1
|
Azani A, Omran SP, Ghasrsaz H, Idani A, Eliaderani MK, Peirovi N, Dokhani N, Lotfalizadeh MH, Rezaei MM, Ghahfarokhi MS, KarkonShayan S, Hanjani PN, Kardaan Z, Navashenagh JG, Yousefi M, Abdolahi M, Salmaninejad A. MicroRNAs as biomarkers for early diagnosis, targeting and prognosis of prostate cancer. Pathol Res Pract 2023; 248:154618. [PMID: 37331185 DOI: 10.1016/j.prp.2023.154618] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 06/09/2023] [Accepted: 06/10/2023] [Indexed: 06/20/2023]
Abstract
Globally, prostate cancer (PC) is leading cause of cancer-related mortality in men worldwide. Despite significant advances in the treatment and management of this disease, the cure rates for PC remains low, largely due to late detection. PC detection is mostly reliant on prostate-specific antigen (PSA) and digital rectal examination (DRE); however, due to the low positive predictive value of current diagnostics, there is an urgent need to identify new accurate biomarkers. Recent studies support the biological role of microRNAs (miRNAs) in the initiation and progression of PC, as well as their potential as novel biomarkers for patients' diagnosis, prognosis, and disease relapse. In the advanced stages, cancer-cell-derived small extracellular vesicles (SEVs) may constitute a significant part of circulating vesicles and cause detectable changes in the plasma vesicular miRNA profile. Recent computational model for the identification of miRNA biomarkers discussed. In addition, accumulating evidence indicates that miRNAs can be utilized to target PC cells. In this article, the current understanding of the role of microRNAs and exosomes in the pathogenesis and their significance in PC prognosis, early diagnosis, chemoresistance, and treatment are reviewed.
Collapse
Affiliation(s)
- Alireza Azani
- Department of Medical Genetics, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sima Parvizi Omran
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Haniyeh Ghasrsaz
- Faculty of Medicine, Mazandaran University of Medical Sciences, Mazandaran, Iran
| | - Asra Idani
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Niloufar Peirovi
- Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Negar Dokhani
- Student Research Committee, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | | | | | | | - Sepideh KarkonShayan
- Social Development and Health Promotion Research Center, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Parisa Najari Hanjani
- Department of Genetics, Faculty of Advanced Technologies in Medicine, Golestan University of Medical Science, Gorgan, Iran
| | - Zahra Kardaan
- Department of Cellular Molecular Biology, Faculty of Life Science and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | | | - Meysam Yousefi
- Department of Medical Genetics, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mitra Abdolahi
- Department of Pathology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Arash Salmaninejad
- Department of Medical Genetics, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Regenerative Medicine, Organ Procurement and Transplantation Multi-Disciplinary Center, Razi Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran.
| |
Collapse
|
2
|
Guan G, Cannon RD, Coates DE, Mei L. Effect of the Rho-Kinase/ROCK Signaling Pathway on Cytoskeleton Components. Genes (Basel) 2023; 14:272. [PMID: 36833199 PMCID: PMC9957420 DOI: 10.3390/genes14020272] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/10/2023] [Accepted: 01/16/2023] [Indexed: 01/22/2023] Open
Abstract
The mechanical properties of cells are important in tissue homeostasis and enable cell growth, division, migration and the epithelial-mesenchymal transition. Mechanical properties are determined to a large extent by the cytoskeleton. The cytoskeleton is a complex and dynamic network composed of microfilaments, intermediate filaments and microtubules. These cellular structures confer both cell shape and mechanical properties. The architecture of the networks formed by the cytoskeleton is regulated by several pathways, a key one being the Rho-kinase/ROCK signaling pathway. This review describes the role of ROCK (Rho-associated coiled-coil forming kinase) and how it mediates effects on the key components of the cytoskeleton that are critical for cell behaviour.
Collapse
Affiliation(s)
- Guangzhao Guan
- Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin 9016, New Zealand
- Department of Oral Diagnostic and Surgical Sciences, Faculty of Dentistry, University of Otago, 310 Great King Street, Dunedin 9016, New Zealand
| | - Richard D. Cannon
- Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin 9016, New Zealand
- Department of Oral Sciences, Faculty of Dentistry, University of Otago, 310 Great King Street, Dunedin 9016, New Zealand
| | - Dawn E. Coates
- Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin 9016, New Zealand
| | - Li Mei
- Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin 9016, New Zealand
- Department of Oral Sciences, Faculty of Dentistry, University of Otago, 310 Great King Street, Dunedin 9016, New Zealand
| |
Collapse
|
3
|
Ye Q, Ying Q, Dai Q, Liao C, Xiao G. Tumor-suppressing effects of miR-381-3p in pediatric acute myeloid leukemia via ROCK1 downregulation. Funct Integr Genomics 2023; 23:43. [PMID: 36658407 DOI: 10.1007/s10142-022-00950-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 12/19/2022] [Accepted: 12/21/2022] [Indexed: 01/21/2023]
Abstract
MicroRNA (miR)-381-3p is the newly discovered tumor-associated miRNA, which is frequently associated with diverse human malignancies; but, it is still unknown about its effect on acute myeloid leukemia (AML) in children. This work focused on exploring miR-381-3p's effect on childhood AML and identifying the possible mechanisms facilitating new treatment development. Using qRT-PCR analysis, miR-381-3p expression remarkably reduced in pediatric AML patients and AML cell lines (HL-60 and U937). Following transfection of miR-381-3p mimic or inhibitor into HL-60 and U937 cells, we conducted MTT assay to evaluate cell proliferation, flow cytometry (FCM) to measured cell apoptosis and cell cycle, whereas Transwell assays to detect cell invasion and migration. Our results demonstrated that miR-381-3p overexpression remarkably repressed cell growth, invasion and migration; additionally, miR-381-3p overexpression resulted in arrest of cell cycle and enhanced cell apoptosis. In contrast, miR-381-3p knockdown led to an opposite effect. Moreover, we predicted miR-381's target gene and validated it by luciferase reporter assay and TargetScan, separately. We identified miR-381-3p's binding site in ROCK1 3'-UTR. As revealed by Western-blot (WB) assay, miR-381-3p overexpression notably suppressed ROCK1 level. Moreover, restoring ROCK1 expression abolished miR-381-3p's inhibition on cell proliferation, invasion and migration. Data in this work indicated the role of miR-381-3p as the tumor suppressor within pediatric AML by targeting ROCK1. Therefore, miR-381-3p might serve as a potential therapeutic target for the treatment of pediatric AML.
Collapse
Affiliation(s)
- Qidong Ye
- Department of Pediatrics, Ningbo First Hospital, Ningbo Hospital of Zhejiang University, No. 59 Liuting Street, Ningbo, 315000, Zhejiang Province, People's Republic of China.
| | - Qianqian Ying
- Department of Pediatrics, Ningbo First Hospital, Ningbo Hospital of Zhejiang University, No. 59 Liuting Street, Ningbo, 315000, Zhejiang Province, People's Republic of China
| | - Qiaoyan Dai
- Department of Pediatrics, Ningbo First Hospital, Ningbo Hospital of Zhejiang University, No. 59 Liuting Street, Ningbo, 315000, Zhejiang Province, People's Republic of China
| | - Cong Liao
- Department of Pediatrics, Ningbo First Hospital, Ningbo Hospital of Zhejiang University, No. 59 Liuting Street, Ningbo, 315000, Zhejiang Province, People's Republic of China
| | - Gang Xiao
- Department of Pediatrics, Ningbo First Hospital, Ningbo Hospital of Zhejiang University, No. 59 Liuting Street, Ningbo, 315000, Zhejiang Province, People's Republic of China
| |
Collapse
|
4
|
Fischer D, Fluegen G, Garcia P, Ghaffari-Tabrizi-Wizsy N, Gribaldo L, Huang RYJ, Rasche V, Ribatti D, Rousset X, Pinto MT, Viallet J, Wang Y, Schneider-Stock R. The CAM Model-Q&A with Experts. Cancers (Basel) 2022; 15:cancers15010191. [PMID: 36612187 PMCID: PMC9818221 DOI: 10.3390/cancers15010191] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/20/2022] [Accepted: 12/24/2022] [Indexed: 12/30/2022] Open
Abstract
The chick chorioallantoic membrane (CAM), as an extraembryonic tissue layer generated by the fusion of the chorion with the vascularized allantoic membrane, is easily accessible for manipulation. Indeed, grafting tumor cells on the CAM lets xenografts/ovografts develop in a few days for further investigations. Thus, the CAM model represents an alternative test system that is a simple, fast, and low-cost tool to study tumor growth, drug response, or angiogenesis in vivo. Recently, a new era for the CAM model in immune-oncology-based drug discovery has been opened up. Although there are many advantages offering extraordinary and unique applications in cancer research, it has also disadvantages and limitations. This review will discuss the pros and cons with experts in the field.
Collapse
Affiliation(s)
- Dagmar Fischer
- Division of Pharmaceutical Technology, Department of Chemistry and Pharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058 Erlangen, Germany
| | - Georg Fluegen
- Department of General, Visceral, Thoracic and Pediatric Surgery (A), Medical Faculty, Heinrich-Heine-University, University Hospital Duesseldorf, 40225 Duesseldorf, Germany
| | - Paul Garcia
- Institute for Advanced Biosciences, Research Center Université Grenoble Alpes (UGA)/Inserm U 1209/CNRS 5309, 38700 La Tronche, France
- R&D Department, Inovotion, 38700 La Tronche, France
| | - Nassim Ghaffari-Tabrizi-Wizsy
- SFL Chicken CAM Lab, Department of Immunology, Otto Loewi Research Center, Medical University of Graz, 8010 Graz, Austria
| | - Laura Gribaldo
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy
| | - Ruby Yun-Ju Huang
- School of Medicine, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
- Graduate Institute of Oncology, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
- Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
| | - Volker Rasche
- Department of Internal Medicine II, Ulm University Medical Center, 89073 Ulm, Germany
| | - Domenico Ribatti
- Department of Translational Biomedicine and Neurosciences, University of Bari “Aldo Moro”, 70124 Bari, Italy
| | | | - Marta Texeira Pinto
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- Ipatimup—Instituto de Patologia e Imunologia Molecular da Universidade do Porto, 4200-135 Porto, Portugal
| | - Jean Viallet
- R&D Department, Inovotion, 38700 La Tronche, France
| | - Yan Wang
- R&D Department, Inovotion, 38700 La Tronche, France
| | - Regine Schneider-Stock
- Experimental Tumorpathology, Institute of Pathology, Universitätsklinikum Erlangen, FAU Erlangen-Nürnberg, 91054 Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Universitätsklinikum Erlangen, FAU Erlangen-Nürnberg, 94054 Erlangen, Germany
- Correspondence: ; Tel.: +49-9131-8526-069
| |
Collapse
|
5
|
Gundu C, Arruri VK, Yadav P, Navik U, Kumar A, Amalkar VS, Vikram A, Gaddam RR. Dynamin-Independent Mechanisms of Endocytosis and Receptor Trafficking. Cells 2022; 11:cells11162557. [PMID: 36010634 PMCID: PMC9406725 DOI: 10.3390/cells11162557] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 08/03/2022] [Accepted: 08/13/2022] [Indexed: 11/16/2022] Open
Abstract
Endocytosis is a fundamental mechanism by which cells perform housekeeping functions. It occurs via a variety of mechanisms and involves many regulatory proteins. The GTPase dynamin acts as a “molecular scissor” to form endocytic vesicles and is a critical regulator among the proteins involved in endocytosis. Some GTPases (e.g., Cdc42, arf6, RhoA), membrane proteins (e.g., flotillins, tetraspanins), and secondary messengers (e.g., calcium) mediate dynamin-independent endocytosis. These pathways may be convergent, as multiple pathways exist in a single cell. However, what determines the specific path of endocytosis is complex and challenging to comprehend. This review summarizes the mechanisms of dynamin-independent endocytosis, the involvement of microRNAs, and factors that contribute to the cellular decision about the specific route of endocytosis.
Collapse
Affiliation(s)
- Chayanika Gundu
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, Telangana, India
| | - Vijay Kumar Arruri
- Department of Neurological Surgery, University of Wisconsin, Madison, WI 53792, USA
| | - Poonam Yadav
- Department of Pharmacology, Central University of Punjab, Bathinda 151001, Punjab, India
| | - Umashanker Navik
- Department of Pharmacology, Central University of Punjab, Bathinda 151001, Punjab, India
| | - Ashutosh Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Kolkata 700054, West Bengal, India
| | - Veda Sudhir Amalkar
- Department of Internal Medicine, Carver College of Medicine, The University of Iowa, Iowa City, IA 52242, USA
| | - Ajit Vikram
- Department of Internal Medicine, Carver College of Medicine, The University of Iowa, Iowa City, IA 52242, USA
| | - Ravinder Reddy Gaddam
- Department of Internal Medicine, Carver College of Medicine, The University of Iowa, Iowa City, IA 52242, USA
- Correspondence:
| |
Collapse
|
6
|
Advances in the Current Understanding of the Mechanisms Governing the Acquisition of Castration-Resistant Prostate Cancer. Cancers (Basel) 2022; 14:cancers14153744. [PMID: 35954408 PMCID: PMC9367587 DOI: 10.3390/cancers14153744] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 07/29/2022] [Indexed: 11/17/2022] Open
Abstract
Despite aggressive treatment and androgen-deprivation therapy, most prostate cancer patients ultimately develop castration-resistant prostate cancer (CRPC), which is associated with high mortality rates. However, the mechanisms governing the development of CRPC are poorly understood, and androgen receptor (AR) signaling has been shown to be important in CRPC through AR gene mutations, gene overexpression, co-regulatory factors, AR shear variants, and androgen resynthesis. A growing number of non-AR pathways have also been shown to influence the CRPC progression, including the Wnt and Hh pathways. Moreover, non-coding RNAs have been identified as important regulators of the CRPC pathogenesis. The present review provides an overview of the relevant literature pertaining to the mechanisms governing the molecular acquisition of castration resistance in prostate cancer, providing a foundation for future, targeted therapeutic efforts.
Collapse
|
7
|
Hatat AS, Benoit-Pilven C, Pucciarelli A, de Fraipont F, Lamothe L, Perron P, Rey A, Giaj Levra M, Toffart AC, Auboeuf D, Eymin B, Gazzeri S. Altered splicing of ATG16-L1 mediates acquired resistance to tyrosine kinase inhibitors of EGFR by blocking autophagy in non-small cell lung cancer. Mol Oncol 2022; 16:3490-3508. [PMID: 35593080 PMCID: PMC9533692 DOI: 10.1002/1878-0261.13229] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 03/25/2022] [Accepted: 05/18/2022] [Indexed: 11/11/2022] Open
Abstract
Despite the initial efficacy of using tyrosine kinase inhibitors of epidermal growth factor receptor (EGFR-TKIs) for treating patients with non-small cell lung cancer (NSCLC), resistance inevitably develops. Recent studies highlight a link between alternative splicing and cancer drug response. Therefore, we aimed to identify deregulated splicing events that play a role in resistance to EGFR-TKI. By using RNA sequencing, reverse transcription PCR (RT-PCR) and RNA interference, we showed that overexpression of a splice variant of the autophagic gene ATG16-L1 that retains exon 8 and encodes the β-isoform of autophagy-related protein 16-1 (ATG16-L1-β) concurs acquired resistance to EGFR-TKI in NSCLC cells. Using matched biopsies, we found increased levels of ATG16-L1-β at the time of progression in 3 of 11 NSCLC patients treated with EGFR-TKI. Mechanistically, gefitinib-induced autophagy was impaired in resistant cells that accumulated ATG16-L1-β. Neutralization of ATG16-L1-β restored autophagy in response to gefitinib, induced apoptosis and inhibited the growth of in ovo tumor xenografts. Conversely, overexpression of ATG16-L1-β in parental sensitive cells prevented gefitinib-induced autophagy and increased cell survival. These results support a role for defective autophagy in acquired resistance to EGFR-TKIs and identify splicing regulation of ATG16-L1 as a therapeutic vulnerability that could be explored for improving EGFR-targeted cancer therapy.
Collapse
Affiliation(s)
- Anne-Sophie Hatat
- Team "RNA splicing, cell signaling and response to therapies", Institute for Advanced Biosciences (IAB), INSERM U1209, CNRS UMR 5309, Grenoble Alpes University, Grenoble, France
| | - Clara Benoit-Pilven
- Laboratory of Biology and Modelling of the Cell, Univ Lyon, ENS de Lyon, Univ Claude Bernard, CNRS UMR 5239, INSERM U1210, 46 Allée d'Italie, Site Jacques Monod, F-69007, Lyon, France
| | - Amélie Pucciarelli
- Team "RNA splicing, cell signaling and response to therapies", Institute for Advanced Biosciences (IAB), INSERM U1209, CNRS UMR 5309, Grenoble Alpes University, Grenoble, France
| | - Florence de Fraipont
- Team "RNA splicing, cell signaling and response to therapies", Institute for Advanced Biosciences (IAB), INSERM U1209, CNRS UMR 5309, Grenoble Alpes University, Grenoble, France.,Molecular Genetic Unit, Grenoble-Alpes University Hospital, Grenoble, France
| | - Lucie Lamothe
- Team "RNA splicing, cell signaling and response to therapies", Institute for Advanced Biosciences (IAB), INSERM U1209, CNRS UMR 5309, Grenoble Alpes University, Grenoble, France
| | - Pascal Perron
- Team "RNA splicing, cell signaling and response to therapies", Institute for Advanced Biosciences (IAB), INSERM U1209, CNRS UMR 5309, Grenoble Alpes University, Grenoble, France
| | - Amandine Rey
- Laboratory of Biology and Modelling of the Cell, Univ Lyon, ENS de Lyon, Univ Claude Bernard, CNRS UMR 5239, INSERM U1210, 46 Allée d'Italie, Site Jacques Monod, F-69007, Lyon, France
| | - Matteo Giaj Levra
- Team "RNA splicing, cell signaling and response to therapies", Institute for Advanced Biosciences (IAB), INSERM U1209, CNRS UMR 5309, Grenoble Alpes University, Grenoble, France.,Thoracic Oncology Unit, Grenoble-Alpes University Hospital, Grenoble, France
| | - Anne-Claire Toffart
- Team "RNA splicing, cell signaling and response to therapies", Institute for Advanced Biosciences (IAB), INSERM U1209, CNRS UMR 5309, Grenoble Alpes University, Grenoble, France.,Thoracic Oncology Unit, Grenoble-Alpes University Hospital, Grenoble, France
| | - Didier Auboeuf
- Laboratory of Biology and Modelling of the Cell, Univ Lyon, ENS de Lyon, Univ Claude Bernard, CNRS UMR 5239, INSERM U1210, 46 Allée d'Italie, Site Jacques Monod, F-69007, Lyon, France
| | - Beatrice Eymin
- Team "RNA splicing, cell signaling and response to therapies", Institute for Advanced Biosciences (IAB), INSERM U1209, CNRS UMR 5309, Grenoble Alpes University, Grenoble, France
| | - Sylvie Gazzeri
- Team "RNA splicing, cell signaling and response to therapies", Institute for Advanced Biosciences (IAB), INSERM U1209, CNRS UMR 5309, Grenoble Alpes University, Grenoble, France
| |
Collapse
|
8
|
Doghish AS, Ismail A, El-Mahdy HA, Elkady MA, Elrebehy MA, Sallam AAM. A review of the biological role of miRNAs in prostate cancer suppression and progression. Int J Biol Macromol 2022; 197:141-156. [PMID: 34968539 DOI: 10.1016/j.ijbiomac.2021.12.141] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 12/20/2021] [Accepted: 12/21/2021] [Indexed: 02/06/2023]
Abstract
Prostate cancer (PC) is the third-leading cause of cancer-related deaths worldwide. Although the current treatment strategies are progressing rapidly, PC is still representing a substantial medical problem for affected patients. Several factors are involved in PC initiation, progression, and treatments failure including microRNAs (miRNAs). The miRNAs are endogenous short non-coding RNA sequence negatively regulating target mRNA expression via degradation or translation repression. miRNAs play a pivotal role in PC pathogenesis through its ability to initiate the induction of cancer stem cells (CSCs) and proliferation, as well as sustained cell cycle, evading apoptosis, invasion, angiogenesis, and metastasis. Furthermore, miRNAs regulate major molecular pathways affecting PC such as the androgen receptor (AR) pathway, p53 pathway, PTEN/PI3K/AKT pathway, and Wnt/β-catenin pathway. Furthermore, miRNAs alter PC therapeutic response towards the androgen deprivation therapy (ADT), chemotherapy and radiation therapy (RT). Thus, the understanding and profiling of the altered miRNAs expression in PC could be utilized as a non-invasive biomarker for the early diagnosis as well as for patient sub-grouping with different prognoses for individualized treatment. Accordingly, in the current review, we summarized in updated form the roles of various oncogenic and tumor suppressor (TS) miRNAs in PC, revealing their underlying molecular mechanisms in PC initiation and progression.
Collapse
Affiliation(s)
- Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt; Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11231 Cairo, Egypt.
| | - Ahmed Ismail
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11231 Cairo, Egypt
| | - Hesham A El-Mahdy
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11231 Cairo, Egypt
| | - Mohamed A Elkady
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11231 Cairo, Egypt
| | - Mahmoud A Elrebehy
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Al-Aliaa M Sallam
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt; Biochemistry Department, Faculty of Pharmacy, Ain-Shams University, Abassia, Cairo 11566, Egypt
| |
Collapse
|
9
|
Shi J, Wei L. Rho Kinases in Embryonic Development and Stem Cell Research. Arch Immunol Ther Exp (Warsz) 2022; 70:4. [PMID: 35043239 PMCID: PMC8766376 DOI: 10.1007/s00005-022-00642-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 12/14/2021] [Indexed: 12/12/2022]
Abstract
The Rho-associated coiled-coil containing kinases (ROCKs or Rho kinases) belong to the AGC (PKA/PKG/PKC) family of serine/threonine kinases and are major downstream effectors of small GTPase RhoA, a key regulator of actin-cytoskeleton reorganization. The ROCK family contains two members, ROCK1 and ROCK2, which share 65% overall identity and 92% identity in kinase domain. ROCK1 and ROCK2 were assumed to be functionally redundant, based largely on their major common activators, their high degree kinase domain homology, and study results from overexpression with kinase constructs or chemical inhibitors. ROCK signaling research has expanded to all areas of biology and medicine since its discovery in 1996. The rapid advance is befitting ROCK’s versatile functions in modulating various cell behavior, such as contraction, adhesion, migration, proliferation, polarity, cytokinesis, and differentiation. The rapid advance is noticeably driven by an extensive linking with clinical medicine, including cardiovascular abnormalities, aberrant immune responsive, and cancer development and metastasis. The rapid advance during the past decade is further powered by novel biotechnologies including CRISPR-Cas and single cell omics. Current consensus, derived mainly from gene targeting and RNA interference approaches, is that the two ROCK isoforms have overlapping and distinct cellular, physiological and pathophysiology roles. In this review, we present an overview of the milestone discoveries in ROCK research. We then focus on the current understanding of ROCK signaling in embryonic development, current research status using knockout and knockin mouse models, and stem cell research.
Collapse
Affiliation(s)
- Jianjian Shi
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, School of Medicine, Indiana University, 1044 West Walnut Street, R4-370, Indianapolis, IN, 46202-5225, USA.
| | - Lei Wei
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, School of Medicine, Indiana University, 1044 West Walnut Street, R4-370, Indianapolis, IN, 46202-5225, USA.
| |
Collapse
|
10
|
Deng T, Xiao Y, Dai Y, Xie L, Li X. Roles of Key Epigenetic Regulators in the Gene Transcription and Progression of Prostate Cancer. Front Mol Biosci 2021; 8:743376. [PMID: 34977151 PMCID: PMC8714908 DOI: 10.3389/fmolb.2021.743376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Accepted: 11/25/2021] [Indexed: 12/24/2022] Open
Abstract
Prostate cancer (PCa) is a top-incidence malignancy, and the second most common cause of death amongst American men and the fifth leading cause of cancer death in men around the world. Androgen receptor (AR), the key transcription factor, is critical for the progression of PCa by regulating a series of target genes by androgen stimulation. A number of co-regulators of AR, including co-activators or co-repressors, have been implicated in AR-mediated gene transcription and PCa progression. Epigenetic regulators, by modifying chromatin integrity and accessibility for transcription regulation without altering DNA sequences, influence the transcriptional activity of AR and further regulate the gene expression of AR target genes in determining cell fate, PCa progression and therapeutic response. In this review, we summarized the structural interaction of AR and epigenetic regulators including histone or DNA methylation, histone acetylation or non-coding RNA, and functional synergy in PCa progression. Importantly, epigenetic regulators have been validated as diagnostic markers and therapeutic targets. A series of epigenetic target drugs have been developed, and have demonstrated the potential to treat PCa alone or in combination with antiandrogens.
Collapse
Affiliation(s)
- Tanggang Deng
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yugang Xiao
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, Guangdong Pharmaceutical University, Guangzhou, China
- School of Clinical Pharmacy, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yi Dai
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, Guangdong Pharmaceutical University, Guangzhou, China
- School of Clinical Pharmacy, Guangdong Pharmaceutical University, Guangzhou, China
| | - Lin Xie
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xiong Li
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, Guangdong Pharmaceutical University, Guangzhou, China
- School of Clinical Pharmacy, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
11
|
Wang W, Yang T, Li D, Huang Y, Bai G, Li Q. LINC00491 promotes cell growth and metastasis through miR-324-5p/ROCK1 in liver cancer. J Transl Med 2021; 19:504. [PMID: 34876144 PMCID: PMC8650505 DOI: 10.1186/s12967-021-03139-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 11/07/2021] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND LINC00491 was involved in some tumors development, but its function in liver cancer has not been reported. This study aimed to investigate LINC00491 expression and function in liver cancer progression. METHODS Sixty liver cancer cases were enrolled. LINC00491, miR-324-5p and rho-associated kinase 1 (ROCK1) expression in liver cancer patients and cells were detected by quantitative reverse transcription-polymerase chain reaction and Western blot. HUH-7 and SK-Hep-1 cells were transfected to modulate LINC00491, miR-324-5p and ROCK1 expression. Cell counting kit-8 assay, colony formation assay, wound healing assay, Transwell experiment, Tunel assay and flow cytometry were performed to detected HUH-7 and SK-Hep-1 cells proliferation, migration, invasion, apoptosis and cell cycle. Biotin-RNA pull-down assay and Dual-Luciferase Reporter Assay was performed to detect the binding among LINC00491, miR-324-5p and ROCK1. Xenograft tumor and lung metastasis was performed using nude mice. Xenograft tumor and lung tissues of mice were experienced immunohistochemistry and hematoxylin-eosin staining. RESULTS LINC00491 was highly expressed in liver cancer cases, associating with poor prognosis. si-LINC00491 inhibited proliferation, colony formation, invasion, migration, and induced cell cycle G1 arrest and apoptosis in HUH-7 and SK-Hep-1 cells. LINC00491 overexpression showed opposite effects. LINC00491 promoted ROCK1 expression by reducing miR-324-5p. miR-324-5p up-regulation or ROCK1 knockdown reversed LINC00491 promotion on liver SK-Hep-1 cells malignant phenotype. LINC00491 facilitated xenograft tumor growth and lung metastasis in mice. CONCLUSION LINC00491 was highly expressed in liver cancer patients, associating with poor prognosis. LINC00491 facilitated liver cancer progression by sponging miR-324-5p/ROCK1. LINC00491 might be a potential treatment target of liver cancer.
Collapse
Affiliation(s)
- Wei Wang
- Department of General Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, China
| | - Tao Yang
- Department of General Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, China
| | - Dongsheng Li
- Department of General Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, China
| | - Yinpeng Huang
- Department of General Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, China
| | - Guang Bai
- Department of General Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, China
| | - Qing Li
- Department of Nephrology, The Third Affiliated Hospital of Jinzhou Medical University, No. 2 Section 5 Heping Road, Jinzhou, 121000, China.
| |
Collapse
|
12
|
Taheri M, Khoshbakht T, Jamali E, Kallenbach J, Ghafouri-Fard S, Baniahmad A. Interaction between Non-Coding RNAs and Androgen Receptor with an Especial Focus on Prostate Cancer. Cells 2021; 10:3198. [PMID: 34831421 PMCID: PMC8619311 DOI: 10.3390/cells10113198] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/10/2021] [Accepted: 11/11/2021] [Indexed: 12/16/2022] Open
Abstract
The androgen receptor (AR) is a member of the nuclear receptor superfamily and has three functional domains, namely the N-terminal, DNA binding, and C-terminal domain. The N-terminal domain harbors potent transactivation functions, whereas the C-terminal domain binds to androgens and antiandrogens used to treat prostate cancer. AR has genomic activity being DNA binding-dependent or through interaction with other DNA-bound transcription factors, as well as a number of non-genomic, non-canonical functions, such as the activation of the ERK, AKT, and MAPK pathways. A bulk of evidence indicates that non-coding RNAs have functional interactions with AR. This type of interaction is implicated in the pathogenesis of human malignancies, particularly prostate cancer. In the current review, we summarize the available data on the role of microRNAs, long non-coding RNAs, and circular RNAs on the expression of AR and modulation of AR signaling, as well as the effects of AR on their expression. Recognition of the complicated interaction between non-coding RNAs and AR has practical importance in the design of novel treatment options, as well as modulation of response to conventional therapeutics.
Collapse
Affiliation(s)
- Mohammad Taheri
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran 1983535511, Iran;
- Institute of Human Genetics, Jena University Hospital, 07747 Jena, Germany;
| | - Tayyebeh Khoshbakht
- Phytochemistry Research Center, Shahid Beheshti University of Medical Sciences, Tehran 1983535511, Iran;
| | - Elena Jamali
- Department of Pathology, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran 1983535511, Iran;
| | - Julia Kallenbach
- Institute of Human Genetics, Jena University Hospital, 07747 Jena, Germany;
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran 1983535511, Iran
| | - Aria Baniahmad
- Institute of Human Genetics, Jena University Hospital, 07747 Jena, Germany;
| |
Collapse
|
13
|
Kimura T, Horikoshi Y, Kuriyagawa C, Niiyama Y. Rho/ROCK Pathway and Noncoding RNAs: Implications in Ischemic Stroke and Spinal Cord Injury. Int J Mol Sci 2021; 22:ijms222111573. [PMID: 34769004 PMCID: PMC8584200 DOI: 10.3390/ijms222111573] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 10/21/2021] [Accepted: 10/24/2021] [Indexed: 01/18/2023] Open
Abstract
Ischemic strokes (IS) and spinal cord injuries (SCI) are major causes of disability. RhoA is a small GTPase protein that activates a downstream effector, ROCK. The up-regulation of the RhoA/ROCK pathway contributes to neuronal apoptosis, neuroinflammation, blood-brain barrier dysfunction, astrogliosis, and axon growth inhibition in IS and SCI. Noncoding RNAs (ncRNAs), such as microRNAs (miRNAs) and long noncoding RNAs (lncRNAs), were previously considered to be non-functional. However, they have attracted much attention because they play an essential role in regulating gene expression in physiological and pathological conditions. There is growing evidence that ROCK inhibitors, such as fasudil and VX-210, can reduce injury in IS and SCI in animal models and clinical trials. Recently, it has been reported that miRNAs are decreased in IS and SCI, while lncRNAs are increased. Inhibiting the Rho/ROCK pathway with miRNAs alleviates apoptosis, neuroinflammation, oxidative stress, and axon growth inhibition in IS and SCI. Further studies are required to explore the significance of ncRNAs in IS and SCI and to establish new strategies for preventing and treating these devastating diseases.
Collapse
Affiliation(s)
- Tetsu Kimura
- Correspondence: ; Tel.: +81-18-884-6175; Fax: +81-18-884-6448
| | | | | | | |
Collapse
|
14
|
Wang C, Chen Q, Xu H. Wnt/β-catenin signal transduction pathway in prostate cancer and associated drug resistance. Discov Oncol 2021; 12:40. [PMID: 35201496 PMCID: PMC8777554 DOI: 10.1007/s12672-021-00433-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 09/01/2021] [Indexed: 11/22/2022] Open
Abstract
Globally, prostate cancer ranks second in cancer burden of the men. It occurs more frequently in black men compared to white or Asian men. Usually, high rates exist for men aged 60 and above. In this review, we focus on the Wnt/β-catenin signal transduction pathway in prostate cancer since many studies have reported that β-catenin can function as an oncogene and is important in Wnt signaling. We also relate its expression to the androgen receptor and MMP-7 protein, both critical to prostate cancer pathogenesis. Some mutations in the androgen receptor also impact the androgen-β-catenin axis and hence, lead to the progression of prostate cancer. We have also reviewed MiRNAs that modulate this pathway in prostate cancer. Finally, we have summarized the impact of Wnt/β-catenin pathway proteins in the drug resistance of prostate cancer as it is a challenging facet of therapy development due to the complexity of signaling pathways interaction and cross-talk.
Collapse
Affiliation(s)
- Chunyang Wang
- Urology Department, PLA General Hospital, Beijing, 100853, China
| | - Qi Chen
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230031, Anhui, China
| | - Huachao Xu
- Department of Urologic Oncology Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230031, Anhui, China.
| |
Collapse
|
15
|
Subramaniam S, Jeet V, Gunter JH, Clements JA, Batra J. Allele-Specific MicroRNA-Mediated Regulation of a Glycolysis Gatekeeper PDK1 in Cancer Metabolism. Cancers (Basel) 2021; 13:cancers13143582. [PMID: 34298795 PMCID: PMC8304593 DOI: 10.3390/cancers13143582] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 07/09/2021] [Accepted: 07/13/2021] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Emerging evidence has revealed that genetic variations in microRNA (miRNA) binding sites called miRSNPs can alter miRNA binding in an allele-specific manner and impart prostate cancer (PCa) risk. Two miRSNPs, rs1530865 (G > C) and rs2357637 (C > A), in the 3' untranslated region of pyruvate dehydrogenase kinase 1 (PDK1) have been previously reported to be associated with PCa risk. However, these results have not been functionally validated. METHODS In silico analysis was used to predict miRNA-PDK1 interactions and was tested using PDK1 knockdown, miRNA overexpression and reporter gene assay. RESULTS PDK1 expression was found to be upregulated in PCa metastasis. Further, our results show that PDK1 suppression reduced the migration, invasion, and glycolysis of PCa cells. Computational predictions showed that miR-3916, miR-3125 and miR-3928 had a higher binding affinity for the C allele than the G allele for the rs1530865 miRSNP which was validated by reporter gene assays. Similarly, miR-2116 and miR-889 had a higher affinity for the A than C allele of the rs2357637 miRSNP. Overexpression of miR-3916 and miR-3125 decreased PDK1 protein levels in cells expressing the rs1530865 SNP C allele, and miR-2116 reduced in cells with the rs2357637 SNP A allele. CONCLUSIONS The present study is the first to report the regulation of the PDK1 gene by miRNAs in an allele-dependent manner and highlights the role of PDK1 in metabolic adaption associated with PCa progression.
Collapse
Affiliation(s)
- Sugarniya Subramaniam
- School of Biomedical Sciences, Faculty of Health, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane 4000, Australia; (S.S.); (V.J.); (J.H.G.); (J.A.C.)
- Australian Prostate Cancer Research Centre-Queensland (APCRC-Q), Translational Research Institute, Queensland University of Technology, Woolloongabba 4102, Australia
| | - Varinder Jeet
- School of Biomedical Sciences, Faculty of Health, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane 4000, Australia; (S.S.); (V.J.); (J.H.G.); (J.A.C.)
- Australian Prostate Cancer Research Centre-Queensland (APCRC-Q), Translational Research Institute, Queensland University of Technology, Woolloongabba 4102, Australia
| | - Jennifer H. Gunter
- School of Biomedical Sciences, Faculty of Health, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane 4000, Australia; (S.S.); (V.J.); (J.H.G.); (J.A.C.)
- Australian Prostate Cancer Research Centre-Queensland (APCRC-Q), Translational Research Institute, Queensland University of Technology, Woolloongabba 4102, Australia
| | - Judith A. Clements
- School of Biomedical Sciences, Faculty of Health, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane 4000, Australia; (S.S.); (V.J.); (J.H.G.); (J.A.C.)
- Australian Prostate Cancer Research Centre-Queensland (APCRC-Q), Translational Research Institute, Queensland University of Technology, Woolloongabba 4102, Australia
| | - Jyotsna Batra
- School of Biomedical Sciences, Faculty of Health, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane 4000, Australia; (S.S.); (V.J.); (J.H.G.); (J.A.C.)
- Australian Prostate Cancer Research Centre-Queensland (APCRC-Q), Translational Research Institute, Queensland University of Technology, Woolloongabba 4102, Australia
- Correspondence: ; Tel.: +61-(0)-734437336
| |
Collapse
|
16
|
Saran U, Chandrasekaran B, Kolluru V, Tyagi A, Nguyen KD, Valadon CL, Shaheen SP, Kong M, Poddar T, Ankem MK, Damodaran C. Diagnostic molecular markers predicting aggressive potential in low-grade prostate cancer. Transl Res 2021; 231:92-101. [PMID: 33279680 DOI: 10.1016/j.trsl.2020.11.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 11/05/2020] [Accepted: 11/30/2020] [Indexed: 12/14/2022]
Abstract
Currently, clinicians rely on clinical nomograms to stratify progression risk at the time of diagnosis in patients with prostate cancer (CaP). However, these tools may not accurately distinguish aggressive potential in low-grade CaP. The current study determined the diagnostic potential of 3 molecular markers (ROCK1, RUNX3, and miR-301a) in terms of their ability to identify which low-grade tumors are likely to progress. Real-time PCR and immunohistochemical analysis were used to assess ROCK1, RUNX3, and miR-301a expression profiles in 118 serum and needle biopsy specimens. Expressions of ROCK1 and miR-301a were found to be significantly higher in Gleason 6 and 7 CaP as compared to BPH, while an inverse trend was observed with RUNX3. Further, incorporation of all 3 molecular markers significantly improved clinical nomograms' diagnostic accuracy and correlated with disease progression. Hence, in conclusion, the inclusion of these 3 molecular markers identified aggressive phenotype and predicted disease progression in low-grade CaP tumors at the time of diagnosis.
Collapse
Affiliation(s)
- Uttara Saran
- Department of Urology, University of Louisville, Louisville, KY
| | | | | | - Ashish Tyagi
- Department of Urology, University of Louisville, Louisville, KY
| | - Kristy D Nguyen
- Department of Urology, University of Louisville, Louisville, KY
| | | | - Saad P Shaheen
- Department of Bioinformatics and Biostatistics, University of Louisville, Louisville, KY
| | | | | | - Murali K Ankem
- Department of Urology, University of Louisville, Louisville, KY
| | | |
Collapse
|
17
|
Keck J, Chambers JP, Yu JJ, Cheng X, Christenson LK, Guentzel MN, Gupta R, Arulanandam BP. Modulation of Immune Response to Chlamydia muridarum by Host miR-135a. Front Cell Infect Microbiol 2021; 11:638058. [PMID: 33928045 PMCID: PMC8076868 DOI: 10.3389/fcimb.2021.638058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 03/15/2021] [Indexed: 11/13/2022] Open
Abstract
Previously, our laboratory established the role of small, noncoding RNA species, i.e., microRNA (miRNA) including miR-135a in anti-chlamydial immunity in infected hosts. We report here chlamydial infection results in decreased miR-135a expression in mouse genital tissue and a fibroblast cell line. Several chemokine and chemokine receptor genes (including CXCL10, CCR5) associated with chlamydial pathogenesis were identified in silico to contain putative miR-135a binding sequence(s) in the 3' untranslated region. The role of miR-135a in the host immune response was investigated using exogenous miR-135a mimic to restore the immune phenotype associated with decreased miR-135a following Chlamydia muridarum (Cm) infection. We observed miR-135a regulation of Cm-primed bone marrow derived dendritic cells (BMDC) via activation of Cm-immune CD4+ T cells for clonal expansion and CCR5 expression. Using a transwell cell migration assay, we explore the role of miR-135a in regulation of genital tract CXCL10 expression and recruitment of CXCR3+ CD4+ T cells via the CXCL10/CXCR3 axis. Collectively, data reported here support miR-135a affecting multiple cellular processes in response to chlamydial infection.
Collapse
Affiliation(s)
- Jonathon Keck
- South Texas Center for Emerging Infectious Diseases, Department of Biology, University of Texas at San Antonio, San Antonio, TX, United States
| | - James P Chambers
- South Texas Center for Emerging Infectious Diseases, Department of Biology, University of Texas at San Antonio, San Antonio, TX, United States
| | - Jieh-Juen Yu
- South Texas Center for Emerging Infectious Diseases, Department of Biology, University of Texas at San Antonio, San Antonio, TX, United States
| | - Xingguo Cheng
- Department of Materials & Bioengineering, Southwest Research Institute, San Antonio, TX, United States
| | - Lane K Christenson
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS, United States
| | - M N Guentzel
- South Texas Center for Emerging Infectious Diseases, Department of Biology, University of Texas at San Antonio, San Antonio, TX, United States
| | - Rishein Gupta
- South Texas Center for Emerging Infectious Diseases, Department of Biology, University of Texas at San Antonio, San Antonio, TX, United States
| | - Bernard P Arulanandam
- South Texas Center for Emerging Infectious Diseases, Department of Biology, University of Texas at San Antonio, San Antonio, TX, United States
| |
Collapse
|
18
|
Yang Y, Liu KY, Liu Q, Cao Q. Androgen Receptor-Related Non-coding RNAs in Prostate Cancer. Front Cell Dev Biol 2021; 9:660853. [PMID: 33869227 PMCID: PMC8049439 DOI: 10.3389/fcell.2021.660853] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 03/12/2021] [Indexed: 12/20/2022] Open
Abstract
Prostate cancer (PCa) is the second leading cause of cancer-related death among men in the United States. Androgen receptor (AR) signaling is the dominant oncogenic pathway in PCa and the main strategy of PCa treatment is to control the AR activity. A large number of patients acquire resistance to Androgen deprivation therapy (ADT) due to AR aberrant activation, resulting in castration-resistant prostate cancer (CRPC). Understanding the molecular mechanisms underlying AR signaling in the PCa is critical to identify new therapeutic targets for PCa patients. The recent advances in high-throughput RNA sequencing (RNA-seq) techniques identified an increasing number of non-coding RNAs (ncRNAs) that play critical roles through various mechanisms in different diseases. Some ncRNAs have shown great potentials as biomarkers and therapeutic targets. Many ncRNAs have been investigated to regulate PCa through direct association with AR. In this review, we aim to comprehensively summarize recent findings of the functional roles and molecular mechanisms of AR-related ncRNAs as AR regulators or targets in the progression of PCa.
Collapse
Affiliation(s)
- Yongyong Yang
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States.,Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Kilia Y Liu
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States.,Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Qi Liu
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States.,Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Qi Cao
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States.,Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
19
|
Zheng K, Hu F, Zhou Y, Zhang J, Zheng J, Lai C, Xiong W, Cui K, Hu YZ, Han ZT, Zhang HH, Chen JG, Man HY, Liu D, Lu Y, Zhu LQ. miR-135a-5p mediates memory and synaptic impairments via the Rock2/Adducin1 signaling pathway in a mouse model of Alzheimer's disease. Nat Commun 2021; 12:1903. [PMID: 33771994 PMCID: PMC7998005 DOI: 10.1038/s41467-021-22196-y] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 02/25/2021] [Indexed: 12/13/2022] Open
Abstract
Aberrant regulation of microRNAs (miRNAs) has been implicated in the pathogenesis of Alzheimer's disease (AD), but most abnormally expressed miRNAs found in AD are not regulated by synaptic activity. Here we report that dysfunction of miR-135a-5p/Rock2/Add1 results in memory/synaptic disorder in a mouse model of AD. miR-135a-5p levels are significantly reduced in excitatory hippocampal neurons of AD model mice. This decrease is tau dependent and mediated by Foxd3. Inhibition of miR-135a-5p leads to synaptic disorder and memory impairments. Furthermore, excess Rock2 levels caused by loss of miR-135a-5p plays an important role in the synaptic disorder of AD via phosphorylation of Ser726 on adducin 1 (Add1). Blocking the phosphorylation of Ser726 on Add1 with a membrane-permeable peptide effectively rescues the memory impairments in AD mice. Taken together, these findings demonstrate that synaptic-related miR-135a-5p mediates synaptic/memory deficits in AD via the Rock2/Add1 signaling pathway, illuminating a potential therapeutic strategy for AD.
Collapse
Affiliation(s)
- Kai Zheng
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fan Hu
- Department of Pathophysiology, Key Lab of Neurological Disorder of Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Yang Zhou
- Department of Pathophysiology, Key Lab of Neurological Disorder of Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Juan Zhang
- Department of Pathophysiology, Key Lab of Neurological Disorder of Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Jie Zheng
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chuan Lai
- Department of Pathophysiology, Key Lab of Neurological Disorder of Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Wan Xiong
- Department of Pathophysiology, Key Lab of Neurological Disorder of Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Ke Cui
- Department of Pathophysiology, Key Lab of Neurological Disorder of Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Ya-Zhuo Hu
- Beijing Key Laboratory of Aging and Geriatrics, National Clinical Research Center for Geriatric Disease, Institute of Geriatrics, Chinese PLA General Hospital and Chinese PLA Medical Academy, Beijing, P. R. China
| | - Zhi-Tao Han
- Beijing Key Laboratory of Aging and Geriatrics, National Clinical Research Center for Geriatric Disease, Institute of Geriatrics, Chinese PLA General Hospital and Chinese PLA Medical Academy, Beijing, P. R. China
| | - Hong-Hong Zhang
- Beijing Key Laboratory of Aging and Geriatrics, National Clinical Research Center for Geriatric Disease, Institute of Geriatrics, Chinese PLA General Hospital and Chinese PLA Medical Academy, Beijing, P. R. China
| | - Jian-Guo Chen
- The Institute of Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Heng-Ye Man
- Department of Biology, Boston University, Boston, MA, USA
| | - Dan Liu
- The Institute of Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Youming Lu
- The Institute of Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, P. R. China.
| | - Ling-Qiang Zhu
- Department of Pathophysiology, Key Lab of Neurological Disorder of Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China.
- The Institute of Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, P. R. China.
| |
Collapse
|
20
|
miR-135a inhibits malignant proliferation and diffusion of non-small cell lung cancer cells by down-regulating ROCK1 protein. Biosci Rep 2021; 40:225118. [PMID: 32484204 PMCID: PMC7295638 DOI: 10.1042/bsr20201276] [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: 04/20/2020] [Revised: 05/21/2020] [Accepted: 05/26/2020] [Indexed: 12/25/2022] Open
Abstract
Objective: To seek the clinical significance and regulatory mechanism of miR-135a and Rho-associated protein kinase 1 (ROCK1) in non-small cell lung cancer (NSCLC). Methods: NSCLC cells were purchased, and miR-135a-mimics, miR-135a-inhibitor, miR-NC, si-ROCK1 and Sh-ROCK1 were transfected into NSCLC cells HCC827 and NCI-H524. qRT-PCR and Western blot were used to detect the expression of miR-135a, ROCK1, Bax, Caspase3, Bcl-2, N-cadherin, vimentin and E-cadherin. MTT, scratch test, Transwell and flow cytometry were used to analyze the cell proliferation, migration, invasion and apoptosis. Results: miR-135a was low expressed in serum of NSCLC group, while ROCK1 was opposite. miR-135a low level or ROCK1 high level was associated with poor prognosis of NSCLC and lower 3-year OS. Over-expression of miR-135a and inhibition of ROCK1 expression could control malignant growth and diffusion of cells and expression of Bcl-2, N-cadherin and vimentin proteins, and promote apoptosis and expression of Bax, Caspase3 and E-cadherin proteins. After transfection of miR-135a-mimics+sh-ROCK1 to HCC827 and NCI-H524, the malignant proliferation and diffusion behavior of the cells were not different from those of the miR-NC group with no transfection sequence. The double luciferase report revealed that miR-135a has a targeting relationship with ROCK1. Conclusion: miR-135a is abnormally down-regulated in NSCLC. As a serum indicator, miR-135a has the potential to diagnose NSCLC and predict prognosis. The up-regulated expression of miR-135a protein can down-regulate the ROCK1 protein, inhibit the malignant proliferation, migration, invasion, EMT and other diffusion behaviors of NSCLC cells, and increase the apoptosis ability of cells.
Collapse
|
21
|
Arrighetti N, Beretta GL. miRNAs as Therapeutic Tools and Biomarkers for Prostate Cancer. Pharmaceutics 2021; 13:380. [PMID: 33805590 PMCID: PMC7999286 DOI: 10.3390/pharmaceutics13030380] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/10/2021] [Accepted: 03/11/2021] [Indexed: 12/14/2022] Open
Abstract
Prostate cancer (PCa) is the fifth cause of tumor-related deaths in man worldwide. Despite the considerable improvement in the clinical management of PCa, several limitations emerged both in the screening for early diagnosis and in the medical treatment. The use of prostate-specific antigen (PSA)-based screening resulted in patients' overtreatment and the standard therapy of patients suffering from locally advanced/metastatic tumors (e.g., radical prostatectomy, radiotherapy, and androgen deprivation therapy) showed time-limited efficacy with patients undergoing progression toward the lethal metastatic castration-resistant PCa (mCRPC). Although valuable alternative therapeutic options have been recently proposed (e.g., docetaxel, cabazitaxel, abiraterone, enzalutamide, and sipuleucel-T), mCRPC remains incurable. Based on this background, there is an urgent need to identify new and more accurate prostate-specific biomarkers for PCa diagnosis and prognosis and to develop innovative medical approaches to counteract mCRPC. In this context, microRNA (miRNAs) emerged as potential biomarkers in prostate tissues and biological fluids and appeared to be promising therapeutic targets/tools for cancer therapy. Here we overview the recent literature and summarize the achievements of using miRNAs as biomarkers and therapeutic targets/tools for fighting PCa.
Collapse
Affiliation(s)
| | - Giovanni Luca Beretta
- Molecular Pharmacology Unit, Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy;
| |
Collapse
|
22
|
Deng B, Deng J, Yi X, Zou Y, Li C. ROCK2 Promotes Osteosarcoma Growth and Glycolysis by Up-Regulating HKII via Phospho-PI3K/AKT Signalling. Cancer Manag Res 2021; 13:449-462. [PMID: 33500659 PMCID: PMC7823140 DOI: 10.2147/cmar.s279496] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Accepted: 10/24/2020] [Indexed: 01/14/2023] Open
Abstract
Background Osteosarcoma (OS) is a malignant bone tumour that exhibits a high mortality. While tumours thrive in a state of malnutrition, the mechanism by which OS cells adapt to metabolic stress through metabolic reprogramming remains unclear. Methods We analysed the expression of ROCK2 in osteosarcoma tissues by RT-qPCR and Western blot. Cell proliferation were analysed using CCK8, EdU and colony formation assays. The level of cell glycolysis was detected by glucose-6 phosphate, glucose consumption, lactate production and ATP levels. Results Herein, our study showed that ROCK2 expression in OS tissues was higher than in adjacent tissues. Functional assays have demonstrated that ROCK2 contributes to the growth of OS cells by inducing aerobic glycolysis. The current study revealed that ROCK2 knockdown decreased the levels of mitochondrial hexokinase II (HKII). And also indicated that ROCK2 served as a key enzyme in glycolysis and that it served an important role in tumour growth. A significant positive correlation was identified between the mRNA and protein expressions of ROCK2 and HKII, further demonstrating that ROCK2-induced glycolysis and proliferation was dependent on HKII expression in OS cells. Mechanistically, ROCK2 promotes HKII expression by activating the phospho-PI3K/AKT signalling pathway. Conclusion Taken together, the results of the current study linked the two drivers of OS growth and aerobic glycolysis and identified a new mechanism of ROCK2 control in OS.
Collapse
Affiliation(s)
- Binbin Deng
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| | - Jianyong Deng
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| | - Xuan Yi
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| | - Yeqing Zou
- Jiangxi Province Key Laboratory of Molecular Medicine, Second Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| | - Chen Li
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| |
Collapse
|
23
|
Grzywa TM, Klicka K, Włodarski PK. Regulators at Every Step-How microRNAs Drive Tumor Cell Invasiveness and Metastasis. Cancers (Basel) 2020; 12:E3709. [PMID: 33321819 PMCID: PMC7763175 DOI: 10.3390/cancers12123709] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/03/2020] [Accepted: 12/07/2020] [Indexed: 02/06/2023] Open
Abstract
Tumor cell invasiveness and metastasis are the main causes of mortality in cancer. Tumor progression is composed of many steps, including primary tumor growth, local invasion, intravasation, survival in the circulation, pre-metastatic niche formation, and metastasis. All these steps are strictly controlled by microRNAs (miRNAs), small non-coding RNA that regulate gene expression at the post-transcriptional level. miRNAs can act as oncomiRs that promote tumor cell invasion and metastasis or as tumor suppressor miRNAs that inhibit tumor progression. These miRNAs regulate the actin cytoskeleton, the expression of extracellular matrix (ECM) receptors including integrins and ECM-remodeling enzymes comprising matrix metalloproteinases (MMPs), and regulate epithelial-mesenchymal transition (EMT), hence modulating cell migration and invasiveness. Moreover, miRNAs regulate angiogenesis, the formation of a pre-metastatic niche, and metastasis. Thus, miRNAs are biomarkers of metastases as well as promising targets of therapy. In this review, we comprehensively describe the role of various miRNAs in tumor cell migration, invasion, and metastasis.
Collapse
Affiliation(s)
- Tomasz M. Grzywa
- Department of Methodology, Medical University of Warsaw, 02-091 Warsaw, Poland; (T.M.G.); (K.K.)
- Doctoral School, Medical University of Warsaw, 02-091 Warsaw, Poland
- Department of Immunology, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Klaudia Klicka
- Department of Methodology, Medical University of Warsaw, 02-091 Warsaw, Poland; (T.M.G.); (K.K.)
- Doctoral School, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Paweł K. Włodarski
- Department of Methodology, Medical University of Warsaw, 02-091 Warsaw, Poland; (T.M.G.); (K.K.)
| |
Collapse
|
24
|
Yuan F, Yun Y, Fan H, Li Y, Lu L, Liu J, Feng W, Chen SY. MicroRNA-135a Protects Against Ethanol-Induced Apoptosis in Neural Crest Cells and Craniofacial Defects in Zebrafish by Modulating the Siah1/p38/p53 Pathway. Front Cell Dev Biol 2020; 8:583959. [PMID: 33134300 PMCID: PMC7561719 DOI: 10.3389/fcell.2020.583959] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 09/14/2020] [Indexed: 12/24/2022] Open
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs that are involved in various biological processes, including apoptosis, by regulating gene expression. This study was designed to test the hypothesis that ethanol-induced downregulation of miR-135a contributes to ethanol-induced apoptosis in neural crest cells (NCCs) by upregulating Siah1 and activating the p38 mitogen-activated protein kinase (MAPK)/p53 pathway. We found that treatment with ethanol resulted in a significant decrease in miR-135a expression in both NCCs and zebrafish embryos. Ethanol-induced downregulation of miR-135a resulted in the upregulation of Siah1 and the activation of the p38 MAPK/p53 pathway and increased apoptosis in NCCs and zebrafish embryos. Ethanol exposure also resulted in growth retardation and developmental defects that are characteristic of fetal alcohol spectrum disorders (FASD) in zebrafish. Overexpression of miRNA-135a significantly reduced ethanol-induced upregulation of Siah1 and the activation of the p38 MAPK/p53 pathway and decreased ethanol-induced apoptosis in NCCs and zebrafish embryos. In addition, ethanol-induced growth retardation and craniofacial defects in zebrafish larvae were dramatically diminished by the microinjection of miRNA-135a mimics. These results demonstrated that ethanol-induced downregulation of miR-135a contributes to ethanol-induced apoptosis in NCCs by upregulating Siah1 and activating the p38 MAPK/p53 pathway and that the overexpression of miRNA-135a can protect against ethanol-induced apoptosis in NCCs and craniofacial defects in a zebrafish model of FASD.
Collapse
Affiliation(s)
- Fuqiang Yuan
- Department of Pharmacology and Toxicology, University of Louisville Health Sciences Center, Louisville, KY, United States.,University of Louisville Alcohol Research Center, Louisville, KY, United States
| | - Yang Yun
- Department of Pharmacology and Toxicology, University of Louisville Health Sciences Center, Louisville, KY, United States.,University of Louisville Alcohol Research Center, Louisville, KY, United States.,College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, China
| | - Huadong Fan
- Department of Pharmacology and Toxicology, University of Louisville Health Sciences Center, Louisville, KY, United States.,University of Louisville Alcohol Research Center, Louisville, KY, United States
| | - Yihong Li
- Department of Pharmacology and Toxicology, University of Louisville Health Sciences Center, Louisville, KY, United States.,University of Louisville Alcohol Research Center, Louisville, KY, United States
| | - Lanhai Lu
- Department of Pharmacology and Toxicology, University of Louisville Health Sciences Center, Louisville, KY, United States.,University of Louisville Alcohol Research Center, Louisville, KY, United States
| | - Jie Liu
- Department of Pharmacology and Toxicology, University of Louisville Health Sciences Center, Louisville, KY, United States.,University of Louisville Alcohol Research Center, Louisville, KY, United States
| | - Wenke Feng
- Department of Pharmacology and Toxicology, University of Louisville Health Sciences Center, Louisville, KY, United States.,University of Louisville Alcohol Research Center, Louisville, KY, United States.,Department of Medicine, University of Louisville, Louisville, KY, United States
| | - Shao-Yu Chen
- Department of Pharmacology and Toxicology, University of Louisville Health Sciences Center, Louisville, KY, United States.,University of Louisville Alcohol Research Center, Louisville, KY, United States
| |
Collapse
|
25
|
Nalairndran G, Hassan Abdul Razack A, Mai C, Fei‐Lei Chung F, Chan K, Hii L, Lim W, Chung I, Leong C. Phosphoinositide-dependent Kinase-1 (PDPK1) regulates serum/glucocorticoid-regulated Kinase 3 (SGK3) for prostate cancer cell survival. J Cell Mol Med 2020; 24:12188-12198. [PMID: 32926495 PMCID: PMC7578863 DOI: 10.1111/jcmm.15876] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 08/19/2020] [Accepted: 08/27/2020] [Indexed: 02/05/2023] Open
Abstract
Prostate cancer (PCa) is the most common malignancy and is the second leading cause of cancer among men globally. Using a kinome-wide lentiviral small-hairpin RNA (shRNA) library screen, we identified phosphoinositide-dependent kinase-1 (PDPK1) as a potential mediator of cell survival in PCa cells. We showed that knock-down of endogenous human PDPK1 induced significant tumour-specific cell death in PCa cells (DU145 and PC3) but not in the normal prostate epithelial cells (RWPE-1). Further analyses revealed that PDPK1 mediates cancer cell survival predominantly via activation of serum/glucocorticoid-regulated kinase 3 (SGK3). Knock-down of endogenous PDPK1 in DU145 and PC3 cells significantly reduced SGK3 phosphorylation while ectopic expression of a constitutively active SGK3 completely abrogated the apoptosis induced by PDPK1. In contrast, no such effect was observed in SGK1 and AKT phosphorylation following PDPK1 knock-down. Importantly, PDPK1 inhibitors (GSK2334470 and BX-795) significantly reduced tumour-specific cell growth and synergized docetaxel sensitivity in PCa cells. In summary, our results demonstrated that PDPK1 mediates PCa cells' survival through SGK3 signalling and suggest that inactivation of this PDPK1-SGK3 axis may potentially serve as a novel therapeutic intervention for future treatment of PCa.
Collapse
Affiliation(s)
- Geetha Nalairndran
- Department of PharmacologyFaculty of MedicineUniversity of MalayaKuala LumpurMalaysia
| | | | - Chun‐Wai Mai
- Center for Cancer and Stem Cell ResearchInstitute for ResearchDevelopment and Innovation (IRDI)International Medical UniversityKuala LumpurMalaysia
- School of PharmacyInternational Medical UniversityKuala LumpurMalaysia
| | - Felicia Fei‐Lei Chung
- Mechanisms of Carcinogenesis Section (MCA)Epigenetics Group (EGE)International Agency for Research on Cancer World Health OrganizationLyonFrance
| | - Kok‐Keong Chan
- School of MedicineInternational Medical UniversityKuala LumpurMalaysia
| | - Ling‐Wei Hii
- Center for Cancer and Stem Cell ResearchInstitute for ResearchDevelopment and Innovation (IRDI)International Medical UniversityKuala LumpurMalaysia
- School of PharmacyInternational Medical UniversityKuala LumpurMalaysia
- School of Postgraduate StudiesInternational Medical UniversityKuala LumpurMalaysia
| | - Wei‐Meng Lim
- Center for Cancer and Stem Cell ResearchInstitute for ResearchDevelopment and Innovation (IRDI)International Medical UniversityKuala LumpurMalaysia
- School of PharmacyInternational Medical UniversityKuala LumpurMalaysia
- School of Postgraduate StudiesInternational Medical UniversityKuala LumpurMalaysia
| | - Ivy Chung
- Department of PharmacologyFaculty of MedicineUniversity of MalayaKuala LumpurMalaysia
- Faculty of MedicineUniversity of Malaya Cancer Research InstituteUniversity of MalayaKuala LumpurMalaysia
| | - Chee‐Onn Leong
- Center for Cancer and Stem Cell ResearchInstitute for ResearchDevelopment and Innovation (IRDI)International Medical UniversityKuala LumpurMalaysia
- School of PharmacyInternational Medical UniversityKuala LumpurMalaysia
| |
Collapse
|
26
|
She JK, Fu DN, Zhen D, Gong GH, Zhang B. LINC01087 is Highly Expressed in Breast Cancer and Regulates the Malignant Behavior of Cancer Cells Through miR-335-5p/Rock1. Onco Targets Ther 2020; 13:9771-9783. [PMID: 33061456 PMCID: PMC7533226 DOI: 10.2147/ott.s255994] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 08/14/2020] [Indexed: 12/19/2022] Open
Abstract
PURPOSE Long non-coding RNA is involved in the genesis and development of various tumors, and it has been found through database screening that LINC01087 is highly expressed in breast cancer (BC), but mechanisms of LINC01087 in BC are still under investigation. Therefore, this study aimed to study relevant mechanisms of LINC01087 in BC to provide potential therapeutic targets for the disease in clinic practice. PATIENTS AND METHODS The qRT-PCR assay was applied to determine the LINC01087 expression in BC, and the cell counting kit-8 (CCK8) assay, transwell assay, and flow cytometry were used to analyze the proliferation, apoptosis, and invasion of breast cancer cells (BCCs), respectively. The Western blot assay was used to determine the ROCK1 expression, and the luciferase reporter gene assay, RNA-binding protein immunoprecipitation (RIP), and RNA pull-down assays were applied to study the interaction between LINC01087 and miR-335-5p. Moreover, tumor xenotransplantation was conducted in nude mice to explore the effects of LINC01087 on BCCs. RESULTS The qRT-PCR assay revealed that the LINC01087 expression in BC tissues was higher than that in corresponding tumor-adjacent tissues, and survival analysis revealed an unfavorable prognosis of patients with high expression of LINC01087. Down-regulation of LINC01087 could slow down the proliferation, invasion, and migration of BCCs and accelerate apoptosis of them in vitro. Luciferase reporter gene assay results revealed that LINC01087 enhanced the expression of ROCK1 by regulating miR-335-5p, and LINC01087 could be adopted as a miR-335-5p sponge to inhibit ROCK1 expression. CONCLUSION LINC01087 is overexpressed in cases with BC, and patients with high expression of it suffer a poor survival. Furthermore, LINC01087 can act as a miR-335-5p sponge to affect the expression of ROCK1 and affect the invasion and migration of BCCs.
Collapse
Affiliation(s)
- Ji-Kai She
- Medicinal Chemistry and Pharmacology Institute, Inner Mongolia University for the Nationalities, Tongliao, Inner Mongolia, People’s Republic of China
- Inner Mongolia Key Laboratory of Mongolian Medicine Pharmacology for Cardio-Cerebral Vascular System, Tongliao, Inner Mongolia, People’s Republic of China
| | - Dan-Ni Fu
- Medicinal Chemistry and Pharmacology Institute, Inner Mongolia University for the Nationalities, Tongliao, Inner Mongolia, People’s Republic of China
- Inner Mongolia Key Laboratory of Mongolian Medicine Pharmacology for Cardio-Cerebral Vascular System, Tongliao, Inner Mongolia, People’s Republic of China
| | - Dong Zhen
- Medicinal Chemistry and Pharmacology Institute, Inner Mongolia University for the Nationalities, Tongliao, Inner Mongolia, People’s Republic of China
- Inner Mongolia Key Laboratory of Mongolian Medicine Pharmacology for Cardio-Cerebral Vascular System, Tongliao, Inner Mongolia, People’s Republic of China
| | - Guo-Hua Gong
- Inner Mongolia Key Laboratory of Mongolian Medicine Pharmacology for Cardio-Cerebral Vascular System, Tongliao, Inner Mongolia, People’s Republic of China
- First Clinical Medical of Inner Mongolia University for Nationalities, Tongliao, Inner Mongolia, People’s Republic of China
| | - Bin Zhang
- Inner Mongolia Key Laboratory of Mongolian Medicine Pharmacology for Cardio-Cerebral Vascular System, Tongliao, Inner Mongolia, People’s Republic of China
- First Clinical Medical of Inner Mongolia University for Nationalities, Tongliao, Inner Mongolia, People’s Republic of China
| |
Collapse
|
27
|
Al-Othman N, Alhendi A, Ihbaisha M, Barahmeh M, Alqaraleh M, Al-Momany BZ. Role of CD44 in breast cancer. Breast Dis 2020; 39:1-13. [PMID: 31839599 DOI: 10.3233/bd-190409] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Breast cancer (BC) is among the most prevalent type of malignancy affecting females worldwide. BC is classified into different types according to the status of the expression of receptors such as estrogen receptor (ER), human epidermal growth factor receptor 2 (HER2), and progesterone receptor (PR). Androgen receptor (AR) appears to be a promising therapeutic target of BC. Binding of 5α-dihydrotestosterone (DHT) to AR controls the expression of microRNA (miRNA) molecules in BC, consequently, affecting protein expression. One of these proteins is the transmembrane glycoprotein cluster of differentiation 44 (CD44). Remarkably, CD44 is a common marker of cancer stem cells in BC. It functions as a co-receptor for a broad diversity of extracellular matrix ligands. Several ligands, primarily hyaluronic acid (HA), can interact with CD44 and mediate its functions. CD44 promotes a variety of functions independently or in cooperation with other cell-surface receptors through activation of varied signaling pathways like Rho GTPases, Ras-MAPK, and PI3K/AKT pathways to regulate cell adhesion, migration, survival, invasion, and epithelial-mesenchymal transition. In this review, we present the relations between AR, miRNA, and CD44 and their roles in BC.
Collapse
Affiliation(s)
- Nihad Al-Othman
- Division of Anatomy, Biochemistry, and Genetics, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Ala' Alhendi
- Division of Anatomy, Biochemistry, and Genetics, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Manal Ihbaisha
- Division of Anatomy, Biochemistry, and Genetics, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Myassar Barahmeh
- Division of Anatomy, Biochemistry, and Genetics, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | | | | |
Collapse
|
28
|
Wang N, Yang Y, Pang M, Du C, Chen Y, Li S, Tian Z, Feng F, Wang Y, Chen Z, Liu B, Rong L. MicroRNA-135a-5p Promotes the Functional Recovery of Spinal Cord Injury by Targeting SP1 and ROCK. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 22:1063-1077. [PMID: 33294293 PMCID: PMC7691148 DOI: 10.1016/j.omtn.2020.08.035] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 08/28/2020] [Indexed: 01/18/2023]
Abstract
Emerging evidence indicates that microRNAs play a pivotal role in neural remodeling after spinal cord injury (SCI). This study aimed to investigate the mechanisms of miR-135a-5p in regulating the functional recovery of SCI by impacting its target genes and downstream signaling. The gene transfection assay and luciferase reporter assay confirmed the target relationship between miR-135a-5p and its target genes (specificity protein 1 [SP1] and Rho-associated kinase [ROCK]1/2). By establishing the H2O2-induced injury model, miR-135a-5p transfection was found to inhibit the apoptosis of PC12 cells by downregulating the SP1 gene, which subsequently induced downregulation of pro-apoptotic proteins (Bax, cleaved caspase-3) and upregulation of anti-apoptotic protein Bcl-2. By measuring the neurite lengths of PC12 cells, miR-135a-5p transfection was found to promote axon outgrowth by downregulating the ROCK1/2 gene, which subsequently caused upregulation of phosphate protein kinase B (AKT) and phosphate glycogen synthase kinase 3β (GSK3β). Use of the rat SCI models showed that miR-135a-5p could increase the Basso, Beattie, and Bresnahan (BBB) scores, indicating neurological function recovery. In conclusion, the miR-135a-5p-SP1-Bax/Bcl-2/caspase-3 and miR-135a-5p-ROCK-AKT/GSK3β axes are involved in functional recovery of SCI by regulating neural apoptosis and axon regeneration, respectively, and thus can be promising effective therapeutic strategies in SCI.
Collapse
Affiliation(s)
- Nanxiang Wang
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, No. 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, People's Republic of China
| | - Yang Yang
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, No. 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, People's Republic of China
| | - Mao Pang
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, No. 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, People's Republic of China
| | - Cong Du
- Cell-Gene Therapy Translational Medicine Research Center, The Third Affiliated Hospital of Sun Yat-Sen University, No. 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, People's Republic of China
| | - Yuyong Chen
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, No. 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, People's Republic of China
| | - Simin Li
- Department of Cariology, Endodontology and Periodontology, University Leipzig, Liebigstrasse 12, 04103 Leipzig, Germany
| | - Zhenming Tian
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, No. 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, People's Republic of China
| | - Feng Feng
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, No. 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, People's Republic of China
| | - Yang Wang
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, No. 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, People's Republic of China
| | - Zhenxiang Chen
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, No. 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, People's Republic of China
| | - Bin Liu
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, No. 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, People's Republic of China
| | - Limin Rong
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, No. 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, People's Republic of China
| |
Collapse
|
29
|
Domińska K, Kowalska K, Urbanek KA, Habrowska-Górczyńska DE, Ochędalski T, Piastowska Ciesielska AW. The Impact of Ang-(1-9) and Ang-(3-7) on the Biological Properties of Prostate Cancer Cells by Modulation of Inflammatory and Steroidogenesis Pathway Genes. Int J Mol Sci 2020; 21:ijms21176227. [PMID: 32872192 PMCID: PMC7504072 DOI: 10.3390/ijms21176227] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 08/23/2020] [Accepted: 08/26/2020] [Indexed: 01/01/2023] Open
Abstract
The local renin–angiotensin system (RAS) plays an important role in the pathophysiology of the prostate, including cancer development and progression. The Ang-(1-9) and Ang-(3-7) are the less known active peptides of RAS. This study examines the influence of these two peptide hormones on the metabolic activity, proliferation and migration of prostate cancer cells. Significant changes in MTT dye reduction were observed depending on the type of angiotensin and its concentration as well as time of incubation. Ang-(1-9) did not regulate the 2D cell division of either prostate cancer lines however, it reduced the size of LNCaP colonies formed in soft agar, maybe through down-regulation of the HIF1a gene. Ang-(3-7) increased the number of PC3 cells in the S phase and improved anchorage-independent growth as well as mobility. In this case, a significant increase in MKI67, BIRC5, and CDH-1 gene expression was also observed as well as all members of the NF-kB family. Furthermore, we speculate that this peptide can repress the proliferation of LNCaP cells by NOS3-mediated G2/M cell cycle arrest. No changes in expression of BIRC5 and BCL2/BAX ratio were observed but a decrease mRNA proapoptotic BAD gene was seen. In the both lines, Ang-(3-7) improved ROCK1 gene expression however, increased VEGF and NOS3 mRNA was only seen in the PC3 or LNCaP cells, respectively. Interestingly, it appears that Ang-(1-9) and Ang-(3-7) can modulate the level of steroidogenic enzymes responsible for converting cholesterol to testosterone in both prostate cancer lines. Furthermore, in PC3 cells, Ang-(1-9) upregulated AR expression while Ang-(3-7) upregulated the expression of both estrogen receptor genes. Ang-(1-9) and Ang-(3-7) can impact on biological properties of prostate cancer cells by modulating inflammatory and steroidogenesis pathway genes, among others.
Collapse
Affiliation(s)
- Kamila Domińska
- Department of Comparative Endocrinology, Medical University of Lodz, Zeligowskiego 7/9, 90-752 Lodz, Poland;
- Correspondence:
| | - Karolina Kowalska
- Department of Cell Cultures and Genomic Analysis, Medical University of Lodz, Zeligowskiego 7/9, 90-752 Lodz, Poland; (K.K.); (K.A.U.); (D.E.H.-G.); (A.W.P.C.)
| | - Kinga Anna Urbanek
- Department of Cell Cultures and Genomic Analysis, Medical University of Lodz, Zeligowskiego 7/9, 90-752 Lodz, Poland; (K.K.); (K.A.U.); (D.E.H.-G.); (A.W.P.C.)
| | - Dominika Ewa Habrowska-Górczyńska
- Department of Cell Cultures and Genomic Analysis, Medical University of Lodz, Zeligowskiego 7/9, 90-752 Lodz, Poland; (K.K.); (K.A.U.); (D.E.H.-G.); (A.W.P.C.)
| | - Tomasz Ochędalski
- Department of Comparative Endocrinology, Medical University of Lodz, Zeligowskiego 7/9, 90-752 Lodz, Poland;
| | - Agnieszka Wanda Piastowska Ciesielska
- Department of Cell Cultures and Genomic Analysis, Medical University of Lodz, Zeligowskiego 7/9, 90-752 Lodz, Poland; (K.K.); (K.A.U.); (D.E.H.-G.); (A.W.P.C.)
| |
Collapse
|
30
|
Shen H, Zheng E, Yang Z, Yang M, Xu X, Zhou Y, Ni J, Li R, Zhao G. YRDC is upregulated in non-small cell lung cancer and promotes cell proliferation by decreasing cell apoptosis. Oncol Lett 2020; 20:43-52. [PMID: 32565932 PMCID: PMC7285791 DOI: 10.3892/ol.2020.11560] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Accepted: 09/20/2019] [Indexed: 02/06/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) is the leading cause of cancer-associated mortality worldwide. yrdC N6-threonylcarbamoltransferase domain containing protein (YRDC) has been demonstrated to be involved in the formation of threonylcarbamoyladenosine in transfer ribonucleic acid. However, the molecular mechanisms underlying NSCLC progression remain largely unclear. The present study revealed that YRDC was upregulated in NSCLC samples compared with adjacent non-cancerous tissues by analyzing datasets obtained from the Gene Expression Omnibus and The Cancer Genome Atlas. Higher expression of YRDC was associated with overall survival time and disease-free survival time in patients with NSCLC, particularly in lung adenocarcinoma. Furthermore, knockdown of YRDC in NSCLS cell lines significantly suppressed cell growth and cell colony formation in vitro. Additionally, the results demonstrated that silencing of YRDC induced apoptosis of A549 cells. Then, the protein-protein interaction networks associated with yrdC N6-threonylcarbamoltransferase domain containing protein (YRDC) in NSCLC were subsequently constructed to investigate the potential molecular mechanism underlying the role of YRDC in NSCLC. The results revealed that YRDC was involved in the regulation of spliceosomes, ribosomes, the p53 signaling pathway, proteasomes, the cell cycle and DNA replication. The present study demonstrated that YRDC may serve as a novel biomarker for the prognosis prediction and treatment of NSCLC.
Collapse
Affiliation(s)
- Haibo Shen
- Cardiothoracic Surgery Department, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang 315010, P.R. China
| | - Enkuo Zheng
- Cardiothoracic Surgery Department, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang 315010, P.R. China
| | - Zhenhua Yang
- Cardiothoracic Surgery Department, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang 315010, P.R. China
| | - Minglei Yang
- Cardiothoracic Surgery Department, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang 315010, P.R. China
| | - Xiang Xu
- Cardiothoracic Surgery Department, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang 315010, P.R. China
| | - Yinjie Zhou
- Cardiothoracic Surgery Department, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang 315010, P.R. China
| | - Junjun Ni
- Cardiothoracic Surgery Department, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang 315010, P.R. China
| | - Rui Li
- Cardiothoracic Surgery Department, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang 315010, P.R. China
| | - Guofang Zhao
- Cardiothoracic Surgery Department, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang 315010, P.R. China
| |
Collapse
|
31
|
Coordinated AR and microRNA regulation in prostate cancer. Asian J Urol 2020; 7:233-250. [PMID: 32742925 PMCID: PMC7385519 DOI: 10.1016/j.ajur.2020.06.003] [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: 08/11/2019] [Revised: 03/22/2020] [Accepted: 04/17/2020] [Indexed: 12/26/2022] Open
Abstract
The androgen receptor (AR) remains a key driver of prostate cancer (PCa) progression, even in the advanced castrate-resistant stage, where testicular androgens are absent. It is therefore of critical importance to understand the molecular mechanisms governing its activity and regulation during prostate tumourigenesis. MicroRNAs (miRs) are small ∼22 nt non-coding RNAs that regulate target gene, often through association with 3′ untranslated regions (3′UTRs) of transcripts. They display dysregulation during cancer progression, can function as oncogenes or tumour suppressors, and are increasingly recognised as targets or regulators of hormonal action. Thus, understanding factors which modulate miRs synthesis is essential. There is increasing evidence for complex and dynamic bi-directional cross-talk between the multi-step miR biogenesis cascade and the AR signalling axis in PCa. This review summarises the wealth of mechanisms by which miRs are regulated by AR, and conversely, how miRs impact AR's transcriptional activity, including that of AR splice variants. In addition, we assess the implications of the convergence of these pathways on the clinical employment of miRs as PCa biomarkers and therapeutic targets.
Collapse
|
32
|
Jin W, Fei X, Wang X, Song Y, Chen F. Detection and Prognosis of Prostate Cancer Using Blood-Based Biomarkers. Mediators Inflamm 2020; 2020:8730608. [PMID: 32454797 PMCID: PMC7218965 DOI: 10.1155/2020/8730608] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 03/24/2020] [Accepted: 04/27/2020] [Indexed: 12/15/2022] Open
Abstract
Prostate cancer (PCa) is second only to lung cancer as a cause of death. Clinical assessment of patients and treatment efficiency therefore depend on the disease being diagnosed as early as possible. However, due to issues regarding the use of prostate-specific antigen (PSA) for screening purposes, PCa management is among the most contentious of healthcare matters. PSA screening is problematic primarily because of diagnosis difficulties and the high rate of false-positive biopsies. Novel PCa biomarkers, such as the Prostate Health Index (PHI) and the 4Kscore, have been proposed in recent times to improve PSA prediction accuracy and have shown higher performance by preventing redundant biopsies. The 4Kscore also shows high precision in determining the risk of developing high-grade PCa, whereas elevated PHI levels suggest that the tumor is aggressive. Some evidence also supports the effectiveness of miRNAs as biomarkers for distinguishing PCa from benign prostatic hyperplasia and for assessing the aggressiveness of the disease. A number of miRNAs that possibly act as tumor inhibitors or oncogenes are impaired in PCa. These new biomarkers are comprehensively reviewed in the present study in terms of their potential use in diagnosing and treating PCa.
Collapse
Affiliation(s)
- Wei Jin
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xiang Fei
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xia Wang
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yan Song
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Fangjie Chen
- Department of Medical Genetics, School of Life Sciences, China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
33
|
Wei Y, Lu S, Hu Y, Guo L, Wu X, Liu X, Sun Y. MicroRNA-135a Regulates VEGFC Expression and Promotes Luteinized Granulosa Cell Apoptosis in Polycystic Ovary Syndrome. Reprod Sci 2020; 27:1436-1442. [PMID: 32016798 DOI: 10.1007/s43032-020-00155-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 12/05/2019] [Indexed: 12/20/2022]
Abstract
Androgen is known to regulate microRNA-135a (miR-135a) and can be regulated by androgen, suggesting that it may contribute to polycystic ovary syndrome (PCOS) with hyperandrogenism. However, its roles and mechanisms of action in PCOS are unknown. In this study, the role and molecular mechanisms underlying miR-135a in granulosa cells (GCs) in PCOS were evaluated. miR-135a expression was upregulated in patients with PCOS and in GCs isolated from patients compared with that in the respective controls (P < 0.01), as determined by RT-qPCR. The overexpression of miR-135a inhibited GC proliferation and induced GC apoptosis, as observed by CCK-8 assay and apoptosis assay. Furthermore, miR-135a overexpression increased the expression of double-strand break maker, γH2AX, as confirmed by western blotting. Our results further suggest that these effects were mediated via downregulation of vascular endothelial growth factor C (VEGFC), which was identified as a direct target of miR-135a. Moreover, levels of VEGFC and miR-135a expression showed a negative correlation. These findings indicate that miR-135a promotes apoptosis and the DNA damage response in GCs in PCOS, likely via VEGFC signaling. This study provides novel insights into GC dysregulation in PCOS and suggests that miR-135a is a promising therapeutic target for PCOS treatment.
Collapse
Affiliation(s)
- Yifang Wei
- Center for Reproductive Medicine, No. 455 Hospital of the Chinese People's Liberation Army, Second Military Medical University, Shanghai, 200052, China. .,Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200135, China.
| | - Shenglian Lu
- Department of Obstetrics and Gynecology, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Yu Hu
- Center for Reproductive Medicine, No. 455 Hospital of the Chinese People's Liberation Army, Second Military Medical University, Shanghai, 200052, China
| | - Li Guo
- Center for Reproductive Medicine, No. 455 Hospital of the Chinese People's Liberation Army, Second Military Medical University, Shanghai, 200052, China
| | - Xiaoyu Wu
- Surgical Intensive Care Unit, The Children's Hospital, Zhejiang University School of Medicine, Zhejiang, 310006, Hangzhou, China
| | - Xing Liu
- Center for Reproductive Medicine, No. 455 Hospital of the Chinese People's Liberation Army, Second Military Medical University, Shanghai, 200052, China
| | - Yun Sun
- Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200135, China. .,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200135, China.
| |
Collapse
|
34
|
Shahbazi R, Baradaran B, Khordadmehr M, Safaei S, Baghbanzadeh A, Jigari F, Ezzati H. Targeting ROCK signaling in health, malignant and non-malignant diseases. Immunol Lett 2020; 219:15-26. [PMID: 31904392 DOI: 10.1016/j.imlet.2019.12.012] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 12/15/2019] [Accepted: 12/30/2019] [Indexed: 12/21/2022]
Abstract
A Rho-associated coiled-coil kinase (ROCK) is identified as a critical downstream effector of GTPase RhoA which contains two isoforms, ROCK1 (also known as p160ROCK and ROKβ) and ROCK2 (also known as Rho-kinase and ROKα), the gene of which is placed on chromosomes 18 (18q11.1) and 2 (2p24), respectively. ROCKs have a principal function in the generation of actin-myosin contractility and regulation of actin cytoskeleton dynamics. They represent a chief role in regulating various cellular functions, such as apoptosis, growth, migration, and metabolism through modulation of cytoskeletal actin synthesis, and cellular contraction through phosphorylation of numerous downstream targets. Emerging evidence has indicated that ROCKs present a significant function in cardiac physiology. Of note, dysregulation of ROCKs involves in several cardiac pathological processes like cardiac hypertrophy, cardiac fibrosis, systemic blood pressure disorder, and pulmonary hypertension. Moreover, ROCKs, in addition to their role in regulating renal arteriolar contraction, glomerular blood flow, and filtration, can also play a role in controlling podocytes, tubular cells, and mesangial cell structure and function. Hyperactivity disorder and over-gene expression of Rho/ROCK have been indicated in different cancers. Furthermore, it seems that increasing the expression of mRNA or ROCK protein has an undesirable effect on patient survival and has a positive impact on the progression and worsening of disease prognosis. This review focuses on the physiological and pathological functions of ROCKs with a particular view on its possible value of ROCK inhibitors as a new therapy in cancers and non-cancer diseases.
Collapse
Affiliation(s)
- Roya Shahbazi
- Department of Pathology, Faculty of Veterinary Medicine, University of Tabriz, 51665-1647, Tabriz, Iran.
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, 51666-14761, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, 51666-14761, Tabriz, Iran.
| | - Monireh Khordadmehr
- Department of Pathology, Faculty of Veterinary Medicine, University of Tabriz, 51665-1647, Tabriz, Iran.
| | - Sahar Safaei
- Immunology Research Center, Tabriz University of Medical Sciences, 51666-14761, Tabriz, Iran.
| | - Amir Baghbanzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, 51666-14761, Tabriz, Iran.
| | - Farinaz Jigari
- Department of Pathology, Faculty of Veterinary Medicine, University of Tabriz, 51665-1647, Tabriz, Iran.
| | - Hamed Ezzati
- Department of Pathology, Faculty of Veterinary Medicine, University of Tabriz, 51665-1647, Tabriz, Iran.
| |
Collapse
|
35
|
Cao Z, Qiu J, Yang G, Liu Y, Luo W, You L, Zheng L, Zhang T. MiR-135a biogenesis and regulation in malignancy: a new hope for cancer research and therapy. Cancer Biol Med 2020; 17:569-582. [PMID: 32944391 PMCID: PMC7476096 DOI: 10.20892/j.issn.2095-3941.2020.0033] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 05/27/2020] [Indexed: 02/06/2023] Open
Abstract
MicroRNAs (miRNAs) are evolutionarily conserved small non-coding RNAs that affect posttranscriptional regulation by binding to the 3′-untranslated region of target messenger RNAs. MiR-135a is a critical miRNA that regulates gene expression, and many studies have focused on its function in cancer research. MiR-135a is dysregulated in various cancers and regulates cancer cell proliferation and invasion via several signaling pathways, such as the MAPK and JAK2/STAT3 pathways. MiR-135a has also been found to promote or inhibit the epithelial-mesenchymal transition and chemoresistance in different cancers. Several studies have discovered the value of miR-135a as a novel biomarker for cancer diagnosis and prognosis. These studies have suggested the potential of therapeutically manipulating miR-135a to improve the outcome of cancer patients. Although these findings have demonstrated the role of miR-135a in cancer progression and clinical applications, a number of questions remain to be answered, such as the dual functional roles of miR-135a in cancer. In this review, we summarize the available studies regarding miR-135a and cancer, including background on the biogenesis and expression of miR-135a in cancer and relevant signaling pathways involved in miR-135a-mediated tumor progression. We also focus on the clinical application of miR-135a as a biomarker in diagnosis and as a therapeutic agent or target in cancer treatment, which will provide a greater level of insight into the translational value of miR-135a.
Collapse
Affiliation(s)
- Zhe Cao
- Department of General Surgery
| | | | | | | | | | - Lei You
- Department of General Surgery
| | - Lianfang Zheng
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Taiping Zhang
- Department of General Surgery.,Clinical Immunology Center, Chinese Academy of Medical Sciences, Beijing 100730, China
| |
Collapse
|
36
|
Xu X, Zhu H, Yang M, Zheng E, Zhou Y, Ni J, Li R, Yang Z, He T, Zhao G. Knockdown of TOR signaling pathway regulator suppresses cell migration and invasion in non-small cell lung cancer via the regulation of epithelial-to-mesenchymal transition. Exp Ther Med 2019; 19:1925-1932. [PMID: 32104250 DOI: 10.3892/etm.2019.8358] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 10/22/2019] [Indexed: 12/27/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is one of the most common cancer types worldwide. Previous studies have indicated that TOR signaling pathway regulator (TIPRL) is involved in the progression of NSCLC. However, the underlying mechanisms of the role of TIPRL in regulating NSCLC metastasis have remained largely elusive. In the present study, the expression pattern of TIPRL in NSCLC was analyzed using The Cancer Genome Atlas (TCGA) dataset. Furthermore, Kaplan-Meier curve analysis was performed to evaluate the prognostic value of TIPRL in NSCLC, using the Kaplan-Meier Plotter and TCGA datasets. Loss-of-function assays were performed to determine the effects of TIPRL on cell migration and invasion. The results suggested that TIPRL was upregulated in NSCLC and positively associated with an advanced Tumor-Node-Metastasis stage. A higher expression level of TIPRL was associated with shorter overall and disease-free survival times in patients with NSCLC. To the best of our knowledge, the present study was the first to report that TIPRL acts as a metastasis promoter in NSCLC. Silencing of TIPRL suppressed A549 cell migration and invasion. Mechanistically, the present study indicated that TIPRL knockdown significantly promoted epithelial-cadherin expression, whereas it suppressed twist and vimentin expression in A549 cells. In conclusion, the present analysis suggested that TIPRL may serve as a biomarker for the prognosis of NSCLC and as a future target for its treatment.
Collapse
Affiliation(s)
- Xiang Xu
- Department of Cardiothoracic Surgery, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang 315010, P.R. China
| | - Huangkai Zhu
- Department of Cardiothoracic Surgery, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang 315010, P.R. China.,Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo, Zhejiang 315000, P.R. China
| | - Minglei Yang
- Department of Cardiothoracic Surgery, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang 315010, P.R. China
| | - Enkuo Zheng
- Department of Cardiothoracic Surgery, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang 315010, P.R. China
| | - Yinjie Zhou
- Department of Cardiothoracic Surgery, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang 315010, P.R. China
| | - Junjun Ni
- Department of Cardiothoracic Surgery, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang 315010, P.R. China
| | - Rui Li
- Department of Cardiothoracic Surgery, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang 315010, P.R. China
| | - Zhenhua Yang
- Department of Cardiothoracic Surgery, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang 315010, P.R. China
| | - Ti He
- Shanghai Genechem Translational Medicine Institute, Shanghai 200120, P.R. China
| | - Guofang Zhao
- Department of Cardiothoracic Surgery, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang 315010, P.R. China
| |
Collapse
|
37
|
Androgen-Regulated microRNAs (AndroMiRs) as Novel Players in Adipogenesis. Int J Mol Sci 2019; 20:ijms20225767. [PMID: 31744106 PMCID: PMC6888160 DOI: 10.3390/ijms20225767] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 11/11/2019] [Accepted: 11/12/2019] [Indexed: 12/14/2022] Open
Abstract
The development, homeostasis, or increase of the adipose tissue is driven by the induction of the adipogenic differentiation (adipogenesis) of undifferentiated mesenchymal stem cells (MSCs). Adipogenesis can be inhibited by androgen stimulation of these MSCs resulting in the transcription initiation or repression of androgen receptor (AR) regulated genes. AR not only regulates the transcription of protein-coding genes but also the transcription of several non-coding microRNAs involved in the posttranscriptional gene regulation (herein designated as AndroMiRs). As microRNAs are largely involved in differentiation processes such as adipogenesis, the involvement of AndroMiRs in the androgen-mediated inhibition of adipogenesis is likely, however, not yet intensively studied. In this review, existing knowledge about adipogenesis-related microRNAs and AndroMiRs is summarized, and putative cross-links are drawn, which are still prone to experimental validation.
Collapse
|
38
|
Yuan S, Luan X, Han G, Guo K, Wang S, Zhang X. LINC01638 lncRNA mediates the postoperative distant recurrence of bladder cancer by upregulating ROCK2. Oncol Lett 2019; 18:5392-5398. [PMID: 31620199 PMCID: PMC6788173 DOI: 10.3892/ol.2019.10924] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 08/06/2019] [Indexed: 01/21/2023] Open
Abstract
It is well established that long intergenic non-protein coding RNA 1638 (LINC01638) promotes the development and progression of breast cancer, whereas its roles in other human diseases are currently unknown. In the present study, expression of LINC01638 and ROCK2 was analyzed using quantitative PCR, ELISA and western blot. Receiver operating characteristic curve was used for diagnostic analysis. Cell transfections were performed to analyze interactions between LINC01638 and ROCK2, while Transwell assays were performed to analyze invasion and migration of the bladder cancer HT-1197 and HT-1376 cell lines. It was observed that LINC01638 and Rho-associated, coiled-coil containing protein kinase 2 (ROCK2) were significantly upregulated in the plasma of patients with early stage (stage I and II) bladder cancer compared with in healthy controls. Upregulation of LINC01638 and ROCK2 distinguished patients with early stage bladder cancer from healthy controls. Plasma levels of LINC01638 and ROCK2 were positively correlated in patients with bladder cancer, but not in healthy controls. A follow-up study after surgical resection revealed that LINC01638 and ROCK2 were further upregulated in patients with distant recurrence, or distant and local recurrence, but not in patients with local recurrence and no recurrence. Overexpression of LINC01638 led to ROCK2 upregulation in bladder cancer cells, whereas ROCK2 overexpression did not significantly affect LINC01638 expression. Overexpression of LINC01638 and ROCK2 mediated the promoted migration and invasion of bladder cancer cells, and ROCK2 small interfering RNA silencing attenuated the enhancing effects of LINC01638 on cancer cell migration and invasion. Therefore, LINC01638 may mediate the postoperative distant recurrence of bladder cancer by upregulating ROCK2.
Collapse
Affiliation(s)
- Shouxian Yuan
- Department of Urology Surgery, The Second People's Hospital of Liaocheng, Linqing, Shandong 252600, P.R. China
| | - Xiuhua Luan
- Department of Urology Surgery, The Second People's Hospital of Liaocheng, Linqing, Shandong 252600, P.R. China
| | - Guiqiang Han
- Department of Urology Surgery, The People's Hospital of Liqing, Linqing, Shandong 252600, P.R. China
| | - Kecun Guo
- Department of Urology Surgery, The Second People's Hospital of Liaocheng, Linqing, Shandong 252600, P.R. China
| | - Shenghui Wang
- Department of Urology Surgery, The Second People's Hospital of Liaocheng, Linqing, Shandong 252600, P.R. China
| | - Xiangkai Zhang
- Department of Urology Surgery, Shanxi Provincial People's Hospital, Taiyuan, Shanxi 030012, P.R. China
| |
Collapse
|
39
|
Up regulation of Rho-associated coiled-coil containing kinase1 (ROCK1) is associated with genetic instability and poor prognosis in prostate cancer. Aging (Albany NY) 2019; 11:7859-7879. [PMID: 31557128 PMCID: PMC6781985 DOI: 10.18632/aging.102294] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 09/14/2019] [Indexed: 12/26/2022]
Abstract
BACKGROUND AND OBJECTIVES Overexpression of the cytoskeleton-modulating kinase ROCK1 has been associated with unfavorable outcome in many cancers, but its impact in prostate cancer is largely unknown. RESULTS A weak ROCK1 staining was found in >90% of normal, and cancerous prostate tissues, but was generally stronger in cancer cells as compared to adjacent normal glands. In cancer, ROCK1 staining was considered weak, moderate, and strong in 22%, 53%, and 18% of cases respectively. Higher ROCK1 expression levels were associated with tumor stage, and Gleason grade, positive nodal stage, positive surgical margin, accelerated cell proliferation and early PSA recurrence in multivariable analysis. ROCK1 up regulation was associated with androgen receptor (AR) expression, TMPRSS2:ERG fusion, genomic deletions of the PTEN tumor suppressor, as well as recurrent deletions at chromosomes 3p, 5q, 6q. Strong ROCK1 staining was found in 3% of AR-negative, but in 27% of strongly AR positive cancers, in 13% of ERG-negative but in 25% of ERG positive cancers, and in 12% of PTEN normal but in 26% of PTEN deleted cancers. CONCLUSIONS This study identifies ROCK1 expression associated with prognosis in prostate cancer. METHODS We tested ROCK1 expression in 12 427 prostate cancer specimens and followed PSA recurrence after prostatectomy.
Collapse
|
40
|
Ai N, Li B, Li L, Li Z, Ji H, Yang G, Yin F. MicroRNA-466 inhibits cancer cell migration and invasion in hepatocellular carcinoma by indirectly mediating the downregulation of ROCK2. Exp Ther Med 2019; 18:1493-1499. [PMID: 31363382 PMCID: PMC6614712 DOI: 10.3892/etm.2019.7709] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 04/18/2019] [Indexed: 01/05/2023] Open
Abstract
MicroRNA-466 was recently characterized as a tumor suppressor with known biological function in prostate cancer. The aim of the current study was to investigate the possible involvement of microRNA-466 in hepatocellular carcinoma (HCC). The current study demonstrated that the expression level of microRNA-466 was significantly downregulated; while the mRNA expression level of Rho-associated coiled-coil containing protein kinase 2 (ROCK2) was significantly upregulated in tumor tissue compared with adjacent healthy tissue samples obtained from patients with HCC. In addition, the relative plasma level of microRNA-466 was significantly decreased, while the relative plasma level of ROCK2 was significantly increased in patients with HCC compared with healthy controls. Expression levels of microRNA-466 and ROCK2 were inversely correlated in tumor tissue but not in adjacent healthy tissue samples obtained from patients with HCC. Plasma levels of microRNA-466 and ROCK2 were inversely correlated in patients with HCC but not in healthy controls. In addition, reduced plasma levels of microRNA-466 may have a diagnostic value in the detection of early stage HCC. MicroRNA-466 overexpression significantly suppressed ROCK2 expression in HCC cells, whereas ROCK2 overexpression did not significantly affect microRNA-466 expression. MicroRNA-466 overexpression significantly suppressed, while ROCK2 overexpression significantly enhanced HCC cell migration and invasion. In addition, ROCK2 overexpression partially reversed the inhibitory effect of microRNA-466 overexpression on HCC cell migration and invasion. Taken together, these results suggest that microRNA-466 may inhibit HCC cell migration and invasion by indirectly mediating the downregulation of ROCK2.
Collapse
Affiliation(s)
- Ning Ai
- Department of Interventional Radiology, The Fourth Hospital of Hebei Medical University, Shijiangzhuang, Hebei 050011, P.R. China
| | - Bo Li
- Department of Interventional Radiology, The Fourth Hospital of Hebei Medical University, Shijiangzhuang, Hebei 050011, P.R. China
| | - Li Li
- Department of Interventional Radiology, The Fourth Hospital of Hebei Medical University, Shijiangzhuang, Hebei 050011, P.R. China
| | - Zhigang Li
- Department of Interventional Radiology, The Fourth Hospital of Hebei Medical University, Shijiangzhuang, Hebei 050011, P.R. China
| | - Hong Ji
- Department of General Surgery, The Second Hospital of Hebei Medical University, Shijiangzhuang, Hebei 050011, P.R. China
| | - Guang Yang
- Department of Interventional Radiology, The Fourth Hospital of Hebei Medical University, Shijiangzhuang, Hebei 050011, P.R. China
| | - Fenghua Yin
- Department of Interventional Radiology, The Fourth Hospital of Hebei Medical University, Shijiangzhuang, Hebei 050011, P.R. China
| |
Collapse
|
41
|
Fernandes RC, Hickey TE, Tilley WD, Selth LA. Interplay between the androgen receptor signaling axis and microRNAs in prostate cancer. Endocr Relat Cancer 2019; 26:R237-R257. [PMID: 30817318 DOI: 10.1530/erc-18-0571] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 02/26/2019] [Indexed: 12/29/2022]
Abstract
The androgen receptor (AR) is a ligand-activated transcription factor that drives prostate cancer. Since therapies that target the AR are the mainstay treatment for men with metastatic disease, it is essential to understand the molecular mechanisms underlying oncogenic AR signaling in the prostate. miRNAs are small, non-coding regulators of gene expression that play a key role in prostate cancer and are increasingly recognized as targets or modulators of the AR signaling axis. In this review, we examine the regulation of AR signaling by miRNAs and vice versa and discuss how this interplay influences prostate cancer growth, metastasis and resistance to therapy. Finally, we explore the potential clinical applications of miRNAs implicated in the regulation of AR signaling in this prevalent hormone-driven disease.
Collapse
Affiliation(s)
- Rayzel C Fernandes
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia
- Freemasons Foundation Centre for Men's Health, Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| | - Theresa E Hickey
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| | - Wayne D Tilley
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia
- Freemasons Foundation Centre for Men's Health, Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| | - Luke A Selth
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia
- Freemasons Foundation Centre for Men's Health, Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
42
|
Liu H, Hou T, Ju W, Xing Y, Zhang X, Yang J. MicroRNA‑122 downregulates Rho‑associated protein kinase 2 expression and inhibits the proliferation of prostate carcinoma cells. Mol Med Rep 2019; 19:3882-3888. [PMID: 30816534 DOI: 10.3892/mmr.2019.9995] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Accepted: 01/17/2019] [Indexed: 11/05/2022] Open
Abstract
MicroRNA‑122 (miR‑122) has been reported to be involved in the pathogenesis of several types of malignancies; however, its role in prostate carcinoma remains unknown. Thus, the current study aimed to investigate the functionality of miR‑122 in prostate carcinoma. Clinical data of 54 patients with prostate carcinoma who were diagnosed and treated in Union Hospital (Wuhan, China) between January 2011 and January 2013 were retrospectively analyzed. The expression levels of miR‑122 and Rho‑associated protein kinase 2 (ROCK2) in prostate tumor and adjacent healthy tissues of patients, as well as in the serum of prostate carcinoma patients and healthy controls, were detected by reverse transcription‑quantitative polymerase chain reaction. Receiver operating characteristic curve and survival curve analyses were used to examine the diagnostic and prognostic values of serum miR‑122 for prostate carcinoma. In addition, miR‑122 mimic was transfected into prostate carcinoma cells, and the effects on cell proliferation and ROCK2 expression were explored by Cell Counting Kit‑8 and western blot assays, respectively. It was observed that miR‑122 was downregulated and ROCK2 was upregulated in tumor tissues as compared with their levels in adjacent healthy tissues. miR‑122 level in the serum was also markedly lower in prostate carcinoma patients in comparison with that in healthy controls. Furthermore, a low serum level of miR‑122 was found to effectively distinguish the prostate carcinoma patients from healthy controls and to be an indicator of poor survival. In prostate carcinoma cells, miR‑122 overexpression inhibited the proliferation and the expression of ROCK2. Taken together, miR‑122 may inhibit the proliferation of prostate carcinoma cells possibly by downregulating ROCK2 expression.
Collapse
Affiliation(s)
- Hong Liu
- Department of Critical Care Medicine, Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Teng Hou
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Wen Ju
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Yifei Xing
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Xiaoping Zhang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Jun Yang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| |
Collapse
|
43
|
Dong Y, Wang J, Du KX, Jia TM, Zhu CL, Zhang Y, Xu FL. MicroRNA-135a participates in the development of astrocytes derived from bacterial meningitis by downregulating HIF-1α. Am J Physiol Cell Physiol 2019; 316:C711-C721. [PMID: 30726113 DOI: 10.1152/ajpcell.00440.2018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Accumulating evidence has highlighted the potential of microRNAs (miRs) as biomarkers in various human diseases. However, the roles of miRs in bacterial meningitis (BM), a severe infectious condition, still remain unclear. Thus, the present study aimed to investigate the effects of miR-135a on proliferation and apoptosis of astrocytes in BM. Neonatal rats were injected with Streptococcus pneumoniae to establish the BM model. The expression of miR-135a and hypoxia-inducible factor 1α (HIF-1α) in the BM rat models were characterized, followed by determination of their interaction. Using gain- and loss-of-function approaches, the effects of miR-135a on proliferation, apoptosis, and expression of glial fibrillary acidic protein (GFAP), in addition to apoptosis-related factors in astrocytes were examined accordingly. The regulatory effect of HIF-1α was also determined along with the overexpression or knockdown of HIF-1α. The results obtained indicated that miR-135a was poorly expressed, whereas HIF-1α was highly expressed in the BM rat models. In addition, restored expression levels of miR-135a were determined to promote proliferation while inhibiting the apoptosis of astrocytes, along with downregulated Bax and Bad, as well as upregulated Bcl-2, Bcl-XL, and GFAP. As a target gene of miR-135a, HIF-1α expression was determined to be diminished by miR-135a. The upregulation of HIF-1α reversed the miR-135a-induced proliferation of astrocytes. Taken together, the key findings of the current study present evidence suggesting that miR-135a can downregulate HIF-1α and play a contributory role in the development of astrocytes derived from BM, providing a novel theoretical perspective for BM treatment approaches.
Collapse
Affiliation(s)
- Yan Dong
- Department of Pediatrics, Third Affiliated Hospital of Zhengzhou University , Zhengzhou , China.,Henan Provincial Key Laboratory of Child Brain Injury , Zhengzhou , China
| | - Jun Wang
- Department of Children Rehabilitation, Third Affiliated Hospital of Zhengzhou University , Zhengzhou , China
| | - Kai-Xian Du
- Department of Pediatrics, Third Affiliated Hospital of Zhengzhou University , Zhengzhou , China
| | - Tian-Ming Jia
- Department of Pediatrics, Third Affiliated Hospital of Zhengzhou University , Zhengzhou , China
| | - Chang-Lian Zhu
- Department of Pediatrics, Third Affiliated Hospital of Zhengzhou University , Zhengzhou , China.,Henan Provincial Key Laboratory of Child Brain Injury , Zhengzhou , China.,Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg , Gothenburg , Sweden
| | - Yan Zhang
- Clinical Laboratory, Henan Red Cross Blood Center , Zhengzhou , China
| | - Fa-Lin Xu
- Department of Pediatrics, Third Affiliated Hospital of Zhengzhou University , Zhengzhou , China
| |
Collapse
|
44
|
Ast V, Kordaß T, Oswald M, Kolte A, Eisel D, Osen W, Eichmüller SB, Berndt A, König R. MiR-192, miR-200c and miR-17 are fibroblast-mediated inhibitors of colorectal cancer invasion. Oncotarget 2018; 9:35559-35580. [PMID: 30473751 PMCID: PMC6238973 DOI: 10.18632/oncotarget.26263] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 10/06/2018] [Indexed: 12/11/2022] Open
Abstract
Colorectal cancer remains a leading cause of cancer-related death worldwide. A previous transcriptomics based study characterized molecular subgroups of which the stromal subgroup was associated with the worst clinical outcome. Micro-RNAs (miRNAs) are well-known regulators of gene expression and can follow a non-linear repression mechanism. We set up a model combining piecewise linear and linear regression and applied this combined regression model to a comprehensive colon adenocarcinoma dataset. We identified miRNAs involved in regulating characteristic gene sets, particularly extracellular matrix remodeling in the stromal subgroup. Comparison of expression data from separated (epithelial) cancer cells and stroma cells or fibroblasts associate these regulatory interactions with infiltrating stromal or tumor-associated fibroblasts. MiR-200c, miR-17 and miR-192 were identified as the most promising candidates regulating genes crucial for extracellular matrix remodeling. We validated our computational findings by in vitro assays. Enforced expression of either miR-200c, miR-17 or miR-192 in untransformed human colon fibroblasts down-regulated 85% of all predicted target genes. Expressing these miRNAs singly or in combination in human colon fibroblasts co-cultured with colon cancer cells considerably reduced cancer cell invasion validating these miRNAs as cancer cell infiltration suppressors in tumor associated fibroblasts.
Collapse
Affiliation(s)
- Volker Ast
- Integrated Research and Treatment Center, Center for Sepsis Control and Care, Jena University Hospital, 07747 Jena, Germany
- Network Modeling, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute Jena, 07745 Jena, Germany
| | - Theresa Kordaß
- GMP & T Cell Therapy Unit, German Cancer Research Center, 69120 Heidelberg, Germany
| | - Marcus Oswald
- Integrated Research and Treatment Center, Center for Sepsis Control and Care, Jena University Hospital, 07747 Jena, Germany
- Network Modeling, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute Jena, 07745 Jena, Germany
| | - Amol Kolte
- Integrated Research and Treatment Center, Center for Sepsis Control and Care, Jena University Hospital, 07747 Jena, Germany
- Network Modeling, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute Jena, 07745 Jena, Germany
| | - David Eisel
- GMP & T Cell Therapy Unit, German Cancer Research Center, 69120 Heidelberg, Germany
| | - Wolfram Osen
- GMP & T Cell Therapy Unit, German Cancer Research Center, 69120 Heidelberg, Germany
| | - Stefan B. Eichmüller
- GMP & T Cell Therapy Unit, German Cancer Research Center, 69120 Heidelberg, Germany
| | - Alexander Berndt
- Institute of Forensic Medicine, Section Pathology, Jena University Hospital, 07747 Jena, Germany
| | - Rainer König
- Integrated Research and Treatment Center, Center for Sepsis Control and Care, Jena University Hospital, 07747 Jena, Germany
- Network Modeling, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute Jena, 07745 Jena, Germany
| |
Collapse
|
45
|
Goradel NH, Mohammadi N, Haghi-Aminjan H, Farhood B, Negahdari B, Sahebkar A. Regulation of tumor angiogenesis by microRNAs: State of the art. J Cell Physiol 2018; 234:1099-1110. [PMID: 30070704 DOI: 10.1002/jcp.27051] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 06/25/2018] [Indexed: 12/21/2022]
Abstract
MicroRNAs (miRNAs, miRs) are small (21-25 nucleotides) endogenous and noncoding RNAs involved in many cellular processes such as apoptosis, development, proliferation, and differentiation via binding to the 3'-untranslated region of the target mRNA and inhibiting its translation. Angiogenesis is a hallmark of cancer, which provides oxygen and nutrition for tumor growth while removing deposits and wastes from the tumor microenvironment. There are many angiogenesis stimulators, among which vascular endothelial growth factor (VEGF) is the most well known. VEGF has three tyrosine kinase receptors, which, following VEGF binding, initiate proliferation, invasion, migration, and angiogenesis of endothelial cells in the tumor environment. One of the tumor microenvironment conditions that induce angiogenesis through increasing VEGF and its receptors expression is hypoxia. Several miRNAs have been identified that affect different targets in the tumor angiogenesis pathway. Most of these miRNAs affect VEGF and its tyrosine kinase receptors expression downstream of the hypoxia-inducible Factor 1 (HIF-1). This review focuses on tumor angiogenesis regulation by miRNAs and the mechanism underlying this regulation.
Collapse
Affiliation(s)
- Nasser H Goradel
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nejad Mohammadi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamed Haghi-Aminjan
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Farhood
- Departments of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Babak Negahdari
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Amirhossein Sahebkar
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
46
|
Zheng Y, Xiang L, Chen M, Xiang C. MicroRNA‑130a inhibits the proliferation, migration and invasive ability of hepatocellular carcinoma cells by downregulating Rho‑kinase 2. Mol Med Rep 2018; 18:3077-3084. [PMID: 30015839 DOI: 10.3892/mmr.2018.9283] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 08/03/2017] [Indexed: 11/06/2022] Open
Abstract
MicroRNA‑130a (miR‑130a) has been reported to be downregulated in hepatocellular carcinoma (HCC). However, the roles and underlying tumor‑suppressive mechanisms of miR‑130a in the pathogenesis of HCC remain unclear. In the current study, reduced expression of miR‑130a was observed in tumor tissues of patients with HCC in addition to in four HCC cell lines, BEL‑7402, MHCC97H, HepG2 and Huh7. Results of methyl thiazolyl tetrazolium (MTT) assays identified decreased growth rates of MHCC97H and HepG2 cells transfected with miR‑130a mimics. The in vitro colony formation assays demonstrated that the number of colonies formed by cells transfected with miR‑130a mimics and cells transfected with miR‑130a inhibitors was lower and higher, respectively, than that formed by the cells transfected with miR‑negative control. In addition, it was identified that overexpression of miR‑130a reduced the migration and invasiveness of MHCC97H and HepG2 cells. Luciferase reporter assays demonstrated that miR‑130a directly targeted the 3'‑untranslated region of Rho‑kinase 2 (ROCK2) mRNA. Northern and western blot analyses indicated that miR‑130a could modulate the mRNA and protein expression of ROCK2. Additionally, small‑interfering RNA‑mediated knockdown of ROCK2 decreased the proliferation, migration and invasiveness of MHCC97H and HepG2 cells. Overall, these observation suggest that miR‑130a is a regulator of ROCK2 and can inhibit proliferation, migration and invasive ability of HCC cells, at least in part, by suppressing the expression of ROCK2. The current study provides further insight into the molecular mechanisms of HCC pathogenesis and suggests a new potential biotarget for HCC treatment.
Collapse
Affiliation(s)
- Yansong Zheng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - Liangguang Xiang
- Department of General Surgery, Fuqing City Hospital, Fuqing, Fujian 350300, P.R. China
| | - Mingliu Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - Chunhui Xiang
- Department of Neurosurgery, The Central Hospital of Enshi Autonomy State, Enshi, Hubei 445000, P.R. China
| |
Collapse
|
47
|
Khatun A, Shimozawa M, Kito H, Kawaguchi M, Fujimoto M, Ri M, Kajikuri J, Niwa S, Fujii M, Ohya S. Transcriptional Repression and Protein Degradation of the Ca 2+-Activated K + Channel K Ca1.1 by Androgen Receptor Inhibition in Human Breast Cancer Cells. Front Physiol 2018; 9:312. [PMID: 29713287 PMCID: PMC5911984 DOI: 10.3389/fphys.2018.00312] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 03/14/2018] [Indexed: 01/14/2023] Open
Abstract
The large-conductance Ca2+-activated K+ channel KCa1.1 plays an important role in the promotion of breast cancer cell proliferation and metastasis. The androgen receptor (AR) is proposed as a therapeutic target for AR-positive advanced triple-negative breast cancer. We herein investigated the effects of a treatment with antiandrogens on the functional activity, activation kinetics, transcriptional expression, and protein degradation of KCa1.1 in human breast cancer MDA-MB-453 cells using real-time PCR, Western blotting, voltage-sensitive dye imaging, and whole-cell patch clamp recording. A treatment with the antiandrogen bicalutamide or enzalutamide for 48 h significantly suppressed (1) depolarization responses induced by paxilline (PAX), a specific KCa1.1 blocker and (2) PAX-sensitive outward currents induced by the depolarizing voltage step. The expression levels of KCa1.1 transcripts and proteins were significantly decreased in MDA-MB-453 cells, and the protein degradation of KCa1.1 mainly contributed to reductions in KCa1.1 activity. Among the eight regulatory β and γ subunits, LRRC26 alone was expressed at high levels in MDA-MB-453 cells and primary and metastatic breast cancer tissues, whereas no significant changes were observed in the expression levels of LRRC26 and activation kinetics of PAX-sensitive outward currents in MDA-MB-453 cells by the treatment with antiandrogens. The treatment with antiandrogens up-regulated the expression of the ubiquitin E3 ligases, FBW7, MDM2, and MDM4 in MDA-MB-453 cells, and the protein degradation of KCa1.1 was significantly inhibited by the respective siRNA-mediated blockade of FBW7 and MDM2. Based on these results, we concluded that KCa1.1 is an androgen-responsive gene in AR-positive breast cancer cells, and its down-regulation through enhancements in its protein degradation by FBW7 and/or MDM2 may contribute, at least in part, to the antiproliferative and antimetastatic effects of antiandrogens in breast cancer cells.
Collapse
Affiliation(s)
- Anowara Khatun
- Division of Pathological Sciences, Department of Pharmacology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Motoki Shimozawa
- Division of Pathological Sciences, Department of Pharmacology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Hiroaki Kito
- Division of Pathological Sciences, Department of Pharmacology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Mayu Kawaguchi
- Division of Pathological Sciences, Department of Pharmacology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Mayu Fujimoto
- Division of Pathological Sciences, Department of Pharmacology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Moe Ri
- Department of Pharmacology, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan
| | - Junko Kajikuri
- Department of Pharmacology, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan
| | - Satomi Niwa
- Division of Pathological Sciences, Department of Pharmacology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Masanori Fujii
- Division of Pathological Sciences, Department of Pharmacology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Susumu Ohya
- Division of Pathological Sciences, Department of Pharmacology, Kyoto Pharmaceutical University, Kyoto, Japan.,Department of Pharmacology, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan
| |
Collapse
|
48
|
Awad D, Pulliam TL, Lin C, Wilkenfeld SR, Frigo DE. Delineation of the androgen-regulated signaling pathways in prostate cancer facilitates the development of novel therapeutic approaches. Curr Opin Pharmacol 2018; 41:1-11. [PMID: 29609138 DOI: 10.1016/j.coph.2018.03.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 03/08/2018] [Indexed: 02/08/2023]
Abstract
Although androgen deprivation therapy (ADT) is initially effective for the treatment of progressive prostate cancer, it inevitably fails due to the onset of diverse resistance mechanisms that restore androgen receptor (AR) signaling. Thus, AR remains a desired therapeutic target even in the relapsed stages of the disease. Given the difficulties in stopping all AR reactivation mechanisms, we propose that the identification of the driver signaling events downstream of the receptor offer viable, alternative therapeutic targets. Here, we summarize recently described, AR-regulated processes that have been demonstrated to promote prostate cancer. By highlighting these signaling events and describing some of the ongoing efforts to pharmacologically modulate these pathways, our goal is to advocate potential new therapeutic targets that would represent an alternative approach for blocking AR actions.
Collapse
Affiliation(s)
- Dominik Awad
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Thomas L Pulliam
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX, USA; Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Chenchu Lin
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Sandi R Wilkenfeld
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Daniel E Frigo
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX, USA; Department of Biology and Biochemistry, University of Houston, Houston, TX, USA; Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Molecular Medicine Program, The Houston Methodist Research Institute, Houston, TX, USA.
| |
Collapse
|
49
|
A multiplatform approach identifies miR-152-3p as a common epigenetically regulated onco-suppressor in prostate cancer targeting TMEM97. Clin Epigenetics 2018; 10:40. [PMID: 29599847 PMCID: PMC5870254 DOI: 10.1186/s13148-018-0475-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 03/19/2018] [Indexed: 12/16/2022] Open
Abstract
Background Prostate cancer (PCa) is a major cause of morbidity and mortality in men worldwide. MicroRNAs are globally downregulated in PCa, especially in poorly differentiated tumors. Nonetheless, the underlying mechanisms are still elusive. Herein, using combined analysis of microRNAs expression and genomewide DNA methylation, we aimed to identify epigenetically downregulated microRNAs in PCa. Results We found that miR-152-3p was underexpressed in PCa and that lower expression levels were associated with promoter hypermethylation in accordance with TCGA dataset analysis. Functional in vitro assays suggest that miR-152-3p suppresses cell viability and invasion potential, whereas it promotes cell cycle arrest at S and G2/M phases. Additionally, miR-152-3p expression was associated with longer disease-free survival in PCa patients from TCGA. Finally, TMEM97, which is overexpressed in PCa, was identified as a novel miR-152-3p target gene. Conclusions Our findings demonstrate the advantages of using a combinatory approach to identify microRNAs downregulated due to aberrant promoter methylation. MiR-152-3p downregulation and promoter methylation was found to be prevalent in primary PCa, which impairs its role in control of cell viability, cell cycle regulation and invasion. Electronic supplementary material The online version of this article (10.1186/s13148-018-0475-2) contains supplementary material, which is available to authorized users.
Collapse
|
50
|
miR-135a inhibits tumor metastasis and angiogenesis by targeting FAK pathway. Oncotarget 2018; 8:31153-31168. [PMID: 28415713 PMCID: PMC5458197 DOI: 10.18632/oncotarget.16098] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 03/01/2017] [Indexed: 01/28/2023] Open
Abstract
Tumor metastasis has been the major cause of recurrence and death in patients with gastric cancer. Here, we find miR-135a has a decreased expression in the metastatic cell lines compared with its parental cell lines by analyzing microRNA array. Further results show that miR-135a is downregulated in the majority of human gastric cancer tissues and cell lines. Decreased expression of miR-135a is associated with TNM stage and poor survival. Besides, regaining miR-135a in gastric cancer cells obviously inhibits tumor growth, migration, invasion and angiogenesis by targeting focal adhesion kinase (FAK) pathway. Bioinformatics analysis and molecular experiments further prove that miR-135a is a novel downstream gene of tumor suppressor p53. Blocking FAK with its inhibitor can also enhance miR-135a expression through inducing p53. In summary, this study reveals the expression and function of miR-135a in gastric cancer and uncovers a novel regulatory mechanism of miR-135a.
Collapse
|