analyzed in normal control and CRC tissues using the GEPIA and Human Protein Atlas website. MISP protein levels in the collected tissues were analyzed using immunohistochemistry. RESULTS MISP mRNA and protein expression levels were significantly increased in CRC tissues compared to adjacent nontumor tissues. Higher MISP protein levels were associated with distant metastasis, recurrence, and lower survival rates. Kaplan-Meier analysis showed that high expression levels of MISP protein were associated with recurrence and death in CRC patients. In addition, a high expression level of MISP protein, lymph node metastasis, and distance metastasis were risk factors for recurrence and a poor prognosis in patients with CRC. CONCLUSION Elevated MISP protein correlated with tumor metastasis, recurrence, and lower survival rates in patients with CRC, and thus, MISP has the potential to become a prognostic marker for CRC.
Collapse
Affiliation(s)
- Jin Li
- Gastrointestinal Surgery, First Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, China
| | - Aimin Zhang
- Department of Gastroenterology, First Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, China
| | - Weilun Li
- Gastrointestinal Surgery, First Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, China
| | - Zeye Duan
- Gastrointestinal Surgery, First Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, China
| | - Suqin Li
- Gastrointestinal Surgery, First Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, China
| | - Yunyan Fan
- Gastrointestinal Surgery, First Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, China
| | - Haiyan Hao
- Infection Disease Department, Baotou City Fourth Hospital, Baotou, China
| |
Collapse
|
2
|
Liang L, Wang L, Liao Z, Ma L, Wang P, Zhao J, Wu J, Yang H. High-yield nanovesicles extruded from dental follicle stem cells promote the regeneration of periodontal tissues as an alternative of exosomes. J Clin Periodontol 2024; 51:1395-1407. [PMID: 38951121 DOI: 10.1111/jcpe.14036] [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: 12/14/2023] [Revised: 05/14/2024] [Accepted: 06/13/2024] [Indexed: 07/03/2024]
Abstract
AIM To identify an optimized strategy for the large-scale production of nanovesicles (NVs) that preserve the biological properties of exosomes (EXOs) for use in periodontal regeneration. MATERIALS AND METHODS NVs from dental follicle stem cells (DFSCs) were prepared through extrusion, and EXOs from DFSCs were isolated. The yield of both extruded NVs (eNVs) and EXOs were quantified through protein concentration and particle number analyses. Their pro-migration, pro-proliferation and pro-osteogenesis capacities were compared subsequently in vitro. Additionally, proteomics analysis was conducted. To further evaluate the periodontal regeneration potential of eNVs and EXOs, they were incorporated into collagen sponges and transplanted into periodontal defects in rats. In vivo imaging and H&E staining were utilized to verify their biodistribution and safety. Micro-Computed Tomography analysis and histological staining were performed to examine the regeneration of periodontal tissues. RESULTS The yield of eNVs was nearly 40 times higher than that of EXOs. Interestingly, in vitro experiments indicated that the pro-migration and pro-proliferation abilities of eNVs were superior, and the pro-osteogenesis potential was comparable to EXOs. More importantly, eNVs exhibited periodontal regenerative potential similar to that of EXOs. CONCLUSIONS Extrusion has proven to be an efficient method for generating numerous eNVs with the potential to replace EXOs in periodontal regeneration.
Collapse
Affiliation(s)
- Lu Liang
- Yunnan Key Laboratory of Stomatology, Kunming, China
- Department of Prosthodontics, kunming Medicine University School and Hospital of Stomatology, Kunming, China
| | - Limeiting Wang
- Yunnan Key Laboratory of Stomatology, Kunming, China
- Department of Prosthodontics, kunming Medicine University School and Hospital of Stomatology, Kunming, China
| | - Zhenhui Liao
- Yunnan Key Laboratory of Stomatology, Kunming, China
- Department of Prosthodontics, kunming Medicine University School and Hospital of Stomatology, Kunming, China
| | - Liya Ma
- Yunnan Key Laboratory of Stomatology, Kunming, China
- Department of Orthodontics, Kunming Medical University School and Hospital of Stomatology, Kunming, China
| | - Pinwen Wang
- Yunnan Key Laboratory of Stomatology, Kunming, China
- Department of Prosthodontics, kunming Medicine University School and Hospital of Stomatology, Kunming, China
| | - Junjie Zhao
- Yunnan Key Laboratory of Stomatology, Kunming, China
- Kunming Medical University School and Hospital of Stomatology, Kunming, China
| | - Jinyan Wu
- Yunnan Key Laboratory of Stomatology, Kunming, China
- Department of Prosthodontics, kunming Medicine University School and Hospital of Stomatology, Kunming, China
- Department of Endodontics, Kunming Medicine University School and Hospital of Stomatology, Kunming, China
| | - Hefeng Yang
- Yunnan Key Laboratory of Stomatology, Kunming, China
- Department of Prosthodontics, kunming Medicine University School and Hospital of Stomatology, Kunming, China
| |
Collapse
|
3
|
Wu J, Ren W, Liu J, Bai X. CUL1 exacerbates glucocorticoid-induced osteoporosis by enhancing ASAP1 ubiquitination. Hormones (Athens) 2024:10.1007/s42000-024-00599-y. [PMID: 39287759 DOI: 10.1007/s42000-024-00599-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 08/26/2024] [Indexed: 09/19/2024]
Abstract
BACKGROUND Glucocorticoid-induced osteoporosis is a leading secondary cause of osteoporosis. Cullin-1 (CUL1) levels are abnormally elevated in patients with osteoporosis, but the underlying mechanism remains unclear. The purpose of this study was to elucidate the mechanism of action of CUL1 in a glucocorticoid (dexamethasone, Dex)-induced osteoporosis model. METHODS C57BL/6J mice were intraperitoneally injected with Dex to establish an osteoporosis model. Mouse femur bone injury and bone formation were detected using hematoxylin-eosin or Masson staining. Apoptosis and cell cycle distribution were determined by flow cytometry. Alkaline phosphatase (ALP) activity and calcified nodules were monitored using ALP and Alizarin Red S staining. The molecular mechanism was validated by co-immunoprecipitation (Co-IP) and ubiquitination assays. RESULTS CUL1 expression was enhanced in the Dex-induced osteoporosis mouse model. CUL1 silencing moderated the Dex-induced cell proliferation and osteogenesis inhibition. Moreover, CUL1 promoted the ubiquitination and degradation of ASAP1 via the SKP1-CUL1-F-box (SCF)-FBXW7 complex. CUL1 induced apoptosis and repressed osteogenesis by ASAP1. CUL1 silencing alleviated the Dex-induced osteoporosis in mice. CONCLUSION CUL1 suppressed osteoblast proliferation and osteogenesis by promoting ASAP1 ubiquitination via the SCF-FBXW7 complex in glucocorticoid-induced osteoporosis.
Collapse
Affiliation(s)
- Jun Wu
- Dalian Medical University, No. 9, West Section of Lushun South Road, Dalian, 116041, Liaoning, P.R. China
- Department of Orthopaedics, The People's Hospital of Liaoning Province, 33 Wenyi Road, Shenyang, 110016, Liaoning, People's Republic of China
| | - Weijian Ren
- Department of Orthopaedics, The People's Hospital of Liaoning Province, 33 Wenyi Road, Shenyang, 110016, Liaoning, People's Republic of China
| | - Jun Liu
- Department of Orthopaedics, The People's Hospital of Liaoning Province, 33 Wenyi Road, Shenyang, 110016, Liaoning, People's Republic of China
| | - Xizhuang Bai
- Department of Orthopaedics, The People's Hospital of Liaoning Province, 33 Wenyi Road, Shenyang, 110016, Liaoning, People's Republic of China.
| |
Collapse
|
4
|
Zhao H, Liu Y, Wu Y, Cheng J, Li Y. Inhibition of ASAP1 Modulates the Tumor Immune Microenvironment and Suppresses Lung Cancer Metastasis via the p-STAT3 Signaling Pathway. Cell Biochem Biophys 2024; 82:2387-2399. [PMID: 38874840 DOI: 10.1007/s12013-024-01349-y] [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] [Accepted: 06/04/2024] [Indexed: 06/15/2024]
Abstract
ADP ribosylation factor guanylate kinase 1 (ASAP1), a key protein regulating cell migration and invasion, has attracted extensive attention in oncological research in recent years. This study aims to explore the effects of ASAP1 inhibition on lung cancer metastasis and its potential mechanisms, particularly how it modulates the tumor immune microenvironment through the p-STAT3 signaling pathway. In this study, shRNA technology was employed to specifically inhibit ASAP1 expression in lung cancer cell lines A549, NCI-H1299, and PC-9. The effects of ASAP1 inhibition on lung cancer cell viability, apoptosis, migration, and invasion were evaluated using CCK-8, TUNEL apoptosis detection, and cell migration and invasion assays. Furthermore, animal experiments were conducted to assess the in vivo effects of ASAP1 inhibition on lung cancer metastasis, and immunohistochemical analysis was performed to investigate changes in immune cells in lung metastasis models, further exploring its impact on the tumor immune microenvironment. The experimental results demonstrated that ASAP1 inhibition significantly reduced lung cancer cell viability, induced apoptosis in A549, NCI-H1299, and PC-9 cells, and suppressed the migration and invasion abilities of these cells. In vivo experiments revealed that ASAP1 inhibition effectively suppressed lung cancer metastasis and altered the tumor immune microenvironment by regulating immune cells. Moreover, we found that ASAP1 inhibition could decrease tumor cell proliferation and induce tumor apoptosis in lung metastasis models by inhibiting the p-STAT3 signaling pathway. This study confirms that ASAP1 inhibition can suppress lung cancer metastasis by modulating the tumor immune microenvironment through the inhibition of the p-STAT3 signaling pathway. These findings provide new targets for lung cancer treatment and a theoretical basis for developing novel strategies against lung cancer metastasis. Future research will further explore the mechanisms of ASAP1 in lung cancer metastasis and how to optimize treatment strategies for lung cancer patients by targeting ASAP1.
Collapse
Affiliation(s)
- Hongye Zhao
- The Department of Dermatology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Yongcun Liu
- The Department of Surgery, Shijiazhuang Traditional Chinese Medicine Hospital, Shijiazhuang, 050011, China
| | - Yunfan Wu
- The Department of Radiation Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Jingge Cheng
- The Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China.
| | - Yishuai Li
- The Department of Thoracic Surgery, Hebei Provincial Key Laboratory of pulmonary disease, Hebei Chest Hospital, Shijiazhuang, 050047, China.
| |
Collapse
|
5
|
Zhao Z, Zhou Y, Lv P, Zhou T, Liu H, Xie Y, Wu Z, Wang X, Zhao H, Zheng J, Jiang X. NSUN4 mediated RNA 5-methylcytosine promotes the malignant progression of glioma through improving the CDC42 mRNA stabilization. Cancer Lett 2024; 597:217059. [PMID: 38876383 DOI: 10.1016/j.canlet.2024.217059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/30/2024] [Accepted: 06/08/2024] [Indexed: 06/16/2024]
Abstract
5-Methylcytosine (m5C) methylation is a significant post-transcriptional modification that play a crucial role in the development and progression of numerous cancers. Whereas the functions and molecular mechanisms underlying m5C methylation in gliomas remain unclear. This study dedicated to explore changes of m5C levels and the clinical significance of the m5C writer NSUN4 in gliomas. We found that high m5C levels were negatively related to prognosis of patients with glioma. Moreover, gain- and loss-of-function experiments revealed the role of NSUN4 in enhancing m5C modification of mRNA to promote the malignant progression of glioma. Mechanistically speaking, NSUN4-mediated m5C alterations regulated ALYREF binding to CDC42 mRNA, thereby impacting the mRNA stability of CDC42. We also demonstrated that CDC42 promoted glioma proliferation, migration, and invasion by activating the PI3K-AKT pathway. Additionally, rescue experiments proved that CDC42 overexpression weaken the inhibitory effect of NSUN4 knockdown on the malignant progression of gliomas in vitro and in vivo. Our findings elucidated that NSUN4-mediated high m5C levels promote ALYREF binding to CDC42 mRNA and regulate its stability, thereby driving the malignant progression of glioma. This provides theoretical support for targeted the treatment of gliomas.
Collapse
Affiliation(s)
- Zhen Zhao
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yujie Zhou
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Peng Lv
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ting Zhou
- Department of Gynaecology and Obstetrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Hanyuan Liu
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Youxi Xie
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zhipeng Wu
- Department of Neurosurgery, Weifang People's Hospital, Weifang, China
| | - Xuan Wang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hongyang Zhao
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Jianglin Zheng
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Xiaobing Jiang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
6
|
Luo Q, Zhang S, Yang F, Feng R, Xu Q, Chen X, Yang S. Role of ADP ribosylation factor guanylate kinase 1 in the malignant biological behavior of gastric cancer. Heliyon 2024; 10:e33255. [PMID: 39021998 PMCID: PMC11253526 DOI: 10.1016/j.heliyon.2024.e33255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 05/29/2024] [Accepted: 06/18/2024] [Indexed: 07/20/2024] Open
Abstract
Aim This study aims to investigate the influence of ASAP1 (ADP ribosylation factor guanylate kinase 1) on the malignant behavior of gastric cancer (GC) cells and to elucidate the potential molecular mechanisms involved in cancer development and progression. Methods We assessed the impact of ASAP1 overexpression and knockdown on GC cell malignancy using CCK8, colony formation, flow cytometry (Annexin V/propidium iodide), Transwell migration, invasion, and scratch assays. Western blot analysis was used to assess the effects of ASAP1 on angiogenesis, matrix metalloproteinases (MMPs), apoptotic proteins, epithelial-mesenchymal transition (EMT)-related proteins, as well as AKT and p-AKT. The influence of ASAP1 knockdown was also evaluated in nude mice bearing BGC823 cell-derived tumors. Results Our findings revealed that ASAP1 was significantly overexpressed in GC cells, enhancing their proliferation, invasion, and migration, while reducing apoptosis. Conversely, ASAP1 knockdown reversed these effects, markedly increasing the expression of cleaved-caspase 3 (Casp3), PARP, and the epithelial marker E-cadherin, and significantly decreasing MMP2, MMP9, VEGFA, and mesenchymal markers such as N-cadherin and vimentin. Additionally, it reduced AKT, and p-AKT levels (P < 0.01). Tumor growth in nude mice was suppressed following ASAP1 knockdown. Conclusion The overexpression of ASAP1 significantly promotes malignant behaviors in GC cells, whereas its knockdown diminishes these effects. This modulation is potentially through the downregulation of VEGFA, leading to reduced angiogenesis, Cleaved-Casp3 and Cleaved-PARP overexpression, and a decrease in MMPs, EMT, AKT, and p-AKT activity.
Collapse
Affiliation(s)
- Qiong Luo
- Departments of Oncology, Fuzhou, Fujian 350001, PR China
| | - Suyun Zhang
- Departments of Oncology, Fuzhou, Fujian 350001, PR China
| | - Fan Yang
- Departments of Pulmonary and Critical Care Medicine, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, PR China
| | - Rui Feng
- Departments of Oncology, Fuzhou, Fujian 350001, PR China
| | - Qian Xu
- Departments of Oncology, Fuzhou, Fujian 350001, PR China
| | - Xiangqi Chen
- Departments of Pulmonary and Critical Care Medicine, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, PR China
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, Fuzhou, Fujian 350001, PR China
| | - Sheng Yang
- Departments of Oncology, Fuzhou, Fujian 350001, PR China
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, Fuzhou, Fujian 350001, PR China
| |
Collapse
|
7
|
Liu J, Yuan Q, Guo H, Guan H, Hong Z, Shang D. Deciphering drug resistance in gastric cancer: Potential mechanisms and future perspectives. Biomed Pharmacother 2024; 173:116310. [PMID: 38394851 DOI: 10.1016/j.biopha.2024.116310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 02/07/2024] [Accepted: 02/17/2024] [Indexed: 02/25/2024] Open
Abstract
Gastric cancer (GC) is a malignant tumor that originates from the epithelium of the gastric mucosa. The latest global cancer statistics show that GC ranks fifth in incidence and fourth in mortality among all cancers, posing a serious threat to public health. While early-stage GC is primarily treated through surgery, chemotherapy is the frontline option for advanced cases. Currently, commonly used chemotherapy regimens include FOLFOX (oxaliplatin + leucovorin + 5-fluorouracil) and XELOX (oxaliplatin + capecitabine). However, with the widespread use of chemotherapy, an increasing number of cases of drug resistance have emerged. This article primarily explores the potential mechanisms of chemotherapy resistance in GC patients from five perspectives: cell death, tumor microenvironment, non-coding RNA, epigenetics, and epithelial-mesenchymal transition. Additionally, it proposes feasibility strategies to overcome drug resistance from four angles: cancer stem cells, tumor microenvironment, natural products, and combined therapy. The hope is that this article will provide guidance for researchers in the field and bring hope to more GC patients.
Collapse
Affiliation(s)
- Jiahua Liu
- First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Qihang Yuan
- First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Hui Guo
- First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Hewen Guan
- First Affiliated Hospital of Dalian Medical University, Dalian, China.
| | - Zhijun Hong
- First Affiliated Hospital of Dalian Medical University, Dalian, China.
| | - Dong Shang
- First Affiliated Hospital of Dalian Medical University, Dalian, China.
| |
Collapse
|
8
|
Shi L, Wang X, Guo S, Gou H, Shang H, Jiang X, Wei C, Wang J, Li C, Wang L, Zhao Z, Yu W, Yu J. TMEM65 promotes gastric tumorigenesis by targeting YWHAZ to activate PI3K-Akt-mTOR pathway and is a therapeutic target. Oncogene 2024; 43:931-943. [PMID: 38341472 PMCID: PMC10959749 DOI: 10.1038/s41388-024-02959-9] [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: 03/13/2023] [Revised: 01/07/2024] [Accepted: 01/26/2024] [Indexed: 02/12/2024]
Abstract
Copy number alterations are crucial for the development of gastric cancer (GC). Here, we identified Transmembrane Protein 65 (TMEM65) amplification by genomic hybridization microarray to profile copy-number variations in GC. TMEM65 mRNA level was significantly up-regulated in GC compared to adjacent normal tissues, and was positively associated with TMEM65 amplification. High TMEM65 expression or DNA copy number predicts poor prognosis (P < 0.05) in GC. Furtherly, GC patients with TMEM65 amplification (n = 129) or overexpression (n = 78) significantly associated with shortened survival. Ectopic expression of TMEM65 significantly promoted cell proliferation, cell cycle progression and cell migration/invasion ability, but inhibited apoptosis (all P < 0.05). Conversely, silencing of TMEM65 in GC cells showed opposite abilities on cell function in vitro and suppressed tumor growth and lung metastasis in vivo (all P < 0.01). Moreover, TMEM65 depletion by VNP-encapsulated TMEM65-siRNA significantly suppressed tumor growth in subcutaneous xenograft model. Mechanistically, TMEM65 exerted oncogenic effects through activating PI3K-Akt-mTOR signaling pathway, as evidenced of increased expression of key regulators (p-Akt, p-GSK-3β, p-mTOR) by Western blot. YWHAZ (Tyrosine 3-Monooxygenase/Tryptophan 5-Monooxygenase) was identified as a direct downstream effector of TMEM65. Direct binding of TMEM65 with YWHAZ in the cytoplasm inhibited ubiquitin-mediated degradation of YWHAZ. Moreover, oncogenic effect of TMEM65 was partly dependent on YWHAZ. In conclusion, TMEM65 promotes gastric tumorigenesis by activating PI3K-Akt-mTOR signaling via cooperating with YWHAZ. TMEM65 overexpression may serve as an independent new biomarker and is a therapeutic target in GC.
Collapse
Affiliation(s)
- Lingxue Shi
- Departments of Endoscopy Center, The First Hospital of Hebei Medical University, Shijiazhuang, China
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong-Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
- The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiaohong Wang
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong-Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
- Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital and Institute, Beijing, China
| | - Shang Guo
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong-Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
- The First Hospital of Hebei Medical University, Shijiazhuang, China
- Gastrointestinal Disease Centre, Hebei Key Laboratory of Colorectal Cancer Precision Diagnosis and Treatment, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Hongyan Gou
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong-Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Haiyun Shang
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong-Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Xiaojia Jiang
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong-Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Chunxian Wei
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong-Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jia Wang
- The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Chao Li
- Departments of Endoscopy Center, The First Hospital of Hebei Medical University, Shijiazhuang, China
- The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Lihong Wang
- The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zengren Zhao
- The First Hospital of Hebei Medical University, Shijiazhuang, China.
- Gastrointestinal Disease Centre, Hebei Key Laboratory of Colorectal Cancer Precision Diagnosis and Treatment, The First Hospital of Hebei Medical University, Shijiazhuang, China.
| | - Weifang Yu
- Departments of Endoscopy Center, The First Hospital of Hebei Medical University, Shijiazhuang, China.
- The First Hospital of Hebei Medical University, Shijiazhuang, China.
| | - Jun Yu
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong-Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
9
|
Tian S, Chen M, Jing W, Meng Q, Wu J. miR-1204 Positioning in 8q24.21 Involved in the Tumorigenesis of Colorectal Cancer by Targeting MASPIN. Protein Pept Lett 2024; 31:544-558. [PMID: 39082173 DOI: 10.2174/0109298665305114240718072029] [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: 02/17/2024] [Revised: 05/16/2024] [Accepted: 06/13/2024] [Indexed: 10/19/2024]
Abstract
BACKGROUND Colorectal cancer remains to be the third leading cause of cancer mortality rates. Despite the diverse effects of the miRNA cluster located in PVT1 of 8q24.21 across various tumors, the specific biological function in colorectal cancer has not been clarified. METHODS The amplification of the miR-1204 cluster was analyzed with the cBioPortal database, while the expression and survival analysis of the miRNAs in the cluster were obtained from several GEO databases of colorectal cancer. To investigate the functional role of miR-1204 in colorectal cancer, overexpression and silencing experiments were performed by miR-1204 mimic and inhibitor transfection in colorectal cancer cell lines, respectively. Then, the effects of miR-1204 on cell proliferation were assessed through CCK-8, colony formation, and Edu assay. In addition, cell migration was evaluated using wound healing and Transwell assay. Moreover, candidate genes identified through RNA sequencing and predicted databases were identified and validated using PCR and western blot. A Dual-luciferase reporter experiment was conducted to identify MASPIN as the target gene of miR-1204. RESULTS In colorectal cancer, the miR-1204 cluster exhibited high amplification, and the expression levels of several cluster miRNAs were also significantly increased. Furthermore, miR-1204 was found to be significantly associated with disease-specific survival according to the analysis of GSE17536. Functional experiments demonstrated that transfection of miR-1204 mimic or inhibitor could enhance or decrease cancer cell proliferation and migration. MASPIN was identified as a target of miR-1204. Additionally, the overexpression of MASPIN partially rescued the effect of miR-1204 mimics on tumorigenic abilities in LOVO cells. CONCLUSION miR-1204 positioning in 8q24.21 promotes the proliferation and migration of colorectal cancer cells by targeting MASPIN.
Collapse
Affiliation(s)
- Simeng Tian
- Department of Surgery, The 1st Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Meilin Chen
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
- Department of Pathology, XiaMen SuSong Hospital, Xia- Men, China
| | - Wanting Jing
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - Qinghui Meng
- Department of Surgery, The 1st Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jie Wu
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| |
Collapse
|
10
|
Zhang X, Yu T, Gao G, Xu J, Lin R, Pan Z, Liu J, Feng W. Cell division cycle 42 effector protein 4 inhibits prostate cancer progression by suppressing ERK signaling pathway. BIOMOLECULES & BIOMEDICINE 2023; 24:840-847. [PMID: 38153517 PMCID: PMC11293231 DOI: 10.17305/bb.2023.9986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 12/09/2023] [Accepted: 12/27/2023] [Indexed: 12/29/2023]
Abstract
Prostate cancer (PCa) is the most common malignancy among men worldwide. The cell division cycle 42 effector protein 4 (CDC42EP4) functions downstream of CDC42, yet its role and molecular mechanisms in PCa remain unexplored. This study aimed to elucidate the role of CDC42EP4 in the progression of PCa and its underlying mechanisms. Bioinformatical analysis indicated that CDC42EP4 expression was significantly lower in PCa tissue compared to normal prostate tissue. Cellular phenotyping analysis suggested that CDC42EP4 markedly inhibited the proliferation, migration, and invasion of PCa cells. Xenograft tumor assays further demonstrated that CDC42EP4 suppressed the growth of PCa cells in vivo. Mechanistically, the study established that CDC42EP4 inhibited the ERK pathway in PCa cells. Additionally, the ERK pathway inhibitor PD0325901 was employed, revealing that PD0325901 significantly nullified the effects of CDC42EP4 on PCa cell proliferation, migration, and invasion. Collectively, our findings demonstrate that CDC42EP4 acts as a critical tumor suppressor gene, inhibiting PCa cell proliferation, migration, and invasion through the ERK pathway, thereby presenting potential targets for PCa therapy.
Collapse
Affiliation(s)
- Xiaowen Zhang
- School of Life Science and Technology, Weifang Medical University, Weifang, China
| | - Tao Yu
- School of Life Science and Technology, Weifang Medical University, Weifang, China
| | - Guojun Gao
- Department of Urology Surgery, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Junbao Xu
- Cancer Center, Shandong Public Health Clinical Center, Shandong, China
| | - Ruihui Lin
- School of Life Science and Technology, Weifang Medical University, Weifang, China
| | - Zhifang Pan
- School of Life Science and Technology, Weifang Medical University, Weifang, China
| | - Jianying Liu
- Department of Nuclear Medicine, The Third Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Weiguo Feng
- School of Life Science and Technology, Weifang Medical University, Weifang, China
| |
Collapse
|
11
|
Chen H, Ma R, Zhou B, Yang X, Duan F, Wang G. Integrated immunological analysis of single-cell and bulky tissue transcriptomes reveals the role of interactions between M0 macrophages and naïve CD4 + T cells in the immunosuppressive microenvironment of cervical cancer. Comput Biol Med 2023; 163:107151. [PMID: 37348264 DOI: 10.1016/j.compbiomed.2023.107151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/27/2023] [Accepted: 06/07/2023] [Indexed: 06/24/2023]
Abstract
In recent decades, the incidence and mortality of cervical cancer have declined in developed countries due to the implementation of screening and vaccination programs. However, cervical cancer remains one of the major culprits of cancer-related deaths in young women. Current studies have found that immune cell-related intercellular communication in the tumor microenvironment has a large impact on the construction of the immunosuppressive microenvironment. In this study, we performed a comprehensive immune analysis on bulk RNA-seq and scRNA-seq data obtained from cervical cancer and revealed that two highly plastic cell populations, M0 macrophages and naïve CD4+ T cells, were significantly correlated with prognosis and clinical phenotypes. Notably, signaling between M0 macrophages and naïve CD4+ T cells as well as intracellular transcription factor activity were significantly altered in the tumor state. Furthermore, we identified overlapping genes between the transcription factor target genes of M0 macrophages or naïve CD4+ T cells and the differentially expressed genes in each type of cell, and these overlapping genes were subsequently subjected to an analysis using the LASSO regression model. Finally, we generated a score index that was significantly associated with the clinical prognosis of cervical cancer. In conclusion, interventions to improve the communication between M0 macrophages and naïve CD4+ T cells may help to improve the immunosuppressive microenvironment of cervical cancer and prevent immune evasion. The relevant molecular mechanisms need to be further validated by experimental and cohort studies.
Collapse
Affiliation(s)
- Huaqiu Chen
- Center of Genetic Testing, The First Affiliated Hospital of Dali University, Dali, Yunnan Province, 671000, China; Department of Laboratory, Xichang People's Hospital, Sichuan, 615000, China
| | - Rong Ma
- Center of Genetic Testing, The First Affiliated Hospital of Dali University, Dali, Yunnan Province, 671000, China; Department of Laboratory, The First People's Hospital of Qujing, Yunnan Province, 655000, China
| | - Bingjie Zhou
- Center of Genetic Testing, The First Affiliated Hospital of Dali University, Dali, Yunnan Province, 671000, China; Maternity and Obstetrics Department of Fangshan District Maternity and Child Health Hospital of Beijing, Fangshan District of Beijing, 102488, China
| | - Xitong Yang
- Center of Genetic Testing, The First Affiliated Hospital of Dali University, Dali, Yunnan Province, 671000, China
| | - Fuhui Duan
- Center of Genetic Testing, The First Affiliated Hospital of Dali University, Dali, Yunnan Province, 671000, China
| | - Guangming Wang
- Center of Genetic Testing, The First Affiliated Hospital of Dali University, Dali, Yunnan Province, 671000, China.
| |
Collapse
|