1
|
Wei W, Zhang Y, Li Y, Huang J, Kang F, Tan S, Lin L, Lu X, Wei H, Wang N. Hypoxia-mediated high expression of TRIM15 promotes malignant progression of high-grade serous ovarian cancer through activation of AKT signaling pathway by K63 ubiquitination. Int J Cancer 2025; 156:2401-2415. [PMID: 40026037 DOI: 10.1002/ijc.35387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 12/24/2024] [Accepted: 02/10/2025] [Indexed: 03/04/2025]
Abstract
The tripartite motif (TRIM) family member TRIM15 is an E3 ubiquitin ligase that is abnormally expressed in a variety of tumors, but its role and mechanism in high-grade serous ovarian cancer (HGSOC) are unclear. Here, we found for the first time that TRIM15 was upregulated in HGSOC and was associated with poor overall survival. Functional experiments showed that TRIM15 drove the proliferation of HGSOC cells and inhibited the apoptosis of tumor cells in vivo and in vitro. In terms of mechanism, we found that TRIM15 contributed to the malignant proliferation of HGSOC cells by promoting the activation of AKT and that there was a direct binding between them. TRIM15 induced lysine-63 (K63) ubiquitination of AKT through its Ring domain, which in turn activated the AKT signaling pathway. In addition, TRIM15-mediated K63 ubiquitination occurs mainly in the pleckstrin homology (PH) domain of AKT. We further identified other proteins and their functions regulated by TRIM15 in HGSOC cells by ubiquitin proteomic analysis. Furthermore, hypoxia-inducible factor-1α promoted TRIM15 transcriptional activation by binding to the hypoxia response elements of the TRIM15 promoter. Our study suggests that TRIM15 induces K63 ubiquitination of the AKT PH domain through its Ring domain and activates the AKT signaling pathway, thereby promoting HGSOC progression. In addition, the abnormally high expression of TRIM15 was associated with the hypoxic microenvironment of HGSOC tissues.
Collapse
Affiliation(s)
- Wei Wei
- Department of Obstetrics and Gynecology, the Second Hospital of Dalian Medical University, Dalian, People's Republic of China
| | - Yang Zhang
- Department of Breast Surgery, the Second Hospital of Dalian Medical University, Dalian, People's Republic of China
| | - Yibing Li
- Department of Obstetrics and Gynecology, the Second Hospital of Dalian Medical University, Dalian, People's Republic of China
| | - Jiazhen Huang
- Department of Obstetrics and Gynecology, the Second Hospital of Dalian Medical University, Dalian, People's Republic of China
| | - Fuli Kang
- Department of Obstetrics and Gynecology, the Second Hospital of Dalian Medical University, Dalian, People's Republic of China
| | - Shuang Tan
- Department of Obstetrics and Gynecology, the Second Hospital of Dalian Medical University, Dalian, People's Republic of China
| | - Lin Lin
- Department of Obstetrics and Gynecology, the Second Hospital of Dalian Medical University, Dalian, People's Republic of China
| | - Xiaohang Lu
- Department of Obstetrics and Gynecology, the Second Hospital of Dalian Medical University, Dalian, People's Republic of China
| | - Heng Wei
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China
| | - Ning Wang
- Department of Obstetrics and Gynecology, the Second Hospital of Dalian Medical University, Dalian, People's Republic of China
| |
Collapse
|
2
|
Wang Y, Wang S, He H, Bai Y, Liu Z, Sabihi SS. Mechanisms of apoptosis-related non-coding RNAs in ovarian cancer: a narrative review. Apoptosis 2025; 30:553-578. [PMID: 39833637 DOI: 10.1007/s10495-024-02074-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/27/2024] [Indexed: 01/22/2025]
Abstract
Ovarian cancer remains a major challenge in oncology due to its complex biology and late-stage diagnosis. Recent advances in molecular biology have highlighted the crucial role of non-coding RNAs (ncRNAs) in regulating apoptosis and cancer progression. NcRNAs, including microRNAs, long non-coding RNAs, and circular RNAs, have emerged as significant players in the molecular networks governing ovarian cancer. Despite these insights, the precise mechanisms by which ncRNAs influence ovarian cancer pathology are not fully understood. This complexity, combined with the heterogeneity of the disease and the development of treatment resistance, poses substantial obstacles to effective therapeutic development. Additionally, the lack of reliable early detection methods further complicates treatment strategies. This manuscript reviews the current state of research on ncRNAs in ovarian cancer, discusses the challenges in translating these findings into clinical applications, and outlines potential future directions. Emphasis is placed on the need for integrated approaches to unravel the intricate roles of ncRNAs, improve early detection, and develop personalized treatment strategies to address the diverse and evolving nature of ovarian cancer. While these findings provide valuable insights, it is crucial to recognize that many results are based on preclinical studies and require further validation to establish their clinical applicability.
Collapse
Affiliation(s)
- Yue Wang
- Department of Obstetrics and Gynecology, Tang Du Hospital, The Air Force Military Medical University, Xi'an, 710038, China
| | - Shirui Wang
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710038, China
| | - Haiyan He
- Department of Obstetrics and Gynecology, Tang Du Hospital, The Air Force Military Medical University, Xi'an, 710038, China
| | - Yingying Bai
- Department of Obstetrics and Gynecology, Tang Du Hospital, The Air Force Military Medical University, Xi'an, 710038, China
| | - Zhuo Liu
- Department of Obstetrics and Gynecology, Xi'an International Medical Center Hospital, Xi'an, 710038, China
| | - Sima-Sadat Sabihi
- Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
3
|
Thibault B, Thole A, D'Angelo R, Basset C, Guillermet-Guibert J. PI3Kα-specific inhibitor BYL-719 synergizes with cisplatin in vitro in PIK3CA-mutated ovarian cancer cells. Sci Rep 2025; 15:6265. [PMID: 39979449 PMCID: PMC11842864 DOI: 10.1038/s41598-025-90714-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 02/14/2025] [Indexed: 02/22/2025] Open
Abstract
Peritoneal carcinomatosis in ovarian cancer is often associated with ascites where cancer cells grow as aggregates. Given the emerging evidence that multicellular growth enhances resistance to conventional therapies, and that patients frequently develop resistance to platinum salts, we investigated the efficiency of PI3K/Akt signalling pathway targeting in multicellular growth and its importance as a potential therapeutic target in cells resistant to platinum salts. Due to its importance in many cancers and to the frequent mutations of its encoding gene PIK3CA, we focused on targeting PI3Kα using BYL-719 (Alpelisib), an isoform-specific inhibitor already used in clinics. We used a panel of 3 ovarian cancer cell lines, SKOV-3, EFO-21 and OVCAR-3, which come from different histological origins and bear different mutations. PI3K targeting drugs inhibit the activity of the PI3K/Akt pathway in all tested ovarian cancer cell lines with a drastic reduction of the phosphorylation of Akt on the serine 473, regardless the histology or the mutational profile. We showed that when cultured in 3D aggregates, ovarian cancer cells are more resistant to the PI3Kα-specific inhibitor BYL-719 and cisplatin compared to 2D monolayers. BYL-719 synergizes with cisplatin in 3D cultures only in PIK3CA-mutated SKOV-3 cells. This drug combination leads to a major cytotoxicity in 3D aggregates of this cell line. Finally, BYL-719 in combination with cisplatin remains active in 3D aggregates of SKOV-3 cells co-cultured with mesenchymal stem cells. We have identified a signalling pathway of interest for the treatment of advanced ovarian cancer in vitro, which could limit the progression of this disease. These data pave the road to investigate whether PI3Kα-specific inhibitor BYL-719 should be proposed in combination with cisplatin, in priority in patients bearing a PIK3CA mutation.
Collapse
Affiliation(s)
- Benoît Thibault
- CRCT, Université de Toulouse, Inserm, CNRS, Centre de Recherches en Cancérologie de Toulouse, Equipe SigDYN, 2 av Hubert Curien, Oncopole de Toulouse, Toulouse, France.
- Labex TOUCAN, ANR, Toulouse, France.
| | - Adrien Thole
- CRCT, Université de Toulouse, Inserm, CNRS, Centre de Recherches en Cancérologie de Toulouse, Equipe SigDYN, 2 av Hubert Curien, Oncopole de Toulouse, Toulouse, France
- Labex TOUCAN, ANR, Toulouse, France
| | - Romina D'Angelo
- CRCT, Université de Toulouse, Inserm, CNRS, Centre de Recherches en Cancérologie de Toulouse, Equipe SigDYN, 2 av Hubert Curien, Oncopole de Toulouse, Toulouse, France
- Labex TOUCAN, ANR, Toulouse, France
| | - Céline Basset
- CRCT, Université de Toulouse, Inserm, CNRS, Centre de Recherches en Cancérologie de Toulouse, Equipe SigDYN, 2 av Hubert Curien, Oncopole de Toulouse, Toulouse, France
- Labex TOUCAN, ANR, Toulouse, France
- Service d'Anatomo-Pathologie, Institut Universitaire du Cancer de Toulouse-Oncopole (IUCT-O), avenue Irène Joliot-Curie, Toulouse, France
| | - Julie Guillermet-Guibert
- CRCT, Université de Toulouse, Inserm, CNRS, Centre de Recherches en Cancérologie de Toulouse, Equipe SigDYN, 2 av Hubert Curien, Oncopole de Toulouse, Toulouse, France.
- Labex TOUCAN, ANR, Toulouse, France.
| |
Collapse
|
4
|
Meng Q, Zheng W, Jiao R, Cui R, Deng Y, Liu R, Wang J, Bai H. MicroRNA 421 induces the formation of high-invasive cell subsets of ovarian cancer from low-invasive cell subsets mediated by exosomes by activating the PI3K/AKT pathway. Am J Cancer Res 2024; 14:2643-2660. [PMID: 38859864 PMCID: PMC11162662 DOI: 10.62347/uhey7375] [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: 01/15/2024] [Accepted: 05/15/2024] [Indexed: 06/12/2024] Open
Abstract
Intratumoral heterogeneity (ITH) results in treatment failure in ovarian cancer (OC). Exosomes are related to the formation of a heterogeneous tumor microenvironment, and microRNAs play a crucial role in the progression of OC. Therefore, we aimed to explore the effect of exosomes and microRNA 421 (miR-421), which is mediated by exosomes, on ITH and the diagnosis of OC. Exosomes derived from A2780 cells with the highest (AHC) or lowest (ALC) invasive/migratory capacity cells (AHE/ALE) were extracted by differential centrifugation. We conducted a series of experiments to verify the role of AHE and miR-421 in promoting the transformation of low-invasive cells to high-invasive cells by regulating the PI3K/AKT pathway, and we also measured the levels of CA125 in serum exosomes. The results of assays showed that the AHE and miR-421, mediated by exosomes, significantly increased the malignancy of ALC cells by activating the PI3K/AKT pathway. The expression of miR-421 was significantly increased in the serum exosomes derived from high-grade serous ovarian cancer (HGSOC) patients. Our findings indicate that MiR-421, mediated by exosomes, could induce the transformation of highly invasive cell subpopulations from subpopulations of OC cells with low invasive potential by activating the PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Qianlong Meng
- Department of Gynecology, Fuxing Hospital, Capital Medical UniversityBeijing, China
- Department of Diagnostics of Clinical Laboratory, Zhejiang HospitalHangzhou, Zhejiang, China
| | - Wei Zheng
- Department of Gynecology, Fuxing Hospital, Capital Medical UniversityBeijing, China
| | - Ruili Jiao
- Department of Obstetrics and Gynecology, Beijing Chaoyang District Maternal and Child Health HospitalBeijing, China
| | - Ran Cui
- Department of Obstetrics and Gynecology, Peking University First HospitalBeijing, China
| | - Yunhan Deng
- Department of Obstetrics and Gynecology, Beijing Chaoyang Hospital, Capital Medical UniversityBeijing, China
| | - Ruizhen Liu
- Department of Obstetrics and Gynecology, Beijing Chaoyang Hospital, Capital Medical UniversityBeijing, China
| | - Jing Wang
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders and National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical UniversityBeijing, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical UniversityBeijing, China
| | - Huimin Bai
- Department of Gynecology, Fuxing Hospital, Capital Medical UniversityBeijing, China
| |
Collapse
|
5
|
Bi X, Liu S, Liu D, Li C. Elucidating the role of Brain-Derived Neurotrophic Factor (BDNF) and its receptor Tyrosine Receptor Kinase B (TrkB) in the development and symptoms of endometriosis. Int J Neurosci 2024:1-7. [PMID: 38287513 DOI: 10.1080/00207454.2023.2285709] [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: 10/26/2023] [Accepted: 11/15/2023] [Indexed: 01/31/2024]
Abstract
Endometriosis (EMs) is a common disease among women of reproductive age, and as of now, the clinical understanding of the etiology of this disease remains unclear. The occurrence of EMs has a profound impact on the reproductive health of women, making early diagnosis and treatment of this disease a pressing challenge in clinical practice. Recent studies have found that Brain-Derived Neurotrophic Factor (BDNF), in combination with its high-affinity receptor Tyrosine Receptor Kinase B (TrkB), participates in the development of EMs and the appearance of clinically relevant symptoms by activating the Mitogen-Activated Protein Kinase (MAPK) pathway, the Phosphatidylinositol 3-Kinase/Protein Kinase B (PI3K/AKT) pathway, and the Phospholipase C-gamma (PLCγ) signaling pathway, or by interacting with other factors. In order to gain a deeper understanding of the pathogenesis related to EMs, this article reviews the roles of BDNF and TrkB in EMs, particularly in terms of aberrant apoptosis and autophagy, cell invasion, proliferation, angiogenesis, oxidative stress, and inflammatory reactions, as well as their relationship with the symptoms associated with EMs.
Collapse
Affiliation(s)
- Xinyi Bi
- Department of Gynaecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, P.R. China
| | - Shulan Liu
- Department of Gynaecology, Gaomi Traditional Chinese Medicine Hospital, Gaomi, P.R. China
| | - Degao Liu
- Center of Obstetrics and Gynecology, Peking University Shenzhen Hospital, Shenzhen, P.R. China
| | - Changzhong Li
- Center of Obstetrics and Gynecology, Peking University Shenzhen Hospital, Shenzhen, P.R. China
- Department of Gynaecology, Shandong provincial hospital affiliated to Shandong first medical university, Jinan, China
- Institute of Obstetrics and Gynecology, Shenzhen PKU-HKUST Medical Center, Shenzhen, P.R. China
- Shenzhen Key Laboratory on Technology for Early Diagnosis of Major Gynecologic Diseases, Shenzhen, P.R. China
| |
Collapse
|
6
|
Pawar NR, Buzza MS, Duru N, Strong AA, Antalis TM. Matriptase drives dissemination of ovarian cancer spheroids by a PAR-2/PI3K/Akt/MMP9 signaling axis. J Cell Biol 2023; 222:e202209114. [PMID: 37737895 PMCID: PMC10515437 DOI: 10.1083/jcb.202209114] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 07/06/2023] [Accepted: 08/14/2023] [Indexed: 09/23/2023] Open
Abstract
The transmembrane serine protease matriptase is a key regulator of both barrier-disruptive and protective epithelial cell-cell interactions. Elevated matriptase is a consistent feature of epithelial ovarian cancers (OvCa), where multicellular spheroids shed from the primary tumor into the peritoneal cavity are critical drivers of metastasis. Dynamic cell-to-cell adhesive contacts are required for spheroid formation and maintenance. Here, we show that overactive matriptase, reflected in an increased ratio of matriptase to its inhibitor hepatocyte growth factor activator inhibitor 1 (HAI-1), disrupts cell-cell contacts to produce loose prometastatic spheroids that display increased mesothelial cell adhesion and submesothelial invasion. We show that these activities are dependent on the matriptase activation of a protease-activated receptor-2 (PAR-2) signaling pathway involving PI3K/Akt and MMP9-induced disruption of cell-cell adhesion by the release of the soluble E-cadherin ectodomain. These data reveal a novel pathological connection between matriptase activation of PAR-2 and disruption of cell-cell adhesion, and support the clinical investigation of this signaling axis as a therapeutic strategy for aggressive metastatic OvCa.
Collapse
Affiliation(s)
- Nisha R. Pawar
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Marguerite S. Buzza
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA
- Research and Development Service, VA Maryland Health Care System, Baltimore, MD, USA
| | - Nadire Duru
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Amando A. Strong
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Toni M. Antalis
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA
- Research and Development Service, VA Maryland Health Care System, Baltimore, MD, USA
| |
Collapse
|
7
|
Nurzadeh M, Ghalandarpoor-Attar SM, Ghalandarpoor-Attar SN, Rabiei M. The sequestosome 1 protein: therapeutic vulnerabilities in ovarian cancer. Clin Transl Oncol 2023; 25:2783-2792. [PMID: 36964889 DOI: 10.1007/s12094-023-03148-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Accepted: 03/04/2023] [Indexed: 03/26/2023]
Abstract
Ovarian cancer (OC) is the most deadly tumor that may develop in a woman's reproductive system. It is also one of the most common causes of death among those who have been diagnosed with cancer in women. An adapter protein known as sequestosome 1(SQSTM1) or p62 is primarily responsible for the transportation, degradation, and destruction of a wide variety of proteins. This adapter protein works in conjunction with the autophagy process as well as the ubiquitin proteasome degradation pathway. In addition, the ability of SQSTM1 to interact with multiple binding partners link SQSTM1 to various pathways in the context of antioxidant defense system and inflammation. In this review, we outline the processes underlying the control that SQSTM1 has on these pathways and how their dysregulation contributes to the development of OC. At the final, the therapeutic approaches based on SQSTM1 targeting have been discussed.
Collapse
Affiliation(s)
- Maryam Nurzadeh
- Fetomaternal Department, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | - Maryam Rabiei
- Obstetrics and Gynecology Department, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
8
|
Tierno D, Azzalini E, Farra R, Drioli S, Felluga F, Lazzarino M, Grassi G, Dapas B, Bonin S. Nanomechanical Characterization of Ovarian Cancer Cell Lines as a Marker of Response to 2c Treatment. Int J Mol Sci 2023; 24:7230. [PMID: 37108391 PMCID: PMC10139025 DOI: 10.3390/ijms24087230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 04/07/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
Epithelial ovarian cancers (EOCs) are a heterogeneous group of tumors with different molecular and clinical features. In past decades, few improvements have been achieved in terms of EOC management and treatment efficacy, such that the 5-year survival rate of patients remained almost unchanged. A better characterization of EOCs' heterogeneity is needed to identify cancer vulnerabilities, stratify patients and adopt proper therapies. The mechanical features of malignant cells are emerging as new biomarkers of cancer invasiveness and drug resistance that can further improve our knowledge of EOC biology and allow the identification of new molecular targets. In this study, we determined the inter and intra-mechanical heterogeneity of eight ovarian cancer cell lines and their association with tumor invasiveness and resistance to an anti-tumoral drug with cytoskeleton depolymerization activity (2c).
Collapse
Affiliation(s)
- Domenico Tierno
- Department of Medical Sciences (DSM), University of Trieste, 34149 Trieste, Italy; (D.T.); (E.A.)
| | - Eros Azzalini
- Department of Medical Sciences (DSM), University of Trieste, 34149 Trieste, Italy; (D.T.); (E.A.)
| | - Rossella Farra
- Department of Life Sciences (DSV), University of Trieste, 34128 Trieste, Italy; (R.F.); (G.G.); (B.D.)
| | - Sara Drioli
- Department of Chemical and Pharmaceutical Sciences (DSCF), University of Trieste, 34127 Trieste, Italy; (S.D.); (F.F.)
| | - Fulvia Felluga
- Department of Chemical and Pharmaceutical Sciences (DSCF), University of Trieste, 34127 Trieste, Italy; (S.D.); (F.F.)
| | - Marco Lazzarino
- Consiglio Nazionale delle Ricerche, Istituto Officina dei Materiali (IOM), 34149 Trieste, Italy;
| | - Gabriele Grassi
- Department of Life Sciences (DSV), University of Trieste, 34128 Trieste, Italy; (R.F.); (G.G.); (B.D.)
| | - Barbara Dapas
- Department of Life Sciences (DSV), University of Trieste, 34128 Trieste, Italy; (R.F.); (G.G.); (B.D.)
| | - Serena Bonin
- Department of Medical Sciences (DSM), University of Trieste, 34149 Trieste, Italy; (D.T.); (E.A.)
| |
Collapse
|
9
|
Czegle I, Huang C, Soria PG, Purkiss DW, Shields A, Wappler-Guzzetta EA. The Role of Genetic Mutations in Mitochondrial-Driven Cancer Growth in Selected Tumors: Breast and Gynecological Malignancies. Life (Basel) 2023; 13:996. [PMID: 37109525 PMCID: PMC10145875 DOI: 10.3390/life13040996] [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: 12/28/2022] [Revised: 03/15/2023] [Accepted: 03/31/2023] [Indexed: 04/29/2023] Open
Abstract
There is an increasing understanding of the molecular and cytogenetic background of various tumors that helps us better conceptualize the pathogenesis of specific diseases. Additionally, in many cases, these molecular and cytogenetic alterations have diagnostic, prognostic, and/or therapeutic applications that are heavily used in clinical practice. Given that there is always room for improvement in cancer treatments and in cancer patient management, it is important to discover new therapeutic targets for affected individuals. In this review, we discuss mitochondrial changes in breast and gynecological (endometrial and ovarian) cancers. In addition, we review how the frequently altered genes in these diseases (BRCA1/2, HER2, PTEN, PIK3CA, CTNNB1, RAS, CTNNB1, FGFR, TP53, ARID1A, and TERT) affect the mitochondria, highlighting the possible associated individual therapeutic targets. With this approach, drugs targeting mitochondrial glucose or fatty acid metabolism, reactive oxygen species production, mitochondrial biogenesis, mtDNA transcription, mitophagy, or cell death pathways could provide further tailored treatment.
Collapse
Affiliation(s)
- Ibolya Czegle
- Department of Internal Medicine and Haematology, Semmelweis University, H-1085 Budapest, Hungary
| | - Chelsea Huang
- Department of Pathology and Laboratory Medicine, Loma Linda University Health, Loma Linda, CA 92354, USA
| | - Priscilla Geraldine Soria
- Department of Pathology and Laboratory Medicine, Loma Linda University Health, Loma Linda, CA 92354, USA
| | - Dylan Wesley Purkiss
- Department of Pathology and Laboratory Medicine, Loma Linda University Health, Loma Linda, CA 92354, USA
| | - Andrea Shields
- Department of Pathology and Laboratory Medicine, Loma Linda University Health, Loma Linda, CA 92354, USA
| | | |
Collapse
|
10
|
Dai Y, Masra N, Zhou L, Yu C, Jin W, Ni H. Hederagenin suppresses glioma cell biological activities via Nur77 in vitro study. Food Sci Nutr 2023; 11:1283-1296. [PMID: 36911825 PMCID: PMC10002964 DOI: 10.1002/fsn3.3163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 11/22/2022] [Indexed: 12/13/2022] Open
Abstract
The aim of this research was to discuss Hederagenin's antitumor effects on glioma by in vitro study. U251 and U87 cell lines were used as research target in our research. In the first step, the different Hed concentrations were treated to U251 and U87 cell lines, and the second step is Nur77 transfection in U251 and U87 with Hed treatment; measuring cell proliferation by MTT and EdU staining; evaluating cell invasion and migration abilities by transwell assay and relative gene and protein expressions by RT-qPCR and WB assay. Compared with NC group, U251 and U87 cell proliferation were significantly depressed with cell apoptosis significantly increasing, and cell invasion and migration abilities were significantly inhibited in Hed-treated groups (p < .05, respectively); however, with Nur77 transfection, the Hed's antitumor effects disappeared. Meanwhile, with Hed supplement, Nur77, PI3K, and AKT gene expressions were significantly downregulated (p < .05, respectively) in Hed-treated groups; and Nur77, p-PI3K, and p-AKT protein expressions were significantly decreased (p < .05, respectively) in Hed-treated groups. Hed had antitumor effects on glioma cell biological activities via Nur77/PI3K/AKT pathway in vitro study.
Collapse
Affiliation(s)
- Yuxiang Dai
- Department of Neurosurgery, Nanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjingChina
| | - Ngarmbaye Masra
- Department of Neurosurgery, Nanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjingChina
| | - Lu Zhou
- Department of Neurosurgery, Nanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjingChina
| | - Chen Yu
- Department of Neurosurgery, Nanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjingChina
| | - Wei Jin
- Department of Neurosurgery, Nanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjingChina
| | - Hongbin Ni
- Department of Neurosurgery, Nanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjingChina
| |
Collapse
|
11
|
Immunotherapeutic Approaches in Ovarian Cancer. Curr Issues Mol Biol 2023; 45:1233-1249. [PMID: 36826026 PMCID: PMC9955550 DOI: 10.3390/cimb45020081] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/27/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Ovarian cancer (OC) is gynecological cancer, and diagnosis and treatment are continuously advancing. Next-generation sequencing (NGS)-based diagnoses have emerged as novel methods for identifying molecules and pathways in cancer research. The NGS-based applications have expanded in OC research for early detection and identification of aberrant genes and dysregulation pathways, demonstrating comprehensive views of the entire transcriptome, such as fusion genes, genetic mutations, and gene expression profiling. Coinciding with advances in NGS-based diagnosis, treatment strategies for OC, such as molecular targeted therapy and immunotherapy, have also advanced. Immunotherapy is effective against many other cancers, and its efficacy against OC has also been demonstrated at the clinical phase. In this review, we describe several NGS-based applications for therapeutic targets of OC, and introduce current immunotherapeutic strategies, including vaccines, checkpoint inhibitors, and chimeric antigen receptor (CAR)-T cell transplantation, for effective diagnosis and treatment of OC.
Collapse
|
12
|
Nokhostin F, Azadehrah M, Azadehrah M. The multifaced role and therapeutic regulation of autophagy in ovarian cancer. Clin Transl Oncol 2022; 25:1207-1217. [PMID: 36534371 DOI: 10.1007/s12094-022-03045-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022]
Abstract
Ovarian cancer (OC) is one of the tumors that occurs most frequently in women. Autophagy is involved in cell homeostasis, biomolecule recycling, and survival, making it a potential target for anti-tumor drugs. It is worth noting that growing evidence reveals a close link between autophagy and OC. In the context of OC, autophagy demonstrates activity as both a tumor suppressor and a tumor promoter, depending on the context. Autophagy's exact function in OC is greatly reliant on the tumor microenvironment (TME) and other conditions, such as hypoxia, nutritional deficiency, chemotherapy, and so on. However, what can be concluded from different studies is that autophagy-related signaling pathways, especially PI3K/AKT/mTOR axis, increase in advanced stages and malignant phenotype of the disease reduces autophagy and ultimately leads to tumor progression. This study sought to present a thorough understanding of the role of autophagy-related signaling pathways in OC and existing therapies targeting these signaling pathways.
Collapse
Affiliation(s)
- Fahimeh Nokhostin
- Department of Obstetrics and Gynecology, Faculty of Medicine, Shahid Sadughi University of Medical Sciences, Yazd, Iran
| | - Mahboobeh Azadehrah
- Cancer Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Malihe Azadehrah
- Cancer Research Center, Golestan University of Medical Sciences, Gorgan, Iran.
| |
Collapse
|
13
|
Jie X, Du M, Zhang M, Jin X, Cai Q, Xu C, Zhang X. Mutation analysis of circulating tumor DNA and paired ascites and tumor tissues in ovarian cancer. Exp Ther Med 2022; 24:542. [PMID: 35978934 PMCID: PMC9366257 DOI: 10.3892/etm.2022.11479] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 05/20/2022] [Indexed: 11/24/2022] Open
Abstract
Circulating tumor DNA (ctDNA) is one conventional type of liquid biopsy that can be collected to dynamically monitor disease status. However, its potential clinical value and concordance with ascites samples or tumor biopsy needs to be evaluated further for patients with ovarian cancer. Therefore, the present study compared the mutation profiles among ctDNA, paired tumor tissue and ascites samples to explore their possible clinical value in ovarian cancer. Targeted next-generation sequencing was used to screen for mutations in 18 peripheral blood samples, six paired ascites samples and eight paired tumor tissues collected from patients with ovarian cancer. Functional analyses were performed using public databases. WebGestalt was used to perform Gene Ontology and pathway enrichment analyses. The cBioPortal for Cancer Genomics was used to assess therapeutic targets. Chilibot and Search Tool for the Retrieval of Interacting Genes/Proteins were used to obtain key genes and their functional interactions. Comparative analysis was performed among the three types of samples using Venn diagram. A total of 104 cancer-associated mutant genes in ctDNA samples, 95 genes in tumor tissues and 44 genes in ascites samples were found. A cluster covering 10 genes, namely NOTCH2, NOTCH3, lysine methyltransferase 2A, PTEN, androgen receptor, DNA-activated protein kinase catalytic subunit, hepatocyte nuclear factor 1 homeobox A, SRC, insulin receptor substrate 2 and SRY-box transcription factor 10, was obtained by Chilibot analysis. This gene panel may have the potential to monitor metastasis and identify therapeutic targets in ovarian cancer. Taken together, the present study focused on the mutant genes in ctDNA, ascites and tumor tissues, and suggested that the integrated information of different samples could be examined to comprehensively reflect the mutational landscape in ovarian cancer. However, procedures and protocols to interpret and utilize the integrated information obtained from various forms of liquid biopsies will require optimization prior to their use for future clinical applications.
Collapse
Affiliation(s)
- Xiaoxiang Jie
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, P.R. China
| | - Ming Du
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, P.R. China
| | - Meng Zhang
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, P.R. China
| | - Xiayu Jin
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, P.R. China
| | - Qingqing Cai
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, P.R. China
| | - Congjian Xu
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, P.R. China
| | - Xiaoyan Zhang
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, P.R. China
| |
Collapse
|
14
|
Zhou M, Wu T, Yuan Y, Dong SJ, Zhang ZM, Wang Y, Wang J. A risk score system based on a six-microRNA signature predicts the overall survival of patients with ovarian cancer. J Ovarian Res 2022; 15:54. [PMID: 35513874 PMCID: PMC9074233 DOI: 10.1186/s13048-022-00980-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 04/12/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Ovarian cancer (OVC) is a devastating disease worldwide; therefore the identification of prognostic biomarkers is urgently needed. We aimed to determine a robust microRNA signature-based risk score system that could predict the overall survival (OS) of patients with OVC. METHODS We extracted the microRNA expression profiles and corresponding clinical data of 467 OVC patients from The Cancer Genome Atlas (TCGA) database and further divided this data into training, validation and complete cohorts. The key prognostic microRNAs for OVC were identified and evaluated by robust likelihood-based survival analysis (RLSA) and multivariable Cox regression. Time-dependent receiver operating characteristic (ROC) curves were then constructed to evaluate the prognostic performance of these microRNAs. A total of 172 ovarian cancer samples and 162 normal ovarian tissues were used to verify the credibility and accuracy of the selected markers of the TCGA cohort by quantitative real-time polymerase chain reaction (PCR). RESULTS We successfully established a risk score system based on a six-microRNA signature (hsa-miR-3074-5p, hsa-miR-758-3p, hsa-miR-877-5p, hsa-miR-760, hsa-miR-342-5p, and hsa-miR-6509-5p). This microRNA based system is able to characterize patients as either high or low risk. The OS of OVC patients, with either high or low risk, was significantly different when compared in the training cohort (p < 0.001), the validation cohort (p < 0.001) and the complete cohort (p < 0.001). Analysis of clinical samples further demonstrated that these microRNAs were aberrantly expressed in OVC tissues. The six-miRNA-based signature was correlated with the prognosis of OVC patients (p < 0.001). CONCLUSIONS The study established a novel risk score system that is predictive of patient prognosis and is a potentially useful guide for the personalized treatment of OVC patients.
Collapse
Affiliation(s)
- Min Zhou
- Department of Gynecologic Cancer, Shaanxi Provincial Cancer Hospital, No. 309 Yanta West Road, Shaanxi, 710061, Xi'an, People's Republic of China
| | - Tao Wu
- Department of Gynecologic Cancer, Shaanxi Provincial Cancer Hospital, No. 309 Yanta West Road, Shaanxi, 710061, Xi'an, People's Republic of China
| | - Yuan Yuan
- Department of Gynecologic Cancer, Shaanxi Provincial Cancer Hospital, No. 309 Yanta West Road, Shaanxi, 710061, Xi'an, People's Republic of China
| | - Shu-Juan Dong
- Department of Obstetrics and Gynecology, Shaanxi Provincial Rehabilitation Hospital, Xi'an, Shaanxi, People's Republic of China
| | - Zhi-Ming Zhang
- Department of Clinical Laboratory, Xi'an Central Hospital, Xi'an, Shaanxi, People's Republic of China
| | - Yan Wang
- Department of Gynecology, Xi'an Central Hospital, No.161 five West Road, Xi'an, Shaanxi, People's Republic of China.
| | - Jing Wang
- Department of Gynecologic Cancer, Shaanxi Provincial Cancer Hospital, No. 309 Yanta West Road, Shaanxi, 710061, Xi'an, People's Republic of China.
| |
Collapse
|
15
|
CTCFL regulates the PI3K-Akt pathway and it is a target for personalized ovarian cancer therapy. NPJ Syst Biol Appl 2022; 8:5. [PMID: 35132075 PMCID: PMC8821627 DOI: 10.1038/s41540-022-00214-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 01/05/2022] [Indexed: 12/04/2022] Open
Abstract
High-grade serous ovarian carcinoma (HGSC) is the most lethal gynecologic malignancy due to the lack of reliable biomarkers, effective treatment, and chemoresistance. Improving the diagnosis and the development of targeted therapies is still needed. The molecular pathomechanisms driving HGSC progression are not fully understood though crucial for effective diagnosis and identification of novel targeted therapy options. The oncogene CTCFL (BORIS), the paralog of CTCF, is a transcriptional factor highly expressed in ovarian cancer (but in rarely any other tissue in females) with cancer-specific characteristics and therapeutic potential. In this work, we seek to understand the regulatory functions of CTCFL to unravel new target genes with clinical relevance. We used in vitro models to evaluate the transcriptional changes due to the presence of CTCFL, followed by a selection of gene candidates using de novo network enrichment analysis. The resulting mechanistic candidates were further assessed regarding their prognostic potential and druggability. We show that CTCFL-driven genes are involved in cytoplasmic membrane functions; in particular, the PI3K-Akt initiators EGFR1 and VEGFA, as well as ITGB3 and ITGB6 are potential drug targets. Finally, we identified the CTCFL targets ACTBL2, MALT1 and PCDH7 as mechanistic biomarkers to predict survival in HGSC. Finally, we elucidated the value of CTCFL in combination with its targets as a prognostic marker profile for HGSC progression and as putative drug targets.
Collapse
|
16
|
Zanjanchi P, Asghari SM, Mohabatkar H, Shourian M, Shafiee Ardestani M. Conjugation of VEGFR1/R2-targeting peptide with gold nanoparticles to enhance antiangiogenic and antitumoral activity. J Nanobiotechnology 2022; 20:7. [PMID: 34983556 PMCID: PMC8725421 DOI: 10.1186/s12951-021-01198-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 12/09/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Inhibition of tumor angiogenesis through simultaneous targeting of vascular endothelial growth factor receptor (VEGFR)-1 and -2 is highly efficacious. An antagonist peptide of VEGFA/VEGFB, referred to as VGB3, can recognize and neutralize both VEGFR1 and VEGFR2 on the endothelial and tumoral cells, thereby inhibits angiogenesis and tumor growth. However, improved efficacy and extending injection intervals is required for its clinical translation. Given that gold nanoparticles (GNPs) can enhance the efficacy of biotherapeutics, we conjugated VGB3 to GNPs to enhance its efficacy and extends the intervals between treatments without adverse effects. RESULTS GNP-VGB3 bound to VEGFR1 and VEGFR2 in human umbilical vein endothelial (HUVE) and 4T1 mammary carcinoma cells. GNP-VGB3 induced cell cycle arrest, ROS overproduction and apoptosis and inhibited proliferation and migration of endothelial and tumor cells more effectively than unconjugated VGB3 or GNP. In a murine 4T1 mammary carcinoma tumor model, GNP-VGB3 more strongly than VGB3 and GNP inhibited tumor growth and metastasis, and increased animal survival without causing weight loss. The superior antitumor effects were associated with durable targeting of VEGFR1 and VEGFR2, thereby inhibiting signaling pathways of proliferation, migration, differentiation, epithelial-to-mesenchymal transition, and survival in tumor tissues. MicroCT imaging and inductively coupled plasma mass spectrometry showed that GNP-VGB3 specifically target tumors and exhibit greater accumulation within tumors than the free GNPs. CONCLUSION Conjugation to GNPs not only improved the efficacy of VGB3 peptide but also extended the intervals between treatments without adverse effects. These results suggest that GNP-VGB3 is a promising candidate for clinical translation.
Collapse
Affiliation(s)
- Pegah Zanjanchi
- Department of Biotechnology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, 8174673441, Iran
| | - S Mohsen Asghari
- Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, 1417614411, Iran.
| | - Hassan Mohabatkar
- Department of Biotechnology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, 8174673441, Iran.
| | - Mostafa Shourian
- Department of Biology, Faculty of Sciences, University of Guilan, Rasht, 4199613776, Iran
| | - Mehdi Shafiee Ardestani
- Department of Radiopharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
17
|
Liu YK, Jia YJ, Liu SH, Shi HJ, Ma J. Low expression of FXYD5 reverses the cisplatin resistance of epithelial ovarian cancer cells. Histol Histopathol 2021; 36:535-545. [PMID: 33570156 DOI: 10.14670/hh-18-310] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVE To investigate the effect of the downregulation of FXYD domain-containing ion transport regulator 5 (FXYD5) on the cisplatin resistance (CisR) of epithelial ovarian cancer (EOC) cells. METHODS A2780-CisR and SKOV3-CisR cells were obtained through repeated administrations of different cisplatin concentrations, and the half-maximal inhibition concentration (IC50) was calculated by MTT assays. After transfection with FXYD5 siRNA-1 and FXYD5 siRNA-2, the IC50 values of the A2780-CisR and SKOV3-CisR cells were also detected by the MTT method. Cell proliferation, migration, invasion and apoptosis were evaluated through 5-ethynyl-2'-deoxyuridine (EdU) DNA synthesis, wound healing, Transwell invasion and Annexin-V-FITC/PI dual-staining assays, respectively. qRT-PCR and Western blotting were conducted to detect mRNA and protein expression. RESULTS Compared with the sensitive parental cells, the A2780-CisR and SKOV3-CisR cells had increased IC50 and FXYD5 expression. FXYD5 siRNA reduced the IC50 value of cisplatin in the A2780-CisR and SKOV3-CisR cells and decreased the expression of ABCG2 (BCRP) and ABCB1 (MDR1). In addition, FXYD5 inhibition reduced the invasion and migration of the A2780-CisR and SKOV3-CisR cells, with upregulation of E-cadherin and downregulation of Snail and Vimentin. Both FXYD5 siRNA-1 and FXYD5 siRNA-2 inhibited the proliferation and promoted the apoptosis of the A2780-CisR and SKOV3-CisR cells with reduced Ki-67 and increased caspase-3. CONCLUSION FXYD5 downregulation may reduce the invasion, migration and EMT formation of EOC cells to increase their sensitivity to cisplatin chemotherapy by inhibiting cell proliferation and promoting cell apoptosis.
Collapse
Affiliation(s)
- Ya-Kun Liu
- Department of Gynecology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China.
| | - Ya-Jing Jia
- Department of Gynecology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Shi-Hao Liu
- Department of Gynecology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Hong-Jie Shi
- Department of Gynecology, People's Hospital of Tang County, Baoding, Hebei, China
| | - Jing Ma
- Department of Gynecology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
18
|
Hassan MK, Waly AA, Elsayed W, Keshk S, Allam WR, El-Khamisy SF. Integrative microRNA and gene expression analysis identifies new epigenetically regulated microRNAs mediating taxane resistance in ovarian cancer. Sci Rep 2021; 11:562. [PMID: 33436648 PMCID: PMC7804410 DOI: 10.1038/s41598-020-78596-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 11/19/2020] [Indexed: 12/18/2022] Open
Abstract
Taxane is a family of front-line chemotherapeutic agents against ovarian cancer (OC). The therapeutic efficacy is frequently counteracted by the development of chemoresistance, leading to high rates of relapse in OC patients. The role(s) of microRNAs (miRNAs) in cancer chemoresistance had been supported by many evidences Epigenetic regulation by miRNAs has been reported to influence cancer development and response to therapeutics, however, their role in OC resistance to paclitaxel (PTX) is unclear. Here, we conducted miRNA profiling in the responsive and PTX-resistant OC cell lines before and after treatment with epigenetic modulators. We reveal 157 miRNAs to be downregulated in the PTX-resistant cells compared to parental controls. The expression of five miRNAs (miRNA-7-5p, -204-3p, -501-5p, -3652 and -4286) was restored after epigenetic modulation, which was further confirmed by qPCR. In silico analysis of the signaling pathways targeted by the selected miRNAs identified the PI3K-AKT pathway as the primary target. Subsequent cDNA array analysis confirmed multiple PI3K-AKT pathway members such as AKT2, PIK3R3, CDKN1A, CCND2 and FGF2 to be upregulated in PTX-resistant cells. STRING analysis showed the deregulated genes in PTX-resistant cells to be primarily involved in cell cycle progression and survival. Thus, high throughput miRNA and cDNA profiling coupled with pathway analysis and data mining provide evidence for epigenetically regulated miRNAs-induced modulation of signaling pathways in PTX resistant OC cells. It paves the way to more in-depth mechanistic studies and new therapeutic strategies to combat chemoresistance.
Collapse
Affiliation(s)
- Mohamed K Hassan
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, Egypt. .,Bitechnology Program, Zoology Department, Faculty of Science, Port Said University, Port Said, Egypt.
| | - Amr A Waly
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, Egypt
| | - Waheba Elsayed
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, Egypt
| | - Sarah Keshk
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, Egypt.,Bitechnology Program, Zoology Department, Faculty of Science, Port Said University, Port Said, Egypt
| | - Walaa Ramadan Allam
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, Egypt
| | - Sherif F El-Khamisy
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, Egypt. .,Department of Molecular Biology and Biotechnology, Krebs Institute, University of Sheffield, Sheffield, UK. .,Institute of Cancer Therapeutics, University of Bradford, Bradford, BD7 1DP, UK.
| |
Collapse
|
19
|
Arora A, Niño JLG, Myaing MZ, Chia S, Arasi B, Ravasio A, Huang RYJ, Dasgupta R, Biro M, Viasnoff V. Two high-yield complementary methods to sort cell populations by their 2D or 3D migration speed. Mol Biol Cell 2020; 31:2779-2790. [PMID: 33085550 PMCID: PMC7851856 DOI: 10.1091/mbc.e20-07-0466] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 10/07/2020] [Accepted: 10/07/2020] [Indexed: 11/28/2022] Open
Abstract
The potential to migrate is one of the most fundamental functions for various epithelial, mesenchymal, and immune cells. Image analysis of motile cell populations, both primary and cultured, typically reveals an intercellular variability in migration speeds. However, cell migration chromatography, the sorting of large populations of cells based on their migratory characteristics, cannot be easily performed. The lack of such methods has hindered our understanding of the direct correlation between the capacity to migrate and other cellular properties. Here, we report two novel, easily implementable and readily scalable methods to sort millions of live migratory cancer and immune cells based on their spontaneous migration in two-dimensional and three-dimensional microenvironments, respectively. Correlative downstream transcriptomic, molecular and functional tests reveal marked differences between the fast and slow subpopulations in patient-derived cancer cells. We further employ our method to reveal that sorting the most migratory cytotoxic T lymphocytes yields a pool of cells with enhanced cytotoxicity against cancer cells. This phenotypic assay opens new avenues for the precise characterization of the mechanisms underlying hither to unexplained heterogeneities in migratory phenotypes within a cell population, and for the targeted enrichment of the most potent migratory leukocytes in immunotherapies.
Collapse
Affiliation(s)
- Aditya Arora
- Mechanobiology Institute, National University of Singapore, Singapore 117411
| | - Jorge Luis Galeano Niño
- EMBL Australia, Single Molecule Science Node, School of Medical Sciences, University of New South Wales 2052, Sydney, Australia
| | - Myint Zu Myaing
- Mechanobiology Institute, National University of Singapore, Singapore 117411
| | - Shumei Chia
- Laboratory of Precision Oncology and Cancer Evolution, Genome Institute of Singapore/A*STAR, Singapore 138632
| | - Bakya Arasi
- Mechanobiology Institute, National University of Singapore, Singapore 117411
| | - Andrea Ravasio
- Mechanobiology Institute, National University of Singapore, Singapore 117411
- Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Ruby Yun-Ju Huang
- Cancer Science Institute of Singapore, National University of Singapore, Centre for Translational Medicine, Singapore 117599
| | - Ramanuj Dasgupta
- Laboratory of Precision Oncology and Cancer Evolution, Genome Institute of Singapore/A*STAR, Singapore 138632
| | - Maté Biro
- Laboratory of Precision Oncology and Cancer Evolution, Genome Institute of Singapore/A*STAR, Singapore 138632
| | - Virgile Viasnoff
- Mechanobiology Institute, National University of Singapore, Singapore 117411
- Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
20
|
Mucinous carcinoma with micropapillary features is morphologically, clinically and genetically distinct from pure mucinous carcinoma of breast. Mod Pathol 2020; 33:1945-1960. [PMID: 32358590 DOI: 10.1038/s41379-020-0554-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 04/05/2020] [Accepted: 04/05/2020] [Indexed: 12/14/2022]
Abstract
Micropapillary features are seen in pure mucinous carcinoma of breast (PMC), which is termed mucinous carcinoma with micropapillary features (MPMC). However, whether MPMC can be identified as a morphologically, clinically or genetically distinct entity from PMC remains controversial. In this study, a retrospective review of 161 cases of breast mucinous carcinoma was conducted to assess the clinicopathologic features, prognostic implications, and genomic alterations of MPMC and PMC. MPMCs were identified in 32% of mucinous carcinomas showing an excellent interobserver agreement (ICC = 0.922). MPMCs occurred at a younger age and exhibited higher nuclear grade, more frequent lymph nodal metastasis, lymphovascular invasion, and HER2 amplification compared with PMCs. Survival analyses revealed that MPMCs show decreased progression-free survival compared with PMCs in both unmatched and matched cohorts. A similar outcome of distant disease-free survival was observed only in the unmatched cohort. However, no statistical difference in recurrence score was observed between MPMC and PMC using a 21-gene assay. Notably, both MPMCs and PMCs displayed low mutation burden, common mutations affecting TTN, GATA3, SF3B1, TP53, recurrent 6q14.1-q27 losses, and 8p11.21-q24.3 gains. GATA3, TP53, and SF3B1 were recurrently mutated in MPMCs, while PIK3CA mutations were exclusively detected in PMCs. Moreover, MPMCs harbored 17q and 20q gains as well as 17p losses, while PMCs displayed gains at 6p. PI3K-Akt, mTOR, ErbB, and focal adhesion pathways were more frequently deregulated in MPMCs than in PMCs, which may responsible for the aggressive tumor behavior of MPMCs. Our findings suggest that MPMC is morphologically, clinically, and genetically distinct from PMC.
Collapse
|
21
|
Dou Z, Gao L, Ren W, Zhang H, Wang X, Li S, Zheng J, Kong X, Chi P, Zhi K. CiRS-7 functions as a ceRNA of RAF-1/PIK3CD to promote metastatic progression of oral squamous cell carcinoma via MAPK/AKT signaling pathways. Exp Cell Res 2020; 396:112290. [PMID: 32956704 DOI: 10.1016/j.yexcr.2020.112290] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Revised: 09/07/2020] [Accepted: 09/15/2020] [Indexed: 12/17/2022]
Abstract
PURPOSE Recent findings have shown that circRNA dysregulation was involved in the development of many types of cancer. However, our knowledge of circRNA in oral squamous cell carcinoma (OSCC) remains elusive. METHODS Here, we explored whether ciRS-7 could function as a ceRNA in promoting metastasis of OSCC via regulating miR-7 activity. The expression levels of ciRS-7 and miR-7 were examined in clinical samples and cell lines by qRT-PCR, and the effects of ectopic expression of ciRS-7 and miR-7 on cell proliferation, migration and invasion were assessed in vitro and in vivo. The effects of ciRS-7 on miR-7 activity were investigated by means of luciferase reporter assay, qRT-PCR and Western blot. In addition, the effects of miR-7 mediated ciRS-7 on the levels of MAPK/AKT signaling proteins were evaluated by Western blot. RESULTS We found that ciRS-7 was highly expressed in OSCC tissues and cell lines compared with normal counterparts. Ectopic expression of ciRS-7 significantly promoted OSCC cell proliferation, migration and invasion through in vitro and in vivo. Based on bioinformatics analysis, qRT-PCR, Western blot and luciferase reporter assays, we determined that ciRS-7 functioned as a sponge for miR-7, resulting in attenuation of miR-7 targets RAF-1 and PIK3CD, which are core components of the MAPK/AKT signaling pathways. Moreover, miR-7 correlated with perineural and lymphovascular invasion in OSCC patients. Further experiments demonstrated that ciRS-7 overexpression could attenuate the anti-tumor effects of miR-7 on OSCC cells. CONCLUSIONS Our results suggested that ciRS-7 can interact directly with miR-7, resulting in upregulation of RAF-1/PIK3CD expression and enhancing metastatic progression of OSCC.
Collapse
Affiliation(s)
- Zhichao Dou
- Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266555, Shandong, PR China; School of Stomatology, Qingdao University, Qingdao, 266000, Shandong, PR China
| | - Ling Gao
- Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266555, Shandong, PR China
| | - Wenhao Ren
- Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266555, Shandong, PR China
| | - Hao Zhang
- Department of Stomatology, People's Hospital, Hubei University of Medicine, Shiyan, 430071, Hubei, PR China
| | - Xiaofei Wang
- Experiment Center of Biomedical Research, Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi, 710049, PR China
| | - Shaoming Li
- Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266555, Shandong, PR China
| | - Jingjing Zheng
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, 266555, Shandong, PR China
| | - Xinjuan Kong
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, PR China
| | - Peng Chi
- Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266555, Shandong, PR China; School of Stomatology, Qingdao University, Qingdao, 266000, Shandong, PR China
| | - Keqian Zhi
- Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266555, Shandong, PR China.
| |
Collapse
|
22
|
Bi X, Lv X, Liu D, Guo H, Yao G, Wang L, Liang X, Yang Y. METTL3-mediated maturation of miR-126-5p promotes ovarian cancer progression via PTEN-mediated PI3K/Akt/mTOR pathway. Cancer Gene Ther 2020; 28:335-349. [PMID: 32939058 DOI: 10.1038/s41417-020-00222-3] [Citation(s) in RCA: 114] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 08/05/2020] [Accepted: 08/21/2020] [Indexed: 02/03/2023]
Abstract
Methyltransferase-like 3 (METTL3) functions as an RNA methyltransferase that controls the modification of N(6)-methyladenosine (m6A) to influence the biosynthesis, decay, and translation of mRNAs. This study aims to investigate the regulation of METTL3-mediated promotion of microRNA-126-5p (miR-126-5p) in the progression of ovarian cancer and to identify the mechanisms in relation to phosphatase and tensin homolog (PTEN) and the PI3K/Akt/mTOR pathway. We found high expression of miR-126-5p in ovarian cancer samples compared to paired adjacent samples, and also in ovarian cancer cell lines. Gain-of-function experiments demonstrated that overexpression of miR-126-5p promoted ovarian cancer cell proliferation, migration, and invasion, and inhibited their apoptosis. Luciferase reporter assay identified that miR-126-5p could directly bind to PTEN. By targeting PTEN, miR-126-5p could activate the PI3K/Akt/mTOR pathway. Furthermore, the RNA methyltransferase METTL3 promoted the maturation of miR-126-5p via the m6A modification of pri-miR-126-5p. Finally, in vitro and in vivo experiments substantiated that silencing of METTL3 impeded the progression and tumorigenesis of ovarian cancer by impairing the miR-126-5p-targeted inhibition of PTEN and thus blocking the PI3K/Akt/mTOR pathway. Coherently, knockdown of METTL3 inhibited the effect of miR-126-5p to upregulate PTEN, and thus prevents PI3K/Akt/mTOR pathway activation, thereby suppressing the development of ovarian cancer. These findings highlight potential targets for the future ovarian cancer treatment as well as tumorigenic mechanisms mediated by m6A modification.
Collapse
Affiliation(s)
- Xuehan Bi
- Department of Obstetrics and Gynecology, the First Hospital of Lanzhou University, Lanzhou, 730000, People's Republic of China.,Department of Obstetrics and Gynecology, the First Hospital of Lanzhou University, Key Laboratory of Gynecologic Oncology Gansu Province, Lanzhou, 730000, People's Republic of China
| | - Xiao Lv
- Department of Obstetrics and Gynecology, the First Hospital of Lanzhou University, Lanzhou, 730000, People's Republic of China.,Department of Obstetrics and Gynecology, the First Hospital of Lanzhou University, Key Laboratory of Gynecologic Oncology Gansu Province, Lanzhou, 730000, People's Republic of China
| | - Dajiang Liu
- Department of Obstetrics and Gynecology, the First Hospital of Lanzhou University, Lanzhou, 730000, People's Republic of China.,Department of Obstetrics and Gynecology, the First Hospital of Lanzhou University, Key Laboratory of Gynecologic Oncology Gansu Province, Lanzhou, 730000, People's Republic of China
| | - Hongtao Guo
- Department of Obstetrics and Gynecology, the First Hospital of Lanzhou University, Lanzhou, 730000, People's Republic of China.,Department of Obstetrics and Gynecology, the First Hospital of Lanzhou University, Key Laboratory of Gynecologic Oncology Gansu Province, Lanzhou, 730000, People's Republic of China
| | - Guang Yao
- Department of Obstetrics and Gynecology, the First Hospital of Lanzhou University, Lanzhou, 730000, People's Republic of China.,Department of Obstetrics and Gynecology, the First Hospital of Lanzhou University, Key Laboratory of Gynecologic Oncology Gansu Province, Lanzhou, 730000, People's Republic of China
| | - Lijuan Wang
- Department of Obstetrics and Gynecology, the First Hospital of Lanzhou University, Lanzhou, 730000, People's Republic of China.,Department of Obstetrics and Gynecology, the First Hospital of Lanzhou University, Key Laboratory of Gynecologic Oncology Gansu Province, Lanzhou, 730000, People's Republic of China
| | - Xiaolei Liang
- Department of Obstetrics and Gynecology, the First Hospital of Lanzhou University, Lanzhou, 730000, People's Republic of China.,Department of Obstetrics and Gynecology, the First Hospital of Lanzhou University, Key Laboratory of Gynecologic Oncology Gansu Province, Lanzhou, 730000, People's Republic of China
| | - Yongxiu Yang
- Department of Obstetrics and Gynecology, the First Hospital of Lanzhou University, Lanzhou, 730000, People's Republic of China. .,Department of Obstetrics and Gynecology, the First Hospital of Lanzhou University, Key Laboratory of Gynecologic Oncology Gansu Province, Lanzhou, 730000, People's Republic of China.
| |
Collapse
|
23
|
Huang ZX, Mao XM, Wu RF, Huang SM, Ding XY, Chen QH, Chen QX. RhoA/ROCK pathway mediates the effect of oestrogen on regulating epithelial-mesenchymal transition and proliferation in endometriosis. J Cell Mol Med 2020; 24:10693-10704. [PMID: 32725958 PMCID: PMC7521234 DOI: 10.1111/jcmm.15689] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 06/19/2020] [Accepted: 07/10/2020] [Indexed: 12/20/2022] Open
Abstract
Endometriosis is a benign gynaecological disease appearing with pelvic pain, rising dysmenorrhoea and infertility seriously impacting on 10% of reproductive‐age females. This research attempts to demonstrate the function and molecular mechanism of RhoA/ROCK pathway on epithelial‐mesenchymal transition (EMT) and proliferation in endometriosis. The expression of Rho family was abnormally changed in endometriotic lesions; in particular, RhoA and ROCK1/2 were significantly elevated. Overexpression of RhoA in human eutopic endometrial epithelial cells (eutopic EECs) enhanced the cell mobility, epithelial‐mesenchymal transition (EMT) and proliferation, and RhoA knockdown exhibited the opposite function. Oestrogen up‐regulated the RhoA activity and expression of RhoA and ROCK1/2. RhoA overexpression reinforced the effect of oestrogen on promoting EMT and proliferation, and RhoA knockdown impaired the effect of oestrogen. oestrogen receptor α (ERα) was involved with the regulation of oestrogen on EMT and proliferation and up‐regulated RhoA activity and expression of RhoA and ROCK1/2. The function of ERα was modulated by the change in RhoA expression. Furthermore, phosphorylated ERK that was enhanced by oestrogen and ERα promoted the protein expression of RhoA/ROCK pathway. Endometriosis mouse model revealed that oestrogen enhanced the size and weight of endometriotic lesions. The expression of RhoA and phosphorylated ERK in mouse endometriotic lesions was significantly elevated by oestrogen. We conclude that abnormal activated RhoA/ROCK pathway in endometriosis is responsible for the function of oestrogen/ERα/ERK signalling, which promoted EMT and proliferation and resulted in the development of endometriosis.
Collapse
Affiliation(s)
| | - Xiao-Mei Mao
- School of Life Sciences, Xiamen University, Xiamen, China
| | - Rong-Feng Wu
- Reproductive Medical Center, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Shao-Min Huang
- Reproductive Medical Center, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Xin-Yu Ding
- The Key Laboratory of Research and Diagnosis of Gynecological Diseases of Xiamen City and Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Qiong-Hua Chen
- The Key Laboratory of Research and Diagnosis of Gynecological Diseases of Xiamen City and Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Qing-Xi Chen
- School of Life Sciences, Xiamen University, Xiamen, China
| |
Collapse
|
24
|
Jiao R, Sun S, Gao X, Cui R, Cao G, Wei H, Wang S, Zhang Z, Bai H. A Polyethylene Glycol-Based Method for Enrichment of Extracellular Vesicles from Culture Supernatant of Human Ovarian Cancer Cell Line A2780 and Body Fluids of High-Grade Serous Carcinoma Patients. Cancer Manag Res 2020; 12:6291-6301. [PMID: 32801874 PMCID: PMC7386806 DOI: 10.2147/cmar.s228288] [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: 08/26/2019] [Accepted: 07/03/2020] [Indexed: 12/21/2022] Open
Abstract
Objective This study tried to evaluate whether 8% polyethylene glycol (PEG) 6000 precipitation combined with differential ultracentrifugation (PPDU) was an efficient and practical method for the enrichment and purification of extracellular vesicles (EVs) derived from the culture supernatant of human ovarian cancer cell line A2780 and from body fluids of patients with high-grade serous carcinoma (HGSC). Methods PPDU was used to enrich and purify the EVs derived from body fluids of patients with HSGC and cell culture supernatant of subclones of human ovarian cancer cell line A2780 with high/low invasive capacity (named as A-H/A-L, respectively). Transmission electron microscope (TEM) and nanoparticle tracking analysis (NTA) were used to identificate the EVs size and distribution. Western blots (WB) were used to detect the expression of CD9, CD63, Alix and Calnexin. The high-purity EVs derived from the cell culture supernatant of A-H/A-L were detected by the protein profile. Expression of integrins (ITGs) αV, β1 and β3 in the EVs derived from body fluids of HGSC patients was also evaluated. Results The diameter of EVs was about 30–260 nm observed under the TEM. Under the NTA identification, the peak size of EVs was ranged from 70 to 159nm. EVs derived from different specimens did not significantly differ in mean size and peak size. Presence of CD9, CD63 and Alix and absence of Calnexin were confirmed in the EVs. The protein concentrations of EVs’ sample extracted from A-H/A-L cell culture supernatant were 0.36µg/µL and 0.20µg/µL, respectively. The total amount of protein obtained from 300ul EVs was 108.02ug and 61.44ug, respectively. Totally, 2397 peptides and 952 proteins were identified by isobaric tags for relative and absolute quantitation (ITRAQ). The expression of ITGαV, β1, and β3 in the EVs from plasma and ascites of HGSC patients was significantly higher than the control group (plasma: all P<0.0001; ascites: P=0.036, 0.001 and 0.004, respectively). The expression level of ITGαV and β1 in EVs of HGSC’s ascites was significantly higher than that in plasma (P= 0.004, 0.001, respectively). The expression of ITGβ3 was also slightly elevated in EVs-derived HGSC patients’ ascites (P=0.492). Conclusion PPDU was an efficient and practical method to enrich EVs from body fluids and cell culture supernatant. The characteristic expression of ITGαV, β1 and β3 in ascites and plasma EVs of patients with HGSC provided useful information on the development of EVs in HGSC.
Collapse
Affiliation(s)
- Ruili Jiao
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, People's Republic of China.,Department of Obstetrics and Gynecology, Maternal and Child Health Hospital, Beijing, People's Republic of China
| | - Shipeng Sun
- Clinical Laboratories, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, People's Republic of China
| | - Xiaoqiong Gao
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, People's Republic of China
| | - Ran Cui
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, People's Republic of China
| | - Guangming Cao
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, People's Republic of China
| | - Huali Wei
- Department of Obstetrics and Gynecology, Emergency General Hospital, Beijing, People's Republic of China
| | - Shuzhen Wang
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, People's Republic of China
| | - Zhenyu Zhang
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, People's Republic of China
| | - Huimin Bai
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, People's Republic of China
| |
Collapse
|
25
|
Cai Z, He S, Li T, Zhao L, Zhang K. Plumbagin inhibits proliferation and promotes apoptosis of ovarian granulosa cells in polycystic ovary syndrome by inactivating PI3K/Akt/mTOR pathway. Anim Cells Syst (Seoul) 2020; 24:197-204. [PMID: 33029296 PMCID: PMC7473319 DOI: 10.1080/19768354.2020.1790416] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is recognized as a general endocrine disease and reproductive disorder. Although evidence indicates that PCOS has a complex etiology and genetic basis, the pathogenic mechanisms and signal pathway in PCOS remain unclear. In this study, the normal structure of follicle and corpus luteum were observed, and no cyst nor hyperemia was observed under the light microscopic study with hematoxylin and eosin (H&E) staining. Eestosterone and progesterone were evaluated by radioimmunoassay in rat serum. The alterations of proliferative ability and cell cycle distribution of each group were assessed by Cell Counting Kit-8 (CCK8) assay and flow cytometry. The protein expression of p-mTOR/mTOR, p-PI3K/PI3K, p-AKT/AKT, and GAPDH were analyzed by western blotting. Both doses of PLB could benefit the ovarian morphology and polycystic property. PLBinduced a suppress effect on the proliferation of rat ovarian granulosa cells. In addition, PLB also induced concentration-dependent apoptosis in rat ovarian granulosa cells. The rat ovarian granulosa cells treated with PLB that the expression levels of p-AKT, p-mTOR, and p-PI3K were significantly decreased in a concentration-dependent manner. PLB not only plays a critical role in attenuating the pathology and polycystic property changes in the ovary but can also induce rat ovarian granulosa cell apoptosis through the PI3K/Akt/mTOR signal pathway. This study showed the innovative role of PLB in the pathogenesis of PCOS and provides a new therapeutic modality for the treatment of PCOS.
Collapse
Affiliation(s)
- Zhaowei Cai
- Reproductive Center, SSL Central Hospital of Dongguan City, Dongguan City, People's Republic of China
| | - Shaojuan He
- Department of Clinical Laboratory, Dongguan People's Hospital, Dongguan City, People's Republic of China
| | - Tao Li
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan City, People's Republic of China
| | - Li Zhao
- Reproductive Center, SSL Central Hospital of Dongguan City, Dongguan City, People's Republic of China
| | - Kerong Zhang
- Department of Gynaecology and Obstetrics and Reproductive Medicine, Second Clinical Medical College of Guangdong Medical, Guangdong Medical University, Dongguan City, People's Republic of China
| |
Collapse
|
26
|
Zhang X, Ishibashi M, Kitatani K, Shigeta S, Tokunaga H, Toyoshima M, Shimada M, Yaegashi N. Potential of Tyrosine Kinase Receptor TIE-1 as Novel Therapeutic Target in High-PI3K-Expressing Ovarian Cancer. Cancers (Basel) 2020; 12:cancers12061705. [PMID: 32604863 PMCID: PMC7352248 DOI: 10.3390/cancers12061705] [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: 05/12/2020] [Revised: 06/17/2020] [Accepted: 06/23/2020] [Indexed: 11/16/2022] Open
Abstract
Tyrosine kinase receptor TIE-1 plays a critical role in angiogenesis and blood-vessel stability. In recent years, increased TIE-1 expression has been observed in many types of cancers; however, the biological significance and underlying mechanisms remain unknown. Thus, in the present study, we investigated the tumor biological functions of TIE-1 in ovarian cancer. The treatment of SKOV3 ovarian-cancer cells with siRNA against TIE-1 decreased the expression of key molecules in the PI3K/Akt signaling pathway, such as p110α and phospho-Akt, suggesting that TIE-1 is related to the PI3K/Akt pathway. Furthermore, the knockdown of TIE-1 significantly decreased cell proliferation in high-PI3K-expressing cell lines (SKOV3, CAOV3) but not low-PI3K-expressing cell lines (TOV112D, A2780). These results suggested that inhibition of TIE-1 decreases cell growth in high-PI3K-expressing cells. Moreover, in low-PI3K-expressing TOV112D ovarian-cancer cells, TIE-1 overexpression induced PI3K upregulation and promoted a PI3K-mediated cell proliferative phenotype. Mechanistically, TIE-1 participates in cell growth and proliferation by regulating the PI3K/Akt signaling pathway. Taken together, our findings strongly implicate TIE-1 as a novel therapeutic target in high-PI3K-expressing ovarian-cancer cells.
Collapse
Affiliation(s)
- Xuewei Zhang
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Tohoku University, Sendai 980-8577, Japan; (X.Z.); (S.S.); (H.T.); (M.S.); (N.Y.)
| | - Masumi Ishibashi
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Tohoku University, Sendai 980-8577, Japan; (X.Z.); (S.S.); (H.T.); (M.S.); (N.Y.)
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 119077, Singapore
- Correspondence: ; Tel.: +81-022-717-7251; Fax: +81-022-717-7258
| | - Kazuyuki Kitatani
- Laboratory of Immunopharmacology, Faculty of Pharmaceutical Sciences, Setsunan University, Hirakata, Osaka 572-8508, Japan;
| | - Shogo Shigeta
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Tohoku University, Sendai 980-8577, Japan; (X.Z.); (S.S.); (H.T.); (M.S.); (N.Y.)
| | - Hideki Tokunaga
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Tohoku University, Sendai 980-8577, Japan; (X.Z.); (S.S.); (H.T.); (M.S.); (N.Y.)
| | - Masafumi Toyoshima
- Department of Obstetrics and Gynecology, Japanese Red Cross Ishinomaki Hospital, Ishinomaki 986-8522, Japan;
| | - Muneaki Shimada
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Tohoku University, Sendai 980-8577, Japan; (X.Z.); (S.S.); (H.T.); (M.S.); (N.Y.)
| | - Nobuo Yaegashi
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Tohoku University, Sendai 980-8577, Japan; (X.Z.); (S.S.); (H.T.); (M.S.); (N.Y.)
| |
Collapse
|
27
|
SNX-2112, an Hsp90 inhibitor, suppresses cervical cancer cells proliferation, migration, and invasion by inhibiting the Akt/mTOR signaling pathway. Med Chem Res 2020. [DOI: 10.1007/s00044-020-02534-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
|
28
|
Ouyang Q, Hu S, Wang G, Hu J, Zhang J, Li L, Hu B, He H, Liu H, Xia L, Wang J. Comparative Transcriptome Analysis Suggests Key Roles for 5-Hydroxytryptamlne Receptors in Control of Goose Egg Production. Genes (Basel) 2020; 11:E455. [PMID: 32331314 PMCID: PMC7230923 DOI: 10.3390/genes11040455] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 04/16/2020] [Accepted: 04/17/2020] [Indexed: 02/06/2023] Open
Abstract
To date, research on poultry egg production performance has only been conducted within inter or intra-breed groups, while those combining both inter- and intra-breed groups are lacking. Egg production performance is known to differ markedly between Sichuan white goose (Anser cygnoides) and Landes goose (Anser anser). In order to understand the mechanism of egg production performance in geese, we undertook this study. Here, 18 ovarian stromal samples from both Sichuan white goose and Landes goose at the age of 145 days (3 individuals before egg production initiation for each breed) and 730 days (3 high- and low egg production individuals during non-laying periods for each breed) were collected to reveal the genome-wide expression profiles of ovarian mRNAs and lncRNAs between these two geese breeds at different physiological stages. Briefly, 58, 347, 797, 777, and 881 differentially expressed genes (DEGs) and 56, 24, 154, 105, and 224 differentially expressed long non-coding RNAs (DElncRNAs) were found in LLD vs. HLD (low egg production Landes goose vs. high egg production Landes goose), LSC vs. HSC (low egg production Sichuan White goose vs. high egg production Sichuan white goose), YLD vs. YSC (young Landes goose vs. young Sichuan white goose), HLD vs. HSC (high egg production Landes goose vs. high egg production Sichuan white goose), and LLD vs. LSC (low egg production Landes goose vs. low egg production Sichuan white goose) groups, respectively. Functional enrichment analysis of these DEGs and DElncRNAs suggest that the "neuroactive ligand-receptor interaction pathway" is crucial for egg production, and particularly, members of the 5-hydroxytryptamine receptor (HTR) family affect egg production by regulating ovarian metabolic function. Furthermore, the big differences in the secondary structures among HTR1F and HTR1B, HTR2B, and HTR7 may lead to their different expression patterns in goose ovaries of both inter- and intra-breed groups. These results provide novel insights into the mechanisms regulating poultry egg production performance.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Jiwen Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; (Q.O.); (S.H.); (G.W.); (J.H.); (J.Z.); (L.L.); (B.H.); (H.H.); (H.L.); (L.X.)
| |
Collapse
|
29
|
Yang Y, Song Z, Wu W, Xu A, Lv S, Ji S. ZnO Quantum Dots Induced Oxidative Stress and Apoptosis in HeLa and HEK-293T Cell Lines. Front Pharmacol 2020; 11:131. [PMID: 32180717 PMCID: PMC7057716 DOI: 10.3389/fphar.2020.00131] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Accepted: 01/30/2020] [Indexed: 12/17/2022] Open
Abstract
Zinc oxide (ZnO) quantum dot (QD) is a promising inexpensive inorganic nanomaterials, of which potential toxic effects on biological systems and human health should be evaluated before biomedical application. In this study, the cytotoxicity of ZnO QDs was assessed using HeLa cervical cancer cell and HEK-293T human embryonic kidney cell lines. Cell viability was significantly decreased by treatment with 50 µg/ml ZnO QDs after only 6 h, and the cytotoxicity of ZnO QDs was higher in HEK-293T than in HeLa cells. ZnO QDs increased the level of reactive oxygen species and decreased the mitochondria membrane potential in a dose-dependent manner. Several gene expression involved in apoptosis was regulated by ZnO QDs, including bcl-2 gene and caspase. In HeLa cells, ZnO QDs significantly increased early and late apoptosis, but only late apoptosis was affected in HEK-293T cells. These findings will be helpful for future research and application of ZnO QDs in biomedicine.
Collapse
Affiliation(s)
- Yanjie Yang
- Provincial Engineering Centre for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Zhenhua Song
- Provincial Engineering Centre for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Weixia Wu
- Provincial Engineering Centre for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Ao Xu
- Provincial Engineering Centre for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Shuangyu Lv
- Provincial Engineering Centre for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Shaoping Ji
- Provincial Engineering Centre for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng, China
| |
Collapse
|
30
|
Liu G, Ruan G, Huang M, Chen L, Sun P. Genome-wide DNA copy number profiling and bioinformatics analysis of ovarian cancer reveals key genes and pathways associated with distinct invasive/migratory capabilities. Aging (Albany NY) 2020; 12:178-192. [PMID: 31895688 PMCID: PMC6977652 DOI: 10.18632/aging.102608] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Accepted: 12/05/2019] [Indexed: 02/07/2023]
Abstract
Ovarian cancer (OC) metastasis presents major hurdles that must be overcome to improve patient outcomes. Recent studies have demonstrated copy number variations (CNVs) frequently contribute to alterations in oncogenic drivers. The present study used a CytoScan HD Array to analyse CNVs and loss of heterozygosity (LOH) in the entire genomes of 6 OC patients and human OC cell lines to determine the genetic target events leading to the distinct invasive/migratory capacities of OC. The results showed that LOH at Xq11.1 and Xp21.1 and gains at 8q21.13 were novel, specific CNVs. Ovarian cancer-related CNVs were then screened by bioinformatics analysis. In addition, transcription factors-target gene interactions were predicted with information from PASTAA analysis. As a result, six genes (i.e., GAB2, AKT1, EGFR, COL6A3, UGT1A1 and UGT1A8) were identified as strong candidates by integrating the above data with gene expression and clinical outcome data. In the transcriptional regulatory network, 4 known cancer-related transcription factors (TFs) interacted with 6 CNV-driven genes. The protein/DNA arrays revealed 3 of these 4 TFs as potential candidate gene-related transcription factors in OC. We then demonstrated that these six genes can serve as potential biomarkers for OC. Further studies are required to elucidate the pathogenesis of OC.
Collapse
Affiliation(s)
- GuiFen Liu
- Laboratory of Gynaecologic Oncology, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou 350001, Fujian Province, China
| | - GuanYu Ruan
- Laboratory of Gynaecologic Oncology, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou 350001, Fujian Province, China
| | - MeiMei Huang
- Laboratory of Gynaecologic Oncology, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou 350001, Fujian Province, China
| | - LiLi Chen
- Laboratory of Gynaecologic Oncology, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou 350001, Fujian Province, China
| | - PengMing Sun
- Laboratory of Gynaecologic Oncology, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou 350001, Fujian Province, China.,Department of Gynaecology, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou 350001, Fujian Province, China
| |
Collapse
|
31
|
Badi RM, Khaleel EF, El-Bidawy MH, Satti HH, Mostafa DG. Exendin-4 Induces Cytotoxic Autophagy in Two Ovarian Cancer Cell Lines through Inhibition of Mtorc1 Mediated by Activation of AMPK and Suppression of Akt. Folia Biol (Praha) 2020; 66:186-203. [PMID: 34087975 DOI: 10.14712/fb2020066050186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Activation of autophagy suppresses ovarian cancer (OC). This in vitro study investigated whether the anti-tumour effect of exendin-4 against OC involves modulation of autophagy and figured out the possible mechanisms of action. SKOV-3 and OVCAR-3 cells (1 × 105/ml) were cultured in DMEM medium and treated with exendin-4 in the presence or absence of chloroquine (CQ), an autophagy inhibitor. In some cases, cells were also treated with exendin- 4 with or without pre-treatment with compound C (CC), an AMPK inhibitor, or insulin-like growth factor (IGF-1), a PI3K/Akt activator. Exendin-4 increased expression of beclin-1 and LC3I/II, suppressed expression of p62, reduced cell survival, migration, and invasion, and increased cell apoptosis and LDH release in both SKOV-3 and OVCAR-3 cells. Besides, exendin-4 reduced phosphorylation of mTORC1, 6SK, 4E-BP1, and Akt but increased phosphorylation of AMPK in both cell lines. These effects were associated with down-regulation of Bcl-2, suppression of nuclear phosphorylation of NF-κB p65, and increased expression of Bax and cleaved caspases 3/8. Chloroquine completely prevented the inhibitory effects of exendin-4 on the cell survival, Bcl-2, NF-κB, and cell invasiveness and abolished its stimulation of cell apoptosis and LDH release. Moreover, only the combined treatment with IGF-1 and CC completely abolished the observed effect of exendin-4 on the expression of beclin-1, LC3I/II, p62, as well as on cell survival, apoptosis, and LDH release. Exendin-4 exhibits a potent anti-tumour cytotoxic effect in SKOV-3 and OVCAR-3 cells by activating the markers of autophagy, mediated by activation of AMPK and inhibition of Akt.
Collapse
Affiliation(s)
- R M Badi
- Department of Physiology, College of Medicine, King Khalid University, Abha, Saudi Arabia
- Department of Physiology, Faculty of Medicine, University of Khartoum, Khartoum, Sudan
| | - E F Khaleel
- Department of Physiology, College of Medicine, King Khalid University, Abha, Saudi Arabia
- Department of Medical Physiology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - M H El-Bidawy
- Department of Medical Physiology, Faculty of Medicine, Cairo University, Cairo, Egypt
- Department of BMS, Division of Physiology, College of Medicine, Prince Sattam Ibn Abdulaziz University, Al-Kharj, Saudi Arabia
| | - H H Satti
- Department of Pathology, College of Medicine, King Khalid University, Abha, Saudi Arabia
- Department of Pathology, Faculty of Medicine, University of Khartoum, Khartoum, Sudan
| | - D G Mostafa
- Department of Physiology, College of Medicine, King Khalid University, Abha, Saudi Arabia
- Department of Medical Physiology, Faculty of Medicine, Assiut University, Assiut, Egypt
| |
Collapse
|
32
|
Ghoneum A, Abdulfattah AY, Said N. Targeting the PI3K/AKT/mTOR/NFκB Axis in Ovarian Cancer. JOURNAL OF CELLULAR IMMUNOLOGY 2020; 2:68-73. [PMID: 32395722 PMCID: PMC7213295 DOI: 10.33696/immunology.1.022] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Ovarian cancer stands as the most lethal gynecologic malignancy and remains the fifth most common gynecologic cancer. Poor prognosis and low five-year survival rate are attributed to nonspecific symptoms at early phases along with a lack of effective treatment at advanced stages. It is thus paramount, that ovarian carcinoma be viewed through several lenses in order to gain a thorough comprehension of its molecular pathogenesis, epidemiology, histological subtypes, hereditary factors, diagnostic approaches, and methods of treatment. Above all, it is crucial to dissect the role that the unique peritoneal tumor microenvironment plays in ovarian cancer progression and metastasis. This short communication seeks to underscore several important aspects of the PI3K/AKT/mTOR/NFκB pathway in the context of ovarian cancer and discuss recent advances in targeting this pathway.
Collapse
Affiliation(s)
- Alia Ghoneum
- Department of Cancer Biology, Wake Forest University School of Medicine, and Comprehensive Cancer Center, Winston Salem, NC 27157, USA
| | - Ammar Yasser Abdulfattah
- Department of Cancer Biology, Wake Forest University School of Medicine, and Comprehensive Cancer Center, Winston Salem, NC 27157, USA
| | - Neveen Said
- Department of Cancer Biology, Wake Forest University School of Medicine, and Comprehensive Cancer Center, Winston Salem, NC 27157, USA
- Department of Pathology, Wake Forest University School of Medicine, and Comprehensive Cancer Center, Winston Salem, NC 27157, USA
- Department of Urology, Wake Forest University School of Medicine, and Comprehensive Cancer Center, Winston Salem, NC 27157, USA
- Wake Forest Baptist Health Sciences, Winston Salem, NC 27157, USA
| |
Collapse
|
33
|
Moioli M, Barra F, Maramai M, Valenzano Menada M, Vellone VG, Costantini S, Ferrero S. Mucinous ovarian cancer: current therapeutic targets, preclinical progress, and experimental drugs. Expert Opin Investig Drugs 2019; 28:1025-1029. [DOI: 10.1080/13543784.2019.1693999] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Melita Moioli
- Academic Unit of Obstetrics and Gynecology, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DiNOGMI), University of Genoa, Genoa, Italy
| | - Fabio Barra
- Academic Unit of Obstetrics and Gynecology, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DiNOGMI), University of Genoa, Genoa, Italy
| | - Mattia Maramai
- Academic Unit of Obstetrics and Gynecology, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DiNOGMI), University of Genoa, Genoa, Italy
| | - Mario Valenzano Menada
- Academic Unit of Obstetrics and Gynecology, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DiNOGMI), University of Genoa, Genoa, Italy
| | - Valerio Gaetano Vellone
- Department of Surgical and Diagnostic Sciences, Ospedale Policlinico San Martino, University of Genoa, Genoa, Italy
| | - Sergio Costantini
- Academic Unit of Obstetrics and Gynecology, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DiNOGMI), University of Genoa, Genoa, Italy
| | - Simone Ferrero
- Academic Unit of Obstetrics and Gynecology, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DiNOGMI), University of Genoa, Genoa, Italy
| |
Collapse
|
34
|
Zhang X, Qin T, Zhu Z, Hong F, Xu Y, Zhang X, Xu X, Ma A. Ivermectin Augments the In Vitro and In Vivo Efficacy of Cisplatin in Epithelial Ovarian Cancer by Suppressing Akt/mTOR Signaling. Am J Med Sci 2019; 359:123-129. [PMID: 32039764 DOI: 10.1016/j.amjms.2019.11.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 10/26/2019] [Accepted: 11/04/2019] [Indexed: 10/25/2022]
Abstract
BACKGROUND The poor outcomes in epithelial ovarian cancer necessitate new treatments. In this work, we systematically analyzed the inhibitory effects of ivermectin and the molecular mechanism of its action in ovarian cancer. METHODS The effects of ivermectin alone and its combination with cisplatin on growth and survival were examined using cultured ovarian cancer cells and a xenograft mouse model. The molecular mechanism of action of ivermectin, focusing on Akt/mTOR signaling, was elucidated. RESULTS Ivermectin arrested growth in the G2/M phase and induced caspase-dependent apoptosis in ovarian cancer, regardless of specific cellular and molecular differences. Ivermectin significantly augmented the inhibitory effect of cisplatin on ovarian cancer cells in a dose-dependent manner. Mechanistically, ivermectin suppressed the phosphorylation of key molecules in the Akt/mTOR signaling pathway in ovarian cancer cells. In addition, overexpression of constitutively active Akt restored ivermectin-induced inhibition of Akt/mTOR, growth arrest and apoptosis. In an ovarian cancer xenograft mouse model, ivermectin alone significantly inhibited tumor growth. In combination with cisplatin, tumor growth was completely reversed over the entire duration of drug treatment without any toxicity. Furthermore, the concentrations of ivermectin used in our study are pharmacologically achievable. CONCLUSIONS Our work suggests that ivermectin may be a useful addition to the treatment armamentarium for ovarian cancer and that targeting Akt/mTOR signaling is a therapeutic strategy to increase chemosensitivity in ovarian cancer.
Collapse
Affiliation(s)
- Xiaohong Zhang
- Department of Obstetrics and Gynecology, The People's Hospital of Hanchuan, Hanchuan, Hubei Province, China
| | - Tingting Qin
- Department of Integrated Traditional Chinese and Western Medicine, Tongren Hospital of Wuhan University (Wuhan Third Hospital), Wuhan, Hubei Province, China
| | - Zhengyan Zhu
- Department of Obstetrics and Gynecology, Tongren Hospital of Wuhan University (Wuhan Third Hospital), Wuhan, Hubei Province, China
| | - Fan Hong
- Department of Integrated Traditional Chinese and Western Medicine, Tongren Hospital of Wuhan University (Wuhan Third Hospital), Wuhan, Hubei Province, China
| | - Yang Xu
- Department of Integrated Traditional Chinese and Western Medicine, Tongren Hospital of Wuhan University (Wuhan Third Hospital), Wuhan, Hubei Province, China
| | - Xiongjie Zhang
- Department of Obstetrics and Gynecology, The People's Hospital of Hanchuan, Hanchuan, Hubei Province, China
| | - Xiaohong Xu
- Department of Obstetrics and Gynecology, The People's Hospital of Hanchuan, Hanchuan, Hubei Province, China
| | - Aiping Ma
- Department of Obstetrics and Gynecology, The People's Hospital of Hanchuan, Hanchuan, Hubei Province, China.
| |
Collapse
|
35
|
ALDH1A2 Is a Candidate Tumor Suppressor Gene in Ovarian Cancer. Cancers (Basel) 2019; 11:cancers11101553. [PMID: 31615043 PMCID: PMC6826427 DOI: 10.3390/cancers11101553] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 09/29/2019] [Accepted: 10/10/2019] [Indexed: 12/22/2022] Open
Abstract
Aldehyde dehydrogenase 1 family member A2 (ALDH1A2) is a rate-limiting enzyme involved in cellular retinoic acid synthesis. However, its functional role in ovarian cancer remains elusive. Here, we found that ALDH1A2 was the most prominently downregulated gene among ALDH family members in ovarian cancer cells, according to complementary DNA microarray data. Low ALDH1A2 expression was associated with unfavorable prognosis and shorter disease-free and overall survival for ovarian cancer patients. Notably, hypermethylation of ALDH1A2 was significantly higher in ovarian cancer cell lines when compared to that in immortalized human ovarian surface epithelial cell lines. ALDH1A2 expression was restored in various ovarian cancer cell lines after treatment with the DNA methylation inhibitor 5-aza-2'-deoxycytidine. Furthermore, silencing DNA methyltransferase 1 (DNMT1) or 3B (DNMT3B) restored ALDH1A2 expression in ovarian cancer cell lines. Functional studies revealed that forced ALDH1A2 expression significantly impaired the proliferation of ovarian cancer cells and their invasive activity. To the best of our knowledge, this is the first study to show that ALDH1A2 expression is regulated by the epigenetic regulation of DNMTs, and subsequently that it might act as a tumor suppressor in ovarian cancer, further suggesting that enhancing ALDH1A2-linked signaling might provide new opportunities for therapeutic intervention in ovarian cancer.
Collapse
|
36
|
Wang Z, Song Y, Han X, Qu P, Wang W. Long noncoding RNA PTENP1 affects the recovery of spinal cord injury by regulating the expression of miR‐19b and miR‐21. J Cell Physiol 2019; 235:3634-3645. [PMID: 31583718 DOI: 10.1002/jcp.29253] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Accepted: 09/03/2019] [Indexed: 12/12/2022]
Affiliation(s)
- Zhan Wang
- Department of Orthopedics Gansu Provincial Hospital Lanzhou Gansu China
- Department of Orthopedics People's Clinical Medical College of Lanzhou University Lanzhou Gansu China
| | - Yuxin Song
- Department of Orthopedics Gansu Provincial Hospital Lanzhou Gansu China
- Department of Orthopedics People's Clinical Medical College of Lanzhou University Lanzhou Gansu China
| | - Xingwen Han
- Department of Orthopedics The First Hospital of Lanzhou University Lanzhou Gansu China
- Department of Orthopedics The First Clinical Medicine College of Lanzhou University Lanzhou Gansu China
| | - Peng Qu
- Department of Orthopedics The First Hospital of Lanzhou University Lanzhou Gansu China
| | - Wenji Wang
- Department of Orthopedics The First Hospital of Lanzhou University Lanzhou Gansu China
- Department of Orthopedics The First Clinical Medicine College of Lanzhou University Lanzhou Gansu China
| |
Collapse
|
37
|
Yu Y, Suryo Rahmanto Y, Shen YA, Ardighieri L, Davidson B, Gaillard S, Ayhan A, Shi X, Xuan J, Wang TL, Shih IM. Spleen tyrosine kinase activity regulates epidermal growth factor receptor signaling pathway in ovarian cancer. EBioMedicine 2019; 47:184-194. [PMID: 31492560 PMCID: PMC6796592 DOI: 10.1016/j.ebiom.2019.08.055] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 08/14/2019] [Accepted: 08/23/2019] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Spleen tyrosine kinase (SYK) is frequently upregulated in recurrent ovarian carcinomas, for which effective therapy is urgently needed. SYK phosphorylates several substrates, but their translational implications remain unclear. Here, we show that SYK interacts with EGFR and ERBB2, and directly enhances their phosphorylation. METHODS We used immunohistochemistry and immunoblotting to assess SYK and EGFR phosphorylation in ovarian serous carcinomas. Association with survival was determined by Kaplan-Meier analysis and the log-rank test. To study its role in EGFR signaling, SYK activity was modulated using a small molecule inhibitor, a syngeneic knockout, and an active kinase inducible system. We applied RNA-seq and phosphoproteomic mass spectrometry to investigate the SYK-regulated EGF-induced transcriptome and downstream substrates. FINDINGS Induced expression of constitutively active SYK130E reduced cellular response to EGFR/ERBB2 inhibitor, lapatinib. Expression of EGFRWT, but not SYK non-phosphorylatable EGFR3F mutant, resulted in paclitaxel resistance, a phenotype characteristic to SYK active ovarian cancers. In tumor xenografts, SYK inhibitor reduces phosphorylation of EGFR substrates. Compared to SYKWT cells, SYKKO cells have an attenuated EGFR/ERBB2-transcriptional activity and responsiveness to EGF-induced transcription. In ovarian cancer tissues, pSYK (Y525/526) levels showed a positive correlation with pEGFR (Y1187). Intense immunoreactivity of pSYK (Y525/526) correlated with poor overall survival in ovarian cancer patients. INTERPRETATION These findings indicate that SYK activity positively modulates the EGFR pathway, providing a biological foundation for co-targeting SYK and EGFR. FUND: Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, NIH/NCI, Ovarian Cancer Research Foundation Alliance, HERA Women's Cancer Foundation and Roseman Foundation. Funders had no role in the design of the study and collection, analysis, and interpretation of data and in writing the manuscript and eventually in the decision to submit the manuscript.
Collapse
Affiliation(s)
- Yu Yu
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Medical Institutions, Baltimore, MD 21231, United States of America; Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD 21205, United States of America.
| | - Yohan Suryo Rahmanto
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Medical Institutions, Baltimore, MD 21231, United States of America; Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD 21205, United States of America
| | - Yao-An Shen
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Medical Institutions, Baltimore, MD 21231, United States of America; Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD 21205, United States of America
| | - Laura Ardighieri
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD 21205, United States of America
| | - Ben Davidson
- Department of Pathology, Oslo University Hospital and Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Norwegian Radium Hospital, 0310 Oslo, Norway
| | - Stephanie Gaillard
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Medical Institutions, Baltimore, MD 21231, United States of America
| | - Ayse Ayhan
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD 21205, United States of America; Department of Pathology, Seirei Mikatahara Hospital, Hamamatsu and Hiroshima Universities Schools of Medicine, Hamamatsu 431-3192, Japan
| | - Xu Shi
- Bradley Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA 22203, United States of America
| | - Jianhua Xuan
- Bradley Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA 22203, United States of America
| | - Tian-Li Wang
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Medical Institutions, Baltimore, MD 21231, United States of America; Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD 21205, United States of America; Department of Gynecology and Obstetrics, Johns Hopkins Medical Institutions, Baltimore, MD 21287, United States of America.
| | - Ie-Ming Shih
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Medical Institutions, Baltimore, MD 21231, United States of America; Department of Gynecology and Obstetrics, Johns Hopkins Medical Institutions, Baltimore, MD 21287, United States of America.
| |
Collapse
|
38
|
Zhang Y, Ye T, Gong S, Hong Z, Zhou X, Liu H, Qu H, Qian J. RNA-sequencing based bone marrow cell transcriptome analysis reveals the potential mechanisms of E'jiao against blood-deficiency in mice. Biomed Pharmacother 2019; 118:109291. [PMID: 31401395 DOI: 10.1016/j.biopha.2019.109291] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 07/20/2019] [Accepted: 07/31/2019] [Indexed: 02/05/2023] Open
Abstract
As a health-care food and traditional Chinese medicine, E'jiao, from the skin of Equus animus L, has been used to nourish blood in China for more than 2000 years. In modern medicine, there are also evidences indicate it has a beneficial effect on chemotherapy-caused blood deficiency. However, its mechanism of action for blood invigoration remains unclear. In the present study, we investigated the hematopoietic effect of E'jiao in 5-Fluorouracil-treated mice. In addition to the counting of bone marrow nucleated cells (BMNCs), flow cytometry was used to detect the population of hematopoietic stem cells (HSCs), and colony-forming unit (CFU) was used to assay the differentiation ability of hematopoietic progenitor cells (HPCs). Gene expression profiles of bone marrow cells were obtained from RNA sequencing (RNA-seq) and differentially expressed genes (DEGs) were analyzed with an emphasis on hematopoiesis-related pathways. The results show that E'jiao promotes the proliferation of both BMNCs and HSCs, as well as the differentiation of HPCs. By providing a hematopoiesis-related molecular regulatory network of E'jiao, we point out that the mechanism of E'jiao is associated with pathways including ECM-receptor interaction, Wnt signaling pathway, PI3K-Akt signaling pathway, TGF-beta signaling pathway, Hematopoietic cell lineage and Osteoclast differentiation, in which Ibsp, Col1a1, Col1a2, Notum, Sost, Dkk1, Irx5, Irx3 and Dcn are the key regulatory molecules. These findings provide valuable molecular basis for the mechanism of action of E'jiao.
Collapse
Affiliation(s)
- Yan Zhang
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China; National Engineering Research Center for Gelatin-based Traditional Chinese Medicine, Dong-E-E-Jiao Co., Ltd., Liaocheng, China
| | - Tingting Ye
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Shuqing Gong
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Zhuping Hong
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Xiangshan Zhou
- National Engineering Research Center for Gelatin-based Traditional Chinese Medicine, Dong-E-E-Jiao Co., Ltd., Liaocheng, China
| | - Haibin Liu
- National Engineering Research Center for Gelatin-based Traditional Chinese Medicine, Dong-E-E-Jiao Co., Ltd., Liaocheng, China.
| | - Haibin Qu
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
| | - Jing Qian
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
| |
Collapse
|
39
|
Cui R, Cao G, Bai H, Zhang Z. LPAR1 regulates the development of intratumoral heterogeneity in ovarian serous cystadenocarcinoma by activating the PI3K/AKT signaling pathway. Cancer Cell Int 2019; 19:201. [PMID: 31384176 PMCID: PMC6664705 DOI: 10.1186/s12935-019-0920-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Accepted: 07/22/2019] [Indexed: 02/08/2023] Open
Abstract
Background To explore the role of lysophosphatidic acid receptor 1 (LPAR1) and its correlation with the PI3K/AKT pathway in the development of intratumoral heterogeneity (ITH) in human ovarian serous cystadenocarcinoma (OSC). Methods Immunohistochemical staining was performed to detect LPAR1 expression in matched primary and recurrent lesions from the same patients. Cell models of ITH were established using the limiting dilution methodology and Transwell invasion/migration assays. LPAR1 expression in the ITH cell models was silenced or upregulated with lentiviral particles, and the biological characteristics were evaluated using various in vitro and in vivo assessments of cell function. The levels of phosphorylated PI3K/AKT (p-PI3K/p-AKT) in LPAR1 knockdown and LPAR1-overexpressing cells were detected. Results The H-scores for LPAR1 staining in the lymphatic metastatic and recurrent lesions were noticeably higher than in the primary tumor lesions from the same patients (P = 0.024/0.031). High LPAR1 expression was associated with worse progression-free survival and overall survival (P = 0.017/0.039). Biological functions in vitro, including invasion, migration, and proliferation, and tumor formation in vivo were decreased in the LPAR1-silenced cells (all P < 0.05). These cellular functions were significantly increased in the LPAR1-overexpressing cells in vitro and in vivo (all P < 0.05). The levels of p-PI3K and p-AKT were significantly decreased in the LPAR1 knockdown cells and significantly increased in the LPAR1-overexpressing cells (all P < 0.05). Conclusions Higher levels of the LPAR1 protein were associated with a poor prognosis. LPAR1 plays essential roles in the invasion, migration, and proliferation of heterogeneous subsets of OSC cell lines and the development of ITH of OSC, possibly by modulating the activity of the PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Ran Cui
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Capital Medical University, No.8, North Road of Workers Stadium, Chaoyang District, Beijing, 100020 China
| | - Guangming Cao
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Capital Medical University, No.8, North Road of Workers Stadium, Chaoyang District, Beijing, 100020 China
| | - Huimin Bai
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Capital Medical University, No.8, North Road of Workers Stadium, Chaoyang District, Beijing, 100020 China
| | - Zhenyu Zhang
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Capital Medical University, No.8, North Road of Workers Stadium, Chaoyang District, Beijing, 100020 China
| |
Collapse
|
40
|
Pivotal Role of AKT2 during Dynamic Phenotypic Change of Breast Cancer Stem Cells. Cancers (Basel) 2019; 11:cancers11081058. [PMID: 31357505 PMCID: PMC6721305 DOI: 10.3390/cancers11081058] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Revised: 07/10/2019] [Accepted: 07/22/2019] [Indexed: 12/12/2022] Open
Abstract
Therapeutic resistance seen in aggressive forms of breast cancer remains challenging for current treatments. More than half of the patients suffer from a disease relapse, most of them with distant metastases. Cancer maintenance, resistance to therapy, and metastatic disease seem to be sustained by the presence of cancer stem cells (CSC) within a tumor. The difficulty in targeting this subpopulation derives from their dynamic interconversion process, where CSC can differentiate to non-CSC, which in turn de-differentiate into cells with CSC properties. Using fluorescent CSC models driven by the expression of ALDH1A 1(aldehyde dehydrogenase 1A1), we confirmed this dynamic phenotypic change in MDA-MB-231 breast cancer cells and to identify Serine/Threonine Kinase 2 (AKT2) as an important player in the process. To confirm the central role of AKT2, we silenced AKT2 expression via small interfering RNA and using a chemical inhibitor (CCT128930), in both CSC and non-CSC from different cancer cell lines. Our results revealed that AKT2 inhibition effectively prevents non-CSC reversion through mesenchymal to epithelial transition, reducing invasion and colony formation ability of both, non-CSC and CSC. Further, AKT2 inhibition reduced CSC survival in low attachment conditions. Interestingly, in orthotopic tumor mouse models, high expression levels of AKT2 were detected in circulating tumor cells (CTC). These findings suggest AKT2 as a promising target for future anti-cancer therapies at three important levels: (i) Epithelial-to-mesenchymal transition (EMT) reversion and maintenance of CSC subpopulation in primary tumors, (ii) reduction of CTC and the likelihood of metastatic spread, and (iii) prevention of tumor recurrence through inhibition of CSC tumorigenic and metastatic potential.
Collapse
|
41
|
PI3K-AKT-mTOR and NFκB Pathways in Ovarian Cancer: Implications for Targeted Therapeutics. Cancers (Basel) 2019; 11:cancers11070949. [PMID: 31284467 PMCID: PMC6679095 DOI: 10.3390/cancers11070949] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 06/10/2019] [Accepted: 06/30/2019] [Indexed: 02/07/2023] Open
Abstract
Ovarian cancer is the most lethal gynecologic malignancy in the United States, with an estimated 22,530 new cases and 13,980 deaths in 2019. Recent studies have indicated that the phosphoinositol 3 kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR), as well as the nuclear factor-κ light chain enhancer of activated B cells (NFκB) pathways are highly mutated and/or hyper-activated in a majority of ovarian cancer patients, and are associated with advanced grade and stage disease and poor prognosis. In this review, we will investigate PI3K/AKT/mTOR and their interconnection with NFκB pathway in ovarian cancer cells.
Collapse
|
42
|
Zhu H, Diao S, Lim V, Hu L, Hu J. FAM83D inhibits autophagy and promotes proliferation and invasion of ovarian cancer cells via PI3K/AKT/mTOR pathway. Acta Biochim Biophys Sin (Shanghai) 2019; 51:509-516. [PMID: 30939187 DOI: 10.1093/abbs/gmz028] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Indexed: 11/13/2022] Open
Abstract
Ovarian cancer is one of the most lethal malignant tumors in women. The family with sequence similarity 83, member D (FAM83D) plays an important role in several cancers, but its function and underlying mechanism in ovarian cancer remain unknown. To investigate the role of FAM83D in ovarian cancer, the expression of FAM83D was determined by immunohistochemistry in tissue microarray slide. Cellular proliferation and invasion were detected by 5-Ethynyl-2'-deoxyuridine assays and transwell invasion assays. The correlations between FAM83D and autophagy were detected by western blot analysis and confocal microscopy. Western blot analysis was used to identify the protein expression of FAM83D, phosphoinositide 3-kinase (PI3K), protein kinase B (AKT), mammalian target of rapamycin (mTOR) and Sequestosome 1 (P62). Tumorigenesis in nude mice was used to explore the function of FAM83D in vivo. We found high expression level of FAM83D in ovarian cancer tissues as compared to the normal ovarian tissues. Knockdown of FAM83D in SKOV3 cells enhanced autophagy and inhibited the proliferation and invasion in vitro, whereas ectopic expression of FAM83D in A2780 cells exerted an opposite effect. Mechanistically, overexpression of FAM83D activated the PI3K/AKT/mTOR pathway, and Torin1 could suppress FAM83D-induced cell proliferation and invasion. In vivo, overexpression FAM83D promoted tumor growth. Overall, FAM83D promoted ovarian cancer cell invasion and proliferation, while inhibited autophagy via the PI3K/AKT/mTOR signaling pathway. Our results suggest that FAM83D may be a candidate oncogene in ovarian cancer, which provides a fresh perspective of FAM83D in ovarian cancer.
Collapse
Affiliation(s)
- Hongtao Zhu
- Department of Obstetrics and Gynecology, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Shuai Diao
- Department of Obstetrics and Gynecology, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Vincent Lim
- Department of Obstetrics and Gynecology, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Lina Hu
- Department of Obstetrics and Gynecology, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Jianguo Hu
- Department of Obstetrics and Gynecology, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| |
Collapse
|
43
|
Egiz M, Usui T, Ishibashi M, Zhang X, Shigeta S, Toyoshima M, Kitatani K, Yaegashi N. La-Related Protein 4 as a Suppressor for Motility of Ovarian Cancer Cells. TOHOKU J EXP MED 2019; 247:59-67. [PMID: 30686809 DOI: 10.1620/tjem.247.59] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The La-related proteins (LARPs) are a family of RNA binding proteins that control the degradation and stabilization of RNAs. As emerging research reveals the biology of each LARP, it is evident that LARPs are dysregulated in some types of cancer. Upregulation of cell motility potentiates the metastatic potential of ovarian cancer cells; however, the roles of LARPs in cell motility remain unknown. In the present study, we investigated the roles of LARPs in the progression of ovarian cancer using SKOV3 human ovarian cancer cells and a public database that integrates microarray-based gene expression data and clinical data. To explore the involvement of LARPs in the cell motility, we performed RNA interference screening for LARP mRNAs in SKOV3 cells. The screening identified LARP4 as a potential suppressor of the formation of lamellipodia. Conversely, enforced expression of LARP4 suppressed the formation of lamellipodia. Moreover, cell migration was significantly increased in LARP4-depleted SKOV3 cells. Mechanistically, LARP4 depletion was associated with the decrease in RhoA protein expression. These results suggest that LARP4 may limit RhoA-dependent cell motility. In a mouse xenograft model with SKOV3 cells, LARP4 depletion potentiated peritoneal metastasis. Upon analysis of a public database of patients with ovarian cancer, the LARP4 mRNA-high expression group (n = 166) showed longer overall survival compared with the LARP4 mRNA-low expression group (n = 489), implying a positive correlation of LARP4 mRNA levels in ovarian cancer tissues with patient prognosis. Taken together, we propose that LARP4 could suppress motility and metastatic potential of ovarian cancer cells.
Collapse
Affiliation(s)
- Mahy Egiz
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Tohoku University
- Department of Obstetrics and Gynecology, Menoufia University Graduate School of Medicine
| | - Toshinori Usui
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Tohoku University
| | - Masumi Ishibashi
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Tohoku University
| | - Xuewei Zhang
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Tohoku University
| | - Shogo Shigeta
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Tohoku University
| | - Masafumi Toyoshima
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Tohoku University
| | - Kazuyuki Kitatani
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Tohoku University
- Tohoku Medical Megabank Organization, Tohoku University
- Laboratory of Immunopharmacology, Faculty of Pharmaceutical Sciences, Setsunan University
| | - Nobuo Yaegashi
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Tohoku University
- Tohoku Medical Megabank Organization, Tohoku University
| |
Collapse
|
44
|
Bu Shen Yang Xue Prescription Has Treating Effect on Endometrial Cancer through FSH/PI3K/AKT/Gankyrin/HIF- α/cyclinD 1 Pathway in Ishikawa Cells. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 2018:8412984. [PMID: 30402135 PMCID: PMC6198581 DOI: 10.1155/2018/8412984] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 09/02/2018] [Accepted: 09/26/2018] [Indexed: 11/22/2022]
Abstract
Background. The formulation of Bu Shen Yang Xue (BSYX) has been clinically used in treating gynecologic disease in China, especially for the development of the endometrium. Endometrial carcinoma is the most common malignant tumor of the female genital tract in developed countries. And few studies have been reported on the antitumor activity of BSYX. Therefore, this study aimed to investigate the effect of BSYX on endometrial cancer and make an initial discussion of the underlining mechanisms in Ishikawa cells. Methods and Results. Firstly, 60 SPF female nude mice were randomly divided into control group, model group, BSYX group, and positive group. The models of subcutaneous tumor xenograft of nude mice were established by injection of human endometrial carcinoma cell line Ishikawa tumor cell suspension. Compared with model group, BSYX reduced effectively tumor volume and changed pathological feature in mice tumor issue. Meanwhile, proteins from tumor issues were detected by western blot analysis. The protein levels of follicle-stimulating hormone receptor (FSHR), p-Akt/Akt, Gankyrin, and cyclinD1 in the model group were higher than those in control group but the expression in BSYX group was lower than that in the model group. The hypoxia inducible factor alpha (HIF-α) protein level in the model group was lower than those in control group and upregulated in BSYX group. In addition, Ishikawa cells were cultured and then exposed to follicle-stimulating hormone (FSH), LY294002, a highly selective PI3K inhibitor and serum containing BSYX, respectively. LY294002 and BSYX markedly decreased the cancer cell viability and migration ability and increased the apoptosis rate. FSH promoted the cancer cell ability and migration ability. LY294002 and BSYX evidently downregulated the proteins levels of FSHR, p-Akt/Akt, Gankyrin, and cyclinD1 and upregulated the expression of HIF-α protein, and FSH was on the opposite. Conclusions. Taken together, our results showed that the formulation of BSYX had antitumor effect on endometrial cancer in vivo and in vitro and was related with FSH/PI3K/AKT/Gankyrin/HIF-α/cyclinD1 transduction pathway.
Collapse
|
45
|
Cai B, Ma L, Nong S, Wu Y, Guo X, Pu J. β-elemene induced anticancer effect in bladder cancer through upregulation of PTEN and suppression of AKT phosphorylation. Oncol Lett 2018; 16:6019-6025. [PMID: 30333873 DOI: 10.3892/ol.2018.9401] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 08/29/2018] [Indexed: 12/25/2022] Open
Abstract
Human bladder cancer is one of the most aggressive tumours known and has shown resistance to traditional chemotherapy, which depends heavily on DNA-damaging drugs. β-elemene is one of the least cytotoxic antitumor agents that are extracted from Curcuma aromatica salisb and it exhibits antitumor effects in many carcinomas. β-elemene has attracted the attention of clinicians and scientists worldwide due to its few side effects and limited effect on the bone marrow. However, the antitumor mechanism of β-elemene remains largely unstudied. In the present study, the expression of the AKT serine/threonine kinase (AKT) signaling pathway in bladder cancer and normal bladder tissue was investigated, and the influence of β-elemene on bladder cancer cells and the mechanisms involved were assessed. The results showed that phosphatase and tensin homolog deleted on chromosome ten (PTEN) was downregulated and phosphorylated-AKT (pAKT) was overexpressed in human bladder cancer. β-elemene significantly suppressed the viability of bladder cancer cells, while leaving normal bladder cells unaffected. In addition, there was an increased number of apoptotic bladder cancer cells following β-elemene treatment, and a significant reduction in cell invasion and migration. Subsequent western blot analyses revealed that bladder cancer cells treated with β-elemene had increased PTEN expression and decreased expression of pAKT. Taken together, these results suggest that β-elemene has an antitumor effect in bladder cancer cells through the upregulation of PTEN and suppression of AKT phosphorylation.
Collapse
Affiliation(s)
- Bo Cai
- Department of Urology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China.,Department of Urology, The First Affiliated Hospital of Suzhou University, Suzhou, Jiangsu 215001, P.R. China
| | - Limin Ma
- Department of Urology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Shaojun Nong
- Department of Urology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - You Wu
- Department of Urology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Xin Guo
- Department of Urology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Jinxian Pu
- Department of Urology, The First Affiliated Hospital of Suzhou University, Suzhou, Jiangsu 215001, P.R. China
| |
Collapse
|
46
|
Zhang F, Chen H, Du J, Wang B, Yang L. Anticancer Activity of Metformin, an Antidiabetic Drug, Against Ovarian Cancer Cells Involves Inhibition of Cysteine-Rich 61 (Cyr61)/Akt/Mammalian Target of Rapamycin (mTOR) Signaling Pathway. Med Sci Monit 2018; 24:6093-6101. [PMID: 30171812 PMCID: PMC6130173 DOI: 10.12659/msm.909745] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Background Ovarian cancer is considered one of the lethal cancers responsible for high mortality and morbidity across the world. The prognosis and the survival rate of ovarian cancer is far from decent. Cysteine-rich 61 (Cyr61) also known as CCN1, is a member of CCN-family of growth factors, reported to be significantly overexpressed in several malignancies which include, but are not limited to, ovarian cancer. Recent studies have revealed that women with type 2 diabetes mellitus have an elevated risk of ovarian cancer. Hence, administration of an antidiabetic drug with anticancer effects such as metformin may act as an effective therapeutic regime against ovarian cancer. Material/Methods Cell viability and apoptosis were examined by MTT and Annexin V/PI double staining respectively. Cell migration was determined by Boyden Chamber assay. Transient knockdown of Cyr61 in ovarian cancer cells was achieved by transecting the cells with siRNA for Cyr61using Lipofectamine 2000. Results Our results indicated that treatment of ovarian cancer cells with metformin caused significant downregulation of Cyr61 protein expression levels ultimately favoring apoptosis. Transient knockdown of Cyr61 resulted in the inhibition of cell proliferation and migration. This was also associated with the concomitant downregulation of pAkt and pmTOR confirming the role of Cyr61 as an upstream modulator of Akt signaling. Conversely the extracellular supplementation of recombinant Cyr61 attenuates the cytotoxic properties of metformin in ovarian cancer cells. Conclusions Taken together, we concluded that metformin exhibits anticancer effects and Cyr61 acts as a direct target for metformin in ovarian cancer cells.
Collapse
Affiliation(s)
- Fengli Zhang
- Infertility Center, Qilu Hospital, Shandong University, Jinan, Shandong, China (mainland).,Department of Obstetrics and Gynecology, Liaocheng People's Hospital, Liaocheng, Shandong, China (mainland)
| | - Huixiao Chen
- Department of Obstetrics and Gynecology, Liaocheng People's Hospital, Liaocheng, Shandong, China (mainland)
| | - Jing Du
- Department of Obstetrics and Gynecology, Liaocheng People's Hospital, Liaocheng, Shandong, China (mainland)
| | - Bin Wang
- Department of Obstetrics and Gynecology, Liaocheng People's Hospital, Liaocheng, Shandong, China (mainland)
| | - Lixiao Yang
- Department of Obstetrics and Gynecology, Liaocheng People's Hospital, Liaocheng, Shandong, China (mainland)
| |
Collapse
|
47
|
Zhu J, Cai Y, Xu K, Ren X, Sun J, Lu S, Chen J, Xu P. Beclin1 overexpression suppresses tumor cell proliferation and survival via an autophagy‑dependent pathway in human synovial sarcoma cells. Oncol Rep 2018; 40:1927-1936. [PMID: 30066884 PMCID: PMC6111547 DOI: 10.3892/or.2018.6599] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 07/17/2018] [Indexed: 12/19/2022] Open
Abstract
Beclin1 is an important autophagy‑related prot-ein, which is involved in both autophagy and apoptosis. In recent years, the antitumor effect of Beclin1 has received increased attention. In the present study, we established a stable Beclin1‑overexpressing cell line with SW982 human synovial sarcoma cells. We found that Beclin1 overexpression decreased the cell viability, inhibited proliferation and induced apoptosis in SW982 cells. The expression levels of Bcl‑2 and PCNA were decreased, while the levels of cleaved‑caspase‑3 and cleaved‑PARP were increased. Beclin1 is closely related with autophagy, thus the autophagy‑related markers LC3 and p62 were detected by western blot analysis, and transmission electron microscopy was used to observe autophagosomes. The results showed that the expression level of LC3II was increased and that of p62 was decreased. Moreover, many double membrane‑enclosed autophagosomes were found in cells with Beclin1 overexpression, which indicated that the autophagic activity was enhanced. To explore the effect of autophagy on the viability of SW982 cells, Atg5 was knocked down using siRNA to inhibit the autophagic activity. We found that autophagy contributed to the decrease in cell viability. Knockdown of Atg5 increased the viability and decreased the apoptotic rate of SW982 cells with Beclin1 overexpression. The expression level of Bcl‑2 was increased, while the expression levels of cleaved‑caspase‑3 and cleaved‑PARP were decreased. We also found that the Akt/Bcl‑2/caspase‑9 pathway was involved. The phosphorylation of AKT was positively correlated with cell viability. The cleavage of caspase‑9 was increased by Beclin1 overexpression and decreased by inhibition of autophagy. Altogether, our results suggested that both autophagy and apoptosis contributed to the antitumor effect of Beclin1 in SW982 cells.
Collapse
Affiliation(s)
- Jialin Zhu
- Department of Joint Surgery, Xi'an Hong Hui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710054, P.R. China
| | - Yongsong Cai
- Department of Joint Surgery, Xi'an Hong Hui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710054, P.R. China
| | - Ke Xu
- Department of Joint Surgery, Xi'an Hong Hui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710054, P.R. China
| | - Xiaoyu Ren
- Department of Joint Surgery, Xi'an Hong Hui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710054, P.R. China
| | - Jian Sun
- Department of Genetics and Molecular Biology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, P.R. China
| | - Shemin Lu
- Department of Genetics and Molecular Biology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, P.R. China
| | - Jinghong Chen
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, Institute of Endemic Diseases, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, P.R. China
| | - Peng Xu
- Department of Joint Surgery, Xi'an Hong Hui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710054, P.R. China
| |
Collapse
|
48
|
Yan T, Liang W, Jiang E, Ye A, Wu Q, Xi M. GINS2 regulates cell proliferation and apoptosis in human epithelial ovarian cancer. Oncol Lett 2018; 16:2591-2598. [PMID: 30013653 DOI: 10.3892/ol.2018.8944] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 05/24/2018] [Indexed: 12/22/2022] Open
Abstract
Go-Ichi-Ni-San 2 (GINS2), also known as partner of Sld five 2, is involved in the initiation of DNA replication and cell cycle progression. GINS2 is abundantly expressed in a number of malignant solid tumors, including breast cancer, melanoma and hepatic carcinoma. However, the functions of GINS2 in epithelial ovarian cancer (EOC) remain unclear. The aim of the present study was to investigate these functions. GINS2 expression was detected in EOC and normal ovarian tissues using immunohistochemistry. To investigate the functions of GINS2 in EOC, GINS2 expression was stably knocked down in SKOV-3 cells using lentiviral short hairpin RNA (shRNA). The expression of GINS2 mRNA and protein in SKOV-3 cells was examined using reverse-transcription quantitative polymerase chain reaction (RT-qPCR) and western blot analyses, respectively. Cell proliferation was determined using high-content screening and MTT assays. Cell cycle progression and apoptosis were detected using flow cytometry. Compared with normal ovarian tissues, EOC tissues expressed increased levels of GINS2 expression (16.7 vs. 58.3%). Increased expression of GINS2 mRNA was also observed in SKOV-3 and OVCAR3 cells. In the investigation of GINS2 functions in EOC, GINS2 expression at the mRNA and protein levels was significantly inhibited by specific GINS2 shRNA. GINS2 knockdown significantly inhibited the proliferation and viability of SKOV-3 cells and induced cell cycle arrest in S phase. Furthermore, GINS2 knockdown in SKOV-3 cells significantly increased cell apoptosis. GINS2 is markedly expressed in EOC tissues and cell lines. Stable GINS2 knockdown in SKOV-3 cells significantly inhibited cell proliferation and induced cell cycle arrest and cell apoptosis. Therefore, GINS2 may be involved in EOC progression.
Collapse
Affiliation(s)
- Ting Yan
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China.,Department of Gynecology, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, P.R. China
| | - Wentong Liang
- Department of Gynecology, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, P.R. China
| | - Enli Jiang
- Department of Gynecology, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, P.R. China
| | - Aizhu Ye
- Department of Clinical Laboratory, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, P.R. China
| | - Qian Wu
- Department of Pathology, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, P.R. China
| | - Mingrong Xi
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
49
|
Zhao Q, Qian Q, Cao D, Yang J, Gui T, Shen K. Role of BMI1 in epithelial ovarian cancer: investigated via the CRISPR/Cas9 system and RNA sequencing. J Ovarian Res 2018; 11:31. [PMID: 29685168 PMCID: PMC5911954 DOI: 10.1186/s13048-018-0406-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 04/13/2018] [Indexed: 01/28/2023] Open
Abstract
Background B-cell-specific Moloney murine leukemia virus integration site 1 (BMI1) might be an appropriate biomarker in the management of epithelial ovarian cancer (EOC). However, the biological role of BMI1 and its relevant molecular mechanism needs further elaboration. Clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 system is an excellent genome-editing tool and is scarcely used in EOC studies. Methods We first applied CRISPR/Cas9 technique to silence BMI1 in EOC cells; thereafter we accomplished various in vivo and in vitro experiments to detect biological behaviors of ovarian cancer cells, including MTT, flow cytometry, Transwell, real-time polymerase chain reaction and western blotting assays, etc.; eventually, we used RNA sequencing to reveal the underlying molecular traits driven by BMI1 in EOC. Results We successfully shut off the expression of BMI1 in EOC cells using CRISPR/Cas9 system, providing an ideal cellular model for investigations of target gene. Silencing BMI1 could reduce cell growth and metastasis, promote cell apoptosis, and enhance the platinum sensitivity of EOC cells. BMI1 might alter extracellular matrix structure and angiogenesis of tumor cells through regulating Focal adhesion and PI3K/AKT pathways. Conclusion BMI1 is a potential biomarker in EOC management, especially for tumor progression and chemo-resistance. Molecular traits, including BMI1 and core genes in Focal adhesion and PI3K/AKT pathways, might be alternatives as therapeutic targets for EOC. Electronic supplementary material The online version of this article (10.1186/s13048-018-0406-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Qianying Zhao
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuaifuyuan, Dongcheng District, Beijing, China.,Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Qiuhong Qian
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuaifuyuan, Dongcheng District, Beijing, China.,Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Shandong, China
| | - Dongyan Cao
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuaifuyuan, Dongcheng District, Beijing, China
| | - Jiaxin Yang
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuaifuyuan, Dongcheng District, Beijing, China
| | - Ting Gui
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuaifuyuan, Dongcheng District, Beijing, China.
| | - Keng Shen
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuaifuyuan, Dongcheng District, Beijing, China.
| |
Collapse
|
50
|
Wang Y, Wang Y, Zhang Z. Adipokine RBP4 drives ovarian cancer cell migration. J Ovarian Res 2018; 11:29. [PMID: 29642915 PMCID: PMC5896151 DOI: 10.1186/s13048-018-0397-9] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 03/12/2018] [Indexed: 12/15/2022] Open
Abstract
Background Obesity has been linked to several types of cancers including ovarian cancer. Retinol binding protein 4 (RBP4) is an adipokine that drives the development of hyperinsulinemia and type II diabetes in obesity patients and animals. Previously, we have identified RBP4 as a serum marker for ovarian cancer. Here we further explored the consequence of RBP4 upregulation in ovarian cancer cells and its molecular mechanism. Results Our results show that RBP4 is overexpressed in ovarian cancer cells to the same extent as in adipose tissues. The overexpression of RBP4 in ovarian cancer cells promotes cancer cell migration and proliferation. At molecular level, cancer progression factors MMP2 and MMP9 are induced in response to RBP4 overexpression. We further investigated which signaling pathways are utilized by RBP4 to activate ovarian cancer cell migration. We found RhoA/Rock1 pathway is turned on and CyclinD1 is upregulated in RBP4 overexpressed cells. Inhibition of RhoA/Rock1 pathway reduces the RBP4-induced MMP2 and MMP9 expression. The RBP4 action is depend on its associated ligand vitamin A/retinol acid (RA) and possibly involves similar pathways as for conferring insulin resistance. Moreover, we show that knockdown of RBP4 significantly reduce cancer cell migration and proliferation as well as expressions of oncogenic factors. Conclusions Our results indicated that RBP4 can drive ovarian cancer cell migration and proliferation via RhoA/Rock1 and ERK pathway. It suggests that RBP4 act as a oncogene in ovarian cancer cells. Thus, RBP4 could be a molecular bridge between obesity and cancers and a potential target for treating obese cancer patients.
Collapse
Affiliation(s)
- Yanyan Wang
- Department of Gynaecology and Obstetrics, Beijing Chaoyang Hospital, Capital Medical University, No. 8 South Road, workers' Stadium, Chaoyang District, Beijing, 100020, China.,The First Affiliated Hospital of Jinzhou Medical University, No.2, people's street, Jinzhou, 121001, China
| | - Yilin Wang
- Department of Gynaecology and Obstetrics, Beijing Chaoyang Hospital, Capital Medical University, No. 8 South Road, workers' Stadium, Chaoyang District, Beijing, 100020, China
| | - Zhenyu Zhang
- Department of Gynaecology and Obstetrics, Beijing Chaoyang Hospital, Capital Medical University, No. 8 South Road, workers' Stadium, Chaoyang District, Beijing, 100020, China.
| |
Collapse
|