1
|
Mashkoor NR, Abed SA, Davoudi A, Jassim ZAA, Faraj ZY, Akbari F, Bajgiran FA, Hedayati M, Salehzadeh A. Synthesis of platinum nanoparticles functionalized with glutamine and conjugated with thiosemicarbazone and their cytotoxic effects on MDA-MB-231 breast cancer cell line and evaluation of CASP-8 gene expression. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024:10.1007/s00210-024-03629-z. [PMID: 39665983 DOI: 10.1007/s00210-024-03629-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 11/11/2024] [Indexed: 12/13/2024]
Abstract
Breast cancer (BC) is the most prevalent form of cancer among women and is a major contributor to cancer-related fatalities. Nanotechnology has provided novel approaches to drug delivery to cancer cells. In this work, we synthesized platinum (Pt) nanoparticles, functionalized them with glutamine, conjugated them with thiosemicarbazone (TSC), and characterized their anticancer effects on the MDA-MB-231 breast cancer cell line. Characteristics of the nanoparticles were assessed by FT-IR, XRD, EDS mapping, SEM, TEM, DLS, and zeta potential measurement. Cell viability was characterized by MTT assay, and cell necrosis/apoptosis levels were determined by flow cytometry. The expression level of the CASP-8 gene was investigated by real-time PCR. Pt@Gln-TSC nanoparticles are spherical, 20-70 nm in diameter in dry form, 662 nm after hydration, and their zeta potential was - 6.6 mV. The 50% inhibitory concentration (IC50) for MDA-MB-231 (breast cancer) and HDF (normal) cell lines was 170 and 348µg/ml, respectively. Also, the IC50 of oxaliplatin drug and TSC on MDA-MB-231 cells was 184 µg/ml and 307 µg/ml, respectively. Treatment with Pt@Gln-TSC nanoparticles caused an increase in cell necrosis and primary apoptosis and elevated the expression of the CASP-8 gene by 2.54 folds. This study shows that Pt@Gln-TSC nanoparticles are significantly more toxic to breast cancer cells than to normal cells and can inhibit MDA-MB-231 cells by activating extrinsic apoptosis.
Collapse
Affiliation(s)
- Nabeel Rahi Mashkoor
- Department of Pathological Analysis, College of Science, AL-Qadisiyah University, Al Diwaniyah, AL-Qadisiyah, Iraq
| | - Salwan Ali Abed
- Environmental Science Department, College of Science, AL-Qadisiyah University, Al Diwaniyah, AL-Qadisiyah, Iraq
| | - Arash Davoudi
- Division of Cytogenetic, Dr. Keshavarz Medical Genetics Lab, Rasht, Iran
- Department of Biology, Rasht Branch, Islamic Azad University, Rasht, Iran
| | | | - Zainab Yousif Faraj
- Scientific Affairs Department, AL-Qadisiyah University, Al Diwaniyah, AL-Qadisiyah, Iraq
| | - Fatemeh Akbari
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | | | | | - Ali Salehzadeh
- Department of Biology, Rasht Branch, Islamic Azad University, Rasht, Iran.
| |
Collapse
|
2
|
McIntyre G, Jackson Z, Colina J, Sekhar S, DiFeo A. miR-181a: regulatory roles, cancer-associated signaling pathway disruptions, and therapeutic potential. Expert Opin Ther Targets 2024; 28:1061-1091. [PMID: 39648331 DOI: 10.1080/14728222.2024.2433687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 11/20/2024] [Indexed: 12/10/2024]
Abstract
INTRODUCTION microRNA-181a (miR-181a) is a crucial post-transcriptional regulator of many mRNA transcripts and noncoding-RNAs, influencing cell proliferation, cancer cell stemness, apoptosis, and immune responses. Its abnormal expression is well-characterized in numerous cancers, establishing it as a significant genomic vulnerability and biomarker in cancer research. AREAS COVERED Here, we summarize miR-181a's correlation with poor patient outcomes across numerous cancers and the mechanisms governing miR-181a's activity and processing. We comprehensively describe miR-181a's involvement in multiple regulatory cancer signaling pathways, cellular processes, and the tumor microenvironment. We also discuss current therapeutic approaches to targeting miR-181a, highlighting their limitations and future potential. EXPERT OPINION miR-181a is a clinically relevant pan-cancer biomarker with potential as a therapeutic target. Its regulatory control of tumorigenic signaling pathways and immune responses positions it as a promising candidate for personalized treatments. The success of miR-181a as a target relies on the development of specific therapeutics platforms. Future research on miR-181a's role in the tumor microenvironment and the RNA binding proteins that regulate its stability will help uncover new techniques to targeting miR-181a. Further research into miR-181a serum levels in patients undergoing therapy will help to better stratify patients and enhance therapeutic success.
Collapse
Affiliation(s)
- Grace McIntyre
- Department of Pathology, Rackham Graduate School, University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Zoe Jackson
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Jose Colina
- Department of Pathology, Rackham Graduate School, University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Sreeja Sekhar
- Department of Pathology, Rackham Graduate School, University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Analisa DiFeo
- Department of Pathology, Rackham Graduate School, University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
3
|
Zhang Y, Guan Y, Zheng X, Li C. Hypoxia-induced miR-181a-5p up-regulation reduces epirubicin sensitivity in breast cancer cells through inhibiting EPDR1/TRPC1 to activate PI3K/AKT signaling pathway. BMC Cancer 2024; 24:167. [PMID: 38308220 PMCID: PMC10835859 DOI: 10.1186/s12885-024-11906-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 01/21/2024] [Indexed: 02/04/2024] Open
Abstract
Breast carcinoma (BC) ranks as a predominant malignancy and constitutes the second principal cause of mortality among women globally. Epirubicin stands as the drug of choice for BC therapeutics. Nevertheless, the emergence of chemoresistance has significantly curtailed its therapeutic efficacy. The resistance mechanisms to Epirubicin remain not entirely elucidated, yet they are conjectured to stem from diminished tumor vascular perfusion and resultant hypoxia consequent to Epirubicin administration. In our investigation, we meticulously scrutinized the Gene Expression Omnibus database for EPDR1, a gene implicated in hypoxia and Epirubicin resistance in BC. Subsequently, we delineated the impact of EPDR1 on cellular proliferation, motility, invasive capabilities, and interstitial-related proteins in BC cells, employing methodologies such as the CCK-8 assay, Transwell assay, and western blot analysis. Our research further unveiled that hypoxia-induced miR-181a-5p orchestrates the regulation of BC cell duplication, migration, invasion, and interstitial-related protein expression via modulation of EPDR1. In addition, we identified TRPC1, a gene associated with EPDR1 expression in BC, and substantiated that EPDR1 influences BC cellular dynamics through TRPC1-mediated modulation of the PI3K/AKT signaling cascade. Our findings underscore the pivotal role of EPDR1 in the development of BC. EPDR1 was found to be expressed at subdued levels in BC tissues, Epirubicin-resistant BC cells, and hypoxic BC cells. The overexpression of EPDR1 curtailed BC cell proliferation, motility, invasiveness, and the expression of interstitial-related proteins. At a mechanistic level, the overexpression of hypoxia-induced miR-181a-5p was observed to inhibit the EPDR1/TRPC1 axis, thereby activating the PI3K/AKT signaling pathway and diminishing the sensitivity to Epirubicin in BC cells. In summation, our study demonstrates that the augmentation of hypoxia-induced miR-181a-5p diminishes Epirubicin sensitivity in BC cells by attenuating EPDR1/TRPC1 expression, thereby invigorating the PI3K/AKT signaling pathway. This exposition offers a theoretical foundation for the application of Epirubicin in BC therapy, marking a significant contribution to the existing body of oncological literature.
Collapse
Affiliation(s)
- Yunwei Zhang
- Department of Breast Surgery, The First Hospital of China Medical University, 155 North Nanjing Street, Shenyang, Liaoning, China
- Department of Breast Clinic, Shenyang Maternity and Child Health Hosital, No. 20, Yuanjiang Street, Shenyang, Liaoning, China
| | - Yunping Guan
- Department of Breast Clinic, Shenyang Maternity and Child Health Hosital, No. 20, Yuanjiang Street, Shenyang, Liaoning, China
| | - Xinyu Zheng
- Department of Breast Surgery, The First Hospital of China Medical University, 155 North Nanjing Street, Shenyang, Liaoning, China.
- Lab 1, Cancer Institute, The First Hospital of China Medical University, 155 North Nanjing Street, Shenyang, Liaoning, China.
| | - Chenyang Li
- Department of Breast Clinic, Shenyang Maternity and Child Health Hosital, No. 20, Yuanjiang Street, Shenyang, Liaoning, China.
| |
Collapse
|
4
|
Perepechaeva ML, Studenikina AA, Grishanova AY, Glushkov AN, Polenok EG, Bajramov PV, Autenshlyus AI. Serum miR-181a and miR-25 in patients with malignant and benign breast diseases. BIOMEDITSINSKAIA KHIMIIA 2023; 69:307-314. [PMID: 37937433 DOI: 10.18097/pbmc20236905307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2023]
Abstract
Breast tumor diseases include a wide range of pathologies that require different approaches to their treatment. MicroRNA (miR) levels, reflecting regulation of the gene expression involved in tumorigenesis, can be diagnostic and prognostic markers of breast diseases. The levels of circulating miR-181a and miR-25 were measured in patients with benign breast diseases (BBD), patients with invasive carcinoma of a nonspecific type (ICNT) and also in conditionally healthy women. Expression of both miRs was higher in patients of both groups as compared to controls; at the same time, the content of serum miR-181a and miR-25 was higher in BBD patients than in ICNT patients. The detected changes may be of interest in the context of precancerous changes in BBD. It seems possible to use them in the future as markers of the pathological process as a part of a large diagnostic panel.
Collapse
Affiliation(s)
- M L Perepechaeva
- Novosibirsk State Medical University, Novosibirsk, Russia; Research Institute of Molecular Biology and Biophysics, Federal Research Center for Fundamental and Translational Medicine, Novosibirsk, Russia
| | - A A Studenikina
- Novosibirsk State Medical University, Novosibirsk, Russia; Research Institute of Molecular Biology and Biophysics, Federal Research Center for Fundamental and Translational Medicine, Novosibirsk, Russia
| | - A Yu Grishanova
- Research Institute of Molecular Biology and Biophysics, Federal Research Center for Fundamental and Translational Medicine, Novosibirsk, Russia
| | - A N Glushkov
- Institute of Human Ecology of the Federal Research Center of Coal and Coal Chemistry SB RAS, Kemerovo, Russia
| | - E G Polenok
- Institute of Human Ecology of the Federal Research Center of Coal and Coal Chemistry SB RAS, Kemerovo, Russia
| | - P V Bajramov
- M.S. Rappoport Kuzbass Clinical Oncologic Dispensary, Kemerovo, Russia
| | - A I Autenshlyus
- Novosibirsk State Medical University, Novosibirsk, Russia; Research Institute of Molecular Biology and Biophysics, Federal Research Center for Fundamental and Translational Medicine, Novosibirsk, Russia
| |
Collapse
|
5
|
Basak M, Sahoo B, Chaudhary DK, Narisepalli SB, Tiwari S, Chitkara D, Mittal A. Human umbilical cord blood-mesenchymal stem cell derived exosomes as an efficient nanocarrier for Docetaxel and miR-125a: Formulation optimization and anti-metastatic behaviour. Life Sci 2023; 322:121621. [PMID: 37001803 DOI: 10.1016/j.lfs.2023.121621] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 03/10/2023] [Accepted: 03/21/2023] [Indexed: 03/31/2023]
Abstract
AIM Exosomes, as a nanocarrier for the co-delivery of biologicals and small anticancer molecules is yet in its infancy. Herein, we investigated hUCBMSC derived exosomes as a biogenic nanocarrier for the co-delivery of tumor suppressor miR-125a and microtubule destabilizing Docetaxel (DTX) to target the proliferative and migratory aggressiveness of the murine TNBC 4T1 cells. MAIN METHODS In this study, hUCBMSCs from the human umbilical cord blood cells (hUCB) were successfully transfected with miR-125a. Thereafter, DTX was encapsulated into both non-transfected and transfected exosomes by optimized mild sonication-incubation technique. The anticancer efficiency of hUCBMSC Exo-DTX and miR-125a Exo-DTX was compared by MTT and morphometric assay. The prominent anti-metastatic behaviour of the latter was confirmed by in-vitro wound healing and transwell invasion assay. Further, the synergistic effect of miR-125a and DTX was confirmed by F-actin and nuclear degradation by confocal and FESEM assay. KEY FINDINGS hUCBMSC exosomes exhibited DTX payload of 8.86 ± 1.97 ng DTX/ μg exosomes and miRNA retention capacity equivalent to 12.31 ± 5.73 %. The co-loaded formulation (miR-125a Exo-DTX) exhibited IC50 at 192.8 ng/ml in 4T1 cells, which is almost 2.36 folds' lower than the free DTX IC50 (472.8 ng/ml). Additionally, miR-125a Exo-DTX treatment caused wound broadening upto 6.14±0.38 % while treatment with free DTX and miR-125a exosomes alone caused 18.71±4.5 % and 77.36±10.4 % of wound closure respectively in 36 h. miR-125a Exo-DTX treatment further exhibited significantly reduced invasiveness of 4T1 cells (by 3.5 ± 1.8 %) along with prominent cytoskeletal degradation and nuclear deformation as compared to the miR-125a exosomes treated group. The miR-125a expressing DTX loaded exosomal formulation clearly demonstrated the synergistic apoptotic and anti-migratory efficiency of the miR-125a Exo-DTX. SIGNIFICANCE The synergistic anticancer and anti-metastatic effect of miR-125a Exo-DTX was observed due to presence of both DTX and miR-125a as the cargo of hUCBMSC derived exosomes.
Collapse
|
6
|
Xu G, Yang Y, Yang J, Xiao L, Wang X, Qin L, Gao J, Xuan R, Wu X, Chen Z, Sun R, Song G. Screening and identification of miR-181a-5p in oral squamous cell carcinoma and functional verification in vivo and in vitro. BMC Cancer 2023; 23:162. [PMID: 36800936 PMCID: PMC9936757 DOI: 10.1186/s12885-023-10600-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Accepted: 02/01/2023] [Indexed: 02/19/2023] Open
Abstract
BACKGROUND Oral squamous cell carcinoma (OSCC) is a common malignant tumor associated with poor prognosis. MicroRNAs (miRNAs) play crucial regulatory roles in the cancer development. However, the role of miRNAs in OSCC development and progression is not well understood. METHODS We sought to establish a dynamic Chinese hamster OSCC animal model, construct miRNA differential expression profiles of its occurrence and development, predict its targets, and perform functional analysis and validation in vitro. RESULTS Using expression and functional analyses, the key candidate miRNA (miR-181a-5p) was selected for further functional research, and the expression of miR-181a-5p in OSCC tissues and cell lines was detected. Subsequently, transfection technology and a nude mouse tumorigenic model were used to explore potential molecular mechanisms. miR-181a-5p was significantly downregulated in human OSCC specimens and cell lines, and decreased miR-181a-5p expression was observed in multiple stages of the Chinese hamster OSCC animal model. Moreover, upregulated miR-181a-5p significantly inhibited OSCC cell proliferation, colony formation, invasion, and migration; blocked the cell cycle; and promoted apoptosis. BCL2 was identified as a target of miR-181a-5p. BCL2 may interact with apoptosis- (BAX), invasion- and migration- (TIMP1, MMP2, and MMP9), and cell cycle-related genes (KI67, E2F1, CYCLIND1, and CDK6) to further regulate biological behavior. Tumor xenograft analysis indicated that tumor growth was significantly inhibited in the high miR-181a-5p expression group. CONCLUSION Our findings indicate that miR-181a-5p can be used as a potential biomarker and provide a novel animal model for mechanistic research on oral cancer.
Collapse
Affiliation(s)
- Guoqiang Xu
- grid.263452.40000 0004 1798 4018Laboratory Animal Center, Shanxi Key Laboratory of Experimental Animal Science and Human Disease Animal Model, Shanxi Medical University, Road Xinjian 56, Taiyuan, 030001 China ,grid.263452.40000 0004 1798 4018Shanxi Medical University School of Basic Medical Science, Taiyuan, 030001 China
| | - Yiyan Yang
- grid.263452.40000 0004 1798 4018Laboratory Animal Center, Shanxi Key Laboratory of Experimental Animal Science and Human Disease Animal Model, Shanxi Medical University, Road Xinjian 56, Taiyuan, 030001 China ,grid.263452.40000 0004 1798 4018Shanxi Medical University School of Basic Medical Science, Taiyuan, 030001 China
| | - Junting Yang
- grid.263452.40000 0004 1798 4018Laboratory Animal Center, Shanxi Key Laboratory of Experimental Animal Science and Human Disease Animal Model, Shanxi Medical University, Road Xinjian 56, Taiyuan, 030001 China ,grid.263452.40000 0004 1798 4018Shanxi Medical University School of Basic Medical Science, Taiyuan, 030001 China
| | - Lanfei Xiao
- grid.263452.40000 0004 1798 4018Laboratory Animal Center, Shanxi Key Laboratory of Experimental Animal Science and Human Disease Animal Model, Shanxi Medical University, Road Xinjian 56, Taiyuan, 030001 China
| | - Xiaotang Wang
- grid.263452.40000 0004 1798 4018Laboratory Animal Center, Shanxi Key Laboratory of Experimental Animal Science and Human Disease Animal Model, Shanxi Medical University, Road Xinjian 56, Taiyuan, 030001 China
| | - Litao Qin
- grid.263452.40000 0004 1798 4018Laboratory Animal Center, Shanxi Key Laboratory of Experimental Animal Science and Human Disease Animal Model, Shanxi Medical University, Road Xinjian 56, Taiyuan, 030001 China
| | - Jiping Gao
- grid.263452.40000 0004 1798 4018Laboratory Animal Center, Shanxi Key Laboratory of Experimental Animal Science and Human Disease Animal Model, Shanxi Medical University, Road Xinjian 56, Taiyuan, 030001 China
| | - Ruijing Xuan
- grid.263452.40000 0004 1798 4018Laboratory Animal Center, Shanxi Key Laboratory of Experimental Animal Science and Human Disease Animal Model, Shanxi Medical University, Road Xinjian 56, Taiyuan, 030001 China
| | - Xiaofen Wu
- grid.263452.40000 0004 1798 4018Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001 China
| | - Zhaoyang Chen
- grid.263452.40000 0004 1798 4018Laboratory Animal Center, Shanxi Key Laboratory of Experimental Animal Science and Human Disease Animal Model, Shanxi Medical University, Road Xinjian 56, Taiyuan, 030001 China
| | - Rui Sun
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China.
| | - Guohua Song
- Laboratory Animal Center, Shanxi Key Laboratory of Experimental Animal Science and Human Disease Animal Model, Shanxi Medical University, Road Xinjian 56, Taiyuan, 030001, China.
| |
Collapse
|
7
|
Swedan HK, Kassab AE, Gedawy EM, Elmeligie SE. Design, synthesis, and biological evaluation of novel ciprofloxacin derivatives as potential anticancer agents targeting topoisomerase II enzyme. J Enzyme Inhib Med Chem 2023; 38:118-137. [PMID: 36305290 PMCID: PMC9635472 DOI: 10.1080/14756366.2022.2136172] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
A series of novel ciprofloxacin (CP) derivatives substituted at the N-4 position with biologically active moieties were designed and synthesised. 14 compounds were 1.02- to 8.66-fold more potent than doxorubicin against T-24 cancer cells. Ten compounds were 1.2- to 7.1-fold more potent than doxorubicin against PC-3 cancer cells. The most potent compounds 6, 7a, 7b, 8a, 9a, and 10c showed significant Topo II inhibitory activity (83-90% at 100 μM concentration). Compounds 6, 8a, and 10c were 1.01- to 2.32-fold more potent than doxorubicin. Compounds 6 and 8a induced apoptosis in T-24 (16.8- and 20.1-fold, respectively compared to control). This evidence was supported by an increase in the level of apoptotic caspase-3 (5.23- and 7.6-fold, sequentially). Both compounds arrested the cell cycle in the S phase in T-24 cancer cells while in PC-3 cancer cells the two compounds arrested the cell cycle in the G1 phase. Molecular docking simulations of compounds 6 and 8a into the Topo II active site rationalised their remarkable Topo II inhibitory activity.
Collapse
Affiliation(s)
- Hadeer K. Swedan
- Central Administration of Research and Health Development, Ministry of Health, and Population (MoHP), Cairo, Egypt
| | - Asmaa E. Kassab
- Faculty of Pharmacy, Department of Pharmaceutical Organic Chemistry, Cairo University, Cairo, Egypt
| | - Ehab M. Gedawy
- Faculty of Pharmacy, Department of Pharmaceutical Organic Chemistry, Cairo University, Cairo, Egypt
- Faculty of Pharmacy and Pharmaceutical Industries, Department of Pharmaceutical Chemistry, Badr University in Cairo (BUC), Badr City, Egypt
| | - Salwa E. Elmeligie
- Faculty of Pharmacy, Department of Pharmaceutical Organic Chemistry, Cairo University, Cairo, Egypt
| |
Collapse
|
8
|
Abdelhaleem EF, Kassab AE, El-Nassan HB, Khalil OM. Design, synthesis, and biological evaluation of new celecoxib analogs as apoptosis inducers and cyclooxygenase-2 inhibitors. Arch Pharm (Weinheim) 2022; 355:e2200190. [PMID: 35976138 DOI: 10.1002/ardp.202200190] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 07/13/2022] [Accepted: 07/20/2022] [Indexed: 11/06/2022]
Abstract
Series of new celecoxib analogs were synthesized to assess their anticancer activity against the MCF-7 cell line. Four compounds, 3a, 3c, 5b, and 5c, showed 1.4-9.2-fold more potent anticancer activity than celecoxib. The antiproliferative activity of the most potent compounds, 3c, 5b, and 5c, seems to be associated well with their ability to induce apoptosis in MCF-7 cells (18-24-fold). This evidence was supported by an increase in the expression of the tumor suppressor gene p53 (4-6-fold), the elevation in the Bax/BCL-2 ratio, and a significant increase in the level of active caspase-7 (4-7-fold). Moreover, compounds 3c and 5c showed significant cyclooxygenase-2 (COX-2) inhibitory activity. They were also docked into the crystal structure of the COX-2 enzyme (PDB ID: 3LN1) to understand their mode of binding.
Collapse
Affiliation(s)
- Eman F Abdelhaleem
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Asmaa E Kassab
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Hala B El-Nassan
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Omneya M Khalil
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
9
|
Sun Y, Zhang J, Wang Y, Wang L, Song M, Khan A, Zhang L, Niu B, Zhao H, Li M, Luo T, He Q, Xie X, Liu Z, Xie J. miR-222-3p is involved in neural tube closure by directly targeting Ddit4 in RA induced NTDs mouse model. Cell Cycle 2021; 20:2372-2386. [PMID: 34779712 DOI: 10.1080/15384101.2021.1982506] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
Previously our results showed miR-222-3p was significantly downregulated in retinoic acid-induced neural tube defect (NTD) mouse model through transcriptome. Down-regulation of miR-222-3p may be a causative biomarker in NTDs. In this study, RNA was extracted from mouse embryos at E8.5, E9.5 and E10.5, and the expression level of miR-222-3p was measured by quantitative real-time PCR analysis. The preliminary mechanism of miR-222-3p in NTDs involved in cell proliferation, apoptosis and migration was investigated in mouse HT-22 cell line. The expression of miR-222-3p was significantly decreased at E8.5, E9.5 and E10.5 developed in mouse embryos which were consistent with our transcriptome sequencing. Suppression of miR-222-3p in HT-22 cells resulted in the inhibition of cell proliferation and migration, cell cycle and apoptosis. Moreover, DNA damage transcript 4 (Ddit4) was identified as a direct and functional target of miR-222-3p. miR-222-3p is negatively regulated by Ddit4. The mutation of binding site of Ddit4 3'UTR abrogated the responsiveness of luciferase reporters to miR-222-3p and showed that Ddit4 expression partially attenuated the function of miR-222-3p. We preliminatively confirmed that low expression of miR-222-3p has reduced the expression of β-catenin, TCF4 and other related genes in the Wnt/β-catenin signaling pathway.Collectively, these results demonstrated that miR-222-3p regulates the Wnt/β-catenin signaling pathway through Ddit4 inhibition in HT-22 cells, resulted in cell proliferation and apoptosis imbalance, and thus led to neural tube defects.
Collapse
Affiliation(s)
- Yuqing Sun
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth, Defect and Cell Regeneration, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Juan Zhang
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth, Defect and Cell Regeneration, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yufei Wang
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth, Defect and Cell Regeneration, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Lei Wang
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth, Defect and Cell Regeneration, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Meiyan Song
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth, Defect and Cell Regeneration, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Ajab Khan
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth, Defect and Cell Regeneration, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Li Zhang
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth, Defect and Cell Regeneration, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Bo Niu
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth, Defect and Cell Regeneration, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Hong Zhao
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth, Defect and Cell Regeneration, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Meining Li
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth, Defect and Cell Regeneration, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Tiane Luo
- Department of Statistics, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Qiwei He
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth, Defect and Cell Regeneration, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xianghui Xie
- Municipal Key Laboratory of Child Development and Nutriomic, Capital Institute of Pediatrics, Beijing, China
| | - Zhizhen Liu
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth, Defect and Cell Regeneration, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jun Xie
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth, Defect and Cell Regeneration, Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
10
|
Paysant H, Hedir S, Justaud F, Weiswald LB, El Dine AN, Soulieman A, Hachem A, Elie N, Brotin E, Denoyelle C, Bignon J, Roussi F, Jouanne M, Tasseau O, Roisnel T, Voisin-Chiret AS, Grée R, Levoin N, Poulain L. Structural revision of the Mcl-1 inhibitor MIM1: synthesis and biological studies on ovarian cancer cells with evaluation of designed analogues. Org Biomol Chem 2021; 19:8968-8987. [PMID: 34596646 DOI: 10.1039/d1ob01521d] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In the area of cancer research, the development of new and potent inhibitors of anti-apoptotic proteins is a very active and promising topic. The small molecule MIM1 has been reported earlier as one of the first selective inhibitors of the anti-apoptotic protein Mcl-1. In the present paper, we first revised the structure of this molecule based on extensive physicochemical analyses. Then we designed and synthesized a focused library of analogues for the corrected structure of MIM1. Next, these molecules were subjected to a panel of in cellulo biological studies, allowing the identification of dual Bcl-xL/Mcl-1 inhibitors, as well as selective Mcl-1 inhibitors. These results have been complemented by fluorescence polarization assays with the Mcl-1 protein. Preliminary structure-activity relationships were discussed and extensive molecular modelling studies allowed us to propose a rationale for the biological activity of this series of new inhibitors, in particular for the selectivity of inhibition of Mcl-1 versus Bcl-xL.
Collapse
Affiliation(s)
- Hippolyte Paysant
- Normandie Univ, UNICAEN, Inserm U1086 ANTICIPE "Unité de Recherche Interdisciplinaire pour la Prévention et le Traitement des Cancers", Caen, France
- UNICANCER, Centre de Lutte Contre le Cancer F. Baclesse, 3 avenue du Général Harris, 14076, Caen, France
| | - Siham Hedir
- Normandie Univ, UNICAEN, Inserm U1086 ANTICIPE "Unité de Recherche Interdisciplinaire pour la Prévention et le Traitement des Cancers", Caen, France
- UNICANCER, Centre de Lutte Contre le Cancer F. Baclesse, 3 avenue du Général Harris, 14076, Caen, France
| | - Frédéric Justaud
- Univ Rennes, CNRS, ISCR (Institut des Sciences Chimiques de Rennes), UMR 6226, F-35000, Rennes, France.
| | - Louis Bastien Weiswald
- Normandie Univ, UNICAEN, Inserm U1086 ANTICIPE "Unité de Recherche Interdisciplinaire pour la Prévention et le Traitement des Cancers", Caen, France
- UNICANCER, Centre de Lutte Contre le Cancer F. Baclesse, 3 avenue du Général Harris, 14076, Caen, France
| | - Assaad Nasr El Dine
- Univ Rennes, CNRS, ISCR (Institut des Sciences Chimiques de Rennes), UMR 6226, F-35000, Rennes, France.
- Laboratoire de Chimie Médicinale et de Produits Naturels, Université Libanaise, Faculté des Sciences et PRASE-EDST, Hadath, Beyrouth, Liban
| | - Ali Soulieman
- Univ Rennes, CNRS, ISCR (Institut des Sciences Chimiques de Rennes), UMR 6226, F-35000, Rennes, France.
- Laboratoire de Chimie Médicinale et de Produits Naturels, Université Libanaise, Faculté des Sciences et PRASE-EDST, Hadath, Beyrouth, Liban
| | - Ali Hachem
- Laboratoire de Chimie Médicinale et de Produits Naturels, Université Libanaise, Faculté des Sciences et PRASE-EDST, Hadath, Beyrouth, Liban
| | - Nicolas Elie
- Normandie Univ, UNICAEN, SF 4206 ICORE, CMABIO3, Caen, France
| | - Emilie Brotin
- Normandie Univ, UNICAEN, Inserm U1086 ANTICIPE "Unité de Recherche Interdisciplinaire pour la Prévention et le Traitement des Cancers", Caen, France
- UNICANCER, Centre de Lutte Contre le Cancer F. Baclesse, 3 avenue du Général Harris, 14076, Caen, France
- Normandie Univ, UNICAEN, SF 4206 ICORE, CMABIO3, Caen, France
- Normandie Univ, UNICAEN, SF4206 ICORE, Plateforme ImpedanCELL, Caen, France
| | - Christophe Denoyelle
- Normandie Univ, UNICAEN, Inserm U1086 ANTICIPE "Unité de Recherche Interdisciplinaire pour la Prévention et le Traitement des Cancers", Caen, France
- UNICANCER, Centre de Lutte Contre le Cancer F. Baclesse, 3 avenue du Général Harris, 14076, Caen, France
- Normandie Univ, UNICAEN, SF 4206 ICORE, CMABIO3, Caen, France
- Normandie Univ, UNICAEN, SF4206 ICORE, Plateforme ImpedanCELL, Caen, France
| | - Jérôme Bignon
- Institut de Chimie des Substances Naturelles CNRS UPR 2301, Université Paris Saclay, Gif-sur-Yvette, France
| | - Fanny Roussi
- Institut de Chimie des Substances Naturelles CNRS UPR 2301, Université Paris Saclay, Gif-sur-Yvette, France
| | - Marie Jouanne
- Normandie Univ, UNICAEN, Centre d'Etudes et de Recherche sur le Médicament de Normandie (CERMN), 14000 Caen, France
| | - Olivier Tasseau
- Univ Rennes, CNRS, ISCR (Institut des Sciences Chimiques de Rennes), UMR 6226, F-35000, Rennes, France.
| | - Thierry Roisnel
- Univ Rennes, CNRS, ISCR (Institut des Sciences Chimiques de Rennes), UMR 6226, F-35000, Rennes, France.
| | - Anne Sophie Voisin-Chiret
- Normandie Univ, UNICAEN, Centre d'Etudes et de Recherche sur le Médicament de Normandie (CERMN), 14000 Caen, France
| | - René Grée
- Univ Rennes, CNRS, ISCR (Institut des Sciences Chimiques de Rennes), UMR 6226, F-35000, Rennes, France.
| | - Nicolas Levoin
- Bioprojet-Biotech, 4 rue du Chesnay Beauregard, BP 96205, 35762, Saint Grégoire, France
| | - Laurent Poulain
- Normandie Univ, UNICAEN, Inserm U1086 ANTICIPE "Unité de Recherche Interdisciplinaire pour la Prévention et le Traitement des Cancers", Caen, France
- UNICANCER, Centre de Lutte Contre le Cancer F. Baclesse, 3 avenue du Général Harris, 14076, Caen, France
| |
Collapse
|
11
|
Design and Synthesis of Novel Betulin Derivatives Containing Thio-/Semicarbazone Moieties as Apoptotic Inducers through Mitochindria-Related Pathways. Molecules 2021; 26:molecules26216356. [PMID: 34770765 PMCID: PMC8587101 DOI: 10.3390/molecules26216356] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/11/2021] [Accepted: 10/19/2021] [Indexed: 11/17/2022] Open
Abstract
Two new series of betulin derivatives with semicarbazone (7a-g) or thiosemicarbazone (8a-g) groups at the C-28 position were synthesized. All compounds were evaluated for their in vitro cytotoxicities in human hepatocellular carcinoma cells (HepG2), human breast carcinoma cells (MCF-7), human lung carcinoma cells (A549), human colorectal cells (HCT-116) and normal human gastric epithelial cells (GES-1). Among these compounds, 8f displayed the most potent cytotoxicity with an IC50 value of 5.86 ± 0.61 μM against MCF-7 cells. Furthermore, the preliminary mechanism studies in MCF-7 cells showed that compound 8f could trigger the intracellular mitochondrial-mediated apoptosis pathway by losing MMP level, which was related with the upregulation of Bax, P53 and cytochrome c expression; the downregulation of Bcl-2 expression; activation of the expression levels of caspase-3, caspase-9, cleaved caspase-3 and cleaved caspase-9; and an increase in the amounts of intracellular reactive oxygen species. These results indicated that compound 8f may be used as a valuable skeleton structure for developing novel antitumor agents.
Collapse
|
12
|
Hua Z, White J, Zhou J. Cancer stem cells in TNBC. Semin Cancer Biol 2021; 82:26-34. [PMID: 34147641 DOI: 10.1016/j.semcancer.2021.06.015] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 06/07/2021] [Accepted: 06/14/2021] [Indexed: 12/24/2022]
Abstract
Triple-negative breast cancer (TNBC) is a broad collection of breast cancer that tests negative for estrogen receptors (ER), progesterone receptors (PR), and excess human epidermal growth factor receptor 2 (HER2) protein. TNBC is considered to have poorer prognosis than other types of breast cancer because of a lack of effective therapeutic targets. The success of precision cancer therapies relies on the clarification of key molecular mechanisms that drive tumor growth and metastasis; however, TNBC is highly heterogeneous in terms of their cellular lineage composition and the molecular nature within each individual case. In particular, the rare and sometimes slow cycling cancer stem cells (CSCs) can provide effective means for TNBC to resist various treatments. Single cell analysis technologies, including single-cell RNA-seq (scRNA-seq) and proteomics, provide an avenue to unravel patient-level intratumoral heterogeneity by identifying CSCs populations, CSC biomarkers and the range of tumor microenvironment cellular constituents that contribute to tumor growth. This review discusses the emerging evidence for the role of CSCs in driving TNBC incidence and the therapeutic implications in manipulating molecular signaling against this rare cell population for the control of this deadly disease.
Collapse
Affiliation(s)
- Zhan Hua
- Department of General Surgery, China-Japan Friendship Hospital, Beijing, 100029, People's Republic of China
| | - Jason White
- Tuskegee University, Center for Cancer Research, Tuskegee, AL, 36830, USA
| | - Jianjun Zhou
- Research Center for Translational Medicine, Cancer Stem Cell Institute, East Hospital, Tongji University School of Medicine, Shanghai, 200120, People's Republic of China.
| |
Collapse
|
13
|
Ansari MA, Thiruvengadam M, Farooqui Z, Rajakumar G, Sajid Jamal QM, Alzohairy MA, Almatroudi A, Alomary MN, Chung IM, Al-Suhaimi EA. Nanotechnology, in silico and endocrine-based strategy for delivering paclitaxel and miRNA: Prospects for the therapeutic management of breast cancer. Semin Cancer Biol 2021; 69:109-128. [PMID: 31891780 DOI: 10.1016/j.semcancer.2019.12.022] [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: 10/04/2019] [Revised: 12/06/2019] [Accepted: 12/25/2019] [Indexed: 02/07/2023]
Abstract
Breast cancer is one of the most prevalent and reoccurring cancers and the second most common reason of death in women. Despite advancements in therapeutic strategies for breast cancer, early tumor recurrence and metastasis in patients indicate resistance to chemotherapeutic medicines, such as paclitaxel due to the abnormal expression of ER and EGF2 in breast cancer cells. Therefore, the development of alternatives to paclitaxel is urgently needed to overcome challenges involving drug resistance. An increasing number of studies has revealed miRNAs as novel natural alternative substances that play a crucial role in regulating several physiological processes and have a close, adverse association with several diseases, including breast cancer. Due to the therapeutic potential of miRNA and paclitaxel in cancer research, the current review focuses on the differential roles of various miRNAs in breast cancer development and treatment. miRNA delivery to a specific target site, the development of paclitaxel and miRNA formulations, and nanotechnological strategies for the delivery of nanopaclitaxel in the management of breast cancer are discussed. These strategies involve improving the cellular uptake and bioavailability and reducing the toxicity of free paclitaxel to achieve accumulation tumor site. Furthermore, a molecular docking study was performed to ascertain the enhanced anticancer activity of the nanoformulation of ANG1005 and Abraxane. An in silico analysis revealed that ANG1005 and Abraxane nanoformulations have superior and significantly enhanced interactions with the proteins α-tubulin and Bcl-2. Therefore, ANG1005 and Abraxane may be more suitable in the therapeutic management of breast cancer than the existing free paclitaxel. miRNAs can revert abnormal gene expression to normalcy; since miRNAs serve as tumor suppressors. Therefore, restoration of particular miRNAs levels as a replacement therapy may be an effective endocrine potential strategy for treating ER positive/ negative breast cancers.
Collapse
Affiliation(s)
- Mohammad Azam Ansari
- Department of Epidemic Diseases Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam, 31441, Saudi Arabia.
| | - Muthu Thiruvengadam
- Department of Crop Science, College of Sanghuh Life Science, Konkuk University, Seoul 05029, Republic of Korea.
| | - Zeba Farooqui
- College of Pharmacy, University of Houston, Houston, TX, 77204, United States
| | - Govindaswamy Rajakumar
- Department of Crop Science, College of Sanghuh Life Science, Konkuk University, Seoul 05029, Republic of Korea.
| | - Qazi Mohammad Sajid Jamal
- Department of Health Informatics, College of Public Health and Health Informatics, Qassim University, Al-Bukayriyah, Saudi Arabia
| | - Mohammad A Alzohairy
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Qassim 51431, Saudi Arabia
| | - Ahmad Almatroudi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Qassim 51431, Saudi Arabia
| | - Mohammad N Alomary
- National Center of Biotechnology, Life Science and Environmental Research Institute, King Abdulaziz City for Science and Technology, P.O. Box 6086, Riyadh, Saudi Arabia
| | - Ill-Min Chung
- Department of Crop Science, College of Sanghuh Life Science, Konkuk University, Seoul 05029, Republic of Korea.
| | - Ebtesam Abdullah Al-Suhaimi
- Department of Biology, College of Science, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam, Saudi Arabia; Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam, 31441, Saudi Arabia
| |
Collapse
|
14
|
Mirzaei S, Gholami MH, Mahabady MK, Nabavi N, Zabolian A, Banihashemi SM, Haddadi A, Entezari M, Hushmandi K, Makvandi P, Samarghandian S, Zarrabi A, Ashrafizadeh M, Khan H. Pre-clinical investigation of STAT3 pathway in bladder cancer: Paving the way for clinical translation. Biomed Pharmacother 2020; 133:111077. [PMID: 33378975 DOI: 10.1016/j.biopha.2020.111077] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/24/2020] [Accepted: 11/27/2020] [Indexed: 02/07/2023] Open
Abstract
Effective cancer therapy requires identification of signaling networks and investigating their potential role in proliferation and invasion of cancer cells. Among molecular pathways, signal transducer and activator of transcription 3 (STAT3) has been of importance due to its involvement in promoting proliferation, and invasion of cancer cells, and mediating chemoresistance. In the present review, our aim is to reveal role of STAT3 pathway in bladder cancer (BC), as one of the leading causes of death worldwide. In respect to its tumor-promoting role, STAT3 is able to enhance the growth of BC cells via inhibiting apoptosis and cell cycle arrest. STAT3 also contributes to metastasis of BC cells via upregulating of MMP-2 and MMP-9 as well as genes in the EMT pathway. BC cells obtain chemoresistance via STAT3 overexpression and its inhibition paves the way for increasing efficacy of chemotherapy. Different molecular pathways such as KMT1A, EZH2, DAB2IP and non-coding RNAs including microRNAs and long non-coding RNAs can function as upstream mediators of STAT3 that are discussed in this review article.
Collapse
Affiliation(s)
- Sepideh Mirzaei
- Department of Biology, Faculty of Science, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | | | - Mahmood Khaksary Mahabady
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Noushin Nabavi
- Research Services, University of Victoria, Victoria, BC, V8W 2Y2, Canada
| | - Amirhossein Zabolian
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | | | - Amirabbas Haddadi
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Maliheh Entezari
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology & Zoonoses, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Pooyan Makvandi
- IstitutoItaliano di Tecnologia, Centre for Micro-BioRobotics, viale Rinaldo Piaggio 34, 56025, Pontedera, Pisa, Italy
| | - Saeed Samarghandian
- Department of Basic Medical Sciences, Neyshabur University of Medical Sciences, Neyshabur, Iran
| | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, 34956, Istanbul, Turkey.
| | - Milad Ashrafizadeh
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, 34956, Istanbul, Turkey; Faculty of Engineering and Natural Sciences, Sabanci University, OrtaMahalle, ÜniversiteCaddesi No. 27, Orhanlı, Tuzla, 34956, Istanbul, Turkey.
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University, Mardan, 23200, Pakistan.
| |
Collapse
|
15
|
Yan L, Li K, Feng Z, Zhang Y, Han R, Ma J, Zhang J, Wu X, Liu H, Jiang Y, Zhang Y, Zhu Y. lncRNA CERS6-AS1 as ceRNA promote cell proliferation of breast cancer by sponging miR-125a-5p to upregulate BAP1 expression. Mol Carcinog 2020; 59:1199-1208. [PMID: 32808708 DOI: 10.1002/mc.23249] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 07/27/2020] [Accepted: 08/07/2020] [Indexed: 12/21/2022]
Abstract
Long noncoding RNAs (lncRNAs) can act as oncogene and tumor suppressor genes in many types of cancers including breast cancer (BC). Our previous study has indicated microRNA (miR)-125a-5p was downregulated and function as a tumor suppressor in BC. However, its upstream regulation mechanism is still unclear. In this study, we used bioinformatics algorithms, RNA pulldown assay, and dual-luciferase reports assay to predict and confirm lncRNA CERS6-AS1 interacted with miR-125a-5p. Then we found CERS6-AS1 was upregulated in BC tissues. Experimental results of tumor growth in nude mice show that CERS6-AS1 promotes tumor growth. Furthermore, CERS6-AS1 regulated BC susceptibility gene 1-associated protein 1 (BAP1) expression via sponging miR-125a-5p via Western blot analysis and quantitative polymerase chain reaction arrays. Finally, we showed that miR-125a-5p had opposing effects to those of CERS6-AS1 on BC cells, demonstrating that CERS6-AS1 may promote cell proliferation and inhibit cell apoptosis via sponging miR-125a-5p. Our results indicated CERS6-AS1 promote BC cell proliferation and inhibit cell apoptosis via sponging miR-125a-5p to upregulate BAP1 expression.
Collapse
Affiliation(s)
- Liang Yan
- Anhui Provincial Key Laboratory of Biological Macro-molecules Research, Wannan Medical College, Wuhu, Anhui, China
| | - Kai Li
- Department of Clinical Diagnostics, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui, China
| | - Zunyong Feng
- Anhui Provincial Key Laboratory of Biological Macro-molecules Research, Wannan Medical College, Wuhu, Anhui, China
| | - Yizongheng Zhang
- Department of Clinical Medicine, The First College of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Renrui Han
- Anhui Provincial Key Laboratory of Biological Macro-molecules Research, Wannan Medical College, Wuhu, Anhui, China
| | - Jinzhu Ma
- Anhui Provincial Key Laboratory of Biological Macro-molecules Research, Wannan Medical College, Wuhu, Anhui, China
| | - Jieling Zhang
- Department of Clinical Diagnostics, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui, China
| | - Xu Wu
- Anhui Provincial Key Laboratory of Biological Macro-molecules Research, Wannan Medical College, Wuhu, Anhui, China
| | - Haijun Liu
- Anhui Provincial Key Laboratory of Biological Macro-molecules Research, Wannan Medical College, Wuhu, Anhui, China
| | - Yuxin Jiang
- Anhui Provincial Key Laboratory of Biological Macro-molecules Research, Wannan Medical College, Wuhu, Anhui, China
| | - Yao Zhang
- Anhui Provincial Key Laboratory of Biological Macro-molecules Research, Wannan Medical College, Wuhu, Anhui, China
| | - Yiping Zhu
- Department of Clinical Diagnostics, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui, China
| |
Collapse
|
16
|
Izadpanah MR, Salehzadeh A, Zaefizadeh M, Nikpasand M. Functionalisation of Fe 3O 4 nanoparticles by 2-((pyrazol-4-yl) methylene) hydrazinecarbothioamide enhances the apoptosis of human breast cancer MCF-7 cells. IET Nanobiotechnol 2020; 14:508-518. [PMID: 32755961 DOI: 10.1049/iet-nbt.2019.0199] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Cancer is a major cause of death. Thus, the incidence and mortality rate of cancer is globally important. Regarding vast problems caused by chemotherapy drugs, efforts have progressed to find new anti-cancer drugs. Pyrazole derivatives are known as components with anti-cancer properties. In here, Fe3O4 nanoparticles were first functionalized with (3-chloropropyl) trimethoxysilane, then 2-((pyrazol-4-yl) methylene) hydrazinecarbothioamide (P) was anchored on the surface of magnetic nanoparticles (PL). The synthesized nano-compounds were characterized using Fourier transform infrared spectroscopy, X-ray diffraction, scanning electron microscopy, Zeta potential, dynamic light scattering, and energy-dispersive x-ray spectrometry analyses. The cytotoxicity effect was evaluated using MTT assay, apoptosis test by Flow cytometry, cell cycle analysis, Caspase-3 activity assay and Hoechst staining on MCF-7 cell line. The high toxicity for tumor cells and low toxicity on normal cells (MCF10A) was considered as an important feature (selectivity index, 10.9). Based on results, the IC50 for P and PL compounds were 157.80 and 131.84 μM/ml respectively. Moreover, apoptosis inducing, nuclear fragmentation, Caspase 3 activity and induction of cell rest in sub-G1 and S phases, were also observed. The inhibitory effect of PL was significantly higher than P, which could be due to the high penetrability of Fe3O4 nanoparticles.
Collapse
Affiliation(s)
| | - Ali Salehzadeh
- Department of Biology, Rasht Branch, Islamic Azad University, Rasht, Iran.
| | - Mohammad Zaefizadeh
- Department of Biology, Ardabil Branch, Islamic Azad University, Ardabil, Iran
| | - Mohammad Nikpasand
- Department of Chemistry, Rasht Branch, Islamic Azad University, Rasht, Iran
| |
Collapse
|
17
|
Wang G, Dong Y, Liu H. Curcumol enhances the anti-tumor effects of metformin via suppressing epithelial-mesenchymal transition in triple-negative breast cancer. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:946. [PMID: 32953746 PMCID: PMC7475397 DOI: 10.21037/atm-20-5438] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Background Triple-negative breast cancer (TNBC) is a severe disease with a high mortality rate. Metformin has been found to possess anti-tumor properties. Curcumol, an active ingredient extracted from curcuma, exerts the protective effect in TNBC cells through inducing apoptosis. However, the effects of curcumol combined with metformin on the treatment of TNBC have yet to be fully established. Methods TNBC cells MDA-MB-231 and MDA-MB-468 cells were used in the study. TNBC cells were treated with curcumol and metformin alone or treated with curcumol combined with metformin. Cell viability was determined using Cell Counting Kit-8 (CCK-8) assay. Cell apoptosis was detected using terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay. The levels of proteins were measured using Western blot. Wound healing assay and Transwell invasion assays were used to determine cell migration and invasion ability, respectively. A xenograft model was established to investigate the tumor growth ability. Immunohistochemistry was performed to determine the expression of Ki-67 and Vascular endothelial growth factor (VEGF). Results In the study, the administration of curcumol alone had no significant effects on the TNBC cells. However, the anti-proliferation, anti-metastasis, and anti-epithelial-mesenchymal transition (EMT) effects of metformin were enhanced by the addition of curcumol. Further, curcumol reversed TNBC cell proliferation, migration, invasion, and EMT induced by rucaparib, and enhanced the effect of metformin on rucaparib-induced TNBC cells. The combination of curcumol and metformin also suppressed tumor growth, EMT marker expression, and the activation of Wnt2/β-Catenin signaling during in vivo experiments. Conclusions The combination of curcumol and metformin enhances the anti-tumor effects of metformin on TNBC via inhibiting EMT. Curcumol combined with metformin may hold promise as a therapeutic strategy for TNBC.
Collapse
Affiliation(s)
- Gangyue Wang
- Department of Breast, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Yi Dong
- Department of Breast, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Heng Liu
- Department of Breast, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
18
|
Hong HC, Chuang CH, Huang WC, Weng SL, Chen CH, Chang KH, Liao KW, Huang HD. A panel of eight microRNAs is a good predictive parameter for triple-negative breast cancer relapse. Theranostics 2020; 10:8771-8789. [PMID: 32754277 PMCID: PMC7392022 DOI: 10.7150/thno.46142] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 06/16/2020] [Indexed: 12/14/2022] Open
Abstract
Rationale: Triple-negative breast cancer (TNBC), which has the highest recurrence rate and shortest survival time of all breast cancers, is in urgent need of a risk assessment method to determine an accurate treatment course. Recently, miRNA expression patterns have been identified as potential biomarkers for diagnosis, prognosis, and personalized therapy. Here, we investigate a combination of candidate miRNAs as a clinically applicable signature that can precisely predict relapse in TNBC patients after surgery. Methods: Four total cohorts of training (TCGA_TNBC and GEOD-40525) and validation (GSE40049 and GSE19783) datasets were analyzed with logistic regression and Gaussian mixture analyses. We established a miRNA signature risk model and identified an 8-miRNA signature for the prediction of TNBC relapse. Results: The miRNA signature risk model identified ten candidate miRNAs in the training set. By combining 8 of the 10 miRNAs (miR-139-5p, miR-10b-5p, miR-486-5p, miR-455-3p, miR-107, miR-146b-5p, miR-324-5p and miR-20a-5p), an accurate predictive model of relapse in TNBC patients was established and was highly correlated with prognosis (AUC of 0.80). Subsequently, this 8-miRNA signature prognosticated relapse in the two validation sets with AUCs of 0.89 and 0.90. Conclusion: The 8-miRNA signature predictive model may help clinicians provide a prognosis for TNBC patients with a high risk of recurrence after surgery and provide further personalized treatment to decrease the chance of relapse.
Collapse
Affiliation(s)
- Hsiao-Chin Hong
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Guangdong Province 518172, China
- School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen, Guangdong Province 518172, China
| | - Cheng-Hsun Chuang
- Institute of Molecular Medicine and Bioengineering, National Chiao Tung University, Hsinchu City 30068, Taiwan, ROC
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu City 30068, Taiwan, ROC
| | - Wei-Chih Huang
- Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsinchu City 30068, Taiwan, ROC
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu City 30068, Taiwan, ROC
- Come True Biomedical Inc., Taichung 408, Taiwan, ROC
| | - Shun-Long Weng
- Department of Obstetrics and Gynecology, Hsinchu MacKay Memorial Hospital, Hsinchu City 300, Taiwan, ROC
- Department of Medicine, MacKay Medical College, New Taipei City 252, Taiwan, ROC
- MacKay Junior College of Medicine, Nursing and Management College, Taipei City 112, Taiwan, ROC
| | - Chia-Hung Chen
- Department of Medical Research, Hsinchu Mackay Memorial Hospital, Hsinchu City 30071, Taiwan, ROC
| | - Kuang-Hsin Chang
- Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsinchu City 30068, Taiwan, ROC
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu City 30068, Taiwan, ROC
| | - Kuang-Wen Liao
- Institute of Molecular Medicine and Bioengineering, National Chiao Tung University, Hsinchu City 30068, Taiwan, ROC
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu City 30068, Taiwan, ROC
- Center for Intelligent Drug Systems and Smart Bio-Devices, National Chiao Tung University, Hsinchu City 30068, Taiwan, ROC
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan, ROC
| | - Hsien-Da Huang
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Guangdong Province 518172, China
- School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen, Guangdong Province 518172, China
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu City 30068, Taiwan, ROC
| |
Collapse
|
19
|
Alexandrova E, Lamberti J, Saggese P, Pecoraro G, Memoli D, Mirici Cappa V, Ravo M, Iorio R, Tarallo R, Rizzo F, Collina F, Cantile M, Di Bonito M, Botti G, Nassa G, Weisz A, Giurato G. Small Non-Coding RNA Profiling Identifies miR-181a-5p as a Mediator of Estrogen Receptor Beta-Induced Inhibition of Cholesterol Biosynthesis in Triple-Negative Breast Cancer. Cells 2020; 9:cells9040874. [PMID: 32260128 PMCID: PMC7226848 DOI: 10.3390/cells9040874] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 03/30/2020] [Accepted: 04/01/2020] [Indexed: 12/17/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is a highly heterogeneous disease, representing the most aggressive breast cancer (BC) subtype with limited treatment options due to a lack of estrogen receptor alpha (ERα), progesterone receptor (PR), and Erb-B2 receptor tyrosine kinase 2 (HER2/neu) expression. Estrogen receptor beta (ERβ) is present in a fraction of TNBC patients, where its expression correlates with improved patient outcomes, supported by the fact that it exerts oncosuppressive effects in TNBC cell models in vitro. ERβ is involved in microRNA-mediated regulation of gene expression in hormone-responsive BC cells and could mediate its actions through small noncoding RNAs (sncRNAs) in TNBCs also. To verify this possibility, smallRNA sequencing was performed on three ERβ-expressing cell lines from different TNBC molecular subtypes. Several sncRNAs resulted modulated by ERβ, with a subset being regulated in a tumor subtype-independent manner. Interestingly, sncRNA profiling of 12 ERβ+and 32 ERβ− primary TNBC biopsies identified 7 microRNAs, 1 PIWI-interacting RNA (piRNA), and 1 transfer RNA (tRNA) differentially expressed in ERβ+ compared to ERβ− tumors and cell lines. Among them, miR-181a-5p was found to be overexpressed in ERβ+ tumors and predicted target key components of the cholesterol biosynthesis pathway previously found to be inhibited by ERβ in TNBC cells.
Collapse
Affiliation(s)
- Elena Alexandrova
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry ‘Scuola Medica Salernitana’, University of Salerno, 84081 Baronissi, Italy
- Genomix4Life Srl, 84081 Baronissi, Italy
| | - Jessica Lamberti
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry ‘Scuola Medica Salernitana’, University of Salerno, 84081 Baronissi, Italy
| | - Pasquale Saggese
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry ‘Scuola Medica Salernitana’, University of Salerno, 84081 Baronissi, Italy
- Division of Pulmonary and Critical Care Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Giovanni Pecoraro
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry ‘Scuola Medica Salernitana’, University of Salerno, 84081 Baronissi, Italy
| | - Domenico Memoli
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry ‘Scuola Medica Salernitana’, University of Salerno, 84081 Baronissi, Italy
| | - Valeria Mirici Cappa
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry ‘Scuola Medica Salernitana’, University of Salerno, 84081 Baronissi, Italy
| | - Maria Ravo
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry ‘Scuola Medica Salernitana’, University of Salerno, 84081 Baronissi, Italy
- Genomix4Life Srl, 84081 Baronissi, Italy
| | | | - Roberta Tarallo
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry ‘Scuola Medica Salernitana’, University of Salerno, 84081 Baronissi, Italy
| | - Francesca Rizzo
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry ‘Scuola Medica Salernitana’, University of Salerno, 84081 Baronissi, Italy
| | - Francesca Collina
- Pathology Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy
| | - Monica Cantile
- Pathology Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy
| | - Maurizio Di Bonito
- Pathology Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy
| | - Gerardo Botti
- Scientific Direction, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy
| | - Giovanni Nassa
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry ‘Scuola Medica Salernitana’, University of Salerno, 84081 Baronissi, Italy
| | - Alessandro Weisz
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry ‘Scuola Medica Salernitana’, University of Salerno, 84081 Baronissi, Italy
- Correspondence: (A.W.); (G.G.); Tel.: + 39-089-965043 (A.W.)
| | - Giorgio Giurato
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry ‘Scuola Medica Salernitana’, University of Salerno, 84081 Baronissi, Italy
- Correspondence: (A.W.); (G.G.); Tel.: + 39-089-965043 (A.W.)
| |
Collapse
|