1
|
Chong ZX. Roles of miRNAs in regulating ovarian cancer stemness. Biochim Biophys Acta Rev Cancer 2024; 1879:189191. [PMID: 39353485 DOI: 10.1016/j.bbcan.2024.189191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 09/02/2024] [Accepted: 09/23/2024] [Indexed: 10/04/2024]
Abstract
Ovarian cancer is one of the gynaecology malignancies with the highest mortality rate. Ovarian cancer stem cell (CSC) is a subpopulation of ovarian cancer cells with increased self-renewability, aggression, metastatic potentials, and resistance to conventional anti-cancer therapy. The emergence of ovarian CSC is a critical factor that promotes treatment resistance and frequent relapse among ovarian cancer patients, leading to poor clinical outcomes. MicroRNA (miRNA) is a short, non-protein-coding RNA that regulates ovarian CSC development. Although multiple original research articles have discussed the CSC-regulatory roles of different miRNAs in ovarian cancer, there is a deficiency of a review article that can summarize the findings from different research papers. To narrow the gap in the literature, this review aimed to provide an up-to-date summary of the CSC-regulatory roles of various miRNAs in modulating ovarian cancer cell stemness. This review will begin by giving an overview of ovarian CSC and the pathways responsible for driving its appearance. Next, the CSC-regulatory roles of miRNAs in controlling ovarian CSC development will be discussed. Overall, more than 60 miRNAs have been reported to play CSC-regulatory roles in the development and progression of ovarian cancer. By targeting various downstream targets, these miRNAs can control the signaling activities of PI3K/AKT, EGFR/ERK, WNT/ß-catenin, NF-kß, Notch, Hippo/YAP, EMT, and DNA repair pathways. Hence, these CSC-modulatory miRNAs have the potential to be used as prognostic biomarkers in predicting the clinical outcomes of ovarian cancer patients. Targeting CSC-promoting miRNAs or increasing the expressions of CSC-repressing miRNAs can help slow ovarian cancer progression. However, more in-depth functional and clinical trials must be carried out to evaluate the suitability, safety, sensitivity, and specificity of these CSC-regulating miRNAs as prognostic biomarkers or therapeutic targets.
Collapse
Affiliation(s)
- Zhi-Xiong Chong
- Faculty of Science and Engineering, University of Nottingham Malaysia, Jalan Broga, 43500 Semenyih, Selangor, Malaysia; NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Centre for Translational Medicine, 14 Medical Drive, #12-01, Singapore 117599; Cancer Science Institute of Singapore, National University of Singapore, Centre for Translational Medicine, 14 Medical Drive, #12-01, Singapore 117599.
| |
Collapse
|
2
|
Meng L, Zhang C, Yu P. Treating cancer through modulating exosomal protein loading and function: The prospects of natural products and traditional Chinese medicine. Pharmacol Res 2024; 203:107179. [PMID: 38615876 DOI: 10.1016/j.phrs.2024.107179] [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: 01/02/2024] [Revised: 03/22/2024] [Accepted: 04/05/2024] [Indexed: 04/16/2024]
Abstract
Exosomes, small yet vital extracellular vesicles, play an integral role in intercellular communication. They transport critical components, such as proteins, lipid bilayers, DNA, RNA, and glycans, to target cells. These vesicles are crucial in modulating the extracellular matrix and orchestrating signal transduction processes. In oncology, exosomes are pivotal in tumor growth, metastasis, drug resistance, and immune modulation within the tumor microenvironment. Exosomal proteins, noted for their stability and specificity, have garnered widespread attention. This review delves into the mechanisms of exosomal protein loading and their impact on tumor development, with a focus on the regulatory effects of natural products and traditional Chinese medicine on exosomal protein loading and function. These insights not only offer new strategies and methodologies for cancer treatment but also provide scientific bases and directions for future clinical applications.
Collapse
Affiliation(s)
- Lulu Meng
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Chao Zhang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China.
| | - Pei Yu
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
3
|
Shen Y, Gao Y, Fu J, Wang C, Tang Y, Chen S, Zhao Y. Lack of Rab27a attenuates foam cell formation and macrophage inflammation in uremic apolipoprotein E knockout mice. J Mol Histol 2023:10.1007/s10735-023-10125-w. [PMID: 37166546 DOI: 10.1007/s10735-023-10125-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Accepted: 05/02/2023] [Indexed: 05/12/2023]
Abstract
As the most common cardiovascular disease, atherosclerosis (AS), is a leading cause of high mortality in patients with chronic renal failure. Rab27a has been reported to regulate the progression of cardiovascular and renal diseases. Nevertheless, little studies investigated the role and mechanism of Rab27a in uremic-accelerated AS (UAAS). An animal model of UAAS was established in apolipoprotein E knockout (apoE-/-) mice using 5/6 nephrectomy (NX). We conducted in vitro and in vivo functional experiments to explore the role of Rab27a in UAAS, including the presence of oxidized low-density lipoprotein (ox-LDL). Rab27a expression was upregulated in the plaque tissues of NX apoE-/- mice. The knockout of Rab27a (Rab27a-/-) reduced AS-induced artery injury, as manifested by the reductions of plaque area, collagen deposition, inflammation and lipid droplet. Besides, cholesterol efflux was increased, while the expression of lipid metabolism-related proteins and the secretions of pro-inflammatory factors were decreased in ox-LDL-induced NX Rab27a-/- apoE-/- mice group. Further, Rab27a deletion inhibited the activation of nuclear factor κB (NF-κB) pathway. In conclusion, our study indicated that Rab27a deficiency attenuated foam cell formation and macrophage inflammation, depending on the NF-κB pathway activation, to inhibit AS progression in uremic apoE-/- mice. This finding may provide a new targeting strategy for UAAS therapy.
Collapse
Affiliation(s)
- Yan Shen
- Department of Nephrology, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Yanta District, Shaanxi, 710061, Xi'an, China.
| | - Yajuan Gao
- Department of Nephrology, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Yanta District, Shaanxi, 710061, Xi'an, China
| | - Jiani Fu
- Department of Nephrology, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Yanta District, Shaanxi, 710061, Xi'an, China
| | - Cui Wang
- Department of Nephrology, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Yanta District, Shaanxi, 710061, Xi'an, China
| | - Yali Tang
- Department of Nephrology, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Yanta District, Shaanxi, 710061, Xi'an, China
| | - Shengnan Chen
- Department of Nephrology, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Yanta District, Shaanxi, 710061, Xi'an, China
| | - Yan Zhao
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| |
Collapse
|
4
|
Ren J, Feng X, Guo Y, Kong D, Wang Y, Xiao J, Jiang W, Feng X, Liu X, Li A, Sun C, He M, Li B, Wang J, Jiang Y, Zheng C. GSK-3β/β-catenin pathway plays crucial roles in the regulation of NK cell cytotoxicity against myeloma cells. FASEB J 2023; 37:e22821. [PMID: 36794671 DOI: 10.1096/fj.202201658rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 01/28/2023] [Accepted: 01/31/2023] [Indexed: 02/17/2023]
Abstract
The plasma cell malignancy, multiple myeloma (MM), has significantly improved by the application of new drugs and autologous hematopoietic stem cell transplantation. However, MM remains incurable. A number of studies have revealed an anti-MM effect of natural killer (NK) cells; however, their clinical efficacy is limited. Furthermore, glycogen synthase kinase (GSK)-3β inhibitors show an antitumor function. In this study, we aimed to evaluate the potential roles of a GSK-3β inhibitor (TWS119) in the regulation of NK cell cytotoxicity against MM. Our results showed that, in the presence of TWS119, the NK cell line, NK-92, and in vitro-expanded primary NK cells exhibited a significantly higher degranulation activity, expression of activating receptors, cellular cytotoxicity, and cytokine secretion when they were exposed to MM cells. Mechanistic studies indicated that TWS119 treatment markedly upregulated RAB27A expression, a key molecule for NK cell degranulation, and induced the colocalization of β-catenin with NF-κB in the nucleus of NK cells. More importantly, GSK-3β inhibition combined with the adoptive transfer of TWS119-treated NK-92 cells significantly reduced tumor volume and prolonged the survival time of myeloma-bearing mice. In summary, our novel findings suggest that targeting GSK-3β through the activation of β-catenin/NF-κB pathway may be an important approach to improve therapeutic efficacy of NK cell transfusion for MM.
Collapse
Affiliation(s)
- Jing Ren
- Department of Hematology, The Second Hospital of Shandong University, Jinan, Shandong, China.,Institute of Biotherapy for Hematological Malignancy, Shandong University, Jinan, Shandong, China
| | - Xiumei Feng
- Department of Hematology, The Fourth People's Hospital of Jinan City, Jinan, Shandong, China
| | - Yanan Guo
- Department of Hematology, The Second Hospital of Shandong University, Jinan, Shandong, China.,Institute of Biotherapy for Hematological Malignancy, Shandong University, Jinan, Shandong, China
| | - Dexiao Kong
- Department of Hematology, The Second Hospital of Shandong University, Jinan, Shandong, China.,Institute of Biotherapy for Hematological Malignancy, Shandong University, Jinan, Shandong, China
| | - Yongjing Wang
- Department of Hematology, The Second Hospital of Shandong University, Jinan, Shandong, China.,Institute of Biotherapy for Hematological Malignancy, Shandong University, Jinan, Shandong, China
| | - Juan Xiao
- Department of Hematology, The Second Hospital of Shandong University, Jinan, Shandong, China.,Institute of Biotherapy for Hematological Malignancy, Shandong University, Jinan, Shandong, China
| | - Wen Jiang
- Institute of Medical Sciences, The Second Hospital of Shandong University, Jinan, Shandong, China
| | - Xiaoli Feng
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong, China
| | - Xiaoli Liu
- Department of Hematology, The Second Hospital of Shandong University, Jinan, Shandong, China.,Institute of Biotherapy for Hematological Malignancy, Shandong University, Jinan, Shandong, China
| | - Ai Li
- Department of Hematology, The Second Hospital of Shandong University, Jinan, Shandong, China.,Institute of Biotherapy for Hematological Malignancy, Shandong University, Jinan, Shandong, China
| | - Congcong Sun
- Department of Hematology, The Second Hospital of Shandong University, Jinan, Shandong, China.,Institute of Biotherapy for Hematological Malignancy, Shandong University, Jinan, Shandong, China
| | - Mingming He
- Department of Hematology, The Second Hospital of Shandong University, Jinan, Shandong, China.,Institute of Biotherapy for Hematological Malignancy, Shandong University, Jinan, Shandong, China
| | - Bingen Li
- R&D Department, Weihai Zhengsheng Biotechnology Co., Ltd, Weihai, China
| | - Juandong Wang
- Department of Hematology, The Second Hospital of Shandong University, Jinan, Shandong, China.,Institute of Biotherapy for Hematological Malignancy, Shandong University, Jinan, Shandong, China
| | - Yang Jiang
- Department of Hematology, The Second Hospital of Shandong University, Jinan, Shandong, China.,Institute of Biotherapy for Hematological Malignancy, Shandong University, Jinan, Shandong, China
| | - Chengyun Zheng
- Department of Hematology, The Second Hospital of Shandong University, Jinan, Shandong, China.,Institute of Biotherapy for Hematological Malignancy, Shandong University, Jinan, Shandong, China
| |
Collapse
|
5
|
Li Q, Zhao H, Dong W, Guan N, Hu Y, Zeng Z, Zhang H, Zhang F, Li Q, Yang J, Xiao W. RAB27A promotes the proliferation and invasion of colorectal cancer cells. Sci Rep 2022; 12:19359. [PMID: 36371494 PMCID: PMC9653419 DOI: 10.1038/s41598-022-23696-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 11/03/2022] [Indexed: 11/13/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most commonly diagnosed cancer types worldwide. Despite significant advances in prevention and diagnosis, CRC is still one of the leading causes of cancer-related mortality globally. RAB27A, the member of RAB27 family of small GTPases, is the critical protein for intracellular secretion and has been reported to promote tumor progression. However, it is controversial for the role of RAB27A in CRC progression, so we explored the exact function of RAB27A in CRC development in this study. Based on the stable colon cancer cell lines of RAB27A knockdown and ectopic expression, we found that RAB27A knockdown inhibited proliferation and clone formation of SW480 colon cancer cells, whereas ectopic expression of RAB27A in RKO colon cancer cells facilitated cell proliferation and clone formation, indicating that RAB27A is critical for colon cancer cell growth. In addition, our data demonstrated that the migration and invasion of colon cancer cells were suppressed by RAB27A knockdown, but promoted by RAB27A ectopic expression. Therefore, RAB27A is identified as an onco-protein in mediating CRC development, which may be a valuable prognostic indicator and potential therapeutic target for CRC.
Collapse
Affiliation(s)
- Qingyan Li
- grid.454145.50000 0000 9860 0426Graduate School of Jinzhou Medical University, Liaoning, 121001 China ,grid.414252.40000 0004 1761 8894Senior Department of Oncology, the Fifth Medical Center of PLA General Hospital, Beijing, 100071 China ,Department of Oncology, Suining Central Hospital, Sichuan, 629300 China
| | - Huixia Zhao
- grid.414252.40000 0004 1761 8894Department of Oncology, 4th Medical Center of PLA General Hospital, Beijing, 100048 China
| | - Weiwei Dong
- grid.414252.40000 0004 1761 8894Senior Department of Oncology, the Fifth Medical Center of PLA General Hospital, Beijing, 100071 China
| | - Na Guan
- grid.454145.50000 0000 9860 0426Graduate School of Jinzhou Medical University, Liaoning, 121001 China ,grid.414252.40000 0004 1761 8894Senior Department of Oncology, the Fifth Medical Center of PLA General Hospital, Beijing, 100071 China
| | - Yanyan Hu
- grid.414252.40000 0004 1761 8894Senior Department of Oncology, the Fifth Medical Center of PLA General Hospital, Beijing, 100071 China
| | - Zhiyan Zeng
- grid.414252.40000 0004 1761 8894Department of Oncology, 4th Medical Center of PLA General Hospital, Beijing, 100048 China
| | - He Zhang
- grid.414252.40000 0004 1761 8894Department of Oncology, 4th Medical Center of PLA General Hospital, Beijing, 100048 China
| | - Fengyun Zhang
- grid.414252.40000 0004 1761 8894Department of Oncology, 4th Medical Center of PLA General Hospital, Beijing, 100048 China
| | - Qiuwen Li
- grid.414252.40000 0004 1761 8894Senior Department of Oncology, the Fifth Medical Center of PLA General Hospital, Beijing, 100071 China
| | - Jingwen Yang
- grid.414252.40000 0004 1761 8894Senior Department of Oncology, the Fifth Medical Center of PLA General Hospital, Beijing, 100071 China
| | - Wenhua Xiao
- grid.454145.50000 0000 9860 0426Graduate School of Jinzhou Medical University, Liaoning, 121001 China ,grid.414252.40000 0004 1761 8894Senior Department of Oncology, the Fifth Medical Center of PLA General Hospital, Beijing, 100071 China
| |
Collapse
|
6
|
Zhang Z, Zhou Y, Jia Y, Wang C, Zhang M, Xu Z. PRR34-AS1 promotes exosome secretion of VEGF and TGF-β via recruiting DDX3X to stabilize Rab27a mRNA in hepatocellular carcinoma. J Transl Med 2022; 20:491. [PMID: 36303180 PMCID: PMC9615160 DOI: 10.1186/s12967-022-03628-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 09/07/2022] [Indexed: 12/24/2022] Open
Abstract
Background Exosomes are deemed to be an important tool of intercellular communicators in cancer cells. Our study investigated the role of PRR34 long non-coding RNA antisense RNA 1 (PRR34-AS1) in regulating exosome secretion in hepatocellular carcinoma (HCC) cells. Methods Quantitative real-time polymerase chain reaction (RT-qPCR) analyzed the expression of PRR34-AS1. We assessed the function of PRR34-AS1 on the biological changes of THLE-3 cells and HCC cells. The downstream interaction between RNAS was assessed by mechanistic experiments. Results PRR34-AS1 expression was upregulated in HCC cells in comparison to THLE-3 cells. PRR34-AS1 depletion repressed HCC cell proliferation, migration and invasion as well as EMT phenotype, while PRR34-AS1 up-regulation accelerated the malignant phenotypes of THLE-3 cells. PRR34-AS1 recruited DDX3X to stabilize the mRNA level of exosomal protein Rab27a. Moreover, PRR34-AS1 facilitated the malignant phenotypes of THLE-3 cells by elevating Rab27a expression to promote the exosome secretion of VEGF and TGF-β in HCC cells. Conclusions The current study revealed a novel function of PRR34-AS1 in accelerating exosome secretion in HCC cells and offered an insight into lncRNA function in the regulation of tumor cell biology. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12967-022-03628-9.
Collapse
Affiliation(s)
- Zhilei Zhang
- Department of Hepatobiliary Surgery, Fourth Hospital of Hebei Medical University, No. 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China.
| | - Ye Zhou
- Department of Hepatobiliary Surgery, Fourth Hospital of Hebei Medical University, No. 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China
| | - Yuming Jia
- Department of Hepatobiliary Surgery, Fourth Hospital of Hebei Medical University, No. 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China
| | - Chao Wang
- Department of Hepatobiliary Surgery, Fourth Hospital of Hebei Medical University, No. 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China
| | - Meng Zhang
- Department of Hepatobiliary Surgery, Fourth Hospital of Hebei Medical University, No. 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China
| | - Zhuo Xu
- Department of Hepatobiliary Surgery, Fourth Hospital of Hebei Medical University, No. 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China
| |
Collapse
|
7
|
Di Vito Nolfi M, Vecchiotti D, Flati I, Verzella D, Di Padova M, Alesse E, Capece D, Zazzeroni F. EV-Mediated Chemoresistance in the Tumor Microenvironment: Is NF-κB a Player? Front Oncol 2022; 12:933922. [PMID: 35814425 PMCID: PMC9257640 DOI: 10.3389/fonc.2022.933922] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 05/23/2022] [Indexed: 11/17/2022] Open
Abstract
Drug resistance is a major impediment to patient survival and remains the primary cause of unsuccessful cancer therapy. Drug resistance occurs in many tumors and is frequently induced by chemotherapy which triggers a defensive response both in cancerous and cancer-associated cells that constitute the tumor microenvironment (TME). Cell to cell communication within the TME is often mediated by extracellular vesicles (EVs) which carry specific tumor-promoting factors able to activate survival pathways and immune escape mechanisms, thus sustaining tumor progression and therapy resistance. NF-κB has been recognized as a crucial player in this context. NF-κB activation is involved in EVs release and EVs, in turn, can trigger NF-κB pathway activation in specific contexts, based on secreting cytotype and their specific delivered cargo. In this review, we discuss the role of NF-κB/EVs interplay that sustain chemoresistance in the TME by focusing on the molecular mechanisms that underlie inflammation, EVs release, and acquired drug resistance.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Daria Capece
- *Correspondence: Francesca Zazzeroni, ; Daria Capece,
| | | |
Collapse
|
8
|
Bourhim T, Villareal MO, Gadhi C, Isoda H. Elucidation of Melanogenesis-Associated Signaling Pathways Regulated by Argan Press Cake in B16 Melanoma Cells. Nutrients 2021; 13:nu13082697. [PMID: 34444857 PMCID: PMC8398289 DOI: 10.3390/nu13082697] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 07/30/2021] [Accepted: 08/02/2021] [Indexed: 12/30/2022] Open
Abstract
The beneficial effect on health of argan oil is recognized worldwide. We have previously reported that the cake that remains after argan oil extraction (argan press-cake or APC) inhibits melanogenesis in B16 melanoma cells in a time-dependent manner without cytotoxicity. In this study, the global gene expression profile of B16 melanoma cells treated with APC extract was determined in order to gain an understanding of the possible mechanisms of action of APC. The results suggest that APC extract inhibits melanin biosynthesis by down-regulating microphthalmia-associated transcription factor (Mitf) and its downstream signaling pathway through JNK signaling activation, and the inhibition of Wnt/β-catenin and cAMP/PKA signaling pathways. APC extract also prevented the transport of melanosomes by down-regulating Rab27a expression. These results suggest that APC may be an important natural skin whitening product and pharmacological agent used for clinical treatment of pigmentary disorders.
Collapse
Affiliation(s)
- Thouria Bourhim
- Faculty of Sciences Semlalia, Cadi Ayyad University, Avenue Prince Moulay Abdellah, B.P. 2390, Marrakesh 40000, Morocco;
| | - Myra O. Villareal
- Alliance for Research on the Mediterranean and North Africa (ARENA), University of Tsukuba, Tennodai 1-1-1, Tsukuba 305-8572, Japan;
- Faculty of Life and Environmental Sciences, University of Tsukuba, Tennodai 1-1-1, Tsukuba 305-8572, Japan
| | - Chemseddoha Gadhi
- Faculty of Sciences Semlalia, Cadi Ayyad University, Avenue Prince Moulay Abdellah, B.P. 2390, Marrakesh 40000, Morocco;
- Alliance for Research on the Mediterranean and North Africa (ARENA), University of Tsukuba, Tennodai 1-1-1, Tsukuba 305-8572, Japan;
- Correspondence: (C.G.); (H.I.)
| | - Hiroko Isoda
- Alliance for Research on the Mediterranean and North Africa (ARENA), University of Tsukuba, Tennodai 1-1-1, Tsukuba 305-8572, Japan;
- Faculty of Life and Environmental Sciences, University of Tsukuba, Tennodai 1-1-1, Tsukuba 305-8572, Japan
- Correspondence: (C.G.); (H.I.)
| |
Collapse
|
9
|
Chang YC, Chan MH, Li CH, Fang CY, Hsiao M, Chen CL. Exosomal Components and Modulators in Colorectal Cancer: Novel Diagnosis and Prognosis Biomarkers. Biomedicines 2021; 9:biomedicines9080931. [PMID: 34440135 PMCID: PMC8391321 DOI: 10.3390/biomedicines9080931] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 07/23/2021] [Accepted: 07/29/2021] [Indexed: 02/07/2023] Open
Abstract
The relatively high incidence and mortality rates for colorectal carcinoma (CRC) make it a formidable malignant tumor. Comprehensive strategies have been applied to predict patient survival and diagnosis. Various clinical regimens have also been developed to improve the therapeutic outcome. Extracellular vesicles (EVs) are recently proposed cellular structures that can be produced by natural or artificial methods and have been extensively studied. In addition to their innate functions, EVs can be manipulated to be drug carriers and exert many biological functions. The composition of EVs, their intravesicular components, and the surrounding tumor microenvironment are closely related to the development of colorectal cancer. Determining the expression profiles of exocytosis samples and using them as indicators for selecting effective combination therapy is an indispensable direction for EV study and should be regarded as a novel prediction platform in addition to cancer stage, prognosis, and other clinical assessments. In this review, we summarize the function, regulation, and application of EVs in the colon cancer research field. We provide an update on and discuss potential values for clinical applications of EVs. Moreover, we illustrate the specific markers, mediators, and genetic alterations of EVs in colorectal carcinogenesis. Furthermore, we outline the vital markers present in the EVs and discuss their plausible uses in colon cancer patient therapy in combination with the currently used clinical strategies. The development and application of these EVs will significantly improve the accuracy of diagnosis, lead to more precise prognoses, and may lead to the improved treatment of colorectal cancer.
Collapse
Affiliation(s)
- Yu-Chan Chang
- Department of Biomedical Imaging and Radiological Science, National Yang-Ming University, Taipei 112, Taiwan;
- Department of Biomedical Imaging and Radiological Science, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Ming-Hsien Chan
- Genomics Research Center, Academia Sinica, Taipei 115, Taiwan; (M.-H.C.); (C.-H.L.)
| | - Chien-Hsiu Li
- Genomics Research Center, Academia Sinica, Taipei 115, Taiwan; (M.-H.C.); (C.-H.L.)
| | - Chih-Yeu Fang
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 350, Taiwan;
| | - Michael Hsiao
- Genomics Research Center, Academia Sinica, Taipei 115, Taiwan; (M.-H.C.); (C.-H.L.)
- Department of Biochemistry, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Correspondence: (M.H.); (C.-L.C.); Tel.: +886-2-2787-1243 (M.H.); +886-2-2736-1661 (ext. 3139) (C.-L.C.); Fax: +886-2-2789-9931 (M.H.)
| | - Chi-Long Chen
- Department of Pathology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- Department of Pathology, Taipei Medical University Hospital, Taipei 110, Taiwan
- Correspondence: (M.H.); (C.-L.C.); Tel.: +886-2-2787-1243 (M.H.); +886-2-2736-1661 (ext. 3139) (C.-L.C.); Fax: +886-2-2789-9931 (M.H.)
| |
Collapse
|
10
|
Brunel A, Bégaud G, Auger C, Durand S, Battu S, Bessette B, Verdier M. Autophagy and Extracellular Vesicles, Connected to rabGTPase Family, Support Aggressiveness in Cancer Stem Cells. Cells 2021; 10:1330. [PMID: 34072080 PMCID: PMC8227744 DOI: 10.3390/cells10061330] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/20/2021] [Accepted: 05/24/2021] [Indexed: 12/22/2022] Open
Abstract
Even though cancers have been widely studied and real advances in therapeutic care have been made in the last few decades, relapses are still frequently observed, often due to therapeutic resistance. Cancer Stem Cells (CSCs) are, in part, responsible for this resistance. They are able to survive harsh conditions such as hypoxia or nutrient deprivation. Autophagy and Extracellular Vesicles (EVs) secretion are cellular processes that help CSC survival. Autophagy is a recycling process and EVs secretion is essential for cell-to-cell communication. Their roles in stemness maintenance have been well described. A common pathway involved in these processes is vesicular trafficking, and subsequently, regulation by Rab GTPases. In this review, we analyze the role played by Rab GTPases in stemness status, either directly or through their regulation of autophagy and EVs secretion.
Collapse
|
11
|
Nakao Y, Fukuda T, Zhang Q, Sanui T, Shinjo T, Kou X, Chen C, Liu D, Watanabe Y, Hayashi C, Yamato H, Yotsumoto K, Tanaka U, Taketomi T, Uchiumi T, Le AD, Shi S, Nishimura F. Exosomes from TNF-α-treated human gingiva-derived MSCs enhance M2 macrophage polarization and inhibit periodontal bone loss. Acta Biomater 2021; 122:306-324. [PMID: 33359765 PMCID: PMC7897289 DOI: 10.1016/j.actbio.2020.12.046] [Citation(s) in RCA: 226] [Impact Index Per Article: 75.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 11/25/2020] [Accepted: 12/18/2020] [Indexed: 02/07/2023]
Abstract
Mesenchymal stem cell (MSC)–derived exosome plays a central role in the cell-free therapeutics involving MSCs and the contents can be customized under disease-associated microenvironments. However, optimal MSC-preconditioning to enhance its therapeutic potential is largely unknown. Here, we show that preconditioning of gingival tissue-derived MSCs (GMSCs) with tumor necrosis factor-alpha (TNF-α) is ideal for the treatment of periodontitis. TNF-α stimulation not only increased the amount of exosome secreted from GMSCs, but also enhanced the exosomal expression of CD73, thereby inducing anti-inflammatory M2 macrophage polarization. The effect of GMSC-derived exosomes on inflammatory bone loss were examined by ligature-induced periodontitis model in mice. Local injection of GMSC-derived exosomes significantly reduced periodontal bone resorption and the number of tartrate-resistant acid phosphatase (TRAP)-positive osteoclasts, and these effects were further enhanced by preconditioning of GMSCs with TNF-α. Thus, GMSC-derived exosomes also exhibited anti-osteoclastogenic activity. Receptor activator of NF-κB ligand (RANKL) expression was regulated by Wnt5a in periodontal ligament cells (PDLCs), and exosomal miR-1260b was found to target Wnt5a-mediated RANKL pathway and inhibit its osteoclastogenic activity. These results indicate that significant ability of the TNF-α-preconditioned GMSC-derived exosomes to regulate inflammation and osteoclastogenesis paves the way for establishment of a therapeutic approach for periodontitis.
Collapse
Affiliation(s)
- Yuki Nakao
- Department of Periodontology, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Takao Fukuda
- Department of Periodontology, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka, Japan; Department of Anatomy and Cell Biology, University of Pennsylvania School of Dental Medicine, Philadelphia, PA, USA
| | - Qunzhou Zhang
- Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, PA, USA
| | - Terukazu Sanui
- Department of Periodontology, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Takanori Shinjo
- Department of Periodontology, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Xiaoxing Kou
- Department of Anatomy and Cell Biology, University of Pennsylvania School of Dental Medicine, Philadelphia, PA, USA; South China Center of Craniofacial Stem Cell Research, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong, China
| | - Chider Chen
- Department of Anatomy and Cell Biology, University of Pennsylvania School of Dental Medicine, Philadelphia, PA, USA; Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, PA, USA
| | - Dawei Liu
- Department of Anatomy and Cell Biology, University of Pennsylvania School of Dental Medicine, Philadelphia, PA, USA; Department of Orthodontics, Peking University School and Stomatology, Peking, China
| | - Yukari Watanabe
- Department of Periodontology, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Chikako Hayashi
- Department of Periodontology, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Hiroaki Yamato
- Department of Periodontology, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Karen Yotsumoto
- Department of Periodontology, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Urara Tanaka
- Department of Periodontology, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Takaharu Taketomi
- Dental and Oral Medical Center, Kurume University School of Medicine, Fukuoka, Japan
| | - Takeshi Uchiumi
- Department of Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Anh D Le
- Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, PA, USA
| | - Songtao Shi
- Department of Anatomy and Cell Biology, University of Pennsylvania School of Dental Medicine, Philadelphia, PA, USA; South China Center of Craniofacial Stem Cell Research, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong, China
| | - Fusanori Nishimura
- Department of Periodontology, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka, Japan.
| |
Collapse
|
12
|
Zhu X, Tian G, Quan J, He P, Liu J. Effects of miR‑340 overexpression and knockdown on the proliferation and metastasis of NSCLC cell lines. Int J Mol Med 2019; 44:643-651. [PMID: 31173161 PMCID: PMC6605470 DOI: 10.3892/ijmm.2019.4213] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Accepted: 05/23/2019] [Indexed: 12/19/2022] Open
Abstract
The present study aimed to investigate the potential biological functions of microRNA‑340 (miR‑340) in non‑small cell lung cancer (NSCLC) beyond its role as a critical regulator of tumorigenesis and tumor progression. The expression levels of miR‑340 and RAB27B were analyzed by reverse transcription‑quantitative polymerase chain reaction. Subsequently, the protein expression levels of RAB27A, RAB27B, RAB9A, RAB11A and BRAB21 were determined by western blot analysis. The expression levels of the aforementioned proteins in NSCLC tissues were analyzed by immunohistochemistry. RAB27B, as a potential target of miR‑340 was investigated via a dual‑luciferase reporter assay. The proliferative ability of PC9, A549 and BEAS‑2B cells was detected with a Cell Counting kit‑8 assay, while the migration and invasion of the NSCLC cells were analyzed using a Transwell assay. The results revealed that the expression levels of miR‑340 in the NSCLC cells were significantly decreased compared with those in normal cells (BEAS‑2B cells). RAB27B was proposed as a potential target gene of miR‑340, and its expression was notably increased in the NSCLC cells. miR‑340 overexpression inhibited the migration and invasion of the NSCLC cells by targeting RAB27B, while the knockdown of miR‑340 exerted opposite effects. On the whole, these findings indicate that the miR‑340/RAB27B axis may be actively involved in the occurrence of NSCLC. Thus, miR‑340 and RAB27B may be novel therapeutic targets for the treatment of NSCLC.
Collapse
Affiliation(s)
- Xidan Zhu
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Gang Tian
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Jing Quan
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Peng He
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Jinbo Liu
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| |
Collapse
|
13
|
Boucher JM, Robich M, Scott SS, Yang X, Ryzhova L, Turner JE, Pinz I, Liaw L. Rab27a Regulates Human Perivascular Adipose Progenitor Cell Differentiation. Cardiovasc Drugs Ther 2019; 32:519-530. [PMID: 30105417 DOI: 10.1007/s10557-018-6813-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE Perivascular adipose tissue (PVAT) surrounds blood vessels and regulates vascular tone through paracrine secretion of cytokines. During conditions promoting cardiometabolic dysfunction, such as obesity, cytokine secretion is altered towards a proinflammatory and proatherogenic profile. Despite the clinical implications for cardiovascular disease, studies addressing the biology of human PVAT remain limited. We are interested in characterizing the resident adipose progenitor cells (APCs) because of their potential role in PVAT expansion during obesity. We also focused on proteins regulating paracrine interactions, including the small GTPase Rab27a, which regulates protein trafficking and secretion. METHODS PVAT from the ascending aorta was collected from patients with severe cardiovascular disease undergoing coronary artery bypass grafting (CABG). Freshly-isolated PVAT was digested and APC expanded in culture for characterizing progenitor markers, evaluating adipogenic potential and assessing the function(s) of Rab27a. RESULTS Using flow cytometry, RT-PCR, and immunoblot, we characterized APC from human PVAT as negative for CD45 and CD31 and expressing CD73, CD105, and CD140A. These APCs differentiate into multilocular, UCP1-producing adipocytes in vitro. Rab27a was detected in interstitial cells of human PVAT in vivo and along F-actin tracks of PVAT-APC in vitro. Knockdown of Rab27a using siRNA in PVAT-APC prior to induction resulted in a marked reduction in lipid accumulation and reduced expression of adipogenic differentiation markers. CONCLUSIONS PVAT-APC from CABG donors express common adipocyte progenitor markers and differentiate into UCP1-containing adipocytes. Rab27a has an endogenous role in promoting the maturation of adipocytes from human PVAT-derived APC.
Collapse
Affiliation(s)
- Joshua M Boucher
- Center for Molecular Medicine, Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME, 04072, USA
| | - Michael Robich
- Center for Molecular Medicine, Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME, 04072, USA
- Division of Thoracic and Cardiac Surgery, Maine Medical Center, Portland, ME, 04102, USA
| | - S Spencer Scott
- Center for Molecular Medicine, Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME, 04072, USA
| | - Xuehui Yang
- Center for Molecular Medicine, Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME, 04072, USA
| | - Larisa Ryzhova
- Center for Molecular Medicine, Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME, 04072, USA
| | - Jacqueline E Turner
- Center for Molecular Medicine, Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME, 04072, USA
| | - Ilka Pinz
- Center for Molecular Medicine, Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME, 04072, USA
| | - Lucy Liaw
- Center for Molecular Medicine, Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME, 04072, USA.
| |
Collapse
|
14
|
Koike S, Yamasaki K, Yamauchi T, Shimada-Omori R, Tsuchiyama K, Aiba S. TRIF and MAVS signaling pathways regulate RAB27A induction and melanosome transfer by TLR3 signaling in human epidermal melanocytes. J Dermatol Sci 2019; 94:306-309. [PMID: 31036445 DOI: 10.1016/j.jdermsci.2019.04.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 04/05/2019] [Accepted: 04/15/2019] [Indexed: 02/02/2023]
Affiliation(s)
- Saaya Koike
- Department of Dermatology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8574, Japan
| | - Kenshi Yamasaki
- Department of Dermatology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8574, Japan.
| | - Takeshi Yamauchi
- Department of Dermatology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8574, Japan
| | - Ryoko Shimada-Omori
- Department of Dermatology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8574, Japan
| | - Kenichiro Tsuchiyama
- Department of Dermatology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8574, Japan
| | - Setsuya Aiba
- Department of Dermatology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8574, Japan
| |
Collapse
|
15
|
Guo D, Lui GYL, Lai SL, Wilmott JS, Tikoo S, Jackett LA, Quek C, Brown DL, Sharp DM, Kwan RYQ, Chacon D, Wong JH, Beck D, van Geldermalsen M, Holst J, Thompson JF, Mann GJ, Scolyer RA, Stow JL, Weninger W, Haass NK, Beaumont KA. RAB27A promotes melanoma cell invasion and metastasis via regulation of pro-invasive exosomes. Int J Cancer 2019; 144:3070-3085. [PMID: 30556600 DOI: 10.1002/ijc.32064] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 11/30/2018] [Indexed: 01/03/2023]
Abstract
Despite recent advances in targeted and immune-based therapies, advanced stage melanoma remains a clinical challenge with a poor prognosis. Understanding the genes and cellular processes that drive progression and metastasis is critical for identifying new therapeutic strategies. Here, we found that the GTPase RAB27A was overexpressed in a subset of melanomas, which correlated with poor patient survival. Loss of RAB27A expression in melanoma cell lines inhibited 3D spheroid invasion and cell motility in vitro, and spontaneous metastasis in vivo. The reduced invasion phenotype was rescued by RAB27A-replete exosomes, but not RAB27A-knockdown exosomes, indicating that RAB27A is responsible for the generation of pro-invasive exosomes. Furthermore, while RAB27A loss did not alter the number of exosomes secreted, it did change exosome size and altered the composition and abundance of exosomal proteins, some of which are known to regulate cancer cell movement. Our data suggest that RAB27A promotes the biogenesis of a distinct pro-invasive exosome population. These findings support RAB27A as a key cancer regulator, as well as a potential prognostic marker and therapeutic target in melanoma.
Collapse
Affiliation(s)
- Dajiang Guo
- The Centenary Institute, The University of Sydney, Newtown, NSW, Australia.,Sydney Medical School, The University of Sydney, Camperdown, NSW, Australia
| | - Goldie Y L Lui
- The Centenary Institute, The University of Sydney, Newtown, NSW, Australia.,Sydney Medical School, The University of Sydney, Camperdown, NSW, Australia
| | - Siew Li Lai
- The Centenary Institute, The University of Sydney, Newtown, NSW, Australia
| | - James S Wilmott
- Sydney Medical School, The University of Sydney, Camperdown, NSW, Australia.,Melanoma Institute Australia, The University of Sydney, North Sydney, NSW, Australia
| | - Shweta Tikoo
- The Centenary Institute, The University of Sydney, Newtown, NSW, Australia.,Sydney Medical School, The University of Sydney, Camperdown, NSW, Australia
| | - Louise A Jackett
- Sydney Medical School, The University of Sydney, Camperdown, NSW, Australia.,Melanoma Institute Australia, The University of Sydney, North Sydney, NSW, Australia.,Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| | - Camelia Quek
- Melanoma Institute Australia, The University of Sydney, North Sydney, NSW, Australia
| | - Darren L Brown
- The Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Danae M Sharp
- The Centenary Institute, The University of Sydney, Newtown, NSW, Australia.,Sydney Medical School, The University of Sydney, Camperdown, NSW, Australia
| | - Rain Y Q Kwan
- The Centenary Institute, The University of Sydney, Newtown, NSW, Australia.,Sydney Medical School, The University of Sydney, Camperdown, NSW, Australia
| | - Diego Chacon
- Centre for Health Technologies and the School of Biomedical Engineering, University of Technology, Sydney, NSW, Australia.,Adult Cancer Program, Lowy Cancer Research Centre, Prince of Wales Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Jason H Wong
- Adult Cancer Program, Lowy Cancer Research Centre, Prince of Wales Clinical School, University of New South Wales, Sydney, NSW, Australia.,School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Dominik Beck
- Centre for Health Technologies and the School of Biomedical Engineering, University of Technology, Sydney, NSW, Australia.,Adult Cancer Program, Lowy Cancer Research Centre, Prince of Wales Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Michelle van Geldermalsen
- The Centenary Institute, The University of Sydney, Newtown, NSW, Australia.,Sydney Medical School, The University of Sydney, Camperdown, NSW, Australia
| | - Jeff Holst
- The Centenary Institute, The University of Sydney, Newtown, NSW, Australia.,Sydney Medical School, The University of Sydney, Camperdown, NSW, Australia
| | - John F Thompson
- Sydney Medical School, The University of Sydney, Camperdown, NSW, Australia.,Melanoma Institute Australia, The University of Sydney, North Sydney, NSW, Australia.,Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| | - Graham J Mann
- Melanoma Institute Australia, The University of Sydney, North Sydney, NSW, Australia.,Centre for Cancer Research, Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW, Australia
| | - Richard A Scolyer
- Sydney Medical School, The University of Sydney, Camperdown, NSW, Australia.,Melanoma Institute Australia, The University of Sydney, North Sydney, NSW, Australia.,Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| | - Jennifer L Stow
- The Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Wolfgang Weninger
- The Centenary Institute, The University of Sydney, Newtown, NSW, Australia.,Discipline of Dermatology, The University of Sydney, Camperdown, NSW, Australia.,Department of Dermatology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| | - Nikolas K Haass
- The Centenary Institute, The University of Sydney, Newtown, NSW, Australia.,Discipline of Dermatology, The University of Sydney, Camperdown, NSW, Australia.,The University of Queensland, The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, QLD, Australia
| | - Kimberley A Beaumont
- The Centenary Institute, The University of Sydney, Newtown, NSW, Australia.,Sydney Medical School, The University of Sydney, Camperdown, NSW, Australia
| |
Collapse
|
16
|
The Crosstalk between Cancer Stem Cells and Microenvironment Is Critical for Solid Tumor Progression: The Significant Contribution of Extracellular Vesicles. Stem Cells Int 2018; 2018:6392198. [PMID: 30532788 PMCID: PMC6247433 DOI: 10.1155/2018/6392198] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 10/02/2018] [Accepted: 10/11/2018] [Indexed: 12/12/2022] Open
Abstract
Several evidences nowadays demonstrated the critical role of the microenvironment in regulating cancer stem cells and their involvement in tumor progression. Extracellular vesicles (EVs) are considered as one of the most effective vehicles of information among cells. Accordingly, a number of studies led to the recognition of stem cell-associated EVs as new complexes able to contribute to cell fate determination of either normal or tumor cells. In this review, we aim to highlight an existing bidirectional role of EV-mediated communication—from cancer stem cells to microenvironment and also from microenvironment to cancer stem cells—in the most widespread solid cancers as prostate, breast, lung, and colon tumors.
Collapse
|
17
|
Yu X, Fang Z, Li G, Zhang S, Liu M, Wang Y. High RASEF expression is associated with a significantly better prognosis in colorectal cancer. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2018; 11:4276-4282. [PMID: 31949824 PMCID: PMC6962987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Accepted: 07/31/2018] [Indexed: 06/10/2023]
Abstract
This study mainly studied the correlation of RASEF expression and the clinical index of colorectal cancer by tissue microarray (TMAs, HCol-Adel180sur-06) containing tissue samples of 90 colorectal cancers. The results showed that RASEF was significantly highly expressed both in nuclei (3.07±1.95 vs 1.83±1.74, P=0.000) and cytoplasm (7.74±2.08 vs 5.83±1.97, P=0.000) compared to their para-carcinoma tissues, which was in line with the data of the Oncomine database. The correlation between RASEF expression and microsatellite instability, analyzed by Spearman's correlation analysis showed that RASEF expression in colorectal cancer cytoplasm was correlated significantly with the mismatch repair genes MLH1 (P=0.037; r=0.227) and MSH6 (P=0.038; r=0.224). Additionally, high RASEF expression was associated with a significantly better prognosis (45.3% vs 8%, P=0.041), which was consistent with the data of the Human Protein Atlas. Subsequently, Cox analysis of multi-factor survival showed that RASEF expression was an independent predictive factor for colorectal cancer (P=0.001). Thus, we speculated that RASEF may be a suppressor gene, and may inhibit the development of colorectal cancer through participating in DNA repair processes.
Collapse
Affiliation(s)
- Xin Yu
- Department of Internal Medicine, Sino-Singapore Eco-City Hospital of Tianjin Medical UniversityTianjin, China
| | - Zhenhuan Fang
- Department of Internal Medicine, Sino-Singapore Eco-City Hospital of Tianjin Medical UniversityTianjin, China
| | - Guodong Li
- Department of General Surgery, The Fourth-Affiliated Hospital of Harbin Medical UniversityHarbin, China
| | - Shujun Zhang
- Department of Pathology, The Fourth-Affiliated Hospital of Harbin Medical UniversityHarbin, China
| | - Ming Liu
- Department of General Surgery, The Fourth-Affiliated Hospital of Harbin Medical UniversityHarbin, China
- Bio-Bank of Department of General Surgery, The Fourth-Affiliated Hospital of Harbin Medical UniversityHarbin, China
| | - Ying Wang
- Shanghai Outdo Biotech Co., Ltd.Shanghai, China
| |
Collapse
|
18
|
Liu J, Gong X, Zhu X, Xue D, Liu Y, Wang P. Rab27A overexpression promotes bladder cancer proliferation and chemoresistance through regulation of NF-κB signaling. Oncotarget 2017; 8:75272-75283. [PMID: 29088864 PMCID: PMC5650419 DOI: 10.18632/oncotarget.20775] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 07/29/2017] [Indexed: 11/25/2022] Open
Abstract
Overexpression of Rab27A has been found in human cancers. However, the clinical significance and biological effects of Rab27A in bladder cancer tissues and cell lines have not been investigated. Here, we checked Rab27A protein in 87 cases of bladder cancer using immunohistochemistry. We found that Rab27A was overexpressed in 39 of 87 (44.8%) cancer cases. Significant association was found between Rab27 and invading depth (p=0.0083). We knocked down Rab27A in 5637 cell line and transfected Rab27A plasmid in BIU-87 cell line. Rab27A depletion inhibited cell growth rate and invasion while its overexpression induced cell growth and invasion. Rab27A also promoted cancer cell growth in vivo. Cell viability and Annexin V/PI staining demonstrated that Rab27A maintained cancer cell survival and reduced apoptosis rate when treated with cisplatin. JC-1 staining showed that Rab27A upregulated mitochondrial membrane potential. Western blot demonstrated that Rab27A overexpression upregulated cyclin D1, cyclin E, p-IκB, p-p65, Bcl-2, cIAP1, cIAP2 protein expression. NF-κB inhibitor BAY 11-7082 abolished the effects of Rab27 on cisplatin resistance and Bcl-2 protein. In conclusion, the present study demonstrated that Rab27A overexpression facilitates bladder cancer growth, invasion and chemoresistance in bladder cancer, possibly through regulation of NF-κB signaling pathway.
Collapse
Affiliation(s)
- Jia Liu
- Department of Urology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Xue Gong
- Department of Urology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Xingwang Zhu
- Department of Urology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Dongwei Xue
- Department of Urology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Yili Liu
- Department of Urology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Ping Wang
- Department of Urology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
19
|
Lei X, Li K, Liu Y, Wang ZY, Ruan BJ, Wang L, Xiang A, Wu D, Lu Z. Co-delivery nanocarriers targeting folate receptor and encapsulating 2-deoxyglucose and α-tocopheryl succinate enhance anti-tumor effect in vivo. Int J Nanomedicine 2017; 12:5701-5715. [PMID: 28848348 PMCID: PMC5557622 DOI: 10.2147/ijn.s135849] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
A combination administration of chemical agents was highlighted to treat tumors. Recently, tumor cell has been found to be different from normal cell in metabolic manner. Most of cancer cells prefer aerobic glycolysis to mitochondrial oxidative phosphorylation (OXPHOS) to satisfy energy and biomass synthesis requirement to survive, grow and proliferate, which provides novel and potential therapeutic targets for chemotherapy. Here, 2-deoxy-d-glucose (2-DG), a potent inhibitor of glucose metabolism, was used to inhibit glycolysis of tumor cells; α-tocopheryl succinate (α-TOS), a water-insoluble vitamin E derivative, was chosen to suppress OXPHOS. Our data demonstrated that the combination treatment of 2-DG and α-TOS could significantly promote the anti-tumor efficiency in vitro compared with administration of the single drug. In order to maximize therapeutic activity and minimize negative side effects, a co-delivery nanocarrier targeting folate receptor (FR) was developed to encapsulate 2-DG and α-TOS simultaneously based on our previous work. Transmission electron microscope, dynamic light scattering method and UV-visible spectrophotometers were used to investigate morphology, size distribution and loading efficiency of the α-TOS-2-DG-loaded and FR-targeted nanoparticles (TDF NPs). The TDF NPs were found to possess a layer-by-layer shape, and the dynamic size was <100 nm. The final encapsulation efficiencies of α-TOS and 2-DG in TDF NPs were 94.3%±1.3% and 61.7%±7.7% with respect to drug-loading capacities of 8.9%±0.8% and 13.2%±2.6%, respectively. Almost no α-TOS release was found within 80 h, and release of 2-DG was sustained and slow within 72 h. The results of FR binding assay and fluorescence biodistribution revealed that TDF NPs could target FR highly expressed on tumor cell in vitro and in vivo. Further, in vivo anti-tumor experiments showed that TDF NPs had an improved biological function with less toxicity. Thus, our work indicates that the co-delivery TDF NPs have a great potential in tumor therapy.
Collapse
Affiliation(s)
- Xiaoying Lei
- State Key Laboratory of Cancer Biology, Department of Pharmacogenomics, School of Pharmacy, The Fourth Military Medical University
| | - Ke Li
- Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, Shaanxi.,Key Laboratory of Biomedical Information Engineering of Education Ministry, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an
| | - Yan Liu
- Genetic Engineering Laboratory of PLA, The Eleventh Institute of Academy of Military Medical Sciences of PLA, Changchun, Jilin, People's Republic of China
| | - Zhen Yu Wang
- State Key Laboratory of Cancer Biology, Department of Pharmacogenomics, School of Pharmacy, The Fourth Military Medical University
| | - Ban Jun Ruan
- State Key Laboratory of Cancer Biology, Department of Pharmacogenomics, School of Pharmacy, The Fourth Military Medical University
| | - Li Wang
- State Key Laboratory of Cancer Biology, Department of Pharmacogenomics, School of Pharmacy, The Fourth Military Medical University
| | - An Xiang
- State Key Laboratory of Cancer Biology, Department of Pharmacogenomics, School of Pharmacy, The Fourth Military Medical University
| | - Daocheng Wu
- Key Laboratory of Biomedical Information Engineering of Education Ministry, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an
| | - Zifan Lu
- State Key Laboratory of Cancer Biology, Department of Pharmacogenomics, School of Pharmacy, The Fourth Military Medical University
| |
Collapse
|
20
|
Qiu GZ, Sun W, Jin MZ, Lin J, Lu PG, Jin WL. The bad seed gardener: Deubiquitinases in the cancer stem-cell signaling network and therapeutic resistance. Pharmacol Ther 2017; 172:127-138. [DOI: 10.1016/j.pharmthera.2016.12.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
21
|
Su WF, Gu Y, Wei ZY, Shen YT, Jin ZH, Yuan Y, Gu XS, Chen G. Rab27a/Slp2-a complex is involved in Schwann cell myelination. Neural Regen Res 2016; 11:1830-1838. [PMID: 28123429 PMCID: PMC5204241 DOI: 10.4103/1673-5374.194755] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Myelination of Schwann cells in the peripheral nervous system is an intricate process involving myelin protein trafficking. Recently, the role and mechanism of the endosomal/lysosomal system in myelin formation were emphasized. Our previous results demonstrated that a small GTPase Rab27a regulates lysosomal exocytosis and myelin protein trafficking in Schwann cells. In this present study, we established a dorsal root ganglion (DRG) neuron and Schwann cell co-culture model to identify the signals associated with Rab27a during myelination. First, Slp2-a, as the Rab27a effector, was endogenously expressed in Schwann cells. Second, Rab27a expression significantly increased during Schwann cell myelination. Finally, Rab27a and Slp2-a silencing in Schwann cells not only reduced myelin protein expression, but also impaired formation of myelin-like membranes in DRG neuron and Schwann cell co-cultures. Our findings suggest that the Rab27a/Slp2-a complex affects Schwann cell myelination in vitro.
Collapse
Affiliation(s)
- Wen-Feng Su
- Jiangsu Key Laboratory of Neuroregeneration, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Yun Gu
- Jiangsu Key Laboratory of Neuroregeneration, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Zhong-Ya Wei
- Jiangsu Key Laboratory of Neuroregeneration, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Yun-Tian Shen
- Jiangsu Key Laboratory of Neuroregeneration, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Zi-Han Jin
- Jiangsu Key Laboratory of Neuroregeneration, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Ying Yuan
- Jiangsu Key Laboratory of Neuroregeneration, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China; Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Xiao-Song Gu
- Jiangsu Key Laboratory of Neuroregeneration, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Gang Chen
- Jiangsu Key Laboratory of Neuroregeneration, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| |
Collapse
|