1
|
Gilardi C, Martins HC, Levone BR, Bianco AL, Bicker S, Germain PL, Gross F, Sungur AÖ, Kisko TM, Stein F, Meinert S, Schwarting RKW, Wöhr M, Dannlowski U, Kircher T, Schratt G. miR-708-5p is elevated in bipolar patients and can induce mood disorder-associated behavior in mice. EMBO Rep 2025; 26:2121-2145. [PMID: 40065182 PMCID: PMC12019553 DOI: 10.1038/s44319-025-00410-y] [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: 11/04/2024] [Revised: 02/21/2025] [Accepted: 02/24/2025] [Indexed: 04/25/2025] Open
Abstract
Mood disorders (MDs) are caused by an interplay of genetic and environmental (GxE) risk factors. However, molecular pathways engaged by GxE risk factors are poorly understood. Using small-RNA sequencing in peripheral blood mononuclear cells (PBMCs), we show that the bipolar disorder (BD)-associated microRNA miR-708-5p is upregulated in healthy human subjects with a high genetic or environmental predisposition for MDs. miR-708-5p is further upregulated in the hippocampus of rats which underwent juvenile social isolation, a model of early life stress. Hippocampal overexpression of miR-708-5p in adult male mice is sufficient to elicit MD-associated behavioral endophenotypes. We further show that miR-708-5p directly targets Neuronatin (Nnat), an endoplasmic reticulum protein. Restoring Nnat expression in the hippocampus of miR-708-5p-overexpressing mice rescues miR-708-5p-dependent behavioral phenotypes. Finally, miR-708-5p is upregulated in PBMCs from patients diagnosed with MD. Peripheral miR-708-5p expression allows to differentiate male BD patients from patients suffering from major depressive disorder (MDD). In summary, we describe a potential functional role for the miR-708-5p/Nnat pathway in MD etiology and identify miR-708-5p as a potential biomarker for the differential diagnosis of MDs.
Collapse
Affiliation(s)
- Carlotta Gilardi
- Laboratory of Systems Neuroscience, Institute for Neuroscience, Department of Health Science and Technology, ETH Zurich, 8057, Zurich, Switzerland
| | - Helena C Martins
- Laboratory of Systems Neuroscience, Institute for Neuroscience, Department of Health Science and Technology, ETH Zurich, 8057, Zurich, Switzerland
| | - Brunno Rocha Levone
- Laboratory of Systems Neuroscience, Institute for Neuroscience, Department of Health Science and Technology, ETH Zurich, 8057, Zurich, Switzerland
| | - Alessandra Lo Bianco
- Laboratory of Systems Neuroscience, Institute for Neuroscience, Department of Health Science and Technology, ETH Zurich, 8057, Zurich, Switzerland
| | - Silvia Bicker
- Laboratory of Systems Neuroscience, Institute for Neuroscience, Department of Health Science and Technology, ETH Zurich, 8057, Zurich, Switzerland
| | - Pierre-Luc Germain
- Laboratory of Systems Neuroscience, Institute for Neuroscience, Department of Health Science and Technology, ETH Zurich, 8057, Zurich, Switzerland
- Laboratory of Molecular and Behavioural Neuroscience, Institute for Neuroscience, Department of Health Science and Technology, ETH Zurich, 8057, Zurich, Switzerland
- Lab of Statistical Bioinformatics, IMLS, University of Zürich, 8057, Zurich, Switzerland
| | - Fridolin Gross
- Laboratory of Systems Neuroscience, Institute for Neuroscience, Department of Health Science and Technology, ETH Zurich, 8057, Zurich, Switzerland
- CNRS UMR5164 ImmunoConcEpT, University of Bordeaux, Bordeaux, France
| | - Ayse Özge Sungur
- Behavioral Neuroscience, Experimental and Biological Psychology, Faculty of Psychology, Philipps-University of Marburg, D-35032, Marburg, Germany
- Center for Mind, Brain and Behavior, Philipps-University Marburg, D-35032, Marburg, Germany
- Laboratory of Biological Psychology, Social and Affective Neuroscience Research Group, Research Unit Brain and Cognition, Faculty of Psychology and Educational Sciences, KU Leuven, B-3000, Leuven, Belgium
- Leuven Brain Institute, KU Leuven, B-3000, Leuven, Belgium
| | - Theresa M Kisko
- Behavioral Neuroscience, Experimental and Biological Psychology, Faculty of Psychology, Philipps-University of Marburg, D-35032, Marburg, Germany
- Center for Mind, Brain and Behavior, Philipps-University Marburg, D-35032, Marburg, Germany
- Laboratory of Biological Psychology, Social and Affective Neuroscience Research Group, Research Unit Brain and Cognition, Faculty of Psychology and Educational Sciences, KU Leuven, B-3000, Leuven, Belgium
- Leuven Brain Institute, KU Leuven, B-3000, Leuven, Belgium
| | - Frederike Stein
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
| | - Susanne Meinert
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Rainer K W Schwarting
- Behavioral Neuroscience, Experimental and Biological Psychology, Faculty of Psychology, Philipps-University of Marburg, D-35032, Marburg, Germany
| | - Markus Wöhr
- Behavioral Neuroscience, Experimental and Biological Psychology, Faculty of Psychology, Philipps-University of Marburg, D-35032, Marburg, Germany
- Center for Mind, Brain and Behavior, Philipps-University Marburg, D-35032, Marburg, Germany
- Laboratory of Biological Psychology, Social and Affective Neuroscience Research Group, Research Unit Brain and Cognition, Faculty of Psychology and Educational Sciences, KU Leuven, B-3000, Leuven, Belgium
- Leuven Brain Institute, KU Leuven, B-3000, Leuven, Belgium
| | - Udo Dannlowski
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Tilo Kircher
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
| | - Gerhard Schratt
- Laboratory of Systems Neuroscience, Institute for Neuroscience, Department of Health Science and Technology, ETH Zurich, 8057, Zurich, Switzerland.
| |
Collapse
|
2
|
Dey Bhowmik A, Shaw P, Gopinatha Pillai MS, Rao G, Dwivedi SKD. Evolving landscape of detection and targeting miRNA/epigenetics for therapeutic strategies in ovarian cancer. Cancer Lett 2024; 611:217357. [PMID: 39615646 DOI: 10.1016/j.canlet.2024.217357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 11/22/2024] [Accepted: 11/25/2024] [Indexed: 12/14/2024]
Abstract
Ovarian cancer (OC) accounts for the highest mortality rates among all gynecologic malignancies. The high mortality of OC is often associated with delayed detection, prolonged latency, enhanced metastatic potential, acquired drug resistance, and frequent recurrence. This review comprehensively explores key aspects of OC, including cancer diagnosis, mechanisms of disease resistance, and the pivotal role of epigenetic regulation, particularly by microRNAs (miRs) in cancer progression. We highlight the intricate regulatory mechanisms governing miR expression within the context of OC and the current status of epigenetic advancement in the therapeutic development and clinical trial progression. Through network analysis we elucidate the regulatory interactions between dysregulated miRs in OC and their targets which are involved in different signaling pathways. By exploring these interconnected facets and critical analysis, we endeavor to provide a nuanced understanding of the molecular dynamics underlying OC, its detection and shedding light on potential avenues for miRs and epigenetics-based therapeutic intervention and management strategies.
Collapse
Affiliation(s)
- Arpan Dey Bhowmik
- Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Pallab Shaw
- Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Mohan Shankar Gopinatha Pillai
- Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Geeta Rao
- Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Shailendra Kumar Dhar Dwivedi
- Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
3
|
Zhang FF, Zhang L, Zhao L, Lu Y, Dong X, Liu YQ, Li Y, Guo S, Zheng SY, Xiao Y, Jiang YZ. The circular RNA Rap1b promotes Hoxa5 transcription by recruiting Kat7 and leading to increased Fam3a expression, which inhibits neuronal apoptosis in acute ischemic stroke. Neural Regen Res 2023; 18:2237-2245. [PMID: 37056143 PMCID: PMC10328278 DOI: 10.4103/1673-5374.369115] [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: 06/21/2022] [Revised: 10/15/2022] [Accepted: 12/30/2022] [Indexed: 02/17/2023] Open
Abstract
Circular RNAs can regulate the development and progression of ischemic cerebral disease. However, it remains unclear whether they play a role in acute ischemic stroke. To investigate the role of the circular RNA Rap1b (circRap1b) in acute ischemic stroke, in this study we established an in vitro model of acute ischemia and hypoxia by subjecting HT22 cells to oxygen and glucose deprivation and a mouse model of acute ischemia and hypoxia by occluding the right carotid artery. We found that circRap1b expression was remarkably down-regulated in the hippocampal tissue of the mouse model and in the HT22 cell model. In addition, Hoxa5 expression was strongly up-regulated in response to circRap1b overexpression. Hoxa5 expression was low in the hippocampus of a mouse model of acute ischemia and in HT22-AIS cells, and inhibited HT22-AIS cell apoptosis. Importantly, we found that circRap1b promoted Hoxa5 transcription by recruiting the acetyltransferase Kat7 to induce H3K14ac modification in the Hoxa5 promoter region. Hoxa5 regulated neuronal apoptosis by activating transcription of Fam3a, a neuronal apoptosis-related protein. These results suggest that circRap1b regulates Hoxa5 transcription and expression, and subsequently Fam3a expression, ultimately inhibiting cell apoptosis. Lastly, we explored the potential clinical relevance of circRap1b and Hoxa5 in vivo. Taken together, these findings demonstrate the mechanism by which circRap1b inhibits neuronal apoptosis in acute ischemic stroke.
Collapse
Affiliation(s)
- Fang-Fang Zhang
- Department of Rehabilitation Medicine, The People’s Hospital of China Medical University (The People’s Hospital of Liaoning Province), Shenyang, Liaoning Province, China
| | - Liang Zhang
- Department of Rehabilitation Medicine, The People’s Hospital of China Medical University (The People’s Hospital of Liaoning Province), Shenyang, Liaoning Province, China
| | - Lin Zhao
- Department of Rehabilitation Medicine, The People’s Hospital of China Medical University (The People’s Hospital of Liaoning Province), Shenyang, Liaoning Province, China
| | - Yu Lu
- Department of Rehabilitation Medicine, The People’s Hospital of China Medical University (The People’s Hospital of Liaoning Province), Shenyang, Liaoning Province, China
| | - Xin Dong
- Department of Rehabilitation Medicine, The People’s Hospital of China Medical University (The People’s Hospital of Liaoning Province), Shenyang, Liaoning Province, China
| | - Yan-Qi Liu
- Department of Rehabilitation Medicine, The People’s Hospital of China Medical University (The People’s Hospital of Liaoning Province), Shenyang, Liaoning Province, China
| | - Yu Li
- Department of Rehabilitation Medicine, The People’s Hospital of China Medical University (The People’s Hospital of Liaoning Province), Shenyang, Liaoning Province, China
| | - Shuang Guo
- Department of Rehabilitation Medicine, The People’s Hospital of China Medical University (The People’s Hospital of Liaoning Province), Shenyang, Liaoning Province, China
| | - Si-Yuan Zheng
- Department of Rehabilitation Medicine, The People’s Hospital of China Medical University (The People’s Hospital of Liaoning Province), Shenyang, Liaoning Province, China
| | - Ying Xiao
- Department of Rehabilitation Medicine, The People’s Hospital of China Medical University (The People’s Hospital of Liaoning Province), Shenyang, Liaoning Province, China
| | - Yu-Zhu Jiang
- Department of Rehabilitation Medicine, The People’s Hospital of China Medical University (The People’s Hospital of Liaoning Province), Shenyang, Liaoning Province, China
| |
Collapse
|
4
|
Blank M, Katsiampoura A, Wachtendorf LJ, Linhardt FC, Tartler TM, Raub D, Azimaraghi O, Chen G, Houle TT, Ferrone C, Eikermann M, Schaefer MS. Association Between Intraoperative Dexamethasone and Postoperative Mortality in Patients Undergoing Oncologic Surgery: A Multicentric Cohort Study. Ann Surg 2023; 278:e105-e114. [PMID: 35837889 DOI: 10.1097/sla.0000000000005526] [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: 11/26/2022]
Abstract
OBJECTIVE We examined the effects of dexamethasone on postoperative mortality, recurrence-free survival, and side effects in patients undergoing oncologic operations. BACKGROUND Dexamethasone prevents nausea and vomiting after anesthesia and may affect cancer proliferation. METHODS A total of 30,561 adult patients undergoing solid cancer resection between 2005 and 2020 were included. Multivariable logistic regression was applied to investigate the effect of dexamethasone on 1-year mortality and recurrence-free survival. Effect modification by the cancer's potential for immunogenicity, defined as a recommendation for checkpoint inhibitor therapy based on the National Comprehensive Cancer Network guidelines, was investigated through interaction term analysis. Key safety endpoints were dexamethasone-associated risk of hyperglycemia >180 mg/dL within 24 hours and surgical site infections within 30 days after surgery. RESULTS Dexamethasone was administered to 38.2% (11,666/30,561) of patients (6.5±2.3 mg). Overall, 3.2% (n=980/30,561) died and 15.4% (n=4718/30,561) experienced cancer recurrence within 1 year of the operation. Dexamethasone was associated with a -0.6% (95% confidence interval: -1.1, -0.2, P =0.007) 1-year mortality risk reduction [adjusted odds ratio (OR adj ): 0.79 (0.67, 0.94), P =0.009; hazard ratio=0.82 (0.69, 0.96), P =0.016] and higher odds of recurrence-free survival [OR adj : 1.28 (1.18, 1.39), P <0.001]. This effect was only present in patients with solid cancers who were defined as not to respond to checkpoint inhibitor therapy [OR adj : 0.70 (0.57, 0.87), P =0.001 vs OR adj : 1.13 (0.85, 1.50), P =0.40]. A high (>0.09 mg/kg) dose of dexamethasone increased the risk of postoperative hyperglycemia [OR adj : 1.55 (1.32, 1.82), P <0.001], but not for surgical site infections [OR adj : 0.84 (0.42, 1.71), P =0.63]. CONCLUSIONS Dexamethasone is associated with decreased 1-year mortality and cancer recurrence in patients undergoing surgical resection of cancers that are not candidates for immune modulators. Dexamethasone increased the risk of postoperative hyperglycemia, however, no increase in surgical site infections was identified.
Collapse
Affiliation(s)
- Michael Blank
- Department of Anesthesia, Critical Care, and Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
- Center for Anesthesia Research Excellence (CARE), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
- Department of Anesthesiology, Montefiore Medical Center, Albert Einstein College of Medicine, New York City, NY
| | - Anastasia Katsiampoura
- Department of Anesthesia, Critical Care, and Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Luca J Wachtendorf
- Department of Anesthesia, Critical Care, and Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
- Center for Anesthesia Research Excellence (CARE), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
- Department of Anesthesiology, Montefiore Medical Center, Albert Einstein College of Medicine, New York City, NY
| | - Felix C Linhardt
- Department of Anesthesia, Critical Care, and Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
- Center for Anesthesia Research Excellence (CARE), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
- Department of Anesthesiology, Montefiore Medical Center, Albert Einstein College of Medicine, New York City, NY
| | - Tim M Tartler
- Department of Anesthesia, Critical Care, and Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
- Center for Anesthesia Research Excellence (CARE), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Dana Raub
- Department of Anesthesia, Critical Care, and Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Omid Azimaraghi
- Center for Anesthesia Research Excellence (CARE), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
- Department of Anesthesiology, Montefiore Medical Center, Albert Einstein College of Medicine, New York City, NY
| | - Guanqing Chen
- Center for Anesthesia Research Excellence (CARE), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Tim T Houle
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston, MA
| | - Cristina Ferrone
- Department of Surgery, Cancer Center, Massachusetts General Hospital, Boston, MA
| | - Matthias Eikermann
- Department of Anesthesiology, Montefiore Medical Center, Albert Einstein College of Medicine, New York City, NY
- Department of Anesthesiology, Essen University Hospital, Essen, Germany
| | - Maximilian S Schaefer
- Department of Anesthesia, Critical Care, and Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
- Center for Anesthesia Research Excellence (CARE), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
- Department of Anesthesiology, Düsseldorf University Hospital, Düsseldorf, Germany
| |
Collapse
|
5
|
Chu YH, Huang YC, Chiu PY, Kuo WH, Pan YR, Kuo YT, Wang RH, Kao YC, Wang YH, Lin YF, Lin KT. Combating breast cancer progression through combination therapy with hypomethylating agent and glucocorticoid. iScience 2023; 26:106597. [PMID: 37128608 PMCID: PMC10148121 DOI: 10.1016/j.isci.2023.106597] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 12/09/2022] [Accepted: 04/03/2023] [Indexed: 05/03/2023] Open
Abstract
Breast cancer is the leading cause of cancer-related death in women. Among breast cancer types, triple-negative breast cancer (TNBC) accounts for 15% of all breast cancers with aggressive tumor behavior. By using bioinformatic approaches, we observed that the microRNA-708 promoter is highly methylated in breast carcinomas, and this methylation is linked to a poor prognosis. Moreover, microRNA-708 expression correlates with better clinical outcomes in TNBC patients. Combination treatment with the hypomethylating agent decitabine and synthetic glucocorticoid significantly increased the expression of microRNA-708, reactivated DNMT-suppressed pathways, and decreased the expression of multiple metastasis-promoting genes such as matrix metalloproteinases (MMPs) and IL-1β, leading to the suppression of breast cancer cell proliferation, migration, and invasion, as well as reduced tumor growth and distant metastasis in the TNBC xenograft mouse model. Overall, our study reveals a therapeutic opportunity in which a combined regimen of decitabine with glucocorticoid may have therapeutic potential in treating TNBC patients.
Collapse
Affiliation(s)
- Yu-Hsin Chu
- Institute of Biotechnology, College of Life Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Yi-Chen Huang
- Institute of Biotechnology, College of Life Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Pei-Yun Chiu
- Institute of Biotechnology, College of Life Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Wen-Hung Kuo
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Yan-Ru Pan
- Institute of Biotechnology, College of Life Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Yuan-Ting Kuo
- Institute of Biotechnology, College of Life Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Rong-Hsuan Wang
- Institute of Biotechnology, College of Life Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Yu-Chin Kao
- Institute of Biotechnology, College of Life Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Yi-Hsiang Wang
- Institute of Molecular Medicine, College of Life Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Yi-Fan Lin
- Institute of Biotechnology, College of Life Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Kai-Ti Lin
- Institute of Biotechnology, College of Life Science, National Tsing Hua University, Hsinchu, Taiwan
- Department of Medical Science, College of Life Science, National Tsing Hua University, Hsinchu, Taiwan
- Corresponding author
| |
Collapse
|
6
|
Buonaiuto R, Neola G, Cecere SC, Caltavituro A, Cefaliello A, Pietroluongo E, De Placido P, Giuliano M, Arpino G, De Angelis C. Glucocorticoid Receptor and Ovarian Cancer: From Biology to Therapeutic Intervention. Biomolecules 2023; 13:biom13040653. [PMID: 37189400 DOI: 10.3390/biom13040653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 03/27/2023] [Accepted: 03/31/2023] [Indexed: 04/08/2023] Open
Abstract
Ovarian cancer (OC) is the leading cause of death from gynecological malignancies worldwide. Fortunately, recent advances in OC biology and the discovery of novel therapeutic targets have led to the development of novel therapeutic agents that may improve the outcome of OC patients. The glucocorticoid receptor (GR) is a ligand-dependent transcriptional factor known for its role in body stress reactions, energy homeostasis and immune regulation. Notably, evidence suggests that GR may play a relevant role in tumor progression and may affect treatment response. In cell culture models, administration of low levels of glucocorticoids (GCs) suppresses OC growth and metastasis. Conversely, high GR expression has been associated with poor prognostic features and long-term outcomes in patients with OC. Moreover, both preclinical and clinical data have shown that GR activation impairs the effectiveness of chemotherapy by inducing the apoptotic pathways and cell differentiation. In this narrative review, we summarize data related to the function and role of GR in OC. To this aim, we reorganized the controversial and fragmented data regarding GR activity in OC and herein describe its potential use as a prognostic and predictive biomarker. Moreover, we explored the interplay between GR and BRCA expression and reviewed the latest therapeutic strategies such as non-selective GR antagonists and selective GR modulators to enhance chemotherapy sensitivity, and to finally provide new treatment options in OC patients.
Collapse
Affiliation(s)
- Roberto Buonaiuto
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| | - Giuseppe Neola
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| | - Sabrina Chiara Cecere
- Oncologia Clinica Sperimentale Uro-Ginecologica, Istituto Nazionale Tumori IRCCS Fondazione G Pascale, 80131 Naples, Italy
| | - Aldo Caltavituro
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| | - Amedeo Cefaliello
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| | - Erica Pietroluongo
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| | - Pietro De Placido
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| | - Mario Giuliano
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| | - Grazia Arpino
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| | - Carmine De Angelis
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| |
Collapse
|
7
|
Melnik D, Cortés-Sánchez JL, Sandt V, Kahlert S, Kopp S, Grimm D, Krüger M. Dexamethasone Selectively Inhibits Detachment of Metastatic Thyroid Cancer Cells during Random Positioning. Cancers (Basel) 2023; 15:cancers15061641. [PMID: 36980530 PMCID: PMC10046141 DOI: 10.3390/cancers15061641] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 02/27/2023] [Accepted: 03/06/2023] [Indexed: 03/11/2023] Open
Abstract
We recently reported that synthetic glucocorticoid dexamethasone (DEX) is able to suppress metastasis-like spheroid formation in a culture of follicular thyroid cancer (FTC)-133 cells cultured under random positioning. We now show that this inhibition was selective for two metastatic thyroid carcinoma cells, FTC-133 and WRO, whereas benign Nthy-ori 3-1 thyrocytes and recurrent ML-1 follicular thyroid cancer cells were not affected by DEX. We then compare Nthy-ori 3-1 and FTC-133 cells concerning their adhesion and mechanosignaling. We demonstrate that DEX disrupts random positioning-triggered p38 stress signaling in FTC-133 cells, thereby antagonizing a variety of biological functions. Thus, DEX treatment of FTC-133 cells is associated with increased adhesiveness, which is mainly caused by the restored, pronounced formation of a normal number of tight junctions. Moreover, we show that Nthy-ori 3-1 and ML-1 cells upregulate the anti-adhesion protein mucin-1 during random positioning, presumably as a protection against mechanical stress. In summary, mechanical stress seems to be an important component in this metastasis model system that is processed differently by metastatic and healthy cells. The balance between adhesion, anti-adhesion and cell–cell connections enables detachment of adherent human cells on the random positioning machine—or not, allowing selective inhibition of thyroid in vitro metastasis by DEX.
Collapse
Affiliation(s)
- Daniela Melnik
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University, 39106 Magdeburg, Germany
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke University, 39106 Magdeburg, Germany
| | - José Luis Cortés-Sánchez
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University, 39106 Magdeburg, Germany
| | - Viviann Sandt
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University, 39106 Magdeburg, Germany
| | - Stefan Kahlert
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke University, 39106 Magdeburg, Germany
- Institute of Anatomy, Otto von Guericke University, 39120 Magdeburg, Germany
| | - Sascha Kopp
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke University, 39106 Magdeburg, Germany
- Core Facility Tissue Engineering, Otto von Guericke University, 39106 Magdeburg, Germany
| | - Daniela Grimm
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University, 39106 Magdeburg, Germany
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke University, 39106 Magdeburg, Germany
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark
| | - Marcus Krüger
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University, 39106 Magdeburg, Germany
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke University, 39106 Magdeburg, Germany
- Correspondence: ; Tel.: +49-391-6757471
| |
Collapse
|
8
|
Lin SC, Liao YC, Chen PM, Yang YY, Wang YH, Tung SL, Chuang CM, Sung YW, Jang TH, Chuang SE, Wang LH. Periostin promotes ovarian cancer metastasis by enhancing M2 macrophages and cancer-associated fibroblasts via integrin-mediated NF-κB and TGF-β2 signaling. J Biomed Sci 2022; 29:109. [PMID: 36550569 PMCID: PMC9784270 DOI: 10.1186/s12929-022-00888-x] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 11/28/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Ovarian cancer has the highest mortality among gynecological cancers due to late diagnosis and lack of effective targeted therapy. Although the study of interplay between cancer cells with their microenvironment is emerging, how ovarian cancer triggers signaling that coordinates with immune cells to promote metastasis is still elusive. METHODS Microarray and bioinformatics analysis of low and highly invasive ovarian cancer cell lines were used to reveal periostin (POSTN), a matrix protein with multifunctions in cancer, with elevated expression in the highly invasive cells. Anchorage independent assay, Western blot, RNA interference, confocal analysis and neutralizing antibody treatment were performed to analyze the effects of POSTN on tumor promotion and to explore the underlying mechanism. Chemotaxis, flow cytometry and cytokine array analyses were undertaken to analyze the involvement of POSTN in cancer-associated fibroblast (CAF) and macrophage modulation. Correlations between POSTN expression levels and clinical characteristics were analyzed using the Oncomine, commercial ovarian cancer cDNA and China Medical University Hospital patient cohort. In vivo effect of POSTN on metastasis was studied using a mouse xenograft model. RESULTS Expression of POSTN was found to be elevated in highly invasive ovarian cancer cells. We observed that POSTN was co-localized with integrin β3 and integrin β5, which was important for POSTN-mediated activation of ERK and NF-κB. Ectopic expression of POSTN enhanced whereas knockdown of POSTN decreased cancer cell migration and invasion in vitro, as well as tumor growth and metastasis in vivo. POSTN enhanced integrin/ERK/NF-κB signaling through an autocrine effect on cancer cells to produce macrophage attracting and mobilizing cytokines including MIP-1β, MCP-1, TNFα and RANTES resulting in increased chemotaxis of THP-1 monocytes and their polarization to M2 macrophages in vitro. In agreement, tumors derived from POSTN-overexpressing SKOV3 harbored more tumor-associated macrophages than the control tumors. POSTN induced TGF-β2 expression from ovarian cancer cells to promote activation of adipose-derived stromal cells to become CAF-like cells expressing alpha smooth muscle actin and fibroblast activation protein alpha. Consistently, increased CAFs were observed in POSTN overexpressing SKOV3 cells-derived metastatic tumors. In clinical relevance, we found that expression of POSTN was positively correlated with advanced-stage diseases and poor overall survival of patients. CONCLUSIONS Our study revealed a POSTN-integrin-NF-κB-mediated signaling and its involvement in enhancing M2 macrophages and CAFs, which could potentially participate in promoting tumor growth. Our results suggest that POSTN could be a useful prognosis marker and potential therapeutic target.
Collapse
Affiliation(s)
- Sheng-Chieh Lin
- grid.254145.30000 0001 0083 6092Graduate Institute of Integrated Medicine and Chinese Medicine Research Center, China Medical University, No. 91, Hsueh-Shih Road, Taichung, 40402 Taiwan ,grid.254145.30000 0001 0083 6092Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Yi-Chu Liao
- grid.59784.370000000406229172Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Po-Ming Chen
- grid.254145.30000 0001 0083 6092Graduate Institute of Integrated Medicine and Chinese Medicine Research Center, China Medical University, No. 91, Hsueh-Shih Road, Taichung, 40402 Taiwan ,grid.452796.b0000 0004 0634 3637Research Assistant Center, Show Chwan Memorial Hospital, Changhua, Taiwan
| | - Ya-Yu Yang
- grid.59784.370000000406229172National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan
| | - Yi-Hsiang Wang
- grid.59784.370000000406229172Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli, Taiwan ,grid.38348.340000 0004 0532 0580Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan
| | - Shiao-Lin Tung
- Department of Hematology and Oncology, Ton-Yen General Hospital, Hsinchu, Taiwan ,Department of Nursing, Hsin Sheng Junior College of Medical Care and Management, Taoyuan, Taiwan
| | - Chi-Mu Chuang
- grid.278247.c0000 0004 0604 5314Department of Obstetrics and Gynecology, Taipei Veterans General Hospital, Taipei, Taiwan ,grid.260539.b0000 0001 2059 7017School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Yu-Wen Sung
- grid.254145.30000 0001 0083 6092Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan ,grid.411508.90000 0004 0572 9415Department of Obstetrics and Gynecology, China Medical University Hospital, Taichung, Taiwan
| | - Te-Hsuan Jang
- grid.59784.370000000406229172National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan ,grid.38348.340000 0004 0532 0580Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan
| | - Shuang-En Chuang
- grid.59784.370000000406229172National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan
| | - Lu-Hai Wang
- grid.254145.30000 0001 0083 6092Graduate Institute of Integrated Medicine and Chinese Medicine Research Center, China Medical University, No. 91, Hsueh-Shih Road, Taichung, 40402 Taiwan ,grid.59784.370000000406229172Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli, Taiwan ,grid.38348.340000 0004 0532 0580Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan
| |
Collapse
|
9
|
Glucocorticoid-induced microRNA-378 signaling mediates the progression of pancreatic cancer by enhancing autophagy. Cell Death Dis 2022; 13:1052. [PMID: 36535942 PMCID: PMC9763328 DOI: 10.1038/s41419-022-05503-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 12/06/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022]
Abstract
Glucocorticoids (GCs) are widely used in tumor therapy to reduce tumor growth, inflammation, edema, and other side effects. Controversially, GCs may also cause the progression of highly aggressive pancreatic ductal adenocarcinoma (PDAC). Because microRNA (miR) and autophagy signaling support the invasive growth of PDAC, we asked whether these mechanisms may be targeted by GCs. Six established human PDAC cell lines, tissue from patients who received GC medication (n = 35) prior to surgery, or not (n = 35), and tumor xenografts were examined by RT‒qPCR, transmission electron microscopy (TEM), monodansylcadaverine (MDC) staining, immunohistochemistry, in situ hybridization, gene array and Kaplan‒Meier analysis with bioinformatics, and MTT, western blot, colony, spheroid, migration, and invasion assays. We found that various GCs, including dexamethasone (DEX), induced typical features of macroautophagy with the appearance of autolysosomes, enhanced LC3-II, decreased SQSTM1/p62 expression and induced epithelial-mesenchymal transition (EMT) and gemcitabine resistance. The GC receptor (GR) antagonist mifepristone (RU486) counteracted DEX-induced autophagy features, suggesting that the GC-GR complex is involved in the induction of autophagy. The autophagy-related miR-378i and miR-378a-3p were selected as the top upregulated candidates, and their high expression in PDAC patient tissue correlated with low survival. siRNA-mediated downregulation of miR-378 inhibited DEX-induced autophagy, and tumor progression. Bioinformatics confirmed the contribution of miR-378 to the regulation of signaling networks involved in GC-induced autophagy and tumor progression. The construction of a molecular docking model revealed stable binding of miR-378 to the DEX-GR complex, suggesting direct regulation. These substantial, novel, in-depth data reveal that GCs favor autophagy-mediated cancer progression by inducing miR-378 and GR binding and implicate GR and miR-378 as new therapeutic targets.
Collapse
|
10
|
Carvalho de Oliveira J, Mathias C, Oliveira VC, Pezuk JA, Brassesco MS. The Double Face of miR-708: A Pan-Cancer Player with Dissociative Identity Disorder. Genes (Basel) 2022; 13:genes13122375. [PMID: 36553642 PMCID: PMC9777992 DOI: 10.3390/genes13122375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 12/08/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Over the last decades, accumulating evidence has shown tumor-dependent profiles of miR-708, being either up- or downregulated, and thus, acting as a "Janus" regulator of oncogenic pathways. Herein, its functional duality was assessed through a thorough review of the literature and further validation in silico using The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. In the literature, miR-708 was found with an oncogenic role in eight tumor types, while a suppressor tumor role was described in seven cancers. This double profile was also found in TCGA and GEO databases, with some tumor types having a high expression of miR-708 and others with low expression compared with non-tumor counterparts. The investigation of validated targets using miRBase, miRTarBase, and miRecords platforms, identified a total of 572 genes that appeared enriched for PI3K-Akt signaling, followed by cell cycle control, p53, Apellin and Hippo signaling, endocrine resistance, focal adhesion, and cell senescence regulations, which are all recognized contributors of tumoral phenotypes. Among these targets, a set of 15 genes shared by at least two platforms was identified, most of which have important roles in cancer cells that influence either tumor suppression or progression. In a clinical scenario, miR-708 has shown to be a good diagnostic and prognosis marker. However, its multitarget nature and opposing roles in diverse human tumors, aligned with insufficient experimental data and the lack of proper delivery strategies, hamper its potential as a sequence-directed therapeutic.
Collapse
Affiliation(s)
| | - Carolina Mathias
- Department of Genetics, Federal University of Paraná, Curitiba 80060-000, Brazil
- Laboratory of Applied Science and Technology in Health, Carlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz), Curitiba 81350-010, Brazil
| | - Verônica Cristina Oliveira
- Department of Biotechnology and Health Innovation, Anhanguera University of São Paulo, Pirituba 05145-200, Brazil
| | - Julia Alejandra Pezuk
- Department of Biotechnology and Health Innovation, Anhanguera University of São Paulo, Pirituba 05145-200, Brazil
| | - María Sol Brassesco
- Biology Department, Faculty of Philosophy, Sciences and Letters at Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-901, Brazil
- Correspondence:
| |
Collapse
|
11
|
Kilanowska A, Ziółkowska A, Stasiak P, Gibas-Dorna M. cAMP-Dependent Signaling and Ovarian Cancer. Cells 2022; 11:cells11233835. [PMID: 36497095 PMCID: PMC9738761 DOI: 10.3390/cells11233835] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 11/23/2022] [Accepted: 11/24/2022] [Indexed: 12/02/2022] Open
Abstract
cAMP-dependent pathway is one of the most significant signaling cascades in healthy and neoplastic ovarian cells. Working through its major effector proteins-PKA and EPAC-it regulates gene expression and many cellular functions. PKA promotes the phosphorylation of cAMP response element-binding protein (CREB) which mediates gene transcription, cell migration, mitochondrial homeostasis, cell proliferation, and death. EPAC, on the other hand, is involved in cell adhesion, binding, differentiation, and interaction between cell junctions. Ovarian cancer growth and metabolism largely depend on changes in the signal processing of the cAMP-PKA-CREB axis, often associated with neoplastic transformation, metastasis, proliferation, and inhibition of apoptosis. In addition, the intracellular level of cAMP also determines the course of other pathways including AKT, ERK, MAPK, and mTOR, that are hypo- or hyperactivated among patients with ovarian neoplasm. With this review, we summarize the current findings on cAMP signaling in the ovary and its association with carcinogenesis, multiplication, metastasis, and survival of cancer cells. Additionally, we indicate that targeting particular stages of cAMP-dependent processes might provide promising therapeutic opportunities for the effective management of patients with ovarian cancer.
Collapse
Affiliation(s)
- Agnieszka Kilanowska
- Department of Anatomy and Histology, Collegium Medicum, University of Zielona Gora, 65-046 Zielona Gora, Poland
- Correspondence: ; Tel.: +48-683-283-148
| | - Agnieszka Ziółkowska
- Department of Anatomy and Histology, Collegium Medicum, University of Zielona Gora, 65-046 Zielona Gora, Poland
| | - Piotr Stasiak
- Department of Anatomy and Histology, Collegium Medicum, University of Zielona Gora, 65-046 Zielona Gora, Poland
| | - Magdalena Gibas-Dorna
- Department of Anatomy and Histology, Collegium Medicum, University of Zielona Gora, 65-046 Zielona Gora, Poland
| |
Collapse
|
12
|
Yadav V, Sharma K, Bhattacharya S, Talwar P, Purohit PK, Saini N. RETRACTED: hsa-miR-23a~27a~24-2 cluster members inhibit aggressiveness of breast cancer cells by commonly targeting NCOA1, NLK and RAP1B. Life Sci 2022; 307:120906. [PMID: 36007610 DOI: 10.1016/j.lfs.2022.120906] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 08/10/2022] [Accepted: 08/18/2022] [Indexed: 11/28/2022]
Abstract
This article has been retracted: please see Elsevier Policy on Article Withdrawal (http://www.elsevier.com/locate/withdrawalpolicy). This article has been retracted at the request of the Editor-in-Chief. The corresponding author notified the journal of three examples of image duplication within the published article (two in Figure 3D and one in Figure 4A), and requested a corrigendum. As per journal policy when considering corrigendum requests, the journal requested the authors to provide source data relating to these affected figures. The editorial team noticed 12 additional suspected image duplications within the supplied source data and the corresponding author was informed. Upon submission of revised source data, the editorial team noticed two new suspected image duplications. The editorial team have concerns about the provenance of the data and therefore the Editor-in-Chief decided to retract the article.
Collapse
Affiliation(s)
- Vikas Yadav
- Functional Genomics Unit, CSIR-Institute of Genomics and Integrative Biology (IGIB), Mall Road, 110007, Delhi, India
| | - Kritika Sharma
- Functional Genomics Unit, CSIR-Institute of Genomics and Integrative Biology (IGIB), Mall Road, 110007, Delhi, India
| | - Sushant Bhattacharya
- Functional Genomics Unit, CSIR-Institute of Genomics and Integrative Biology (IGIB), Mall Road, 110007, Delhi, India
| | - Puneet Talwar
- Institute of Human Behaviour & Allied Sciences (IHBAS), Delhi, India
| | - Paresh Kumar Purohit
- Functional Genomics Unit, CSIR-Institute of Genomics and Integrative Biology (IGIB), Mall Road, 110007, Delhi, India; Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Neeru Saini
- Functional Genomics Unit, CSIR-Institute of Genomics and Integrative Biology (IGIB), Mall Road, 110007, Delhi, India; Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
13
|
Chen S, Li QH, Chen X, Bao HJ, Wu W, Shen F, Lu BF, Jiang RQ, Zong ZH, Zhao Y. SNORA70E promotes the occurrence and development of ovarian cancer through pseudouridylation modification of RAP1B and alternative splicing of PARPBP. J Cell Mol Med 2022; 26:5150-5164. [PMID: 36056690 PMCID: PMC9575132 DOI: 10.1111/jcmm.17540] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 08/03/2022] [Accepted: 08/26/2022] [Indexed: 11/27/2022] Open
Abstract
The present study demonstrated for the first time that SNORA70E, which belongs to box H/ACA small nucleolar noncoding RNAs (snoRNAs) who could bind and induce pseudouridylation of RNAs, was significantly elevated in ovarian cancer tissues and was an unfavourable prognostic factor of ovarian cancer. The over‐expression of SNORA70E showed increased cell proliferation, invasion and migration in vitro and induced tumour growth in vivo. Further research found that SNORA70E regulates RAS‐Related Protein 1B (RAP1B) mRNA through pseudouracil modification by combing with the pyrimidine synthase Dyskerin Pseudouridine Synthase 1 (DKC1) and increase RAP1B protein level. What's more, the silencing of DKC1/RAP1B in SNORA70E overexpression cells both inhibited cell proliferation, migration and invasion through reducing β‐catenin, PI3K, AKT1, mTOR, and MMP9 protein levels. Besides, RNA‐Seq results revealed that SNORA70E regulates the alternative splicing of PARP‐1 binding protein (PARPBP), leading to the 4th exon‐skipping in PARPBP‐88, forming a new transcript PARPBP‐15, which promoted cell invasion, migration and proliferation. Finally, ASO‐mediated silencing of SNORA70E could inhibit ovarian cancer cell proliferation, invasion, migration ability in vitro and inhibit tumorigenicity in vivo. In conclusion, SNORA70E promotes the occurrence and development of ovarian cancer through pseudouridylation modification of RAP1B and alternative splicing of PARPBP. Our results demonstrated that SNORA70E may be a new diagnostic and therapeutic target for ovarian cancer.
Collapse
Affiliation(s)
- Shuo Chen
- Department of Obstetrics and Gynecology, Department of Gynecologic Oncology Research Office, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qian-Hui Li
- Department of Obstetrics and Gynecology, Department of Gynecologic Oncology Research Office, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xi Chen
- Department of Obstetrics and Gynecology, Department of Gynecologic Oncology Research Office, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Hai-Juan Bao
- Department of Obstetrics and Gynecology, Department of Gynecologic Oncology Research Office, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wu Wu
- Department of Obstetrics and Gynecology, Department of Gynecologic Oncology Research Office, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Fan Shen
- Department of Obstetrics and Gynecology, Department of Gynecologic Oncology Research Office, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Bing-Feng Lu
- Department of Obstetrics and Gynecology, Department of Gynecologic Oncology Research Office, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ru-Qi Jiang
- Department of Obstetrics and Gynecology, Department of Gynecologic Oncology Research Office, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhi-Hong Zong
- Department of Obstetrics and Gynecology, Department of Gynecologic Oncology Research Office, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yang Zhao
- Department of Obstetrics and Gynecology, Department of Gynecologic Oncology Research Office, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
14
|
Differentially Expressed microRNAs in Peritoneal Dialysis Effluent-Derived Exosomes from the Patients with Ultrafiltration Failure. Genet Res (Camb) 2022; 2022:2276175. [PMID: 36101746 PMCID: PMC9452989 DOI: 10.1155/2022/2276175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 08/01/2022] [Accepted: 08/12/2022] [Indexed: 11/20/2022] Open
Abstract
Background Ultrafiltration failure remains one of the most severe complications of long-term peritoneal dialysis (PD), which results in death. This study aimed to characterize the circulating exosomal microRNA (miRNA) profiles associated with ultrafiltration failure and explore its underlying mechanisms. Methods Exosomes were isolated from the peritoneal dialysis effluent (PDE) of patients with ultrafiltration failure or success using the ultracentrifugation method, and then transmission electron microscopy (TEM), nanoparticle tracking analysis (NTA), and western blot were used for exosome characterization. After that, the isolated exosomes were sent for small RNA sequencing, and eight differentially expressed miRNAs (DE-miRNAs) were chosen for RT-qPCR validation. Results TEM, NTA, and western blot revealed that exosomes were successfully isolated. After sequencing, 70 DE-miRNAs involved in ultrafiltration were identified, including 41 upregulated ones and 29 downregulated ones. Functional analyses revealed that these DE-miRNAs were significantly enriched in pathways of cancer, ubiquitin-mediated proteolysis, axon orientation, and the Rap1 and Ras signaling pathways. In addition, the consistency rate of RT-qPCR and sequencing results was 75%, which indicated the relatively high reliability of the sequencing data. Conclusions Our findings implied that these DE-miRNAs may be potential biomarkers of ultrafiltration failure, which would help us to discover novel therapeutic targets/pathways for ultrafiltration failure in patients with end-stage renal disease.
Collapse
|
15
|
Cammarata G, Barraco N, Giusti I, Gristina V, Dolo V, Taverna S. Extracellular Vesicles-ceRNAs as Ovarian Cancer Biomarkers: Looking into circRNA-miRNA-mRNA Code. Cancers (Basel) 2022; 14:cancers14143404. [PMID: 35884464 PMCID: PMC9324482 DOI: 10.3390/cancers14143404] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/09/2022] [Accepted: 07/12/2022] [Indexed: 02/08/2023] Open
Abstract
Simple Summary Patients with ovarian cancer have a very poor chance of long-term survival, usually due to advanced disease at the time of diagnosis. Emerging evidence suggests that extracellular vesicles contain noncoding RNAs such as microRNAs, piwiRNAs, circular RNAs, and long noncoding RNAs, with regulatory effects on ovarian cancer. In this review, we focus on ovarian cancer-associated circular RNA shuttled by extracellular vesicles as mediators of cancer progression and novel biomarkers in liquid biopsy. We propose a circular-RNA–microRNA-mRNA code that can reveal the regulatory network created by extracellular vesicles, noncoding RNAs, and mRNAs in ovarian cancer. Future research in this field will help to identify novel diagnostic biomarkers and druggable therapeutic targets, which will ultimately benefit patients. Abstract Ovarian cancer (OC) is one of the most lethal gynecologic malignancies in females worldwide. OC is frequently diagnosed at an advanced stage due to a lack of specific symptoms and effective screening tests, resulting in a poor prognosis for patients. Age, genetic alterations, and family history are the major risk factors for OC pathogenesis. Understanding the molecular mechanisms underlying OC progression, identifying new biomarkers for early detection, and discovering potential targets for new drugs are urgent needs. Liquid biopsy (LB), used for cancer detection and management, consists of a minimally invasive approach and practical alternative source to investigate tumor alterations by testing extracellular vesicles (EVs), circulating tumor cells, tumor-educated platelets, and cell-free nucleic acids. EVs are nanosize vesicles shuttling proteins, lipids, and nucleic acids, such as DNA, RNA, and non-coding RNAs (ncRNAs), that can induce phenotypic reprogramming of target cells. EVs are natural intercellular shuttles for ncRNAs, such as microRNAs (miRNAs) and circular-RNAs (circRNAs), known to have regulatory effects in OC. Here we focus on the involvement of circRNAs and miRNAs in OC cancer progression. The circRNA-microRNA-mRNA axis has been investigated with Circbank and miRwalk analysis, unraveling the intricate and detailed regulatory network created by EVs, ncRNAs, and mRNAs in OC.
Collapse
Affiliation(s)
- Giuseppe Cammarata
- Institute of Translational Pharmacology (IFT), National Research Council of Italy (CNR), 90146 Palermo, Italy
- Correspondence: (G.C.); (S.T.)
| | - Nadia Barraco
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy; (N.B.); (V.G.)
| | - Ilaria Giusti
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (I.G.); (V.D.)
| | - Valerio Gristina
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy; (N.B.); (V.G.)
| | - Vincenza Dolo
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (I.G.); (V.D.)
| | - Simona Taverna
- Institute of Translational Pharmacology (IFT), National Research Council of Italy (CNR), 90146 Palermo, Italy
- Correspondence: (G.C.); (S.T.)
| |
Collapse
|
16
|
Mousavi SM, Derakhshan M, Baharloii F, Dashti F, Mirazimi SMA, Mahjoubin-Tehran M, Hosseindoost S, Goleij P, Rahimian N, Hamblin MR, Mirzaei H. Non-coding RNAs and glioblastoma: Insight into their roles in metastasis. Mol Ther Oncolytics 2022; 24:262-287. [PMID: 35071748 PMCID: PMC8762369 DOI: 10.1016/j.omto.2021.12.015] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Glioma, also known as glioblastoma multiforme (GBM), is the most prevalent and most lethal primary brain tumor in adults. Gliomas are highly invasive tumors with the highest death rate among all primary brain malignancies. Metastasis occurs as the tumor cells spread from the site of origin to another site in the brain. Metastasis is a multifactorial process, which depends on alterations in metabolism, genetic mutations, and the cancer microenvironment. During recent years, the scientific study of non-coding RNAs (ncRNAs) has led to new insight into the molecular mechanisms involved in glioma. Many studies have reported that ncRNAs play major roles in many biological procedures connected with the development and progression of glioma. Long ncRNAs (lncRNAs), microRNAs (miRNAs), and circular RNAs (circRNAs) are all types of ncRNAs, which are commonly dysregulated in GBM. Dysregulation of ncRNAs can facilitate the invasion and metastasis of glioma. The present review highlights some ncRNAs that have been associated with metastasis in GBM. miRNAs, circRNAs, and lncRNAs are discussed in detail with respect to their relevant signaling pathways involved in metastasis.
Collapse
Affiliation(s)
- Seyed Mojtaba Mousavi
- Department of Neurosciences and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Derakhshan
- Department of Pathology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Fatereh Baharloii
- Department of Cardiology, Chamran Cardiovascular Research Education Hospital, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Fatemeh Dashti
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Seyed Mohammad Ali Mirazimi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Maryam Mahjoubin-Tehran
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Saereh Hosseindoost
- Brain and Spinal Cord Research Center, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Pouya Goleij
- Department of Genetics, Faculty of Biology, Sana Institute of Higher Education, Sari, Iran
| | - Neda Rahimian
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences (IUMS), Tehran, Iran
- Department of Internal Medicine, Firoozgar Hospital, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Michael R. Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa
- Radiation Biology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
17
|
Kalfeist L, Galland L, Ledys F, Ghiringhelli F, Limagne E, Ladoire S. Impact of Glucocorticoid Use in Oncology in the Immunotherapy Era. Cells 2022; 11:770. [PMID: 35269392 PMCID: PMC8909189 DOI: 10.3390/cells11050770] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/18/2022] [Accepted: 02/18/2022] [Indexed: 12/11/2022] Open
Abstract
Thanks to their anti-inflammatory, anti-oedema, and anti-allergy properties, glucocorticoids are among the most widely prescribed drugs in patients with cancer. The indications for glucocorticoid use are very wide and varied in the context of cancer and include the symptomatic management of cancer-related symptoms (compression, pain, oedema, altered general state) but also prevention or treatment of common side effects of anti-cancer therapies (nausea, allergies, etc.) or immune-related adverse events (irAE). In this review, we first give an overview of the different clinical situations where glucocorticoids are used in oncology. Next, we describe the current state of knowledge regarding the effects of these molecules on immune response, in particular anti-tumour response, and we summarize available data evaluating how these effects may interfere with the efficacy of immunotherapy using immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Laura Kalfeist
- Platform of Transfer in Cancer Biology, Georges-Francois Leclerc Center, 21000 Dijon, France; (L.K.); (L.G.); (F.L.); (F.G.); (E.L.)
- UMR INSERM 1231 “Lipides Nutrition Cancer”, 21000 Dijon, France
| | - Loïck Galland
- Platform of Transfer in Cancer Biology, Georges-Francois Leclerc Center, 21000 Dijon, France; (L.K.); (L.G.); (F.L.); (F.G.); (E.L.)
- UMR INSERM 1231 “Lipides Nutrition Cancer”, 21000 Dijon, France
- Department of Medical Oncology, Georges-François Leclerc Center, 21000 Dijon, France
| | - Fanny Ledys
- Platform of Transfer in Cancer Biology, Georges-Francois Leclerc Center, 21000 Dijon, France; (L.K.); (L.G.); (F.L.); (F.G.); (E.L.)
- UMR INSERM 1231 “Lipides Nutrition Cancer”, 21000 Dijon, France
| | - François Ghiringhelli
- Platform of Transfer in Cancer Biology, Georges-Francois Leclerc Center, 21000 Dijon, France; (L.K.); (L.G.); (F.L.); (F.G.); (E.L.)
- UMR INSERM 1231 “Lipides Nutrition Cancer”, 21000 Dijon, France
- Department of Medical Oncology, Georges-François Leclerc Center, 21000 Dijon, France
- School of Medicine, University of Burgundy Franche-Comté, 21000 Dijon, France
| | - Emeric Limagne
- Platform of Transfer in Cancer Biology, Georges-Francois Leclerc Center, 21000 Dijon, France; (L.K.); (L.G.); (F.L.); (F.G.); (E.L.)
- UMR INSERM 1231 “Lipides Nutrition Cancer”, 21000 Dijon, France
| | - Sylvain Ladoire
- Platform of Transfer in Cancer Biology, Georges-Francois Leclerc Center, 21000 Dijon, France; (L.K.); (L.G.); (F.L.); (F.G.); (E.L.)
- UMR INSERM 1231 “Lipides Nutrition Cancer”, 21000 Dijon, France
- Department of Medical Oncology, Georges-François Leclerc Center, 21000 Dijon, France
- School of Medicine, University of Burgundy Franche-Comté, 21000 Dijon, France
| |
Collapse
|
18
|
Role and Involvement of TENM4 and miR-708 in Breast Cancer Development and Therapy. Cells 2022; 11:cells11010172. [PMID: 35011736 PMCID: PMC8750459 DOI: 10.3390/cells11010172] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/31/2021] [Accepted: 01/02/2022] [Indexed: 12/20/2022] Open
Abstract
Teneurin 4 (TENM4) is a transmembrane protein that is codified by the ODZ4 gene and is involved in nervous system development, neurite outgrowth, and neuronal differentiation. In line with its involvement in the nervous system, TENM4 has also been implicated in several mental disorders such as bipolar disorder, schizophrenia, and autism. TENM4 mutations and rearrangements have recently been identified in a number of tumors. This, combined with impaired expression in tumors, suggests that it may potentially be involved in tumorigenesis. Most of the TENM4 mutations that are observed in tumors occur in breast cancer, in which TENM4 plays a role in cells’ migration and stemness. However, the functional role that TENM4 plays in breast cancer still needs to be better evaluated, and further studies are required to better understand the involvement of TENM4 in breast cancer progression. Herein, we review the currently available data for TENM4′s role in breast cancer and propose its use as both a novel target with which to ameliorate patient prognosis and as a potential biomarker. Moreover, we also report data on the tumorigenic role of miR-708 deregulation and the possible use of this miRNA as a novel therapeutic molecule, as miR-708 is spliced out from TENM4 mRNA.
Collapse
|
19
|
Hu Q, Gao M, Zhang D, Leng B, Wang J, Liu Q, He S, Zhi W, Zhou Z. De novo assembly and transcriptome characterization: Novel insights into the mechanisms of primary ovarian cancer in Microtus fortis. Mol Med Rep 2021; 25:64. [PMID: 34958106 PMCID: PMC8767550 DOI: 10.3892/mmr.2021.12580] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 09/02/2021] [Indexed: 11/30/2022] Open
Abstract
The natural incidence of primary epithelial ovarian cancer (OVC) in adult female voles of some established strains of Microtus fortis is relatively high. M. fortis OVC has some pathological similarities to human epithelial OVC, therefore M. fortis represents the latest and most valuable animal model for studying human OVC. The lack of available genetic information for M. fortis limits the use of common immunological methods; thus, high-throughput sequencing technologies have been used to reveal the mechanisms of primary OVC in M. fortis. The individuals with cancer were diagnosed using histopathologic hematoxylin and eosin staining. The present study used RNA-sequencing (RNA-seq) technology to establish a de novo assembly of the M. fortis transcriptome produced 339,830 unigenes by the short reads assembly program Trinity. Comparisons were made between OVC and healthy ovarian tissue (OV) and between fallopian tube cancer (FTC) and healthy fallopian tube (FT) tissues using RNA-seq analysis. A total of 3,434 differentially expressed genes (DEGs) were identified in OVC tissue compared with OV tissue using RNA-Seq by Expectation-Maximization software, including 1,950 significantly upregulated and 1,484 significantly downregulated genes. There were 2,817 DEGs identified in the FTC tissues compared with the FT tissue, including 1,762 significantly upregulated and 1,055 significantly downregulated genes. Pathway enrichment analysis revealed that upregulated transcripts in the OVC vs. OV groups were involved in cell growth and proliferation-associated pathways, whereas the downregulated DEGS in the OVC vs. OV groups were enriched in steroid biosynthesis-related pathways. Furthermore, the tumor suppressor gene, p53, was downregulated in the FTC and OVC compared with the FT and OV groups, respectively; whereas, genes that promoted cell migration, such as Ras-related protein Rap-1b, Ras homolog family member A and RAC1, were upregulated. In summary, to the best of our knowledge, the present study characterized the M. fortis de novo transcriptome of OV and FT tissues and to perform RNA-seq quantification to analyze the differences in healthy and cancerous OV and FT tissues. These results identified pathways that differed between cancerous and healthy M. fortis tissues. Analysis of these pathways may help to reveal the pathogenesis of primary OVC in M. fortis in future work.
Collapse
Affiliation(s)
- Qi Hu
- Department of Laboratory Animal Science, Xiangya Medical College, Central South University, Changsha, Hunan 410013, P.R. China
| | - Mingyue Gao
- Department of Bioinformatics Center, NEOMICS Institute, Shenzhen, Guangdong 518118, P.R. China
| | - Du Zhang
- Department of Bioinformatics Center, NEOMICS Institute, Shenzhen, Guangdong 518118, P.R. China
| | - Bingfeng Leng
- Department of Bioinformatics Center, NEOMICS Institute, Shenzhen, Guangdong 518118, P.R. China
| | - Junwen Wang
- Department of Bioinformatics Center, NEOMICS Institute, Shenzhen, Guangdong 518118, P.R. China
| | - Qian Liu
- Department of Laboratory Animal Science, Xiangya Medical College, Central South University, Changsha, Hunan 410013, P.R. China
| | - Shuangyan He
- Department of Laboratory Animal Science, Xiangya Medical College, Central South University, Changsha, Hunan 410013, P.R. China
| | - Wenling Zhi
- Department of Laboratory Animal Science, Xiangya Medical College, Central South University, Changsha, Hunan 410013, P.R. China
| | - Zhijun Zhou
- Department of Laboratory Animal Science, Xiangya Medical College, Central South University, Changsha, Hunan 410013, P.R. China
| |
Collapse
|
20
|
Li AL, Chou CY, Chen CL, Wu KL, Lin SC, Chen HC, Wang MC, Chang CC, Hsu BG, Wu MS, Ma N, Huang CC. The MicroRNA Prediction Models as Ancillary Diagnosis Biomarkers for Urothelial Carcinoma in Patients With Chronic Kidney Disease. Front Med (Lausanne) 2021; 8:726214. [PMID: 34660637 PMCID: PMC8517232 DOI: 10.3389/fmed.2021.726214] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 09/07/2021] [Indexed: 12/24/2022] Open
Abstract
Urothelial carcinoma is a common urological cancer in chronic kidney disease patients. Cystoscopy and urine cytology are the clinical diagnostic tools for UC. However, cystoscopy is an invasive procedure, while urine cytology showed low sensitivity for low-grade urothelial tumors. High accuracy with non-invasive tools for UC is needed for CKD patients. Our study collected a total of 272 urine and 138 plasma samples to detect the miRNA expression levels for establishing UC signatures from CKD patients. Seventeen candidate miRNAs of biofluids were selected and confirmed by qRT-PCR. Our results showed that urinary miR-1274a and miR-30a-5p expression levels were significantly lower but miR-19a-5p expression levels were higher in UC when compared with CKD. In plasma samples, miR-155-5p, miR-19b-1-5p, miR-378, and miR-636 showed significantly lower expression in UC compared to those with CKD. The Kaplan-Meier curve showed that lower expression of miR-19a, miR-19b, miR-636 and miR-378, and higher expression of miR-708-5p were associated with poor prognosis in patients with bladder cancer. In addition, we produced classifiers for predicting UC by multiple logistic regression. The urine signature was developed with four miRNAs, and the AUC was 0.8211. Eight miRNA expression levels from both urine and plasma samples were examined, and the AUC was 0.8595. Two miRNA classifiers and the nomograms could improve the drawbacks of current UC biomarker screenings for patients with CKD.
Collapse
Affiliation(s)
- An-Lun Li
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan, Taiwan
| | - Che-Yi Chou
- Division of Nephrology, Department of Internal Medicine, Asia University Hospital, Taichung, Taiwan
| | - Chien-Lung Chen
- Department of Nephrology, Landseed International Hospital, Taoyuan, Taiwan
| | - Kun-Lin Wu
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan, Taiwan.,Division of Nephrology, Department of Internal Medicine, Taoyuan Armed Forces General Hospital, Taoyuan, Taiwan
| | - Shih-Chieh Lin
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Hung-Chun Chen
- Division of Nephrology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ming-Cheng Wang
- Division of Nephrology, Cheng Kung University Hospital, Tainan, Taiwan
| | - Chia-Chu Chang
- Division of Nephrology, Department of Internal Medicine, Kuang Tien General Hospital, Taichung, Taiwan.,Department of Nutrition, Hungkuang University, Taichung, Taiwan
| | - Bang-Gee Hsu
- Division of Nephrology, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | - Mai-Szu Wu
- Division of Nephrology, Taipei Medical University and Hospitals, Taipei, Taiwan
| | - Nianhan Ma
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan, Taiwan
| | - Chiu-Ching Huang
- Division of Nephrology and The Kidney Institute, China Medical University and Hospitals, Taichung, Taiwan
| |
Collapse
|
21
|
Pu J, Zhou X, Liu J, Hou P, Ji M. Therapeutic potential and deleterious effect of glucocorticoids on azoxymethane/dextran sulfate sodium-induced colorectal cancer in mice. Am J Cancer Res 2021; 11:4866-4883. [PMID: 34765297 PMCID: PMC8569368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 09/15/2021] [Indexed: 06/13/2023] Open
Abstract
Glucocorticoids (GCs) are widely used in the treatment of various autoimmune and inflammatory diseases, including inflammatory bowel disease (IBD). However, the effect of GCs on the progression of colitis-associated colorectal cancer (CAC) has not been well explored. In this study, we first established a colorectal cancer model induced by azoxymethane and dextran sulfate sodium (AOM/DSS) and a colitis model induced by DSS in mice. Dexamethasone (DEX) was then administered at different periods of time to determine its effect on tumorigenesis and tumor progression. Meanwhile, body weight, stool property and fecal blood of mice were recorded. At the end of this study, the number and load of tumors were evaluated, and the expression of proteins associated with cell proliferation was analyzed. To evaluate the inflammation in colon, we detected the level of pro-inflammatory cytokine TNFα, and the mucosal infiltration of inflammatory cells. Our results revealed that AOM injection followed by three cycles of drinking water containing 1.5% DSS successfully induced multiple tumor formation in mouse colon and rectum. Both early and late DEX intervention suppressed tumor growth in mouse colorectum, and downregulated the expression of PCNA and cyclin D1. Moreover, DEX treatment significantly inhibited TNFα production, mucosal infiltration of inflammatory cells, and the activity of MAPK/JNK pathway, particularly early DEX intervention. However, we also found that DEX treatment deteriorated the general state of mouse manifested by greater loss of body weight and rectal bleeding. In summary, both early and late DEX interventions significantly ameliorate colonic inflammation and inhibit the progression of AOM/DSS-induced colorectal cancer, at least partly due to the inhibition of MAPK/JNK pathway. It is noteworthy that the deleterious effect on the general condition of mouse may limit the duration of GCs treatment.
Collapse
Affiliation(s)
- Jun Pu
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi’an Jiaotong UniversityXi’an 710061, Shaanxi, China
| | - Xinrui Zhou
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi’an Jiaotong UniversityXi’an 710061, Shaanxi, China
| | - Jiaxin Liu
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi’an Jiaotong UniversityXi’an 710061, Shaanxi, China
| | - Peng Hou
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi’an Jiaotong UniversityXi’an 710061, Shaanxi, China
- Department of Endocrinology, The First Affiliated Hospital of Xi’an Jiaotong UniversityXi’an 710061, Shaanxi, China
| | - Meiju Ji
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi’an Jiaotong UniversityXi’an 710061, Shaanxi, China
- Center for Translational Medicine, The First Affiliated Hospital of Xi’an Jiaotong UniversityXi’an 710061, Shaanxi, China
| |
Collapse
|
22
|
Zhang Z, Wang R, Luo R, Zhu J, Huang X, Liu W, Liu F, Feng F, Qu W. An Activatable Theranostic Nanoprobe for Dual-Modal Imaging-Guided Photodynamic Therapy with Self-Reporting of Sensitizer Activation and Therapeutic Effect. ACS NANO 2021; 15:5366-5383. [PMID: 33705106 DOI: 10.1021/acsnano.0c10916] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Intelligent systems that offer traceable cancer therapy are highly desirable for precision medicine. Although photodynamic therapy (PDT) has been approved in the clinic for decades, determining where the tumor is, when to irradiate, and how long to expose to light still confuse the clinicians. Patients are always suffering from the phototoxicity of the photosensitizer in nonmalignant tissues. Herein, an activatable theranostic agent, ZnPc@TPCB nanoparticles (NPs), is prepared by doping a photosensitizer, ZnPc, with an aggregation-induced emission probe, TPCB. The assembled or disassembled ZnPc@TPCB NPs in various phases have behaved differently in fluorescence intensity, photoacoustic (PA) signals, and PDT efficiency. The intact nanoparticles are non-emissive in aqueous media while showing strong PA signals and low PDT efficiency, which can eliminate the phototoxicity and self-monitor their distribution and image the tumors' location. Disassembling of the NPs leads to the release of ZnPc and its red fluorescence turn-on to self-report the photosensitizer's activation. Upon light irradiation, the reactive oxygen species (ROS) generated by ZnPc can induce cell apoptosis and activate the ROS sensor, TPCB, which will yield intense orange-red fluorescence and instantly predict the therapeutic effect. Moreover, enhanced PDT efficacy is achieved via the GSH-depleting adjuvant quinone methide produced by the activated TPCB. The well-designed ZnPc@TPCB NPs have shown promising potential for finely controlled PDT with good biosafety and broad application prospects in individual therapy, which may inspire the development of precision medicine.
Collapse
Affiliation(s)
- Zhongtao Zhang
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Ruyi Wang
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Renjie Luo
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing 211198, China
| | - Jiaxin Zhu
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Xiaoxian Huang
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Wenyuan Liu
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing 211198, China
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing 211198, China
| | - Fulei Liu
- The Joint Laboratory of China Pharmaceutical University and Taian City Central Hospital, Taian City Central Hospital, Taian 271000, China
- Pharmaceutical Department, Taian City Central Hospital, Taian 271000, China
| | - Feng Feng
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China
- Jiangsu Food and Pharmaceutical Science College, Huaian 223003, China
| | - Wei Qu
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China
- Key Laboratory of Biomedical Functional Materials, China Pharmaceutical University, Nanjing 211198, China
| |
Collapse
|
23
|
Molecular and Pharmacological Characterization of the Interaction between Human Geranylgeranyltransferase Type I and Ras-Related Protein Rap1B. Int J Mol Sci 2021; 22:ijms22052501. [PMID: 33801503 PMCID: PMC7958859 DOI: 10.3390/ijms22052501] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 02/20/2021] [Accepted: 02/25/2021] [Indexed: 12/30/2022] Open
Abstract
Geranylgeranyltransferase type-I (GGTase-I) represents an important drug target since it contributes to the function of many proteins that are involved in tumor development and metastasis. This led to the development of GGTase-I inhibitors as anti-cancer drugs blocking the protein function and membrane association of e.g., Rap subfamilies that are involved in cell differentiation and cell growth. In the present study, we developed a new NanoBiT assay to monitor the interaction of human GGTase-I and its substrate Rap1B. Different Rap1B prenylation-deficient mutants (C181G, C181S, and ΔCQLL) were designed and investigated for their interaction with GGTase-I. While the Rap1B mutants C181G and C181S still exhibited interaction with human GGTase-I, mutant ΔCQLL, lacking the entire CAAX motif (defined by a cysteine residue, two aliphatic residues, and the C-terminal residue), showed reduced interaction. Moreover, a specific, peptidomimetic and competitive CAAX inhibitor was able to block the interaction of Rap1B with GGTase-I. Furthermore, activation of both Gαs-coupled human adenosine receptors, A2A (A2AAR) and A2B (A2BAR), increased the interaction between GGTase-I and Rap1B, probably representing a way to modulate prenylation and function of Rap1B. Thus, A2AAR and A2BAR antagonists might be promising candidates for therapeutic intervention for different types of cancer that overexpress Rap1B. Finally, the NanoBiT assay provides a tool to investigate the pharmacology of GGTase-I inhibitors.
Collapse
|
24
|
Mohammadi L, Mosayyebi B, Imani M, Rahmati M. Dexamethasone Reduces Cell Adhesion and Migration of T47D Breast Cancer Cell Line. Anticancer Agents Med Chem 2020; 22:2494-2501. [PMID: 33319693 DOI: 10.2174/1871520621666201214150427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 10/16/2020] [Accepted: 10/26/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Aberrant expression of cell adhesion molecules and matrix metalloproteinase (MMPs) plays a pivotal role in tumor biological processes including progression and metastasis of cancer cells. Targeting these processes and detailed understanding of their underlying molecular mechanism is an essential step in cancer treatment. Dexamethasone (Dex) is a type of synthetic corticosteroid hormone used as adjuvant therapy in combination with current cancer treatments such as chemotherapy in order to alleviate its side effects like acute nausea and vomiting. Recent evidences have suggested that Dex may have antitumor characteristics. OBJECTIVE Dex affects the migration and adhesion of T47D breast cancer cells as well as cell adhesion molecules e.g., cadherin and integrin, and MMPs by regulating the expression levels of associated genes. METHODS In this study, we evaluated the cytotoxicity of Dex on the T47D breast cancer cell line through MTT assay. Cell adhesion assay and wound healing assay were performed to determine the impact of Dex on cell adhesion and cell migration, respectively. Moreover, real-time PCR was used to measure the levels of α and β integrin, E-cadherin, N-cadherin, MMP-2, and MMP-9. RESULTS Dex decreased the viability of T47D cells in a time and dose-dependent manner. Cell adhesion and migration of T47D cells were reduced upon Dex treatment. The expression of α and β integrin, E-cadherin, N-cadherin, MMP-2, and MMP-9 were altered in response to the Dex treatment. CONCLUSION Our findings demonstrated that Dex may have a role in the prevention of metastasis in this cell line.
Collapse
Affiliation(s)
- Leila Mohammadi
- Student Research Committee, Tabriz University of Medical Science, Tabriz. Iran
| | - Bashir Mosayyebi
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz. Iran
| | - Mahsa Imani
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz. Iran
| | - Mohammad Rahmati
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz. Iran
| |
Collapse
|
25
|
Pang JM, Huang YC, Sun SP, Pan YR, Shen CY, Kao MC, Wang RH, Wang LH, Lin KT. Effects of synthetic glucocorticoids on breast cancer progression. Steroids 2020; 164:108738. [PMID: 33065150 DOI: 10.1016/j.steroids.2020.108738] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 09/08/2020] [Accepted: 09/21/2020] [Indexed: 12/18/2022]
Abstract
Glucocorticoids (GCs) are widely prescribed as adjuvant therapy for breast cancer patients. Unlike other steroid hormone receptors, the GC receptor is not considered an oncogene. Research in the past few years has revealed the complexity of GC-mediated signaling, but it remains puzzling whether GCs promote or inhibit tumor progression in different cancer types. Here we evaluated the potential of using a synthetic GC, dexamethasone (DEX), in the treatment of breast cancer. We found that the administration of low-dose DEX suppressed tumor growth and distant metastasis in the MCF-7 and MDA-MB-231 xenograft mouse model, whereas treatment with high-dose DEX enhanced tumor growth and metastasis, respectively. Treatment of breast cancer cells with DEX inhibited cell adhesion, migration, and invasion in a dose-dependent manner. The DEX-mediated inhibition of cell adhesion, migration, and invasion is partly through induction of microRNA-708 and subsequent Rap1B-mediated signaling in MDA-MB-231 cells. On the other hand, in MCF-7 cells, DEX-suppressed cell migration is independent from microRNA-708 mediated signaling. Overall, our data reveal that DEX acts as a double-edged sword during breast-cancer progression and metastasis: Lower concentrations inhibit breast cancer tumor growth and metastasis, whereas higher concentrations may play an undesired role to promote breast cancer progression.
Collapse
Affiliation(s)
- Jia Meng Pang
- Institute of Biotechnology, College of Life Science, National Tsing Hua University, Hsinchu 300, Taiwan; Department of Medical Science, College of Life Science, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Yi-Chen Huang
- Institute of Biotechnology, College of Life Science, National Tsing Hua University, Hsinchu 300, Taiwan; Department of Life Science, College of Life Science, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Shu-Pin Sun
- Biotechnology R&D Center, National Taiwan University Hospital Hsin-Chu Branch, Hsinchu 300, Taiwan
| | - Yan-Ru Pan
- Institute of Biotechnology, College of Life Science, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Chia-Yi Shen
- Institute of Biotechnology, College of Life Science, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Ming-Chien Kao
- Institute of Biotechnology, College of Life Science, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Rong-Hsuan Wang
- Institute of Biotechnology, College of Life Science, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Lu-Hai Wang
- Chinese Medicine Research Center and Institute of Integrated Medicine, China Medical University, 91, Hsueh Road, Taichung City 40402, Taiwan
| | - Kai-Ti Lin
- Institute of Biotechnology, College of Life Science, National Tsing Hua University, Hsinchu 300, Taiwan; Department of Medical Science, College of Life Science, National Tsing Hua University, Hsinchu 300, Taiwan.
| |
Collapse
|
26
|
Slow-Freezing Cryopreservation Ensures High Ovarian Tissue Quality Followed by In Vivo and In Vitro Methods and Is Safe for Fertility Preservation. ACTA ACUST UNITED AC 2020; 56:medicina56100547. [PMID: 33086522 PMCID: PMC7603126 DOI: 10.3390/medicina56100547] [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: 08/30/2020] [Revised: 10/15/2020] [Accepted: 10/16/2020] [Indexed: 01/03/2023]
Abstract
Background and objectives: Cancer incidence is growing with younger patients diagnosed with this disease every year. Improved cancer diagnostics and treatment lead to better survival of cancer patients. However, after aggressive chemo- or radiotherapy, cancer survivors suffer from various degrees of subfertility or infertility. Several fertility preservation technologies have been developed for young cancer patients: cryopreservation of germ cells, embryos, or reproductive tissues. The best results have been shown by cryopreservation of sperm and embryos. Yet the success of using cryopreserved oocytes or reproductive tissues (ovarian and testicular) is still insufficient. Therefore, this study was designed to assess the vitality, viability, general quality, and safety of frozen-thawed human ovarian tissue for retransplantation using modern molecular tests. Materials and Methods: The new miRNA array test was used to evaluate miRNA expression in thawed ovarian tissue in combination with standard xenotransplantation and pathological examination of microslides. Results: Our results demonstrated that slow freezing is an efficient way (80%) to cryopreserve ovarian tissue with no structural damage afterwards. We have shown that xenotransplantation into immunodeficient mice, histology, and immunohistochemistry could be potentially replaced by more recent molecular methods. Conclusions: The latter method has shown that altered expression of miRNAs might be used as identifiers of normal/damaged tissue after further analysis. Newer, safer, and more specific approaches need to be developed in order to eliminate the risk of disease reoccurrence.
Collapse
|
27
|
Nguyen VHL, Yue C, Du KY, Salem M, O’Brien J, Peng C. The Role of microRNAs in Epithelial Ovarian Cancer Metastasis. Int J Mol Sci 2020; 21:ijms21197093. [PMID: 32993038 PMCID: PMC7583982 DOI: 10.3390/ijms21197093] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 09/22/2020] [Accepted: 09/23/2020] [Indexed: 12/11/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is the deadliest gynecological cancer, and the major cause of death is mainly attributed to metastasis. MicroRNAs (miRNAs) are a group of small non-coding RNAs that exert important regulatory functions in many biological processes through their effects on regulating gene expression. In most cases, miRNAs interact with the 3′ UTRs of target mRNAs to induce their degradation and suppress their translation. Aberrant expression of miRNAs has been detected in EOC tumors and/or the biological fluids of EOC patients. Such dysregulation occurs as the result of alterations in DNA copy numbers, epigenetic regulation, and miRNA biogenesis. Many studies have demonstrated that miRNAs can promote or suppress events related to EOC metastasis, such as cell migration, invasion, epithelial-to-mesenchymal transition, and interaction with the tumor microenvironment. In this review, we provide a brief overview of miRNA biogenesis and highlight some key events and regulations related to EOC metastasis. We summarize current knowledge on how miRNAs are dysregulated, focusing on those that have been reported to regulate metastasis. Furthermore, we discuss the role of miRNAs in promoting and inhibiting EOC metastasis. Finally, we point out some limitations of current findings and suggest future research directions in the field.
Collapse
Affiliation(s)
- Vu Hong Loan Nguyen
- Department of Biology, York University, Toronto, ON M3J 1P3, Canada; (V.H.L.N.); (C.Y.); (K.Y.D.); (M.S.); (J.O.)
| | - Chenyang Yue
- Department of Biology, York University, Toronto, ON M3J 1P3, Canada; (V.H.L.N.); (C.Y.); (K.Y.D.); (M.S.); (J.O.)
| | - Kevin Y. Du
- Department of Biology, York University, Toronto, ON M3J 1P3, Canada; (V.H.L.N.); (C.Y.); (K.Y.D.); (M.S.); (J.O.)
| | - Mohamed Salem
- Department of Biology, York University, Toronto, ON M3J 1P3, Canada; (V.H.L.N.); (C.Y.); (K.Y.D.); (M.S.); (J.O.)
| | - Jacob O’Brien
- Department of Biology, York University, Toronto, ON M3J 1P3, Canada; (V.H.L.N.); (C.Y.); (K.Y.D.); (M.S.); (J.O.)
| | - Chun Peng
- Department of Biology, York University, Toronto, ON M3J 1P3, Canada; (V.H.L.N.); (C.Y.); (K.Y.D.); (M.S.); (J.O.)
- Centre for Research in Biomolecular Interactions, York University, Toronto, ON M3J 1P3, Canada
- Correspondence:
| |
Collapse
|
28
|
Looi CK, Hii LW, Ngai SC, Leong CO, Mai CW. The Role of Ras-Associated Protein 1 (Rap1) in Cancer: Bad Actor or Good Player? Biomedicines 2020; 8:334. [PMID: 32906721 PMCID: PMC7555474 DOI: 10.3390/biomedicines8090334] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/02/2020] [Accepted: 09/05/2020] [Indexed: 02/05/2023] Open
Abstract
Metastasis is known as the most life-threatening event in cancer patients. In principle, the immune system can prevent tumor development. However, dysfunctional T cells may fail to eliminate the tumor cells effectively and provide additional survival advantages for tumor proliferation and metastasis. Constitutive activation of Ras-associated protein1 (Rap1) has not only led to T cell anergy, but also inhibited autophagy and supported cancer progression through various oncogenic events. Inhibition of Rap1 activity with its negative regulator, Rap1GAP, impairs tumor progression. However, active Rap1 reduces tumor invasion in some cancers, indicating that the pleiotropic effects of Rap1 signaling in cancers could be cancer-specific. All in all, targeting Rap1 signaling and its regulators could potentially control carcinogenesis, metastasis, chemoresistance and immune evasion. Rap1GAP could be a promising therapeutic target in combating cancer.
Collapse
Affiliation(s)
- Chin-King Looi
- School of Postgraduate Study, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia; (C.-K.L.); (L.-W.H.)
| | - Ling-Wei Hii
- School of Postgraduate Study, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia; (C.-K.L.); (L.-W.H.)
- School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia;
| | - Siew Ching Ngai
- School of Biosciences, Faculty of Science and Engineering, University of Nottingham Malaysia, Semenyih, Selangor 43500, Malaysia;
| | - Chee-Onn Leong
- School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia;
- Centre for Cancer and Stem Cells Research, Institute for Research, Development, and Innovation (IRDI), International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia
| | - Chun-Wai Mai
- School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia;
- Centre for Cancer and Stem Cells Research, Institute for Research, Development, and Innovation (IRDI), International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia
| |
Collapse
|
29
|
Alpha KM, Xu W, Turner CE. Paxillin family of focal adhesion adaptor proteins and regulation of cancer cell invasion. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 355:1-52. [PMID: 32859368 PMCID: PMC7737098 DOI: 10.1016/bs.ircmb.2020.05.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The paxillin family of proteins, including paxillin, Hic-5, and leupaxin, are focal adhesion adaptor/scaffolding proteins which localize to cell-matrix adhesions and are important in cell adhesion and migration of both normal and cancer cells. Historically, the role of these proteins in regulating the actin cytoskeleton through focal adhesion-mediated signaling has been well documented. However, studies in recent years have revealed additional functions in modulating the microtubule and intermediate filament cytoskeletons to affect diverse processes including cell polarization, vesicle trafficking and mechanosignaling. Expression of paxillin family proteins in stromal cells is also important in regulating tumor cell migration and invasion through non-cell autonomous effects on the extracellular matrix. Both paxillin and Hic-5 can also influence gene expression through a variety of mechanisms, while their own expression is frequently dysregulated in various cancers. Accordingly, these proteins may serve as valuable targets for novel diagnostic and treatment approaches in cancer.
Collapse
Affiliation(s)
- Kyle M Alpha
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Weiyi Xu
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Christopher E Turner
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, Syracuse, NY, United States.
| |
Collapse
|
30
|
Zhou Z, Xu H, Duan Y, Liu B. MicroRNA-101 suppresses colorectal cancer progression by negative regulation of Rap1b. Oncol Lett 2020; 20:2225-2231. [PMID: 32782539 PMCID: PMC7400857 DOI: 10.3892/ol.2020.11791] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 11/07/2019] [Indexed: 02/07/2023] Open
Abstract
Colorectal cancer (CRC) is the fourth most lethal malignancy and is the second most common cause of cancer-associated mortality worldwide. The development of high-throughput sequencing has enabled the identification of potential biomarkers for the diagnosis and treatment of various types of cancer. Although microRNA-101 (miR-101) has been demonstrated to be a potential biomarker of CRC, its detailed mechanisms remain to be fully discovered. In the present study, overall survival analysis was applied to determine the association between miR-101 and CRC prognosis. Reverse transcription-quantitative PCR (RT-qPCR) was used to examine gene expression levels in tissues and cells. Cell proliferative and apoptotic activities were determined by MTT and flow cytometry assays, respectively. Wound healing and Transwell assays were used to examine CRC cell migration and invasion, respectively. In the present study, RT-qPCR analysis indicated that miR-101 was significantly downregulated in CRC tissues and cells. However, clinical data collected from The Cancer Genome Atlas revealed no significant association between the expression levels of miR-101 and the prognosis of CRC. Additionally, miR-101 inhibited the progression of CRC by directly binding to the 3′-untranslated region of Ras-related protein Rap1b (Rap1b). This was associated with downregulation of Rap1b expression. Furthermore, the overexpression of Rap1b promoted miR-101 mimic-attenuated CRC cell progression. The present study demonstrated that miR-101 may be involved in the repression of the CRC progression by forming a negative feedback loop with Rap1b. The findings revealed the interaction between miR-101 and Rap1b during the progression of CRC, which could aid the development of therapeutic strategies.
Collapse
Affiliation(s)
- Zhiyuan Zhou
- Department of General Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200011, P.R. China
| | - Hang Xu
- Department of General Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200011, P.R. China
| | - Yantao Duan
- Department of General Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200011, P.R. China
| | - Bin Liu
- Department of General Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200011, P.R. China
| |
Collapse
|
31
|
Monteleone NJ, Lutz CS. miR-708-5p targets oncogenic prostaglandin E2 production to suppress a pro-tumorigenic phenotype in lung cancer cells. Oncotarget 2020; 11:2464-2483. [PMID: 32655834 PMCID: PMC7335672 DOI: 10.18632/oncotarget.27614] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 05/14/2020] [Indexed: 12/13/2022] Open
Abstract
Many cancers maintain an inflammatory microenvironment to promote their growth. Lung cancer is of particular importance, as it is the deadliest cancer worldwide. One inflammatory pathway commonly dysregulated in cancer is the metabolism of arachidonic acid (AA) by Cyclooxygenase-2 (COX-2) and microsomal Prostaglandin E Synthase 1 (mPGES-1) into Prostaglandin E2 (PGE2). While researchers have identified PGE2's pro-tumorigenic functions, the mechanisms governing overexpression of COX-2 and mPGES-1 are incompletely understood. MicroRNAs (miRNAs) are important post-transcriptional regulators commonly dysregulated in cancer. Interestingly, miR-708-5p (miR-708) is predicted to target both COX-2 and mPGES-1. In this study, we show that high miR-708 expression is associated with survival rates in lung squamous cell carcinoma patients. miR-708 also represses PGE2 production by suppressing both COX-2 and mPGES-1 expression in lung cancer cells. miR-708 regulation of COX-2 and mPGES-1 is mediated through targeting of their 3' untranslated regions (UTRs). Moreover, miR-708 decreases proliferation, survival, and migration of lung cancer cells, which can be partially attributed to miR-708's inhibition of PGE2 signaling. Lastly, we identify novel miR-708 predicted targets and possible regulators of miR-708 expression in lung cancer. Collectively, these data demonstrate that dysregulated miR-708 expression contributes to exacerbated PGE2 production, leading to an enhanced pro-tumorigenic phenotype in lung cancer cells.
Collapse
Affiliation(s)
- Nicholas J. Monteleone
- Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers Biomedical & Health Sciences, New Jersey Medical School, School of Graduate Studies, Newark, NJ 07103, USA
| | - Carol S. Lutz
- Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers Biomedical & Health Sciences, New Jersey Medical School, School of Graduate Studies, Newark, NJ 07103, USA
| |
Collapse
|
32
|
Abstract
Introduction: Ovarian carcinoma (OC) is the leading cause of death in women with gynecologic cancers. Most patients are diagnosed at an advanced stage with a low five-year survival rate of 20-30%. Discovering novel biomarkers for early detection and outcome prediction of OC is an urgent medical need. miRNAs, a group of small non-coding RNAs, play critical roles in multiple biologic processes and cancer pathogenesis.Areas covered: We provide an in-depth look at the functions of miRNAs in OC, particularly focusing on their roles in chemoresistance and metastasis in OC. We also discuss the biological and clinical significance of miRNAs in exosomes and expand on long non-coding RNA which acts as ceRNA of miRNAs.Expert opinion: miRNAs participate in many biological processes including proliferation, apoptosis, chemoresistance, metastasis, epithelial-mesenchymal transition, and cancer stem cell. They will substantially contribute to our understanding of OC pathogenesis. Given their resistance to the degradation of ribonucleases and availability in plasma exosomes, miRNAs may serve as emerging biomarkers for cancer detection, therapeutic assessment, and prognostic prediction. Being a messenger, exosomal miRNAs are crucial for the crosstalk between cancer cells and stromal cells in tumor microenvironment.
Collapse
Affiliation(s)
- Huilin Zhang
- Department of Surgical Pathology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Bingjian Lu
- Department of Surgical Pathology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| |
Collapse
|
33
|
Fan M, Ma X, Wang F, Zhou Z, Zhang J, Zhou D, Hong Y, Wang Y, Wang G, Dong Q. MicroRNA-30b-5p functions as a metastasis suppressor in colorectal cancer by targeting Rap1b. Cancer Lett 2020; 477:144-156. [PMID: 32112903 DOI: 10.1016/j.canlet.2020.02.021] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 01/24/2020] [Accepted: 02/14/2020] [Indexed: 12/24/2022]
Abstract
Colorectal liver metastasis (CRLM) is the leading cause of death in patients with colorectal cancer (CRC). MiR-30b-5p can function as an oncogene or tumor suppressor in cancers, but its role in CRLM is still unknown. Here, we found that miR-30b-5p overexpression suppressed the invasion, migration, adhesion, and motility of HCT116 and LoVo cells. The expression of EMT (Zeb1, Snail, and vimentin) and adhesion-related proteins (p-paxillin and p-Src) was decreased. We validated Rap1b, a Ras family small GTPase that regulates cell adhesion and mobility, as the direct and functional target of miR-30b-5p. Rap1b overexpression rescued the aggressive characteristics of CRC cells that were inhibited by miR-30b-5p. Rap1b knockdown suppressed invasion and migration and decreased CRC cell-matrix adhesion and spreading, which was consistent with the results of miR-30b-5p overexpression. Further in vivo experiments demonstrated that miR-30b-5p overexpression inhibited CRLM, but Rap1b rescue attenuated the inhibitory effect of miR-30b-5p. In addition, miR-30b-5p was downregulated in CRC specimens, and Rap1b showed a negative correlation with miR-30b-5p expression in primary CRC and LM tissues. These results indicate that miR-30b-5p functions as a metastasis suppressor by targeting Rap1b and may provide a new target for the treatment of CRLM.
Collapse
Affiliation(s)
- Mengjing Fan
- Biomedical Research Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Ximei Ma
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Feifan Wang
- Department of Urology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Zhuha Zhou
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jing Zhang
- Biomedical Research Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Difan Zhou
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yiyang Hong
- Biomedical Research Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yihong Wang
- Department of Pathology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Guanyu Wang
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Qinghua Dong
- Biomedical Research Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China; Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, China.
| |
Collapse
|
34
|
Zou X, Zhu C, Zhang L, Zhang Y, Fu F, Chen Y, Zhou J. MicroRNA-708 Suppresses Cell Proliferation and Enhances Chemosensitivity of Cervical Cancer Cells to cDDP by Negatively Targeting Timeless. Onco Targets Ther 2020; 13:225-235. [PMID: 32021269 PMCID: PMC6966141 DOI: 10.2147/ott.s227015] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 11/22/2019] [Indexed: 01/10/2023] Open
Abstract
Purpose Cervical cancer is the fourth most common cause of cancer-associated mortality in women worldwide. Previous studies have reported that microRNAs (miRNAs) are involved in multiple biological aspects of cancer progression by regulating gene expression. Here, we investigated the role of microRNA-708 (miR-708) in cervical cancer. Methods The expression levels of miR-708 in cervical cancer tissues and paired-normal cervical tissues were tested by quantitative polymerase chain reaction (qPCR). The interaction between miR-708 and Timeless was identified by bioinformatics method, dual-luciferase reporter assay, and Western blotting. The effects of over-expression of miR-708 on cell proliferation and cisplatin sensitivity were determined by Cell Counting Kit-8 (CCK-8) and colony formation assay. Cell cycle and apoptosis were analyzed by flow cytometry. DNA damage induced by over-expression of miR-708 was determined by comet assay. Expression levels of the genes involved in repair of DNA damage were analyzed by Western blotting. Results MiR-708 was down-regulated in cervical cancer tissues compared with paired-normal cervical tissues. By bioinformatics method, Western blotting, and dual-luciferase reporter assay, we found that Timeless was a direct target of miR-708. Furthermore, miR-708 suppressed cellular viability, colony formation, promoted apoptosis, and induced DNA damage levels. MiR-708 also enhanced chemosensitivity of cervical cancer cells to cDDP via impairing the ATR/CHK1 signaling pathway. Conclusion We conclude that miR-708 suppresses cell proliferation, facilitates cisplatin efficacy, and impairs DNA repair pathway in cervical cancer cells. These results demonstrate that miR-708 might be a candidate therapeutic target for future cervical cancer therapy.
Collapse
Affiliation(s)
- Xinwei Zou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China.,Clinical Research Center of Obstetrics and Gynecology, Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, People's Republic of China.,Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Chenjie Zhu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China.,Clinical Research Center of Obstetrics and Gynecology, Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, People's Republic of China.,Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Lin Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Yi Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Fengqing Fu
- Clinical Research Center of Obstetrics and Gynecology, Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, People's Republic of China.,Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Youguo Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China.,Clinical Research Center of Obstetrics and Gynecology, Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, People's Republic of China.,Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Jinhua Zhou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China.,Clinical Research Center of Obstetrics and Gynecology, Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, People's Republic of China.,Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| |
Collapse
|
35
|
Identification of microRNAs that Regulate the MAPK Pathway in Human Cumulus Cells from PCOS Women with Insulin Resistance. Reprod Sci 2020; 27:833-844. [PMID: 32046427 DOI: 10.1007/s43032-019-00086-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 09/08/2019] [Indexed: 12/14/2022]
Abstract
Polycystic ovary syndrome (PCOS) is one of the most common gynaecological endocrine disorders, and more than 60% of PCOS patients have varying degrees of insulin resistance (IR). The regulatory role of microRNAs (miRNAs) at post-transcriptional levels in human cumulus cells relating to IR in PCOS remains unclear. In this case-control study, 26 PCOS patients with IR (PCOS-IR) and 24 patients without IR (PCOS-control) were enrolled. We determined the differentially expressed miRNA and mRNA using next-generation sequencing technology, and these miRNAs and mRNAs were validated by quantitative real-time polymerase chain reaction (PCR). These miRNA regulating pathways (e.g., MAPK pathway) were analysed by bioinformatics analysis, and the Rap1b was demonstrated to be targeted by miR-612 based on quantitative real-time PCR, western blot and luciferase activity assay. A total of 59 known miRNAs and 617 differentially expressed genes were identified that differentially expressed between PCOS-IR and PCOS-control cumulus cells. Moreover, the potential regulating roles of miRNAs and their targeting genes in pathophysiology of IR and PCOS were analysed by gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway annotation, and several key processes were enriched, such as MAPK activity. Furthermore, Rap1b, a regulator of the MAPK pathway, was demonstrated to be suppressed directly by miR-612 in PCOS-IR cumulus cells based on negative expression correlation validation, dual luciferase activity assay and reduction of Rap1b expression after miR-612 mimics transfection. Our results suggested that miRNAs and their targeted pathways in ovarian cumulus cells may play important roles in the aetiology and pathophysiology of PCOS with IR.
Collapse
|
36
|
Feng T, Zhu Z, Jin Y, Wang H, Mao X, Liu D, Li Y, Lu L, Zuo G. The microRNA‑708‑5p/ZEB1/EMT axis mediates the metastatic potential of osteosarcoma. Oncol Rep 2019; 43:491-502. [PMID: 31894343 PMCID: PMC6967104 DOI: 10.3892/or.2019.7452] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 11/22/2019] [Indexed: 12/19/2022] Open
Abstract
MicroRNA‑708‑5p (miR‑708‑5p) and epithelial‑to‑mesenchymal transition (EMT) have been widely identified to contribute to the pathogenesis and progression of multiple cancers. However, the connection between miR‑708‑5p and EMT has not been sufficiently clarified. Therefore, our research aimed to investigate the impact of miR‑708‑5p on EMT and the metastasis of osteosarcoma (OS). We first analyzed the differentially expressed microRNAs (DEmiRNAs) from the GSE70367 dataset. We found that the expression of miR‑708‑5p was lower in OS cells. Overexpression of miR‑708‑5p was able to impair the migration and invasion of OS cells. Moreover, miR‑708‑5p inhibited EMT of OS cells MG63 and SaOS‑2, wherein E‑cadherin was increased, and N‑cadherin, vimentin, and Snail were decreased. Semaphorin 4C (SEMA4C), mitogen‑activated protein kinase kinase kinase 3 (MAP3K3), and zinc finger E‑box‑binding homeobox 1 (ZEB1) were predicted as target genes of miR‑708‑5p by bioinformatics method. Only ZEB1, one of the EMT‑inducing transcription factors, was validated as the direct target gene of miR‑708‑5p in OS cells through dual‑luciferase reporter assay and Western blot analysis. Knockdown of ZEB1 was found to inhibit the metastasis of MG63 and SaOS‑2 cells, whereas ZEB1 over-expression promoted their metastasis. In summary, miR‑708‑5p impaired the metastasis and EMT of OS, which was found to be mediated by inhibition of ZEB1.
Collapse
Affiliation(s)
- Tianyu Feng
- Key Laboratory of Diagnostic Medicine Designated by The Chinese Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Zhongkai Zhu
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Yaqian Jin
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Hao Wang
- Key Laboratory of Diagnostic Medicine Designated by The Chinese Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Xiaohan Mao
- Key Laboratory of Diagnostic Medicine Designated by The Chinese Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Dan Liu
- Key Laboratory of Diagnostic Medicine Designated by The Chinese Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Yiling Li
- Key Laboratory of Diagnostic Medicine Designated by The Chinese Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Lixia Lu
- Key Laboratory of Diagnostic Medicine Designated by The Chinese Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Guowei Zuo
- Key Laboratory of Diagnostic Medicine Designated by The Chinese Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
37
|
Senthil Kumar KJ, Gokila Vani M, Hsieh HW, Lin CC, Liao JW, Chueh PJ, Wang SY. MicroRNA-708 activation by glucocorticoid receptor agonists regulate breast cancer tumorigenesis and metastasis via downregulation of NF-κB signaling. Carcinogenesis 2019; 40:335-348. [PMID: 30726934 DOI: 10.1093/carcin/bgz011] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 01/06/2019] [Accepted: 01/22/2019] [Indexed: 02/06/2023] Open
Abstract
Therapeutic administration of glucocorticoids (GCs) is frequently used as add-on chemotherapy for palliative purposes during breast cancer treatment. Recent studies have shown that GC treatment induces microRNA-708 in ovarian cancer cells, resulting in impaired tumor cell proliferation and metastasis. However, the regulatory functions of GCs on miR-708 and its downstream target genes in human breast cancer cells (BCCs) are poorly understood. In this study, we found that treatment with either the synthetic GC dexamethasone (DEX) or the natural GC mimic, antcin A (ATA) significantly increased miR-708 expression by transactivation of glucocorticoid receptor alpha (GRα) in MCF-7 and MDA-MB-231 human BCCs. Induction of miR-708 by GR agonists resulted in inhibition of cell proliferation, cell-cycle progression, cancer stem cell (CSC)-like phenotype and metastasis of BCCs. In addition, GR agonist treatment or miR-708 mimic transfection remarkably inhibited IKKβ expression and suppressed nuclear factor-kappaB (NF-κB) activity and its downstream target genes, including COX-2, cMYC, cyclin D1, Matrix metalloproteinase (MMP)-2, MMP-9, CD24, CD44 and increased p21CIP1 and p27KIP1 that are known to be involved in proliferation, cell-cycle progression, metastasis and CSC marker protein. BCCs xenograft models indicate that treatment with GR agonists significantly reduced tumor growth, weight and volume. Overall, our data strongly suggest that GR agonists induced miR-708 and downstream suppression of NF-κB signaling, which may be applicable as a novel therapeutic intervention in breast cancer treatment.
Collapse
Affiliation(s)
- K J Senthil Kumar
- Department of Forestry, National Chung Hsing University, Taichung, Taiwan.,National Chung Hsing University/University of California at Davis, Plant and Food Biotechnology Center, National Chung Hsing University, Taichung, Taiwan
| | - M Gokila Vani
- Department of Forestry, National Chung Hsing University, Taichung, Taiwan.,National Chung Hsing University/University of California at Davis, Plant and Food Biotechnology Center, National Chung Hsing University, Taichung, Taiwan
| | | | | | - Jiunn-Wang Liao
- Graduate Institute of Veterinary Pathology, National Chung Hsing University, Taichung, Taiwan
| | - Pin-Ju Chueh
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan.,Department of Biotechnology, Asia University, Taichung, Taiwan
| | - Sheng-Yang Wang
- Department of Forestry, National Chung Hsing University, Taichung, Taiwan.,National Chung Hsing University/University of California at Davis, Plant and Food Biotechnology Center, National Chung Hsing University, Taichung, Taiwan.,Agricultural Biotechnology Research Center, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
38
|
Schneider E, Pochert N, Ruess C, MacPhee L, Escano L, Miller C, Krowiorz K, Delsing Malmberg E, Heravi-Moussavi A, Lorzadeh A, Ashouri A, Grasedieck S, Sperb N, Kumar Kopparapu P, Iben S, Staffas A, Xiang P, Rösler R, Kanduri M, Larsson E, Fogelstrand L, Döhner H, Döhner K, Wiese S, Hirst M, Keith Humphries R, Palmqvist L, Kuchenbauer F, Rouhi A. MicroRNA-708 is a novel regulator of the Hoxa9 program in myeloid cells. Leukemia 2019; 34:1253-1265. [DOI: 10.1038/s41375-019-0651-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 10/09/2019] [Accepted: 11/13/2019] [Indexed: 12/31/2022]
|
39
|
Wang Y, Huang J, Chen W, Wang R, Kao M, Pan Y, Chan S, Tsai K, Kung H, Lin K, Wang L. Dysregulation of cystathionine γ-lyase promotes prostate cancer progression and metastasis. EMBO Rep 2019; 20:e45986. [PMID: 31468690 PMCID: PMC6776913 DOI: 10.15252/embr.201845986] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 08/06/2019] [Accepted: 08/08/2019] [Indexed: 12/12/2022] Open
Abstract
Hydrogen sulfide (H2 S), an endogenous signaling gaseous molecule, is involved in various physiological activities, including vessel relaxation, regulation of cellular bioenergetics, inflammation, and angiogenesis. By using xenograft orthotopic implantation of prostate cancer PC3 cells and subsequently comparing bone metastatic with primary tumor-derived cancer cells, we find that H2 S-producing enzyme cystathionine γ-lyase (CTH) is upregulated in bone-metastatic PC3 cells. Clinical data further reveal that the expression of CTH is elevated in late-stage prostate cancer patients, and higher CTH expression correlates with poor survival from The Cancer Genome Atlas (TCGA) prostate cancer RNA-seq datasets. CTH promotes NF-κB nuclear translocation through H2 S-mediated sulfhydration on cysteine-38 of the NF-κB p65 subunit, resulting in increased IL-1β expression and H2 S-induced cell invasion. Knockdown of CTH in PC3 cells results in the suppression of tumor growth and distant metastasis, while overexpression of CTH in DU145 cells promotes primary tumor growth and lymph node metastasis in the orthotopic implanted xenograft mouse model. Together, our findings provide evidence that CTH generated H2 S promotes prostate cancer progression and metastasis through IL-1β/NF-κB signaling pathways.
Collapse
Affiliation(s)
- Yi‐Hsiang Wang
- Institute of Molecular and Genomic MedicineNational Health Research InstitutesZhunanMiaoli CountyTaiwan
- Institute of Molecular MedicineCollege of Life ScienceNational Tsing Hua UniversityHsinchuTaiwan
| | - Jo‐Ting Huang
- Institute of Molecular and Genomic MedicineNational Health Research InstitutesZhunanMiaoli CountyTaiwan
| | - Wen‐Ling Chen
- Institute of Molecular and Genomic MedicineNational Health Research InstitutesZhunanMiaoli CountyTaiwan
| | - Rong‐Hsuan Wang
- Institute of BiotechnologyCollege of Life ScienceNational Tsing Hua UniversityHsinchuTaiwan
| | - Ming‐Chien Kao
- Institute of BiotechnologyCollege of Life ScienceNational Tsing Hua UniversityHsinchuTaiwan
| | - Yan‐Ru Pan
- Institute of BiotechnologyCollege of Life ScienceNational Tsing Hua UniversityHsinchuTaiwan
| | - Shih‐Hsuan Chan
- Institute of Molecular and Genomic MedicineNational Health Research InstitutesZhunanMiaoli CountyTaiwan
- Institute of Molecular MedicineCollege of Life ScienceNational Tsing Hua UniversityHsinchuTaiwan
- Chiese Medicine Research CenterInstitute of Integrated MedicineChina Medical UniversityTaichung CityTaiwan
| | - Kuo‐Wang Tsai
- Department of Medical Education and ResearchKaohsiung Veterans General HospitalKaohsiungTaiwan
- Institute of Biomedical SciencesNational Sun Yat‐Sen UniversityKaohsiungTaiwan
- Department of Chemical BiologyNational Pingtung University of EducationPingtungTaiwan
| | - Hsing‐Jien Kung
- Institute of Molecular and Genomic MedicineNational Health Research InstitutesZhunanMiaoli CountyTaiwan
- PhD Program for Cancer Biology and Drug DiscoveryTaipei Medical UniversityTaipeiTaiwan
| | - Kai‐Ti Lin
- Institute of Molecular and Genomic MedicineNational Health Research InstitutesZhunanMiaoli CountyTaiwan
- Institute of BiotechnologyCollege of Life ScienceNational Tsing Hua UniversityHsinchuTaiwan
| | - Lu‐Hai Wang
- Institute of Molecular and Genomic MedicineNational Health Research InstitutesZhunanMiaoli CountyTaiwan
- Chiese Medicine Research CenterInstitute of Integrated MedicineChina Medical UniversityTaichung CityTaiwan
| |
Collapse
|
40
|
Zhu X, Qiu J, Zhang T, Yang Y, Guo S, Li T, Jiang K, Zahoor A, Deng G, Qiu C. MicroRNA-188-5p promotes apoptosis and inhibits cell proliferation of breast cancer cells via the MAPK signaling pathway by targeting Rap2c. J Cell Physiol 2019; 235:2389-2402. [PMID: 31541458 DOI: 10.1002/jcp.29144] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 08/23/2019] [Indexed: 12/28/2022]
Abstract
Breast cancer is a common malignancy that is highly lethal with poor survival rates and immature therapeutics that urgently needs more effective and efficient therapies. MicroRNAs are intrinsically involved in different cancer remedies, but their mechanism in breast cancer has not been elucidated for prospective treatment. The function and mechanism of microRNA-188-5p (miR-188) have not been thoroughly investigated in breast cancer. In our study, we found that the expression of miR-188 in breast cancer tissues was obviously reduced. Our findings also revealed the abnormal overexpression of miR-188 in 4T1 and MCF-7 cells significantly suppressed cell proliferation and migration and also enhanced apoptosis. miR-188 induced cell cycle arrest in the G1 phase. To illuminate the molecular mechanism of miR-188, Rap2c was screened as a single target gene by bioinformatics database analysis and was further confirmed by dual-luciferase assay. Moreover, Rap2c was found to be a vital molecular switch for the mitogen-activated protein kinase signaling pathway in tumor progression by decreasing apoptosis and promoting proliferation and migration. In conclusion, our results revealed that miR-188 is a cancer progression suppressor and a promising future target for breast cancer therapy.
Collapse
Affiliation(s)
- Xinying Zhu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Jinxia Qiu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Tao Zhang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Yaping Yang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Shuai Guo
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Tianshun Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Kangfeng Jiang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Arshad Zahoor
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Ganzhen Deng
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Changwei Qiu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
41
|
Sun SN, Hu S, Shang YP, Li LY, Zhou H, Chen JS, Yang JF, Li J, Huang Q, Shen CP, Xu T. Relevance function of microRNA-708 in the pathogenesis of cancer. Cell Signal 2019; 63:109390. [PMID: 31419576 DOI: 10.1016/j.cellsig.2019.109390] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Revised: 08/10/2019] [Accepted: 08/10/2019] [Indexed: 02/07/2023]
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs that regulate gene expression post-transcriptionally responsible for regulating >70% of human genes. MicroRNA-708 (miR-708) is encoded in the intron 1 of the Odd Oz/ten-m homolog 4 (ODZ4) gene. Numerous researches have confirmed that the abnormal expressed miR-708 is involved in the regulation of multiple types of cancer. Notably, the expression level of miR-708 was higher in lung cancer, bladder cancer (BC) and colorectal cancer (CRC) cell lines while lower in hepatocellular carcinoma (HCC), prostate cancer (PC), gastric cancer (GC) and so on. This review provides a current view on the association between miR-708 and several cancers and focuses on the recent studies of miR-708 regulation, discussing its potential as an epigenetic biomarker and therapeutic target for these cancers. In particular, the regulated mechanisms and clinical application of miR-708 in these cancers are also discussed.
Collapse
Affiliation(s)
- Si-Nan Sun
- The First Affiliation Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Shuang Hu
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; The Key laboratory of Anti-inflammatory and Immune medicines, Ministry of Education, Institute for Liver Diseases of Anhui Medical University, Hefei 230032, China
| | | | - Liang-Yun Li
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; The Key laboratory of Anti-inflammatory and Immune medicines, Ministry of Education, Institute for Liver Diseases of Anhui Medical University, Hefei 230032, China
| | - Hong Zhou
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; The Key laboratory of Anti-inflammatory and Immune medicines, Ministry of Education, Institute for Liver Diseases of Anhui Medical University, Hefei 230032, China
| | - Jia-Si Chen
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; The Key laboratory of Anti-inflammatory and Immune medicines, Ministry of Education, Institute for Liver Diseases of Anhui Medical University, Hefei 230032, China
| | - Jun-Fa Yang
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; The Key laboratory of Anti-inflammatory and Immune medicines, Ministry of Education, Institute for Liver Diseases of Anhui Medical University, Hefei 230032, China
| | - Jun Li
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; The Key laboratory of Anti-inflammatory and Immune medicines, Ministry of Education, Institute for Liver Diseases of Anhui Medical University, Hefei 230032, China
| | - Qiang Huang
- The First Affiliation Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China.
| | - Chuan-Pu Shen
- Teaching and Research Department of Traditional Chinese Medicine, Anhui Medical University, Hefei 230032, China.
| | - Tao Xu
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; The Key laboratory of Anti-inflammatory and Immune medicines, Ministry of Education, Institute for Liver Diseases of Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
42
|
Cao XM. Role of miR-337-3p and its target Rap1A in modulating proliferation, invasion, migration and apoptosis of cervical cancer cells. Cancer Biomark 2019; 24:257-267. [PMID: 30883336 DOI: 10.3233/cbm-181225] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
OBJECTIVE To investigate the role of miR-337-3p targeting Rap1A in modulating proliferation, invasion, migration and apoptosis of cervical cancer cells. METHODS The expression levels of miR-337-3p and Rap1A in cervical cancer tissues and normal tissues were evaluated through quantitative Real-time PCR (qRT-PCR) and Western blotting; and correlations of miR-337-3p with clinicopathological characteristics and prognosis of patients were also analyzed. Besides, human cervical cancer cell line HeLa cells were randomly divided into five groups (Mock, NC, miR-337-3p mimic, Rap1A, and miR-337-3p mimic + Rap1A groups). CCK-8 assay was utilized to measure cell proliferation, flow cytometry to evaluate cell apoptosis, and wound-healing and Transwell assays to detect cell migration and invasion. RESULTS Cervical cancer tissues presented a significant decrease in miR-337-3p and a remarkable increase in Rap1A protein. Besides, the expression levels of miR-337-3p and Rap1A were closely related to the major clinicopathological characteristics of cervical cancer; and patients with high-miR-337-3p-expression had the higher 5-year survival rate (all p< 0.05). When compared to Mock group, cells in miR-337-3p mimic group were suppressed in proliferation, migration, and invasion, but significantly promoted in apoptosis; meanwhile, cells in the Rap1A group showed changes in a completely opposite trend (all p< 0.05). Moreover, Rap1A can reverse the effect of miR-337-3p mimic on cell proliferation, invasion, migration and apoptosis (all p< 0.05). CONCLUSION MiR-337-3p was discovered to be decreased in cervical cancer, and miR-337-3p up-regulation may inhibit the proliferation, migration and invasion and promote the apoptosis of cervical cancer cells via down-regulating Rap1A.
Collapse
|
43
|
Tan W, Tang H, Jiang X, Ye F, Huang L, Shi D, Li L, Huang X, Li L, Xie X, Xie X. Metformin mediates induction of miR-708 to inhibit self-renewal and chemoresistance of breast cancer stem cells through targeting CD47. J Cell Mol Med 2019; 23:5994-6004. [PMID: 31273952 PMCID: PMC6714171 DOI: 10.1111/jcmm.14462] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 02/26/2019] [Accepted: 05/20/2019] [Indexed: 12/24/2022] Open
Abstract
Breast cancer stem cells (BCSCs) have been considered responsible for cancer progression, recurrence, metastasis and drug resistance. However, the mechanisms by which cells acquire self-renewal and chemoresistance properties are remaining largely unclear. Herein, we evaluated the role of miR-708 and metformin in BCSCs, and found that the expression of miR-708 is significantly down-regulated in BCSCs and tumour tissues, and correlates with chemotherapy response and prognosis. Moreover, miR-708 markedly inhibits sphere formation, CD44+ /CD24- ratio, and tumour initiation and increases chemosensitivity of BCSCs. Mechanistically, miR-708 directly binds to cluster of differentiation 47 (CD47), and regulates tumour-associated macrophage-mediated phagocytosis. On the other hand, CD47 is essential for self-renewal, tumour initiation and chemoresistance of BCSCs, and correlates with the prognosis of breast cancer patients. In addition, the anti-type II diabetes drug metformin are found to be involved in the miR-708/CD47 signalling pathway. Therefore, our study demonstrated that miR-708 plays an important tumour suppressor role in BCSCs self-renewal and chemoresistance, and the miR-708/CD47 regulatory axis may represent a novel therapeutic mechanism of metformin in BCSCs.
Collapse
Affiliation(s)
- Weige Tan
- Department of Breast Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Hailin Tang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Xinhua Jiang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Feng Ye
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Lili Huang
- Guangzhou School of Medicine, Guangzhou Medical University, Guangzhou, China
| | - Dingbo Shi
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Laisheng Li
- Department of Laboratory Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaojia Huang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Li Li
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Xiaoming Xie
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Xinhua Xie
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| |
Collapse
|
44
|
Wang P, Gu J, Wang K, Shang J, Wang W. miR-206 inhibits thyroid cancer proliferation and invasion by targeting RAP1B. J Cell Biochem 2019; 120:18927-18936. [PMID: 31245877 DOI: 10.1002/jcb.29213] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 05/30/2019] [Indexed: 01/18/2023]
Abstract
Thyroid cancer (TC) is one of the primary tumors arisen from endocrine system. The purpose of this study was to investigate the underlying mechanism by which RAP1B (Ras-related protein Rap-1b) modulates microRNA (miR)-206 related effects on TC cells. Expression of miR-206 and RAP1B was analyzed in cells and tissues. miR-206 mimics or inhibitors and RAP1B vector were used in functional experiments to investigate the effects of miR-206 and RAP1B on cell activities including proliferation, migration, and invasion. Luciferase assay was performed to explore the association between miR-206 and RAP1B. The influence of miR-206 on tumorigenesis of TC cells was investigated using an ex vivo model. Our results demonstrated the reduce of miR-206 in TC tissues and cell lines in which RAP1B was increased. Overexpression of miR-206 significantly inhibited the functional capacities of TPC-1 cells including proliferation, invasion, and migration, most likely, through reducing the expression of RAP1B. Xenograft experiment showed that increased miR-206 could effectively inhibit the tumorigenesis of TC cells. Our study showed that miR-206 negatively regulated cell activities of proliferation, invasion, and migration in TC via suppressing RAP1B expression, suggesting that miR-206 exerts a vital role in TC.
Collapse
Affiliation(s)
- Peng Wang
- Department of Head and Neck Surgery, Institute of Cancer Research and Basic Medical Sciences of Chinese Academy of Sciences, Cancer Hospital of University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Zhejiang, China
| | - Jialei Gu
- Department of Head and Neck Surgery, Institute of Cancer Research and Basic Medical Sciences of Chinese Academy of Sciences, Cancer Hospital of University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Zhejiang, China
| | - Kejing Wang
- Department of Head and Neck Surgery, Institute of Cancer Research and Basic Medical Sciences of Chinese Academy of Sciences, Cancer Hospital of University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Zhejiang, China
| | - Jinbiao Shang
- Department of Head and Neck Surgery, Institute of Cancer Research and Basic Medical Sciences of Chinese Academy of Sciences, Cancer Hospital of University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Zhejiang, China
| | - Wendong Wang
- Department of Head and Neck Surgery, Institute of Cancer Research and Basic Medical Sciences of Chinese Academy of Sciences, Cancer Hospital of University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Zhejiang, China
| |
Collapse
|
45
|
Chen Y, Deng X, Chen W, Shi P, Lian M, Wang H, Wang K, Qian D, Xiao D, Long H. Silencing of microRNA-708 promotes cell growth and epithelial-to-mesenchymal transition by activating the SPHK2/AKT/β-catenin pathway in glioma. Cell Death Dis 2019; 10:448. [PMID: 31171769 PMCID: PMC6554356 DOI: 10.1038/s41419-019-1671-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 04/22/2019] [Accepted: 05/19/2019] [Indexed: 02/06/2023]
Abstract
Aberrant microRNA-708 (miR-708) expression is frequently reported in cancer studies; however, its role in glioma has not been examined in detail. We investigated miR-708 function in glioma and revealed that miR-708 expression was significantly down-regulated in glioma tissues and cell lines. Restoration of miR-708 inhibited glioma cell growth and invasion both in vitro and in vivo. The oncogene SPHK2 (sphingosine kinase 2) was identified as a downstream target of miR-708 using luciferase and western blot assays. miR-708 inhibited AKT/β-catenin signaling, which is activated by SPHK2. In addition, we revealed that miR-708 was transcriptionally repressed by EZH2 (enhancer of zeste homolog 2)-induced histone H3 lysine 27 trimethylation and promoter methylation. In summary, our findings revealed that miR-708 is a glioma tumor suppressor and suggest that miR-708 is a potential therapeutic target for glioma patients.
Collapse
Affiliation(s)
- Yan Chen
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Cancer Immunotherapy Research and Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, Southern Medical University, Guangzhou, China
| | - Xubin Deng
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | | | - Pengwei Shi
- Department of Emergency, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Mei Lian
- Guangdong Provincial Key Laboratory of Cancer Immunotherapy Research and Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, Southern Medical University, Guangzhou, China
| | - Hongxiao Wang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Kewan Wang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Dadi Qian
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Dong Xiao
- Guangdong Provincial Key Laboratory of Cancer Immunotherapy Research and Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, Southern Medical University, Guangzhou, China.
| | - Hao Long
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
46
|
Frame FM, Maitland NJ. Epigenetic Control of Gene Expression in the Normal and Malignant Human Prostate: A Rapid Response Which Promotes Therapeutic Resistance. Int J Mol Sci 2019; 20:E2437. [PMID: 31108832 PMCID: PMC6566891 DOI: 10.3390/ijms20102437] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 04/24/2019] [Accepted: 04/29/2019] [Indexed: 12/12/2022] Open
Abstract
A successful prostate cancer must be capable of changing its phenotype in response to a variety of microenvironmental influences, such as adaptation to treatment or successful proliferation at a particular metastatic site. New cell phenotypes emerge by selection from the large, genotypically heterogeneous pool of candidate cells present within any tumor mass, including a distinct stem cell-like population. In such a multicellular model of human prostate cancer, flexible responses are primarily governed not only by de novo mutations but appear to be dominated by a combination of epigenetic controls, whose application results in treatment resistance and tumor relapse. Detailed studies of these individual cell populations have resulted in an epigenetic model for epithelial cell differentiation, which is also instructive in explaining the reported high and inevitable relapse rates of human prostate cancers to a multitude of treatment types.
Collapse
Affiliation(s)
- Fiona M Frame
- The Cancer Research Unit, Department of Biology, University of York, Heslington, York YO10 5DD, UK.
| | - Norman J Maitland
- The Cancer Research Unit, Department of Biology, University of York, Heslington, York YO10 5DD, UK.
| |
Collapse
|
47
|
Baghdadi MB, Firmino J, Soni K, Evano B, Di Girolamo D, Mourikis P, Castel D, Tajbakhsh S. Notch-Induced miR-708 Antagonizes Satellite Cell Migration and Maintains Quiescence. Cell Stem Cell 2018; 23:859-868.e5. [PMID: 30416072 DOI: 10.1016/j.stem.2018.09.017] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Revised: 07/18/2018] [Accepted: 09/27/2018] [Indexed: 12/31/2022]
Abstract
Critical features of stem cells include anchoring within a niche and activation upon injury. Notch signaling maintains skeletal muscle satellite (stem) cell quiescence by inhibiting differentiation and inducing expression of extracellular components of the niche. However, the complete spectrum of how Notch safeguards quiescence is not well understood. Here, we perform Notch ChIP-sequencing and small RNA sequencing in satellite cells and identify the Notch-induced microRNA-708, which is a mirtron that is highly expressed in quiescent cells and sharply downregulated in activated cells. We employ in vivo and ex vivo functional studies, in addition to live imaging, to show that miR-708 regulates quiescence and self-renewal by antagonizing cell migration through targeting the transcripts of the focal-adhesion-associated protein Tensin3. Therefore, this study identifies a Notch-miR708-Tensin3 axis and suggests that Notch signaling can regulate satellite cell quiescence and transition to the activation state through dynamic regulation of the migratory machinery.
Collapse
Affiliation(s)
- Meryem B Baghdadi
- Stem Cells and Development, Department of Developmental & Stem Cell Biology, Institut Pasteur, Paris 75015, France; CNRS UMR 3738, Institut Pasteur, Paris 75015, France; Sorbonne Universités, UPMC, University of Paris 06, IFD-ED 515, Paris 75252, France
| | - Joao Firmino
- Bioimaging and Optics platform (BIOP), School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - Kartik Soni
- Stem Cells and Development, Department of Developmental & Stem Cell Biology, Institut Pasteur, Paris 75015, France; CNRS UMR 3738, Institut Pasteur, Paris 75015, France
| | - Brendan Evano
- Stem Cells and Development, Department of Developmental & Stem Cell Biology, Institut Pasteur, Paris 75015, France; CNRS UMR 3738, Institut Pasteur, Paris 75015, France
| | - Daniela Di Girolamo
- Stem Cells and Development, Department of Developmental & Stem Cell Biology, Institut Pasteur, Paris 75015, France; Dipartimento di Medicina Clinica e Chirurgica, Università degli Studi di Napoli Frederico II, 80131 Naples, Italy
| | | | - David Castel
- UMR8203 "Vectorologie et Thérapeutiques Anticancéreuses," CNRS, Gustave Roussy, Université Paris-Sud, Université Paris-Saclay, 94805 Villejuif, France; Département de Cancérologie de l'Enfant et de l'Adolescent, Gustave Roussy, Université Paris-Sud, Université Paris-Saclay, 94805 Villejuif, France
| | - Shahragim Tajbakhsh
- Stem Cells and Development, Department of Developmental & Stem Cell Biology, Institut Pasteur, Paris 75015, France; CNRS UMR 3738, Institut Pasteur, Paris 75015, France.
| |
Collapse
|
48
|
Duan Y, Dong Y, Dang R, Hu Z, Yang Y, Hu Y, Cheng J. MiR-122 inhibits epithelial mesenchymal transition by regulating P4HA1 in ovarian cancer cells. Cell Biol Int 2018; 42:1564-1574. [PMID: 30136751 DOI: 10.1002/cbin.11052] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 08/18/2018] [Indexed: 02/01/2023]
Affiliation(s)
- Yanan Duan
- Department of Gynecology; The Fourth Hospital of Hebei Medical University; Shijiazhuang 050011 People's Republic of China
| | - Yinghui Dong
- Department of Oncology; Shijiazhuang First Hospital; Shijiazhuang 050011 People's Republic of China
| | - Rongguang Dang
- Department of Oncology; Shijiazhuang First Hospital; Shijiazhuang 050011 People's Republic of China
| | - Zhihui Hu
- Department of Oncology; Shijiazhuang First Hospital; Shijiazhuang 050011 People's Republic of China
| | - Ye Yang
- Department of Oncology; Shijiazhuang First Hospital; Shijiazhuang 050011 People's Republic of China
| | - Yueyun Hu
- Department of Oncology; Shijiazhuang First Hospital; Shijiazhuang 050011 People's Republic of China
| | - Jianxin Cheng
- Department of Gynecology; The Fourth Hospital of Hebei Medical University; Shijiazhuang 050011 People's Republic of China
| |
Collapse
|
49
|
Significance of PI3K/AKT signaling pathway in metastasis of esophageal squamous cell carcinoma and its potential as a target for anti-metastasis therapy. Oncotarget 2018; 8:38755-38766. [PMID: 28418888 PMCID: PMC5503569 DOI: 10.18632/oncotarget.16333] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 02/20/2017] [Indexed: 02/06/2023] Open
Abstract
Metastasis is the most lethal hallmark of esophageal squamous cell carcinoma (ESCC). The aim of the study is to identify key signaling pathways that control metastasis in ESCC. Highly invasive ESCC sublines (designated I3 cells) were established through three rounds of selection of cancer cells invading through matrigel-coated chambers. Gene expression profile of one of the I3 sublines was compared with that of its parental cell line using cDNA microarray analysis. Gene ontology and pathway analyses of the differentially expressed genes (both upregulated and downregulated) indicated that genes associated with cellular movement and the AKT pathway were associated with increased cancer cell invasiveness. Western blot analysis confirmed increased phosphorylated AKT (p-AKT), N-cadherin and decreased E-cadherin expression in the I3 cells. Immunohistochemistry was used to evaluate the clinical significance of p-AKT expression in ESCC, and the results showed higher p-AKT nuclear expression in lymph node metastases when compared with primary carcinoma. Inactivation of the PI3K/AKT pathway with specific inhibitors, or with PTEN overexpression, resulted in reversed cadherin switching and inhibited cancer cell motility. Inhibition of the pathway by treatment with wortmannin markedly suppressed experimental metastasis in nude mice. Our data demonstrated the importance of the PI3K/AKT signaling pathway in ESCC metastasis and support PI3K/AKT as a valid therapeutic target in treatment of metastatic ESCC.
Collapse
|
50
|
Chang JW, Kuo WH, Lin CM, Chen WL, Chan SH, Chiu MF, Chang IS, Jiang SS, Tsai FY, Chen CH, Huang PH, Chang KJ, Lin KT, Lin SC, Wang MY, Uen YH, Tu CW, Hou MF, Tsai SF, Shen CY, Tung SL, Wang LH. Wild-type p53 upregulates an early onset breast cancer-associated gene GAS7 to suppress metastasis via GAS7-CYFIP1-mediated signaling pathway. Oncogene 2018; 37:4137-4150. [PMID: 29706651 PMCID: PMC6062498 DOI: 10.1038/s41388-018-0253-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Revised: 01/07/2018] [Accepted: 03/14/2018] [Indexed: 01/13/2023]
Abstract
The early onset breast cancer patients (age ≤ 40) often display higher incidence of axillary lymph node metastasis, and poorer five-year survival than the late-onset patients. To identify the genes and molecules associated with poor prognosis of early onset breast cancer, we examined gene expression profiles from paired breast normal/tumor tissues, and coupled with Gene Ontology and public data base analysis. Our data showed that the expression of GAS7b gene was lower in the early onset breast cancer patients as compared to the elder patients. We found that GAS7 was associated with CYFIP1 and WAVE2 complex to suppress breast cancer metastasis via blocking CYFIP1 and Rac1 protein interaction, actin polymerization, and β1-integrin/FAK/Src signaling. We further demonstrated that p53 directly regulated GAS7 gene expression, which was inversely correlated with p53 mutations in breast cancer specimens. Our study uncover a novel regulatory mechanism of p53 in early onset breast cancer progression through GAS7-CYFIP1-mediated signaling pathways.
Collapse
Affiliation(s)
- Jer-Wei Chang
- Institute of Molecular and Genomic Medicine, National Health Research Institute, Miaoli, Taiwan
| | - Wen-Hung Kuo
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Chiao-Mei Lin
- Institute of Molecular and Genomic Medicine, National Health Research Institute, Miaoli, Taiwan
| | - Wen-Ling Chen
- Institute of Molecular and Genomic Medicine, National Health Research Institute, Miaoli, Taiwan
| | - Shih-Hsuan Chan
- Institute of Molecular and Genomic Medicine, National Health Research Institute, Miaoli, Taiwan.,Institute of Molecular Medicine, College of Life Science, National Tsing Hua University, Hsinchu, Taiwan.,Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Meng-Fan Chiu
- Institute of Molecular and Genomic Medicine, National Health Research Institute, Miaoli, Taiwan
| | - I-Shou Chang
- National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan
| | - Shih-Sheng Jiang
- National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan
| | - Fang-Yu Tsai
- National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan
| | - Chung-Hsing Chen
- National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan
| | - Pei-Hsin Huang
- Department of Pathology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - King-Jen Chang
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan.,Taiwan Adventist Hospital, Taipei, Taiwan
| | - Kai-Ti Lin
- Institute of Biotechnology, College of Life Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Sheng-Chieh Lin
- College of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Ming-Yang Wang
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Yih-Huei Uen
- Department of Surgery, Asia University Hospital, Taichung, Taiwan
| | - Chi-Wen Tu
- Department of General Surgery, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi, Taiwan
| | - Ming-Feng Hou
- Department of Surgery, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Surgery, Kaohsiung Municipal Hsiao Kang Hospital, Kaohsiung, Taiwan.,Division of Breast Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Shih-Feng Tsai
- Institute of Molecular and Genomic Medicine, National Health Research Institute, Miaoli, Taiwan
| | - Chen-Yang Shen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Shiao-Lin Tung
- Department of Hematology and Oncology, Ton-Yen General Hospital, Hsinchu, Taiwan
| | - Lu-Hai Wang
- Institute of Molecular and Genomic Medicine, National Health Research Institute, Miaoli, Taiwan. .,College of Chinese Medicine, China Medical University, Taichung, Taiwan.
| |
Collapse
|