1
|
Ye N, Huang J, Zhang Y, Yang Y. Ghrelin suppresses apoptosis and autophagy in osteoarthritis synovial cells by modulating the ADORA2B/PI3K/Akt/mTOR signaling pathway. J Orthop 2025; 68:27-33. [PMID: 39995546 PMCID: PMC11846923 DOI: 10.1016/j.jor.2025.01.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 01/13/2025] [Accepted: 01/26/2025] [Indexed: 02/26/2025] Open
Abstract
Given the pivotal role that apoptosis and autophagy play in the pathogenesis of osteoarthritis (OA), the current study aims to examine the regulatory effects of ghrelin on these processes via the ADORA2B/PI3K/Akt/mTOR signaling pathway. Serum levels of ghrelin were measured in both OA patients and healthy controls using an ELISA kit. Cell proliferation was evaluated through the Cell Counting Kit-8 (CCK-8) assay, while Western blot analysis was utilized to determine the expression levels of autophagy-related proteins (LC3II/I, BECLIN-1) and apoptosis markers (BAX, Bcl-2), as well as to assess the activation status of the PI3K/Akt/mTOR signaling pathway in OA synovial cells. These analyses were performed under conditions of ADORA2B and mTOR silencing, as well as in control settings. The results revealed that ghrelin expression was significantly reduced in the serum of OA patients. Furthermore, ghrelin was found to enhance synovial cell proliferation while simultaneously inhibiting apoptosis and autophagy, as evidenced by lowered expression levels of LC3/I, BECLIN-1, and BAX, alongside an increase in Bcl-2 expression. This modulation occurred through the regulation of the PI3K/Akt/mTOR signaling pathway mediated by ADORA2B. These findings underscore the role of ghrelin in the progression of osteoarthritis by influencing synovial cell activity through the ADORA2B/PI3K/Akt/mTOR pathway, thus laying the groundwork for investigating targeted therapeutic strategies in clinical practice.
Collapse
Affiliation(s)
- Nan Ye
- Department of Orthopaedic Surgery Center B, The Second Affiliated Hospital of Inner Mongolia Medical University, Keerqin Road No. 59, Hohhot, 010090, PR China
| | - Jian Huang
- Department of Orthopaedic Surgery Center B, The Second Affiliated Hospital of Inner Mongolia Medical University, Keerqin Road No. 59, Hohhot, 010090, PR China
| | - Yuanzhi Zhang
- Department of Orthopaedic Surgery Center B, The Second Affiliated Hospital of Inner Mongolia Medical University, Keerqin Road No. 59, Hohhot, 010090, PR China
| | - Yifeng Yang
- Department of Orthopaedic Surgery Center B, The Second Affiliated Hospital of Inner Mongolia Medical University, Keerqin Road No. 59, Hohhot, 010090, PR China
| |
Collapse
|
2
|
Li X, Niu C, Yi G, Zhang Y, Jin W, Zhang Z, Zhang W, Li B. Quercetin inhibits the epithelial-mesenchymal transition and reverses CDK4/6 inhibitor resistance in breast cancer by regulating circHIAT1/miR-19a-3p/CADM2 axis. PLoS One 2024; 19:e0305612. [PMID: 38990915 PMCID: PMC11239024 DOI: 10.1371/journal.pone.0305612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 05/31/2024] [Indexed: 07/13/2024] Open
Abstract
Breast cancer (BC) cells have a high risk of metastasis due to epithelial-mesenchymal transition (EMT). Palbociclib (CDK4/6 inhibitor) is an approved drug for BC treatment. However, the drug resistance and metastasis can impair the treatment outcome of Palbociclib. Understanding the mechanisms of EMT and Palbociclib drug resistance in BC is conducive to the formulation of novel therapeutic strategy. Here, we investigated the role of circHIAT1/miR-19a-3p/CADM2 axis in modulating EMT and Palbociclib resistance in BC. circHIAT1 and CADM2 were down-regulated in BC tissues and cell lines, and miR-19a-3p showed an up-regulation. circHIAT1 could interact with miR-19a-3p and suppress its activity, while miR-19a-3p functioned to negatively regulate CADM2. Forced over-expression of circHIAT1 could impaired the EMT status and migratory ability of BC cells, and this effect was inhibited by miR-19a-3p mimic. In addition, we also generated Palbociclib resistant BC cells, and showed that circHIAT1 and CADM2 were down-regulated in the resistant BC cells while miR-19a-3p showed an up-regulation. Forced circHIAT1 over-expression re-sensitized BC cells to Palbociclib treatment. Quercetin, a bioactive flavonoid, could suppressed the migration and invasion of BC cells, and re-sensitized BC cells to Palbociclib. The anti-cancer effect of quercetin could be attributed to its regulatory effect on circHIAT1/miR-19a-3p/CADM2 axis. In vivo tumorigenesis experiment further revealed that quercetin administration enhanced the anti-cancer effect of Palbociclib, an effect was dependent on the up-regulation of circHIAT1 by quercetin. In summary, this study identified quercetin as a potential anti-cancer compound to reverse Palbociclib resistance and impair EMT in BC cells by targeting circHIAT1/miR-19a-3p/CADM2 axis.
Collapse
Affiliation(s)
- Xiaogang Li
- Department of General Surgery, Affiliated Hospital of Yunnan University, Kunming, Yunnan, China
| | - Chao Niu
- Department of General Surgery, Affiliated Hospital of Yunnan University, Kunming, Yunnan, China
| | - Guoqiang Yi
- Department of General Surgery, Affiliated Hospital of Yunnan University, Kunming, Yunnan, China
| | - Yuan Zhang
- Department of General Surgery, Affiliated Hospital of Yunnan University, Kunming, Yunnan, China
| | - Wendi Jin
- Department of General Surgery, Affiliated Hospital of Yunnan University, Kunming, Yunnan, China
| | - Zhiping Zhang
- Department of General Surgery, Affiliated Hospital of Yunnan University, Kunming, Yunnan, China
| | - Wanfu Zhang
- Department of General Surgery, Affiliated Hospital of Yunnan University, Kunming, Yunnan, China
| | - Bo Li
- Department of General Surgery, Affiliated Hospital of Yunnan University, Kunming, Yunnan, China
| |
Collapse
|
3
|
Ma Q, Ye S, Liu H, Zhao Y, Mao Y, Zhang W. HMGA2 promotes cancer metastasis by regulating epithelial-mesenchymal transition. Front Oncol 2024; 14:1320887. [PMID: 38361784 PMCID: PMC10867147 DOI: 10.3389/fonc.2024.1320887] [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: 10/13/2023] [Accepted: 01/09/2024] [Indexed: 02/17/2024] Open
Abstract
Epithelial-mesenchymal transition (EMT) is a complex physiological process that transforms polarized epithelial cells into moving mesenchymal cells. Dysfunction of EMT promotes the invasion and metastasis of cancer. The architectural transcription factor high mobility group AT-hook 2 (HMGA2) is highly overexpressed in various types of cancer (e.g., colorectal cancer, liver cancer, breast cancer, uterine leiomyomas) and significantly correlated with poor survival rates. Evidence indicated that HMGA2 overexpression markedly decreased the expression of epithelial marker E-cadherin (CDH1) and increased that of vimentin (VIM), Snail, N-cadherin (CDH2), and zinc finger E-box binding homeobox 1 (ZEB1) by targeting the transforming growth factor beta/SMAD (TGFβ/SMAD), mitogen-activated protein kinase (MAPK), and WNT/beta-catenin (WNT/β-catenin) signaling pathways. Furthermore, a new class of non-coding RNAs (miRNAs, circular RNAs, and long non-coding RNAs) plays an essential role in the process of HMGA2-induced metastasis and invasion of cancer by accelerating the EMT process. In this review, we discuss alterations in the expression of HMGA2 in various types of cancer. Furthermore, we highlight the role of HMGA2-induced EMT in promoting tumor growth, migration, and invasion. More importantly, we discuss extensively the mechanism through which HMGA2 regulates the EMT process and invasion in most cancers, including signaling pathways and the interacting RNA signaling axis. Thus, the elucidation of molecular mechanisms that underlie the effects of HMGA2 on cancer invasion and patient survival by mediating EMT may offer new therapeutic methods for preventing cancer progression.
Collapse
Affiliation(s)
- Qing Ma
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
| | - Sisi Ye
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
| | - Hong Liu
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
| | - Yu Zhao
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
| | - Yan Mao
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
| | - Wei Zhang
- Emergency Department of West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
| |
Collapse
|
4
|
De Martino M, Pellecchia S, Decaussin-Petrucci M, Testa D, Meireles Da Costa N, Pallante P, Chieffi P, Fusco A, Esposito F. Drug-induced inhibition of HMGA and EZH2 activity as a possible therapy for anaplastic thyroid carcinoma. Cell Cycle 2023; 22:2552-2565. [PMID: 38165007 PMCID: PMC10936675 DOI: 10.1080/15384101.2023.2298027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 11/22/2023] [Accepted: 12/14/2023] [Indexed: 01/03/2024] Open
Abstract
Anaplastic thyroid carcinoma (ATC) is one of the most aggressive and lethal neoplasms in humans, and just limited progresses have been made to extend patient survival and decrease ATC-associated mortality. Thus, the identification of novel therapeutic strategies for treating ATC is needed. Recently, our group has identified two proteins with oncogenic activity, namely HMGA1 and EZH2, with pivotal roles in ATC cancer progression. Therefore, we tested the ability of trabectedin, a HMGA1-targeting drug, and GSK126, an inhibitor of EZH2 enzymatic activity, to impair cell viability of four ATC-derived cell lines. In the present study, we first confirmed the overexpression of HMGA1 and EZH2 in all ATC-derived cell lines and tissues compared to the normal primary thyroid cells and tissues. Then, treatment of the ATC cell lines with trabectedin and GSK126 resulted in a drastic induction of apoptotic cell death, which increased when the ATC cell lines were treated with a combination of both drugs. Conversely, normal primary human thyroid cells did not show any significant reduction in their viability when exposed to the same drugs. Noteworthy, both drugs induced the deregulation of EZH2- and HMGA1-controlled genes. Altogether, these findings propose the combination of trabectedin and GSK126 as possible novel strategy for ATC therapy.
Collapse
Affiliation(s)
- Marco De Martino
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche (DMMBM), Istituto per l’Endocrinologia e l’Oncologia Sperimentale (IEOS) “G. Salvatore”, Consiglio Nazionale delle Ricerche (CNR) c/o, Università degli Studi di Napoli “Federico II”, Naples, Italy
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Simona Pellecchia
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche (DMMBM), Istituto per l’Endocrinologia e l’Oncologia Sperimentale (IEOS) “G. Salvatore”, Consiglio Nazionale delle Ricerche (CNR) c/o, Università degli Studi di Napoli “Federico II”, Naples, Italy
| | | | - Domenico Testa
- Clinic of Otorhinolaryngology, Head and Neck Surgery Unit, Department of Anesthesiology, Surgical and Emergency Science, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Nathalia Meireles Da Costa
- Programa de Carcinogênese Molecular, Instituto Nacional de Câncer - INCA, Rua André Cavalcanti, Rio de Janeiro, Brazil
| | - Pierlorenzo Pallante
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche (DMMBM), Istituto per l’Endocrinologia e l’Oncologia Sperimentale (IEOS) “G. Salvatore”, Consiglio Nazionale delle Ricerche (CNR) c/o, Università degli Studi di Napoli “Federico II”, Naples, Italy
| | - Paolo Chieffi
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Alfredo Fusco
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche (DMMBM), Istituto per l’Endocrinologia e l’Oncologia Sperimentale (IEOS) “G. Salvatore”, Consiglio Nazionale delle Ricerche (CNR) c/o, Università degli Studi di Napoli “Federico II”, Naples, Italy
- Programa de Carcinogênese Molecular, Instituto Nacional de Câncer - INCA, Rua André Cavalcanti, Rio de Janeiro, Brazil
| | - Francesco Esposito
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche (DMMBM), Istituto per l’Endocrinologia e l’Oncologia Sperimentale (IEOS) “G. Salvatore”, Consiglio Nazionale delle Ricerche (CNR) c/o, Università degli Studi di Napoli “Federico II”, Naples, Italy
| |
Collapse
|
5
|
Lu H, Xie D, Qu B, Li M, He Y, Liu W. Emodin prevents renal ischemia-reperfusion injury via suppression of p53-mediated cell apoptosis based on network pharmacology. Heliyon 2023; 9:e15682. [PMID: 37215853 PMCID: PMC10195913 DOI: 10.1016/j.heliyon.2023.e15682] [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: 08/31/2022] [Revised: 04/16/2023] [Accepted: 04/18/2023] [Indexed: 05/24/2023] Open
Abstract
Background Previous evidence indicated that emodin has significant advantages for preventing acute kidney injury (AKI). However, the mechanisms responsible for these effects of emodin have yet to be elucidated. Methods We first used network pharmacology and molecular docking to identify the core targets of emodin for AKI and performed a range of experiments to validate this result. Pretreatment with emodin for 7 days, the rats were treated with bilateral renal artery clipping for 45 min to identify the prevention effect. Hypoxia/reoxygenation (H/R), and vancomycin - induced renal tubular epithelial cells (HK-2 cells) were treated with emodin to explore the related molecular mechanism. Results Network pharmacology and molecular docking showed that anti-apoptosis might be the core mechanism responsible for the action of emodin on AKI; this anti-apoptotic effect appears to because by regulation p53-related signaling pathway. Our data showed that pretreatment with emodin significantly improved renal function and renal tubular injury in renal I/R model rats (P < 0.05. The prevention effect of emodin was proved to be related to anti - apoptosis of HK-2 cells, possibly by downregulating the levels of p53, cleaved-caspase-3, pro-caspase-9, and upregulated the levels of Bcl-2. The efficacy and mechanism of emodin on anti - apoptosis was also confirmed in vancomycin - induced HK-2 cells. Meanwhile, the data also showed that emodin promoted angiogenesis in I/R damaged kidneys and H/R-induced HK-2 cells, which was associated with decreasing HIF-1α levels and increasing VEGF levels. Conclusions Our findings indicated that the preventive effect of emodin on AKI is probably attributable to anti-apoptosis response and promoting angiogenesis effect.
Collapse
Affiliation(s)
- Hongmei Lu
- Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100700, China
- Department of Clinical Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610072, China
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Beijing, 100700, China
| | - Dengpiao Xie
- Department of Clinical Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610072, China
| | - Bo Qu
- Department of Clinical Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610072, China
| | - Mingquan Li
- Department of Clinical Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610072, China
| | - Yuhua He
- Department of Clinical Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610072, China
| | - Weijing Liu
- Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100700, China
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Beijing, 100700, China
| |
Collapse
|
6
|
High Mobility Group A 1 Expression as a Poor Prognostic Marker Associated with Tumor Invasiveness in Gastric Cancer. Life (Basel) 2022; 12:life12050709. [PMID: 35629376 PMCID: PMC9146826 DOI: 10.3390/life12050709] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 04/30/2022] [Accepted: 05/06/2022] [Indexed: 01/05/2023] Open
Abstract
The prognosis of advanced gastric cancer remains poor. Overexpression of high mobility group A 1 (HMGA1) in breast cancer and neuroblastoma indicates a poor prognosis. However, the relationship between HMGA1 expression and gastric cancer development remains unclear. Treatment strategies can be developed by identifying potential markers associated with gastric cancer. We used a constructed tissue array and performed hematoxylin and eosin and immunohistochemical staining. We quantified the staining results and performed statistical analysis to evaluate the relationship between HMGA1 expression and prognosis. HMGA1 expression was related to the expression of Ki-67, caspase3, CD31, N-cadherin, fibronectin, pAkt, and pErk. In the Kaplan–Meier graph, higher HMGA1 expression levels were associated with a relatively poor survival rate (p = 0.04). High expression of HMGA1 leads to a low survival rate, which is associated with HMGA1, proliferation, apoptosis, angiogenesis, epithelial-mesenchymal transition, and tyrosine kinase.
Collapse
|
7
|
Mansoori B, Terp MG, Mohammadi A, Pedersen CB, Ditzel HJ, Baradaran B, Gjerstorff MF. HMGA2 Supports Cancer Hallmarks in Triple-Negative Breast Cancer. Cancers (Basel) 2021; 13:5197. [PMID: 34680349 PMCID: PMC8533747 DOI: 10.3390/cancers13205197] [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: 10/03/2021] [Accepted: 10/11/2021] [Indexed: 12/13/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer that exhibits a high proliferation rate and early metastasis leading to a poor prognosis. HMGA2 is a DNA binding transcriptional regulator implicated in tumorigenesis. Here, we demonstrate that the HMGA2 promoter is demethylated in TNBC tumors, leading to increased expression of HMGA2 at both mRNA and protein levels. Importantly, high HMGA2 levels in TNBC tumors are correlated with poor prognosis. To detail the role of HMGA2 in TNBC development and progression, we studied its effect on core cancer phenotypes. Stable knockdown of HMGA2 in TNBC cells revealed that HMGA2 may support cell proliferation, cell migration and invasion. In addition, HMGA2 knockdown decreased cancer stem cell (CSC) features. Importantly, we found that silencing HMGA2 inhibited NF-kB signaling and lead to decreased expression of the downstream molecules IL-6 and IL-8 and reduced STAT3 pathway activation. Our results demonstrate that HMGA2 supports cancer hallmarks in TNBC and may represent a promising target for TNBC treatment.
Collapse
Affiliation(s)
- Behzad Mansoori
- Immunology Research Center, Tabriz University of Medical Sciences, Golghasht St., Tabriz 51666-14731, Iran; (B.M.); (B.B.)
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, J. B. Winsløws Vej 25, 3, DK-5000 Odense C, Denmark; (M.G.T.); (A.M.); (C.B.P.); (H.J.D.)
- Aging Research Institute, Physical Medicine and Rehabilitation Research Center, Tabriz University of Medical Sciences, Golghasht St., Tabriz 51656-65811, Iran
| | - Mikkel Green Terp
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, J. B. Winsløws Vej 25, 3, DK-5000 Odense C, Denmark; (M.G.T.); (A.M.); (C.B.P.); (H.J.D.)
| | - Ali Mohammadi
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, J. B. Winsløws Vej 25, 3, DK-5000 Odense C, Denmark; (M.G.T.); (A.M.); (C.B.P.); (H.J.D.)
| | - Christina Bøg Pedersen
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, J. B. Winsløws Vej 25, 3, DK-5000 Odense C, Denmark; (M.G.T.); (A.M.); (C.B.P.); (H.J.D.)
| | - Henrik Jørn Ditzel
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, J. B. Winsløws Vej 25, 3, DK-5000 Odense C, Denmark; (M.G.T.); (A.M.); (C.B.P.); (H.J.D.)
- Department of Oncology, Odense University Hospital, DK-5000 Odense C, Denmark
- Academy of Geriatric Cancer Research (AgeCare), Odense University Hospital, DK-5000 Odense C, Denmark
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Golghasht St., Tabriz 51666-14731, Iran; (B.M.); (B.B.)
| | - Morten Frier Gjerstorff
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, J. B. Winsløws Vej 25, 3, DK-5000 Odense C, Denmark; (M.G.T.); (A.M.); (C.B.P.); (H.J.D.)
- Department of Oncology, Odense University Hospital, DK-5000 Odense C, Denmark
- Academy of Geriatric Cancer Research (AgeCare), Odense University Hospital, DK-5000 Odense C, Denmark
| |
Collapse
|
8
|
Mansoori B, Najafi S, Mohammadi A, AsadollahSeraj H, Savadi P, Mansoori B, Nazari A, Mokhtarzadeh A, Roshani E, Duijf PH, Cho WCS, Baradaran B. The synergy between miR-486-5p and tamoxifen causes profound cell death of tamoxifen-resistant breast cancer cells. Biomed Pharmacother 2021; 141:111925. [PMID: 34323695 DOI: 10.1016/j.biopha.2021.111925] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 06/08/2021] [Accepted: 07/12/2021] [Indexed: 11/24/2022] Open
Abstract
Breast cancer (BC) is the most common type of malignancy in women. A subset of breast cancers show resistance to endocrine-based therapies. The estrogen receptor (ER) plays a critical role in developing hormone-dependent BC. Loss of ER contributes to resistance to tamoxifen therapy and may contribute to mortality. Thus, it is crucial to overcome this problem. Here, using luciferase reporter assays, qRT-PCR, and Western blot analyses, we demonstrate that the microRNA miR-486-5p targets HMGA1 mRNA, decreasing its mRNA and protein levels in ER-positive (ER+) BC cells. Consistently, miR-486-5p is significantly downregulated, whereas HMGA1 is considerably upregulated in ER+ BC samples. Remarkably, while both miR-486-5p and tamoxifen individually cause G2/M cell cycle arrest, combination treatment synergistically causes profound cell death, specifically in tamoxifen-resistant ER+ cells but not in tamoxifen-sensitive ER+ cells. Combined treatment with miR-486-5p and tamoxifen also additively reduces cell migration, invasion, colony formation, mammary spheroid formation and a CD24-CD44+ cell population, representing decreased cancer stemness. However, these phenomena are independent of the tamoxifen responsiveness of the ER+ BC cells. Thus, miR-486-5p and tamoxifen exhibit additive and synergistic tumor-suppressive effects, most importantly causing profound cell death specifically in tamoxifen-resistant BC cells. Therefore, our work suggests that combining miR-486-5p replacement therapy with tamoxifen treatment is a promising strategy to treat endocrine therapy-resistant BC.
Collapse
Affiliation(s)
- Behzad Mansoori
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Souzan Najafi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Mohammadi
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | | | - Pouria Savadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behnaz Mansoori
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Afsaneh Nazari
- Department of Genetics, Faculty of Basic Sciences, Islamic Azad University, Zanjan Branch, Zanjan, Iran
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elmira Roshani
- Department of Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Pascal Hg Duijf
- Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Australia; Centre for Data Science, Queensland University of Technology, Brisbane, Australia; University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, Australia
| | | | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
9
|
Abedi Gaballu F, Cho WCS, Dehghan G, Zarebkohan A, Baradaran B, Mansoori B, Abbaspour-Ravasjani S, Mohammadi A, Sheibani N, Aghanejad A, Ezzati Nazhad Dolatabadi J. Silencing of HMGA2 by siRNA Loaded Methotrexate Functionalized Polyamidoamine Dendrimer for Human Breast Cancer Cell Therapy. Genes (Basel) 2021; 12:1102. [PMID: 34356120 PMCID: PMC8303903 DOI: 10.3390/genes12071102] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 07/10/2021] [Accepted: 07/16/2021] [Indexed: 02/06/2023] Open
Abstract
The transcription factor high mobility group protein A2 (HMGA2) plays an important role in the pathogenesis of some cancers including breast cancer. Polyamidoamine dendrimer generation 4 is a kind of highly branched polymeric nanoparticle with surface charge and highest density peripheral groups that allow ligands or therapeutic agents to attach it, thereby facilitating target delivery. Here, methotrexate (MTX)- modified polyamidoamine dendrimer generation 4 (G4) (G4/MTX) was generated to deliver specific small interface RNA (siRNA) for suppressing HMGA2 expression and the consequent effects on folate receptor (FR) expressing human breast cancer cell lines (MCF-7, MDA-MB-231). We observed that HMGA2 siRNA was electrostatically adsorbed on the surface of the G4/MTX nanocarrier for constructing a G4/MTX-siRNA nano-complex which was verified by changing the final particle size and zeta potential. The release of MTX and siRNA from synthesized nanocomplexes was found in a time- and pH-dependent manner. We know that MTX targets FR. Interestingly, G4/MTX-siRNA demonstrates significant cellular internalization and gene silencing efficacy when compared to the control. Besides, the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay demonstrated selective cell cytotoxicity depending on the folate receptor expressing in a dose-dependent manner. The gene silencing and protein downregulation of HMGA2 by G4/MTX-siRNA was observed and could significantly induce cell apoptosis in MCF-7 and MDA-MB-231 cancer cells compared to the control group. Based on the findings, we suggest that the newly developed G4/MTX-siRNA nano-complex may be a promising strategy to increase apoptosis induction through HMGA2 suppression as a therapeutic target in human breast cancer.
Collapse
Affiliation(s)
- Fereydoon Abedi Gaballu
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5166-15731, Iran; (F.A.G.); (B.B.); (B.M.)
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz 51666-16471, Iran
| | | | - Gholamreza Dehghan
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz 51666-16471, Iran
| | - Amir Zarebkohan
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz 5166-15731, Iran;
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5166-15731, Iran; (F.A.G.); (B.B.); (B.M.)
| | - Behzad Mansoori
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5166-15731, Iran; (F.A.G.); (B.B.); (B.M.)
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, 5230 Odense, Denmark;
| | | | - Ali Mohammadi
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, 5230 Odense, Denmark;
| | - Nader Sheibani
- McPherson Eye Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53726, USA;
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, WI 53726, USA
| | - Ayuob Aghanejad
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz 51666-16471, Iran;
| | | |
Collapse
|
10
|
Mansoori B, Duijf PHG, Mohammadi A, Safarzadeh E, Ditzel HJ, Gjerstorff MF, Cho WCS, Baradaran B. MiR-142-3p targets HMGA2 and suppresses breast cancer malignancy. Life Sci 2021; 276:119431. [PMID: 33785332 DOI: 10.1016/j.lfs.2021.119431] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/17/2021] [Accepted: 03/23/2021] [Indexed: 11/26/2022]
Abstract
MicroRNAs (miRNAs) have the ability to regulate gene expression programs in cells. Hence, altered expression of miRNAs significantly contributes to breast cancer development and progression. Here, we demonstrate that the miRNA miR-142-3p directly targets the 3' untranslated region of HMGA2, which encodes an onco-embryonic protein that is overexpressed in most cancers, including breast cancer. Down regulation of miR-142-3p predicting poor patient survival in grade 3 breast cancer (P-value = 0.045). MiR-142-3p downregulates HMGA2 mRNA and protein levels. Higher miR-142-3p and lower HMGA2 expressed are found in breast cancer versus normal breast tissue (P-value<0.05), and their levels inversely correlate in breast cancers (P-value = 1.46 × 10-4). We demonstrate that miR-142-3p induces apoptosis and G2/M cell cycle arrest in breast cancer cells. In addition, it inhibits breast cancer stem cell properties and decreases SOX2, NANOG, ALDH and c-Myc expression. MiR-142-3p also decreases cell proliferation through inhibition of the ERK/AKT/STAT3 signaling pathways. Finally, pathway analyses of patient samples suggest that these mechanisms also acting in the tumors of breast cancer patients. Thus, our work identifies HMGA2 as a direct miR-142-3p target and indicates that miR-142-3p is an important suppressor of breast cancer oncogenesis. This identifies miR-142-3p may candidate as a therapeutic molecule for breast cancer treatment.
Collapse
Affiliation(s)
- Behzad Mansoori
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark; Aging Research Institute, Physical Medicine and Rehabilitation Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Pascal H G Duijf
- Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Australia; University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, Australia
| | - Ali Mohammadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Elham Safarzadeh
- Department of Microbiology and Immunology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Henrik J Ditzel
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark; Department of Oncology, Odense University Hospital, Odense, Denmark; Academy of Geriatric Cancer Research (AgeCare), Odense University Hospital, Odense, Denmark
| | - Morten F Gjerstorff
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark; Department of Oncology, Odense University Hospital, Odense, Denmark; Academy of Geriatric Cancer Research (AgeCare), Odense University Hospital, Odense, Denmark
| | | | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
11
|
Derakhshani A, Rostami Z, Safarpour H, Shadbad MA, Nourbakhsh NS, Argentiero A, Taefehshokr S, Tabrizi NJ, Kooshkaki O, Astamal RV, Singh PK, Taefehshokr N, Alizadeh N, Silvestris N, Baradaran B. From Oncogenic Signaling Pathways to Single-Cell Sequencing of Immune Cells: Changing the Landscape of Cancer Immunotherapy. Molecules 2021; 26:2278. [PMID: 33920054 PMCID: PMC8071039 DOI: 10.3390/molecules26082278] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 04/09/2021] [Accepted: 04/09/2021] [Indexed: 12/19/2022] Open
Abstract
Over the past decade, there have been remarkable advances in understanding the signaling pathways involved in cancer development. It is well-established that cancer is caused by the dysregulation of cellular pathways involved in proliferation, cell cycle, apoptosis, cell metabolism, migration, cell polarity, and differentiation. Besides, growing evidence indicates that extracellular matrix signaling, cell surface proteoglycans, and angiogenesis can contribute to cancer development. Given the genetic instability and vast intra-tumoral heterogeneity revealed by the single-cell sequencing of tumoral cells, the current approaches cannot eliminate the mutating cancer cells. Besides, the polyclonal expansion of tumor-infiltrated lymphocytes in response to tumoral neoantigens cannot elicit anti-tumoral immune responses due to the immunosuppressive tumor microenvironment. Nevertheless, the data from the single-cell sequencing of immune cells can provide valuable insights regarding the expression of inhibitory immune checkpoints/related signaling factors in immune cells, which can be used to select immune checkpoint inhibitors and adjust their dosage. Indeed, the integration of the data obtained from the single-cell sequencing of immune cells with immune checkpoint inhibitors can increase the response rate of immune checkpoint inhibitors, decrease the immune-related adverse events, and facilitate tumoral cell elimination. This study aims to review key pathways involved in tumor development and shed light on single-cell sequencing. It also intends to address the shortcomings of immune checkpoint inhibitors, i.e., their varied response rates among cancer patients and increased risk of autoimmunity development, via applying the data from the single-cell sequencing of immune cells.
Collapse
Affiliation(s)
- Afshin Derakhshani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 51656-65811, Iran; (A.D.); (M.A.S.); (S.T.); (N.J.T.); (R.V.A.); (N.A.)
- IRCCS Istituto Tumori “Giovanni Paolo II” of Bari, 70124 Bari, Italy;
| | - Zeinab Rostami
- Student Research Committee, Birjand University of Medical Sciences, Birjand 97178-53577, Iran; (Z.R.); (O.K.)
| | - Hossein Safarpour
- Cellular & Molecular Research Center, Birjand University of Medical Sciences, Birjand 97178-53577, Iran;
| | - Mahdi Abdoli Shadbad
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 51656-65811, Iran; (A.D.); (M.A.S.); (S.T.); (N.J.T.); (R.V.A.); (N.A.)
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz 51666-14766, Iran
| | | | | | - Sina Taefehshokr
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 51656-65811, Iran; (A.D.); (M.A.S.); (S.T.); (N.J.T.); (R.V.A.); (N.A.)
| | - Neda Jalili Tabrizi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 51656-65811, Iran; (A.D.); (M.A.S.); (S.T.); (N.J.T.); (R.V.A.); (N.A.)
| | - Omid Kooshkaki
- Student Research Committee, Birjand University of Medical Sciences, Birjand 97178-53577, Iran; (Z.R.); (O.K.)
| | - Reza Vaezi Astamal
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 51656-65811, Iran; (A.D.); (M.A.S.); (S.T.); (N.J.T.); (R.V.A.); (N.A.)
| | - Pankaj Kumar Singh
- Principal Research Technologist, Department of Radiation Oncology, Mayo Clinic, 4500 San Pablo Rd S, Jacksonville, FL 32224, USA;
| | - Nima Taefehshokr
- Department of Microbiology and Immunology, Center for Human Immunology, The University of Western Ontario, London, ON N6A 5C1, Canada;
| | - Nazila Alizadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 51656-65811, Iran; (A.D.); (M.A.S.); (S.T.); (N.J.T.); (R.V.A.); (N.A.)
| | - Nicola Silvestris
- IRCCS Istituto Tumori “Giovanni Paolo II” of Bari, 70124 Bari, Italy;
- Department of Biomedical Sciences and Human Oncology, University of Bari “Aldo Moro”, 70124 Bari, Italy
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 51656-65811, Iran; (A.D.); (M.A.S.); (S.T.); (N.J.T.); (R.V.A.); (N.A.)
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz 51666-14766, Iran
| |
Collapse
|
12
|
Lotfinejad P, Kazemi T, Safaei S, Amini M, Roshani Asl E, Baghbani E, Sandoghchian Shotorbani S, Jadidi Niaragh F, Derakhshani A, Abdoli Shadbad M, Silvestris N, Baradaran B. PD-L1 silencing inhibits triple-negative breast cancer development and upregulates T-cell-induced pro-inflammatory cytokines. Biomed Pharmacother 2021; 138:111436. [PMID: 33667790 DOI: 10.1016/j.biopha.2021.111436] [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: 11/13/2020] [Revised: 02/21/2021] [Accepted: 02/23/2021] [Indexed: 12/22/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is an invasive tumor with a high incidence of distant metastasis and poor prognosis. In TNBC cells, high PD-L1 expression can induce an immunosuppressive tumor microenvironment, repressing the anti-tumoral immune responses. Although FDA-approved agents targeting the PD-1/PD-L1 axis are potent to eliminate tumoral cells, their immune-related adverse events have become worrisome. As the regulator of gene expression, siRNAs can directly target PD-L1 in breast cancer cells. The gene modification of tumoral PD-L1 can reduce our reliance on the current method of targeting the PD-L1/PD-1 axis. We initiated the study with bioinformatics analysis; the results indicated that TNBC and the MDA-MB-231 cells significantly overexpressed PD-L1 compared to other breast cancer subtypes and cell lines. Our results demonstrated that PD-L1 silencing substantially reduced PD-L1 expression at mRNA and protein levels in MDA-MB-231 cells. Moreover, our results demonstrated that PD-L1 knockdown reduced cancer cell proliferation and induced apoptosis via intrinsic and extrinsic apoptosis pathways. We observed that PD-L1 silencing effectively inhibited the migration of TNBC cells. Further investigation also displayed that silencing of PD-L1 in breast cancer cells induced T-cell cytotoxic function by upregulating the gene expression of pro-inflammatory cytokines, i.e., IL-2, IFN-γ, and TNF-α, and downregulating the gene expression of anti-inflammatory cytokines, i.e., IL-10, and TGF-β, in a co-culture system.
Collapse
Affiliation(s)
- Parisa Lotfinejad
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Tohid Kazemi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sahar Safaei
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Amini
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elmira Roshani Asl
- Department of Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Elham Baghbani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | - Afshin Derakhshani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; IRCCS Istituto Tumori "Giovanni Paolo II" of Bari, Italy
| | - Mahdi Abdoli Shadbad
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nicola Silvestris
- IRCCS Istituto Tumori "Giovanni Paolo II" of Bari, Italy; Department of Biomedical Sciences and Human Oncology DIMO-University of Bari, Bari, Italy.
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Tabriz University of Medical Sciences, Tabriz, Iran; Pharmaceutical Analysis Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
13
|
HMGA2 as a Critical Regulator in Cancer Development. Genes (Basel) 2021; 12:genes12020269. [PMID: 33668453 PMCID: PMC7917704 DOI: 10.3390/genes12020269] [Citation(s) in RCA: 111] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 02/01/2021] [Accepted: 02/08/2021] [Indexed: 02/07/2023] Open
Abstract
The high mobility group protein 2 (HMGA2) regulates gene expression by binding to AT-rich regions of DNA. Akin to other DNA architectural proteins, HMGA2 is highly expressed in embryonic stem cells during embryogenesis, while its expression is more limited at later stages of development and in adulthood. Importantly, HMGA2 is re-expressed in nearly all human malignancies, where it promotes tumorigenesis by multiple mechanisms. HMGA2 increases cancer cell proliferation by promoting cell cycle entry and inhibition of apoptosis. In addition, HMGA2 influences different DNA repair mechanisms and promotes epithelial-to-mesenchymal transition by activating signaling via the MAPK/ERK, TGFβ/Smad, PI3K/AKT/mTOR, NFkB, and STAT3 pathways. Moreover, HMGA2 supports a cancer stem cell phenotype and renders cancer cells resistant to chemotherapeutic agents. In this review, we discuss these oncogenic roles of HMGA2 in different types of cancers and propose that HMGA2 may be used for cancer diagnostic, prognostic, and therapeutic purposes.
Collapse
|
14
|
Ma W, Xue N, Zhang J, Wang D, Yao X, Lin L, Xu Q. circUBAP2 regulates osteosarcoma progression via the miR‑204‑3p/HMGA2 axis. Int J Oncol 2021; 58:298-311. [PMID: 33650644 PMCID: PMC7864148 DOI: 10.3892/ijo.2021.5178] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 01/19/2021] [Indexed: 12/11/2022] Open
Abstract
Circular RNA (circRNA/circ)-ubiquitin associated protein 2 (UBAP2), a newly recognized circRNA, serves a functional role in several types of tumor, including ovarian cancer, colorectal cancer and osteosarcoma. However, the precise roles and molecular mechanism under-lying circUBAP2 in osteosarcoma (OS) are not completely understood. In the present study, the expression levels of circUBAP2, microRNA (miR)-204-3p and (HMGA2) were evaluated via reverse transcription-quantitative PCR in OS tissues and cells. OS cell proliferation, migration, invasion and apoptosis were assessed by performing Cell Counting Kit-8, Transwell and flow cytometry assays, respectively. HMGA2 protein expression levels were determined via western blot-ting. Dual-luciferase reporter assays were performed to verify the interaction between circUBAP2 and miR-204-3p, and between miR-204-3p and HMGA2. An RNA immunoprecipitation (RIP) assay was conducted to confirm the interaction between circUBAP2 and miR-204-3p. The results demonstrated that circUBAP2 expression was significantly upregulated in OS tissues and cell lines compared with para-cancerous tissues and hFOB1.19 cells, respectively. In addition, high circUBAP2 expression levels in patients with OS were associated with a lower survival rate compared with lower expression levels in patients with OS. The functional assays revealed that circUBAP2 knockdown significantly inhibited OS cell proliferation, migration and invasion, but increased OS cell apoptosis compared with the small interfering RNA-negative control (si-NC) group. The dual-luciferase reporter and RIP assay results confirmed that circUBAP2 bound to miR-204-3p. Moreover, miR-204-3p expression was significantly downregulated in OS tissues compared with paracancerous tissues, and miR-204-3p expression was negatively correlated with circUBAP2 expression in OS tissues. Collectively, the results demonstrated that miR-204-3p was associated with circUBAP2 knockdown-mediated inhibition of OS cell malignant behavior. Moreover, miR-204-3p was also identified as one of the direct targets of HMGA2. Collectively, the results indicated that compared with the si-NC group, circUBAP2 knockdown significantly inhibited OS cell malignant behavior by binding to miR-204-3p, which subsequently regulated HMGA2 expression. Therefore, the present study demonstrated that circUBAP2 expression was upregulated in OS, and circUBAP2 regulated OS cell malignant behavior via the miR-204-3p/HMGA2 axis.
Collapse
Affiliation(s)
- Weiguo Ma
- Department of Clinical Laboratory, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan 450008, P.R. China
| | - Ning Xue
- Department of Clinical Laboratory, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan 450008, P.R. China
| | - Junhua Zhang
- Department of Clinical Laboratory, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan 450008, P.R. China
| | - Dan Wang
- Department of Clinical Laboratory, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan 450008, P.R. China
| | - Xiaobin Yao
- Department of Clinical Laboratory, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan 450008, P.R. China
| | - Lin Lin
- Department of Clinical Laboratory, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan 450008, P.R. China
| | - Qingxia Xu
- Department of Clinical Laboratory, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan 450008, P.R. China
| |
Collapse
|
15
|
Synthesis and in vitro and in vivo biological evaluation of novel derivatives of flexicaulin A as antiproliferative agents. Eur J Med Chem 2020; 208:112789. [PMID: 32883640 DOI: 10.1016/j.ejmech.2020.112789] [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: 06/14/2020] [Revised: 08/25/2020] [Accepted: 08/25/2020] [Indexed: 11/21/2022]
Abstract
As our research focuses on anticancer drugs, a series of novel derivatives of flexicaulin A (FA), an ent-kaurene diterpene, condensed with an aromatic ring were synthesized, and their antiproliferative activities against four human cancer cell lines (TE-1, EC109, MCF-7, and MGC-803) were evaluated. The activities of most of the new compounds were better than those of FA. Compound 2y exhibited the best activity with an IC50 value reaching 0.13 μM against oesophageal cancer cells (EC109 cells). The IC50 values for 2y in normal cells (GES-1 cells and HUVECs) were 0.52 μM and 0.49 μM, respectively. Subsequent mechanistic investigations found that compound 2y can inhibit the proliferation of cancer cells and cell cloning. In addition, 2y could reduce the mitochondrial membrane potential, increase the apoptosis rate, and increase the ROS level in EC109 cells. Moreover, 2y can upregulate the expression of ROS/JNK pathway-related proteins (p-ASK1, p-MKK4, p-JNK, and p-Cjun (ser63)) and pro-apoptotic proteins (Bax, Bad, and Bim). In vivo experiments showed that 2y can inhibit tumour growth in nude mice. The mechanism involves an increase in protein expression in the ROS pathway, leading to changes in apoptosis-related proteins. In addition, compound 2y shows low toxicity. These results indicate that compound 2y holds promising potential as an antiproliferative agent.
Collapse
|
16
|
Pegoraro S, Ros G, Sgubin M, Petrosino S, Zambelli A, Sgarra R, Manfioletti G. Targeting the intrinsically disordered architectural High Mobility Group A (HMGA) oncoproteins in breast cancer: learning from the past to design future strategies. Expert Opin Ther Targets 2020; 24:953-969. [PMID: 32970506 DOI: 10.1080/14728222.2020.1814738] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Triple-negative breast cancer (TNBC) is the most difficult breast cancer subtype to treat because of its heterogeneity and lack of specific therapeutic targets. High Mobility Group A (HMGA) proteins are chromatin architectural factors that have multiple oncogenic functions in breast cancer, and they represent promising molecular therapeutic targets for this disease. AREAS COVERED We offer an overview of the strategies that have been exploited to counteract HMGA oncoprotein activities at the transcriptional and post-transcriptional levels. We also present the possibility of targeting cancer-associated factors that lie downstream of HMGA proteins and discuss the contribution of HMGA proteins to chemoresistance. EXPERT OPINION Different strategies have been exploited to counteract HMGA protein activities; these involve interfering with their nucleic acid binding properties and the blocking of HMGA expression. Some approaches have provided promising results. However, some unique characteristics of the HMGA proteins have not been exploited; these include their extensive protein-protein interaction network and their intrinsically disordered status that present the possibility that HMGA proteins could be involved in the formation of proteinaceous membrane-less organelles (PMLO) by liquid-liquid phase separation. These unexplored characteristics could open new pharmacological avenues to counteract the oncogenic contributions of HMGA proteins.
Collapse
Affiliation(s)
- Silvia Pegoraro
- Department of Life Sciences, University of Trieste , Trieste, Italy
| | - Gloria Ros
- Department of Life Sciences, University of Trieste , Trieste, Italy
| | - Michela Sgubin
- Department of Life Sciences, University of Trieste , Trieste, Italy
| | - Sara Petrosino
- Department of Life Sciences, University of Trieste , Trieste, Italy
| | | | - Riccardo Sgarra
- Department of Life Sciences, University of Trieste , Trieste, Italy
| | | |
Collapse
|
17
|
Ghrelin promotes the osteogenic differentiation of rMSCs via miR-206 and the ERK1/2 pathway. Cytotechnology 2020; 72:707-713. [PMID: 32700168 DOI: 10.1007/s10616-020-00413-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 07/19/2020] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE Mesenchymal stem cells (MSCs) can differentiate into chondroblasts, adipocytes, or osteoblasts under appropriate stimulation. Ghrelin, an endogenous ligand for the growth hormone secretagogue receptor (GHSR), stimulates growth hormone (GH) secretion and exerts both orexigenic and adipogenic effects. The ERK1/2 signaling pathway is known to trigger osteogenic differentiation of rabbit bone marrow-derived mesenchymal stromal cells. In the present study, the function of miR-206 in the ghrelin-mediated osteogenic differentiation of rabbit bone marrow-derived mesenchymal stromal cells (rMSCs) was explored. METHODS The expression of miR-206 was detected by qPCR, and phosphorylated ERK1/2 and the protein expression levels of ALP, RUNX2, and Osterix were assessed by western blotting. RESULTS Ghrelin inhibited the expression of miR-206 to promote the osteogenic differentiation of rMSCs. Moreover, ghrelin increased the phosphorylation of ERK1/2, while overexpression of miR-206 suppressed ERK1/2 phosphorylation, indicating that miR-206 can regulate the ERK1/2 pathway. Further, inhibition of ERK1/2 had no influence on miR-206 expression; however, the phosphorylation of ERK1/2 was decreased, and the protein expression levels of ALP, RUNX2, and Osterix were downregulated. CONCLUSIONS Ghrelin promotes the osteogenic differentiation of rMSCs via miR-206 and the ERK1/2 pathway.
Collapse
|
18
|
Aali M, Mesgarzadeh AH, Najjary S, Abdolahi HM, Kojabad AB, Baradaran B. Evaluating the role of microRNAs alterations in oral squamous cell carcinoma. Gene 2020; 757:144936. [PMID: 32640301 DOI: 10.1016/j.gene.2020.144936] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 05/31/2020] [Accepted: 07/01/2020] [Indexed: 12/16/2022]
Abstract
Oral squamous cell carcinoma (OSCC) accounts for nearly 90 percent of oral cavity malignancies and is one of the most widespread oral cancers in the world. The microRNAs (miRNAs or miRs) have an important role in cellular processes comprising cell cycle, differentiation, and also apoptosis. MiRNAs are also implicated in the progression of cancers, including OSCC, through a variety of signaling pathways. One of the most significant signaling pathways in OSCC is the PI3K / Akt pathway that has been illustrated to be under the tight regulation of miRNAs. Deregulation or activation of the PI3K / Akt pathway due to mutations has been revealed to be implicated in the development of oral cancer. According to studies, more than 47% of HNSCC and around 38% of OSCC samples indicate at least one molecular alteration in this signaling pathway. The potential of miRNAs for their use as therapeutic tools in the diagnosis as well as treatment of numerous diseases have been confirmed. In the current review, we summarize miRNAs and their possible mechanisms as well as their functions in OSCC advancement and progression.
Collapse
Affiliation(s)
- Mehdi Aali
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Tabriz University of Medical Sciences, Tabriz, Iran; Dental and Periodontal Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Hossein Mesgarzadeh
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Tabriz University of Medical Sciences, Tabriz, Iran; Dental and Periodontal Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shiva Najjary
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Mashhadi Abdolahi
- Tabriz Health Services Management Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
19
|
Unachukwu U, Chada K, D’Armiento J. High Mobility Group AT-Hook 2 (HMGA2) Oncogenicity in Mesenchymal and Epithelial Neoplasia. Int J Mol Sci 2020; 21:ijms21093151. [PMID: 32365712 PMCID: PMC7246488 DOI: 10.3390/ijms21093151] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 04/27/2020] [Accepted: 04/28/2020] [Indexed: 12/31/2022] Open
Abstract
High mobility group AT-hook 2 (HMGA2) has been associated with increased cell proliferation and cell cycle dysregulation, leading to the ontogeny of varied tumor types and their metastatic potentials, a frequently used index of disease prognosis. In this review, we deepen our understanding of HMGA2 pathogenicity by exploring the mechanisms by which HMGA2 misexpression and ectopic expression induces mesenchymal and epithelial tumorigenesis respectively and distinguish the pathogenesis of benign from malignant mesenchymal tumors. Importantly, we highlight the regulatory role of let-7 microRNA family of tumor suppressors in determining HMGA2 misexpression events leading to tumor pathogenesis and focused on possible mechanisms by which HMGA2 could propagate lymphangioleiomyomatosis (LAM), benign mesenchymal tumors of the lungs. Lastly, we discuss potential therapeutic strategies for epithelial and mesenchymal tumorigenesis based on targeting the HMGA2 signaling pathway.
Collapse
Affiliation(s)
- Uchenna Unachukwu
- Department of Anesthesiology, Columbia University Medical Center, 630 West 168th Street, P&S 12-402, New York, NY 10032, USA;
| | - Kiran Chada
- Department of Biochemistry & Molecular Biology; Robert Wood Johnson Medical School, 675 Hoes Lane, Piscataway, NJ 08854, USA;
| | - Jeanine D’Armiento
- Department of Anesthesiology, Columbia University Medical Center, 630 West 168th Street, P&S 12-402, New York, NY 10032, USA;
- Correspondence: ; Tel.: +212-305-3745
| |
Collapse
|
20
|
Kooshkaki O, Rezaei Z, Rahmati M, Vahedi P, Derakhshani A, Brunetti O, Baghbanzadeh A, Mansoori B, Silvestris N, Baradaran B. MiR-144: A New Possible Therapeutic Target and Diagnostic/Prognostic Tool in Cancers. Int J Mol Sci 2020; 21:ijms21072578. [PMID: 32276343 PMCID: PMC7177921 DOI: 10.3390/ijms21072578] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 04/02/2020] [Accepted: 04/04/2020] [Indexed: 12/14/2022] Open
Abstract
MicroRNAs (miRNAs) are small and non-coding RNAs that display aberrant expression in the tissue and plasma of cancer patients when tested in comparison to healthy individuals. In past decades, research data proposed that miRNAs could be diagnostic and prognostic biomarkers in cancer patients. It has been confirmed that miRNAs can act either as oncogenes by silencing tumor inhibitors or as tumor suppressors by targeting oncoproteins. MiR-144s are located in the chromosomal region 17q11.2, which is subject to significant damage in many types of cancers. In this review, we assess the involvement of miR-144s in several cancer types by illustrating the possible target genes that are related to each cancer, and we also briefly describe the clinical applications of miR-144s as a diagnostic and prognostic tool in cancers.
Collapse
Affiliation(s)
- Omid Kooshkaki
- Student Research Committee, Birjand University of Medical Sciences, Birjand 9717853577, Iran;
- Department of Immunology, Birjand University of Medical Sciences, Birjand 9717853577, Iran
| | - Zohre Rezaei
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand 9717853577, Iran;
- Department of Biology, University of Sistan and Baluchestan, Zahedan 9816745845, Iran
| | - Meysam Rahmati
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz 5166/15731, Iran;
| | - Parviz Vahedi
- Department of Anatomical Sciences, Maragheh University of Medical Sciences, Maragheh 5165665931, Iran;
| | - Afshin Derakhshani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5165665811, Iran; (A.D.); (A.B.)
| | - Oronzo Brunetti
- Medical Oncology Unit—IRCCS Istituto Tumori “Giovanni Paolo II” of Bari, 70124 Bari, Italy;
| | - Amir Baghbanzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5165665811, Iran; (A.D.); (A.B.)
| | - Behzad Mansoori
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, 5230 Odense, Denmark;
| | - Nicola Silvestris
- Medical Oncology Unit—IRCCS Istituto Tumori “Giovanni Paolo II” of Bari, 70124 Bari, Italy;
- Department of Biomedical Sciences and Human Oncology DIMO—University of Bari, 70124 Bari, Italy
- Correspondence: (N.S.); (B.B.); Tel.: +39-0805555419 (N.S.); +98-413-3371440 (B.B.)
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5165665811, Iran; (A.D.); (A.B.)
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz 5166614766, Iran
- Correspondence: (N.S.); (B.B.); Tel.: +39-0805555419 (N.S.); +98-413-3371440 (B.B.)
| |
Collapse
|
21
|
Kang H, Tong C, Li C, Luo J. miR-497 plays a key role in Tanshinone IIA-attenuated proliferation in OCI-AML3 cells via the MAPK/ERK1/2 pathway. Cytotechnology 2020; 72:427-432. [PMID: 32162175 DOI: 10.1007/s10616-020-00389-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 02/29/2020] [Accepted: 03/06/2020] [Indexed: 12/11/2022] Open
Abstract
Acute myelod leukemia (AML), as a uncontrolled proliferation of cells, was arrested differentiation of progenitor cells. The present study aimed to explore Tanshinone IIA (TIIA) effects on OCI-AML3 and the involvement of the MAPK signaling pathway and miR-497 in TIIA-mediated effects. Cell growth percentage was detected using a cell counting kit. Expression of miR-497 was detected by qPCR. Phosphorylated ERK1/2, JNK and p38 were assessed using western blot. The growth percentage of OCI-AML3 decreased and the effected time increased with increasing TIIA concentration. The miR-497 was upregulated and the p-ERK1/2 was decreased when the TIIA added. TIIA cannot influence the p-ERK1/2. Hence, the proliferation of OCI-AML3 cells was raising. However, when the p-ERK1/2 was inhibited, there no influence on the miR-497 expression after TIIA added. TIIA upregulates miR-497, and decrease the p-ERK1/2 expression, when TIIA simulated OCI-AML3 cell in vitro. And in miR-497 might be involved in the regulation of proliferation in this process.
Collapse
Affiliation(s)
- Hongyang Kang
- Department of Hematology, The Second Hospital of Hebei Medical University, Shijiazhuang, China.,Department of Hematology, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Changqing Tong
- Department of Hematology, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Chaonan Li
- Department of Hematology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jianmin Luo
- Department of Hematology, The Second Hospital of Hebei Medical University, Shijiazhuang, China.
| |
Collapse
|
22
|
Mansoori B, Duijf PHG, Mohammadi A, Najafi S, Roshani E, Shanehbandi D, Hajiasgharzadeh K, Shirjang S, Ditzel HJ, Kazemi T, Mokhtarzadeh A, Gjerstorff MF, Baradaran B. Overexpression of HMGA2 in breast cancer promotes cell proliferation, migration, invasion and stemness. Expert Opin Ther Targets 2020; 24:255-265. [PMID: 32172636 DOI: 10.1080/14728222.2020.1736559] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Accepted: 02/18/2020] [Indexed: 01/07/2023]
Abstract
Despite improved therapeutic strategies for early-stage breast cancer, the most common cancer type in women, relapse remains common and the underlying mechanisms for this progression remain poorly understood. To gain more insight, we studied the DNA-binding protein HMGA2 in breast cancer development and stemness. We demonstrated that HMGA2 is overexpressed in breast cancer tissues at the mRNA and protein levels (P value <0.0001). HMGA2 knockdown and overexpression in breast cancer cells revealed that HMGA2 promotes cell proliferation and protects against apoptosis via the intrinsic pathway. HMGA2 knockdown also causes cell cycle arrest in G2/M phase. In addition, we found that HMGA2 increases breast cancer cell migration and invasion (P value <0.001) and promotes the acquisition of cancer stem cell features, both in vitro, in colony formation (P value <0.01) and spheroid assays, and in breast cancer tissues. Overexpression of HMGA2 in breast cancer spurs the acquisition of several hallmarks of cancer, including increased cell proliferation, migration, invasion and stemness, and decreased apoptosis. Thus, targeting HMGA2 could represent an effective strategy to block breast cancer progression.
Collapse
Affiliation(s)
- Behzad Mansoori
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Aging Research Institute, Physical Medicine and Rehabilitation Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Pascal H G Duijf
- University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, Australia
| | - Ali Mohammadi
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Souzan Najafi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elmira Roshani
- Department of Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Dariush Shanehbandi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Solmaz Shirjang
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Henrik J Ditzel
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Department of Oncology, Institute for Clinical Research, Odense University Hospital, Odense, Denmark
| | - Tohid Kazemi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Morten F Gjerstorff
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
23
|
Emam MA, Khattab HI, Hegazy MG. Assessment of anticancer activity of Pulicaria undulata on hepatocellular carcinoma HepG2 cell line. Tumour Biol 2019; 41:1010428319880080. [PMID: 31603389 DOI: 10.1177/1010428319880080] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Searching for new sources of safe nutraceuticals antitumor drugs is an important issue. Consequentially, this study designed to assess the antitumor activity of Pulicaria undulata extract in vitro in the treatment of hepatocellular carcinoma HepG2 cell line. Aerial parts of P. undulata plants were collected, used for phytochemical analysis, and assessed for anticancer activity. The antitumor activity was evaluated through studying the cell viability and apoptotic pathway. The gas chromatography-mass spectrometry phytochemical analysis revealed that P. undulata is a promising new source of several known antioxidant and antitumor compounds which could participate in drug development and exploration of alternative strategies to the harmful synthetic antitumor drugs. P. undulata stifled HepG2 cell viability in a concentration-dependent manner. Meanwhile, P. undulata tempted substantial apoptosis in HepG2 cells and enhanced the expression of miR-34a. However, the mRNA expression level of antiapoptotic B-cell lymphoma-2 was markedly decreased by P. undulata treatment. Moreover, P. undulata increased the protein expression of proapoptotic p53 and caspase 3/9 with reducing B-cell lymphoma-2 protein expression level. Thus, P. undulata induced apoptosis in the HepG2 cells by overexpression of miR-34a which regulates p53/B-cell lymphoma-2/caspases signaling pathway. These findings were well appreciated with morphological studies of cells treated with P. undulata. In conclusion, P. undulata could be a probable candidate agent for the initiation of cell apoptosis in HepG2 and thereby can serve as promising therapeutic agent for treatment of hepatocellular carcinoma which should attract further studies.
Collapse
Affiliation(s)
- Manal A Emam
- Biochemistry Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Hemmat I Khattab
- Botany Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Marwa Ga Hegazy
- Biochemistry Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| |
Collapse
|
24
|
Xu SJ, Zhang F, Wang LJ, Hao MH, Yang XJ, Li NN, Ji HL, Xu P. Flavonoids of Rosa roxburghii Tratt offers protection against radiation induced apoptosis and inflammation in mouse thymus. Apoptosis 2019; 23:470-483. [PMID: 29995207 DOI: 10.1007/s10495-018-1466-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
The present study evaluated the protective effect of the natural compound flavonoids of Rosa roxburghii Tratt (FRT) against γ-radiation-induced apoptosis and inflammation in mouse thymus cells in vivo and in vitro. Thymus cells and mice were exposed to 60Co γ-ray at a dose of 6 Gy. The radiation treatment induced significant cell apoptosis and inflammation. Radiation increased the expressions of cleaved caspase 3/8-10, AIF, and PARP-1, and FRT could mitigate their activation and inhibit subsequent apoptosis in the thymus both in vitro or in vivo. Irradiation increased the mRNA expression of ICAM-1/VCAM-1, IL-1α/IL-6 and TNF-α/NF-κB. Our results also indicated that FRT alleviated gene expression of some inflammatory factors such as ICAM-1/VCAM-1, TNF-α/NF-κB, but not IL-1α/IL-6. Irradiation increased the protein expression levels of ICAM-1/VCAM-1, IL-1α/IL-6 and TNF-α/NF-Κb, and our results also indicated that FRT alleviated protein level expression of certain inflammatory factors such as ICAM-1, IL-1α/IL-6, TNF-α/NF-κB, but not VCAM-1. Our results suggested that FRT enhanced radioprotection at least partially by regulating caspase 3/8-10, AIF, and PARP-1 to reduce apoptosis and by regulating ICAM-1, IL-1α/IL-6, TNF-α/NF-κB to reduce inflammation.
Collapse
Affiliation(s)
- Sai-Juan Xu
- Department of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Fan Zhang
- Department of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Li-Juan Wang
- Department of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Ming-Hua Hao
- Department of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Xian-Jun Yang
- International Joint Research Laboratory for Recombiant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Na-Na Li
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, 453003, Henan, China.
| | - Hong-Long Ji
- Department of Cellular and Molecular Biology, University of Texas Health Northeast, Tyler, TX, 75708, USA. .,Institute of Lung and Molecular Therapy, Xinxiang Medical University, Xinxiang, 453003, Henan, China.
| | - Ping Xu
- Department of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, Henan, China.
| |
Collapse
|
25
|
Mansoori B, Mohammadi A, Gjerstorff MF, Shirjang S, Asadzadeh Z, Khaze V, Holmskov U, Kazemi T, Duijf PHG, Baradaran B. miR-142-3p is a tumor suppressor that inhibits estrogen receptor expression in ER-positive breast cancer. J Cell Physiol 2019; 234:16043-16053. [PMID: 30741415 DOI: 10.1002/jcp.28263] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 01/16/2019] [Accepted: 01/22/2019] [Indexed: 01/24/2023]
Abstract
Estrogen receptors (ERs) are involved in the development of many types of malignant tumors, in particular, breast cancer. Among others, ERs affect cell growth, proliferation, and differentiation. The microRNA (miRNA) miR-142-3p has been shown to inhibit carcinogenesis by regulating various cellular processes, including cell cycle progression, cell migration, apoptosis, and invasion. It does so via targeting molecules involved in a range of signaling pathways. We surgically collected 20 ER-positive breast cancer samples, each with matched adjacent normal breast tissue, and measured the expression of miR-142-3p via quantitative real-time polymerase chain reaction (qRT-PCR). Bioinformatics methods, luciferase reporter assay, qRT-PCR, and western blot analysis were used to assess whether miR-142-3p could target ESR1, which encodes the estrogen receptor, in ER-positive breast cancer cells and patient samples. We also restored miRNA expression and performed cell viability, cytotoxicity, and colony formation assays. Western blot analysis and qRT-PCR were used to study the expression of apoptosis and stemness markers. We found that miR-142-3p is downregulated in ER-positive breast cancers. Restoration of miR-142-3p expression in ER-positive breast cancer cells reduced cell viability, induced apoptosis via the intrinsic pathway and decreased both colony formation and the expression of stem cell markers. Bioinformatic analysis predicted miR-142-3p could bind to 3'-untranslated region ESR1 messenger RNA (mRNA). Consistently, we demonstrated that miR-142-3p reduced luciferase activity in ER-positive breast cancer cells, and decreased ESR1 expression in both mRNA and protein levels. The results revealed miR-142-3p and ESR1 expression correlated negatively in ER-positive breast cancer samples. The results suggest miR-142-3p acts as a tumor suppressor via multiple mechanisms. Thus, restoration of miR-142-3p expression, for example, via miRNA replacement therapy, may represent an effective strategy for the treatment of ER-positive breast cancer patients.
Collapse
Affiliation(s)
- Behzad Mansoori
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Mohammadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Morten F Gjerstorff
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Solmaz Shirjang
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zahra Asadzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vahid Khaze
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Uffe Holmskov
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Tohid Kazemi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Pascal H G Duijf
- University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, Australia
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
26
|
Zhang S, Mo Q, Wang X. Oncological role of HMGA2 (Review). Int J Oncol 2019; 55:775-788. [PMID: 31432151 DOI: 10.3892/ijo.2019.4856] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 05/17/2019] [Indexed: 11/06/2022] Open
Abstract
The high mobility group A2 (HMGA2) protein is a non‑histone architectural transcription factor that modulates the transcription of several genes by binding to AT‑rich sequences in the minor groove of B‑form DNA and alters the chromatin structure. As a result, HMGA2 influences a variety of biological processes, including the cell cycle process, DNA damage repair process, apoptosis, senescence, epithelial‑mesenchymal transition and telomere restoration. In addition, the overexpression of HMGA2 is a feature of malignancy, and its elevated expression in human cancer predicts the efficacy of certain chemotherapeutic agents. Accumulating evidence has suggested that the detection of HMGA2 can be used as a routine procedure in clinical tumour analysis.
Collapse
Affiliation(s)
- Shizhen Zhang
- Department of Breast Surgery, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| | - Qiuping Mo
- Department of Surgical Oncology and Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Xiaochen Wang
- Department of Breast Surgery, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| |
Collapse
|
27
|
Lu H, Hao L, Yang H, Chen J, Liu J. miRNA-34a suppresses colon carcinoma proliferation and induces cell apoptosis by targeting SYT1. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2019; 12:2887-2897. [PMID: 31934125 PMCID: PMC6949727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Accepted: 04/23/2019] [Indexed: 06/10/2023]
Abstract
BACKGROUND MicroRNAs are emerging as the important regulators in cancer-related processes. This research were performed to find the function and mechanism of miR-34a effect on colon cancer. METHODS In this study, we examined the expression of miR-34a in colon cancer tissues and cell lines by qRT-PCR. In vitro cell functional assays studies were built to define miR-34a and SYT1 function involved in cell growth, migration, and invasion and apoptosis. EGFP reporter assay was used to determine the relationship of SYT1 and miR-181a. To confirmed the relationship between SYT1 and miR-34a, the SYT1 restoration rescued miR-34a mediated growth and inhibited cell apoptosis were detect. RESULT Our studies show that microRNA-34a (miR-34a) is downregulated in human colon cancer relative to normal colon mucosal epithelial cells, and downexpression of miR-34a promotes cell proliferation, migration, and invasion, nevertheless overexpression of miR-34 facilitates cell apoptosis in vitro. Furthermore, SYT1 3'-UTR is found to be down-regulated directly by miR-34a, demonstrating that SYT1 is a important target of miR-34a in colon cancer. The knockdown of SYT1 markedly inhibits colon cancer cell proliferation, migration, and invasion, and induces cell apoptosis, indicating that SYT1 may function as an oncogene in colon cancer. The restoration of SYT1 expression can counteract the effect of miR-34a on cell proliferation, and induces cell apoptosis, of colon cancer cells. CONCLUSION Together, these results indicate that miR-34a is a new regulator of SYT1, and both miR-34a and SYT1 play the important roles in the pathogenesis of colon cancer.
Collapse
Affiliation(s)
- Haichao Lu
- Department of Gastrointestinal Surgery, The First People's Hospital of Nanning Nanning, Guangxi Zhuang Autonomous Region, China
| | - Liang Hao
- Department of Gastrointestinal Surgery, The First People's Hospital of Nanning Nanning, Guangxi Zhuang Autonomous Region, China
| | - Hongfan Yang
- Department of Gastrointestinal Surgery, The First People's Hospital of Nanning Nanning, Guangxi Zhuang Autonomous Region, China
| | - Jianshe Chen
- Department of Gastrointestinal Surgery, The First People's Hospital of Nanning Nanning, Guangxi Zhuang Autonomous Region, China
| | - Jinxin Liu
- Department of Gastrointestinal Surgery, The First People's Hospital of Nanning Nanning, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
28
|
Shirmohamadi M, Eghbali E, Najjary S, Mokhtarzadeh A, Kojabad AB, Hajiasgharzadeh K, Lotfinezhad P, Baradaran B. Regulatory mechanisms of microRNAs in colorectal cancer and colorectal cancer stem cells. J Cell Physiol 2019; 235:776-789. [PMID: 31264216 DOI: 10.1002/jcp.29042] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 06/13/2019] [Indexed: 12/12/2022]
Abstract
Colorectal cancer (CRC) is one of the most lethal and hard-to-treat cancers in the world, which in its advanced stages, surgery and chemotherapy are the main common treatment approaches. The microRNAs (miRNAs), as novel markers for CRC detection, promote their regulatory effects via the 3'-untranslated binding region (3'-UTR) of target messenger RNA in posttranscriptional regulation of genes and also play a pivotal role in modulating resistance to chemotherapeutic agents. These small noncoding RNAs have also a critical role in CRC stem cells (CRCSCs) regulation, comprising self-renewal, differentiation, and tumorigenesis. Cancer stem cells (CSCs) are distinctive cell types inside a tumor tissue that are believed to derive from normal somatic stem cells. The CSCs have self-renewal abilities, angiogenesis, as well as specific surface markers expression characteristics. Furthermore, they are frequently criticized for tumor maintenance, treatment resistance, tumor development, and distant metastasis. In this review, we discuss the current understandings of CRCSCs and their environment with a focus on the role of miRNAs on the regulation of CSCs and their targeting application in CRC treatment.
Collapse
Affiliation(s)
- Masoud Shirmohamadi
- Liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elham Eghbali
- Medical Radiation Sciences Research Group, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shiva Najjary
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | - Parisa Lotfinezhad
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
29
|
Mansoori B, Mohammadi A, Naghizadeh S, Gjerstorff M, Shanehbandi D, Shirjang S, Najafi S, Holmskov U, Khaze V, Duijf PHG, Baradaran B. miR-330 suppresses EMT and induces apoptosis by downregulating HMGA2 in human colorectal cancer. J Cell Physiol 2019; 235:920-931. [PMID: 31241772 DOI: 10.1002/jcp.29007] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 05/31/2019] [Indexed: 12/24/2022]
Abstract
MicroRNAs (miRNAs) are important molecular regulatorsof cellular signaling and behavior. They alter gene expression by targeting messenger RNAs, including those encoding transcriptional regulators, such as HMGA2. While HMGA2 is oncogenic in various tumors, miRNAs may be oncogenic or tumor suppressive. Here, we investigate the expression of HMGA2 and the miRNA miR-330 in a patient with colorectal cancer (CRC) samples and their effects on oncogenic cellular phenotypes. We found that HMGA2 expression is increased and miR-330 expression is decreased in CRCs and each predicts poor long-term patient survival. Stably increased miR-330 expression in human colorectal cancer cells (HCT116) and SW480 CRC cell lines downregulate the oncogenic expression of HMGA2, a predicted miR-330 target. Additionally, this promotes apoptosis and decreases cell migration and viability. Consistently, it also decreases protein-level expression of markers for epithelial-to-mesenchymal-transition (Snail-1, E-cadherin, and Vascular endothelial growth factor receptors) and transforming growth factor β signaling (SMAD3), as well as phospho- Protein kinase B (AKT) and phospho-STAT3 levels. We conclude that miR-330 acts as a tumor suppressor miRNA in CRC by suppressing HMGA2 expression and reducing cell survival, proliferation, and migration. Thus, we identify miR-330 as a promising candidate for miRNA replacement therapy for patients with CRC.
Collapse
Affiliation(s)
- Behzad Mansoori
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.,Aging Research Institute, Physical Medicine and Rehabilitation Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Mohammadi
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Sanaz Naghizadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Morten Gjerstorff
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Dariush Shanehbandi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Solmaz Shirjang
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Souzan Najafi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Uffe Holmskov
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Vahid Khaze
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Pascal H G Duijf
- Translational Research Institute, University of Queensland Diamantina Institute, The University of Queensland, Brisbane, Australia
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
30
|
Wang Y, Wang C. microRNA-211-3p has a Role in the Effects of Lipopolysaccharide on Endoplasmic Reticulum Stress in Cultured Human Skin Fibroblasts. Med Sci Monit Basic Res 2019; 25:164-168. [PMID: 31221950 PMCID: PMC6607940 DOI: 10.12659/msmbr.915379] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Background Lipopolysaccharide (LPS) in bacterial infection of skin wounds delays wound healing. This study aimed to investigate the effects of LPS and endoplasmic reticulum stress in cultured skin fibroblasts and microRNA-211-3p (miR-211-3p) signaling. Material/Methods Human skin fibroblasts were cultured in increasing concentrations of LPS at 0 ng/ml, 5 ng/ml, 10 ng/ml, and 20 ng/ml for 0, 12 h, 24 h, 36 h, and 48 h. Cell proliferation was determined using the MTT assay. Protein expression levels of the transcription factors GRP78, CHOP, p-JNK, and the endoplasmic reticulum stress apoptosis proteins, caspase-12 and Bcl-2, were determined by Western blot. The expression of miR-211-3p in human skin fibroblasts was detected by quantitative polymerase chain reaction (qPCR). Results Cell proliferation of human skin fibroblasts decreased with increasing concentrations of LPS in a dose-dependent and time-dependent way. Protein levels of GRP78, CHOP, p-JNK, caspase-12, and Bcl-2 were increased 8 h and 12 h after LPS treatment compared with 0 h and 4 h after treatment. However, the expression of miR-211-3p was decreased in human skin fibroblasts after treatment with LPS. When miR-211-3p was overexpressed, the endoplasmic reticulum stress/CHOP related proteins, including GRP78, CHOP, p-JNK, caspase-12, and Bcl-2 were unchanged after the addition of LPS. Overexpression of miR-211-3p also reduced inhibitory effects of LPS on the growth of human skin fibroblasts. Conclusions This study showed that microRNA-211-3p had a role in the effects of LPS on endoplasmic reticulum stress and CHOP activation in cultured human skin fibroblasts.
Collapse
Affiliation(s)
- Yongxiang Wang
- Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China (mainland)
| | - Chunyan Wang
- Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China (mainland)
| |
Collapse
|
31
|
Mansoori B, Mohammadi A, Asadzadeh Z, Shirjang S, Minouei M, Abedi Gaballu F, Shajari N, Kazemi T, Gjerstorff MF, Duijf PHG, Baradaran B. HMGA2 and Bach-1 cooperate to promote breast cancer cell malignancy. J Cell Physiol 2019; 234:17714-17726. [PMID: 30825204 DOI: 10.1002/jcp.28397] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 01/25/2019] [Accepted: 01/28/2019] [Indexed: 12/12/2022]
Abstract
During breast cancer progression, tumor cells acquire multiple malignant features. The transcription factors and cell cycle regulators high mobility group A2 (HMGA2) and BTB and CNC homology 1 (Bach-1) are overexpressed in several cancers, but the mechanistic understanding of how HMGA2 and Bach-1 promote cancer development has been limited. We found that HMGA2 and Bach-1 are overexpressed in breast cancer tissues and their expression correlates positively in tumors but not in normal tissues. Individual HMGA2 or Bach-1 knockdown downregulates expression of both proteins, suggesting a mutual stabilizing effect between the two proteins. Importantly, combined HMGA2 and Bach-1 knockdown additively decrease cell proliferation, migration, epithelial-to-mesenchymal transition, and colony formation, while promoting apoptotic cell death via upregulation of caspase-3 and caspase-9. First the first time, we show that HMGA2 and Bach-1 overexpression in tumors correlate positively and that the proteins cooperatively suppress a broad range of malignant cellular properties, such as proliferation, migration, clonogenicity, and evasion of apoptotic cell death. Thus, our observations suggest that combined targeting of HMGA2 and Bach1 may be an effective therapeutic strategy to treat breast cancer.
Collapse
Affiliation(s)
- Behzad Mansoori
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Mohammadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zahra Asadzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Solmaz Shirjang
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahsa Minouei
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Neda Shajari
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Tohid Kazemi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Morten F Gjerstorff
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Pascal H G Duijf
- University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, Australia
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
32
|
Sadeghiyeh N, Sehati N, Mansoori B, Mohammadi A, Shanehbandi D, Khaze V, Baradaran B. MicroRNA-145 replacement effect on growth and migration inhibition in lung cancer cell line. Biomed Pharmacother 2018; 111:460-467. [PMID: 30594785 DOI: 10.1016/j.biopha.2018.12.094] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 12/14/2018] [Accepted: 12/19/2018] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Lung cancer is the main cause of cancer death in males and females worldwide. Reduced expression of miR-145 has been reported in many types of cancers. In this study, we transfected miR-145 into lung cancer cells by vector-based miR-145, and investigated the effects of this intervention on growth and migration inhibition of cancer cells as well on the expression of targeted genes. METHODS IC50 of Geneticin (G418) antibiotic was measured using MTT test in NSCLC cell lines. miR-145 was transfected into lung cancer cells by jetPEI. qRT-PCR was used to evaluate the transcript level of the miR-145 and expression for KRAS, MMP-9, vimentin, caspase-3, caspase-8 and caspase-9 genes in A549 cells. MTT assay was used to evaluate the proliferation inhibition of cancer cells. Wound healing assay was used to check the migration status of transfected lung cancer cells. The apoptosis induction was assessed by DAPI staining assay. RESULTS The MTT assay showed that the IC50 of Genticin was 494.1 μg/ml. The results of the qRT-PCR showed increased expression level of miR-145 and downregulation of KRAS, MMP-9, and vimentin expression in A549 transfected cells compared with the control group. The MTT assay results demonstrated inhibition of cancer cell proliferation after miR-145 replacement. Wound healing assay results revealed that migration was reduced upon miR-145 transfection. The transfected cell displayed increased apoptosis rate by inducing caspase-3 and caspase-9 mRNA expression. CONCLUSION The results of this study showed that increased miR-145 expression exerted a critical role in subsiding the growth, survival, and migration of lung cancer cell line.
Collapse
Affiliation(s)
- Navaz Sadeghiyeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nasser Sehati
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Mansoori
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Mohammadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Dariush Shanehbandi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vahid Khaze
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
33
|
Mohammadi A, Mansoori B, Savadi P, Khaze V, Minouei M, McMillan NAJ, Hallaj-Nezhadi S, Baradaran B. Targeting of high mobility group A2 by small interfering RNA-loaded nanoliposome-induced apoptosis and migration inhibition in gastrointestinal cancer cells. J Cell Biochem 2018; 120:9203-9212. [PMID: 30507008 DOI: 10.1002/jcb.28196] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 11/08/2018] [Indexed: 12/14/2022]
Abstract
BACKGROUND Considering the complex nature of gastrointestinal cancer, different methods including surgery, radiotherapy, and chemotherapy are considered for the treatment. Novel strategies including silencing of oncogenes using safe delivery systems could be considered as a novel approach in colorectal cancer treatment. The aim of this study was to investigate the silencing effect of high mobility group A2 (HMGA2) small interfering RNA (siRNA)-loaded nanoliposomes on gastrointestinal cancers. METHODS The siRNA-lipoplexes were prepared using dioleoyl trimethylammonium propane (DOTAP)/cholesterol (Chol)/1, 2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE) through the freeze-drying of a monophase solution method. The size, polydispersity index (PDI), and zeta-potential of nanoliposomes were determined using Zetasizer analyzer. The morphology of the nanoliposomes was determined by transmission electron microscopy (TEM). The agarose gel-retardation assay was carried out to confirm the loading of siRNAs into liposome. The silencing of the HMGA2 in cancer cells was evaluated by quantitative reverse-transcription polymerase chain reaction (qRT-PCR). The effect of liposomes on cell cytotoxicity was studied by MTT assay. The inhibitory effect of siRNA-loaded liposomes was evaluated by a wound-healing assay. The apoptosis induction was investigated via the annexin V/propidium iodide assay. RESULTS The size, PDI, and zeta-potential of the prepared liposomes were found to be 350 nm, 0.67, and 86.3 mV, respectively. They were spherical in shape and could efficiently associate with siRNA. The results of gene silencing showed that the optimum condition of HMGA2 silencing was 80 pmol HMGA2 and 24 hours after treatment in each cancer cell lines. MTT assays indicated that silencing of HMGA2 in optimal condition could reduce the viability of the cancer cells more than 60% in the three cell lines. The result of the apoptosis assay showed more than 50% of the cell deaths related to the apoptosis in all three cell lines. The gene expression evaluation confirmed that apoptosis was induced via the intrinsic pathway inducing both caspase-3 and -9 expressions. Also, the reduction in Bcl2 expression confirmed the activation apoptosis pathway in the treated cancer cells. The wound-healing assay showed the suppression of cancer cell migration after treatment with the prepared nanoliposomes. CONCLUSION The results of this study showed the HMGA2 siRNA-loaded nanoliposomes could be effective in the treatment of gastrointestinal cancers.
Collapse
Affiliation(s)
- Ali Mohammadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Mansoori
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Pouria Savadi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vahid Khaze
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahsa Minouei
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nigel A J McMillan
- School of Medical Sciences and Menzies Health Institute Queensland, Griffith University, Southport, Australia
| | | | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
34
|
Khordadmehr M, Shahbazi R, Ezzati H, Jigari-Asl F, Sadreddini S, Baradaran B. Key microRNAs in the biology of breast cancer; emerging evidence in the last decade. J Cell Physiol 2018; 234:8316-8326. [PMID: 30422324 DOI: 10.1002/jcp.27716] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 10/16/2018] [Indexed: 12/12/2022]
Abstract
microRNAs (miRNAs) are a family of small noncoding RNAs that play a pivotal role in the regulation of main biological and physiological processes, including cell cycle regulation, proliferation, differentiation, apoptosis, stem cell maintenance, and organ development. Dysregulation of these tiny molecules has been related to different human diseases, such as cancer. It has been estimated that more than 50% of these noncoding RNA sequences are placed on fragile sites or cancer-associated genomic regions. After the discovery of the first specific miRNA signatures in breast cancer, many studies focused on the involvement of these small RNAs in the pathophysiology of breast tumors and their possible clinical implications as reliable prognostic biomarkers or as a new therapeutic approach. Therefore, the present review will focus on the recent findings on the involvement of miRNAs in the biology of breast cancer associated with their clinical implications.
Collapse
Affiliation(s)
- Monireh Khordadmehr
- Department of Pathology, Faculty of Veterinary, Medicine, University of Tabriz, Tabriz, East Azerbaijan, Iran
| | - Roya Shahbazi
- Department of Pathology, Faculty of Veterinary, Medicine, University of Tabriz, Tabriz, East Azerbaijan, Iran
| | - Hamed Ezzati
- Department of Pathology, Faculty of Veterinary, Medicine, University of Tabriz, Tabriz, East Azerbaijan, Iran
| | - Farinaz Jigari-Asl
- Department of Pathology, Faculty of Veterinary, Medicine, University of Tabriz, Tabriz, East Azerbaijan, Iran
| | - Sanam Sadreddini
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
35
|
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
|
36
|
Wang Y, Liu Z, Shen J. MicroRNA-421-targeted PDCD4 regulates breast cancer cell proliferation. Int J Mol Med 2018; 43:267-275. [PMID: 30365117 PMCID: PMC6257841 DOI: 10.3892/ijmm.2018.3932] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 09/26/2018] [Indexed: 12/19/2022] Open
Abstract
MicroRNAs (miRNAs) are expressed aberrantly in various types of cancer, and negatively regulate the expression of target genes which may be useful in therapeutic strategies in several biological processes. In the present study, the expression levels and the effects of miRNA (miR)-421 in breast cancer tissues and MCF-7 and MDA-MB-231 cells were evaluated to elucidate therapeutic targets in breast cancer cells. The putative targets of miR-421 were predicted by bioinformatics approaches, and the expression levels of miR-421 were measured in MCF-7 and MDA-MB-231 cells by reverse transcription-quantitative polymerase chain reaction analysis following miR-421 knockdown. The rates of cell proliferation, migration capacity, invasiveness and apoptosis were determined in miR-421 inhibitor-transfected MCF-7 and MDA-MB-231 cells. The expression levels of target proteins regulated by miR-421 in MCF-7 and MDA-MB-231 cells were analyzed by western blot analysis. miR-421 was increased significantly in breast cancer tissues and cells, and was regulated by miR-421 antisense oligonucleotides. The knockdown of miR-421 in MCF-7 and MDA-MB-231 cells decreased cell proliferation, migration capacity and invasiveness, and promoted apoptosis compared with control groups. The expression of target protein programmed cell death 4 (PDCD4) were decreased in MCF-7 and MDA-MB-231 cells transfected with miR-421 inhibitors. These results suggested a correlation between miR-421 and PDCD4, and physiological functions of breast cancer cells, suggesting that miR-421 may be a potential strategy in the therapy of breast cancer.
Collapse
Affiliation(s)
- Yiwei Wang
- Tianjin First Center Hospital, Tianjin 300192, P.R. China
| | - Zipeng Liu
- Hanzhong Central Hospital, Hanzhong, Shaanxi 723000, P.R. China
| | - Jian Shen
- The Second Affiliated Hospital of Xi'an Medical University, Xi'an, Shaanxi 710038, P.R. China
| |
Collapse
|
37
|
Naghizadeh S, Mansoori B, Mohammadi A, Kafil HS, Mousavi Z, Sakhinia E, Baradaran B. Effects of HMGA2 gene downregulation by siRNA on lung carcinoma cell migration in A549 cell lines. J Cell Biochem 2018; 120:5024-5032. [PMID: 30317663 DOI: 10.1002/jcb.27778] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 09/06/2018] [Indexed: 01/08/2023]
Abstract
BACKGROUND Although there are multiple treatments for lung cancer, the death rate of this cancer remains high because of metastasis in earlier stages. So a novel treatment for overcoming metastasis is urgently needed. Overexpression of high-mobility group AT-hook 2 (HMGA2), a nonhistone chromosomal protein has been observed in metastatic cancers. So, we suggested that HMGA2 upregulation may play a critical role in treating lung cancer. METHODS The A549 cells were transfected with specific HMGA2 small interfering RNA (siRNA) using transfection reagent. Relative HMGA2 and matrix metallopeptidase 1 (MMP1), C-X-C chemokine receptor type 4 (CXCR4), vimentin, and E-cadherin messenger RNA expression levels were measured by quantitative real-time polymerase chain reaction. To diagnose cytotoxic effect of HMGA2 siRNA and other components of transfection process, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay was applied. The migration capacity after transfection with HMGA2 siRNA was detected by wound-healing assay. RESULTS HMGA2 siRNA significantly reduced HMGA2 expression in a dose-dependent manner 48 hours after transfection. Expression levels of MMP1, vimentin, and CXCR4 were reduced, but E-cadherin level was not changed meaningfully. HMGA2 knockdown significantly reduced cell survival rate and also led to the inhibition of cell migration. CONCLUSIONS Our results indicated that RNA interference by downregulation of HMGA2 gene expression and affecting downstream genes led to the inhibition of cell migration and proliferation. Therefore, HMGA2 siRNA might be an alternative treatment option for metastatic lung cancer.
Collapse
Affiliation(s)
- Sanaz Naghizadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Mansoori
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Mohammadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Samadi Kafil
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.,Drug Applied Research Center, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zohreh Mousavi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ebrahim Sakhinia
- Drug Applied Research Center, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
38
|
Ghasabi M, Mansoori B, Mohammadi A, Duijf PH, Shomali N, Shirafkan N, Mokhtarzadeh A, Baradaran B. MicroRNAs in cancer drug resistance: Basic evidence and clinical applications. J Cell Physiol 2018; 234:2152-2168. [PMID: 30146724 DOI: 10.1002/jcp.26810] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 04/30/2018] [Indexed: 12/19/2022]
Abstract
Development of drug resistance has considerably limited the efficacy of cancer treatments, including chemotherapy and targeted therapies. Hence, understanding the molecular mechanisms underpinning the innate or the acquired resistance to these therapies is critical to improve drug efficiency and clinical outcomes. Several studies have implicated microRNAs (miRNA) in this process. MiRNAs repress gene expression by specific binding to complementary sequences in the 3' region of target messenger RNAs (mRNAs), followed by target mRNA degradation or blocked translation. By targeting molecules specific to a particular pathway within tumor cells, the new generation of cancer treatment strategies has shown significant advantages over conventional chemotherapy. However, the long-term efficacy of targeted therapies often remains poor, because tumor cells develop resistance to such therapeutics. Targeted therapies often involve monoclonal antibodies (mAbs), such as those blocking the ErB/HER tyrosine kinases, epidermal growth factor receptor (cetuximab) and HER2 (trastuzumab), and those inhibiting vascular endothelial growth factor receptor signaling (e.g., bevacizumab). Even though these are among the most used agents in tumor medicine, clinical response to these drugs is reduced due to the emergence of drug resistance as a result of toxic effects in the tumor microenvironment. Research on different types of human cancers has revealed that aberrant expression of miRNAs promotes resistance to the aforementioned drugs. In this study, we review the mechanisms of tumor cell resistance to mAb therapies and the role of miRNAs therein. Emerging treatment strategies combine therapies using innovative miRNA mimics or antagonizers with conventional approaches to maximize outcomes of patients with cancer.
Collapse
Affiliation(s)
- Mehri Ghasabi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Mansoori
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Mohammadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Pascal Hg Duijf
- University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, Australia
| | - Navid Shomali
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Naghmeh Shirafkan
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
39
|
Zaleski M, Kobilay M, Schroeder L, Debald M, Semaan A, Hettwer K, Uhlig S, Kuhn W, Hartmann G, Holdenrieder S. Improved sensitivity for detection of breast cancer by combination of miR-34a and tumor markers CA 15-3 or CEA. Oncotarget 2018; 9:22523-22536. [PMID: 29854296 PMCID: PMC5976482 DOI: 10.18632/oncotarget.25077] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 03/02/2018] [Indexed: 12/13/2022] Open
Abstract
Background MicroRNAs biomarkers have shown value for diagnosis and prognosis of various cancers. Combination with established tumor markers has rarely been done. Results Breast cancer patients had significantly higher serum RNA loads (AUC 0.665), lower miR-34a (AUC 0.772), higher CEA and CA 15-3 levels (AUCs 0.717 and 0.721) than healthy controls. miR-34a correlated with tumor stage and hormone receptor status. There was no significant difference between groups for all other miRNAs. Combination of miR-34a with CEA or CA 15-3 led to improved AUCs of 0.844 and 0.800, respectively. Sensitivity of miR-34a and CA 15-3 reached 56.1% at 95% specificity. When compared with benign breast diseases, combination of miR-34a (AUC 0.719) and CEA (0.623) or CA 15-3 (0.619) resulted in improved performances (0.794 and 0.741). Sensitivity of miR-34a and CA 15-3 reached 53.7% at 95% specificity. Conclusion While miR-34a provides valuable information for diagnosis and staging, combination with tumor markers CA15-3 or CEA improves the sensitivity for breast cancer detection. Patients and Methods The diagnostic relevance of the miR-21, miR-34a, miR-92a, miR-155, miR-222 and miR-let-7c was tested in sera of 103 individuals (55 breast cancer, 20 benign breast diseases, 28 healthy controls). MiRNAs were detected by quantitative rt-PCR after extraction and reverse transcription. Cel-miR-39 and miR-16 were used for normalization. Established tumor markers CEA, CA 15-3, CA 19-9 and CA 125 were measured by automatized immunoassays. Diagnostic performance was tested by areas under the curve (AUC) of receiver operating characteristic (ROC) curves and sensitivities at 90% and 95% specificity.
Collapse
Affiliation(s)
- Martin Zaleski
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Makbule Kobilay
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Lars Schroeder
- Department of Gynecology and Obstetrics, University Hospital Bonn, Bonn, Germany.,Center for Integrated Oncology (CIO) Köln/Bonn, Bonn, Germany
| | - Manuel Debald
- Department of Gynecology and Obstetrics, University Hospital Bonn, Bonn, Germany.,Center for Integrated Oncology (CIO) Köln/Bonn, Bonn, Germany
| | | | - Karina Hettwer
- QuoData Statistics, Dresden, Germany.,Joint Research and Services Center for Biomarker Evaluation in Oncology, Bonn/Dresden, Germany
| | - Steffen Uhlig
- QuoData Statistics, Dresden, Germany.,Joint Research and Services Center for Biomarker Evaluation in Oncology, Bonn/Dresden, Germany
| | - Walther Kuhn
- Department of Gynecology and Obstetrics, University Hospital Bonn, Bonn, Germany.,Center for Integrated Oncology (CIO) Köln/Bonn, Bonn, Germany
| | - Gunther Hartmann
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany.,Center for Integrated Oncology (CIO) Köln/Bonn, Bonn, Germany
| | - Stefan Holdenrieder
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany.,Center for Integrated Oncology (CIO) Köln/Bonn, Bonn, Germany.,Joint Research and Services Center for Biomarker Evaluation in Oncology, Bonn/Dresden, Germany
| |
Collapse
|
40
|
Giancotti V, Bergamin N, Cataldi P, Rizzi C. Epigenetic Contribution of High-Mobility Group A Proteins to Stem Cell Properties. Int J Cell Biol 2018; 2018:3698078. [PMID: 29853899 PMCID: PMC5941823 DOI: 10.1155/2018/3698078] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Revised: 03/01/2018] [Accepted: 03/18/2018] [Indexed: 02/07/2023] Open
Abstract
High-mobility group A (HMGA) proteins have been examined to understand their participation as structural epigenetic chromatin factors that confer stem-like properties to embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs), and cancer stem cells (CSCs). The function of HMGA was evaluated in conjunction with that of other epigenetic factors such as histones and microRNAs (miRs), taking into consideration the posttranscriptional modifications (PTMs) of histones (acetylation and methylation) and DNA methylation. HMGA proteins were coordinated or associated with histone and DNA modification and the expression of the factors related to pluripotency. CSCs showed remarkable differences compared with ESCs and iPSCs.
Collapse
Affiliation(s)
- Vincenzo Giancotti
- Department of Life Science, University of Trieste, Trieste, Italy
- Trieste Proteine Ricerche, Palmanova, Udine, Italy
| | - Natascha Bergamin
- Division of Pathology, Azienda Ospedaliero-Universitaria, Udine, Italy
| | - Palmina Cataldi
- Division of Pathology, Azienda Ospedaliero-Universitaria, Udine, Italy
| | - Claudio Rizzi
- Division of Pathology, Azienda Ospedaliero-Universitaria, Udine, Italy
| |
Collapse
|
41
|
Abstract
BACKGROUND Breast cancer has a high prevalence among women worldwide. Tumor invasion and metastasis still remains an open issue that causes most of the therapeutic failures and remains the prime cause of patient mortality. Hence, there is an unmet need to develop the most effective therapeutic approach with the lowest side effects and highest cytotoxicity that will effectively arrest or eradicate metastasis. METHODS An MTT assay and scratch test were used to assess the cytotoxicity and migration effects of Urtica dioica on the breast cancer cells. The QRT-PCR was used to study the expression levels of miR-21, MMP1, MMP9, MMP13, CXCR4, vimentin, and E-cadherin. RESULTS The results of gene expression in tumoral groups confirmed the overexpression of miR-21, MMP1, MMP9, MMP13, vimentin, and CXCR4, and the lower expression of E-cadherin compared to control groups (P<0.05). Moreover, the results of the MTT assay show that Urtica dioica significantly inhibited breast cancer cell proliferation. Moreover, findings from the scratch assay exhibited the inhibitory effects of Urtica dioica on the migration of breast cancer cell lines. CONCLUSION Urtica dioica extract could inhibit cancer cell migration by regulating miR-21, MMP1, MMP9, MMP13, vimentin, CXCR4, and E-Cadherin. Moreover, our findings demonstrated that the extract could decrease miR-21 expression, which substantially lessens the overexpressed MMP1, MMP9, MMP13, vimentin, and CXCR4 and increases E-cadherin in the tumoral group.
Collapse
|
42
|
Ma J, Li D, Kong FF, Yang D, Yang H, Ma XX. miR-302a-5p/367-3p-HMGA2 axis regulates malignant processes during endometrial cancer development. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:19. [PMID: 29391048 PMCID: PMC5796297 DOI: 10.1186/s13046-018-0686-6] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 01/23/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND Metastasis is one of the main reasons for treatment failure in endometrial cancer. Notably, high mobility group AT-hook 2 (HMGA2) has been recognized as a driving factor of tumour metastasis. microRNAs (miRNAs) are powerful posttranscriptional regulators of HMGA2. METHODS The binding sites of miR-302a-5p and miR-367-3p on HMGA2 mRNA were identified using bioinformatics prediction software and were validated via luciferase assay. The expression levels of miR-302a-5p and miR-367-3p were detected using quantitative real-time PCR and in situ hybridization. Western blotting and immunohistochemistry were used to detect the levels of HMGA2 and epithelial-mesenchymal transition pathway-related proteins. Co-immunoprecipitation was used to detect protein interactions. The roles of miR-302a-5p and miR-367-3p in the regulation of HMGA2 during the progression of endometrial cancer were investigated using both in vitro and in vivo assays. RESULTS In the present study, high HMGA2 expression was correlated with poor clinical outcomes in endometrial cancer. The binding sites of miRNAs on HMGA2 mRNA were identified using bioinformatics prediction software and were validated via luciferase assay. In the endometrial cancer cell lines Ishikawa and HEC-1A, the overexpression of miR-302a-5p/367-3p significantly inhibited the expression of HMGA2 mRNA. In endometrial cancer tissues, we showed that miR-302a-5p and miR-367-3p were significantly downregulated and thus inversely correlated with HMGA2. The miR-302a-5p and miR-367-3p expression levels were closely correlated with FIGO stage and lymph node metastasis. High expression of miR-302a-5p/367-3p was correlated with high survival rates in endometrial cancer. In addition, miR-302a-5p/367-3p suppressed the malignant behaviour of endometrial carcinoma cells via the inhibition of HMGA2 expression. CONCLUSION Our findings indicate that miR-302a-5p/367-3p-mediated expression of HMGA2 regulates the malignant behaviour of endometrial carcinoma cells, which suggests that the miR-302a-5p/367-3p-HMGA2 axis may be a predictive biomarker of endometrial cancer metastasis and patient survival and a potential therapeutic target in metastatic endometrial cancer.
Collapse
Affiliation(s)
- Jian Ma
- Department of Obstetrics and Gynecology, Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Da Li
- Department of Obstetrics and Gynecology, Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Fan-Fei Kong
- Department of Obstetrics and Gynecology, Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Di Yang
- Department of Obstetrics and Gynecology, Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Hui Yang
- Department of Obstetrics and Gynecology, Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Xiao-Xin Ma
- Department of Obstetrics and Gynecology, Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
| |
Collapse
|
43
|
Galdiero F, Bello AM, Spina A, Capiluongo A, Liuu S, De Marco M, Rosati A, Capunzo M, Napolitano M, Vuttariello E, Monaco M, Califano D, Turco MC, Chiappetta G, Vinh J, Chiappetta G. Identification of BAG3 target proteins in anaplastic thyroid cancer cells by proteomic analysis. Oncotarget 2018; 9:8016-8026. [PMID: 29487711 PMCID: PMC5814278 DOI: 10.18632/oncotarget.23858] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 10/30/2017] [Indexed: 11/25/2022] Open
Abstract
BAG3 protein is an apoptosis inhibitor and is highly expressed in Anaplastic Thyroid Cancer. We investigated the entire set of proteins modulated by BAG3 silencing in the human anaplastic thyroid 8505C cancer cells by using the Stable-Isotope Labeling by Amino acids in Cell culture strategy combined with mass spectrometry analysis. By this approach we identified 37 up-regulated and 54 down-regulated proteins in BAG3-silenced cells. Many of these proteins are reportedly involved in tumor progression, invasiveness and resistance to therapies. We focused our attention on an oncogenic protein, CAV1, and a tumor suppressor protein, SERPINB2, that had not previously been reported to be modulated by BAG3. Their expression levels in BAG3-silenced cells were confirmed by qRT-PCR and western blot analyses, disclosing two novel targets of BAG3 pro-tumor activity. We also examined the dataset of proteins obtained by the quantitative proteomics analysis using two tools, Downstream Effect Analysis and Upstream Regulator Analysis of the Ingenuity Pathways Analysis software. Our analyses confirm the association of the proteome profile observed in BAG3-silenced cells with an increase in cell survival and a decrease in cell proliferation and invasion, and highlight the possible involvement of four tumor suppressor miRNAs and TP53/63 proteins in BAG3 activity.
Collapse
Affiliation(s)
- Francesca Galdiero
- Functional Genomic Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Napoli, Italia
| | - Anna Maria Bello
- Functional Genomic Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Napoli, Italia
| | - Anna Spina
- Functional Genomic Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Napoli, Italia
| | - Anna Capiluongo
- Functional Genomic Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Napoli, Italia
| | - Sophie Liuu
- ESPCI ParisTech, Spectrométrie de Masse Biologique et Protéomique (SMBP), USR3149 CNRS, Paris, France
| | | | - Alessandra Rosati
- Biouniversa s.r.l., University of Salerno, Fisciano, Italy.,Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Baronissi (SA), Italy
| | - Mario Capunzo
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Baronissi (SA), Italy
| | - Maria Napolitano
- Functional Genomic Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Napoli, Italia
| | - Emilia Vuttariello
- Functional Genomic Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Napoli, Italia
| | - Mario Monaco
- Functional Genomic Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Napoli, Italia
| | - Daniela Califano
- Functional Genomic Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Napoli, Italia
| | - Maria Caterina Turco
- Biouniversa s.r.l., University of Salerno, Fisciano, Italy.,Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Baronissi (SA), Italy.,"SS. Giovanni di Dio e Ruggi d'Aragona-Schola Medica Salernitana", University of Salerno Hospital, Salerno, Italy
| | - Gennaro Chiappetta
- Functional Genomic Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Napoli, Italia
| | - Joëlle Vinh
- ESPCI ParisTech, Spectrométrie de Masse Biologique et Protéomique (SMBP), USR3149 CNRS, Paris, France
| | - Giovanni Chiappetta
- ESPCI ParisTech, Spectrométrie de Masse Biologique et Protéomique (SMBP), USR3149 CNRS, Paris, France
| |
Collapse
|
44
|
An analysis of suppressing migratory effect on human urinary bladder cancer cell line by silencing of snail-1. Biomed Pharmacother 2017; 96:545-550. [DOI: 10.1016/j.biopha.2017.10.044] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Revised: 10/05/2017] [Accepted: 10/09/2017] [Indexed: 11/24/2022] Open
|
45
|
Jafarlou M, Shanehbandi D, Dehghan P, Mansoori B, Othman F, Baradaran B. Enhancement of chemosensitivity by simultaneously silencing of Mcl-1 and Survivin genes using small interfering RNA in human myelomonocytic leukaemia. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2017; 46:1792-1798. [PMID: 29113504 DOI: 10.1080/21691401.2017.1392969] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Acute myeloid leukaemia (AML) is a genetically heterogeneous, severe and rapidly progressing disease triggered by blocking granulocyte or monocyte differentiation and maturation. Overexpression of myeloid cell leukaemia-1 (Mcl-1) and Survivin is associated with drug resistance, tumour progression and inhibition of apoptotic mechanisms in leukaemia and several cancers. In the present study, we examined the combined effect of etoposide and dual siRNA-mediated silencing of Mcl-1 and Survivin on U-937 AML cells. The AML cells were co-transfected with Mcl-1 and Survivin-specific siRNAs and genes silencing were confirmed by quantitative real-time PCR and Western blotting. Subsequently, MTT assay was used for the evaluation of cytotoxic effects by dual siRNA and etoposide on their own and in combination. For the studying of apoptosis, DNA-histone ELISA and annexin-V/FITC assays were performed. Co-transfection of Mcl-1 and Survivin siRNA significantly blocked their expression at the mRNA and protein levels, leading to the induction of apoptosis and strong inhibition of growth (p < .05). Besides, combined treatment of etoposide with Mcl-1 and Survivin siRNAs co-transfection leads to synergistically enhance etoposide-induced cytotoxic and apoptotic effects (p < .05). The results showed that Mcl-1 and Survivin play a major role in the U937 cells survival and their resistance relative to etoposide. Thus, Mcl-1 and Survivin can be considered as promising molecular targets for the treatment of AML. The combination treatment with etoposide, and siRNA-mediated silencing of corresponding genes may be a novel strategy in chemoresistance AML treatment.
Collapse
Affiliation(s)
- Mahdi Jafarlou
- a Immunology Research Center , Tabriz University of Medical Sciences , Tabriz , Iran.,b Department of Human Anatomy, Faculty of Medicine and Health Sciences, UPM , Selangor , Malaysia
| | - Dariush Shanehbandi
- a Immunology Research Center , Tabriz University of Medical Sciences , Tabriz , Iran
| | - Parvin Dehghan
- c Nutrition Research Center, Tabriz University of Medical Sciences , Tabriz , Iran
| | - Behzad Mansoori
- a Immunology Research Center , Tabriz University of Medical Sciences , Tabriz , Iran
| | - F Othman
- b Department of Human Anatomy, Faculty of Medicine and Health Sciences, UPM , Selangor , Malaysia
| | - Behzad Baradaran
- a Immunology Research Center , Tabriz University of Medical Sciences , Tabriz , Iran
| |
Collapse
|
46
|
Shajari N, Davudian S, Kazemi T, Mansoori B, Salehi S, Khaze Shahgoli V, Shanehbandi D, Mohammadi A, Duijf PHG, Baradaran B. Silencing of BACH1 inhibits invasion and migration of prostate cancer cells by altering metastasis-related gene expression. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2017; 46:1495-1504. [PMID: 28889753 DOI: 10.1080/21691401.2017.1374284] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Cancer lethality is mainly caused by metastasis. Therefore, understanding the nature of the genes involved in this process has become a priority. BACH1, a basic leucine zipper transcription factor, has been shown to transcriptionally regulate expression of a range of genes that are associated with breast cancer metastasis. However, the exact role and the underlying molecular mechanism of BACH1 in prostate cancer remain unclear. This study aims to explore the expression of BACH1 in prostate cancer tissues and the effect of BACH1 suppression on prostate cancer cell behavior. MATERIALS AND METHODS In this study, we used quantitative real-time PCR (qRT-PCR) to measure BACH1 expression in prostate adenocarcinoma tissues and two metastasis-derived prostate cancer cell lines, DU145 and LNCaP. We also used immunohistochemical (IHC) staining to measure BACH1 protein expression in prostate adenocarcinoma and matched normal tissue samples. In the following BACH1 expression was silenced in DU145 cells using siRNA as well. Knockdown was confirmed by qRT-PCR and Western blotting. The cytotoxic effects of BACH1-siRNA on DU145 cells were determined using an MTT assay. The migration and invasive capacity of DU145 cells were examined by scratch wound healing assay and matrigel invasion assay, respectively. We also used qRT-PCR to study the effect of BACH1 silencing on the expression levels of metastasis-related genes. RESULTS We find that the expression of BACH1 mRNA and protein in prostate cancer tissues is significantly higher than in matched normal prostate tissues (p < .05). In addition, DU145 and LNCaP cells exhibited 4.25-fold and 3.45-fold higher levels of BACH1 compared to HFF cell line. BACH1-siRNA significantly reduced both mRNA and protein expression levels in DU145 cells. More importantly, we show that BACH1 promotes key features of metastasis, as BACH1-siRNA treatment significantly reduced cell invasion and migration by changing the expression levels of a number of metastasis-related genes in vitro. CONCLUSIONS BACH1 is overexpressed in prostate cancer. Because this promotes invasion and migration, it may facilitate metastasis of prostate cancer. Thus, BACH1 is a potential therapeutic target for metastatic prostate cancer. BACH1 silencing therapy can be considered as a novel and effective adjuvant in prostate cancer targeted therapies.
Collapse
Affiliation(s)
- Neda Shajari
- a Immunology Research Center , Tabriz University of Medical Sciences , Tabriz , Iran.,b Student Research Committee , Tabriz University of Medical Sciences , Tabriz , Iran
| | - Sadaf Davudian
- a Immunology Research Center , Tabriz University of Medical Sciences , Tabriz , Iran.,b Student Research Committee , Tabriz University of Medical Sciences , Tabriz , Iran
| | - Tohid Kazemi
- a Immunology Research Center , Tabriz University of Medical Sciences , Tabriz , Iran
| | - Behzad Mansoori
- a Immunology Research Center , Tabriz University of Medical Sciences , Tabriz , Iran.,b Student Research Committee , Tabriz University of Medical Sciences , Tabriz , Iran
| | - Shima Salehi
- a Immunology Research Center , Tabriz University of Medical Sciences , Tabriz , Iran.,b Student Research Committee , Tabriz University of Medical Sciences , Tabriz , Iran
| | - Vahid Khaze Shahgoli
- a Immunology Research Center , Tabriz University of Medical Sciences , Tabriz , Iran
| | - Dariush Shanehbandi
- a Immunology Research Center , Tabriz University of Medical Sciences , Tabriz , Iran
| | - Ali Mohammadi
- a Immunology Research Center , Tabriz University of Medical Sciences , Tabriz , Iran
| | - Pascal H G Duijf
- c University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute , Brisbane , Australia
| | - Behzad Baradaran
- a Immunology Research Center , Tabriz University of Medical Sciences , Tabriz , Iran.,c University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute , Brisbane , Australia
| |
Collapse
|
47
|
Gao X, Dai M, Li Q, Wang Z, Lu Y, Song Z. HMGA2 regulates lung cancer proliferation and metastasis. Thorac Cancer 2017; 8:501-510. [PMID: 28752530 PMCID: PMC5582513 DOI: 10.1111/1759-7714.12476] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 06/13/2017] [Accepted: 06/13/2017] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND This study aimed to explore the effects of HMGA2 on cell proliferation and metastases in lung cancer and its underlying mechanism. METHODS HMGA2 expression in lung cancer tissues and its association with overall survival were analyzed based on data from a public database. The roles of HMGA2 were validated via loss-of-function and gain-of-function experiments in vitro. HMGA2 regulation by microRNA-195 (miR-195) was validated by real time-PCR, Western blotting, and luciferase reporter assays. RESULTS HMGA2 was upregulated and associated with reduced overall survival in patients with lung adenocarcinoma. HMGA2 knockdown suppressed the proliferation and motility of H1299 cells, while HMGA2 ectopic expression in A549 cells increased cell proliferation and migration. HMGA2 affected cell apoptosis through caspase 3/9 and Bcl-2, and regulated epithelial-to-mesenchymal transition by targeting Twist 1. Moreover, miR-195 was found to directly target the 3' untranslated region of HMGA2 messenger RNA and suppress its expression in lung cancer. CONCLUSION This study demonstrated that HMGA2, regulated by miR-195, played important roles in proliferation, metastases, and epithelial-to-mesenchymal transition in lung cancer. HMGA2 might serve as a biomarker and potential therapeutic target for lung cancer treatment.
Collapse
Affiliation(s)
- Xiaotian Gao
- Department of Cardiothoracic Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China.,Department of Cardiac Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ming Dai
- Department of Cardiothoracic Surgery, Central People's Hospital of Zhanjiang, Zhanjiang, China
| | - Qinglan Li
- Department of Respiratory Medicine, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Zhigang Wang
- Department of Cardiothoracic Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Yonglin Lu
- Department of Respiratory Medicine, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Zeqing Song
- Department of Respiratory Medicine, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
48
|
Karimi L, Mansoori B, shanebandi D, Mohammadi A, Aghapour M, Baradaran B. Function of microRNA-143 in different signal pathways in cancer: New insights into cancer therapy. Biomed Pharmacother 2017; 91:121-131. [DOI: 10.1016/j.biopha.2017.04.060] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 04/05/2017] [Accepted: 04/13/2017] [Indexed: 01/05/2023] Open
|
49
|
Tavanafar F, Safaralizadeh R, Hosseinpour-Feizi MA, Mansoori B, Shanehbandi D, Mohammadi A, Baradaran B. Restoration of miR-143 expression could inhibit migration and growth of MDA-MB-468 cells through down-regulating the expression of invasion-related factors. Biomed Pharmacother 2017; 91:920-924. [DOI: 10.1016/j.biopha.2017.04.119] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 04/23/2017] [Accepted: 04/27/2017] [Indexed: 01/16/2023] Open
|
50
|
Sharula, Wu Z. Regulation of Apoptosis by SYB in HepG2 Liver Cancer Cells is Mediated by the P53/Caspase 9 Axis. Anticancer Agents Med Chem 2017; 17:941-947. [PMID: 28356025 PMCID: PMC5543571 DOI: 10.2174/1871520617666170327161433] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 03/06/2017] [Accepted: 03/18/2017] [Indexed: 12/20/2022]
Abstract
Objective: To explore the function of miR-34a in promotion of apoptosis by SYB. Methods: In this study, the most effective concentration of SYB was determined by measuring cell proliferation. Relative miR-34a mRNA levels were detected by quantitative RT-PCR. Apoptosis was assessed using Annexin-V/PI assays, whereas protein levels of p53, caspase 3, caspase 9, caspase 8 and Bcl2 were evaluated by western blotting. Results: Minimum HepG2 cell growth was observed after 36h of exposure to 150 nmol/L SYB. miR-34a expression was highest 40min after the addition of SYB. SYB slightly decreased the abundance of Bcl-2, but increased the abundance of p53, caspase 3, caspase 9 and caspase 8. SYB failed to alter miR-34a expression when p53 was inhibited. Bcl-2 abundance remained low over time, whereas the abundance of caspase 3, caspase 9 and caspase 8 gradually increased. Inhibition of p53 promoted HepG2 cell growth in comparison with that of the control group. miR-34a was silenced to assess the role of miR-34a in the inhibitory effect of SYB on HepG2 cell growth. When p53 was silenced, protein abundance of Bcl2, caspase 3, caspase 8 and caspase 9 remained unchanged following the addition of SYB; moreover, HepG2 cell growth was increased. Conlusion: SYB represents a promising therapeutic approach for liver cancer patients.
Collapse
Affiliation(s)
- Sharula
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016China
| | - Zhongjun Wu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016China
| |
Collapse
|