1
|
Aloufi A, Aubee J, Vargas KM, Apprey V, Thompson K, Copeland R, Kanaan Y, Ricks-Santi L, Brim H, Abbas M. Vitamin D receptor polymorphisms and associated miRNAs in the development of breast cancer in African American women. Gene 2024; 927:148695. [PMID: 38945313 PMCID: PMC11462433 DOI: 10.1016/j.gene.2024.148695] [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: 01/08/2024] [Revised: 05/31/2024] [Accepted: 06/12/2024] [Indexed: 07/02/2024]
Abstract
Breast cancer (BCa) is a prevalent form of cancer in women, exhibiting varying rates and distribution across different ethnic groups. Among these groups, African American (AA) women have the highest incidence of BCa and the lowest levels of Vitamin D (VD). Numerous studies have explored the connection between variations in the VDR gene and BCa risk, particularly in different populations, but research on the AA population remains limited. Epigenetic modifications, including specific microRNAs (miRNAs), can influence gene expression without altering the genetic code and have been implicated in cancer initiation and progression. Our hypothesis suggests that VDR gene variations may increase BCa risk in AA women and that changes in miRNA expression profiles could contribute to BCa development. Using data from the 1000 Genome Project, we identified five VDR gene variants with significant frequency differences between AA and European-American (EA) populations. We genotyped 404 African American BCa cases and controls for five variants using TaqMan® assays. SNPstats assessed their association with BCa risk. The rs1544410 variant's recessive model (A/A) showed a decreased BCa risk in AA (odds ratio 0.33, 95% CI: 0.15-0.73, p-value 0.0041). Conversely, the rs2853563 variant's recessive model (A/A) was linked to an increased BCa risk (odds ratio 4.04, 95% CI: 1.49-10.95, p-value 0.0022). We investigated miRNA expression influenced by VD in HCC1806 Triple-Negative Breast Cancer (TNBC) cell lines with the A/A allele for rs2853563. nCounter® Nanostring technology assessed miRNA profiles after calcitriol treatment. Our results indicated that calcitriol treatment led to reduced expression of six miRNAs, four of which are associated with tumor suppression in the presence of the AA genotype in TNBC cell lines. These findings suggest that specific VDR genotypes could have a potential effect on the miRNAs expression which could potentially serve as markers for cell proliferation in TNBC.
Collapse
Affiliation(s)
- Abrar Aloufi
- Howard University, Department of Microbiology, Washington, DC, USA
| | - Joseph Aubee
- Howard University, Department of Microbiology, Washington, DC, USA
| | - Kevin Monsalve Vargas
- MedStar Georgetown University Hospital, Pre/Postoperative Services, Washington, DC, USA
| | - Victor Apprey
- The National Human Genome Center, Howard University, Washington, DC, USA
| | - Karl Thompson
- Howard University, Department of Microbiology, Washington, DC, USA
| | - Robert Copeland
- Howard University, Department of Microbiology, Washington, DC, USA
| | - Yasmine Kanaan
- Howard University, Department of Microbiology, Washington, DC, USA
| | | | - Hassan Brim
- Howard University, Department of Pathology, Washington, DC, USA.
| | - Muneer Abbas
- Howard University, Department of Microbiology, Washington, DC, USA; The National Human Genome Center, Howard University, Washington, DC, USA.
| |
Collapse
|
2
|
Baziyar MA, Hosseini A, Jandel F. The role of palbociclib on the alterations in CDKN2, CCNE1, E2F3, MDM2 expressions as target genes of miR-141. PLoS One 2024; 19:e0306545. [PMID: 39116089 PMCID: PMC11309483 DOI: 10.1371/journal.pone.0306545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 06/18/2024] [Indexed: 08/10/2024] Open
Abstract
INTRODUCTION According to WHO, Breast cancer is widely considered to be the first or second cause of cancer-related death almost universally. Cell cycle disruption, either in the form of uncontrolled expression of cyclins or because of the suspension in negative regulatory proteins (CDK inhibitors), was found to cause breast cancer. Palbociclib as specific CDK4/6 inhibitor is used for the treatment of ER+ metastatic cancers. In this study, we are looking to investigate the effect of palbociclib on breast cancer cells and evaluate the changes in the expression of some genes involved in the cell cycle as target genes of miR-141 after treatment with this drug. We used MCF7 as functional estrogen and non-invasive and MDA-MB-231 cell lines as triple-negative type of breast cancer and a model for more aggressive. METHOD & MATERIALS MCF7 and MDA-MB-231 cell lines were cultured in DMEM medium. After counting cells and measuring viability, Palbociclib was administered at varying doses using the IC50 obtained from MTT, with the treatment given at two time points of 24 and 72 hours. RNA was extracted from untreated and treated cells and RNAs were converted to cDNA in the end. Gene expression changes were investigated by real-time PCR. Data management and analysis were conducted using GraphPad Prism 5.01 software. RESULT AND CONCLUSION Among investigated genes, E2F3 gene was not significantly affected by Palbociclib in any of cell lines and time points. Besides, the expression of CCNE1 gene was significantly suppressed. It seems this drug was unable to reduce the expression of MDM2 gene significantly in triple negative (MDA-MB-231) cancer cells; however, a decrease was observed in luminal A (MCF-7) cells. CDKN2A and miR-141 genes expression increased significantly after treatment which can be aligned with palbociclib in proliferation inhibition.
Collapse
Affiliation(s)
- Mohammad Ali Baziyar
- Department of Medical Biotechnology, School of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Arshad Hosseini
- Department of Medical Biotechnology, School of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Farinush Jandel
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| |
Collapse
|
3
|
Ribeiro HG, Galdino OA, de Souza KSC, Rosa Neta AP, Lin-Wang HT, Cunha-Neto E, de Rezende AA, Silbiger VN. Unraveling the role of miRNAs as biomarkers in Chagas cardiomyopathy: Insights into molecular pathophysiology. PLoS Negl Trop Dis 2024; 18:e0011865. [PMID: 38300899 PMCID: PMC10833550 DOI: 10.1371/journal.pntd.0011865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2024] Open
Abstract
BACKGROUND Chagas cardiomyopathy (ChCM) is a severe form of Chagas disease and a major cause of cardiovascular morbidity and mortality. The dysregulation of the immune response leads to cardiac remodeling and functional disruptions, resulting in life-threatening complications. Conventional diagnostic methods have limitations, and therapeutic response evaluation is challenging. MicroRNAs (miRNAs), important regulators of gene expression, show potential as biomarkers for diagnosis and prognosis. AIM This review aims to summarize experimental findings on miRNA expression in ChCM and explore the potential of these miRNAs as biomarkers of Chagas disease. METHODS The search was conducted in the US National Library of Medicine MEDLINE/PubMed public database using the terms "Chagas cardiomyopathy" OR "Chagas disease" AND "microRNA" OR "miRNA" OR "miR." Additionally, bioinformatics analysis was performed to investigate miRNA-target interactions and explore enrichment pathways of gene ontology biological processes and molecular functions. RESULTS The miR-21, miR-146b, miR-146a, and miR-155 consistently exhibited up-regulation, whereas miR-145 was down-regulated in ChCM. These specific miRNAs have been linked to fibrosis, immune response, and inflammatory processes in heart tissue. Moreover, the findings from various studies indicate that these miRNAs have the potential as biomarkers for the disease and could be targeted in therapeutic strategies for ChCM. CONCLUSION In this review, we point out miR-21, miR-146b, miR-146a, miR-155, and miR-145-5p role in the complex mechanisms of ChCM. These miRNAs have been shown as potential biomarkers for precise diagnosis, reliable prognostic evaluation, and effective treatment strategies in the ChCM.
Collapse
Affiliation(s)
- Heriks Gomes Ribeiro
- Department of Clinical and Toxicological Analysis, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Ony Araújo Galdino
- Department of Clinical and Toxicological Analysis, Federal University of Rio Grande do Norte, Natal, Brazil
| | | | - Antonia Pereira Rosa Neta
- Department of Clinical and Toxicological Analysis, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Hui Tzu Lin-Wang
- Molecular Biology Laboratory, Dante Pazzanese Institute of Cardiology, São Paulo, Brazil
| | - Edecio Cunha-Neto
- Laboratory of Immunology, Heart Institute (InCor), University of São Paulo, School of Medicine, São Paulo, Brazil
- Division of Clinical Immunology and Allergy, University of São Paulo, School of Medicine, São Paulo, Brazil
- Institute for Investigation in Immunology (iii), INCT, São Paulo, Brazil
| | - Adriana Augusto de Rezende
- Department of Clinical and Toxicological Analysis, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Vivian Nogueira Silbiger
- Department of Clinical and Toxicological Analysis, Federal University of Rio Grande do Norte, Natal, Brazil
- Translational Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
| |
Collapse
|
4
|
Yin J, Gu T, Chaudhry N, Davidson NE, Huang Y. Epigenetic modulation of antitumor immunity and immunotherapy response in breast cancer: biological mechanisms and clinical implications. Front Immunol 2024; 14:1325615. [PMID: 38268926 PMCID: PMC10806158 DOI: 10.3389/fimmu.2023.1325615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Accepted: 12/22/2023] [Indexed: 01/26/2024] Open
Abstract
Breast cancer (BC) is the most common non-skin cancer and the second leading cause of cancer death in American women. The initiation and progression of BC can proceed through the accumulation of genetic and epigenetic changes that allow transformed cells to escape the normal cell cycle checkpoint control. Unlike nucleotide mutations, epigenetic changes such as DNA methylation, histone posttranslational modifications (PTMs), nucleosome remodeling and non-coding RNAs are generally reversible and therefore potentially responsive to pharmacological intervention. Epigenetic dysregulations are critical mechanisms for impaired antitumor immunity, evasion of immune surveillance, and resistance to immunotherapy. Compared to highly immunogenic tumor types, such as melanoma or lung cancer, breast cancer has been viewed as an immunologically quiescent tumor which displays a relatively low population of tumor-infiltrating lymphocytes (TIL), low tumor mutational burden (TMB) and modest response rates to immune checkpoint inhibitors (ICI). Emerging evidence suggests that agents targeting aberrant epigenetic modifiers may augment host antitumor immunity in BC via several interrelated mechanisms such as enhancing tumor antigen presentation, activation of cytotoxic T cells, inhibition of immunosuppressive cells, boosting response to ICI, and induction of immunogenic cell death (ICD). These discoveries have established a highly promising basis for using combinatorial approaches of epigenetic drugs with immunotherapy as an innovative paradigm to improve outcomes of BC patients. In this review, we summarize the current understanding of how epigenetic processes regulate immune cell function and antitumor immunogenicity in the context of the breast tumor microenvironment. Moreover, we discuss the therapeutic potential and latest clinical trials of the combination of immune checkpoint blockers with epigenetic agents in breast cancer.
Collapse
Affiliation(s)
- Jun Yin
- The University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Tiezheng Gu
- The University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Norin Chaudhry
- Department of Internal Medicine, Division of Hematology, Oncology, and Blood and Marrow Transplantation, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Nancy E. Davidson
- Fred Hutchinson Cancer Center, University of Washington, Seattle, WA, United States
| | - Yi Huang
- Department of Internal Medicine, Division of Hematology, Oncology, and Blood and Marrow Transplantation, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, United States
| |
Collapse
|
5
|
Garrido-Cano I, Adam-Artigues A, Lameirinhas A, Blandez JF, Candela-Noguera V, Lluch A, Bermejo B, Sancenón F, Cejalvo JM, Martínez-Máñez R, Eroles P. Delivery of miR-200c-3p Using Tumor-Targeted Mesoporous Silica Nanoparticles for Breast Cancer Therapy. ACS APPLIED MATERIALS & INTERFACES 2023; 15:38323-38334. [PMID: 37549382 PMCID: PMC10436244 DOI: 10.1021/acsami.3c07541] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 07/25/2023] [Indexed: 08/09/2023]
Abstract
Despite advances in breast cancer treatment, it remains the leading cause of cancer-related death in women worldwide. In this context, microRNAs have emerged as potential therapeutic targets but still present some limitations for in vivo applications. Particularly, miR-200c-3p is a well-known tumor suppressor microRNA that inhibits tumor progression and metastasis in breast cancer through downregulating ZEB1 and ZEB2. Based on the above, we describe the design and validation of a nanodevice using mesoporous silica nanoparticles for miR-200c-3p delivery for breast cancer treatment. We demonstrate the biocompatibility of the synthesized nanodevices as well as their ability to escape from endosomes/lysosomes and inhibit tumorigenesis, invasion, migration, and proliferation of tumor cells in vitro. Moreover, tumor targeting and effective delivery of miR-200c-3p from the nanoparticles in vivo are confirmed in an orthotopic breast cancer mouse model, and the therapeutic efficacy is also evidenced by a decrease in tumor size and lung metastasis, while showing no signs of toxicity. Overall, our results provide evidence that miR-200c-3p-loaded nanoparticles are a potential strategy for breast cancer therapy and a safe and effective system for tumor-targeted delivery of microRNAs.
Collapse
Affiliation(s)
- Iris Garrido-Cano
- Biomedical
Research Institute INCLIVA, Valencia 46010, Spain
- Instituto
Interuniversitario de Investigación de Reconocimiento Molecular
y Desarrollo Tecnológico (IDM), Universitat Politècnica
de València, Universitat de València, Valencia 46010, Spain
| | | | - Ana Lameirinhas
- Biomedical
Research Institute INCLIVA, Valencia 46010, Spain
| | - Juan F. Blandez
- Instituto
Interuniversitario de Investigación de Reconocimiento Molecular
y Desarrollo Tecnológico (IDM), Universitat Politècnica
de València, Universitat de València, Valencia 46010, Spain
- CIBER
de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid 28029, Spain
- Unidad
Mixta de Investigación en Nanomedicina y Sensores, Universitat Politècnica de València,
IIS La Fe, Valencia 46026, Spain
| | - Vicente Candela-Noguera
- Instituto
Interuniversitario de Investigación de Reconocimiento Molecular
y Desarrollo Tecnológico (IDM), Universitat Politècnica
de València, Universitat de València, Valencia 46010, Spain
| | - Ana Lluch
- Biomedical
Research Institute INCLIVA, Valencia 46010, Spain
- Centro
de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid 28029, Spain
- Universitat
de València, Valencia 46010, Spain
- Clinical
Oncology Department, Hospital Clínico
Universitario de Valencia, Valencia 46010, Spain
| | - Begoña Bermejo
- Biomedical
Research Institute INCLIVA, Valencia 46010, Spain
- Centro
de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid 28029, Spain
- Clinical
Oncology Department, Hospital Clínico
Universitario de Valencia, Valencia 46010, Spain
| | - Felix Sancenón
- Instituto
Interuniversitario de Investigación de Reconocimiento Molecular
y Desarrollo Tecnológico (IDM), Universitat Politècnica
de València, Universitat de València, Valencia 46010, Spain
- CIBER
de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid 28029, Spain
- Unidad
Mixta de Investigación en Nanomedicina y Sensores, Universitat Politècnica de València,
IIS La Fe, Valencia 46026, Spain
- Unidad
Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades
y Nanomedicina. Universitat Politècnica de Valencia, Centro de Investigación Príncipe Felipe, Valencia 46012, Spain
| | - Juan Miguel Cejalvo
- Biomedical
Research Institute INCLIVA, Valencia 46010, Spain
- Centro
de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid 28029, Spain
- Clinical
Oncology Department, Hospital Clínico
Universitario de Valencia, Valencia 46010, Spain
| | - Ramón Martínez-Máñez
- Instituto
Interuniversitario de Investigación de Reconocimiento Molecular
y Desarrollo Tecnológico (IDM), Universitat Politècnica
de València, Universitat de València, Valencia 46010, Spain
- CIBER
de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid 28029, Spain
- Unidad
Mixta de Investigación en Nanomedicina y Sensores, Universitat Politècnica de València,
IIS La Fe, Valencia 46026, Spain
- Unidad
Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades
y Nanomedicina. Universitat Politècnica de Valencia, Centro de Investigación Príncipe Felipe, Valencia 46012, Spain
| | - Pilar Eroles
- Biomedical
Research Institute INCLIVA, Valencia 46010, Spain
- Centro
de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid 28029, Spain
- Universitat
de València, Valencia 46010, Spain
| |
Collapse
|
6
|
Gahlawat AW, Witte T, Sinn P, Schott S. Circulating cf-miRNA as a more appropriate surrogate liquid biopsy marker than cfDNA for ovarian cancer. Sci Rep 2023; 13:5503. [PMID: 37015943 PMCID: PMC10073086 DOI: 10.1038/s41598-023-32243-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 03/24/2023] [Indexed: 04/06/2023] Open
Abstract
Ovarian cancer (OC) is an aggressive disease, primarily diagnosed in late stages with only 20% of patients surviving more than 5 years. Liquid biopsy markers have great potential to improve current diagnostic and prognostic methods. Here, we compared miRNAs and DNA methylation in matched plasma, whole blood and tissues as a surrogate marker for OC. We found that while both cfDNA and cf-miRNAs levels were upregulated in OC compared to patients with benign lesions or healthy controls, only cf-miRNA levels were an independent prognosticator of survival. Following on our previous work, we found members of the miR-200 family, miR-200c and miR-141 to be upregulated in both plasma and matched tissues of OC patients which correlated with adverse clinical features. We could also show that the upregulation of miR-200c and -141 correlated with promoter DNA hypomethylation in tissues, but not in plasma or matched whole blood samples. As cf-miRNAs are more easily obtained and very stable in blood, we conclude that they might serve as a more appropriate surrogate liquid biopsy marker than cfDNA for OC.
Collapse
Affiliation(s)
- Aoife Ward Gahlawat
- Department of Gynaecology and Obstetrics, University Hospital of Heidelberg, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany.
- National Center for Tumor Diseases (NCT), University Hospital of Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Tania Witte
- Department of Gynaecology and Obstetrics, University Hospital of Heidelberg, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
| | - Peter Sinn
- Department of Pathology, University Hospital of Heidelberg, Heidelberg, Germany
| | - Sarah Schott
- Department of Gynaecology and Obstetrics, University Hospital of Heidelberg, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
| |
Collapse
|
7
|
Fotakopoulos G, Georgakopoulou VE, Spandidos DA, Papalexis P, Angelopoulou E, Aravantinou-Fatorou A, Trakas N, Trakas I, Brotis AG. Role of miR‑200 family in brain metastases: A systematic review. Mol Clin Oncol 2023; 18:15. [PMID: 36798467 PMCID: PMC9926042 DOI: 10.3892/mco.2023.2611] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 01/18/2023] [Indexed: 01/26/2023] Open
Abstract
Brain metastasis (BM) represents the single most severe neurological complication of systemic cancer. The prognosis of patients with BM is poor, irrespective of the implemented treatment. The present study performed a systematic review of the literature using three online databases (PubMed, Scopus and Web of Science). Recently, a number of small RNA molecules, the microRNAs (miRNAs/miRs), have attracted increasing scientific attention. Members of the miR-200 family, which includes five miRNAs (miR-141, miR-200a, miR-200b, miR-200c and miR-429) appear to play pivotal roles in cancer initiation and metastasis. Indeed, a systematic review of the pertinent literature revealed that miR-200 family members regulate the brain metastatic cascade, particularly by modulating epithelial-to-mesenchymal transition. That holds true for the major representatives of BM, including lung and breast cancer, as well as for other less frequent secondary lesions originating from melanoma and the gastrointestinal tract. Therefore, the miRNAs may serve as potential diagnostic and/or prognostic markers, and under specific circumstances, as invaluable therapeutic targets. However, the available clinical evidence is relatively limited. A number of studies have suggested that the miR-200 family members are accurate prognostic markers of survival and resistance to chemotherapy in patients with breast cancer. Similarly, they may prove helpful in differentiating a metastatic lesion from a malignant glioma, or a hemangioblastoma from a renal cell carcinoma in patients with von Hippel Lindau syndrome, based on a cerebrospinal fluid sample. However, currently, there is no known therapeutic role for miR-200 family members in the setting of BM.
Collapse
Affiliation(s)
- George Fotakopoulos
- Department of Neurosurgery, General University Hospital of Larissa, 41221 Larissa, Greece,Correspondence to: Dr George Fotakopoulos, Department of Neurosurgery, General University Hospital of Larissa, Mezourlo, 41221 Larissa, Greece
| | - Vasiliki Epameinondas Georgakopoulou
- Department of Infectious Diseases and COVID-19 Unit, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Demetrios A. Spandidos
- Laboratory of Clinical Virology, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Petros Papalexis
- Unit of Endocrinology, First Department of Internal Medicine, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece,Department of Biomedical Sciences, University of West Attica, 12243 Athens, Greece
| | - Efthalia Angelopoulou
- Department of Neurology, Eginitio University Hospital, National and Kapodistrian University of Athens, 11528 Athens, Greece
| | - Aikaterini Aravantinou-Fatorou
- First Department of Internal Medicine, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Nikolaos Trakas
- Department of Biochemistry, Sismanogleio Hospital, 15126 Athens, Greece
| | - Ilias Trakas
- Department of Infectious Diseases and COVID-19 Unit, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Alexandros G. Brotis
- Department of Neurosurgery, General University Hospital of Larissa, 41221 Larissa, Greece
| |
Collapse
|
8
|
Tovarnytska A. Breastfeeding impact on patent ductus arteriosus closure in preterm newborns. CHILD`S HEALTH 2022; 17:184-191. [DOI: 10.22141/2224-0551.17.4.2022.1515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
Background. Patent ductus arteriosus (PDA) is a serious problem in clinical neonatology and pediatrics. Today treatment of PDA remains debatable because of high risk of complications with medical and invasive treatment. The purpose of the study was to determine the dependence of PDA closure on the type of feeding the newborn. Materials and methods. We have made a retrospective analysis of 300 case histories of children in neonatal units. We selected two comparison groups: the main group consisted of breastfed babies born at 37 weeks gestation (n = 102); controls — preterm infants who received artificial feeding since birth (n = 198). Results. At discharge from the hospital, 44 (14.7 ± 2.0 %) children had PDA (p < 0.05). The percentage of infants on artificial feeding with PDA was 2.3 times higher compared to breastfed babies: 18.2 ± 2.7 % versus 7.8 ± 2.7 % (p = 0.017 by χ2 test). Respiratory distress syndrome was more common in the control group as well: 72.7 ± 3.2 % versus 46.1 ± 4.9 % (p < 0.001). Additionally, the group of formula-fed children more often had severe complications of respiratory distress syndrome: respiratory failure type III — 25.8 ± 3.1 % versus12.7 ± 3.3 % (р = 0.009); development of bronchopulmonary dysplasia — 13.1 ± 2.4 % versus 2.9 ± 1.7 % (p = 0.005). The need for invasive mechanical ventilation in the intensive care unit was observed much less often in breastfed children. Moreover, the level of respiratory disorders (type II respiratory failure) in both groups differed insignificantly (p = 0.742): 24.5 ± 4.3 % in the main group and 26.3 ± 3.1 % in controls. Conclusions. Breastfeeding from the first days of life contributes to the closure of the ductus arteriosus and the favorable course of respiratory distress syndrome in premature babies.
Collapse
|
9
|
Sundararajan V, Burk UC, Bajdak-Rusinek K. Revisiting the miR-200 Family: A Clan of Five Siblings with Essential Roles in Development and Disease. Biomolecules 2022; 12:biom12060781. [PMID: 35740906 PMCID: PMC9221129 DOI: 10.3390/biom12060781] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/29/2022] [Accepted: 06/01/2022] [Indexed: 12/07/2022] Open
Abstract
Over two decades of studies on small noncoding RNA molecules illustrate the significance of microRNAs (miRNAs/miRs) in controlling multiple physiological and pathological functions through post-transcriptional and spatiotemporal gene expression. Among the plethora of miRs that are essential during animal embryonic development, in this review, we elaborate the indispensable role of the miR-200 family (comprising miR-200a, -200b, 200c, -141, and -429) in governing the cellular functions associated with epithelial homeostasis, such as epithelial differentiation and neurogenesis. Additionally, in pathological contexts, miR-200 family members are primarily involved in tumor-suppressive roles, including the reversal of the cancer-associated epithelial–mesenchymal transition dedifferentiation process, and are dysregulated during organ fibrosis. Moreover, recent eminent studies have elucidated the crucial roles of miR-200s in the pathophysiology of multiple neurodegenerative diseases and tissue fibrosis. Lastly, we summarize the key studies that have recognized the potential use of miR-200 members as biomarkers for the diagnosis and prognosis of cancers, elaborating the application of these small biomolecules in aiding early cancer detection and intervention.
Collapse
Affiliation(s)
- Vignesh Sundararajan
- Cancer Science Institute of Singapore, National University of Singapore, Center for Translational Medicine, Singapore 117599, Singapore;
| | - Ulrike C. Burk
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany;
| | - Karolina Bajdak-Rusinek
- Department of Medical Genetics, Faculty of Medical Sciences, Medical University of Silesia, 40-752 Katowice, Poland
- Correspondence: ; Tel.: +48-32-208-8382
| |
Collapse
|
10
|
Su K, Peng Y, Yu H. Development of a Prognostic Model Based on Pyroptosis-Related Genes in Pancreatic Adenocarcinoma. DISEASE MARKERS 2022; 2022:9141117. [PMID: 35677632 PMCID: PMC9169203 DOI: 10.1155/2022/9141117] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 05/12/2022] [Accepted: 05/19/2022] [Indexed: 11/17/2022]
Abstract
Background The importance of pyroptosis in tumorigenesis and cancer progression is becoming increasingly apparent. However, the efficacy of using pyroptosis-related genes (PRGs) in predicting the prognosis of pancreatic adenocarcinoma (PAAD) patients is unknown. Methods This investigation used two databases to obtain expression data for PAAD patients. Differentially expressed PRGs (DEPRGs) were identified between PAAD and control samples. Several bioinformatic approaches were used to analyze the biological functions of DEPRGs and to identify prognostic DERPGs. A miRNA-prognostic DEPRG-transcription factor (TF) regulatory network was created via the miRNet online tool. A risk score model was created after each patient's risk score was calculated. The microenvironments of the low- and high-risk groups were assessed using xCell, the expression of immune checkpoints was determined, and gene set variation analysis (GSVA) was performed. Finally, the efficacy of certain potential drugs was predicted using the pRRophetic algorithm, and the results in the high- and low-risk groups were compared. Results A total of 13 DEPRGs were identified between PAAD and control samples. Functional enrichment analysis showed that the DEPRGs had a close relationship with inflammation. In univariate and multivariate Cox regression analyses, GSDMC, IRF1, and PLCG1 were identified as prognostic biomarkers in PAAD. The results of the miRNA-prognostic DEPRG-TF regulatory network showed that GSDMC, IRF1, and PLCG1 were regulated by both specific and common miRNAs and TFs. Based on the risk score and other independent prognostic indicators, a nomogram with a good ability to predict the survival of PAAD patients was developed. By evaluating the tumor microenvironment, we observed that the immune and metabolic microenvironments of the two groups were substantially different. In addition, individuals in the low-risk group were more susceptible to axitinib and camptothecin, whereas lapatinib might be preferred for patients in the high-risk group. Conclusion Our study revealed the prognostic value of PRGs in PAAD and created a reliable model for predicting the prognosis of PAAD patients. Our findings will benefit the prognostication and treatment of PAAD patients.
Collapse
Affiliation(s)
- Kaifeng Su
- Medical Faculty of Ludwig-Maximilians-University of Munich, University Hospital of LMU Munich, Munich, Germany
| | - Yang Peng
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Haochen Yu
- Medical Faculty of Ludwig-Maximilians-University of Munich, University Hospital of LMU Munich, Munich, Germany
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
11
|
Navarro-Manzano E, Luengo-Gil G, González-Conejero R, García-Garre E, García-Martínez E, García-Torralba E, Chaves-Benito A, Vicente V, Ayala de la Peña F. Prognostic and Predictive Effects of Tumor and Plasma miR-200c-3p in Locally Advanced and Metastatic Breast Cancer. Cancers (Basel) 2022; 14:cancers14102390. [PMID: 35625994 PMCID: PMC9139340 DOI: 10.3390/cancers14102390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 05/07/2022] [Accepted: 05/10/2022] [Indexed: 11/16/2022] Open
Abstract
While the role of miR-200c in cancer progression has been established, its expression and prognostic role in breast cancer is not completely understood. The predictive role of miR-200c in response to chemotherapy has also been suggested by some studies, but only limited clinical evidence is available. The purpose of this study was to investigate miR-200c-3p in the plasma and primary tumor of BC patients. The study design included two cohorts involving women with locally advanced (LABC) and metastatic breast cancer. Tumor and plasma samples were obtained before and after treatment. We found that miR-200c-3p was significantly higher in the plasma of BC patients compared with the controls. No correlation of age with plasma miR-200c-3p was found for controls or for BC patients. MiR-200c-3p tumor expression was also associated with poor overall survival in LABC patients treated with neoadjuvant chemotherapy, independently of pathological complete response or clinical stage. Our findings suggest that plasmatic miR-200c-3p levels could be useful for BC staging, while the tumor expression of miR-200c-3p might provide further prognostic information beyond residual disease in BC treated with neoadjuvant chemotherapy.
Collapse
Affiliation(s)
- Esther Navarro-Manzano
- Department of Hematology and Medical Oncology, Hospital Universitario Morales Meseguer, 30008 Murcia, Spain; (E.N.-M.); (G.L.-G.); (R.G.-C.); (E.G.-G.); (E.G.-M.); (E.G.-T.); (V.V.)
- Centro Regional de Hemodonación, 30003 Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria, IMIB, 30120 Murcia, Spain
- Department of Medicine, Medical School, University of Murcia, 30001 Murcia, Spain;
| | - Ginés Luengo-Gil
- Department of Hematology and Medical Oncology, Hospital Universitario Morales Meseguer, 30008 Murcia, Spain; (E.N.-M.); (G.L.-G.); (R.G.-C.); (E.G.-G.); (E.G.-M.); (E.G.-T.); (V.V.)
- Centro Regional de Hemodonación, 30003 Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria, IMIB, 30120 Murcia, Spain
- Department of Medicine, Medical School, University of Murcia, 30001 Murcia, Spain;
| | - Rocío González-Conejero
- Department of Hematology and Medical Oncology, Hospital Universitario Morales Meseguer, 30008 Murcia, Spain; (E.N.-M.); (G.L.-G.); (R.G.-C.); (E.G.-G.); (E.G.-M.); (E.G.-T.); (V.V.)
- Centro Regional de Hemodonación, 30003 Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria, IMIB, 30120 Murcia, Spain
- Department of Medicine, Medical School, University of Murcia, 30001 Murcia, Spain;
| | - Elisa García-Garre
- Department of Hematology and Medical Oncology, Hospital Universitario Morales Meseguer, 30008 Murcia, Spain; (E.N.-M.); (G.L.-G.); (R.G.-C.); (E.G.-G.); (E.G.-M.); (E.G.-T.); (V.V.)
- Instituto Murciano de Investigación Biosanitaria, IMIB, 30120 Murcia, Spain
| | - Elena García-Martínez
- Department of Hematology and Medical Oncology, Hospital Universitario Morales Meseguer, 30008 Murcia, Spain; (E.N.-M.); (G.L.-G.); (R.G.-C.); (E.G.-G.); (E.G.-M.); (E.G.-T.); (V.V.)
- Instituto Murciano de Investigación Biosanitaria, IMIB, 30120 Murcia, Spain
- Medical School, Universidad Católica San Antonio, 30107 Murcia, Spain
| | - Esmeralda García-Torralba
- Department of Hematology and Medical Oncology, Hospital Universitario Morales Meseguer, 30008 Murcia, Spain; (E.N.-M.); (G.L.-G.); (R.G.-C.); (E.G.-G.); (E.G.-M.); (E.G.-T.); (V.V.)
- Instituto Murciano de Investigación Biosanitaria, IMIB, 30120 Murcia, Spain
| | - Asunción Chaves-Benito
- Department of Medicine, Medical School, University of Murcia, 30001 Murcia, Spain;
- Department of Pathology, Hospital Universitario Morales Meseguer, 30008 Murcia, Spain
| | - Vicente Vicente
- Department of Hematology and Medical Oncology, Hospital Universitario Morales Meseguer, 30008 Murcia, Spain; (E.N.-M.); (G.L.-G.); (R.G.-C.); (E.G.-G.); (E.G.-M.); (E.G.-T.); (V.V.)
- Centro Regional de Hemodonación, 30003 Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria, IMIB, 30120 Murcia, Spain
- Department of Medicine, Medical School, University of Murcia, 30001 Murcia, Spain;
| | - Francisco Ayala de la Peña
- Department of Hematology and Medical Oncology, Hospital Universitario Morales Meseguer, 30008 Murcia, Spain; (E.N.-M.); (G.L.-G.); (R.G.-C.); (E.G.-G.); (E.G.-M.); (E.G.-T.); (V.V.)
- Instituto Murciano de Investigación Biosanitaria, IMIB, 30120 Murcia, Spain
- Department of Medicine, Medical School, University of Murcia, 30001 Murcia, Spain;
- Correspondence: ; Tel.: +34-968360900
| |
Collapse
|
12
|
Müller Coan BG, Cesarman E, Acencio ML, Elgui de Oliveira D. Latent Membrane Protein 1 (LMP1) from Epstein-Barr Virus (EBV) Strains M81 and B95.8 Modulate miRNA Expression When Expressed in Immortalized Human Nasopharyngeal Cells. Genes (Basel) 2022; 13:353. [PMID: 35205397 PMCID: PMC8871543 DOI: 10.3390/genes13020353] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 02/12/2022] [Accepted: 02/15/2022] [Indexed: 12/01/2022] Open
Abstract
The Epstein-Barr virus (EBV) is a ubiquitous γ herpesvirus strongly associated with nasopharyngeal carcinomas, and the viral oncogenicity in part relies on cellular effects of the viral latent membrane protein 1 (LMP1). It was previously described that EBV strains B95.8 and M81 differ in cell tropism and the activation of the lytic cycle. Nonetheless, it is unknown whether LMP1 from these strains have different effects when expressed in nasopharyngeal cells. Thus, herein we evaluated the effects of EBV LMP1 derived from viral strains B95.8 and M81 and expressed in immortalized nasopharyngeal cells NP69SV40T in the regulation of 91 selected cellular miRNAs. We found that cells expressing either LMP1 behave similarly in terms of NF-kB activation and cell migration. Nonetheless, the miRs 100-5p, 192-5p, and 574-3p were expressed at higher levels in cells expressing LMP1 B95.8 compared to M81. Additionally, results generated by in silico pathway enrichment analysis indicated that LMP1 M81 distinctly regulate genes involved in cell cycle (i.e., RB1), mRNA processing (i.e., NUP50), and mitochondrial biogenesis (i.e., ATF2). In conclusion, LMP1 M81 was found to distinctively regulate miRs 100-5p, 192-5p, and 574-3p, and the in silico analysis provided valuable clues to dissect the molecular effects of EBV LMP1 expressed in nasopharyngeal cells.
Collapse
Affiliation(s)
- Barbara G. Müller Coan
- Biosciences Institute of Botucatu, São Paulo State University (UNESP), Botucatu 18618-689, SP, Brazil;
| | - Ethel Cesarman
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10065, USA;
| | - Marcio Luis Acencio
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Belvaux, L-4367 Luxembourg, Luxembourg;
| | - Deilson Elgui de Oliveira
- Department of Pathology, Medical School, São Paulo State University (UNESP), Botucatu, SP, 18618-687, Brazil
- ViriCan, Institute for Biotechnology (IBTEC), São Paulo State University (UNESP), Botucatu, SP, 18607-440, Brazil
| |
Collapse
|
13
|
Exosome-derived miR-200a promotes esophageal cancer cell proliferation and migration via the mediating Keap1 expression. Mol Cell Biochem 2022; 477:1295-1308. [PMID: 35137328 DOI: 10.1007/s11010-022-04353-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 01/06/2022] [Indexed: 12/18/2022]
Abstract
Previous studies have reported that exosomes bearing certain microRNAs (miRNAs) are related to the physiological functions of different types of cancer cells. Our study aimed to elucidate the role of miR-200a in esophageal squamous cell carcinoma (ESCC). We observed that miR-200a expression is higher in esophageal carcinoma cells, tissues, and exosomes than in normal cells and healthy tissues. We showed that exosome-shuttled miR-200a promotes the proliferation, migration, and invasion of esophageal cells and inhibits apoptosis, thereby leading to the progression of ESCC. We showed that miR-200a exerts its effects through its interaction with Keap1, thus altering the Keap1/Nrf2 signaling pathway. Our results suggest that exosome-shuttled miR-200a might be useful as a biomarker for prognosis in patients with ESCC.
Collapse
|
14
|
Cavallari I, Ciccarese F, Sharova E, Urso L, Raimondi V, Silic-Benussi M, D’Agostino DM, Ciminale V. The miR-200 Family of microRNAs: Fine Tuners of Epithelial-Mesenchymal Transition and Circulating Cancer Biomarkers. Cancers (Basel) 2021; 13:5874. [PMID: 34884985 PMCID: PMC8656820 DOI: 10.3390/cancers13235874] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/17/2021] [Accepted: 11/18/2021] [Indexed: 12/13/2022] Open
Abstract
The miR-200 family of microRNAs (miRNAs) includes miR-200a, miR-200b, miR-200c, miR-141 and miR-429, five evolutionarily conserved miRNAs that are encoded in two clusters of hairpin precursors located on human chromosome 1 (miR-200b, miR-200a and miR-429) and chromosome 12 (miR-200c and miR-141). The mature -3p products of the precursors are abundantly expressed in epithelial cells, where they contribute to maintaining the epithelial phenotype by repressing expression of factors that favor the process of epithelial-to-mesenchymal transition (EMT), a key hallmark of oncogenic transformation. Extensive studies of the expression and interactions of these miRNAs with cell signaling pathways indicate that they can exert both tumor suppressor- and pro-metastatic functions, and may serve as biomarkers of epithelial cancers. This review provides a summary of the role of miR-200 family members in EMT, factors that regulate their expression, and important targets for miR-200-mediated repression that are involved in EMT. The second part of the review discusses the potential utility of circulating miR-200 family members as diagnostic/prognostic biomarkers for breast, colorectal, lung, ovarian, prostate and bladder cancers.
Collapse
Affiliation(s)
- Ilaria Cavallari
- Veneto Institute of Oncology IOV–IRCCS, 35128 Padova, Italy; (I.C.); (F.C.); (E.S.); (L.U.); (V.R.); (M.S.-B.)
| | - Francesco Ciccarese
- Veneto Institute of Oncology IOV–IRCCS, 35128 Padova, Italy; (I.C.); (F.C.); (E.S.); (L.U.); (V.R.); (M.S.-B.)
| | - Evgeniya Sharova
- Veneto Institute of Oncology IOV–IRCCS, 35128 Padova, Italy; (I.C.); (F.C.); (E.S.); (L.U.); (V.R.); (M.S.-B.)
| | - Loredana Urso
- Veneto Institute of Oncology IOV–IRCCS, 35128 Padova, Italy; (I.C.); (F.C.); (E.S.); (L.U.); (V.R.); (M.S.-B.)
- Department of Surgery, Oncology and Gastroenterology, University of Padua, 35128 Padova, Italy
| | - Vittoria Raimondi
- Veneto Institute of Oncology IOV–IRCCS, 35128 Padova, Italy; (I.C.); (F.C.); (E.S.); (L.U.); (V.R.); (M.S.-B.)
| | - Micol Silic-Benussi
- Veneto Institute of Oncology IOV–IRCCS, 35128 Padova, Italy; (I.C.); (F.C.); (E.S.); (L.U.); (V.R.); (M.S.-B.)
| | - Donna M. D’Agostino
- Veneto Institute of Oncology IOV–IRCCS, 35128 Padova, Italy; (I.C.); (F.C.); (E.S.); (L.U.); (V.R.); (M.S.-B.)
- Department of Biomedical Sciences, University of Padua, 35131 Padova, Italy
| | - Vincenzo Ciminale
- Veneto Institute of Oncology IOV–IRCCS, 35128 Padova, Italy; (I.C.); (F.C.); (E.S.); (L.U.); (V.R.); (M.S.-B.)
- Department of Surgery, Oncology and Gastroenterology, University of Padua, 35128 Padova, Italy
| |
Collapse
|
15
|
Ma X, Ying Y, Sun J, Xie H, Li J, He L, Wang W, Chen S, Shen H, Yi J, Luo J, Wang X, Zheng X, Liu B, Xie L. circKDM4C enhances bladder cancer invasion and metastasis through miR-200bc-3p/ZEB1 axis. Cell Death Dis 2021; 7:365. [PMID: 34811353 PMCID: PMC8608878 DOI: 10.1038/s41420-021-00712-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 10/04/2021] [Accepted: 10/11/2021] [Indexed: 01/22/2023]
Abstract
Circular RNAs (circRNAs) play essential roles in human bladder cancer (BCa) development, however, unusual expression patterns and functional dysfunction of circRNAs in BCa have not been evaluated. In this study, we validated that circKDM4C (hsa_circ_0001839), derived from the KDM4C gene, is elevated in BCa cell lines as well as tissues. Functionally, overexpression of circKDM4C significantly enhances, and silencing of circKDM4C suppresses migration and invasion capabilities of BCa cells. Mechanistically, circKDM4C can directly interact with miR-200b-3p and miR-200c-3p as a miRNA sponge, which enhances the expression of ZEB1 and promotes mesenchymal phenotype. Conclusively, our findings indicate that circKDM4C may act as a pro-oncogenic factor in BCa invasion and metastasis via the circKDM4C/miR-200bc-3p/ZEB1 axis, which is a potential biomarker or therapeutic target for bladder cancer.
Collapse
Affiliation(s)
- Xueyou Ma
- grid.452661.20000 0004 1803 6319Department of Urology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003 China ,grid.13402.340000 0004 1759 700XCancer Center, Zhejiang University, Hangzhou, 310058 China
| | - Yufan Ying
- grid.452661.20000 0004 1803 6319Department of Urology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003 China ,grid.13402.340000 0004 1759 700XCancer Center, Zhejiang University, Hangzhou, 310058 China
| | - Jiazhu Sun
- grid.452661.20000 0004 1803 6319Department of Urology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003 China ,grid.13402.340000 0004 1759 700XCancer Center, Zhejiang University, Hangzhou, 310058 China
| | - Haiyun Xie
- grid.452661.20000 0004 1803 6319Department of Urology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003 China ,grid.13402.340000 0004 1759 700XCancer Center, Zhejiang University, Hangzhou, 310058 China
| | - Jiangfeng Li
- grid.452661.20000 0004 1803 6319Department of Urology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003 China ,grid.13402.340000 0004 1759 700XCancer Center, Zhejiang University, Hangzhou, 310058 China
| | - Liujia He
- grid.452661.20000 0004 1803 6319Department of Urology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003 China ,grid.13402.340000 0004 1759 700XCancer Center, Zhejiang University, Hangzhou, 310058 China
| | - Weiyu Wang
- grid.452661.20000 0004 1803 6319Department of Urology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003 China ,grid.13402.340000 0004 1759 700XCancer Center, Zhejiang University, Hangzhou, 310058 China
| | - Shiming Chen
- grid.452661.20000 0004 1803 6319Department of Urology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003 China ,grid.13402.340000 0004 1759 700XCancer Center, Zhejiang University, Hangzhou, 310058 China
| | - Haixiang Shen
- grid.452661.20000 0004 1803 6319Department of Urology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003 China ,grid.13402.340000 0004 1759 700XCancer Center, Zhejiang University, Hangzhou, 310058 China
| | - Jiahe Yi
- grid.452661.20000 0004 1803 6319Department of Urology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003 China ,grid.13402.340000 0004 1759 700XCancer Center, Zhejiang University, Hangzhou, 310058 China
| | - Jindan Luo
- grid.452661.20000 0004 1803 6319Department of Urology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003 China ,grid.13402.340000 0004 1759 700XCancer Center, Zhejiang University, Hangzhou, 310058 China
| | - Xiao Wang
- grid.452661.20000 0004 1803 6319Department of Urology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003 China ,grid.13402.340000 0004 1759 700XCancer Center, Zhejiang University, Hangzhou, 310058 China
| | - Xiangyi Zheng
- grid.452661.20000 0004 1803 6319Department of Urology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003 China ,grid.13402.340000 0004 1759 700XCancer Center, Zhejiang University, Hangzhou, 310058 China
| | - Ben Liu
- Department of Urology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China. .,Cancer Center, Zhejiang University, Hangzhou, 310058, China.
| | - Liping Xie
- Department of Urology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China. .,Cancer Center, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
16
|
Sindhu KJ, Venkatesan N, Karunagaran D. MicroRNA Interactome Multiomics Characterization for Cancer Research and Personalized Medicine: An Expert Review. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2021; 25:545-566. [PMID: 34448651 DOI: 10.1089/omi.2021.0087] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
MicroRNAs (miRNAs) that are mutually modulated by their interacting partners (interactome) are being increasingly noted for their significant role in pathogenesis and treatment of various human cancers. Recently, miRNA interactome dissected with multiomics approaches has been the subject of focus since individual tools or methods failed to provide the necessary comprehensive clues on the complete interactome. Even though single-omics technologies such as proteomics can uncover part of the interactome, the biological and clinical understanding still remain incomplete. In this study, we present an expert review of studies involving multiomics approaches to identification of miRNA interactome and its application in mechanistic characterization, classification, and therapeutic target identification in a variety of cancers, and with a focus on proteomics. We also discuss individual or multiple miRNA-based interactome identification in various pathological conditions of relevance to clinical medicine. Various new single-omics methods that can be integrated into multiomics cancer research and the computational approaches to analyze and predict miRNA interactome are also highlighted in this review. In all, we contextulize the power of multiomics approaches and the importance of the miRNA interactome to achieve the vision and practice of predictive, preventive, and personalized medicine in cancer research and clinical oncology.
Collapse
Affiliation(s)
- K J Sindhu
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, India
| | - Nalini Venkatesan
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, India
| | - Devarajan Karunagaran
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, India
| |
Collapse
|
17
|
Mandal S, Bandyopadhyay S, Tyagi K, Roy A. Recent advances in understanding the molecular role of phosphoinositide-specific phospholipase C gamma 1 as an emerging onco-driver and novel therapeutic target in human carcinogenesis. Biochim Biophys Acta Rev Cancer 2021; 1876:188619. [PMID: 34454048 DOI: 10.1016/j.bbcan.2021.188619] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/04/2021] [Accepted: 08/21/2021] [Indexed: 02/07/2023]
Abstract
Phosphoinositide metabolism is crucial intracellular signaling system that regulates a plethora of biological functions including mitogenesis, cell proliferation and division. Phospholipase C gamma 1 (PLCγ1) which belongs to phosphoinositide-specific phospholipase C (PLC) family, is activated by many extracellular stimuli including hormones, neurotransmitters, growth factors and modulates several cellular and physiological functions necessary for tumorigenesis such as cell survival, migration, invasion and angiogenesis by generating inositol 1,4,5-triphosphate (IP3) and diacylglycerol (DAG) via hydrolysis of phosphatidylinositol 4,5-biphosphate (PIP2). Cancer remains as a leading cause of global mortality and aberrant expression and regulation of PLCγ1 is linked to a plethora of deadly human cancers including carcinomas of the breast, lung, pancreas, stomach, prostate and ovary. Although PLCγ1 cross-talks with many onco-drivers and signaling circuits including PI3K, AKT, HIF1-α and RAF/MEK/ERK cascade, its precise role in carcinogenesis is not completely understood. This review comprehensively discussed the status quo of this ubiquitously expressed phospholipase as a tumor driver and highlighted its significance as a novel therapeutic target in cancer. Furthermore, we have highlighted the significance of somatic driver mutations in PLCG1 gene and molecular roles of PLCγ1 in several major human cancers, a knowledgebase that can be utilized to develop novel, isoform-specific small molecule inhibitors of PLCγ1.
Collapse
Affiliation(s)
- Supratim Mandal
- Department of Microbiology, University of Kalyani, Kalyani, Nadia, West Bengal 741235, India.
| | - Shrabasti Bandyopadhyay
- Department of Microbiology, University of Kalyani, Kalyani, Nadia, West Bengal 741235, India
| | - Komal Tyagi
- Amity Institute of Molecular Medicine & Stem Cell Research, Amity University, Sector 125, Noida, Uttar Pradesh 201303, India
| | - Adhiraj Roy
- Amity Institute of Molecular Medicine & Stem Cell Research, Amity University, Sector 125, Noida, Uttar Pradesh 201303, India.
| |
Collapse
|
18
|
Phospholipase Signaling in Breast Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021. [PMID: 33983572 DOI: 10.1007/978-981-32-9620-6_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/23/2023]
Abstract
Breast cancer progression results from subversion of multiple intra- or intercellular signaling pathways in normal mammary tissues and their microenvironment, which have an impact on cell differentiation, proliferation, migration, and angiogenesis. Phospholipases (PLC, PLD and PLA) are essential mediators of intra- and intercellular signaling. They hydrolyze phospholipids, which are major components of cell membrane that can generate many bioactive lipid mediators, such as diacylglycerol, phosphatidic acid, lysophosphatidic acid, and arachidonic acid. Enzymatic processing of phospholipids by phospholipases converts these molecules into lipid mediators that regulate multiple cellular processes, which in turn can promote breast cancer progression. Thus, dysregulation of phospholipases contributes to a number of human diseases, including cancer. This review describes how phospholipases regulate multiple cancer-associated cellular processes, and the interplay among different phospholipases in breast cancer. A thorough understanding of the breast cancer-associated signaling networks of phospholipases is necessary to determine whether these enzymes are potential targets for innovative therapeutic strategies.
Collapse
|
19
|
Rahimian N, Razavi ZS, Aslanbeigi F, Mirkhabbaz AM, Piroozmand H, Shahrzad MK, Hamblin MR, Mirzaei H. Non-coding RNAs related to angiogenesis in gynecological cancer. Gynecol Oncol 2021; 161:896-912. [PMID: 33781555 DOI: 10.1016/j.ygyno.2021.03.020] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 03/19/2021] [Indexed: 02/07/2023]
Abstract
Gynecological cancer affects the female reproductive system, including ovarian, uterine, endometrial, cervical, vulvar, and vaginal tumors. Non-coding RNAs (ncRNAs), and in particular microRNAs, function as regulatory molecules, which can control gene expression in a post-transcriptional manner. Normal physiological processes like cellular proliferation, differentiation, and apoptosis, and pathological processes such as oncogenesis and metastasis are regulated by microRNAs. Numerous reports have shown a direct role of microRNAs in the modulation of angiogenesis in gynecological cancer, via targeting pro-angiogenic factors and signaling pathways. Understanding the molecular mechanism involved in the regulation of angiogenesis by microRNAs may lead to new treatment options. Recently the regulatory role of some long non-coding RNAs in gynecological cancer has also been explored, but the information on this function is more limited. The aim of this article is to explore the pathways responsible for angiogenesis, and to what extent ncRNAs may be employed as biomarkers or therapeutic targets in gynecological cancer.
Collapse
Affiliation(s)
- Neda Rahimian
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | | | | | | | - Haleh Piroozmand
- Faculty of Veterinary Sciences, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Mohammad Karim Shahrzad
- Department of Internal Medicine and endocrinology, Shohadae Tajrish Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa.
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
20
|
Fontana A, Barbano R, Dama E, Pasculli B, Rendina M, Morritti MG, Melocchi V, Castelvetere M, Valori VM, Ravaioli S, Bravaccini S, Ciuffreda L, Graziano P, Maiello E, Copetti M, Fazio VM, Esteller M, Bianchi F, Parrella P. Combined analysis of miR-200 family and its significance for breast cancer. Sci Rep 2021; 11:2980. [PMID: 33536459 PMCID: PMC7859396 DOI: 10.1038/s41598-021-82286-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 01/18/2021] [Indexed: 12/30/2022] Open
Abstract
While the molecular functions of miR-200 family have been deeply investigated, a role for these miRNAs as breast cancer biomarkers remains largely unexplored. In the attempt to clarify this, we profiled the miR-200 family members expression in a large cohort of breast cancer cases with a long follow-up (H-CSS cohort) and in TCGA-BRCA cohort. Overall, miR-200 family was found upregulated in breast tumors with respect to normal breast tissues while downregulated in more aggressive breast cancer molecular subtypes (i.e. Luminal B, HER2 and triple negative), consistently with their function as repressors of the epithelial-to-mesenchymal transition (EMT). In particular miR-141-3p was found differentially expressed in breast cancer molecular subtypes in both H-CSS and TCGA-BRCA cohorts, and the combined analysis of all miR-200 family members demonstrated a slight predictive accuracy on H-CSS cancer specific survival at 12 years (survival c-statistic: 0.646; 95%CI 0.538–0.754).
Collapse
Affiliation(s)
- Andrea Fontana
- Fondazione IRCCS Casa Sollievo della Sofferenza, UO di Biostatistica, San Giovanni Rotondo, FG, Italy
| | - Raffaela Barbano
- Laboratorio Di Oncologia, Fondazione IRCCS Casa Sollievo della Sofferenza, Viale Padre Pio, 71013, San Giovanni Rotondo, FG, Italy
| | - Elisa Dama
- Cancer Biomarkers Lab, ISBREMIT, Institute for Stem-Cell Biology, Regenerative Medicine and Innovative Therapies, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, FG, Italy
| | - Barbara Pasculli
- Laboratorio Di Oncologia, Fondazione IRCCS Casa Sollievo della Sofferenza, Viale Padre Pio, 71013, San Giovanni Rotondo, FG, Italy
| | - Michelina Rendina
- Laboratorio Di Oncologia, Fondazione IRCCS Casa Sollievo della Sofferenza, Viale Padre Pio, 71013, San Giovanni Rotondo, FG, Italy
| | - Maria Grazia Morritti
- Fondazione IRCCS Casa Sollievo della Sofferenza, UO di Oncologia, San Giovanni Rotondo, FG, Italy
| | - Valentina Melocchi
- Cancer Biomarkers Lab, ISBREMIT, Institute for Stem-Cell Biology, Regenerative Medicine and Innovative Therapies, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, FG, Italy
| | - Marina Castelvetere
- Fondazione IRCCS Casa Sollievo della Sofferenza, UO di Anatomia Patologica, San Giovanni Rotondo, FG, Italy
| | - Vanna Maria Valori
- Fondazione IRCCS Casa Sollievo della Sofferenza, UO di Oncologia, San Giovanni Rotondo, FG, Italy
| | - Sara Ravaioli
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Sara Bravaccini
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Luigi Ciuffreda
- Fondazione IRCCS Casa Sollievo della Sofferenza, UO di Chirurgia Senologica, San Giovanni Rotondo, FG, Italy
| | - Paolo Graziano
- Fondazione IRCCS Casa Sollievo della Sofferenza, UO di Anatomia Patologica, San Giovanni Rotondo, FG, Italy
| | - Evaristo Maiello
- Fondazione IRCCS Casa Sollievo della Sofferenza, UO di Oncologia, San Giovanni Rotondo, FG, Italy
| | - Massimiliano Copetti
- Fondazione IRCCS Casa Sollievo della Sofferenza, UO di Biostatistica, San Giovanni Rotondo, FG, Italy
| | - Vito Michele Fazio
- Laboratorio Di Oncologia, Fondazione IRCCS Casa Sollievo della Sofferenza, Viale Padre Pio, 71013, San Giovanni Rotondo, FG, Italy
| | - Manel Esteller
- Josep Carreras Leukaemia Research Institute (IJC), Badalona, Barcelona, Catalonia, Spain.,Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain.,Physiological Sciences Department, School of Medicine and Health Sciences, University of Barcelona, Barcelona, Catalonia, Spain.,Institució Catalana de Recerca I Estudis Avançats (ICREA), Barcelona, Catalonia, Spain
| | - Fabrizio Bianchi
- Cancer Biomarkers Lab, ISBREMIT, Institute for Stem-Cell Biology, Regenerative Medicine and Innovative Therapies, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, FG, Italy
| | - Paola Parrella
- Laboratorio Di Oncologia, Fondazione IRCCS Casa Sollievo della Sofferenza, Viale Padre Pio, 71013, San Giovanni Rotondo, FG, Italy.
| |
Collapse
|
21
|
Abstract
Despite the decline in death rate from breast cancer and recent advances in targeted therapies and combinations for the treatment of metastatic disease, metastatic breast cancer remains the second leading cause of cancer-associated death in U.S. women. The invasion-metastasis cascade involves a number of steps and multitudes of proteins and signaling molecules. The pathways include invasion, intravasation, circulation, extravasation, infiltration into a distant site to form a metastatic niche, and micrometastasis formation in a new environment. Each of these processes is regulated by changes in gene expression. Noncoding RNAs including microRNAs (miRNAs) are involved in breast cancer tumorigenesis, progression, and metastasis by post-transcriptional regulation of target gene expression. miRNAs can stimulate oncogenesis (oncomiRs), inhibit tumor growth (tumor suppressors or miRsupps), and regulate gene targets in metastasis (metastamiRs). The goal of this review is to summarize some of the key miRNAs that regulate genes and pathways involved in metastatic breast cancer with an emphasis on estrogen receptor α (ERα+) breast cancer. We reviewed the identity, regulation, human breast tumor expression, and reported prognostic significance of miRNAs that have been documented to directly target key genes in pathways, including epithelial-to-mesenchymal transition (EMT) contributing to the metastatic cascade. We critically evaluated the evidence for metastamiRs and their targets and miRNA regulation of metastasis suppressor genes in breast cancer progression and metastasis. It is clear that our understanding of miRNA regulation of targets in metastasis is incomplete.
Collapse
Affiliation(s)
- Belinda J Petri
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY, 40292, USA
| | - Carolyn M Klinge
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY, 40292, USA.
| |
Collapse
|
22
|
Mishra N, Raina K, Agarwal R. Deciphering the role of microRNAs in mustard gas-induced toxicity. Ann N Y Acad Sci 2020; 1491:25-41. [PMID: 33305460 DOI: 10.1111/nyas.14539] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 10/26/2020] [Accepted: 11/01/2020] [Indexed: 12/16/2022]
Abstract
Mustard gas (sulfur mustard, SM), a highly vesicating chemical warfare agent, was first deployed in warfare in 1917 and recently during the Iraq-Iran war (1980s) and Syrian conflicts (2000s); however, the threat of exposure from stockpiles and old artillery shells still looms large. Whereas research has been long ongoing on SM-induced toxicity, delineating the precise molecular pathways is still an ongoing area of investigation; thus, it is important to attempt novel approaches to decipher these mechanisms and develop a detailed network of pathways associated with SM-induced toxicity. One such avenue is exploring the role of microRNAs (miRNAs) in SM-induced toxicity. Recent research on the regulatory role of miRNAs provides important results to fill in the gaps in SM toxicity-associated mechanisms. In addition, differentially expressed miRNAs can also be used as diagnostic markers to determine the extent of toxicity in exposed individuals. Thus, in our review, we have summarized the studies conducted so far in cellular and animal models, including human subjects, on the expression profiles and roles of miRNAs in SM- and/or SM analog-induced toxicity. Further detailed research in this area will guide us in devising preventive strategies, diagnostic tools, and therapeutic interventions against SM-induced toxicity.
Collapse
Affiliation(s)
- Neha Mishra
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado-Anschutz Medical Campus, Aurora, Colorado
| | - Komal Raina
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado-Anschutz Medical Campus, Aurora, Colorado.,Department of Pharmaceutical Sciences, South Dakota State University, Brookings, South Dakota
| | - Rajesh Agarwal
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado-Anschutz Medical Campus, Aurora, Colorado
| |
Collapse
|
23
|
Li Y, Li L. Bioinformatic screening for candidate biomarkers and their prognostic values in endometrial cancer. BMC Genet 2020; 21:113. [PMID: 32962636 PMCID: PMC7510080 DOI: 10.1186/s12863-020-00898-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Accepted: 08/11/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Endometrial cancer is a common gynecological cancer with annually increasing incidence worldwide. However, the biomarkers that provide prognosis and progression for this disease remain elusive. RESULTS Two eligible human endometrial cancer datasets (GSE17025 and GSE25405) were selected for the study. A total of 520 differentially expressed mRNAs and 30 differentially expressed miRNAs were identified. These mRNAs were mainly enriched in cell cycle, skeletal system development, vasculature development, oocyte maturation, and oocyte meiosis signalling pathways. A total of 160 pairs of differentially expressed miRNAs and mRNAs, including 22 differentially expressed miRNAs and 71 overlapping differentially expressed mRNAs, were validated in endometrial cancer samples using starBase v2.0 project. The prognosis analysis revealed that Cyclin E1 (CCNE1, one of the 82 hub genes, which correlated with hsa-miR-195 and hsa-miR-424) was significantly linked to a worse overall survival in endometrial cancer patients. CONCLUSIONS The hub genes and differentially expressed miRNAs identified in this study might be used as prognostic biomarkers for endometrial cancer and molecular targets for its treatment.
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, 530021, People's Republic of China.,Department of Gynecology and obstetrics, Shangyu People's Hospital, Shangyu, Zhejiang, 312300, People's Republic of 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, 530021, People's Republic of China.
| |
Collapse
|
24
|
Bozgeyik E, Tepe NB, Bozdag Z. Identification of microRNA expression signature for the diagnosis and prognosis of cervical squamous cell carcinoma. Pathol Res Pract 2020; 216:153159. [PMID: 32841775 DOI: 10.1016/j.prp.2020.153159] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 07/29/2020] [Accepted: 08/01/2020] [Indexed: 01/08/2023]
Abstract
Cervical cancer is the fourth leading cause of cancer death among women globally. The prognosis of cervical cancer patients differs considerably, and clinical outcomes are difficult to predict. Given the significant roles of miRNAs in human cancers, identification of novel and reliable miRNA biomarkers is important for targeted cervical cancer therapy. In the present study, we aimed to reveal biological significance of miR-200a, miR-423, miR-34a, miR-193a, and miR-455 for the prognosis and diagnosis of cervical cancer and their association with the clinical outcomes of patients. Distinct expression profiles of miRNAs in formalin-fixed paraffin-embedded tissue samples of patients and healthy controls were evaluated using qRT-PCR. We identified miR-200a, miR-455, and miR-34a were significantly downregulated in cervical squamous cell carcinoma tissues compared to normal cervix tissue from healthy controls. Both miR-455 and miR-34a confer a promising diagnostic factor for the cervical cancer while miR-200a showed no significance in ROC analysis. Notably, low expression of miR-34a was markedly associated with the poor overall survival of cervical cancer patients as revealed by Kaplan-Meier survival analysis. Also, univariate and multivariate analysis indicated miR-34a expression as an independent prognostic factor. Consequently, our results underline the importance of distinct expression miRNAs in cervical squamous cell carcinoma.
Collapse
Affiliation(s)
- Esra Bozgeyik
- Department of Medical Services and Techniques, Vocational School of Health Services, Adiyaman University, Adiyaman, Turkey.
| | | | - Zehra Bozdag
- Department of Pathology, University of Gaziantep, Gaziantep, Turkey
| |
Collapse
|
25
|
Ghazi T, Nagiah S, Dhani S, Chuturgoon AA. Fusaric acid-induced epigenetic modulation of hepatic H3K9me3 triggers apoptosis in vitro and in vivo. Epigenomics 2020; 12:955-972. [PMID: 32762452 DOI: 10.2217/epi-2019-0284] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Aim: To determine the effect of the food-borne mycotoxin, fusaric acid (FA) on miR-200a, SUV39H1-mediated H3K9me3, genome integrity and apoptosis in human liver (HepG2) cells and C57BL/6 mice livers. Materials & methods: MiR-200a, Sirt1, SUV39H1-mediated H3K9me3, genome integrity and apoptosis was measured in HepG2 cells and C57BL/6 mice livers using qPCR, western blot, DNA electrophoresis and luminometry. Results: FA: upregulated miR-200a and decreased Sirt1 expression in HepG2 cells and mice livers; decreased expression of SUV39H1 and KDM4B, thus decreasing H3K9me3 and increasing H3K9me1; increased cell mortality via apoptosis. Conclusion: FA induced apoptosis by upregulating miR-200a and decreasing SUV39H1-mediated H3K9me3 in HepG2 cells and mice livers.
Collapse
Affiliation(s)
- Terisha Ghazi
- Discipline of Medical Biochemistry & Chemical Pathology, School of Laboratory Medicine & Medical Science, College of Health Sciences, Howard College Campus, University of KwaZulu-Natal, Durban 4041, South Africa
| | - Savania Nagiah
- Discipline of Medical Biochemistry & Chemical Pathology, School of Laboratory Medicine & Medical Science, College of Health Sciences, Howard College Campus, University of KwaZulu-Natal, Durban 4041, South Africa
| | - Shanel Dhani
- Discipline of Medical Biochemistry & Chemical Pathology, School of Laboratory Medicine & Medical Science, College of Health Sciences, Howard College Campus, University of KwaZulu-Natal, Durban 4041, South Africa
| | - Anil A Chuturgoon
- Discipline of Medical Biochemistry & Chemical Pathology, School of Laboratory Medicine & Medical Science, College of Health Sciences, Howard College Campus, University of KwaZulu-Natal, Durban 4041, South Africa
| |
Collapse
|
26
|
Khalife H, Skafi N, Fayyad-Kazan M, Badran B. MicroRNAs in breast cancer: New maestros defining the melody. Cancer Genet 2020; 246-247:18-40. [PMID: 32805688 DOI: 10.1016/j.cancergen.2020.08.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 07/07/2020] [Accepted: 08/03/2020] [Indexed: 02/06/2023]
Abstract
MicroRNAs, short non-coding single-stranded RNAs, are important regulators and gatekeepers of the coding genes in the human genome. MicroRNAs are highly conserved among species and expressed in different tissues and cell types. They are involved in almost all the biological processes as apoptosis, proliferation, cell cycle arrest and differentiation. Playing all these roles, it is not surprising that the deregulation of the microRNA profile causes a number of diseases including cancer. Breast cancer, the most commonly diagnosed malignancy in women, accounts for the highest cancer-related deaths worldwide. Different microRNAs were shown to be up or down regulated in breast cancer. MicroRNAs can function as oncogenes or tumor suppressors according to their targets. In this review, the most common microRNAs implicated in breast cancer are fully illustrated with their targets. Besides, the review highlights the effect of exosomal microRNA on breast cancer and the effect of microRNAs on drug and therapies resistance as well as the miRNA-based therapeutic strategies used until today.
Collapse
Affiliation(s)
- Hoda Khalife
- Laboratory of Cancer biology and Molecular Immunology, Faculty of Sciences-I, Lebanese University, Hadath, Beirut, Lebanon.
| | - Najwa Skafi
- Laboratory of Cancer biology and Molecular Immunology, Faculty of Sciences-I, Lebanese University, Hadath, Beirut, Lebanon.
| | - Mohammad Fayyad-Kazan
- Laboratory of Cancer biology and Molecular Immunology, Faculty of Sciences-I, Lebanese University, Hadath, Beirut, Lebanon; Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut, Lebanon.
| | - Bassam Badran
- Laboratory of Cancer biology and Molecular Immunology, Faculty of Sciences-I, Lebanese University, Hadath, Beirut, Lebanon.
| |
Collapse
|
27
|
Gao S, Duan H, An D, Yi X, Li J, Liao C. Knockdown of long non-coding RNA LINC00467 inhibits glioma cell progression via modulation of E2F3 targeted by miR-200a. Cell Cycle 2020; 19:2040-2053. [PMID: 32684096 PMCID: PMC7469466 DOI: 10.1080/15384101.2020.1792127] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 11/28/2019] [Indexed: 12/19/2022] Open
Abstract
Studies have found that LINC00467 is an important regulator of cancer. However, the function of LINC00467 in glioma cell is unclear. Therefore, this experimental design based on LINC00467 to explore its mechanism of action in glioma cell. RT-qPCR was used to detect the expression of LINC0046 and miR-200a in glioma cell lines. MTT assay, Edru assay and Transwell assay and flow cytometry were used to detect the effects of LINC0046 and miR-200a on PC cell proliferation, migration and apoptosis. Target gene prediction and screening, luciferase reporter assays were used to validate downstream target genes for LINC0046 and miR-200a. Western blotting was used to detect the protein expression of E2F3. The tumor changes in mice were detected by in vivo experiments in nude mice. LINC00467 was up-regulated in glioma cells. Knockdown of LINC00467 inhibited the viability, migration and invasion of glioma cells. In glioma cells, miR-200a was significantly reduced, while E2F3 was significantly rised. LINC00467 negatively regulated the expression of miR-200a in gliomas, while miR-200a negatively regulated the expression of E2F3 in gliomas. INC00467 promoted the development of glioma by inhibiting miR-200a and promoting E2F3 expression. LINC00467 may be a potential therapeutic target for gliomas.
Collapse
Affiliation(s)
- Shuzi Gao
- Department of Cerebrovascular Diseases, Zhuhai People’s Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai City, Guangdong Province, PR. China
| | - Haixia Duan
- Department of Ophthalmology, Zhuhai Hospital of Integrated Traditional Chinese and Western Medicine, Zhuhai City, Guangdong Province, PR. China
| | - Dezhu An
- Department of Neurosurgery, Zhuhai People’s Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai City, Guangdong Province, PR. China
| | - Xinfeng Yi
- Department of Neurosurgery, Zhuhai People’s Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai City, Guangdong Province, PR. China
| | - Jiayan Li
- Department of Neurosurgery, Zhuhai People’s Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai City, Guangdong Province, PR. China
| | - Changchun Liao
- Department of Neurosurgery, Zhuhai People’s Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai City, Guangdong Province, PR. China
| |
Collapse
|
28
|
Abdalla F, Singh B, Bhat HK. MicroRNAs and gene regulation in breast cancer. J Biochem Mol Toxicol 2020; 34:e22567. [DOI: 10.1002/jbt.22567] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 06/01/2020] [Accepted: 06/18/2020] [Indexed: 12/15/2022]
Affiliation(s)
- Fatma Abdalla
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy University of Missouri‐Kansas City Kansas City Missouri
| | - Bhupendra Singh
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy University of Missouri‐Kansas City Kansas City Missouri
- Eurofins Lancaster Laboratories Lancaster PA 17605
| | - Hari K. Bhat
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy University of Missouri‐Kansas City Kansas City Missouri
| |
Collapse
|
29
|
Abdoli Shadbad M, Hajiasgharzadeh K, Baradaran B. Cross-talk between myeloid-derived suppressor cells and Mucin1 in breast cancer vaccination: On the verge of a breakthrough. Life Sci 2020; 258:118128. [PMID: 32710947 DOI: 10.1016/j.lfs.2020.118128] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 07/16/2020] [Accepted: 07/17/2020] [Indexed: 01/22/2023]
Abstract
Although breast cancer is one of the leading troublesome cancers, the available therapeutic options have not fulfilled the desired outcomes. Immune-based therapy has gained special attention for breast cancer treatment. Although this approach is highly tolerable, its low response rate has rendered it as an undesirable approach. This review aims to describe the essential oncogenic pathways involved in breast cancer, elucidate the immunosuppression and oncogenic effect of Mucin1, and introduce myeloid-derived suppressor cells, which are the main culprits of anti-tumoral immune response attenuation. The various auto-inductive loops between Mucin1 and myeloid-derived suppressor cells are focal in the suppression of anti-tumoral immune responses in patients with breast cancer. These cross-talks between the Mucin1 and myeloid-derived suppressor cells can be the underlying causes of immunotherapy's impotence for patients with breast cancer. This approach can pave the road for the development of a potent vaccine for patients with breast cancer and is translated into clinical settings.
Collapse
Affiliation(s)
| | - Khalil Hajiasgharzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Connective Tissue Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Pharmaceutical Analysis Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
30
|
Çınar Ayan İ, Çetinkaya S, Dursun HG, Süntar İ. Bioactive Compounds of Rheum ribes L. and its Anticancerogenic Effect via Induction of Apoptosis and miR-200 Family Expression in Human Colorectal Cancer Cells. Nutr Cancer 2020; 73:1228-1243. [DOI: 10.1080/01635581.2020.1792947] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- İlknur Çınar Ayan
- Department of Medical Biology, Medical Faculty, Necmettin Erbakan University, Meram, Konya, Turkey
| | - Sümeyra Çetinkaya
- Biotechnology Research Center of Ministry of Agriculture and Forestry, Yenimahalle, Ankara, Turkey
| | - Hatice Gül Dursun
- Department of Medical Biology, Medical Faculty, Necmettin Erbakan University, Meram, Konya, Turkey
| | - İpek Süntar
- Department of Pharmacognosy Faculty of Pharmacy, Gazi University, Etiler, Ankara, Turkey
| |
Collapse
|
31
|
Nabih HK. Crosstalk between NRF2 and Dicer through metastasis regulating MicroRNAs; mir-34a, mir-200 family and mir-103/107 family. Arch Biochem Biophys 2020; 686:108326. [DOI: 10.1016/j.abb.2020.108326] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 02/27/2020] [Accepted: 03/01/2020] [Indexed: 12/16/2022]
|
32
|
Cao Q, Wang N, Ren L, Tian J, Yang S, Cheng H. miR-125a-5p post-transcriptionally suppresses GALNT7 to inhibit proliferation and invasion in cervical cancer cells via the EGFR/PI3K/AKT pathway. Cancer Cell Int 2020; 20:117. [PMID: 32308562 PMCID: PMC7147043 DOI: 10.1186/s12935-020-01209-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 04/06/2020] [Indexed: 02/08/2023] Open
Abstract
Background The carcinogenesis and progression of cervical cancer is a complex process in which numerous microRNAs are involved. The purpose of this study is to investigate the role of miR-125a-5p in progression of cervical cancer. Methods RT-qPCR was used to detect the expression of miR-125a-5p and GALNT7 in cervical cancer tissues and cell lines. Then, the miR-125a-5p mimic, miR-125a-5p inhibitor, GALNT7 siRNA, or/and pcDNA-GALNT7 were respectively transfected into HeLa and Caski cervical cancer cells, and Cell Counting kit-8 assay, Transwell assay and flow cytometry analysis were respectively used to observe cell proliferation, invasion and apoptosis. Subsequently, luciferase reporter gene assay was employed in confirming the target relationship between miR-125a-5p and GALNT7. MiR-125a-5p mimic or/and pcDNA-GALNT7 were transfected into the cervical cancer cells at the absence of epidermal growth factor (EGF) or not, and the pcDNA-GALNT7 was transfected into the cervical cancer cells at the absence of inhibitors of multiple kinases or not. Furthermore, the effect of miR-125a-5p on tumor growth was also studied using a xenograft model of nude mice. Results MiR-125a-5p was down-regulated in both cervical cancer tissues and cell lines and it inhibited cell proliferation and invasion of cervical cancer cells. MiR-125a-5p directly targeted and post-transcriptionally downregulated GALNT7 that was strongly upregulated in cervical cancer tissues and cell lines. Similar to the effect of miR-125a-5p mimic, silencing GALNT7 inhibited proliferation and invasion of cervical cancer cells. In addition, miR-125a-5p overexpression could counteract both GALNT7- and EGF-induced cell proliferation and invasion. GALNT7 promoted cell proliferation and invasion by activating the EGFR/PI3K/AKT kinase pathway, which could be abated by the inhibitors of the kinases. Moreover, the role of miR-125a-5p inhibited tumor formation in cervical cancer by suppressing the expression of GALNT7 in vivo. Conclusion In conclusion, miR-125a-5p suppressed cervical cancer progression by post-transcriptionally downregulating GALNT7 and inactivating the EGFR/PI3K/AKT pathway.
Collapse
Affiliation(s)
- Qinxue Cao
- Department Gynecology, Huaihe Hospital of Henan University, No.8 Baobei Road, Kaifeng, 475000 Henan Province China
| | - Ning Wang
- Department Gynecology, Huaihe Hospital of Henan University, No.8 Baobei Road, Kaifeng, 475000 Henan Province China
| | - Lu Ren
- Department Gynecology, Huaihe Hospital of Henan University, No.8 Baobei Road, Kaifeng, 475000 Henan Province China
| | - Jun Tian
- Department Gynecology, Huaihe Hospital of Henan University, No.8 Baobei Road, Kaifeng, 475000 Henan Province China
| | - Shaoqin Yang
- Department Gynecology, Huaihe Hospital of Henan University, No.8 Baobei Road, Kaifeng, 475000 Henan Province China
| | - Hailing Cheng
- Department Gynecology, Huaihe Hospital of Henan University, No.8 Baobei Road, Kaifeng, 475000 Henan Province China
| |
Collapse
|
33
|
Tfaily MA, Nassar F, Sellam LS, Amir-Tidadini ZC, Asselah F, Bourouba M, Rihab N. miRNA expression in advanced Algerian breast cancer tissues. PLoS One 2020; 15:e0227928. [PMID: 32040529 PMCID: PMC7010257 DOI: 10.1371/journal.pone.0227928] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 01/02/2020] [Indexed: 11/18/2022] Open
Abstract
Breast cancer is one of the commonest cancers among Algerian females. Compared to Western populations, the median age of diagnosis of breast cancer is much lower in Algeria. The objective of this study is to explore the expression of several miRNAs reported to be deregulated in breast cancer. The miRNAs miR-21, miR-125b, miR-100, miR-425-5p, miR-200c, miR-183 and miR-182 were studied on tumor and normal adjacent Algerian breast tissues using quantitative reverse transcription real time PCR, and the results were analyzed according to clinical characteristics. Compared to the normal adjacent tissues, miR-21, miR-183, miR-182, miR-425-5p and miR-200c were found to be upregulated while miR-100 and miR-125b were insignificantly deregulated. A positive correlation was noted among miR-183, miR-182 and miR-200c and among miR-425-5p, miR-183, miR-200c and miR-21. Further global miRNA microarray profiling studies can aid in finding ethnic specific miRNA biomarkers in the Algerian breast cancer population.
Collapse
Affiliation(s)
- Mohamad Ali Tfaily
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Farah Nassar
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Leila-Sarah Sellam
- Department of Cell and Molecular Biology, Team Cytokines and Nitric oxide synthases, Faculty of Biology, University of Sciences and Technology Houari Boumediene USTHB, Algiers, Algeria
| | | | - Fatima Asselah
- Central Laboratory for Anatomopathology, Mustapha Pacha Hospital, Algiers, Algeria
| | - Mehdi Bourouba
- Department of Cell and Molecular Biology, Team Cytokines and Nitric oxide synthases, Faculty of Biology, University of Sciences and Technology Houari Boumediene USTHB, Algiers, Algeria
- * E-mail: (RN); (MB)
| | - Nasr Rihab
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- * E-mail: (RN); (MB)
| |
Collapse
|
34
|
Pillman KA, Scheer KG, Hackett-Jones E, Saunders K, Bert AG, Toubia J, Whitfield HJ, Sapkota S, Sourdin L, Pham H, Le TD, Cursons J, Davis MJ, Gregory PA, Goodall GJ, Bracken CP. Extensive transcriptional responses are co-ordinated by microRNAs as revealed by Exon-Intron Split Analysis (EISA). Nucleic Acids Res 2019; 47:8606-8619. [PMID: 31372646 PMCID: PMC6895270 DOI: 10.1093/nar/gkz664] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 07/16/2019] [Accepted: 07/30/2019] [Indexed: 12/29/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) has been a subject of intense scrutiny as it facilitates metastasis and alters drug sensitivity. Although EMT-regulatory roles for numerous miRNAs and transcription factors are known, their functions can be difficult to disentangle, in part due to the difficulty in identifying direct miRNA targets from complex datasets and in deciding how to incorporate 'indirect' miRNA effects that may, or may not, represent biologically relevant information. To better understand how miRNAs exert effects throughout the transcriptome during EMT, we employed Exon-Intron Split Analysis (EISA), a bioinformatic technique that separates transcriptional and post-transcriptional effects through the separate analysis of RNA-Seq reads mapping to exons and introns. We find that in response to the manipulation of miRNAs, a major effect on gene expression is transcriptional. We also find extensive co-ordination of transcriptional and post-transcriptional regulatory mechanisms during both EMT and mesenchymal to epithelial transition (MET) in response to TGF-β or miR-200c respectively. The prominent transcriptional influence of miRNAs was also observed in other datasets where miRNA levels were perturbed. This work cautions against a narrow approach that is limited to the analysis of direct targets, and demonstrates the utility of EISA to examine complex regulatory networks involving both transcriptional and post-transcriptional mechanisms.
Collapse
Affiliation(s)
- Katherine A Pillman
- Centre for Cancer Biology, an alliance of SA Pathology and University of South Australia, Adelaide, SA, Australia.,ACRF Cancer Genomics Facility, Centre for Cancer Biology, SA Pathology, Adelaide, Australia
| | - Kaitlin G Scheer
- Centre for Cancer Biology, an alliance of SA Pathology and University of South Australia, Adelaide, SA, Australia
| | - Emily Hackett-Jones
- Centre for Cancer Biology, an alliance of SA Pathology and University of South Australia, Adelaide, SA, Australia
| | - Klay Saunders
- Centre for Cancer Biology, an alliance of SA Pathology and University of South Australia, Adelaide, SA, Australia
| | - Andrew G Bert
- Centre for Cancer Biology, an alliance of SA Pathology and University of South Australia, Adelaide, SA, Australia
| | - John Toubia
- Centre for Cancer Biology, an alliance of SA Pathology and University of South Australia, Adelaide, SA, Australia.,ACRF Cancer Genomics Facility, Centre for Cancer Biology, SA Pathology, Adelaide, Australia
| | - Holly J Whitfield
- Bioinformatics Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Sunil Sapkota
- Centre for Cancer Biology, an alliance of SA Pathology and University of South Australia, Adelaide, SA, Australia
| | - Laura Sourdin
- Centre for Cancer Biology, an alliance of SA Pathology and University of South Australia, Adelaide, SA, Australia
| | - Hoang Pham
- School of Information Technology and Mathematical Sciences, University of South Australia, Mawson Lakes, SA, Australia
| | - Thuc D Le
- School of Information Technology and Mathematical Sciences, University of South Australia, Mawson Lakes, SA, Australia
| | - Joseph Cursons
- School of Information Technology and Mathematical Sciences, University of South Australia, Mawson Lakes, SA, Australia.,Department of Medical Biology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria, Australia
| | - Melissa J Davis
- School of Information Technology and Mathematical Sciences, University of South Australia, Mawson Lakes, SA, Australia.,Department of Medical Biology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria, Australia.,Department of Biochemistry, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria, Australia
| | - Philip A Gregory
- Centre for Cancer Biology, an alliance of SA Pathology and University of South Australia, Adelaide, SA, Australia.,School of Medicine, Discipline of Medicine, University of Adelaide, SA, Australia
| | - Gregory J Goodall
- Centre for Cancer Biology, an alliance of SA Pathology and University of South Australia, Adelaide, SA, Australia.,School of Medicine, Discipline of Medicine, University of Adelaide, SA, Australia
| | - Cameron P Bracken
- Centre for Cancer Biology, an alliance of SA Pathology and University of South Australia, Adelaide, SA, Australia.,School of Medicine, Discipline of Medicine, University of Adelaide, SA, Australia
| |
Collapse
|
35
|
MiR-200 family and cancer: From a meta-analysis view. Mol Aspects Med 2019; 70:57-71. [DOI: 10.1016/j.mam.2019.09.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 09/12/2019] [Accepted: 09/17/2019] [Indexed: 12/20/2022]
|
36
|
Gilyazova IR, Klimentova EA, Bulygin KV, Izmailov AA, Bermisheva MA, Galimova EF, Safiullin RI, Galimov SN, Pavlov VN, Khusnutdinova EK. MicroRNA-200 family expression analysis in metastatic clear cell renal cell carcinoma patients. Cancer Gene Ther 2019; 27:768-772. [PMID: 31680118 DOI: 10.1038/s41417-019-0149-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 10/10/2019] [Accepted: 10/16/2019] [Indexed: 12/13/2022]
Abstract
The aim of this study is to analyse the of expression levels of microRNA-200 family members in patients with metastatic clear cell renal cell carcinoma (ccRCC). Analysis of microRNA expression was performed on 23 paired DNA samples extracted from kidney tumour tissue and the surrounding normal renal parenchyma. MicroRna-200c was found to have significantly lower expression (in kidney tumour tissue compared to normal renal parenchyma. No other microRna-200 family members showed statistically significant differences in expression levels between tumour and normal kidney tissue. Recent data suggest that the role of microRNA-200c in tumour pathogenesis is rather contradictory, and the underlying mechanisms by which microRNA-200c affects the carcinogenic potential of malignant cells remains unclear and requires further investigation at the molecular level.
Collapse
Affiliation(s)
- Irina R Gilyazova
- Institute of Biochemistry and Genetics, Ufa Federal Research Center of the Russian Academy of Sciences, Ufa, Russian Federation.,Bashkir State Medical University, Ufa, Russian Federation
| | - Elizaveta A Klimentova
- Institute of Biochemistry and Genetics, Ufa Federal Research Center of the Russian Academy of Sciences, Ufa, Russian Federation
| | - Kirill V Bulygin
- Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russian Federation. .,M.V. Lomonosov Moscow State University, Moscow, Russian Federation.
| | | | - Marina A Bermisheva
- Institute of Biochemistry and Genetics, Ufa Federal Research Center of the Russian Academy of Sciences, Ufa, Russian Federation
| | | | | | | | | | - Elsa K Khusnutdinova
- Institute of Biochemistry and Genetics, Ufa Federal Research Center of the Russian Academy of Sciences, Ufa, Russian Federation.,Bashkir State Medical University, Ufa, Russian Federation
| |
Collapse
|
37
|
Jin W, Mulas F, Gaertner B, Sui Y, Wang J, Matta I, Zeng C, Vinckier N, Wang A, Nguyen-Ngoc KV, Chiou J, Kaestner KH, Frazer KA, Carrano AC, Shih HP, Sander M. A Network of microRNAs Acts to Promote Cell Cycle Exit and Differentiation of Human Pancreatic Endocrine Cells. iScience 2019; 21:681-694. [PMID: 31733514 PMCID: PMC6889369 DOI: 10.1016/j.isci.2019.10.063] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 09/30/2019] [Accepted: 10/28/2019] [Indexed: 12/12/2022] Open
Abstract
Pancreatic endocrine cell differentiation is orchestrated by the action of transcription factors that operate in a gene regulatory network to activate endocrine lineage genes and repress lineage-inappropriate genes. MicroRNAs (miRNAs) are important modulators of gene expression, yet their role in endocrine cell differentiation has not been systematically explored. Here we characterize miRNA-regulatory networks active in human endocrine cell differentiation by combining small RNA sequencing, miRNA over-expression, and network modeling approaches. Our analysis identified Let-7g, Let-7a, miR-200a, miR-127, and miR-375 as endocrine-enriched miRNAs that drive endocrine cell differentiation-associated gene expression changes. These miRNAs are predicted to target different transcription factors, which converge on genes involved in cell cycle regulation. When expressed in human embryonic stem cell-derived pancreatic progenitors, these miRNAs induce cell cycle exit and promote endocrine cell differentiation. Our study delineates the role of miRNAs in human endocrine cell differentiation and identifies miRNAs that could facilitate endocrine cell reprogramming.
Collapse
Affiliation(s)
- Wen Jin
- Departments of Pediatrics and Cellular & Molecular Medicine, Pediatric Diabetes Research Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Francesca Mulas
- Departments of Pediatrics and Cellular & Molecular Medicine, Pediatric Diabetes Research Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Bjoern Gaertner
- Departments of Pediatrics and Cellular & Molecular Medicine, Pediatric Diabetes Research Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Yinghui Sui
- Departments of Pediatrics and Cellular & Molecular Medicine, Pediatric Diabetes Research Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jinzhao Wang
- Departments of Pediatrics and Cellular & Molecular Medicine, Pediatric Diabetes Research Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Ileana Matta
- Departments of Pediatrics and Cellular & Molecular Medicine, Pediatric Diabetes Research Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Chun Zeng
- Departments of Pediatrics and Cellular & Molecular Medicine, Pediatric Diabetes Research Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Nicholas Vinckier
- Departments of Pediatrics and Cellular & Molecular Medicine, Pediatric Diabetes Research Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Allen Wang
- Departments of Pediatrics and Cellular & Molecular Medicine, Pediatric Diabetes Research Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Kim-Vy Nguyen-Ngoc
- Departments of Pediatrics and Cellular & Molecular Medicine, Pediatric Diabetes Research Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Joshua Chiou
- Departments of Pediatrics and Cellular & Molecular Medicine, Pediatric Diabetes Research Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Klaus H Kaestner
- Department of Genetics and Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Kelly A Frazer
- Department of Pediatrics, Institute for Genomic Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Andrea C Carrano
- Departments of Pediatrics and Cellular & Molecular Medicine, Pediatric Diabetes Research Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Hung-Ping Shih
- Department of Translational Research and Cellular Therapeutics, Diabetes and Metabolic Research Institute, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Maike Sander
- Departments of Pediatrics and Cellular & Molecular Medicine, Pediatric Diabetes Research Center, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
38
|
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
|
39
|
Kaban K, Salva E, Akbuga J. Modulation of the dual-faced effects of miR-141 with chitosan/miR-141 nanoplexes in breast cancer cells. J Gene Med 2019; 21:e3116. [PMID: 31389101 DOI: 10.1002/jgm.3116] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 07/24/2019] [Accepted: 07/24/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND miR-141, known as a tumor suppressive microRNA, is downregulated in breast cancer. However, recent contrasting studies report that it also acts as oncogene when it is upregulated. The present study aimed to investigate whether miR-141 is a tumor suppressor or oncogenic when it reaches normal levels in chitosan/miR-141 nanoplexes. METHODS Chitosan nanoplexes were prepared using simple complexation method. Nanoplexes were characterized by a gel retardation assay and zeta potential and particle size measurements. To determine the expression level of miR-141, a quantitative real-time polymerase chain reaction was performed. The effects of miR-141 mimics were investigated with respect to angiogenesis by vascular endothelial growth factor (VEGF), epithelial-mesenchymal transition (EMT) by E-cadherin, metastasis by Igfbp-4 and Tinagl1 enzyme-linked immunosorbent assays, invasion by an invasion chamber, and apoptosis by Annexin V. RESULTS The miR-141 expression levels of MDA-MB-231 and MDA-MB-435 cells by administration of chitosan/mimic miR-141 nanoplexes reached endogenous miR-141 levels of a non-tumorigenic epithelial breast cell line, MCF-10A. According to our results, metastasis, VEGF, EMT and invasion in breast cancer cells were diminished, whereas apoptosis increased by 1.5- and 2.4-fold in breast cancer cell lines as a result of the miR-141 mimics. CONCLUSIONS In conclusion, we have demonstrated that administration of miR-141 mimics at the determined doses to breast cancer cells revealed a tumor suppressor effect, and not the oncogenic face. The delivery of miR-141 by chitosan nanoplexes presents a promising approach for the suppression of breast cancer.
Collapse
Affiliation(s)
- Kubra Kaban
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Marmara University, Istanbul, Turkey
| | - Emine Salva
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Inonu University, Malatya, Turkey
| | - Julide Akbuga
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Marmara University, Istanbul, Turkey
| |
Collapse
|
40
|
Bradley MW, Aiello KA, Ponnapalli SP, Hanson HA, Alter O. GSVD- and tensor GSVD-uncovered patterns of DNA copy-number alterations predict adenocarcinomas survival in general and in response to platinum. APL Bioeng 2019; 3:036104. [PMID: 31463421 PMCID: PMC6701977 DOI: 10.1063/1.5099268] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 08/06/2019] [Indexed: 12/14/2022] Open
Abstract
More than a quarter of lung, uterine, and ovarian adenocarcinoma (LUAD, USEC, and OV) tumors are resistant to platinum drugs. Only recently and only in OV, patterns of copy-number alterations that predict survival in response to platinum were discovered, and only by using the tensor GSVD to compare Agilent microarray platform-matched profiles of patient-matched normal and primary tumor DNA. Here, we use the GSVD to compare whole-genome sequencing (WGS) and Affymetrix microarray profiles of patient-matched normal and primary LUAD, USEC, and OV tumor DNA. First, the GSVD uncovers patterns similar to one Agilent OV pattern, where a loss of most of the chromosome arm 6p combined with a gain of 12p encode for transformation. Like the Agilent OV pattern, the WGS LUAD and Affymetrix LUAD, USEC, and OV patterns are correlated with shorter survival, in general and in response to platinum. Like the tensor GSVD, the GSVD separates these tumor-exclusive genotypes from experimental inconsistencies. Second, by identifying the shorter survival phenotypes among the WGS- and Affymetrix-profiled tumors, the Agilent pattern proves to be a technology-independent predictor of survival, independent also of the best other indicator at diagnosis, i.e., stage. Third, like no other indicator, the pattern predicts the overall survival of OV patients experiencing progression-free survival, in general and in response to platinum. We conclude that comparative spectral decompositions, such as the GSVD and tensor GSVD, underlie a mathematically universal description of the relationships between a primary tumor's genotype and a patient's overall survival phenotype, which other methods miss.
Collapse
Affiliation(s)
| | | | - Sri Priya Ponnapalli
- Scientific Computing and Imaging Institute, University of Utah, Salt Lake City, Utah 84112, USA
| | | | | |
Collapse
|
41
|
LncRNA XIST enhanced TGF-β2 expression by targeting miR-141-3p to promote pancreatic cancer cells invasion. Biosci Rep 2019; 39:BSR20190332. [PMID: 31213574 PMCID: PMC6603275 DOI: 10.1042/bsr20190332] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 05/30/2019] [Accepted: 06/12/2019] [Indexed: 11/17/2022] Open
Abstract
The level of expression of long non-coding RNA (LncRNA) X-inactive specific transcript (XIST) is up-regulated in pancreatic cancer (PC). However, the role of XIST in PC and the underlying mechanism are still unknown. The present study aimed to elucidate how XIST participates in PC and its potential target, miR-141-3p. We detected the XIST expression in PC tissues and cells by qRT-PCR. Cell proliferation was measured using a CCK8 kit, and the migration and invasion of cells was measured by Transwell assay. Silencing XIST and miR-141-3p was performed with transfection by Lipofectamine kit. Binding assay was conducted by luciferase reporter assay. Protein expression was examined by Western blot. These results indicate that (i) XIST is highly expressed in tumor tissues while miR-141-3p is down-regulated. (ii) Silencing XIST inhibits the pancreatic cell proliferation, migration and invasion. (iii) MiR-141-3p inhibitor alleviates the inhibitory effect by siXIST in PC cell lines. (iv) MiR-141-3p directly interacts with XIST and also negatively regulates transforming growth factor-β 2 (TGF-β2) expression. (v) Overexpression of XIST attenuates the inhibition of TGF-β2 expression by miR-141-3p. The conclusion, is that XIST could promote proliferation, migration and invasion of PC cells via miR-141-5p/TGF-β2 axis.
Collapse
|
42
|
Tang W, Zhou Y, Sun D, Dong L, Xia J, Yang B. Oncogenic role of phospholipase C-γ1 in progression of hepatocellular carcinoma. Hepatol Res 2019; 49:559-569. [PMID: 30623526 DOI: 10.1111/hepr.13309] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 12/30/2018] [Accepted: 01/04/2019] [Indexed: 12/13/2022]
Abstract
AIM Phospholipase C-γ1 (PLCG1) was previously found to be involved in a variety of oncogenic behaviors such as cell motility, cell proliferation, cell migration, and invasion. However, its function in hepatocellular carcinoma (HCC) was unknown. Here, we explored the expression pattern and function of PLCG1 in HCC progression. METHODS Expression of PLCG1 was examined by western blotting in hepatoma cells and human tumor tissues. Expression was also detected by immunohistochemistry in 150 HCC clinical samples, and its clinical significance was analyzed. The influence of PLCG1 on HCC carcinogenesis were determined in vitro and in vivo. The underlying mechanisms were explored by detecting the expression of critical molecules of signaling pathways. RESULTS The results showed that PLCG1 was overexpressed in hepatoma cell lines and clinical HCC tissues. Increased PLCG1 expression in tumor tissues was remarkably correlated with poor clinical features of HCC. Patients with positive PLCG1 expression in tumor tissues had shorter overall survival and relapse-free survival. Phospholipase C gamma 1 could substantially promote cell proliferation, anchor growth, and cell invasion in vitro. The in vivo study showed that inhibition of PLCG1 in hepatoma cells significantly repressed tumor growth in nude mice. Furthermore, we showed that PLCG1 might exert its function by activating the mitogen-activated protein kinase and nuclear factor-κB signaling pathways. CONCLUSION Our data indicated that PLCG1 could act as an oncogene in HCC carcinogenesis and could serve as a valuable prognostic marker and potential therapeutic target for HCC.
Collapse
Affiliation(s)
- Wenqing Tang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yi Zhou
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Dalong Sun
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ling Dong
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Institute of Liver disease, Fudan University, Shanghai, China
| | - Jinglin Xia
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China.,Minhang Hospital, Fudan University, Shanghai, China
| | - Biwei Yang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
43
|
Zhang Y, Chen J, Wang Y, Wang D, Cong W, Lai BS, Zhao Y. Multilayer network analysis of miRNA and protein expression profiles in breast cancer patients. PLoS One 2019; 14:e0202311. [PMID: 30946749 PMCID: PMC6448837 DOI: 10.1371/journal.pone.0202311] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 03/19/2019] [Indexed: 12/21/2022] Open
Abstract
MiRNAs and proteins play important roles in different stages of breast tumor development and serve as biomarkers for the early diagnosis of breast cancer. A new algorithm that combines machine learning algorithms and multilayer complex network analysis is hereby proposed to explore the potential diagnostic values of miRNAs and proteins. XGBoost and random forest algorithms were employed to screen the most important miRNAs and proteins. Maximal information coefficient was applied to assess intralayer and interlayer connection. A multilayer complex network was constructed to identify miRNAs and proteins that could serve as biomarkers for breast cancer. Proteins and miRNAs that are nodes in the network were subsequently categorized into two network layers considering their distinct functions. The betweenness centrality was used as the first measurement of the importance of the nodes within each single layer. The degree of the nodes was chosen as the second measurement to map their signalling pathways. By combining these two measurements into one score and comparing the difference of the same candidate between normal tissue and cancer tissue, this novel multilayer network analysis could be applied to successfully identify molecules associated with breast cancer.
Collapse
Affiliation(s)
- Yang Zhang
- Harbin Institute of Technology (Shenzhen), Shenzhen, Guangdong, China
| | - Jiannan Chen
- Harbin Institute of Technology (Shenzhen), Shenzhen, Guangdong, China
| | - Yu Wang
- Harbin Institute of Technology (Shenzhen), Shenzhen, Guangdong, China
| | - Dehua Wang
- Harbin Institute of Technology (Shenzhen), Shenzhen, Guangdong, China
| | - Weihui Cong
- Harbin Institute of Technology (Shenzhen), Shenzhen, Guangdong, China
| | - Bo Shiun Lai
- Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Yi Zhao
- Harbin Institute of Technology (Shenzhen), Shenzhen, Guangdong, China
| |
Collapse
|
44
|
Wu Y, Shi W, Tang T, Wang Y, Yin X, Chen Y, Zhang Y, Xing Y, Shen Y, Xia T, Guo C, Pan Y, Jin L. miR-29a contributes to breast cancer cells epithelial-mesenchymal transition, migration, and invasion via down-regulating histone H4K20 trimethylation through directly targeting SUV420H2. Cell Death Dis 2019; 10:176. [PMID: 30792382 PMCID: PMC6385178 DOI: 10.1038/s41419-019-1437-0] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 01/01/2019] [Accepted: 01/08/2019] [Indexed: 12/31/2022]
Abstract
Breast cancer is the most prevalent cancer in women worldwide, which remains incurable once metastatic. Breast cancer stem cells (BCSCs) are a small subset of breast cancer cells which are essential in tumor formation, metastasis, and drug resistance. microRNAs (miRNAs) play important roles in the breast cancer cells and BCSCs by regulating specific genes. In this study, we found that miR-29a was up-regulated in BCSCs, in aggressive breast cancer cell line and in breast cancer tissues. We also confirmed suppressor of variegation 4–20 homolog 2 (SUV420H2), which is a histone methyltransferase that specifically trimethylates Lys-20 of histone H4 (H4K20), as the target of miR-29a. Both miR-29a overexpression and SUV420H2 knockdown in breast cancer cells promoted their migration and invasion in vitro and in vivo. Furthermore, we discovered that SUV420H2-targeting miR-29a attenuated the repression of connective tissue growth factor (CTGF) and growth response protein-1 (EGR1) by H4K20 trimethylation and promoted the EMT progress of breast cancer cells. Taken together, our findings reveal that miR-29a plays critical roles in the EMT and metastasis of breast cancer cells through targeting SUV420H2. These findings may provide new insights into novel molecular therapeutic targets for breast cancer.
Collapse
Affiliation(s)
- You Wu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of life Science and Technology, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, Jiangsu province, China
| | - Wanyue Shi
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of life Science and Technology, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, Jiangsu province, China
| | - Tingting Tang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of life Science and Technology, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, Jiangsu province, China
| | - Yidong Wang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of life Science and Technology, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, Jiangsu province, China
| | - Xin Yin
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of life Science and Technology, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, Jiangsu province, China
| | - Yanlin Chen
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of life Science and Technology, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, Jiangsu province, China
| | - Yanfeng Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of life Science and Technology, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, Jiangsu province, China
| | - Yun Xing
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of life Science and Technology, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, Jiangsu province, China
| | - Yumeng Shen
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of life Science and Technology, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, Jiangsu province, China
| | - Tiansong Xia
- Department of Breast Surgery, Breast Disease Center of Jiangsu Province, First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu province, China
| | - Changying Guo
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of life Science and Technology, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, Jiangsu province, China
| | - Yi Pan
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of life Science and Technology, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, Jiangsu province, China.
| | - Liang Jin
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of life Science and Technology, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, Jiangsu province, China.
| |
Collapse
|
45
|
Jiang LP, Wang SR, Chung HK, Buddula S, Wang JY, Rao JN. miR-222 represses expression of zipcode binding protein-1 and phospholipase C-γ1 in intestinal epithelial cells. Am J Physiol Cell Physiol 2019; 316:C415-C423. [PMID: 30649922 DOI: 10.1152/ajpcell.00165.2018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Both zipcode binding protein-1 (ZBP1) and phospholipase C-γ1 (PLCγ1) are intimately involved in many aspects of early intestinal mucosal repair after acute injury, but the exact mechanisms that control their cellular abundances remain largely unknown. The present study shows that microRNA-222 (miR-222) interacts with the mRNAs encoding ZBP1 and PLCγ1 and regulates ZBP1 and PLCγ1 expression in intestinal epithelial cells (IECs). The biotinylated miR-222 bound specifically to the ZBP1 and PLCγ1 mRNAs in IECs. Ectopically expressed miR-222 precursor destabilized the ZBP1 and PLCγ1 mRNAs and consequently lowered the levels of cellular ZBP1 and PLCγ1 proteins. Conversely, decreasing the levels of cellular miR-222 by transfection with its antagonism increased the stability of the ZBP1 and PLCγ1 mRNAs and increased the levels of ZBP1 and PLCγ1 proteins. Overexpression of miR-222 also inhibited cell migration over the wounded area, which was partially abolished by overexpressing ZBP1 and PLCγ1. Furthermore, prevention of the increased levels of ZBP1 and PLCγ1 in the miR-222-silenced cells by transfection with specific small interfering RNAs targeting ZBP1 or PLCγ1 mRNA inhibited cell migration after wounding. These findings indicate that induced miR-222 represses expression of ZBP1 and PLCγ1 at the posttranscriptional level, thus inhibiting IEC migration during intestinal epithelial restitution after wounding.
Collapse
Affiliation(s)
- Li-Ping Jiang
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine , Baltimore, Maryland
| | - Shelley R Wang
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine , Baltimore, Maryland.,Baltimore Veterans Affairs Medical Center , Baltimore, Maryland
| | - Hee Kyoung Chung
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine , Baltimore, Maryland.,Baltimore Veterans Affairs Medical Center , Baltimore, Maryland
| | - Saharsh Buddula
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine , Baltimore, Maryland
| | - Jian-Ying Wang
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine , Baltimore, Maryland.,Baltimore Veterans Affairs Medical Center , Baltimore, Maryland.,Department of Pathology, University of Maryland School of Medicine , Baltimore, Maryland
| | - Jaladanki N Rao
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine , Baltimore, Maryland.,Baltimore Veterans Affairs Medical Center , Baltimore, Maryland
| |
Collapse
|
46
|
Liang C, Yu S, Wong KC, Luo J. A novel semi-supervised model for miRNA-disease association prediction based on [Formula: see text]-norm graph. J Transl Med 2018; 16:357. [PMID: 30547813 PMCID: PMC6295065 DOI: 10.1186/s12967-018-1741-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 12/10/2018] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND Identification of miRNA-disease associations has attracted much attention recently due to the functional roles of miRNAs implicated in various biological and pathological processes. Great efforts have been made to discover the potential associations between miRNAs and diseases both experimentally and computationally. Although reliable, the experimental methods are in general time-consuming and labor-intensive. In comparison, computational methods are more efficient and applicable to large-scale datasets. METHODS In this paper, we propose a novel semi-supervised model to predict miRNA-disease associations via [Formula: see text]-norm graph. Specifically, we first recalculate the miRNA functional similarities as well as the disease semantic similarities based on the latest version of MeSH descriptors and HMDD. We then update the similarity matrices and association matrix iteratively in both miRNA space and disease space. The optimized association matrices from each space are combined together as the final output. RESULTS Compared with four state-of-the-art prediction methods, our method achieved favorable performance with AUCs of 0.943 and 0.946 in both global LOOCV and local LOOCV, respectively. In addition, we carried out three types of case studies on five common human diseases, and most of the top 50 predicted miRNAs were confirmed to be associated with the investigated diseases by four databases dbDEMC, PheomiR, miR2Disease and miRwayDB. Specifically, our results provided potential evidence that miRNAs within the same family or cluster were likely to play functional roles together in given diseases. CONCLUSIONS Taken together, the experimental results clearly demonstrated the utility of the proposed method. We anticipated that our method could serve as a reliable and efficient tool for miRNA-disease association prediction.
Collapse
Affiliation(s)
- Cheng Liang
- School of Information Science and Engineering, Shandong Normal University, Jinan, 250358 China
| | - Shengpeng Yu
- School of Information Science and Engineering, Shandong Normal University, Jinan, 250358 China
| | - Ka-Chun Wong
- Department of Computer Science, City University of Hong Kong, Kowloon Tong, 999077 Hong Kong
| | - Jiawei Luo
- College of Information Science and Engineering, Hunan University, Changsha, 410082 China
| |
Collapse
|
47
|
Vahidian F, Mohammadi H, Ali-Hasanzadeh M, Derakhshani A, Mostaan M, Hemmatzadeh M, Baradaran B. MicroRNAs and breast cancer stem cells: Potential role in breast cancer therapy. J Cell Physiol 2018; 234:3294-3306. [PMID: 30362508 DOI: 10.1002/jcp.27246] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Accepted: 07/24/2018] [Indexed: 12/19/2022]
Abstract
MicroRNAs (miRNAs) can control cancer and cancer stem cells (CSCs), and this topic has drawn immense attention recently. Stem cells are a tiny population of a bulk of tumor cells that have enormous potential in expansion and metastasis of the tumor. miRNA have a crucial role in the management of the function of stem cells. This role is to either promote or suppress the tumor. In this review, we investigated the function and different characteristics of CSCs and function of the miRNAs that are related to them. We also demonstrated the role and efficacy of these miRNAs in breast cancer and breast cancer stem cells (BCSC). Eventually, we revealed the metastasis, tumor formation, and their role in the apoptosis process. Also, the therapeutic potential of miRNA as an effective method for the treatment of BCSC was described. Extensive research is required to investigate the employment or suppression of these miRNAs for therapeutics approached in different cancers in the future.
Collapse
Affiliation(s)
- Fatemeh Vahidian
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Mohammadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Ali-Hasanzadeh
- Department of Immunology, School of Medicine, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Afshin Derakhshani
- Department of Immunology, Birjand University of Medical Sciences, Birjand, Iran.,Student Research Committee, Birjand University of Medical Sciences, Birjand, Iran
| | - Masoud Mostaan
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Islamic Azad university, Tabriz, Iran
| | - Maryam Hemmatzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
48
|
Liu C, Hu W, Li LL, Wang YX, Zhou Q, Zhang F, Song-Yang YY, Zhu W, Sun CC, Li DJ. Roles of miR-200 family members in lung cancer: more than tumor suppressors. Future Oncol 2018; 14:2875-2886. [PMID: 30208739 DOI: 10.2217/fon-2018-0155] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
miRNAs are a class of single-stranded noncoding RNAs, which have no coding potential, but modulate many molecular mechanisms including cancer pathogenesis. miRNAs participate in cell proliferation, differentiation, apoptosis, as well as carcinogenesis or cancer progression, and their involvement in lung cancer has been recently shown. They are suggested to have bidirectional functions on important cancer-related genes so as to enhance or attenuate tumor genesis. Epithelial-mesenchymal transition (EMT) is a fundamental process which contributes to integrity of organogenesis and tissue differentiation as well as tissue repair, organ fibrosis and the progression of carcinoma, and several miRNAs were suggested to form the network regulating EMT in lung cancer, among which, miR-200 family members (miR-200a, miR-200b, miR-200c, miR-429 and miR-141) play crucial roles in the suppression of EMT.
Collapse
Affiliation(s)
- Cong Liu
- Department of Occupational & Environmental Health, Wuhan University School of Health Sciences, Wuhan, Hubei 430071, PR China
| | - Wei Hu
- Department of Occupational & Environmental Health, Wuhan University School of Health Sciences, Wuhan, Hubei 430071, PR China
| | - Lin-Lin Li
- Department of Occupational & Environmental Health, Wuhan University School of Health Sciences, Wuhan, Hubei 430071, PR China
| | - Yu-Xuan Wang
- Department of Occupational & Environmental Health, Wuhan University School of Health Sciences, Wuhan, Hubei 430071, PR China
| | - Qun Zhou
- Department of Occupational & Environmental Health, Wuhan University School of Health Sciences, Wuhan, Hubei 430071, PR China
| | - Feng Zhang
- Department of Occupational & Environmental Health, Wuhan University School of Health Sciences, Wuhan, Hubei 430071, PR China
| | - Yi-Yan Song-Yang
- Department of Occupational & Environmental Health, Wuhan University School of Health Sciences, Wuhan, Hubei 430071, PR China
| | - Wei Zhu
- Department of Occupational & Environmental Health, Wuhan University School of Health Sciences, Wuhan, Hubei 430071, PR China
| | - Cheng-Chao Sun
- Department of Occupational & Environmental Health, Wuhan University School of Health Sciences, Wuhan, Hubei 430071, PR China
| | - De-Jia Li
- Department of Occupational & Environmental Health, Wuhan University School of Health Sciences, Wuhan, Hubei 430071, PR China
| |
Collapse
|
49
|
Tang T, Yang Z, Zhu Q, Wu Y, Sun K, Alahdal M, Zhang Y, Xing Y, Shen Y, Xia T, Xi T, Pan Y, Jin L. Up-regulation of miR-210 induced by a hypoxic microenvironment promotes breast cancer stem cells metastasis, proliferation, and self-renewal by targeting E-cadherin. FASEB J 2018; 32:fj201801013R. [PMID: 30188754 DOI: 10.1096/fj.201801013r] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Breast cancer stem cells (BCSCs), a small subset of breast cancer cells with stem cell-like properties, are essential in tumor formation, metastasis, resistance to anticancer therapies, and cancer recurrence. MicroRNAs (miRNAs) are involved in tumorigenicity by regulating specific oncogenes and tumor-suppressor genes, and their roles in BCSCs are becoming apparent. A novel, 3-dimensional (3D), semisolid culture system was established to culture MCF-7 spheroid cells with high percentage of BCSCs. The differences in miRNA expression among the MCF-7 parental cells, BCSC-enriched MCF-7 spheroid cells, and CD44+/CD24- MCF-7 cells were evaluated by miRNA microarray, and the high expression of miR-210 in MCF-7 spheroid cells and CD44+/CD24- MCF-7 cells was verified by quantitative RT-PCR. MCF-7 cells were cultured in a hypoxic chamber to detect the effect of hypoxia on miR-210 expression and the stemness of the cells. The 3-(4,5-dimethylthiazol-2- yl)-2,5-diphenyl tetrazolium bromide (MTT), transwell, and sphere-formation assays were performed to detect the proliferation, migration, and self-renewal ability of miR-210-overexpressed MCF-7 cells and MCF-7 spheroid cells with miR-210 knocked down. The target of miR-210 was validated with a dual-luciferase reporter assay and Western blotting. In vivo xenograft assay and metastasis assay were performed to study the effects of miR-210 targeting E-cadherin on BCSCs growth and lung metastasis, and the tumors were assessed by immunohistochemistry and immunofluorescence. We developed a novel 3D, semisolid culture system to culture MCF-7 spheroid cells, which are enriched in BCSCs, and found, by performing miRNAs expression profiling, miR-210 was up-regulated in those cells compared with MCF-7 parental cells. High miR-210 expression was also detected in CD44+/CD24- MCF-7 cells and human CD44+/CD24- breast cancer cells, which was demonstrated to be partially due to the hypoxic microenvironment around BCSCs in MCF-7 spheroids or solid tumors. Ectopic expression of miR-210 in MCF-7 cells promoted their migration, invasion, proliferation, and self-renewal in both in vitro and in vivo studies. We further reported that miR-210 suppressed E-cadherin expression by targeting the open reading frame region of E-cadherin mRNA and by up-regulation of E-cadherin transcription repressor, Snail. Accordingly, E-cadherin overexpression compromises the migration, invasion, proliferation, and self-renewal ability of miR-210-overexpressed MCF-7 both in vitro and in vivo. These findings reveal a novel regulatory pathway centered on hypoxia-mediated miR-210 targeting of E-cadherin, which contributes to the properties and breast tumorigenesis of BCSCs.-Tang, T., Yang, Z., Zhu, Q., Wu, Y., Sun, K., Alahdal, M., Zhang, Y., Xing, Y., Shen, Y., Xia, T., Xi, T., Pan, Y., Jin, L. Up-regulation of miR-210 induced by a hypoxic microenvironment promotes breast cancer stem cells metastasis, proliferation, and self-renewal by targeting E-cadherin.
Collapse
Affiliation(s)
- Tingting Tang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Zhaocong Yang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Qinhua Zhu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - You Wu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Kun Sun
- Nanjing No. 3 Senior School, Nanjing, China
| | - Murad Alahdal
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yanfeng Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yun Xing
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yumeng Shen
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Tiansong Xia
- Department of Breast Surgery, Breast Disease Center of Jiangsu Province, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Tao Xi
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yi Pan
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Liang Jin
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
50
|
WIPF1 antagonizes the tumor suppressive effect of miR-141/200c and is associated with poor survival in patients with PDAC. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:167. [PMID: 30041660 PMCID: PMC6056910 DOI: 10.1186/s13046-018-0848-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 07/16/2018] [Indexed: 12/31/2022]
Abstract
Background Aberrant expression of Wiskott–Aldrich syndrome protein interacting protein family member 1 (WIPF1) contributes to the invasion and metastasis of several malignancies. However, the role of WIPF1 in human pancreatic ductal adenocarcinoma (PDAC) remains poorly understood. Methods Human pancreatic cancer samples from PDAC patients were collected for methylation analysis. Bioinformatic prediction program and luciferase reporter assay were used to identify microRNAs regulating WIPF1 expression. The association between WIPF1 expression and the overall survival (OS) of patients with PDAC was evaluated by using The Cancer Genome Atlas (TCGA) database. The roles of miR-141/200c and WIPF1 on the invasion and metastasis of PDAC cells were investigated both in vitro and in vivo. Results We found that compared to the surrounding non-cancerous tissues, there was significantly increased methylation of miR-200c and miR-141 in human PDAC tissues that resulted in their silencing, whereas the members of the other cluster of miR-200 family including miR-200a, miR-200b and miR-429 were hypomethylated. Our data show that forced expression of miR-141 or miR-200c suppressed invasion and metastasis of PDAC cells both in vitro and in xenograft and identified WIPF1 as a direct target of miR-141 and miR-200c. Both miR-141 and miR-200c inhibit WIPF1 by directly interacting with its 3′-untranslated region. Remarkably, silencing of WIPF1 blocked PDAC growth and metastasis both in vitro and in vivo, whereas forced WIPF1 overexpression antagonized the tumor suppressive effect of miR-141/200c. Additionally, by using TCGA database we showed that high expression of WIPF1 correlated with poor survival in patients with PDAC. Moreover, we show that miR-141 and miR-200c blocked YAP/TAZ expression by suppressing WIPF1. Conclusions We have identified WIPF1 as an oncoprotein in PDAC and a direct target of miR-141/miR-200c. We have also defined the miR-141/200c-WIPF1-YAP/TAZ as a novel signaling pathway that is involved in the regulation of the invasion and metastasis of human PDAC cells. Electronic supplementary material The online version of this article (10.1186/s13046-018-0848-6) contains supplementary material, which is available to authorized users.
Collapse
|