1
|
Zehtabi M, Ghaedrahmati F, Dari MAG, Moramezi F, Kempisty B, Mozdziak P, Farzaneh M. Emerging biologic and clinical implications of miR-182-5p in gynecologic cancers. Clin Transl Oncol 2025; 27:2367-2382. [PMID: 39661239 DOI: 10.1007/s12094-024-03822-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 11/28/2024] [Indexed: 12/12/2024]
Abstract
MicroRNAs (miRNAs) have emerged as important regulators of gene expression in various biological processes, including cancer. miR-182-5p has gained attention for its potential implications in gynecologic cancers, including breast, ovarian, endometrial, and cervical cancers. miR-182-5p dysregulation has been associated with multiple facets of tumor biology in gynecologic cancers, including tumor initiation, progression, metastasis, and therapeutic response. Studies have highlighted its involvement in key signaling pathways and cellular processes that contribute to cancer development and progression. In addition, miR-182-5p has shown potential as a diagnostic and prognostic biomarker, with studies demonstrating its correlation with clinicopathological features and patient outcomes. Furthermore, the therapeutic potential of miR-182-5p is being explored in gynecologic cancers. Strategies such as miRNA mimics or inhibitors targeting miR-182-5p have shown promise in preclinical and early clinical studies. These approaches aim to modulate miR-182-5p expression, restoring normal cellular functions and potentially enhancing treatment responses. Understanding the biologic and clinical implications of miR-182-5p in gynecologic cancers is crucial for the development of targeted therapeutic strategies and personalized medicine approaches. Further investigations are needed to unravel the specific target genes and pathways regulated by miR-182-5p. It is important to consider the emerging biologic and clinical implications of miR-182-5p in gynecologic cancers.
Collapse
Affiliation(s)
- Mojtaba Zehtabi
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farhoodeh Ghaedrahmati
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mahrokh Abouali Gale Dari
- Department of Obstetrics and Gynecology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Farideh Moramezi
- Fertility, Infertility and Perinatology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Bartosz Kempisty
- Department of Human Morphology and Embryology, Division of Anatomy, Faculty of Medicine, Wrocław Medical University, Wrocław, Poland
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University, Torun, Poland
- Physiology Graduate Faculty North, Carolina State University, Raleigh, NC, 27695, USA
- Center of Assisted Reproduction Department of Obstetrics and Gynecology, University Hospital and Masaryk University, Brno, Czech Republic
| | - Paul Mozdziak
- Physiology Graduate Faculty North, Carolina State University, Raleigh, NC, 27695, USA
| | - Maryam Farzaneh
- Fertility, Infertility and Perinatology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
- Clinical Research Development Unit, Imam Khomeini Hospital, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
2
|
Ishaq Y, Rauff B, Alzahrani B, Ikram A, Javed H, Abdullah I, Mujtaba G. Bioinformatics and Experimental Insights Into miR-182, hsa_circ_0070269, and circ-102,166 as Therapeutic Targets for HCV-Associated HCC. Cancer Rep (Hoboken) 2024; 7:e70049. [PMID: 39617640 PMCID: PMC11608829 DOI: 10.1002/cnr2.70049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 09/28/2024] [Accepted: 10/03/2024] [Indexed: 01/05/2025] Open
Abstract
AIMS Hepatocellular carcinoma (HCC) is a type of malignant tumor and the sixth leading cause of death worldwide. It is caused by HBV, HCV infection, and alcohol consumption. MicroRNAs are typically small, non-coding RNAs that are involved in the regulation of mRNA expression. Recent studies revealed miRNAs' regulatory roles in liver cancer, linked to risk factors like HCV, HBV infection, alcoholism, drug use, and auto-immune hepatic disorders. Circular RNAs also belong to the class of non-coding RNAs; they act as ceRNAs to regulate miRNA expression and regulate different oncogenic pathways in HCC progression. This study aimed to check the hsa_circ_0070269, circ-102,166 (hsa_circ_0004913), and miR-182 expression in HCV induced HCC patients. METHODS Data analysis was used to find out studies related to the role of hsa_circ_0070269, circ-102,166, and miR-182 in HCC; miR-182 targeted genes, their role in different diseases; and miR-182 interactions with hsa_circ_0070269 and circ-102,166 in the HCC. It was revealed that the hsa_circ_0070269, circ-102,166, and miR-182 correlations in HCV induced HCC have not been explored yet. Therefore, to validate data from literature mining, expression analysis of dysregulated hsa_circ_0070269, circ-102,166, and miR-182 was performed in HCV induced HCC patients using RT-PCR. RESULTS It was found that miR-182 was significantly upregulated and acts as an oncomiRNA in HCV induced HCC, and hsa_circ_0070269 and circ-102,166 were downregulated in HCV induced HCC. We have identified that miR-182 relative expression level was significantly high (p < 0.0029), while has_circ_0070269 (p < 0.002) and circ-102,166 (p < 0.002) were significantly downregulated in HCV-HCC patients as compared to expression in healthy individuals. CONCLUSION Our data revealed that miR-182 acts as an oncomiRNA in HCC development. Hsa_circ_0070269 and circ-102,166 are highly expressed in healthy controls compared to HCV induced HCC patients, can sponge miR-182 expression by acting as tumor suppressors, and can be used as biomarkers and targets for HCC treatment.
Collapse
Affiliation(s)
- Yasmeen Ishaq
- Institute of Molecular Biology and Biotechnology (IMBB)University of Lahore (UOL)LahorePakistan
| | - Bisma Rauff
- Department of Biomedical EngineeringUET LahoreNarowalPakistan
| | - Badr Alzahrani
- Department of Clinical Laboratory Sciences, College of Applied Medical SciencesJouf UniversitySakakaSaudi Arabia
| | - Aqsa Ikram
- Institute of Molecular Biology and Biotechnology (IMBB)University of Lahore (UOL)LahorePakistan
| | - Hasnain Javed
- Provincial Public Health reference lab LahorePunjab AIDS Control ProgramLahorePakistan
| | - Imran Abdullah
- Institute of Nuclear Medicine & Oncology (INMOL) Cancer HospitalLahorePakistan
| | - Ghulam Mujtaba
- Institute of Nuclear Medicine & Oncology (INMOL) Cancer HospitalLahorePakistan
| |
Collapse
|
3
|
Liu X, Zhang J, Wang Z, Yan M, Xu M, Li G, Shender V, Wei J, Li J, Shao C, Zhang S, Kong B, Song K, Liu Z. Splicing Factor PQBP1 Curtails BAX Expression to Promote Ovarian Cancer Progression. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306229. [PMID: 38342602 PMCID: PMC11022708 DOI: 10.1002/advs.202306229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 11/27/2023] [Indexed: 02/13/2024]
Abstract
Splicing factor polyglutamine binding protein-1 (PQBP1) is abundantly expressed in the central nervous system during development, and mutations in the gene cause intellectual disability. However, the roles of PQBP1 in cancer progression remain largely unknown. Here, it is shown that PQBP1 overexpression promotes tumor progression and indicates worse prognosis in ovarian cancer. Integrative analysis of spyCLIP-seq and RNA-seq data reveals that PQBP1 preferentially binds to exon regions and modulates exon skipping. Mechanistically, it is shown that PQBP1 regulates the splicing of genes related to the apoptotic signaling pathway, including BAX. PQBP1 promotes BAX exon 2 skipping to generate a truncated isoform that undergoes degradation by nonsense-mediated mRNA decay, thus making cancer cells resistant to apoptosis. In contrast, PQBP1 depletion or splice-switching antisense oligonucleotides promote exon 2 inclusion and thus increase BAX expression, leading to inhibition of tumor growth. Together, the results demonstrate an oncogenic role of PQBP1 in ovarian cancer and suggest that targeting the aberrant splicing mediated by PQBP1 has therapeutic potential in cancer treatment.
Collapse
Affiliation(s)
- Xihan Liu
- Key Laboratory of Experimental TeratologyMinistry of EducationDepartment of Obstetrics and GynecologyQilu HospitalDepartment of Cell BiologySchool of Basic Medical ScienceShandong UniversityJinan250012China
- Advanced Medical Research InstituteShandong UniversityJinan250012China
| | - Jiaojiao Zhang
- Key Laboratory of Experimental TeratologyMinistry of EducationDepartment of Obstetrics and GynecologyQilu HospitalDepartment of Cell BiologySchool of Basic Medical ScienceShandong UniversityJinan250012China
| | - Zixiang Wang
- Key Laboratory of Experimental TeratologyMinistry of EducationDepartment of Obstetrics and GynecologyQilu HospitalDepartment of Cell BiologySchool of Basic Medical ScienceShandong UniversityJinan250012China
- Advanced Medical Research InstituteShandong UniversityJinan250012China
| | - Mingyao Yan
- Key Laboratory of Experimental TeratologyMinistry of EducationDepartment of Obstetrics and GynecologyQilu HospitalDepartment of Cell BiologySchool of Basic Medical ScienceShandong UniversityJinan250012China
| | - Meining Xu
- Key Laboratory of Experimental TeratologyMinistry of EducationDepartment of Obstetrics and GynecologyQilu HospitalDepartment of Cell BiologySchool of Basic Medical ScienceShandong UniversityJinan250012China
| | - Gaoyuan Li
- Key Laboratory of Experimental TeratologyMinistry of EducationDepartment of Obstetrics and GynecologyQilu HospitalDepartment of Cell BiologySchool of Basic Medical ScienceShandong UniversityJinan250012China
| | - Victoria Shender
- Center for Precision Genome Editing and Genetic Technologies for BiomedicineFederal Research and Clinical Center of Physical‐Chemical Medicine of Federal Medical Biological AgencyMoscow119435Russia
| | - Jian‐jun Wei
- Department of PathologyNorthwestern University School of MedicineChicagoIL60611USA
| | - Jianqiao Li
- Department of OphthalmologyQilu HospitalShandong UniversityJinan250012China
| | - Changshun Shao
- Key Laboratory of Radiation Medicine and ProtectionInstitutes for Translational MedicineSoochow UniversitySuzhou215127China
| | - Shiqian Zhang
- Key Laboratory of Experimental TeratologyMinistry of EducationDepartment of Obstetrics and GynecologyQilu HospitalDepartment of Cell BiologySchool of Basic Medical ScienceShandong UniversityJinan250012China
| | - Beihua Kong
- Key Laboratory of Experimental TeratologyMinistry of EducationDepartment of Obstetrics and GynecologyQilu HospitalDepartment of Cell BiologySchool of Basic Medical ScienceShandong UniversityJinan250012China
| | - Kun Song
- Key Laboratory of Experimental TeratologyMinistry of EducationDepartment of Obstetrics and GynecologyQilu HospitalDepartment of Cell BiologySchool of Basic Medical ScienceShandong UniversityJinan250012China
| | - Zhaojian Liu
- Key Laboratory of Experimental TeratologyMinistry of EducationDepartment of Obstetrics and GynecologyQilu HospitalDepartment of Cell BiologySchool of Basic Medical ScienceShandong UniversityJinan250012China
- Advanced Medical Research InstituteShandong UniversityJinan250012China
| |
Collapse
|
4
|
Flores-Colón M, Rivera-Serrano M, Reyes-Burgos VG, Rolón JG, Pérez-Santiago J, Marcos-Martínez MJ, Valiyeva F, Vivas-Mejía PE. MicroRNA Expression Profiles in Human Samples and Cell Lines Revealed Nine miRNAs Associated with Cisplatin Resistance in High-Grade Serous Ovarian Cancer. Int J Mol Sci 2024; 25:3793. [PMID: 38612604 PMCID: PMC11011404 DOI: 10.3390/ijms25073793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 03/23/2024] [Accepted: 03/26/2024] [Indexed: 04/14/2024] Open
Abstract
Metastasis and drug resistance are major contributors to cancer-related fatalities worldwide. In ovarian cancer (OC), a staggering 70% develop resistance to the front-line therapy, cisplatin. Despite proposed mechanisms, the molecular events driving cisplatin resistance remain unclear. Dysregulated microRNAs (miRNAs) play a role in OC initiation, progression, and chemoresistance, yet few studies have compared miRNA expression in OC samples and cell lines. This study aimed to identify key miRNAs involved in the cisplatin resistance of high-grade-serous-ovarian-cancer (HGSOC), the most common gynecological malignancy. MiRNA expression profiles were conducted on RNA isolated from formalin-fixed-paraffin-embedded human ovarian tumor samples and HGSOC cell lines. Nine miRNAs were identified in both sample types. Targeting these with oligonucleotide miRNA inhibitors (OMIs) reduced proliferation by more than 50% for miR-203a, miR-96-5p, miR-10a-5p, miR-141-3p, miR-200c-3p, miR-182-5p, miR-183-5p, and miR-1206. OMIs significantly reduced migration for miR-183-5p, miR-203a, miR-296-5p, and miR-1206. Molecular pathway analysis revealed that the nine miRNAs regulate pathways associated with proliferation, invasion, and chemoresistance through PTEN, ZEB1, FOXO1, and SNAI2. High expression of miR-1206, miR-10a-5p, miR-141-3p, and miR-96-5p correlated with poor prognosis in OC patients according to the KM plotter database. These nine miRNAs could be used as targets for therapy and as markers of cisplatin response.
Collapse
Affiliation(s)
- Marienid Flores-Colón
- Department of Biochemistry, University of Puerto Rico, Medical Sciences Campus, San Juan, PR 00936, USA; (M.F.-C.); (V.G.R.-B.)
- Comprehensive Cancer Center, University of Puerto Rico, San Juan, PR 00936, USA; (M.R.-S.); (J.P.-S.); (F.V.)
| | - Mariela Rivera-Serrano
- Comprehensive Cancer Center, University of Puerto Rico, San Juan, PR 00936, USA; (M.R.-S.); (J.P.-S.); (F.V.)
- Department of Biology, University of Puerto Rico, Rio Piedras Campus, San Juan, PR 00936, USA
| | - Víctor G. Reyes-Burgos
- Department of Biochemistry, University of Puerto Rico, Medical Sciences Campus, San Juan, PR 00936, USA; (M.F.-C.); (V.G.R.-B.)
- Comprehensive Cancer Center, University of Puerto Rico, San Juan, PR 00936, USA; (M.R.-S.); (J.P.-S.); (F.V.)
| | - José G. Rolón
- School of Medicine, University of Puerto Rico, Medical Sciences Campus, San Juan, PR 00936, USA;
| | - Josué Pérez-Santiago
- Comprehensive Cancer Center, University of Puerto Rico, San Juan, PR 00936, USA; (M.R.-S.); (J.P.-S.); (F.V.)
| | - María J. Marcos-Martínez
- Department of Pathology and Laboratory Medicine, University of Puerto Rico, Medical Sciences Campus, San Juan, PR 00936, USA;
| | - Fatima Valiyeva
- Comprehensive Cancer Center, University of Puerto Rico, San Juan, PR 00936, USA; (M.R.-S.); (J.P.-S.); (F.V.)
| | - Pablo E. Vivas-Mejía
- Department of Biochemistry, University of Puerto Rico, Medical Sciences Campus, San Juan, PR 00936, USA; (M.F.-C.); (V.G.R.-B.)
- Comprehensive Cancer Center, University of Puerto Rico, San Juan, PR 00936, USA; (M.R.-S.); (J.P.-S.); (F.V.)
| |
Collapse
|
5
|
Madrigal T, Ortega-Bernal D, Herrera LA, González-De la Rosa CH, Domínguez-Gómez G, Aréchaga-Ocampo E, Díaz-Chávez J. Mutant p53 Gain-of-Function Induces Migration and Invasion through Overexpression of miR-182-5p in Cancer Cells. Cells 2023; 12:2506. [PMID: 37887350 PMCID: PMC10605582 DOI: 10.3390/cells12202506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/29/2023] [Accepted: 10/11/2023] [Indexed: 10/28/2023] Open
Abstract
The master-key TP53 gene is a tumor suppressor that is mutated in more than 50% of human cancers. Some p53 mutants lose their tumor suppressor activity and acquire new oncogenic functions, known as a gain of function (GOF). Recent studies have shown that p53 mutants can exert oncogenic effects through specific miRNAs. We identified the differentially expressed miRNA profiles of the three most frequent p53 mutants (p53R273C, p53R248Q, and p53R175H) after their transfection into the Saos-2 cell line (null p53) as compared with p53WT transfected cells. The associations between these miRNAs and the signaling pathways in which they might participate were identified with miRPath Software V3.0. QRT-PCR was employed to validate the miRNA profiles. We observed that p53 mutants have an overall negative effect on miRNA expression. In the global expression profile of the human miRNome regulated by the p53R273C mutant, 72 miRNAs were underexpressed and 35 overexpressed; in the p53R175H miRNAs profile, our results showed the downregulation of 93 and upregulation of 10 miRNAs; and in the miRNAs expression profile regulated by the p53R248Q mutant, we found 167 decreased and 6 increased miRNAs compared with p53WT. However, we found overexpression of some miRNAs, like miR-182-5p, in association with processes such as cell migration and invasion. In addition, we explored whether the induction of cell migration and invasion by the p53R48Q mutant was dependent on miR-182-5p because we found overexpression of miR-182-5p, which is associated with processes such as cell migration and invasion. Inhibition of mutant p53R248Q and miR-182-5p increased FOXF2-MTSS1 levels and decreased cell migration and invasion. In summary, our results suggest that p53 mutants increase the expression of miR-182-5p, and this miRNA is necessary for the p53R248Q mutant to induce cell migration and invasion in a cancer cell model.
Collapse
Affiliation(s)
- Tzitzijanik Madrigal
- Unidad de Investigación en Cáncer, Instituto de Investigaciones Biomédicas-Universidad Nacional Autónoma de México, Instituto Nacional de Cancerología, San Fernando 22, Sección XVI, Tlalpan, CDMX, Mexico City 14080, Mexico; (T.M.); (L.A.H.)
- Departamento de Ciencias Biológicas y de la Salud, UAM Iztapalapa, Mexico City 09340, Mexico
| | - Daniel Ortega-Bernal
- Departamento de Atención a la Salud, UAM Xochimilco, Mexico City 04960, Mexico;
- Departamento de Ciencias Naturales, Unidad Cuajimalpa, Universidad Autonóma Metropolitana, Mexico City 05348, Mexico; (C.H.G.-D.l.R.); (E.A.-O.)
| | - Luis A. Herrera
- Unidad de Investigación en Cáncer, Instituto de Investigaciones Biomédicas-Universidad Nacional Autónoma de México, Instituto Nacional de Cancerología, San Fernando 22, Sección XVI, Tlalpan, CDMX, Mexico City 14080, Mexico; (T.M.); (L.A.H.)
- Escuela de Medicina y Ciencias de la Salud-Tecnológico de Monterrey, Mexico City 14380, Mexico
| | - Claudia Haydée González-De la Rosa
- Departamento de Ciencias Naturales, Unidad Cuajimalpa, Universidad Autonóma Metropolitana, Mexico City 05348, Mexico; (C.H.G.-D.l.R.); (E.A.-O.)
| | - Guadalupe Domínguez-Gómez
- Subdirección de Investigación Clínica, Instituto Nacional de Cancerología, Mexico City 14080, Mexico;
| | - Elena Aréchaga-Ocampo
- Departamento de Ciencias Naturales, Unidad Cuajimalpa, Universidad Autonóma Metropolitana, Mexico City 05348, Mexico; (C.H.G.-D.l.R.); (E.A.-O.)
| | - José Díaz-Chávez
- Unidad de Investigación en Cáncer, Instituto de Investigaciones Biomédicas-Universidad Nacional Autónoma de México, Instituto Nacional de Cancerología, San Fernando 22, Sección XVI, Tlalpan, CDMX, Mexico City 14080, Mexico; (T.M.); (L.A.H.)
| |
Collapse
|
6
|
Pal S, Bhowmick S, Sharma A, Sierra-Fonseca JA, Mondal S, Afolabi F, Roy D. Lymphatic vasculature in ovarian cancer. Biochim Biophys Acta Rev Cancer 2023; 1878:188950. [PMID: 37419192 PMCID: PMC10754213 DOI: 10.1016/j.bbcan.2023.188950] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 06/29/2023] [Accepted: 06/29/2023] [Indexed: 07/09/2023]
Abstract
Ovarian cancer (OVCA) is the second most common gynecological cancer and one of the leading causes of cancer related mortality among women. Recent studies suggest that among ovarian cancer patients at least 70% of the cases experience the involvement of lymph nodes and metastases through lymphatic vascular network. However, the impact of lymphatic system in the growth, spread and the evolution of ovarian cancer, its contribution towards the landscape of ovarian tissue resident immune cells and their metabolic responses is still a major knowledge gap. In this review first we present the epidemiological aspect of the OVCA, the lymphatic architecture of the ovary, we discuss the role of lymphatic circulation in regulation of ovarian tumor microenvironment, metabolic basis of the upregulation of lymphangiogenesis which is often observed during progression of ovarian metastasis and ascites development. Further we describe the implication of several mediators which influence both lymphatic vasculature as well as ovarian tumor microenvironment and conclude with several therapeutic strategies for targeting lymphatic vasculature in ovarian cancer progression in present day.
Collapse
Affiliation(s)
- Sarit Pal
- Department of Medical Physiology, College of Medicine, Texas A&M University, Bryan, TX 77843, United States
| | - Sramana Bhowmick
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata 700073, West Bengal, India
| | - Anurag Sharma
- Pathology and Laboratory Medicine, Cleveland Clinic, Cleveland, OH, United States
| | | | - Susmita Mondal
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata 700073, West Bengal, India
| | - Favour Afolabi
- Department of Biological Sciences, Alcorn State University, Lorman, MS 39096, United States
| | - Debarshi Roy
- Department of Biological Sciences, Alcorn State University, Lorman, MS 39096, United States.
| |
Collapse
|
7
|
Miśkiewicz J, Mielczarek-Palacz A, Gola JM. MicroRNAs as Potential Biomarkers in Gynecological Cancers. Biomedicines 2023; 11:1704. [PMID: 37371799 PMCID: PMC10296063 DOI: 10.3390/biomedicines11061704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 05/25/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
MicroRNAs are non-coding transcripts that, thanks to the ability to regulate the mRNA of target genes, can affect the expression of genes encoding tumor suppressors and oncogenes. They can control many important cellular processes, including apoptosis, differentiation, growth, division, and metabolism. Therefore, miRNAs play an important role in the development of many cancers, including gynecological cancers. Ovarian cancer, endometrial cancer, cervical cancer, and vulvar cancer are the most common cancers in women and are a frequent cause of death. The heterogeneity of the pathogenesis of these gynecological diseases makes the diagnostic process a significant obstacle for modern medicine. To date, many studies have been carried out, in which particular attention has been paid to the molecular pathomechanism of these diseases, with particular emphasis on miRNAs. To date, the changed profile of many miRNAs, which influenced the promotion of proliferation, migration, invasion processes and the simultaneous inhibition of programmed cell death, has been proven many times. Detailed understanding of the molecular effects of miRNAs in the above-mentioned gynecological cancers will enable the development of potential predictive and prognostic biomarkers, as well as the optimization of the diagnostic process.
Collapse
Affiliation(s)
- Joanna Miśkiewicz
- Department of Immunology and Serology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 41-200 Sosnowiec, Poland; (J.M.); (A.M.-P.)
| | - Aleksandra Mielczarek-Palacz
- Department of Immunology and Serology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 41-200 Sosnowiec, Poland; (J.M.); (A.M.-P.)
| | - Joanna Magdalena Gola
- Department of Molecular Biology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 41-200 Sosnowiec, Poland
| |
Collapse
|
8
|
Two oncomiRs, miR-182-5p and miR-103a-3p, Involved in Intravenous Leiomyomatosis. Genes (Basel) 2023; 14:genes14030712. [PMID: 36980984 PMCID: PMC10048324 DOI: 10.3390/genes14030712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/10/2023] [Accepted: 03/11/2023] [Indexed: 03/17/2023] Open
Abstract
Leiomyomas, also referred to as fibroids, belong to the most common type of benign tumors developing in the myometrium of the uterus. Intravenous leiomyomatosis (IVL) tends to be regarded as a rare type of uterine leiomyoma. IVL tumors are characterized by muscle cell masses developing within the uterine and extrauterine venous system. The underlying mechanism responsible for the proliferation of these lesions is still unknown. The aim of the study was to investigate the expression of the two epigenetic factors, oncomiRs miR-182-5p and miR-103a-3p, in intravenous leiomyomatosis. This study was divided into two stages: initially, miR-182-5p and miR-103a-3p expression was assessed in samples coming from intravenous leiomyomatosis localized in myometrium (group I, n = 6), intravenous leiomyomatosis beyond the uterus (group II; n = 5), and the control group, i.e., intramural leiomyomas (group III; n = 9). The expression level of miR-182-5p was significantly higher in samples coming from intravenous leiomyomatosis (group I and group II) as compared to the control group (p = 0.029 and p = 0.024, respectively). In the second part of the study, the expression levels of the studied oncomiRs were compared between seven samples delivered from one woman during a four-year observation. The long-term follow-up of one patient demonstrated significantly elevated levels of both studied oncomiRs in intravenous leiomyomatosis in comparison to intramural leiomyoma samples.
Collapse
|
9
|
Kandettu A, Adiga D, Devi V, Suresh PS, Chakrabarty S, Radhakrishnan R, Kabekkodu SP. Deregulated miRNA clusters in ovarian cancer: Imperative implications in personalized medicine. Genes Dis 2022; 9:1443-1465. [PMID: 36157483 PMCID: PMC9485269 DOI: 10.1016/j.gendis.2021.12.026] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 12/04/2021] [Accepted: 12/31/2021] [Indexed: 11/25/2022] Open
Abstract
Ovarian cancer (OC) is one of the most common and fatal types of gynecological cancer. OC is usually detected at the advanced stages of the disease, making it highly lethal. miRNAs are single-stranded, small non-coding RNAs with an approximate size ranging around 22 nt. Interestingly, a considerable proportion of miRNAs are organized in clusters with miRNA genes placed adjacent to one another, getting transcribed together to result in miRNA clusters (MCs). MCs comprise two or more miRNAs that follow the same orientation during transcription. Abnormal expression of the miRNA cluster has been identified as one of the key drivers in OC. MC exists both as tumor-suppressive and oncogenic clusters and has a significant role in OC pathogenesis by facilitating cancer cells to acquire various hallmarks. The present review summarizes the regulation and biological function of MCs in OC. The review also highlights the utility of abnormally expressed MCs in the clinical management of OC.
Collapse
Affiliation(s)
- Amoolya Kandettu
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Divya Adiga
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Vasudha Devi
- Department of Pharmacology, Centre for Cardiovascular Pharmacology, Melaka Manipal Medical College, Manipal Academy of Higher Education, Manipal Campus, Manipal, Karnataka 576104, India
| | - Padmanaban S. Suresh
- School of Biotechnology, National Institute of Technology, Calicut, Kerala 673601, India
| | - Sanjiban Chakrabarty
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
- Center for DNA Repair and Genome Stability (CDRGS), Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Raghu Radhakrishnan
- Department of Oral Pathology, Manipal College of Dental Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Shama Prasada Kabekkodu
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
- Center for DNA Repair and Genome Stability (CDRGS), Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| |
Collapse
|
10
|
Serra M, Pal R, Puliga E, Sulas P, Cabras L, Cusano R, Giordano S, Perra A, Columbano A, Kowalik MA. mRNA-miRNA networks identify metabolic pathways associated to the anti-tumorigenic effect of thyroid hormone on preneoplastic nodules and hepatocellular carcinoma. Front Oncol 2022; 12:941552. [PMID: 36203462 PMCID: PMC9530455 DOI: 10.3389/fonc.2022.941552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 08/30/2022] [Indexed: 11/13/2022] Open
Abstract
Background Thyroid hormones (THs) inhibit hepatocellular carcinoma (HCC) through different mechanisms. However, whether microRNAs play a role in the antitumorigenic effect of THs remains unknown. Methods By next generation sequencing (NGS) we performed a comprehensive comparative miRNomic and transcriptomic analysis of rat hepatic preneoplastic lesions exposed or not to a short-term treatment with triiodothyronine (T3). The expression of the most deregulated miRs was also investigated in rat HCCs, and in human hepatoma cell lines, treated or not with T3. Results Among miRs down-regulated in preneoplastic nodules following T3, co-expression networks revealed those targeting thyroid hormone receptor-β (Thrβ) and deiodinase1, and Oxidative Phosphorylation. On the other hand, miRs targeting members of the Nrf2 Oxidative Pathway, Glycolysis, Pentose Phosphate Pathway and Proline biosynthesis – all involved in the metabolic reprogramming displayed by preneoplastic lesions– were up-regulated. Notably, while the expression of most miRs deregulated in preneoplastic lesions was not altered in HCC or in hepatoma cells, miR-182, a miR known to target Dio1 and mitochondrial complexes, was down-deregulated by T3 treatment at all stages of hepatocarcinogenesis and in hepatocarcinoma cell lines. In support to the possible critical role of miR-182 in hepatocarcinogenesis, exogenous expression of this miR significantly impaired the inhibitory effect of T3 on the clonogenic growth capacity of human HCC cells. Conclusions This work identified several miRNAs, so far never associated to T3. In addition, the precise definition of the miRNA-mRNA networks elicited by T3 treatment gained in this study may provide a better understanding of the key regulatory events underlying the inhibitory effect of T3 on HCC development. In this context, T3-induced down-regulation of miR-182 appears as a promising tool.
Collapse
Affiliation(s)
- Marina Serra
- Department of Biomedical Sciences, Unit of Oncology and Molecular Pathology, University of Cagliari, Cagliari, Italy
| | - Rajesh Pal
- Department of Biomedical Sciences, Unit of Oncology and Molecular Pathology, University of Cagliari, Cagliari, Italy
| | - Elisabetta Puliga
- Department of Oncology, University of Turin, Turin, Italy
- Candiolo Cancer Institute-Fondazione del Piemonte per l'Oncologia (FPO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Candiolo, Italy
| | - Pia Sulas
- Department of Biomedical Sciences, Unit of Oncology and Molecular Pathology, University of Cagliari, Cagliari, Italy
| | - Lavinia Cabras
- Department of Biomedical Sciences, Unit of Oncology and Molecular Pathology, University of Cagliari, Cagliari, Italy
| | - Roberto Cusano
- Centro di Ricerca, Sviluppo e Studi Superiori in Sardegna (CRS4), Pula, Italy
| | - Silvia Giordano
- Department of Oncology, University of Turin, Turin, Italy
- Candiolo Cancer Institute-Fondazione del Piemonte per l'Oncologia (FPO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Candiolo, Italy
| | - Andrea Perra
- Department of Biomedical Sciences, Unit of Oncology and Molecular Pathology, University of Cagliari, Cagliari, Italy
| | - Amedeo Columbano
- Department of Biomedical Sciences, Unit of Oncology and Molecular Pathology, University of Cagliari, Cagliari, Italy
- *Correspondence: Amedeo Columbano, ; Marta Anna Kowalik,
| | - Marta Anna Kowalik
- Department of Biomedical Sciences, Unit of Oncology and Molecular Pathology, University of Cagliari, Cagliari, Italy
- *Correspondence: Amedeo Columbano, ; Marta Anna Kowalik,
| |
Collapse
|
11
|
Fu Y, Liu H, Long M, Song L, Meng Z, Lin S, Zhang Y, Qin J. Icariin attenuates the tumor growth by targeting miR-1-3p/TNKS2/Wnt/β-catenin signaling axis in ovarian cancer. Front Oncol 2022; 12:940926. [PMID: 36185280 PMCID: PMC9516086 DOI: 10.3389/fonc.2022.940926] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 08/18/2022] [Indexed: 11/20/2022] Open
Abstract
Purpose Despite various therapy advances, ovarian cancer remains an incurable disease for which survival rates have only modestly improved. Natural products are important sources of anti-cancer lead compounds. Icariin exhibited broad anti-cancer efficacy. However, the mechanism of icariin against ovarian cancer is poorly elucidated. Methods Cell viability was detected to evaluate the effect of icariin on SKOV-3 cells. The cell cycle and apoptosis were analyzed. The transcript of SKOV-3 cells was profiled by RNA-seq. GSEA and DEGs analyses were performed to interpret gene expression data. Western blot and TOP/FOP flash assay were applied to detect Wnt/β-catenin signaling. MiRDB database and dual-luciferase reporter assay was applied to study the regulation of miR-1-3p on TNKS2. Anti-tumor efficacy of icariin was evaluated by xenograft mouse model. Immunohistochemistry was performed with antibodies against Ki67. Results Icariin significantly suppressed the proliferation of SKOV-3 cells. Furthermore, icariin stalled cell cycle and induced apoptosis by blocking TNKS2/Wnt/β-catenin pathway through upregulating the level of miR-1-3p. Finally, icariin dramatically suppressed tumor growth in vivo. Conclusions In this study, we demonstrated for the first time that icariin significantly attenuated the growth of ovarian tumor in xenograft mouse model. Furthermore, we systematically revealed that icariin attenuates the tumor progression by suppressing TNKS2/Wnt/β-catenin signaling via upregulating the level of miR-1-3p in ovarian cancer with transcriptome analysis.
Collapse
Affiliation(s)
- Yanjin Fu
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Haiquan Liu
- Huizhou Traditional Chinese Medicine Hospital, Guangzhou University of Traditional Chinese Medicine, Huizhou, Guangdong, China
| | - Mengsha Long
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Linliang Song
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Zuyu Meng
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Shaozi Lin
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Yiyao Zhang
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - JiaJia Qin
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
- *Correspondence: JiaJia Qin,
| |
Collapse
|
12
|
Stieg DC, Wang Y, Liu LZ, Jiang BH. ROS and miRNA Dysregulation in Ovarian Cancer Development, Angiogenesis and Therapeutic Resistance. Int J Mol Sci 2022; 23:ijms23126702. [PMID: 35743145 PMCID: PMC9223852 DOI: 10.3390/ijms23126702] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/05/2022] [Accepted: 06/14/2022] [Indexed: 12/11/2022] Open
Abstract
The diverse repertoires of cellular mechanisms that progress certain cancer types are being uncovered by recent research and leading to more effective treatment options. Ovarian cancer (OC) is among the most difficult cancers to treat. OC has limited treatment options, especially for patients diagnosed with late-stage OC. The dysregulation of miRNAs in OC plays a significant role in tumorigenesis through the alteration of a multitude of molecular processes. The development of OC can also be due to the utilization of endogenously derived reactive oxygen species (ROS) by activating signaling pathways such as PI3K/AKT and MAPK. Both miRNAs and ROS are involved in regulating OC angiogenesis through mediating multiple angiogenic factors such as hypoxia-induced factor (HIF-1) and vascular endothelial growth factor (VEGF). The NAPDH oxidase subunit NOX4 plays an important role in inducing endogenous ROS production in OC. This review will discuss several important miRNAs, NOX4, and ROS, which contribute to therapeutic resistance in OC, highlighting the effective therapeutic potential of OC through these mechanisms.
Collapse
Affiliation(s)
- David C. Stieg
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA; (D.C.S.); (L.-Z.L.)
| | - Yifang Wang
- Department of Pathology, Anatomy & Cell Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA;
| | - Ling-Zhi Liu
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA; (D.C.S.); (L.-Z.L.)
| | - Bing-Hua Jiang
- Department of Pathology, Anatomy & Cell Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA;
- Correspondence:
| |
Collapse
|
13
|
Interactions between miRNAs and Double-Strand Breaks DNA Repair Genes, Pursuing a Fine-Tuning of Repair. Int J Mol Sci 2022; 23:ijms23063231. [PMID: 35328651 PMCID: PMC8954595 DOI: 10.3390/ijms23063231] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/06/2022] [Accepted: 03/09/2022] [Indexed: 02/04/2023] Open
Abstract
The repair of DNA damage is a crucial process for the correct maintenance of genetic information, thus, allowing the proper functioning of cells. Among the different types of lesions occurring in DNA, double-strand breaks (DSBs) are considered the most harmful type of lesion, which can result in significant loss of genetic information, leading to diseases, such as cancer. DSB repair occurs through two main mechanisms, called non-homologous end joining (NHEJ) and homologous recombination repair (HRR). There is evidence showing that miRNAs play an important role in the regulation of genes acting in NHEJ and HRR mechanisms, either through direct complementary binding to mRNA targets, thus, repressing translation, or by targeting other genes involved in the transcription and activity of DSB repair genes. Therefore, alteration of miRNA expression has an impact on the ability of cells to repair DSBs, which, in turn, affects cancer therapy sensitivity. This latter gives account of the importance of miRNAs as regulators of NHEJ and HRR and places them as a promising target to improve cancer therapy. Here, we review recent reports demonstrating an association between miRNAs and genes involved in NHEJ and HRR. We employed the Web of Science search query TS (“gene official symbol/gene aliases*” AND “miRNA/microRNA/miR-”) and focused on articles published in the last decade, between 2010 and 2021. We also performed a data analysis to represent miRNA–mRNA validated interactions from TarBase v.8, in order to offer an updated overview about the role of miRNAs as regulators of DSB repair.
Collapse
|
14
|
Ma C, He D, Tian P, Wang Y, He Y, Wu Q, Jia Z, Zhang X, Zhang P, Ying H, Jin ZB, Hu G. miR-182 targeting reprograms tumor-associated macrophages and limits breast cancer progression. Proc Natl Acad Sci U S A 2022; 119:e2114006119. [PMID: 35105806 PMCID: PMC8833194 DOI: 10.1073/pnas.2114006119] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 12/28/2021] [Indexed: 12/12/2022] Open
Abstract
The protumor roles of alternatively activated (M2) tumor-associated macrophages (TAMs) have been well established, and macrophage reprogramming is an important therapeutic goal. However, the mechanisms of TAM polarization remain incompletely understood, and effective strategies for macrophage targeting are lacking. Here, we show that miR-182 in macrophages mediates tumor-induced M2 polarization and can be targeted for therapeutic macrophage reprogramming. Constitutive miR-182 knockout in host mice and conditional knockout in macrophages impair M2-like TAMs and breast tumor development. Targeted depletion of macrophages in mice blocks the effect of miR-182 deficiency in tumor progression while reconstitution of miR-182-expressing macrophages promotes tumor growth. Mechanistically, cancer cells induce miR-182 expression in macrophages by TGFβ signaling, and miR-182 directly suppresses TLR4, leading to NFκb inactivation and M2 polarization of TAMs. Importantly, therapeutic delivery of antagomiR-182 with cationized mannan-modified extracellular vesicles effectively targets macrophages, leading to miR-182 inhibition, macrophage reprogramming, and tumor suppression in multiple breast cancer models of mice. Overall, our findings reveal a crucial TGFβ/miR-182/TLR4 axis for TAM polarization and provide rationale for RNA-based therapeutics of TAM targeting in cancer.
Collapse
Affiliation(s)
- Chengxin Ma
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Dasa He
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Pu Tian
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yuan Wang
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yunfei He
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Qiuyao Wu
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Zhenchang Jia
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xue Zhang
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Peiyuan Zhang
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Hao Ying
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Zi-Bing Jin
- Beijing Institute of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing Key Lab of Ophthalmology and Visual Sciences, Beijing 100005, China
| | - Guohong Hu
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China;
| |
Collapse
|
15
|
Todorova VK, Byrum SD, Gies AJ, Haynie C, Smith H, Reyna NS, Makhoul I. Circulating Exosomal microRNAs as Predictive Biomarkers of Neoadjuvant Chemotherapy Response in Breast Cancer. Curr Oncol 2022; 29:613-630. [PMID: 35200555 PMCID: PMC8870357 DOI: 10.3390/curroncol29020055] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/17/2022] [Accepted: 01/24/2022] [Indexed: 12/24/2022] Open
Abstract
Background: Neoadjuvant chemotherapy (NACT) is an increasingly used approach for treatment of breast cancer. The pathological complete response (pCR) is considered a good predictor of disease-specific survival. This study investigated whether circulating exosomal microRNAs could predict pCR in breast cancer patients treated with NACT. Method: Plasma samples of 20 breast cancer patients treated with NACT were collected prior to and after the first cycle. RNA sequencing was used to determine microRNA profiling. The Cancer Genome Atlas (TCGA) was used to explore the expression patterns and survivability of the candidate miRNAs, and their potential targets based on the expression levels and copy number variation (CNV) data. Results: Three miRNAs before that NACT (miR-30b, miR-328 and miR-423) predicted pCR in all of the analyzed samples. Upregulation of miR-127 correlated with pCR in triple-negative breast cancer (TNBC). After the first NACT dose, pCR was predicted by exo-miR-141, while miR-34a, exo-miR182, and exo-miR-183 predicted non-pCR. A significant correlation between the candidate miRNAs and the overall survival, subtype, and metastasis in breast cancer, suggesting their potential role as predictive biomarkers of pCR. Conclusions: If the miRNAs identified in this study are validated in a large cohort of patients, they might serve as predictive non-invasive liquid biopsy biomarkers for monitoring pCR to NACT in breast cancer.
Collapse
Affiliation(s)
- Valentina K. Todorova
- Division of Medical Oncology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
- Correspondence:
| | - Stephanie D. Byrum
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (S.D.B.); (A.J.G.)
| | - Allen J. Gies
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (S.D.B.); (A.J.G.)
| | - Cade Haynie
- Biology Department, Ouachita Baptist University, Arkadelphia, AR 71998, USA; (C.H.); (H.S.); (N.S.R.)
| | - Hunter Smith
- Biology Department, Ouachita Baptist University, Arkadelphia, AR 71998, USA; (C.H.); (H.S.); (N.S.R.)
| | - Nathan S. Reyna
- Biology Department, Ouachita Baptist University, Arkadelphia, AR 71998, USA; (C.H.); (H.S.); (N.S.R.)
| | - Issam Makhoul
- Division of Medical Oncology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
| |
Collapse
|
16
|
Li Y, Wang S, Li P, Li Y, Liu Y, Fang H, Zhang X, Liu Z, Kong B. Rad50 promotes ovarian cancer progression through NF-κB activation. J Cell Mol Med 2021; 25:10961-10972. [PMID: 34734468 PMCID: PMC8642684 DOI: 10.1111/jcmm.17017] [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: 11/30/2020] [Revised: 09/20/2021] [Accepted: 09/30/2021] [Indexed: 01/12/2023] Open
Abstract
Rad50 is a component of MRN (Mre11-Rad50-Nbs1), which participates in DNA double-strand break repair and DNA-damage checkpoint activation. Here, we sought to investigate the clinical and functional significance of Rad50 in high-grade serous ovarian cancer (HGSOC). We found that Rad50 was frequently upregulated in HGSOCs and enhanced Rad50 expression inversely correlated with patient survival. In addition, ectopic expression of Rad50 promoted proliferation/invasion and induced EMT of ovarian cancer cells, whereas knockdown of Rad50 led to decreased aggressive behaviors. Mechanistic investigations revealed that Rad50 induced aggressiveness in HGSOC via activation of NF-κB signaling pathway. Moreover, we identified CARD9 as an interacting protein of Rad50 in ovarian cancer cells and the activation of NF-κB pathway by Rad50 is CARD9 dependent. Our findings provide evidence that Rad50 exhibits oncogenic property via NF-κB activation in HGSOC.
Collapse
Affiliation(s)
- Yinuo Li
- Key Laboratory of Experimental Teratology, Ministry of Education, Department of Obstetrics and Gynecology, Qilu Hospital, Department of Cell Biology, School of Basic Medical SciencesCheeloo College of MedicineShandong UniversityJinanChina
| | - Shourong Wang
- Key Laboratory of Experimental Teratology, Ministry of Education, Department of Obstetrics and Gynecology, Qilu Hospital, Department of Cell Biology, School of Basic Medical SciencesCheeloo College of MedicineShandong UniversityJinanChina
| | - Peng Li
- Key Laboratory of Experimental Teratology, Ministry of Education, Department of Obstetrics and Gynecology, Qilu Hospital, Department of Cell Biology, School of Basic Medical SciencesCheeloo College of MedicineShandong UniversityJinanChina
| | - Yingwei Li
- Key Laboratory of Experimental Teratology, Ministry of Education, Department of Obstetrics and Gynecology, Qilu Hospital, Department of Cell Biology, School of Basic Medical SciencesCheeloo College of MedicineShandong UniversityJinanChina
| | - Yao Liu
- Key Laboratory of Experimental Teratology, Ministry of Education, Department of Obstetrics and Gynecology, Qilu Hospital, Department of Cell Biology, School of Basic Medical SciencesCheeloo College of MedicineShandong UniversityJinanChina
| | - Haiya Fang
- Department of Obstetrics & GynecologyJinhua Hospital of Zhejiang UniversityJinhuaChina
| | - Xiyu Zhang
- Key Laboratory of Experimental Teratology, Ministry of Education, Department of Obstetrics and Gynecology, Qilu Hospital, Department of Cell Biology, School of Basic Medical SciencesCheeloo College of MedicineShandong UniversityJinanChina
- Advanced Medical Research InstituteCheeloo College of MedicineShandong UniversityJinanChina
| | - Zhaojian Liu
- Key Laboratory of Experimental Teratology, Ministry of Education, Department of Obstetrics and Gynecology, Qilu Hospital, Department of Cell Biology, School of Basic Medical SciencesCheeloo College of MedicineShandong UniversityJinanChina
- Advanced Medical Research InstituteCheeloo College of MedicineShandong UniversityJinanChina
| | - Beihua Kong
- Key Laboratory of Experimental Teratology, Ministry of Education, Department of Obstetrics and Gynecology, Qilu Hospital, Department of Cell Biology, School of Basic Medical SciencesCheeloo College of MedicineShandong UniversityJinanChina
| |
Collapse
|
17
|
Wei JJ. Linking altered microRNA expression to racial disparities in uterine serous carcinoma. Gynecol Oncol 2021; 163:446-447. [PMID: 34836615 DOI: 10.1016/j.ygyno.2021.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 10/29/2021] [Accepted: 11/03/2021] [Indexed: 11/19/2022]
Affiliation(s)
- Jian-Jun Wei
- Department of Pathology, Department Obstetrics and Gynecology, Northwestern University, Feinberg School of Medicine, 251 East Huron Street, Feinberg 7-334, Chicago, IL 60611, USA.
| |
Collapse
|
18
|
Mirahmadi Y, Nabavi R, Taheri F, Samadian MM, Ghale-Noie ZN, Farjami M, Samadi-khouzani A, Yousefi M, Azhdari S, Salmaninejad A, Sahebkar A. MicroRNAs as Biomarkers for Early Diagnosis, Prognosis, and Therapeutic Targeting of Ovarian Cancer. JOURNAL OF ONCOLOGY 2021; 2021:3408937. [PMID: 34721577 PMCID: PMC8553480 DOI: 10.1155/2021/3408937] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 09/27/2021] [Indexed: 02/06/2023]
Abstract
Ovarian cancer is the major cause of gynecologic cancer-related mortality. Regardless of outstanding advances, which have been made for improving the prognosis, diagnosis, and treatment of ovarian cancer, the majority of the patients will die of the disease. Late-stage diagnosis and the occurrence of recurrent cancer after treatment are the most important causes of the high mortality rate observed in ovarian cancer patients. Unraveling the molecular mechanisms involved in the pathogenesis of ovarian cancer may help find new biomarkers and therapeutic targets for ovarian cancer. MicroRNAs (miRNAs) are small noncoding RNAs that regulate gene expression, mostly at the posttranscriptional stage, through binding to mRNA targets and inducing translational repression or degradation of target via the RNA-induced silencing complex. Over the last two decades, the role of miRNAs in the pathogenesis of various human cancers, including ovarian cancer, has been documented in multiple studies. Consequently, these small RNAs could be considered as reliable markers for prognosis and early diagnosis. Furthermore, given the function of miRNAs in various cellular pathways, including cell survival and differentiation, targeting miRNAs could be an interesting approach for the treatment of human cancers. Here, we review our current understanding of the most updated role of the important dysregulation of miRNAs and their roles in the progression and metastasis of ovarian cancer. Furthermore, we meticulously discuss the significance of miRNAs as prognostic and diagnostic markers. Lastly, we mention the opportunities and the efforts made for targeting ovarian cancer through inhibition and/or stimulation of the miRNAs.
Collapse
Affiliation(s)
- Yegane Mirahmadi
- Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Genetics Research Centre, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Fourough Taheri
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Mohammad Mahdi Samadian
- Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zari Naderi Ghale-Noie
- Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Genetics Research Centre, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahsa Farjami
- Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Genetics Research Centre, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Abbas Samadi-khouzani
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Meysam Yousefi
- Department of Medical Genetics, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Sara Azhdari
- Department of Anatomy and Embryology, School of Medicine, Bam University of Medical Sciences, Bam, Iran
| | - Arash Salmaninejad
- Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Genetics Research Centre, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Genetics, Faculty of Medicine, Guilan University of Medical Sciences, Guilan, Iran
| | - Amirhossein Sahebkar
- Applied Biomedical 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
|
19
|
Xiao D, Xie J, Wu S. MicroRNA-182 is a potential biomarker for prognosis of gastric cancer: A protocol for meta-analysis and bioinformatics analysis. Medicine (Baltimore) 2021; 100:e25830. [PMID: 34087825 PMCID: PMC8183740 DOI: 10.1097/md.0000000000025830] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 04/16/2021] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Being the second leading cause of cancer death in the world, gastric cancer is a common malignant tumor in digestive system. Most patients were diagnosed in advanced stage and had poor prognosis. In recent years, related studies have displayed that MicroRNA-182 (miRNA-182) can promote the proliferation, infiltration, metastasis and drug resistance of tumor cells, so it can be used as a new molecular marker for the early diagnosis, prognosis, and treatment of tumors. However, the expression and prognosis of miRNA-182 in gastric cancer are not clear. Therefore, this study conducted a meta-analysis to further clarify the relationship between the expression of miRNA-182 in gastric cancer and prognosis. In addition, a bioinformatics analysis was adopted to further analyze the possible molecular mechanism of miRNA-182, so as to provide a theoretical basis for the diagnosis, treatment, and prognosis of patients suffering from gastric cancer. METHODS The following electronic databases were searched on computer: Wanfang, Chinese Biomedical Literature Database, Chinese National Knowledge Infrastructure, the Chongqing VIP Chinese Science and Technology Periodical Database, PubMed, Embase, and Web of Science databases. The retrieval time is set to build the database until April 2021. Combined hazard ratios (HRs) and 95% confidence intervals (95% CIs) were used to evaluate the effects of miRNA-182 on the prognosis of gastric cancer. Stata 16.0 software was applied for the meta-analysis. The expression of miRNA-182 in gastric cancer was analyzed by Gene Expression Omnibus database and The Cancer Genome Atlas database. The survival curve of miRNA-182 differential expression was analyzed by OncomiR. The target genes of miRNA-182 were predicted by TargetScan, miRBase, miRTarBase, starBase V2.0, and miRWalk. The target genes were obtained by the intersection of Wayne diagram. DAVID database was used for gene ontology (GO) and Kyoto encyclopedia of genes and genomes enrichment analysis. STRING database and Cytoscape were applied to construct Protein-protein interaction network to obtain key genes (hub gene). The expression of hub gene in gastric cancer was analyzed by gene expression profiling interactive analysis. The survival curve between hub gene and prognosis of gastric cancer was drawn by Kaplan-Meier Plotter database. TIMER database was used to analyze the relationship between hub gene expression and immune cell infiltration in gastric cancer. RESULTS The results of this meta-analysis will be submitted to a peer-reviewed journal for publication. CONCLUSION This study provides high-quality evidence support for the expression of miRNA-182 and the prognosis of gastric cancer. Through bioinformatics analysis, we further discussed the mechanism of miRNA-182 in gastric cancer and the understanding of related pathways. OSF REGISTRATION NUMBER DOI 10.17605/OSF.IO/EHJ6X.
Collapse
|
20
|
Liu J, Yoo J, Ho JY, Jung Y, Lee S, Hur SY, Choi YJ. Plasma-derived exosomal miR-4732-5p is a promising noninvasive diagnostic biomarker for epithelial ovarian cancer. J Ovarian Res 2021; 14:59. [PMID: 33910598 PMCID: PMC8082916 DOI: 10.1186/s13048-021-00814-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 04/20/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Exosomal miRNAs regulate gene expression and play important roles in several diseases. We used exosomal miRNA profiling to investigate diagnostic biomarkers of epithelial ovarian cancer (EOC). METHODS In total, 55 individuals were enrolled, comprising healthy (n = 21) and EOC subjects (n = 34). Small mRNA (smRNA) sequencing and real-time PCR (RT-PCR) were performed to identify potential biomarkers. Receiver operating characteristic (ROC) curves were conducted to determine biomarker sensitivity and specificity. RESULTS Using smRNA sequencing, we identified seven up-regulated (miR-4732-5p, miR-877-5p, miR-574-3p, let-7a-5p, let-7b-5p, let-7c-5p, and let-7f-5p) and two down-regulated miRNAs (miR-1273f and miR-342-3p) in EOC patients when compared with healthy subjects. Of these, miR-4732-5p and miR-1273f were the most up-regulated and down-regulated respectively, therefore they were selected for RT-PCR analysis. Plasma derived exosomal miR-4732-5p had an area under the ROC curve of 0.889, with 85.7% sensitivity and 82.4% specificity in distinguishing EOC patients from healthy subjects (p<0.0001) and could be a potential biomarker for monitoring the EOC progression from early stage to late stage (p = 0.018). CONCLUSIONS Plasma derived exosomal miR-4732-5p may be a promising candidate biomarker for diagnosing EOC.
Collapse
Affiliation(s)
- Jingjing Liu
- Department of Obstetrics and Gynecology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Obstetrics and Gynecology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jigeun Yoo
- Department of Obstetrics and Gynecology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jung Yoon Ho
- Department of Obstetrics and Gynecology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yuyeon Jung
- Department of Obstetrics and Gynecology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sanha Lee
- Department of Obstetrics and Gynecology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Soo Young Hur
- Department of Obstetrics and Gynecology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Youn Jin Choi
- Department of Obstetrics and Gynecology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| |
Collapse
|
21
|
HMGA2 as a Critical Regulator in Cancer Development. Genes (Basel) 2021; 12:genes12020269. [PMID: 33668453 PMCID: PMC7917704 DOI: 10.3390/genes12020269] [Citation(s) in RCA: 111] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 02/01/2021] [Accepted: 02/08/2021] [Indexed: 02/07/2023] Open
Abstract
The high mobility group protein 2 (HMGA2) regulates gene expression by binding to AT-rich regions of DNA. Akin to other DNA architectural proteins, HMGA2 is highly expressed in embryonic stem cells during embryogenesis, while its expression is more limited at later stages of development and in adulthood. Importantly, HMGA2 is re-expressed in nearly all human malignancies, where it promotes tumorigenesis by multiple mechanisms. HMGA2 increases cancer cell proliferation by promoting cell cycle entry and inhibition of apoptosis. In addition, HMGA2 influences different DNA repair mechanisms and promotes epithelial-to-mesenchymal transition by activating signaling via the MAPK/ERK, TGFβ/Smad, PI3K/AKT/mTOR, NFkB, and STAT3 pathways. Moreover, HMGA2 supports a cancer stem cell phenotype and renders cancer cells resistant to chemotherapeutic agents. In this review, we discuss these oncogenic roles of HMGA2 in different types of cancers and propose that HMGA2 may be used for cancer diagnostic, prognostic, and therapeutic purposes.
Collapse
|
22
|
Immunohistochemical Expression Status of p53, CD44v9, and Ki-67 in a Series of Fallopian Tube Lesions of High-grade Serous Carcinoma. Int J Gynecol Pathol 2021; 40:419-426. [PMID: 34397764 DOI: 10.1097/pgp.0000000000000738] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Pelvic high-grade serous carcinoma (HGSC) has been postulated to arise via a stepwise accumulation of (epi)genetic alterations from normal epithelium to secretory cell outgrowth (SCOUT), p53 signature, and serous tubal intraepithelial carcinoma (STIC) to invasive HGSC. The aim of this study is to investigate alterations in p53 and CD44v9 expression and the status of Ki-67 labeling index in a series of fallopian tube lesions of HGSC patients. A total of 45 specimens were analyzed in 16 patients with HGSC, and their lesions were categorized as follows: morphologically normal fallopian tube epithelium (FTE, n=6 samples), SCOUT (n=5), p53 signature (n=4), dormant STIC (n=8), active STIC (n=6), and HGSC (n=16). Morphologic features and immunohistochemical expression patterns of the p53 protein, CD44v9 protein, and Ki-67 antigen were blindly evaluated by 2 pathologists. Increased nuclear p53 protein accumulation was observed in p53 signature, dormant STIC, active STIC and HGSC compared with normal FTE and SCOUT (P<0.001). Immunohistochemistry scores of CD44v9 protein expression were significantly higher in normal FTE, SCOUT, and p53 signature than in dormant STIC, active STIC, and HGSC (P<0.001). Both active STIC and HGSC had significantly higher Ki-67 labeling indices than normal FTE, SCOUT, p53 signature and dormant STIC (P<0.001). CD44v9 loss contributes to the stepwise progression of p53 signature to dormant STIC. In conclusion, p53 mutation followed by CD44v9 loss may be involved in the evolution of STIC, which may confer positive clonal selection with a growth and survival advantage.
Collapse
|
23
|
Nguyen VHL, Yue C, Du KY, Salem M, O’Brien J, Peng C. The Role of microRNAs in Epithelial Ovarian Cancer Metastasis. Int J Mol Sci 2020; 21:ijms21197093. [PMID: 32993038 PMCID: PMC7583982 DOI: 10.3390/ijms21197093] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 09/22/2020] [Accepted: 09/23/2020] [Indexed: 12/11/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is the deadliest gynecological cancer, and the major cause of death is mainly attributed to metastasis. MicroRNAs (miRNAs) are a group of small non-coding RNAs that exert important regulatory functions in many biological processes through their effects on regulating gene expression. In most cases, miRNAs interact with the 3′ UTRs of target mRNAs to induce their degradation and suppress their translation. Aberrant expression of miRNAs has been detected in EOC tumors and/or the biological fluids of EOC patients. Such dysregulation occurs as the result of alterations in DNA copy numbers, epigenetic regulation, and miRNA biogenesis. Many studies have demonstrated that miRNAs can promote or suppress events related to EOC metastasis, such as cell migration, invasion, epithelial-to-mesenchymal transition, and interaction with the tumor microenvironment. In this review, we provide a brief overview of miRNA biogenesis and highlight some key events and regulations related to EOC metastasis. We summarize current knowledge on how miRNAs are dysregulated, focusing on those that have been reported to regulate metastasis. Furthermore, we discuss the role of miRNAs in promoting and inhibiting EOC metastasis. Finally, we point out some limitations of current findings and suggest future research directions in the field.
Collapse
Affiliation(s)
- Vu Hong Loan Nguyen
- Department of Biology, York University, Toronto, ON M3J 1P3, Canada; (V.H.L.N.); (C.Y.); (K.Y.D.); (M.S.); (J.O.)
| | - Chenyang Yue
- Department of Biology, York University, Toronto, ON M3J 1P3, Canada; (V.H.L.N.); (C.Y.); (K.Y.D.); (M.S.); (J.O.)
| | - Kevin Y. Du
- Department of Biology, York University, Toronto, ON M3J 1P3, Canada; (V.H.L.N.); (C.Y.); (K.Y.D.); (M.S.); (J.O.)
| | - Mohamed Salem
- Department of Biology, York University, Toronto, ON M3J 1P3, Canada; (V.H.L.N.); (C.Y.); (K.Y.D.); (M.S.); (J.O.)
| | - Jacob O’Brien
- Department of Biology, York University, Toronto, ON M3J 1P3, Canada; (V.H.L.N.); (C.Y.); (K.Y.D.); (M.S.); (J.O.)
| | - Chun Peng
- Department of Biology, York University, Toronto, ON M3J 1P3, Canada; (V.H.L.N.); (C.Y.); (K.Y.D.); (M.S.); (J.O.)
- Centre for Research in Biomolecular Interactions, York University, Toronto, ON M3J 1P3, Canada
- Correspondence:
| |
Collapse
|
24
|
Construction of an miRNA-mRNA regulatory network in colorectal cancer with bioinformatics methods. Anticancer Drugs 2020; 30:588-595. [PMID: 30601194 DOI: 10.1097/cad.0000000000000745] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Colorectal cancer (CRC) is the third most common cancer worldwide. This study aimed to explore the regulatory mechanisms of miRNAs in CRC. Differentially expressed miRNAs (DEMs) and differentially expressed genes (DEGs) in CRC tissue samples compared with control samples in mRNA and miRNA datasets were screened. Functional and pathway enrichment analysis of the DEGs was carried out. Targets of the DEMs were identified. Overlaps between the DEGs and targets of DEMs were selected. The miRNA-mRNA regulatory network of these overlaps was constructed and visualized. The candidate genes selected were validated by quantitative real-time PCR. DEGs were identified and considered DEGs-1 and DEGs-2. A total of 584 genes in DEGs-1 and 527 genes in DEGs-2 were obtained, including 465 overlaps, and 44 DEMs were identified. The overlaps were enriched in 46 Gene Ontology terms and 19 Kyoto Encyclopedia of Genes and Genomes pathways. Moreover, 137 overlapped genes between targets of the DEMs and the 465 overlaps were obtained. The miRNA-mRNA regulating network of the 137 overlapped genes was constructed. Extracellular matrix-related proteins and pathways might play critical roles in the development of CRC. The quantitative real-time PCR results of the candidates were in agreement with the bioinformatics analysis. miR-128, miR-182, and miR-143 might be key miRNAs regulating cell proliferation and metastasis of CRC.
Collapse
|
25
|
Nazari-Shafti TZ, Neuber S, Garcia Duran A, Xu Z, Beltsios E, Seifert M, Falk V, Stamm C. Human mesenchymal stromal cells and derived extracellular vesicles: Translational strategies to increase their proangiogenic potential for the treatment of cardiovascular disease. Stem Cells Transl Med 2020; 9:1558-1569. [PMID: 32761804 PMCID: PMC7695640 DOI: 10.1002/sctm.19-0432] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 05/14/2020] [Accepted: 05/18/2020] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) offer great potential for the treatment of cardiovascular diseases (CVDs) such as myocardial infarction and heart failure. Studies have revealed that the efficacy of MSCs is mainly attributed to their capacity to secrete numerous trophic factors that promote angiogenesis, inhibit apoptosis, and modulate the immune response. There is growing evidence that MSC‐derived extracellular vesicles (EVs) containing a cargo of lipids, proteins, metabolites, and RNAs play a key role in this paracrine mechanism. In particular, encapsulated microRNAs have been identified as important positive regulators of angiogenesis in pathological settings of insufficient blood supply to the heart, thus opening a new path for the treatment of CVD. In the present review, we discuss the current knowledge related to the proangiogenic potential of MSCs and MSC‐derived EVs as well as methods to enhance their biological activities for improved cardiac tissue repair. Increasing our understanding of mechanisms supporting angiogenesis will help optimize future approaches to CVD intervention.
Collapse
Affiliation(s)
- Timo Z Nazari-Shafti
- Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Berlin, Germany.,German Centre for Cardiovascular Research, Partner Site Berlin, Berlin, Germany.,Berlin Institute of Health Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Sebastian Neuber
- Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Berlin, Germany.,German Centre for Cardiovascular Research, Partner Site Berlin, Berlin, Germany.,Berlin Institute of Health Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Ana Garcia Duran
- Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Berlin, Germany.,Berlin Institute of Health Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin-Brandenburg School for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Zhiyi Xu
- Berlin Institute of Health Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Eleftherios Beltsios
- Berlin Institute of Health Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Martina Seifert
- Berlin Institute of Health Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Institute of Medical Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt- Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Volkmar Falk
- Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Berlin, Germany.,German Centre for Cardiovascular Research, Partner Site Berlin, Berlin, Germany.,Berlin Institute of Health Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Division of Cardiovascular Surgery, University of Zurich, Zurich, Switzerland
| | - Christof Stamm
- Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Berlin, Germany.,German Centre for Cardiovascular Research, Partner Site Berlin, Berlin, Germany.,Berlin Institute of Health Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
26
|
Alshamrani AA. Roles of microRNAs in Ovarian Cancer Tumorigenesis: Two Decades Later, What Have We Learned? Front Oncol 2020; 10:1084. [PMID: 32850313 PMCID: PMC7396563 DOI: 10.3389/fonc.2020.01084] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Accepted: 05/29/2020] [Indexed: 12/24/2022] Open
Abstract
Ovarian cancer is one of the top gynecological malignancies that cause deaths among females in the United States. At the molecular level, significant progress has been made in our understanding of ovarian cancer development and progression. MicroRNAs (miRNAs) are short, single-stranded, highly conserved non-coding RNA molecules (19–25 nucleotides) that negatively regulate target genes post-transcriptionally. Over the last two decades, mounting evidence has demonstrated the aberrant expression of miRNAs in different human malignancies, including ovarian carcinomas. Deregulated miRNAs can have profound impacts on various cancer hallmarks by repressing tumor suppressor genes. This review will discuss up-to-date knowledge of how the aberrant expression of miRNAs and their targeted genes drives ovarian cancer initiation, proliferation, survival, and resistance to chemotherapies. Understanding the mechanisms by which these miRNAs affect these hallmarks should allow the development of novel therapeutic strategies to treat these lethal malignancies.
Collapse
Affiliation(s)
- Ali A Alshamrani
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
27
|
Knarr M, Avelar RA, Sekhar SC, Kwiatkowski LJ, Dziubinski ML, McAnulty J, Skala S, Avril S, Drapkin R, DiFeo A. miR-181a initiates and perpetuates oncogenic transformation through the regulation of innate immune signaling. Nat Commun 2020; 11:3231. [PMID: 32591511 PMCID: PMC7320168 DOI: 10.1038/s41467-020-17030-w] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 06/08/2020] [Indexed: 01/17/2023] Open
Abstract
Genomic instability (GI) predisposes cells to malignant transformation, however the molecular mechanisms that allow for the propagation of cells with a high degree of genomic instability remain unclear. Here we report that miR-181a is able to transform fallopian tube secretory epithelial cells through the inhibition of RB1 and stimulator-of-interferon-genes (STING) to propagate cells with a high degree of GI. MiR-181a targeting of RB1 leads to profound nuclear defects and GI generating aberrant cytoplasmic DNA, however simultaneous miR-181a mediated inhibition of STING allows cells to bypass interferon mediated cell death. We also found that high miR-181a is associated with decreased IFNγ response and lymphocyte infiltration in patient tumors. DNA oncoviruses are the only known inhibitors of STING that allow for cellular transformation, thus, our findings are the first to identify a miRNA that can downregulate STING expression to suppress activation of intrinsic interferon signaling. This study introduces miR-181a as a putative biomarker and identifies the miR-181a-STING axis as a promising target for therapeutic exploitation.
Collapse
Affiliation(s)
- Matthew Knarr
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, 44106, USA.,Department of Obstetrics & Gynecology, The University of Michigan, Ann Arbor, MI, 48109, USA.,Department of Pathology, The University of Michigan, Ann Arbor, MI, 48109, USA
| | - Rita A Avelar
- Department of Obstetrics & Gynecology, The University of Michigan, Ann Arbor, MI, 48109, USA.,Department of Pathology, The University of Michigan, Ann Arbor, MI, 48109, USA.,The Rogel Cancer Center, The University of Michigan, Ann Arbor, MI, 48109, USA
| | - Sreeja C Sekhar
- Department of Obstetrics & Gynecology, The University of Michigan, Ann Arbor, MI, 48109, USA.,Department of Pathology, The University of Michigan, Ann Arbor, MI, 48109, USA.,The Rogel Cancer Center, The University of Michigan, Ann Arbor, MI, 48109, USA
| | - Lily J Kwiatkowski
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Michele L Dziubinski
- Department of Obstetrics & Gynecology, The University of Michigan, Ann Arbor, MI, 48109, USA.,Department of Pathology, The University of Michigan, Ann Arbor, MI, 48109, USA.,The Rogel Cancer Center, The University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jessica McAnulty
- Department of Obstetrics & Gynecology, The University of Michigan, Ann Arbor, MI, 48109, USA.,Department of Pathology, The University of Michigan, Ann Arbor, MI, 48109, USA.,The Rogel Cancer Center, The University of Michigan, Ann Arbor, MI, 48109, USA
| | - Stephanie Skala
- Department of Pathology, The University of Michigan, Ann Arbor, MI, 48109, USA
| | - Stefanie Avril
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, 44106, USA.,Department of Pathology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Ronny Drapkin
- Penn Ovarian Cancer Research Center, Department of Obstetrics and Gynecology, Perelman School of Medicine, University of Pennsylvania, Pennsylvania, PA, USA
| | - Analisa DiFeo
- Department of Obstetrics & Gynecology, The University of Michigan, Ann Arbor, MI, 48109, USA. .,Department of Pathology, The University of Michigan, Ann Arbor, MI, 48109, USA. .,The Rogel Cancer Center, The University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
28
|
Akyay OZ, Gov E, Kenar H, Arga KY, Selek A, Tarkun İ, Canturk Z, Cetinarslan B, Gurbuz Y, Sahin B. Mapping the Molecular Basis and Markers of Papillary Thyroid Carcinoma Progression and Metastasis Using Global Transcriptome and microRNA Profiling. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2020; 24:148-159. [PMID: 32073999 DOI: 10.1089/omi.2019.0188] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Papillary thyroid carcinoma (PTC) is the most common type of thyroid cancer (TC). In a subgroup of patients with PTC, the disease progresses to an invasive stage or in some cases to distant organ metastasis. At present, there is an unmet clinical and diagnostic need for early identification of patients with PTC who are at risk of disease progression or metastasis. In this study, we report several molecular leads and potential biomarker candidates of PTC metastasis for further translational research. The study design was based on comparisons of PTC in three different groups using cross-sectional sampling: Group 1, PTC localized to the thyroid (n = 20); Group 2, PTC with extrathyroidal progression (n = 22); and Group 3, PTC with distant organ metastasis (n = 20). Global transcriptome and microRNAs (miRNA) analyses were conducted using an initial screening set comprising nine formalin-fixed paraffin-embedded PTC samples obtained from three independent patients per study group. The findings were subsequently validated by quantitative real-time polymerase chain reaction (qRT-PCR) using the abovementioned independent patient sample set (n = 62). Comparative analyses of differentially expressed miRNAs showed that miR-193-3p, miR-182-5p, and miR-3607-3p were novel miRNAs associated with PTC metastasis. These potential miRNA biomarkers were associated with TC metastasis and miRNA-target gene associations, which may provide important clinicopathological information on metastasis. Our findings provide new molecular leads for further translational biomarker research, which could facilitate the identification of patients at risk of PTC disease progression or metastasis.
Collapse
Affiliation(s)
- Ozlem Zeynep Akyay
- Department of Endocrinology and Metabolism, Sanliurfa Mehmet Akif İnan Education and Research Hospital, Health Sciences University, Sanliurfa, Turkey
| | - Esra Gov
- Department of Bioengineering, Faculty of Engineering, Adana Alparslan Turkes Science and Technology University, Adana, Turkey
| | - Halime Kenar
- Experimental and Clinical Research Center, Diabetes and Obesity Research Laboratory, Kocaeli University, Kocaeli, Turkey
| | - Kazım Yalcın Arga
- Department of Bioengineering, Faculty of Engineering, Marmara University, Istanbul, Turkey
| | - Alev Selek
- Department of Endocrinology and Metabolism, School of Medicine, Kocaeli University, Kocaeli, Turkey
| | - İlhan Tarkun
- Department of Endocrinology and Metabolism, Anadolu Medical Center, İstanbul, Turkey
| | - Zeynep Canturk
- Department of Endocrinology and Metabolism, School of Medicine, Kocaeli University, Kocaeli, Turkey
| | - Berrin Cetinarslan
- Department of Endocrinology and Metabolism, School of Medicine, Kocaeli University, Kocaeli, Turkey
| | - Yesim Gurbuz
- Department of Pathology, School of Medicine, Kocaeli University, Kocaeli, Turkey
| | - Busra Sahin
- Department of Pathology, School of Medicine, Kocaeli University, Kocaeli, Turkey
| |
Collapse
|
29
|
Mansouri M, Peymani M, Mohamadynejad P. A genetic variant in the flanking region of miR-182 could decrease the susceptibility to the breast cancer risk in the iranian population. NUCLEOSIDES NUCLEOTIDES & NUCLEIC ACIDS 2020; 39:806-817. [PMID: 31994428 DOI: 10.1080/15257770.2019.1704778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Breast cancer is one of the most malignant tumors in the world. It is, in fact, the second leading cause of cancer death in women. Recent research has identified the role of miR-182 in this disease as an oncogene agent. In this study, the association of rs4541843 in the flanking region of the miR-182 sequence with the susceptibility to breast cancer risk has been studied in the Iranian population. By using the PCR_RFLP, the genotype rs4541843 was determined in 161 patients and 164 control subjects. The genotypes of the individuals were analyzed statistically to find the association between rs4541843 and the breast cancer incidence and its pathological characteristics. The results revealed that due to the dominance of the G allele, the frequency of GG + AG genotypes, as compared with AA, had a significant correlation with the incidence of this disease in controls and cases (P = 0.022; OR = 0.523). Moreover, the genotypes AG and AA could significantly decrease the susceptibility to the breast cancer risk; also in the presence of the A allele (OR, 0.565; P = 0.015), the incidence of the disease could be decreased. Our results indicated that this SNP was associated with the breast cancer risk of the Iranian population. We suppose that rs4541843 may influence the processing of the mature miRNA by affecting the cleavage of Drosha. Therefore, this SNP can be considered as a candidate genetic marker for the susceptibility to breast cancer in the Iranian women.
Collapse
Affiliation(s)
- Maryam Mansouri
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Maryam Peymani
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Parisa Mohamadynejad
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| |
Collapse
|
30
|
Szczepaniak A, Fichna J, Zielińska M. Opioids in Cancer Development, Progression and Metastasis: Focus on Colorectal Cancer. Curr Treat Options Oncol 2020; 21:6. [PMID: 31970561 PMCID: PMC6976545 DOI: 10.1007/s11864-019-0699-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
OPINION STATEMENT So far, opioids have been successfully used to reduce cancer pain in patients in order to improve their quality of life. However, the use of opioids leads to numerous side effects such as constipation, drowsiness, nausea, itching, increased sweating and hormonal changes. In this review, we described the action of opioids in several molecular pathways significant for maintenance of the intestinal homeostasis including the impact on the intestinal epithelium integrity, changes in microbiome composition, modulation of the immune system or induction of apoptosis and inhibition of angiogenesis. We summed up the role of individual opioids in the processes involved in the growth and development of cancer and elucidated if targeting opioid receptors may constitute novel therapeutic option in colon cancer.
Collapse
Affiliation(s)
- Adrian Szczepaniak
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - Jakub Fichna
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - Marta Zielińska
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland.
| |
Collapse
|
31
|
Li Y, Li L. Prognostic values and prospective pathway signaling of MicroRNA-182 in ovarian cancer: a study based on gene expression omnibus (GEO) and bioinformatics analysis. J Ovarian Res 2019; 12:106. [PMID: 31703725 PMCID: PMC6839211 DOI: 10.1186/s13048-019-0580-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 10/10/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Ovarian carcinoma (OC) is a common cause of death among women with gynecological cancer. MicroRNAs (miRNAs) are believed to have vital roles in tumorigenesis of OC. Although miRNAs are broadly recognized in OC, the role of has-miR-182-5p (miR-182) in OC is still not fully elucidated. METHODS We evaluated the significance of miR-182 expression in OC by using analysis of a public dataset from the Gene Expression Omnibus (GEO) database and a literature review. Furthermore, we downloaded three mRNA datasets of OC and normal ovarian tissues (NOTs), GSE14407, GSE18520 and GSE36668, from GEO to identify differentially expressed genes (DEGs). Then the targeted genes of hsa-miR-182-5p (TG_miRNA-182-5p) were predicted using miRWALK3.0. Subsequently, we analyzed the gene overlaps integrated between DEGs in OC and predicted target genes of miR-182 by Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis. STRING and Cytoscape were used to construct a protein-protein interaction (PPI) network and the prognostic effects of the hub genes were analyzed. RESULTS A common pattern of up-regulation for miR-182 in OC was found in our review of the literature. A total of 268 DEGs, both OC-related and miR-182-related, were identified, of which 133 genes were discovered from the PPI network. A number of DEGs were enriched in extracellular matrix organization, pathways in cancer, focal adhesion, and ECM-receptor interaction. Two hub genes, MCM3 and GINS2, were significantly associated with worse overall survival of patients with OC. Furthermore, we identified covert miR-182-related genes that might participate in OC by network analysis, such as DCN, AKT3, and TIMP2. The expressions of these genes were all down-regulated and negatively correlated with miR-182 in OC. CONCLUSIONS Our study suggests that miR-182 is essential for the biological progression of OC.
Collapse
Affiliation(s)
- Yaowei Li
- Department of Gynecologic Oncology, Affiliated Tumor Hospital of Guangxi Medical University, Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Ministry of Education, Nanning, Guangxi, China
- Department of Gynecology and obstetrics, Shangyu People's Hospital, Shangyu, Zhejiang, China
| | - Li Li
- Department of Gynecologic Oncology, Affiliated Tumor Hospital of Guangxi Medical University, Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Ministry of Education, Nanning, Guangxi, China.
| |
Collapse
|
32
|
Záveský L, Jandáková E, Weinberger V, Minář L, Hanzíková V, Dušková D, Drábková LZ, Hořínek A. Ovarian Cancer: Differentially Expressed microRNAs in Tumor Tissue and Cell-Free Ascitic Fluid as Potential Novel Biomarkers. Cancer Invest 2019; 37:440-452. [PMID: 31530033 DOI: 10.1080/07357907.2019.1663208] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Ovarian cancer is the deadliest gynecologic cancer. The large-scale microRNA (miRNA) expression profiling and individual miRNA validation was performed to find potential novel biomarkers for ovarian cancer. The most consistent overexpression of miRs-200b-3p, 135 b-5p and 182-5p was found in both ascitic fluid and tumors and suggests their potential as oncogenes. miR-451a was consistently underexpressed so may be a tumor suppressor. Results were inconsistent for miR-204-5p, which was overexpressed in ascitic fluid but underexpressed in tumor tissue. miR-203a-3p was generally overexpressed but this failed to be proved in independent sample set in tissue validation.
Collapse
Affiliation(s)
- Luděk Záveský
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University and General University Hospital in Prague , Prague 2 , Czech Republic
| | - Eva Jandáková
- Department of Pathology, Masaryk University and University Hospital Brno , Brno , Czech Republic
| | - Vít Weinberger
- Department of Obstetrics and Gynecology, Masaryk University and University Hospital Brno , Brno , Czech Republic
| | - Luboš Minář
- Department of Obstetrics and Gynecology, Masaryk University and University Hospital Brno , Brno , Czech Republic
| | - Veronika Hanzíková
- Faculty Transfusion Center, General University Hospital in Prague , Prague 2 , Czech Republic
| | - Daniela Dušková
- Faculty Transfusion Center, General University Hospital in Prague , Prague 2 , Czech Republic
| | | | - Aleš Hořínek
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University and General University Hospital in Prague , Prague 2 , Czech Republic.,3rd Department of Medicine, Department of Endocrinology and Metabolism, First Faculty of Medicine, Charles University and General University Hospital in Prague , Prague 2 , Czech Republic
| |
Collapse
|
33
|
Jiang Y, Zhou Z, Fei R, Zhou X, Wang J, Tao Y, Li J, Chen T. Role of miR-182-5p overexpression in trichloroethylene-induced abnormal cell cycle functions in human HepG2 cells. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2019; 82:920-927. [PMID: 31524101 DOI: 10.1080/15287394.2019.1666550] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Trichloroethylene (TCE), a widely used industrial solvent, occurs frequently in the global environment. TCE was found to induce hepatocarcinogenesis in mice and one of the underlying mechanisms was reported to involve miR-182-5p overexpression. Subsequently, miR-182-5p overexpression was shown to contribute to chemical-induced enhanced cell proliferation in mouse liver cells by targeting the gene Cited2. The aim of this study was to compare our findings in mice with those in a human hepatoma cell line HepG2. Data demonstrated that TCE at 0.1mM exerted no marked effect on human hepatoma cell line HepG2 cell migration, cell cycle, apoptosis, and DNA damage, but significantly stimulated cell proliferation rate and increased mRNA expression levels of proliferating cell nuclear antigen (PCNA), a cell proliferation biomarker. In addition, TCE enhanced miR-182-5p expression levels but lowered Cited2 mRNA expression. In summary, data showed that similar to mouse liver cells, TCE exposure also upregulated cells miR-182-5p expression and inhibited Cited2 expression in human hepatoma cell line HepG2. Our results suggest that the TCE-mediated alterations in the observed cellular functions involve interaction with miR-182-5p. It is of interest that utilization of liver cancer tissues from the Cancer Genome Atlas (TCGA) database also demonstrated that upregulated miR-182-5p expression and reduced Cited2 mRNA expression was detected suggesting that TCE-induced hepatocarcinogenesis involved processes similar to those in humans.
Collapse
Affiliation(s)
- Yan Jiang
- Medical College, Soochow University , Suzhou , China
| | - Zijie Zhou
- Medical College, Soochow University , Suzhou , China
| | - Ren Fei
- Medical College, Soochow University , Suzhou , China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University , Suzhou , China
| | - Xuan Zhou
- Medical College, Soochow University , Suzhou , China
| | - Jin Wang
- Medical College, Soochow University , Suzhou , China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University , Suzhou , China
| | - Yizhou Tao
- Medical College, Soochow University , Suzhou , China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University , Suzhou , China
| | - Jianxiang Li
- Medical College, Soochow University , Suzhou , China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University , Suzhou , China
| | - Tao Chen
- Medical College, Soochow University , Suzhou , China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University , Suzhou , China
| |
Collapse
|
34
|
Khan S, Ayub H, Khan T, Wahid F. MicroRNA biogenesis, gene silencing mechanisms and role in breast, ovarian and prostate cancer. Biochimie 2019; 167:12-24. [PMID: 31493469 DOI: 10.1016/j.biochi.2019.09.001] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 09/01/2019] [Indexed: 12/21/2022]
Abstract
Micro-ribonucleic acids (miRNAs) are important class of short regulatory RNA molecules involved in regulation of several essential biological processes. In addition to Dicer and Drosha, over the past few years several other gene products are discovered that regulates miRNA biogenesis pathways. Similarly, various models of molecular mechanisms underlying miRNA mediated gene silencing have been uncovered through which miRNA contribute in diverse physiological and pathological processes. Dysregulated miRNA expression has been reported in many cancers manifesting tumor suppressive or oncogenic role. In this review, critical overview of recent findings in miRNA biogenesis, silencing mechanisms and specifically the role of miRNA in breast, ovarian and prostate cancer will be described. Recent advancements in miRNA research summarized in this review will enhance the molecular understanding of miRNA biogenesis and mechanism of action. Also, role of miRNAs in pathogenesis of breast, ovarian and prostate cancer will provide the insights for the use of miRNAs as biomarker or therapeutic agents for the cancers.
Collapse
Affiliation(s)
- Sanna Khan
- Department of Biotechnology, COMSATS University Islamabad, Abbottabad Campus, 22060, Pakistan
| | - Humaira Ayub
- Department of Biotechnology, COMSATS University Islamabad, Abbottabad Campus, 22060, Pakistan
| | - Taous Khan
- Department of Pharmacy, COMSATS University Islamabad, Abbottabad Campus, 22060, Pakistan
| | - Fazli Wahid
- Department of Biotechnology, COMSATS University Islamabad, Abbottabad Campus, 22060, Pakistan.
| |
Collapse
|
35
|
Kim G, Kim J, Han SY, Hwang IG, Kim HS, Min H. The effects of BRCA1 expression on the chemosensitivity of gastric cancer cells to platinum agents. Oncol Lett 2019; 17:5023-5029. [PMID: 31186713 PMCID: PMC6507359 DOI: 10.3892/ol.2019.10169] [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: 08/27/2018] [Accepted: 02/20/2019] [Indexed: 01/30/2023] Open
Abstract
Breast cancer type 1 susceptibility protein (BRCA1) is a tumor suppressor gene that encodes a nuclear phosphoprotein, which is involved in homologous recombination to repair DNA double strand breaks and maintain genome stability. When BRCA1 is mutated or altered, DNA damage may not be effectively repaired, which leads to DNA replication errors and cancer growth. Accordingly, people carrying a mutation in the BRCA1 gene possess an increased risk of several types of cancer, including breast and ovarian cancer. Previous clinical studies have reported an association between BRCA1 expression level and the incidence of gastric cancer; however, to the best of our knowledge, an in vitro study has not been performed to support these clinical observations. Therefore, the present study evaluated BRCA1 expression levels in gastric cancer cell lines. In addition, the IC50 values of cisplatin and oxaliplatin in each cell line were determined to investigate a potential correlation between BRCA1 expression level and chemosensitivity to platinum agents. The present results revealed that the BRCA1 expression level in gastric cancer is variable and associated with the treatment response to platinum-based chemotherapy. This suggests that BRCA1 may serve as a therapeutic marker for platinum-based chemotherapy in gastric cancer.
Collapse
Affiliation(s)
- Geon Kim
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Jisu Kim
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Su-Young Han
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| | - In Gyu Hwang
- Department of Internal Medicine, College of Medicine, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Hee Sung Kim
- Department of Pathology, College of Medicine, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Hyeyoung Min
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| |
Collapse
|
36
|
Abstract
MicroRNAs are critical post-transcriptional regulators of a majority of genes, of which the FOXO family of transcription factors is no exception. Here, we describe generalizable methods, including 3' UTR reporter assays and western blotting after microRNA manipulation, to test if a candidate miRNA (miR-182) directly targets a candidate (FOXO3) gene product. We also provide guidance on candidate miRNA selection and unbiased miRNA-target identification methods.
Collapse
Affiliation(s)
- Doug Hanniford
- Department of Pathology, New York University Langone Health, New York, NY, USA
- Interdisciplinary Melanoma Cooperative Group, New York University Langone Health, New York, NY, USA
| | - Eva Hernando
- Department of Pathology, New York University Langone Health, New York, NY, USA.
- Interdisciplinary Melanoma Cooperative Group, New York University Langone Health, New York, NY, USA.
| |
Collapse
|
37
|
Chartron E, Theillet C, Guiu S, Jacot W. Targeting homologous repair deficiency in breast and ovarian cancers: Biological pathways, preclinical and clinical data. Crit Rev Oncol Hematol 2018; 133:58-73. [PMID: 30661659 DOI: 10.1016/j.critrevonc.2018.10.012] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 09/25/2018] [Accepted: 10/30/2018] [Indexed: 12/16/2022] Open
Abstract
Mutation or epigenetic silencing of homologous recombination (HR) repair genes is characteristic of a growing proportion of triple-negative breast cancers (TNBCs) and high-grade serous ovarian carcinomas. Defects in HR lead to genome instability, allowing cells to acquire the multiple genetic alterations essential for cancer development. However, this deficiency can also be exploited by using DNA damaging agents or by targeting compensatory repair pathways. A noteworthy example is treatment of TNBC and epithelial ovarian cancer harboring BRCA1/2 germline mutations using platinum salts and/or PARP inhibitors. Dramatic responses to PARP inhibitors may support a wider use in the HR-deficient population beyond those with mutated germline BRCA1 and 2. In this review, we discuss HR deficiency hallmarks as predictive biomarkers for platinum salt and PARP inhibitor sensitivity for selecting patients affected by TNBC or epithelial ovarian cancer who could benefit from these therapeutic options.
Collapse
Affiliation(s)
- Elodie Chartron
- Department of medical oncology, Montpellier Academic Hospital, Montpellier, France
| | - Charles Theillet
- IRCM, INSERM, Université de Montpellier, ICM, Montpellier, France
| | - Séverine Guiu
- Department of Medical Oncology, Institut du Cancer de Montpellier, Montpellier, France
| | - William Jacot
- IRCM, INSERM, Université de Montpellier, ICM, Montpellier, France; Department of Medical Oncology, Institut du Cancer de Montpellier, Montpellier, France.
| |
Collapse
|
38
|
Huang YX, Nie XG, Li GD, Fan DS, Song LL, Zhang XL. Downregulation of microRNA‑182 inhibits cell viability, invasion and angiogenesis in retinoblastoma through inhibition of the PI3K/AKT pathway and CADM2 upregulation. Int J Oncol 2018; 53:2615-2626. [PMID: 30320366 DOI: 10.3892/ijo.2018.4587] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 07/02/2018] [Indexed: 11/06/2022] Open
Abstract
Retinoblastoma (RB) is a well‑vascularized tumor dependent on angiogenesis. The present study aimed to explore whether microRNA (miR)‑182 regulates cell viability, invasion and angiogenesis in RB via the phosphatidylinositol‑3‑OH kinase (PI3K)/protein kinase B (AKT) signaling pathway and by targeting cell adhesion molecule 2 (CADM2). The expression levels of miR‑182 and CADM2 were initially detected in RB tissues from patients with RB who underwent ophthalmectomy, and normal retinal tissues collected from other trauma patients who underwent eye enucleation. To determine whether CADM2 was targeted by miR‑182, a dual luciferase reporter assay was conducted. Subsequently, Y79 and WERI‑Rb‑1 RB cells were transfected with a miR‑182 mimic or miR‑182 inhibitor, or small interfering RNA against CADM2, in order to investigate the effects of miR‑182 on viability and invasion, which were detected using MTT and Transwell assays, respectively. In addition, to determine whether the regulatory mechanism underlying the effects of miR‑182 was associated with the PI3K/AKT signaling pathway, the expression levels of associated genes were detected by reverse transcription‑quantitative polymerase chain reaction and western blot analysis. A xenograft tumor model in nude mice was also established, in order to evaluate the effects of miR‑182 on tumor growth and angiogenesis. The results indicated that miR‑182 expression was increased and CADM2 expression was reduced in RB tissues; CADM2 was confirmed to be targeted and negatively regulated by miR‑182. When the expression of miR‑182 was downregulated, cell viability, invasion, tumor volume and angiogenesis were significantly decreased. Furthermore, the expression levels of PI3K/AKT signaling pathway‑associated genes were increased in response to miR‑182 overexpression or CADM2 silencing. Taken together, these results suggested that inhibition of miR‑182 may suppress cell viability, invasion and angiogenesis in RB through inactivation of the PI3K/AKT pathway and CADM2 upregulation. This mechanism may reveal a novel potential therapeutic target.
Collapse
Affiliation(s)
- Yan-Xia Huang
- Department of Ophthalmology, Luoyang Central Hospital, Luoyang, Henan 471009, P.R. China
| | - Xin-Gang Nie
- Department of Ophthalmology, Luoyang Central Hospital, Luoyang, Henan 471009, P.R. China
| | - Guang-Da Li
- Department of Ophthalmology, Linyi People's Hospital, Linyi, Shandong 276003, P.R. China
| | - Dong-Sheng Fan
- Department of Ophthalmology, Luoyang Central Hospital, Luoyang, Henan 471009, P.R. China
| | - Li-Li Song
- Department of Ophthalmology, Luoyang Central Hospital, Luoyang, Henan 471009, P.R. China
| | - Xin-Lin Zhang
- Department of Ophthalmology, Luoyang Central Hospital, Luoyang, Henan 471009, P.R. China
| |
Collapse
|
39
|
Oxidative stress-induced miRNAs modulate AKT signaling and promote cellular senescence in uterine leiomyoma. J Mol Med (Berl) 2018; 96:1095-1106. [PMID: 30097674 DOI: 10.1007/s00109-018-1682-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 07/12/2018] [Accepted: 08/06/2018] [Indexed: 02/08/2023]
Abstract
Uterine leiomyomas (ULM) grow under high oxidative stress due to a hypoxic microenvironment and defects in redox metabolism. AKT is one major pathway activated by reactive oxygen species (ROS) that maintains ULM growth and survival. We previously reported that AKT inactivated by AKT inhibitors can significantly induce cellular senescence in ULM cells. Since some miRNAs are induced by AKT inhibitors in an ROS-dependent manner, we proposed that these miRNAs may modulate AKT function and cellular senescence in ULM. We therefore established ex vivo models of a three-dimensional ULM spheroid culture system to study the role of miRNAs in cellular senescence. Four miRNAs, miR-29b, miR-181a, miR-182, and miR-200c, were found to induce cellular senescence in primary ULM and myometrium spheroid cultures when stably overexpressed. miR-181a and miR-182 were found to repress AKT3 and CCND2, respectively. Correspondingly, RNAi of AKT3 or CCND2 also induced cellular senescence and G0/G1 arrest. Thus, miR-181a and miR-182 may drive cellular senescence in ULM by repressing AKT3 and CCND2 activity, respectively. We further demonstrated that senescent ULM cells can be effectively removed by BH3 mimetic ABT263, which provides a new therapeutic venue for the treatment of ULM. Our findings suggest that miRNAs are potent modulators in regulating the ROS-AKT-cell cycle axis in uterine leiomyoma. KEY MESSAGES A subset of oxidative stress-induced miRNAs is involved in AKT signaling in uterine leiomyoma. Overexpression of miR-181a and miR-182 resulted in cellular senescence in leiomyoma through repression of AKT3 and CCND2, respectively. Silencing of AKT3 and CCND2 drives leiomyoma cell into senescence and cycle arrest. Application of our newly developed 3D leiomyoma spheroids can provide a quick and reliable ex vivo model for cytopathologic and functional analysis. BH3 mimetics can effectively reduce the viability of miRNA-mediated senescent cells in leiomyoma.
Collapse
|
40
|
Taking the Tube: From Normal Fallopian Tube Epithelium to Ovarian High-grade Serous Carcinoma. Clin Obstet Gynecol 2018; 60:697-710. [PMID: 29045296 DOI: 10.1097/grf.0000000000000313] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Detailed pathologic studies over the past decade suggest a distal fallopian tube origin for the majority of "ovarian" high-grade serous carcinomas (HGSC). This review will summarize molecular alterations observed in tubal precursors for HGSC, namely p53 signatures and serous tubal intraepithelial carcinomas, and in nonmalignant fallopian tube epithelial cells obtained from women at increased genetic risk for HGSC. Recent experiments investigating the impact of follicular fluid exposure and retrograde menstruation on tumor development in the fallopian tube will also be discussed. These data will be reconciled with traditional ovarian cancer risk factors related to reproductive history.
Collapse
|
41
|
Javadi H, Lotfi AS, Hosseinkhani S, Mehrani H, Amani J, Soheili ZS, Hojati Z, Kamali M. The combinational effect of E6/E7 siRNA and anti-miR-182 on apoptosis induction in HPV16-positive cervical cells. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2018; 46:727-736. [PMID: 29873516 DOI: 10.1080/21691401.2018.1468770] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
In the present research, we assumed that reducing the amounts of E6 and E7 oncoproteins by a specific siRNA sequence and recovering p53 and RB proteins, along with the recovery of the FOXO1 protein by applying anti-miR-182, would increase apoptosis and reduce proliferation rate in cancer cells. The HPV16-positive CaSki cervical cancer cell line was used. 48 hours after transfection of siRNA for targeting E6 and E7 oncoproteins and anti-miR-182, expression of its cellular targets p53, p21 and FOXO1 was assessed by real-time PCR, western blot analysis and immunocytofluorescence staining. In all treatments, apoptosis rate and viability were evaluated using Annexin-V-FITC apoptosis detection kits and MTT assays, respectively. Among the designed siRNAs, E6-1 and E7-2 proved the most effective in reducing E6 and E7 expressions by increasing the apoptotic rates to 12.4% and 16%, respectively, after 48 hours. Also, using anti-miR-182 increased apoptotic rate to 12.7% 48 hours after transfection of cervical cancer cells. The combinational use of either E6-1 or E7-2 siRNAs with anti-miR-182 resulted in a rise in apoptosis to 19.3% and 26%, respectively, higher than those obtained from the individual application of either without anti-miR-182. The simultaneous use of siRNA E6-1 and siRNA E7-2 with cisplatin increased sensitivity to cisplatin and reduced the viability of the cancer cells as compared to the use of cisplatin alone. The simultaneous use of cisplatin and anti-miR-182 had no considerable effect on viability or apoptosis rate compared to cisplatin alone.
Collapse
Affiliation(s)
- Hamidreza Javadi
- a Nanobiotechnology Research Center , Baqiyatallah University of Medical Sciences , Tehran , Iran.,b Department of Molecular Medicine , Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology , Tehran , Iran
| | - Abbas Sahebghadam Lotfi
- c Department of Clinical Biochemistry, Faculty of Medicine , Tarbiat Modares University , Tehran , Iran
| | - Saman Hosseinkhani
- d Department of Biochemistry, Faculty of Basic Sciences , Tarbiat Modares University , Tehran , Iran
| | - Hossein Mehrani
- e Department of Biochemistry, Faculty of Science , Islamic Azad University Branch of Neyshabur , Neyshabur , Iran
| | - Jafar Amani
- f Applied Microbiology Research Center, System Biology and Poisonings Institute , Baqiyatallah University of Medical Sciences , Tehran , Iran
| | - Zahra Soheila Soheili
- b Department of Molecular Medicine , Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology , Tehran , Iran
| | - Zahra Hojati
- b Department of Molecular Medicine , Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology , Tehran , Iran
| | - Mehdi Kamali
- a Nanobiotechnology Research Center , Baqiyatallah University of Medical Sciences , Tehran , Iran
| |
Collapse
|
42
|
Hou D, Liu Z, Xu X, Liu Q, Zhang X, Kong B, Wei JJ, Gong Y, Shao C. Increased oxidative stress mediates the antitumor effect of PARP inhibition in ovarian cancer. Redox Biol 2018; 17:99-111. [PMID: 29684820 PMCID: PMC6006521 DOI: 10.1016/j.redox.2018.03.016] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 03/23/2018] [Accepted: 03/26/2018] [Indexed: 01/09/2023] Open
Abstract
PARP inhibitors have been widely tested in clinical trials, especially for the treatment of breast cancer and ovarian cancer, and were shown to be highly successful. Because PARP primarily functions in sensing and repairing DNA strand breaks, the therapeutic effect of PARP inhibition is generally believed to be attributed to impaired DNA repair. We here report that oxidative stress is also increased by PARP inhibition and mediates the antitumor effect. We showed that PARP1 is highly expressed in specimens of high grade serous ovarian carcinoma and its activity is required for unperturbed proliferation of ovarian cancer cells. Inhibition or depletion of PARP leads to not only an increase in DNA damage, but also an elevation in the levels of reactive oxygen species (ROS). Importantly, antioxidant N-acetylcysteine (NAC) significantly attenuated the induction of DNA damage and the perturbation of proliferation by PARP inhibition or depletion. We further showed that NADPH oxidases 1 and 4 were significantly upregulated by PARP inhibition and were partially responsible for the induction of oxidative stress. Depletion of NOX1 and NOX4 partially rescued the growth inhibition of PARP1-deficient tumor xenografts. Our findings suggest that in addition to compromising the repair of DNA damage, PARP inhibition or depletion may exert extra antitumor effect by elevating oxidative stress in ovarian cancer cells. PARP1 is overexpressed in ovarian cancer. PARP inhibition increases oxidative stress and oxidative DNA damage. PARP inhibition increases ROS by upregulating NOX1 and NOX4. Oxidative stress mediates the antitumor effect of PARP inhibition.
Collapse
Affiliation(s)
- Dong Hou
- Key Laboratory of Experimental Teratology, Ministry of Education/Department of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Zhaojian Liu
- Department of Cell Biology, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Xiuhua Xu
- Key Laboratory of Experimental Teratology, Ministry of Education/Department of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Qiao Liu
- Key Laboratory of Experimental Teratology, Ministry of Education/Department of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Xiyu Zhang
- Key Laboratory of Experimental Teratology, Ministry of Education/Department of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Beihua Kong
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Jian-Jun Wei
- Department of Pathology, Northwestern University School of Medicine, Chicago, IL, USA
| | - Yaoqin Gong
- Key Laboratory of Experimental Teratology, Ministry of Education/Department of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Changshun Shao
- Key Laboratory of Experimental Teratology, Ministry of Education/Department of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, Shandong 250012, China; The First Affiliated Hospital of Soochow University and State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University, 199 Ren Ai Road, Suzhou, Jiangsu 215123, China.
| |
Collapse
|
43
|
Lin LT, Liu SY, Leu JD, Chang CY, Chiou SH, Lee TC, Lee YJ. Arsenic trioxide-mediated suppression of miR-182-5p is associated with potent anti-oxidant effects through up-regulation of SESN2. Oncotarget 2018; 9:16028-16042. [PMID: 29662624 PMCID: PMC5882315 DOI: 10.18632/oncotarget.24678] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 02/24/2018] [Indexed: 01/15/2023] Open
Abstract
Arsenic trioxide (ATO) is a traditional Chinese medicine that can induce oxidative stress for treatment of cancer cells. However, ATO may generate anti-oxidative responses to compromise the cytotoxic effect, but the underlying mechanisms remain unclear. Here we found that ATO could inhibit miR-182-5p expression in patient-derived primary S1 glioblastoma (GBM) cells accompanied by up-regulation of Sestrin-2 (SESN2) mRNA, a known anti-oxidant molecule. This phenomenon was also detected in a U87MG glioma cell line, human lung adenocarcinoma H1299 cell line and A549 cell line. Pretreatment with a free radical scavenger N-acetylcysteine (NAC) reduced the oxidative stress induced by ATO. Concomitantly, ATO mediated suppression of miR-182-5p and enhancement of SESN2 expression were also compromised. The MTT assay further showed that ATO induced cytotoxicity was enhanced by transfection of miR-182-5p mimics. Overexpression of miR-182-5p mimics significantly suppressed the expression of SENS2 and a firefly luciferase reporter gene fused to 3’- untranslated region (UTR) of SESN2 mRNA. Use of ribonucleoprotein immunoprecipitation (RNP-IP), ATO mediated suppression of miR-182-5p led to the stabilization of SESN2 mRNA as a result of Argonaute-2 (AGO2) dependent gene silencing. Furthermore, high expression of miR-182-5p and low expression of SESN2 mRNA tend to be associated with longer survival of glioma or lung cancer patients using public available gene expression datasets and online tools for prediction of clinical outcomes. Taken together, current data suggest that the miR-182-5p/SENS2 pathway is involved in ATO induced anti-oxidant responses, which may be important for the design of novel strategy for cancer treatment.
Collapse
Affiliation(s)
- Liang-Ting Lin
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei, Taiwan.,Current address: Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong
| | - Shin-Yi Liu
- Department of Radiation Oncology, MacKay Memorial Hospital, Taipei, Taiwan
| | - Jyh-Der Leu
- Division of Radiation Oncology, Taipei City Hospital Ren Ai Branch, Taipei, Taiwan.,Institute of Neuroscience, National Chengchi University, Taipei, Taiwan
| | - Chun-Yuan Chang
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Shih-Hwa Chiou
- Department of Medical Research and Education, Taipei Veterans General Hospital, Taipei, Taiwan.,Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Institute of Pharmacology, National Yang-Ming University, Taipei, Taiwan
| | - Te-Chang Lee
- Institute of Pharmacology, National Yang-Ming University, Taipei, Taiwan.,Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yi-Jang Lee
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei, Taiwan.,Biophotonics and Molecular Imaging Research Center (BMIRC), National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
44
|
Segura MF, Jubierre L, Li S, Soriano A, Koetz L, Gaziel-Sovran A, Masanas M, Kleffman K, Dankert JF, Walsh MJ, Hernando E. Krüppel-like factor 4 (KLF4) regulates the miR-183~96~182 cluster under physiologic and pathologic conditions. Oncotarget 2018; 8:26298-26311. [PMID: 28412746 PMCID: PMC5432258 DOI: 10.18632/oncotarget.15459] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 02/06/2017] [Indexed: 12/11/2022] Open
Abstract
MicroRNAs (miRNAs) are a class of endogenous non-coding small RNAs that post-transcriptionally control the translation and stability of target mRNAs in a sequence-dependent manner. MiRNAs are essential for key cellular processes including proliferation, differentiation, cell death and metabolism, among others. Consequently, alterations of miRNA expression contribute to developmental defects and a myriad of diseases.The expression of miRNAs can be altered by several mechanisms including gene copy number alterations, aberrant DNA methylation, defects of the miRNA processing machinery or unscheduled expression of transcription factors. In this work, we sought to analyze the regulation of the miR-182 cluster, located at the 7q32 locus, which encodes three different miRNAs that are abundantly expressed in human embryonic stem cells and de-regulated in cancer. We have found that the Krüppel-like factor 4 (KLF4) directly regulates miR-182 cluster expression in human embryonic stem cells (hESCs) and in melanoma tumors, in which the miR-182 cluster is highly expressed and has a pro-metastatic role. Furthermore, higher KLF4 expression was found to be associated with metastatic progression and poor patient outcome. Loss of function experiments revealed that KLF4 is required for melanoma cell maintenance. These findings provide new insights into the regulation of the miR-182 cluster expression and new opportunities for therapeutic intervention in tumors in which the KLF4-miR-182 cluster axis is deregulated.
Collapse
Affiliation(s)
- Miguel F Segura
- Department of Pathology, New York University School of Medicine, New York, NY, USA.,Interdisciplinary Melanoma Cooperative Group, New York University Perlmutter Cancer Institute, NYU School of Medicine, New York, NY, USA.,Laboratory of Translational Research in Child and Adolescent Cancer, Vall d'Hebron Research Institute (VHIR)-UAB, Barcelona, Spain
| | - Luz Jubierre
- Laboratory of Translational Research in Child and Adolescent Cancer, Vall d'Hebron Research Institute (VHIR)-UAB, Barcelona, Spain
| | - SiDe Li
- Departments of Structural and Chemical Biology, Genetics and Genomic Sciences and Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Aroa Soriano
- Laboratory of Translational Research in Child and Adolescent Cancer, Vall d'Hebron Research Institute (VHIR)-UAB, Barcelona, Spain
| | - Lisa Koetz
- Department of Pathology, New York University School of Medicine, New York, NY, USA.,Interdisciplinary Melanoma Cooperative Group, New York University Perlmutter Cancer Institute, NYU School of Medicine, New York, NY, USA
| | - Avital Gaziel-Sovran
- Department of Pathology, New York University School of Medicine, New York, NY, USA.,Interdisciplinary Melanoma Cooperative Group, New York University Perlmutter Cancer Institute, NYU School of Medicine, New York, NY, USA
| | - Marc Masanas
- Laboratory of Translational Research in Child and Adolescent Cancer, Vall d'Hebron Research Institute (VHIR)-UAB, Barcelona, Spain
| | - Kevin Kleffman
- Department of Pathology, New York University School of Medicine, New York, NY, USA.,Interdisciplinary Melanoma Cooperative Group, New York University Perlmutter Cancer Institute, NYU School of Medicine, New York, NY, USA
| | - John F Dankert
- Department of Pathology, New York University School of Medicine, New York, NY, USA.,Interdisciplinary Melanoma Cooperative Group, New York University Perlmutter Cancer Institute, NYU School of Medicine, New York, NY, USA
| | - Martin J Walsh
- Departments of Structural and Chemical Biology, Genetics and Genomic Sciences and Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Eva Hernando
- Department of Pathology, New York University School of Medicine, New York, NY, USA.,Interdisciplinary Melanoma Cooperative Group, New York University Perlmutter Cancer Institute, NYU School of Medicine, New York, NY, USA
| |
Collapse
|
45
|
Tian S, Zhang M, Chen X, Liu Y, Lou G. MicroRNA-595 sensitizes ovarian cancer cells to cisplatin by targeting ABCB1. Oncotarget 2018; 7:87091-87099. [PMID: 27893429 PMCID: PMC5349973 DOI: 10.18632/oncotarget.13526] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 11/09/2016] [Indexed: 12/20/2022] Open
Abstract
Ovarian cancer is among the leading cause of cancer-related deaths in females. In this study, we demonstrated that miR-595 expression was downregulated in the ovarian cancer tissues and cell lines. miR-595 expression was lower in the lymph node metastases tissues than in the primary ovarian cancer tissues and normal tissues. Furthermore, miR-595 overexpression suppressed the ovarian cancer cell proliferation, colony formation and invasion and promoted the sensitivity of ovarian cancer cell to cisplatin. We identified ABCB1 as a direct target gene of miR-595 in the ovarian cancer cell. ABCB1 expression was upregulated in the ovarian cancer tissues and cell lines. Morevoer, the expression level of ABCB1 was inversely correlated with miR-595 in the ovarian cancer tissues. In addition, overexpression of ABCB1 decreased the miR-595-overexpressing HO8910PM and SKOV-3 cell sensitivity to cisplatin. Ectopic expression of ABCB1 promoted the miR-595-overexpressing HO8910PM and SKOV-3 cell proliferation, colony formation and invasion. These data suggested that miR-595 acted a tumor suppressor role in ovarian cancer development and increased the sensitivity of ovarian cancer to cisplatin.
Collapse
Affiliation(s)
- Songyu Tian
- Department of Gynecology Oncology, Cancer Hospital of Harbin Medical University, Harin, 150081, Heilongjiang, China
| | - Mingyue Zhang
- Department of Anaesthesiology, Cancer Hospital of Harbin Medical University, Harin, 150081, Heilongjiang, China
| | - Xiuwei Chen
- Department of Gynecology Oncology, Cancer Hospital of Harbin Medical University, Harin, 150081, Heilongjiang, China
| | - Yunduo Liu
- Department of Gynecology Oncology, Cancer Hospital of Harbin Medical University, Harin, 150081, Heilongjiang, China
| | - Ge Lou
- Department of Gynecology Oncology, Cancer Hospital of Harbin Medical University, Harin, 150081, Heilongjiang, China
| |
Collapse
|
46
|
Ma Y, Liang AJ, Fan YP, Huang YR, Zhao XM, Sun Y, Chen XF. Dysregulation and functional roles of miR-183-96-182 cluster in cancer cell proliferation, invasion and metastasis. Oncotarget 2018; 7:42805-42825. [PMID: 27081087 PMCID: PMC5173173 DOI: 10.18632/oncotarget.8715] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 03/31/2016] [Indexed: 02/07/2023] Open
Abstract
Previous studies have reported aberrant expression of the miR-183-96-182 cluster in a variety of tumors, which indicates its' diagnostic or prognostic value. However, a key characteristic of the miR-183-96-182 cluster is its varied expression levels, and pleomorphic functional roles in different tumors or under different conditions. In most tumor types, the cluster is highly expressed and promotes tumorigenesis, cancer progression and metastasis; yet tumor suppressive effects have also been reported in some tumors. In the present study, we discuss the upstream regulators and the downstream target genes of miR-183-96-182 cluster, and highlight the dysregulation and functional roles of this cluster in various tumor cells. Newer insights summarized in this review will help readers understand the different facets of the miR-183-96-182 cluster in cancer development and progression.
Collapse
Affiliation(s)
- Yi Ma
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - A-Juan Liang
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Yu-Ping Fan
- Reproductive Medicine Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yi-Ran Huang
- Department of Urology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiao-Ming Zhao
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Yun Sun
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Xiang-Feng Chen
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China.,Department of Urology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
47
|
Weidle UH, Birzele F, Kollmorgen G, Nopora A. Potential microRNA-related Targets for Therapeutic Intervention with Ovarian Cancer Metastasis. Cancer Genomics Proteomics 2018; 15:1-15. [PMID: 29275359 PMCID: PMC5822180 DOI: 10.21873/cgp.20061] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 10/09/2017] [Accepted: 10/17/2017] [Indexed: 02/06/2023] Open
Abstract
Treatment of disseminated epithelial ovarian cancer (EOC) is an unmet medical need. Therefore, the identification along with preclinical and clinical validation of new targets is an issue of high importance. In this review we focus on microRNAs that mediate metastasis of EOC. We summarize up-regulated metastasis-promoting and down-regulated metastasis-suppressing microRNAs. We focus on preclinical in vitro and in vivo functions as well as their metastasis-related clinical correlations. Finally, we outline modalities for therapeutic intervention and critical issues of microRNA-based therapeutics in the context of metastatic EOC.
Collapse
Affiliation(s)
- Ulrich H Weidle
- Roche Innovation Center Munich, Roche Diagnostics GmbH, Penzberg, Germany
| | - Fabian Birzele
- Roche Innovation Center Basel, F. Hofman La Roche, Basel, Switzerland
| | - Gwen Kollmorgen
- Roche Innovation Center Munich, Roche Diagnostics GmbH, Penzberg, Germany
| | - Adam Nopora
- Roche Innovation Center Munich, Roche Diagnostics GmbH, Penzberg, Germany
| |
Collapse
|
48
|
Zeleniak AE, Huang W, Fishel ML, Hill R. PTEN-Dependent Stabilization of MTSS1 Inhibits Metastatic Phenotype in Pancreatic Ductal Adenocarcinoma. Neoplasia 2017; 20:12-24. [PMID: 29175021 PMCID: PMC5714254 DOI: 10.1016/j.neo.2017.10.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 10/10/2017] [Accepted: 10/23/2017] [Indexed: 12/19/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) presents at metastatic stage in over 50% of patients. With a survival rate of just 2.7% for patients presenting with distant disease, it is imperative to uncover novel mechanisms capable of suppressing metastasis in PDAC. Previously, we reported that the loss of metastasis suppressor protein 1 (MTSS1) in PDAC cells results in significant increase in cellular migration and invasion. Conversely, we also found that overexpressing MTSS1 in metastatic PDAC cell lines corresponds with not only decreased metastatic phenotype, but also greater overall survival. While it is known that MTSS1 is downregulated in late-stage PDAC, the mechanism behind that loss has not yet been elucidated. Here, we build off our previous findings to present a novel regulatory mechanism for the stabilization of MTSS1 via the tumor suppressor protein phosphatase and tensin homolog (PTEN). We show that PTEN loss in PDAC cells results in a decrease in MTSS1 expression and increased metastatic potential. Additionally, we demonstrate that PTEN forms a complex with MTSS1 in order to stabilize and protect it from proteasomal degradation. Finally, we show that the inflammatory tumor microenvironment, which makes up over 90% of PDAC tumor bulk, is capable of downregulating PTEN expression through secretion of miRNA-23b, potentially uncovering a novel extrinsic mechanism of MTSS1 regulation. Collectively, these data offer new insight into the role and regulation of MTSS1in suppressing tumor cell invasion and migration and help shed light as to what molecular mechanisms could be leading to early cell dissemination in PDAC.
Collapse
Affiliation(s)
- Ann E Zeleniak
- Harper Cancer Research Institute, University of Notre Dame, South Bend, IN 46556, USA; Integrated Biomedical Sciences Program, University of Notre Dame, South Bend, IN 46556, USA
| | - Wei Huang
- Harper Cancer Research Institute, University of Notre Dame, South Bend, IN 46556, USA; Department of Biological Sciences, University of Notre Dame, South Bend, IN 46556, USA
| | - Melissa L Fishel
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Pediatrics, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Pancreatic Cancer Signature Center, Indianapolis, IN 46202, USA
| | - Reginald Hill
- Harper Cancer Research Institute, University of Notre Dame, South Bend, IN 46556, USA; Department of Biological Sciences, University of Notre Dame, South Bend, IN 46556, USA.
| |
Collapse
|
49
|
Braicu OL, Budisan L, Buiga R, Jurj A, Achimas-Cadariu P, Pop LA, Braicu C, Irimie A, Berindan-Neagoe I. miRNA expression profiling in formalin-fixed paraffin-embedded endometriosis and ovarian cancer samples. Onco Targets Ther 2017; 10:4225-4238. [PMID: 28894379 PMCID: PMC5584916 DOI: 10.2147/ott.s137107] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Endometriosis is an inflammatory pathology associated with a negative effect on life quality. Recently, this pathology was connected to ovarian cancer, in particular with endometrioid ovarian cancer. microRNAs (miRNAs) are a class of RNA transcripts ~19–22 nucleotides in length, the altered miRNA pattern being connected to pathological status. miRNAs are highly stable transcripts, and these can be assessed from formalin-fixed paraffin-embedded (FFPE) samples leading to the identification of miRNAs that could be developed as diagnostic and prognostic biomarkers, in particular those involved in malignant transformation. The aim of our study was to evaluate miRNA expression pattern in FFPE samples from endometriosis and ovarian cancer patients using PCR-array technology and also to compare the differential expression pattern in ovarian cancer versus endometriosis. For the PCR-array study, we have used nine macrodissected FFPE samples from endometriosis tissue, eight samples of ovarian cancers and five normal ovarian tissues. Quantitative real-time PCR (qRT-PCR) was used for data validation in a new patient cohort of 17 normal samples, 33 endometriosis samples and 28 ovarian cancer macrodissected FFPE samples. Considering 1.5-fold expression difference as a cut-off level and a P-value <0.05, we have identified four miRNAs being overexpressed in endometrial tissue, while in ovarian cancer 15 were differentially expressed (nine overexpressed and six downregulated). The expression level was confirmed by qRT-PCR for miR-93, miR-141, miR-155, miR-429, miR-200c, miR-205 and miR-492. Using the interpretative program Ingenuity Pathway Analysis revealed several deregulated pathways due to abnormal miRNA expression in endometriosis and ovarian cancer, which in turn is responsible for pathogenesis; this differential expression of miRNAs can be exploited as a therapeutic target. A higher number of altered miRNAs were detected in endometriosis versus ovarian cancer tissue, most of them being linked with epithelial-to-mesenchymal transition.
Collapse
Affiliation(s)
- Ovidiu-Leonard Braicu
- Department of Surgery, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca
| | - Liviuta Budisan
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy
| | - Rares Buiga
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy.,Pathology Department, The Oncology Institute Prof. Dr. Ion Chiricuta, Cluj-Napoca
| | - Ancuta Jurj
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy
| | - Patriciu Achimas-Cadariu
- Department of Surgery, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca.,Department of Surgical Oncology, The Oncology Institute Prof. Dr. Ion Chiricuta, Cluj-Napoca
| | - Laura Ancuta Pop
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy
| | - Cornelia Braicu
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy
| | - Alexandru Irimie
- Department of Surgery, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca.,Department of Surgical Oncology, The Oncology Institute Prof. Dr. Ion Chiricuta, Cluj-Napoca
| | - Ioana Berindan-Neagoe
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy.,MEDFUTURE-Research Center for Advanced Medicine, University of Medicine and Pharmacy Iuliu-Hatieganu, Cluj-Napoca.,Department of Functional Genomics, Proteomics and Experimental Pathology, The Oncology Institute Prof. Dr. Ion Chiricuta, Cluj-Napoca, Romania
| |
Collapse
|
50
|
miR-130a upregulates mTOR pathway by targeting TSC1 and is transactivated by NF-κB in high-grade serous ovarian carcinoma. Cell Death Differ 2017; 24:2089-2100. [PMID: 28800130 DOI: 10.1038/cdd.2017.129] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Revised: 06/23/2017] [Accepted: 07/05/2017] [Indexed: 11/08/2022] Open
Abstract
Activation of mammalian target of rapamycin (mTOR) signaling pathway is associated with poor prognosis of epithelial ovarian cancer. The TSC1-TSC2 complex is a critical negative regulator of mTOR signaling. Here, we demonstrated that TSC1 was frequently downregulated in high-grade serous ovarian carcinoma (HGSOC) and low TSC1 expression level is associated with advanced tumor stage. We next identified miR-130a to be a negative regulator of TSC1 by targeting its 3'UTR. miR-130a was overexpressed in HGSOC and could drive proliferation and invasion/metastasis of ovarian cancer cells. miR-130a could also attenuate rapamycin/starvation-induced autophagy. Ectopic TSC1 expression could block the effects of miR-130a on cell proliferation, migration and autophagy. Finally, we found that miR-130a expression could be upregulated by inflammatory factors and was transactivated by NF-κB. Therefore, our findings establish a crosstalk between inflammation and mTOR signaling that is mediated by miR-130a, which might have a pivotal role in the initiation and progression of HGSOC.
Collapse
|