1
|
Yan Z, He L, Yuan J, Niu Y, Shuai S, Luo S, Du C, Rao H. The splicing factor SRRM2 modulates two S6K kinases to promote colorectal cancer growth. Oncogene 2025; 44:1284-1299. [PMID: 39956864 DOI: 10.1038/s41388-025-03307-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 01/16/2025] [Accepted: 02/10/2025] [Indexed: 02/18/2025]
Abstract
The mechanistic target of rapamycin (mTOR) pathway plays a critical role in cell growth and metabolic homeostasis. The ribosomal protein S6 kinases S6K1 and S6K2 are the major effectors of the mTOR pathway key to translation efficiency, but the underlying regulatory mechanisms remain largely unclear. In this study, we searched for mTOR regulators and found that the splicing factor SRRM2 modulates the levels of S6K1 and S6K2, thereby activating the mTOR-S6K pathway. Interestingly, SRRM2 facilitates the expression of S6K2 by modulating alternative splicing, and enhances the stability of the S6K1 protein by regulating the E3 ubiquitin ligase WWP2. Moreover, SRRM2 is highly expressed in colorectal cancer (CRC) tissues and is associated with a poor prognosis. SRRM2 promotes CRC growth in vitro and in vivo. Combined, these data reveal an oncogenic role of SRRM2 in CRC through activating the mTOR-S6K pathway by two different approaches, further suggesting SRRM2 as a potential therapeutic target for CRC.
Collapse
Affiliation(s)
- Zhengwei Yan
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Luling He
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Jiawei Yuan
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Yulong Niu
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Shimin Shuai
- Department of Human Cell Biology and Genetics, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Shiwen Luo
- School of Medicine, Nanchang University, Nanchang, Jiangxi, China
| | - Changzheng Du
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, China
- Beijing Tsinghua Changgung Hospital & Tsinghua University School of Medicine, 168 Litang Road, Changping District, Beijing, 102218, PR China
| | - Hai Rao
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, China.
- Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen, China.
| |
Collapse
|
2
|
Cheng D, Liu Z, Sun R, Jiang Y, Zeng Z, Zhao R, Mo Z. Overexpression of mir-489-3p inhibits proliferation and migration of non-small cell lung cancer cells by suppressing the HER2/PI3K/AKT/Snail signaling pathway. Heliyon 2024; 10:e35832. [PMID: 39224367 PMCID: PMC11367056 DOI: 10.1016/j.heliyon.2024.e35832] [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: 12/19/2023] [Revised: 08/02/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024] Open
Abstract
Background Lung cancer is a highly prevalent malignancy with significant morbidity and mortality rates. MiR-489-3p, a microRNA, has been identified as a regulator of tumor cell proliferation and invasion. Its expression is downregulated in non-small cell lung cancer (NSCLC). Elucidating the molecular mechanisms underlying miR-489-3p's role in NSCLC pathogenesis is crucial for identifying potential diagnostic and therapeutic targets. Methods To investigate the molecular mechanism of miR-489-3p in NSCLC, this study utilized A549, a commonly used NSCLC cell line. MiR-489-3p mimics and inhibitors were transfected into A549 cells. Additionally, co-transfection experiments using wortmannin, an inhibitor of the PI3K/AKT pathway, were performed. Expression of miR-489-3p and related proteins was analyzed by Western blotting and quantitative real-time PCR (qRT-PCR). Cell migration and proliferation were assessed by wound healing and colony formation assays, respectively. Results Overexpression of miR-489-3p significantly inhibited the proliferation and migration of A549 cells. This inhibitory effect was further enhanced upon co-transfected with wortmannin. Analysis of human lung specimens showed increased expression of HER2, PI3K, and AKT in lung adenocarcinoma tissues compared to adjacent non-cancerous tissues. Conclusions These findings suggest that miR-489-3p overexpression may inhibit NSCLC cell proliferation and migration by suppressing the HER2/PI3K/AKT/Snail signaling pathway. This study elucidates miR-489-3p's molecular mechanisms in NSCLC and provides experimental basis for identifying early diagnostic markers and novel therapeutic targets.
Collapse
Affiliation(s)
- Di Cheng
- The First Affiliated Hospital of Shaoyang University, Shaoyang, 422000, Hunan, China
- Guangxi Key Laboratory of Diabetic Systems Medicine, Department of Histology and Embryology, Guilin Medical University, Guilin, 541199, Guangxi, China
| | - Zhong Liu
- The First Affiliated Hospital of Shaoyang University, Shaoyang, 422000, Hunan, China
- Joint Laboratory of Chronic Disease Prevention and Research in Guilin Medical University & Hunan Mingshun, Shaodong, 422800, China
| | - Renren Sun
- Guangxi Key Laboratory of Diabetic Systems Medicine, Department of Histology and Embryology, Guilin Medical University, Guilin, 541199, Guangxi, China
| | - Yun Jiang
- The First Affiliated Hospital of Shaoyang University, Shaoyang, 422000, Hunan, China
| | - Zhaoming Zeng
- Joint Laboratory of Chronic Disease Prevention and Research in Guilin Medical University & Hunan Mingshun, Shaodong, 422800, China
| | - Rui Zhao
- The First Affiliated Hospital of Shaoyang University, Shaoyang, 422000, Hunan, China
| | - Zhongcheng Mo
- Guangxi Key Laboratory of Diabetic Systems Medicine, Department of Histology and Embryology, Guilin Medical University, Guilin, 541199, Guangxi, China
- Joint Laboratory of Chronic Disease Prevention and Research in Guilin Medical University & Hunan Mingshun, Shaodong, 422800, China
| |
Collapse
|
3
|
Jia Z, Wan X. Correlation and influencing factors analysis of colorectal polyps with Helicobacter pylori Infection and p-S6K1 expression. BMC Infect Dis 2023; 23:794. [PMID: 37964239 PMCID: PMC10644558 DOI: 10.1186/s12879-023-08791-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] [Received: 05/10/2023] [Accepted: 11/04/2023] [Indexed: 11/16/2023] Open
Abstract
OBJECTIVE To investigate the correlation between colorectal polyps (CRP) and Helicobacter pylori (H. pylori) infection, and the correlation between CRP and the expression of phosphorylated ribosomal protein S6 kinase (p-S6K1). Besides, its related influencing factors were determined in the present study. METHODS A total of 191 subjects who underwent colonoscopy in our hospital from January 2020 to February 2022 were selected for this study. Among them, 141 patients were diagnosed with CRP, and the other 50 subjects were no significant colorectal abnormalities. 141 CRP patients were divided into H. pylori-positive group (n = 89) and H. pylori-negative group (n = 52) according to the results of the H. pylori test. The expression of p-S6K1 in CRP tissue was detected. The relationship between the p-S6K1 expression and the clinicopathological characteristics of CRP patients was analyzed. The logistic analysis of factors influencing the occurrence of CRP was performed. RESULTS There were significant differences in pathological type, site of disease, the number and size of polyps between the H. pylori negative group and the H. pylori positive group (P < 0.001, P = 0.037, P = 0.042 and P = 0.039). The percentage of the p-S6K1 positive expression in polyp tissues was higher than that in normal tissue and parapolyp tissues (P < 0.001). The p-S6K1 negative group showed significant difference in the number and pathological type of polyps and the presence or absence of a pedicle as compared with the p-S6K1 positive group (P = 0.006, P < 0.001 and P = 0.012). Logistic multifactor analysis showed that BMI, H. pylori infection, smoking history, ApoB, Lp(a) and the p-S6K1 positive expression were all risk factors for the development of CRP (P = 0.025, P = 0.020, P = 0.010, P = 0.005, P = 0.043 and P < 0.001). CONCLUSION H. pylori infection was closely related to the pathological type, location, and the number and size of CRP. p-S6K1 was highly expressed in CRP, and was positively related to the number, the pathological type and pedicle of polyps. H. pylori infection and the positive p-S6K1 expression were independent risk factors for CRP. By exploring the association between H. pylori infection as well as p-S6K1 and CRP, it is hoped that it will help to formulate a more rigorous colorectal cancer screening program for H. pylori-positive individuals, and at the same time find a new direction for the prevention of CRP and colorectal cancer, and provide some help for future research.
Collapse
Affiliation(s)
- Zeming Jia
- Department of General Surgery, Xiangya Hospital, Central South University, No. 87 Xiangya Road, 410008, Changsha, Hunan, P R China
| | - Xiaoping Wan
- Department of General Surgery, Xiangya Hospital, Central South University, No. 87 Xiangya Road, 410008, Changsha, Hunan, P R China.
| |
Collapse
|
4
|
Li H, Cheng Z, Jiang B, Shao X, Xu M. Prognosis value and positive association of Rab1A/IL4Rα aberrant expression in gastric cancer. Sci Rep 2023; 13:6964. [PMID: 37117331 PMCID: PMC10147632 DOI: 10.1038/s41598-023-33955-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 04/21/2023] [Indexed: 04/30/2023] Open
Abstract
Gastric cancer (GC) is the most common gastrointestinal cancer and the leading cause of worldwide cancer-associated mortality. Several GC patients are diagnosed at the advanced stage with an unsatisfactory 5-year survival rate. Rab1A was significantly associated with IL4Rα expression in non-small cell lung cancer. However, their potential correlation in expression and prognosis remains largely unknown in GC. In this study, Rab1A/IL-4Rα was significantly increased in GC than in para-cancerous tissues, and Rab1A/IL-4Rα overexpression caused poor prognosis among GC patients. Rab1A expression was significantly correlated with IL-4Rα expression in GC tissues, as determined by IHC analysis. In addition, the mRNA expression of Rab1A was closely linked with the IL-4Rα mRNA expression in GC tissue expressed by qPCR. Furthermore, the Kaplan-Meier analysis demonstrated that the group with negative Rab1A and IL-4Rα expression had longer 5-year survival rates than the other group. Besides, the group with positive Rab1A and IL-4Rα expression had a worse prognosis than the other group. Finally, nomograms revealed the overall 3 and 5-year survival determined crucial roles of Rab1A/IL-4Rα expression in predicting the prognosis of GC patients. Therefore, Rab1A/IL-4Rα is vital in GC, providing a novel perspective on targeted GC therapy.
Collapse
Affiliation(s)
- Haoran Li
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241000, China
| | - Zhengwu Cheng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241000, China
| | - Bin Jiang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241000, China
| | - Xinyu Shao
- Department of Gastroenterology, Suzhou Municipal Hospital, Affiliated Suzhou Hospital of Nanjing Medical University, No. 242 Guangji Road, Suzhou, 215006, Jiangsu, China.
| | - Menglin Xu
- Department of Oncology, The First Affiliated Hospital of Wannan Medical College, No. 2 Zheshan West Road, Jinghu District, Wuhu, 241000, Anhui, China.
| |
Collapse
|
5
|
Gu M, Jiang B, Li H, Zhu D, Jiang Y, Xu W. Aldolase A promotes cell proliferation and cisplatin resistance via the EGFR pathway in gastric cancer. Am J Transl Res 2022; 14:6586-6595. [PMID: 36247245 PMCID: PMC9556458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 08/03/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND Gastric cancer is the third leading cause of cancer-related mortality worldwide, and the 5-year survival rate remains poor, globally. Overexpression of Aldolase A (ALDOA) has been linked to tumor cell proliferation and metastasis in numerous cancer types, including pancreatic, colorectal, hepatocellular carcinoma, and lung cancer. Although the significance of ALDOA as a potential biomarker in GC prognosis has been reported, its potential role and possible mechanism of ALDOA in GC cell sensitivity to chemotherapy remains to be elucidated. METHODS The GEPIA platform and clinical samples were used to investigate ALDOA expression in GC tumors and neighboring normal tissues. The CCK8 and colony formation tests were used to examine whether ALDOA increased GC cell proliferation and decreased resistance to the chemotherapy drug cisplatin. Furthermore, the underlying molecular mechanisms were elucidated. RESULTS Overexpression of ADOLA was seen in GC tumors and GC cells. Prognostic markers, i.e., invasion depth, tumor size, and metastasis of lymph node were all negatively impacted by ADOLA overexpression. Following ADOLA knockdown, in vitro proliferation of AGS cells was decreased and drug resistance was reduced. Conversely, ADOLA overexpression exhibited an inverse effect in MKN45 cells. ALDOA knockdown dramatically slowed the development of GC tumors in in vivo experiments. Mechanistically, ADOLA regulated the activity of epidermal growth factor receptor (EGFR), its downstream molecue the extracellular signal-regulated kinase 1/2 (ERK1/2) and protein kinase B (AKT) signaling pathway in GC cells. Moreover, in the absence of EGFR, ALDOA overexpression had no effect on GC cell growth. CONCLUSION In the EGFR signaling pathway, ADOLA boosted the proliferation and cisplatin resistance of GC cells, making it a viable GC therapeutic target.
Collapse
Affiliation(s)
- Menghui Gu
- Department of Gastrointestinal Surgery, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical UniversitySuzhou 215002, China
| | - Bin Jiang
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of Wannan Medical CollegeWuhu 241000, China
| | - Hao Li
- Department of Gastrointestinal Surgery, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical UniversitySuzhou 215002, China
| | - Dawei Zhu
- Department of Gastrointestinal Surgery, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical UniversitySuzhou 215002, China
| | - Yaqi Jiang
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of Wannan Medical CollegeWuhu 241000, China
| | - Wei Xu
- Department of Gastrointestinal Surgery, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical UniversitySuzhou 215002, China
| |
Collapse
|
6
|
Zhou J, Fan Q, Li J, Wu J, Huang J, Zhang Y, He X. Knockdown of MAGE-A6 enhanced the irradiation sensitivity of non-small cell lung cancer cells by activating the AMPK pathway. ENVIRONMENTAL TOXICOLOGY 2022; 37:1711-1722. [PMID: 35285568 DOI: 10.1002/tox.23519] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 01/25/2022] [Accepted: 03/06/2022] [Indexed: 06/14/2023]
Abstract
Non-small cell lung cancer is a common respiratory tumor. The mortality rate of lung cancer patients has continued to rise in recent years. Several studies revealed that the expression of melanoma antigen 6 (MAGE-A6) promoted the development of multiple types of cancer. In addition, the suppression of AMPK pathway could restrict the radiosensitization of prostate cancer cells. Inhibition of MAGE-A6 activated the AMPK pathway in colorectal cancer cells. However, whether the MAGE-A6 could regulate the radiosensitivity of non-small cell lung cancer cells by regulating of the AMPK pathway is unclear. In this study, we established the MAGE-A6 knockdown in A549 and H1299 cells. Next, the apoptosis and proliferation of these cells were detected by the flow cytometry analysis and colony formation assay after the irradiation, respectively. Then, the expression of p-AMPKα1 and p-S6K1 in these cells was explored by the western blotting. After that, we inhibited the expression of AMPKα1 in MAGE-A6 knockdown cells. The proliferation and apoptosis of these cells were detected with colony formation assay and flow cytometry analysis. Finally, the tumor formation of these cells was detected in nude mice. Our results showed that inhibition of MAGE-A6 suppressed the proliferation and aggravated the apoptosis of A549 and H1299 cells after the irradiation. Knockdown of MAGE-A6 activated the expression of p-AMPKα1 and repressed the expression of p-S6K1 in these cells. Suppression of AMPKα1 in MAGE-A6 knockdown cells abolished these effects. Knockdown of MAGE-A6 also enhanced the radiosensitivity of these cells in vivo. These results suggested that inhibition of MAGE-A6 promoted the radiosensitivity of non-small cell lung cancer cells by activating AMPK pathway. Therefore, MAGE-6 has the potential to be explored as the therapeutic target for the treatment of non-small cell lung cancer in clinical.
Collapse
Affiliation(s)
- Jialiang Zhou
- Department of Radiation Oncology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Qiang Fan
- Department of Radiation Oncology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Jie Li
- Department of Interventional Radiology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Jia Wu
- Department of Radiation Oncology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Jianfeng Huang
- Department of Radiation Oncology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Yunxia Zhang
- Department of Radiation Oncology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Xiuyun He
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
7
|
Rah B, Rather RA, Bhat GR, Baba AB, Mushtaq I, Farooq M, Yousuf T, Dar SB, Parveen S, Hassan R, Mohammad F, Qassim I, Bhat A, Ali S, Zargar MH, Afroze D. JAK/STAT Signaling: Molecular Targets, Therapeutic Opportunities, and Limitations of Targeted Inhibitions in Solid Malignancies. Front Pharmacol 2022; 13:821344. [PMID: 35401182 PMCID: PMC8987160 DOI: 10.3389/fphar.2022.821344] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 02/14/2022] [Indexed: 12/24/2022] Open
Abstract
JAK/STAT signaling pathway is one of the important regulatory signaling cascades for the myriad of cellular processes initiated by various types of ligands such as growth factors, hormones, and cytokines. The physiological processes regulated by JAK/STAT signaling are immune regulation, cell proliferation, cell survival, apoptosis and hematopoiesis of myeloid and non-myeloid cells. Dysregulation of JAK/STAT signaling is reported in various immunological disorders, hematological and other solid malignancies through various oncogenic activation mutations in receptors, downstream mediators, and associated transcriptional factors such as STATs. STATs typically have a dual role when explored in the context of cancer. While several members of the STAT family are involved in malignancies, however, a few members which include STAT3 and STAT5 are linked to tumor initiation and progression. Other STAT members such as STAT1 and STAT2 are pivotal for antitumor defense and maintenance of an effective and long-term immune response through evolutionarily conserved programs. The effects of JAK/STAT signaling and the persistent activation of STATs in tumor cell survival; proliferation and invasion have made the JAK/STAT pathway an ideal target for drug development and cancer therapy. Therefore, understanding the intricate JAK/STAT signaling in the pathogenesis of solid malignancies needs extensive research. A better understanding of the functionally redundant roles of JAKs and STATs may provide a rationale for improving existing cancer therapies which have deleterious effects on normal cells and to identifying novel targets for therapeutic intervention in solid malignancies.
Collapse
|
8
|
The Timing and Duration of Folate Restriction Differentially Impacts Colon Carcinogenesis. Nutrients 2021; 14:nu14010016. [PMID: 35010891 PMCID: PMC8746403 DOI: 10.3390/nu14010016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 12/17/2021] [Accepted: 12/17/2021] [Indexed: 01/27/2023] Open
Abstract
Diet plays a crucial role in the development of colorectal cancer (CRC). Of particular importance, folate, present in foods and supplements, is a crucial modulator of CRC risk. The role of folate, and, specifically, the synthetic variant, folic acid, in the primary prevention of CRC has not been fully elucidated. Animal studies varied considerably in the timing, duration, and supplementation of folates, leading to equivocal results. Our work attempts to isolate these variables to ascertain the role of folic acid in CRC initiation, as we previously demonstrated that folate restriction conferred protection against CRC initiation in a β-pol haploinsufficient mouse model. Here we demonstrated that prior adaptation to folate restriction altered the response to carcinogen exposure in wild-type C57BL/6 mice. Mice adapted to folate restriction for 8 weeks were protected from CRC initiation compared to mice placed on folate restriction for 1 week, irrespective of antibiotic supplementation. Through analyses of mTOR signaling, DNA methyltransferase, and DNA repair, we have identified factors that may play a critical role in the differential responses to folate restriction. Furthermore, the timing and duration of folate restriction altered these pathways differently in the absence of carcinogenic insult. These results represent novel findings, as we were able to show that, in the same model and under controlled conditions, folate restriction produced contrasting results depending on the timing and duration of the intervention.
Collapse
|
9
|
Zhang S, Lin X, Hou Q, Hu Z, Wang Y, Wang Z. Regulation of mTORC1 by amino acids in mammalian cells: A general picture of recent advances. ACTA ACUST UNITED AC 2021; 7:1009-1023. [PMID: 34738031 PMCID: PMC8536509 DOI: 10.1016/j.aninu.2021.05.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 05/13/2021] [Accepted: 05/18/2021] [Indexed: 12/11/2022]
Abstract
The mechanistic target of rapamycin complex 1 (mTORC1) integrates various types of signal inputs, such as energy, growth factors, and amino acids to regulate cell growth and proliferation mainly through the 2 direct downstream targets, eukaryotic translation initiation factor 4E-binding protein 1 (4EBP1) and ribosomal protein S6 kinase 1 (S6K1). Most of the signal arms upstream of mTORC1 including energy status, stress signals, and growth factors converge on the tuberous sclerosis complex (TSC) - Ras homologue enriched in brain (Rheb) axis. Amino acids, however, are distinct from other signals and modulate mTORC1 using a unique pathway. In recent years, the transmission mechanism of amino acid signals upstream of mTORC1 has been gradually elucidated, and some sensors or signal transmission pathways for individual amino acids have also been discovered. With the help of these findings, we propose a general picture of recent advances, which demonstrates that various amino acids from lysosomes, cytoplasm, and Golgi are sensed by their respective sensors. These signals converge on mTORC1 and form a huge and complicated signal network with multiple synergies, antagonisms, and feedback mechanisms.
Collapse
Affiliation(s)
- Shizhe Zhang
- Key Laboratory of Ruminant Nutrition and Physiology, College of Animal Science and Technology, Shandong Agricultural University, No. 61, Daizong Street, Tai'an, Shandong, China
| | - Xueyan Lin
- Key Laboratory of Ruminant Nutrition and Physiology, College of Animal Science and Technology, Shandong Agricultural University, No. 61, Daizong Street, Tai'an, Shandong, China
| | - Qiuling Hou
- Key Laboratory of Ruminant Nutrition and Physiology, College of Animal Science and Technology, Shandong Agricultural University, No. 61, Daizong Street, Tai'an, Shandong, China
| | - Zhiyong Hu
- Key Laboratory of Ruminant Nutrition and Physiology, College of Animal Science and Technology, Shandong Agricultural University, No. 61, Daizong Street, Tai'an, Shandong, China
| | - Yun Wang
- Key Laboratory of Ruminant Nutrition and Physiology, College of Animal Science and Technology, Shandong Agricultural University, No. 61, Daizong Street, Tai'an, Shandong, China
| | - Zhonghua Wang
- Key Laboratory of Ruminant Nutrition and Physiology, College of Animal Science and Technology, Shandong Agricultural University, No. 61, Daizong Street, Tai'an, Shandong, China
| |
Collapse
|
10
|
Rab1A promotes cell proliferation and migration by upregulating Gli1 in colorectal cancer. Sci Rep 2021; 11:16243. [PMID: 34376787 PMCID: PMC8355269 DOI: 10.1038/s41598-021-95798-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Accepted: 07/31/2021] [Indexed: 11/16/2022] Open
Abstract
Rab1A, as a highly conserved small guanosine triphosphatase (GTPase), plays contentious roles in different types of cancers. The role of Rab1A in colorectal cancer (CRC) has been described in previous studies, but the molecular mechanisms of Rab1A in CRC remain far from being addressed. In the present study, we found that Rab1A expression was significantly upregulated in CRC tissues and increased Rab1A expression correlated with tumor size, lymph node metastasis (LNM) and tumor-node-metastasis (TNM) stage of CRC patients. We also found that Rab1A exerts its promotive effect on CRC cell proliferation, migration and EMT progress. Further mechanistic experiments showed that glioma-associated oncogene-1 (Gli1), as a key transcriptional factor of the Hedgehog pathway, was implicated in Rab1A-mediated regulation of CRC cell proliferation and migration. In addition, Rab1A upregulated Gli1 expression through Smoothened homolog (SMO)-independent pathway. Finally, Rab1A activated mechanistic target of rapamycin (mTOR) signaling in CRC cells. Collectively, our results define Rab1A as a novel regulator of Gli1 to promote CRC cell proliferation and migration, and suggest that the Rab1A/mTOR/Gli1 axis may serve as a promising therapeutic target for the treatment of CRC.
Collapse
|
11
|
Abstract
Autophagy is a fundamental cellular process that has important roles in innate and adaptive immunity against a broad range of microbes. Many pathogenic microbes have evolved mechanisms to evade or exploit autophagy. It has been previously demonstrated that induction of autophagy can suppress the intracellular survival of mycobacteria, and several PE_PGRS family proteins of Mycobacterium tuberculosis have been proposed to act as inhibitors of autophagy to promote mycobacterial survival. However, the mechanisms by which these effectors inhibit autophagy have not been defined. Here, we report detailed studies of M. tuberculosis deletion mutants of two genes, pe_pgrs20 and pe_pgrs47, that we previously reported as having a role in preventing autophagy of infected host cells. These mutants resulted in increased autophagy and reduced intracellular survival of M. tuberculosis in macrophages. This phenotype was accompanied by increased cytokine production and antigen presentation by infected cells. We further demonstrated that autophagy inhibition by PE_PGRS20 and PE_PGRS47 resulted from canonical autophagy rather than autophagy flux inhibition. Using macrophages transfected to express PE_PGRS20 or PE_PGRS47, we showed that these proteins inhibited autophagy initiation directly by interacting with Ras-related protein Rab1A. Silencing of Rab1A in mammalian cells rescued the survival defects of the pe_pgrs20 and pe_pgrs47 deletion mutant strains and reduced cytokine secretion. To our knowledge, this is the first study to identify mycobacterial effectors that directly interact with host proteins responsible for autophagy initiation. IMPORTANCE Tuberculosis is a significant global infectious disease caused by infection of the lungs with Mycobacterium tuberculosis, which then resides and replicates mainly within host phagocytic cells. Autophagy is a complex host cellular process that helps control intracellular infections and enhance innate and adaptive immune responses. During coevolution with humans, M. tuberculosis has acquired various mechanisms to inhibit host cellular processes, including autophagy. We identified two related M. tuberculosis proteins, PE_PGRS20 and PE_PGRS47, as the first reported examples of specific mycobacterial effectors interfering with the initiation stage of autophagy. Autophagy regulation by these PE_PGRS proteins leads to increased bacterial survival in phagocytic cells and increased autophagic degradation of mycobacterial antigens to stimulate adaptive immune responses. A better understanding of how M. tuberculosis regulates autophagy in host cells could facilitate the design of new and more effective therapeutics or vaccines against tuberculosis.
Collapse
|
12
|
Radanova M, Mihaylova G, Nazifova-Tasinova N, Levkova M, Tasinov O, Ivanova D, Mihaylova Z, Donev I. Oncogenic Functions and Clinical Significance of Circular RNAs in Colorectal Cancer. Cancers (Basel) 2021; 13:3395. [PMID: 34298612 PMCID: PMC8303601 DOI: 10.3390/cancers13143395] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/13/2021] [Accepted: 07/02/2021] [Indexed: 12/11/2022] Open
Abstract
Colorectal cancer (CRC) is ranked as the second most commonly diagnosed disease in females and the third in males worldwide. Therefore, the finding of new more reliable biomarkers for early diagnosis, for prediction of metastasis, and resistance to conventional therapies is an important challenge in overcoming the disease. The current review presents circular RNAs (circRNAs) with their unique features as potential prognostic and diagnostic biomarkers in CRC. The review highlights the mechanism of action and the role of circRNAs with oncogenic functions in the CRC as well as the association between their expression and clinicopathological characteristics of CRC patients. The comprehension of the role of oncogenic circRNAs in CRC pathogenesis is growing rapidly and the next step is using them as suitable new drug targets in the personalized treatment of CRC patients.
Collapse
Affiliation(s)
- Maria Radanova
- Department of Biochemistry, Molecular Medicine and Nutrigenomics, Medical University of Varna, 9000 Varna, Bulgaria; (M.R.); (G.M.); (N.N.-T.); (O.T.); (D.I.)
- Laboratory of Molecular Pathology, University Hospital “St. Marina”, 9000 Varna, Bulgaria
| | - Galya Mihaylova
- Department of Biochemistry, Molecular Medicine and Nutrigenomics, Medical University of Varna, 9000 Varna, Bulgaria; (M.R.); (G.M.); (N.N.-T.); (O.T.); (D.I.)
| | - Neshe Nazifova-Tasinova
- Department of Biochemistry, Molecular Medicine and Nutrigenomics, Medical University of Varna, 9000 Varna, Bulgaria; (M.R.); (G.M.); (N.N.-T.); (O.T.); (D.I.)
| | - Mariya Levkova
- Department of Medical Genetics, Molecular Medicine and Nutrigenomics, Medical University of Varna, 9000 Varna, Bulgaria;
| | - Oskan Tasinov
- Department of Biochemistry, Molecular Medicine and Nutrigenomics, Medical University of Varna, 9000 Varna, Bulgaria; (M.R.); (G.M.); (N.N.-T.); (O.T.); (D.I.)
| | - Desislava Ivanova
- Department of Biochemistry, Molecular Medicine and Nutrigenomics, Medical University of Varna, 9000 Varna, Bulgaria; (M.R.); (G.M.); (N.N.-T.); (O.T.); (D.I.)
| | - Zhasmina Mihaylova
- Clinic of Medical Oncology, Military Medical Academy, 1000 Sofia, Bulgaria;
| | - Ivan Donev
- Clinic of Medical Oncology, Hospital Nadezhda, 1000 Sofia, Bulgaria
| |
Collapse
|
13
|
Jin H, Tang Y, Yang L, Peng X, Li B, Fan Q, Wei S, Yang S, Li X, Wu B, Huang M, Tang S, Liu J, Li H. Rab GTPases: Central Coordinators of Membrane Trafficking in Cancer. Front Cell Dev Biol 2021; 9:648384. [PMID: 34141705 PMCID: PMC8204108 DOI: 10.3389/fcell.2021.648384] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 05/10/2021] [Indexed: 12/11/2022] Open
Abstract
Tumor progression involves invasion, migration, metabolism, autophagy, exosome secretion, and drug resistance. Cargos transported by membrane vesicle trafficking underlie all of these processes. Rab GTPases, which, through coordinated and dynamic intracellular membrane trafficking alongside cytoskeletal pathways, determine the maintenance of homeostasis and a series of cellular functions. The mechanism of vesicle movement regulated by Rab GTPases plays essential roles in cancers. Therefore, targeting Rab GTPases to adjust membrane trafficking has the potential to become a novel way to adjust cancer treatment. In this review, we describe the characteristics of Rab GTPases; in particular, we discuss the role of their activation in the regulation of membrane transport and provide examples of Rab GTPases regulating membrane transport in tumor progression. Finally, we discuss the clinical implications and the potential as a cancer therapeutic target of Rab GTPases.
Collapse
Affiliation(s)
- Hongyuan Jin
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Yuanxin Tang
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Liang Yang
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Xueqiang Peng
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Bowen Li
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Qin Fan
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Shibo Wei
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Shuo Yang
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Xinyu Li
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Bo Wu
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Mingyao Huang
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Shilei Tang
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Jingang Liu
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Hangyu Li
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
14
|
Huang L, Luo S, Zhang X, Cai Y, Xue F, Hu H, Zeng Z, Lin T, Wang F, Wang W, Zhang S, Kang L. Distinct Genomic Landscape of Colorectal Mucinous Carcinoma Determined via Comprehensive Genomic Profiling: Steps to a New Treatment Strategy. Front Oncol 2021; 11:603564. [PMID: 34026601 PMCID: PMC8139246 DOI: 10.3389/fonc.2021.603564] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 04/08/2021] [Indexed: 12/17/2022] Open
Abstract
Colorectal mucinous carcinoma (MC) is associated with inferior prognosis and response to treatment compared to adenocarcinoma (AC). The molecular landscapes of MC and adenocarcinoma with mucous composition (AMC) are not well-defined. We aimed to describe the genomic landscape of MC and AMC in a large colorectal cancer cohort. Tumor samples from patients with MC, AMC, or AC were analyzed using next-generation sequencing. MC had a molecular signature distinct from that of AC; genomic features were similar between AMC and MC but not between AMC and AC. HER2 amplification and TP53 and APC mutation rates were lower, whereas SMAD4, PIK3CA, ACVR2A, KMT2D, LRP1, TGFBR2, GRIN2A, BRAF V600E, PTEN, and BRCA2 mutation rates were higher in MC than in AC. The mutation frequencies in MAPK, PI3K, and TGF- pathways were higher, whereas those of cell cycle proteins and Wnt were lower in MC and AMC than in AC. The proportion of hypermutated tumors was significantly higher in MC and AMC than in AC. As MC has a distinct molecular signature from AC, immunotherapy can be potentially applied in treating MC. Similar molecular profiles of AMC and MC suggest that treatment strategies for MC, but not AC, can be used for AMC treatment.
Collapse
Affiliation(s)
- Liang Huang
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.,Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Shuanglin Luo
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.,Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Xingwei Zhang
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.,Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yonghua Cai
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.,Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Fangqin Xue
- Department of Gastrointestinal Surgery, Fujian Provincial Hospital, Fuzhou, China
| | - Huanxin Hu
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.,Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Ziwei Zeng
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.,Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Tengjiao Lin
- Department of Research and Development, OrigiMed, Shanghai, China
| | - Fei Wang
- Department of Research and Development, OrigiMed, Shanghai, China
| | - Weifeng Wang
- Department of Research and Development, OrigiMed, Shanghai, China
| | - Sen Zhang
- Department of Colorectal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Liang Kang
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.,Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
15
|
Wang G, Wang X, Han M, Wang X. Loss of DAB2IP Contributes to Cell Proliferation and Cisplatin Resistance in Gastric Cancer. Onco Targets Ther 2021; 14:979-988. [PMID: 33603402 PMCID: PMC7884952 DOI: 10.2147/ott.s289722] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 12/31/2020] [Indexed: 12/24/2022] Open
Abstract
Objective Resistance to chemotherapeutic drugs, such as cisplatin, has been one of the major problems adversely affecting the clinical prognosis of patients with gastric cancer (GC). Disabled Homolog 2-Interacting Protein (DAB2IP) status is one of the major factors involved in sensitivity to chemotherapy in multiple cancer types. In the present study, we aimed to investigate the potential roles of DAB2IP in GC cell proliferation and cisplatin resistance. Materials and Methods DAB2IP expression was detected in human GC tissues using immunohistochemistry (IHC). The role of DAB2IP in regulating GC cell proliferation and cisplatin resistance was explored by genetic manipulation. Western blot analysis was used to determine the molecular signaling to explain the mechanism of the observed DAB2IP effects in GC. Results DAB2IP expression was downregulated in human GC tissues and low DAB2IP expression predicted poor prognosis. Moreover, our data provided evidence that DAB2IP upregulation impaired cell proliferation property and sensitized GC cells to cisplatin while DAB2IP depletion possessed the opposite effects. Mechanistically, we showed that DAB2IP could inhibit the phosphorylation and activation of protein kinase B (AKT) and extracellular signal-regulated kinase (ERK), and the enhanced proliferation ability induced by DAB2IP knockdown was greatly impaired after incubation with AKT or ERK inhibitor. Conclusion DAB2IP modulates GC cell proliferation and sensitivity to cisplatin potentially via regulation of AKT and ERK signaling pathway, indicating that DAB2IP may serve as a potential prognostic biomarker and therapeutic target for treatment of GC.
Collapse
Affiliation(s)
- Guannan Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241000, People's Republic of China
| | - Xu Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241000, People's Republic of China
| | - Meng Han
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241000, People's Republic of China
| | - Xiaoming Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241000, People's Republic of China
| |
Collapse
|
16
|
Xu M, Shao X, Li H, Zhang Z, Zhou C, Cheng Z. Clinical value and potential association of Rab1A and FoxM1 aberrant expression in colorectal cancer. Sci Rep 2020; 10:20160. [PMID: 33214609 PMCID: PMC7678875 DOI: 10.1038/s41598-020-77182-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 11/05/2020] [Indexed: 11/21/2022] Open
Abstract
Colorectal carcinoma (CRC) is one of the most common malignancies with a dismal 5-year survival rate. Our recent study indicated that Rab1A expression was closely related to GLI1 expression. A previous study shows that aberrant overexpression of GLI1 promotes colorectal cancer metastasis via FoxM1 overexpression. However, the potential correlation between Rab1A and FoxM1 in CRC remains elusive. Immunohistochemistry was performed to investigate the association of the expression of Rab1A and FoxM1 and to determine the prognosis in 135 CRC tissue and adjacent normal tissues. Using Oncomine datasets, we found that Rab1A and FoxM1 mRNA were obviously upregulated in CRC tissues compared to normal tissues. Additionally, the expression of Rab1A and FoxM1 was significantly higher in CRC tissues than that in normal tissues. Rab1A expression was positively correlated with FoxM1 expression in CRC, especially in TNM stage III. In addition, Rab1A and FoxM1 overexpression was found to be significantly correlated with poor prognosis in CRC patients. Besides, both high expression of Rab1A and FoxM1 led to a worse prognosis than anyone low group, and both low expression of Rab1A and FoxM1 had a better prognosis than the anyone low group. Therefore, Rab1A and FoxM1 play crucial roles and could be used as clinical biomarkers in CRC.
Collapse
Affiliation(s)
- Menglin Xu
- Department of Oncology, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241000, China
| | - Xinyu Shao
- Department of Gastroenterology, The Affiliated Suzhou Hospital of Nanjing Medical University, No.242 Guangji Road, Suzhou, 215006, Jiangsu Province, China
| | - Haoran Li
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wannan Medical College, No. 2 Zheshan West Road, Jinghu District, Wuhu, 241000, Anhui Province, China
| | - Zhengrong Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wannan Medical College, No. 2 Zheshan West Road, Jinghu District, Wuhu, 241000, Anhui Province, China
| | - Chunli Zhou
- Department of Gastroenterology, The Affiliated Suzhou Hospital of Nanjing Medical University, No.242 Guangji Road, Suzhou, 215006, Jiangsu Province, China.
| | - Zhengwu Cheng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wannan Medical College, No. 2 Zheshan West Road, Jinghu District, Wuhu, 241000, Anhui Province, China.
| |
Collapse
|
17
|
Han T, Cheng Z, Xu M, Wang X, Wu J, Fang X. Yes-Associated Protein Contributes to Cell Proliferation and Migration of Gastric Cancer via Activation of Gli1. Onco Targets Ther 2020; 13:10867-10876. [PMID: 33149604 PMCID: PMC7603417 DOI: 10.2147/ott.s266449] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Accepted: 09/04/2020] [Indexed: 12/21/2022] Open
Abstract
Objective In the present study, we aimed to explore the potential oncogenic property and the internal mechanism of yes-associated protein (YAP) in gastric cancer (GC). Materials and Methods YAP protein levels were evaluated in human GC tissues and paired normal tissues using immunohistochemistry (IHC). The role of YAP in regulating GC cell proliferation and migration was verified by genetic manipulation in vitro. Western blot analysis was used to determine the molecular signaling to explain the mechanism of the observed YAP effects in GC. Results Nuclear YAP protein expression was upregulated in GC tissues, and high nuclear YAP level was significantly correlated with lymph node metastasis (LNM) and tumor node metastasis (TNM) stage in patients suffered from GC. YAP knockdown inhibited GC cell proliferation, migration and epithelial-mesenchymal transition (EMT) progress in vitro, whereas YAP elevation did the opposite. YAP regulated glioma-associated oncogene-1 (Gli1) expression independent of smoothened homolog (SMO). YAP modulated protein kinase B (AKT)/mechanistic target of rapamycin (mTOR) signaling pathway in GC cells. Conclusion YAP enhanced GC cell proliferation and migration potentially via its regulation of Gli1 expression through the non-classical Hedgehog pathway, indicating suppression of YAP/Gli1 signaling axis may highlight a new entry point for combination therapy of GC.
Collapse
Affiliation(s)
- Ting Han
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wannan Medical College, Wuhu 241000, People's Republic of China
| | - Zhengwu Cheng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wannan Medical College, Wuhu 241000, People's Republic of China
| | - Menglin Xu
- Department of Oncology, The First Affiliated Hospital of Wannan Medical College, Wuhu 241000, People's Republic of China
| | - Xiaoming Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wannan Medical College, Wuhu 241000, People's Republic of China
| | - Jian Wu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wannan Medical College, Wuhu 241000, People's Republic of China
| | - Xiaosan Fang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wannan Medical College, Wuhu 241000, People's Republic of China
| |
Collapse
|
18
|
Kong Q, Fan Q, Ma X, Li J, Ma R. CircRNA circUGGT2 Contributes to Hepatocellular Carcinoma Development via Regulation of the miR-526b-5p/RAB1A Axis. Cancer Manag Res 2020; 12:10229-10241. [PMID: 33116877 PMCID: PMC7571581 DOI: 10.2147/cmar.s263985] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 09/25/2020] [Indexed: 12/22/2022] Open
Abstract
Background Hepatocellular carcinoma (HCC) is a common malignant tumor in the world. Circular RNA hsa_circ_0008274 (circUGGT2) is reported to be upregulated in HCC tissues. Notwithstanding, the role and regulatory mechanism of circUGGT2 in HCC are indistinct. Methods Quantitative real-time polymerase chain reaction (qRT-PCR) was implemented to examine the levels of circUGGT2, microRNA (miR)-526b-5p, and ras-related protein Rab-1A (RAB1A) mRNA in HCC tissues and cells. Cell proliferation and colony formation were assessed with 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazoliumbromide (MTT) or colony formation assays. The levels of cyclin D1, proliferating cell nuclear antigen (PCNA), and RAB1A were detected with Western blotting. Cell cycle progression, migration, and invasion were evaluated by using flow cytometry or transwell assays. The relationship between circUGGT2 or RAB1A and miR-526b-5p was verified via dual-luciferase reporter and/or RNA pull-down assays. Xenograft assay was executed to confirm the role of circUGGT2 in vivo. Results We observed that circUGGT2 and RAB1A were upregulated while miR-526b-5p was downregulated in HCC tissues and cells. CircUGGT2 silencing suppressed tumor growth in vivo and curbed proliferation, colony formation, cell cycle progression, migration, and invasion of HCC cells in vitro. Mechanically, circUGGT2 regulated RAB1A expression via competitively binding to miR-526b-5p. Also, the inhibitory influence of circUGGT2 silencing on the malignancy of HCC cells was overturned by miR-526b-5p inhibitor. Furthermore, RAB1A overexpression reversed the suppressive influence of miR-526b-5p mimic on the malignancy of HCC cells. Conclusion CircUGGT2 silencing inhibited HCC development via modulating the miR-526b-5p/RAB1A axis, providing a possible target for HCC treatment.
Collapse
Affiliation(s)
- Qingling Kong
- Office of Hospital Infection Control, People's Hospital of Rizhao, Rizhao 276826, People's Republic of China
| | - Qing Fan
- Department of Hemodialysis, People's Hospital of Rizhao, Rizhao 276826, People's Republic of China
| | - Xianbin Ma
- Department of Clinical Laboratory, People's Hospital of Rizhao, Rizhao 276826, People's Republic of China
| | - Jian Li
- Department of Interventional Radiology, People's Hospital of Rizhao, Rizhao 276826, People's Republic of China
| | - Rong Ma
- Department of General Medicine, People's Hospital of Rizhao, Rizhao 276826, People's Republic of China
| |
Collapse
|
19
|
Cheng Z, Shao X, Xu M, Zhou C, Wang J. ENO1 Acts as a Prognostic Biomarker Candidate and Promotes Tumor Growth and Migration Ability Through the Regulation of Rab1A in Colorectal Cancer. Cancer Manag Res 2019; 11:9969-9978. [PMID: 32063722 PMCID: PMC6884970 DOI: 10.2147/cmar.s226429] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 10/30/2019] [Indexed: 12/23/2022] Open
Abstract
Background Colorectal carcinoma (CRC) is one of the most common malignancies with a dismal 5‐year survival rate. The glycolytic enzyme α-enolase (ENO1) is overexpressed in multiple cancers and is involved in tumor cell proliferation and metastasis. However, its clinical significance, biological role, and underlying molecular mechanisms in CRC are still unclear. The aim of the present study was to investigate the potential role of ENO1 in the initiation and development of CRC. Patients and methods The in situ expression of ENO1 in CRC and adjacent normal tissues was examined by immunohistochemistry. The effects of ENO1 on the in vitro proliferation and migration of CRC cell lines were investigated by MTT, colony formation, and Transwell assays. Finally, the in vivo tumorigenic capacity of ENO1 was assessed in a mouse model. Results ENO1 was overexpressed in CRC tissues and significantly correlated with the clinicopathological parameters. Furthermore, Rab1A was also overexpressed in CRC tissues and was positively correlated to that of ENO1. The high expression levels of both ENO1 and Rab1A led to significantly worse prognosis of CRC patients compared to either alone. Furthermore, knockdown of ENO1 significantly inhibited CRC cells proliferation and migration in vitro and reduced xenograft growth in vivo via the concomitant downregulation of Rab1A. Conclusion The ENO1/Rab1A signaling axis is involved in CRC progression and is a potential biomarker for the treatment of CRC.
Collapse
Affiliation(s)
- Zhengwu Cheng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wannan Medical College, Wuhu 241000, People's Republic of China
| | - Xinyu Shao
- Department of Gastroenterology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou 215006, People's Republic of China
| | - Menglin Xu
- Department of Oncology, The First Affiliated Hospital of Wannan Medical College, Wuhu 241000, People's Republic of China
| | - Chunli Zhou
- Department of Gastroenterology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou 215006, People's Republic of China
| | - Junfeng Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wannan Medical College, Wuhu 241000, People's Republic of China
| |
Collapse
|
20
|
Expression analysis and implication of Rab1A in gastrointestinal relevant tumor. Sci Rep 2019; 9:13384. [PMID: 31527621 PMCID: PMC6746845 DOI: 10.1038/s41598-019-49786-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 08/31/2019] [Indexed: 12/20/2022] Open
Abstract
Gastrointestinal cancers have become increasingly prevalent worldwide. Previous studies have reported an oncogenic function of Rab1A in colorectal cancer and hepatocellular carcinomas via the mTOR pathway. However, the exact role of Rab1A in gastrointestinal cancers remains elusive. We detected significantly higher expression of Rab1A in the gastrointestinal tumor tissues compared to that in other cancer types following an in silico analysis of TGCA and GTEX databases. Furthermore, Rab1A was overexpressed in the gastrointestinal tumor tissues compared to the para-tumor tissues. Although Rab1A expression levels were not associated with the tumor-lymph node-metastasis (TNM) stage, Rab1A overexpression in the tumor tissues of a gastric cancer (GC) cohort was strongly correlated with poor prognosis in the patients. In addition, Rab1A knockdown significantly inhibited the in vitro proliferation and migration abilities of GC cells, as well as the growth of GC xenografts in vivo. Furthermore, a positive correlation was observed between Rab1A expression levels and that of different upstream/downstream mTOR targets. Taken together, Rab1A regulates the PI3K-AKT-mTORC1 pathway through the mTORC1 complex consisting of mTORC1, Rheb and Rab1A, and is a promising therapeutic target in GC.
Collapse
|