151
|
Mazzoccoli L, Robaina MC, Apa AG, Bonamino M, Pinto LW, Queiroga E, Bacchi CE, Klumb CE. MiR-29 silencing modulates the expression of target genes related to proliferation, apoptosis and methylation in Burkitt lymphoma cells. J Cancer Res Clin Oncol 2018; 144:483-497. [PMID: 29318382 DOI: 10.1007/s00432-017-2575-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 12/28/2017] [Indexed: 12/22/2022]
Abstract
PURPOSE Burkitt lymphoma (BL) is a B-cell lymphoma frequently diagnosed in children. It is characterized by MYC translocations, which lead to the constitutive expression of the MYC oncogene. MYC contributes to miR-29 repression through an E-box MYC binding site on the miR-29b-1/miR-29a promoter region. We evaluated the role of miR-29a/b/c and their predicted targets in BL pathogenesis. METHODS Mature sequences of miR-29a/b/c were transfected to the BL cell lines BL41 and Raji, and evaluated for DNMT3B, MCL1, BIM, CDK6, AKT and TCL1 protein expression as well as for MCL-1 and CDK6 mRNA expression. BL cells were treated with 5-aza-2'-deoxycytidine (decitabine) and evaluated for miR29 expressions and methylation status. DNMT3B inhibition was performed by DNMT3B siRNA. RESULTS Ectopic expression of miR-29s in BL cells decreased CDK6, DNMT3B, TCL1 and MCL-1 protein levels, but CDK6 and MCL-1 mRNA expression was unaffected by miR-29. Decitabine enhanced miR-29 expression levels and decreased CDK6 protein expression. Additionally, inhibition of DNMT3B by siRNA increased miR-29a/b expression. Notably, the miR-29a/b1 and miR-29b2/c promoter genes showed methylated CpG sequences that were demethylated after decitabine treatments. Furthermore, MYC-negative tumours had higher levels of miR-29 expression compared with MYC-translocated cases, suggesting that MYC regulates miR-29 in BL tumours. CONCLUSIONS Our results suggest a significant role for miR-29s in BL pathogenesis in altering the expression of targets involved in critical cancer pathways, such as cell cycle control, apoptosis inhibition and DNA methylation. Moreover, methylation-mediated miR-29 epigenetic silencing may occur during BL development.
Collapse
Affiliation(s)
- Luciano Mazzoccoli
- Programa de Pesquisa em Hemato-Oncologia Molecular, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
| | - Marcela Cristina Robaina
- Programa de Pesquisa em Hemato-Oncologia Molecular, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
| | | | - Martin Bonamino
- Programa de Carcinogênese Molecular, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
- Fundação Instituto Oswaldo Cruz, Vice-presidência de Pesquisa e Laboratórios de Referência, Rio de Janeiro, Brazil
| | | | - Eduardo Queiroga
- Laboratório Bacchi, Consultoria em Patologia, Botucatu, São Paulo, Brazil
| | - Carlos E Bacchi
- Laboratório Bacchi, Consultoria em Patologia, Botucatu, São Paulo, Brazil
| | - Claudete Esteves Klumb
- Programa de Pesquisa em Hemato-Oncologia Molecular, Instituto Nacional de Câncer, Rio de Janeiro, Brazil.
- Laboratório de Hemato-Oncologia Celular e Molecular, Instituto Nacional de Câncer, Praça da Cruz Vermelha, 23, 6° andar, ala C, Rio de Janeiro, RJ, CEP: 20230-130, Brazil.
| |
Collapse
|
152
|
Abstract
Lung cancer is the leading cause of cancer deaths worldwide and over 80% of lung cancer patients are classified as having non-small cell lung cancer. Although there have been technological advancements in the early detection and standard treatment of lung cancer, it is often diagnosed at an advanced stage and is chemoresistant to most available drugs. A number of studies have demonstrated that microRNA is able to modulate various tumorigenic processes, including progression and metastasis, in various mechanisms. In this review we examine the most recent achievements in microRNA and lung cancer treatment and summarize the research progress on the reciprocal regulation between microRNA and epigenetic modifications, as both have been intensively studied in lung cancer. Epigenetic modifications on the human genome regulate gene and microRNA expression at the transcriptional level; inversely, microRNA can also transcriptionally cleave and/or translationally repress the expression of several key enzymes involved in epigenetic processes such as DNA methylation and histone modification. Better understanding of reciprocal regulation between microRNA and epigenetic modifications will underlie the development of novel microRNA orientated diagnostic and therapeutic strategies relating to lung cancer in the near future.
Collapse
Affiliation(s)
- Rajeev Kumar
- Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA
| | - Yaguang Xi
- Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA
| |
Collapse
|
153
|
Yuan Q, Sun T, Ye F, Kong W, Jin H. MicroRNA-124-3p affects proliferation, migration and apoptosis of bladder cancer cells through targeting AURKA. Cancer Biomark 2018; 19:93-101. [PMID: 28269755 DOI: 10.3233/cbm-160427] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVE The aim of this study was to establish the relationship between miR-124-3p and Aurora A kinase (AURKA) in bladder cancer (BC). METHODS The expressions of miR-124-3p and AURKA in BC tissues and cell lines were detected using RT-PCR and western blot. BC cells were transfected with miR-124-3p mimics and AURKA siRNA. After this cell proliferation, migration, cell cycle and apoptosis were measured using CCK-8, colony formation assay, wound healing assay and cytometry tests. The relationship between miR-124-3p and AURKA was confirmed with luciferase reporter assay. Mice xenograft models were constructed to examine the effects of AURKA on BC in vivo. RESULTS MiR-124-3p expression was significantly down-regulated in BC tissues and cell lines, while AURKA was significantly up-regulated compared to normal samples. MiR-124-3p targeted AURKA and decreased its expression. Transfection of miR-124-3p mimics and AURKA siRNA was shown to down-regulate BC cell proliferation and migration as well as induce cell apoptosis. As suggested by xenograft models, the inhibition of AURKA can effectively suppress tumor growth. CONCLUSION MiR-124-3p has significant impact on proliferation, migration and apoptosis of BC cells by targeting AURKA.
Collapse
Affiliation(s)
- Qiuyue Yuan
- Cancer Cente, The First Hospital of Jilin University, Changchun, Jilin 130021, China.,Cancer Cente, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Tingge Sun
- Cancer Cente, The First Hospital of Jilin University, Changchun, Jilin 130021, China.,Cancer Cente, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Feng Ye
- Cancer Cente, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Weisheng Kong
- BASO Cell Science and Technology Co., Ltd., Zhuhai, Guangdong 519015, China
| | - Haofan Jin
- Cancer Cente, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| |
Collapse
|
154
|
Sun Y, Xu R, Huang J, Yao Y, Pan X, Chen Z, Ma G. Insulin-like growth factor-1-mediated regulation of miR-193a expression promotes the migration and proliferation of c-kit-positive mouse cardiac stem cells. Stem Cell Res Ther 2018; 9:41. [PMID: 29467020 PMCID: PMC5822561 DOI: 10.1186/s13287-017-0762-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 12/13/2017] [Accepted: 12/22/2017] [Indexed: 12/13/2022] Open
Abstract
Background C-kit-positive cardiac stem cells (CSCs) have been shown to be a promising candidate treatment for myocardial infarction and heart failure. Insulin-like growth factor (IGF)-1 is an anabolic growth hormone that regulates cellular proliferation, differentiation, senescence, and death in various tissues. Although IGF-1 promotes the migration and proliferation of c-kit-positive mouse CSCs, the underlying mechanism remains unclear. Methods Cells were isolated from adult mouse hearts, and c-kit-positive CSCs were separated using magnetic beads. The cells were cultured with or without IGF-1, and c-kit expression was measured by Western blotting. IGF-1 induced CSC proliferation and migration, as measured through Cell Counting Kit-8 (CCK-8) and Transwell assays, respectively. The miR-193a expression was measured by quantitative real-time PCR (qPCR) assays. Results IGF-1 enhanced c-kit expression in c-kit-positive CSCs. The activities of the phosphoinositol 3-kinase (PI3K)/AKT signaling pathway and DNA methyltransferases (DNMTs) were enhanced, and their respective inhibitors LY294002 and 5-azacytidine (5-AZA) blunted c-kit expression. Based on the results of quantitative real-time PCR (qPCR) assays, the expression of miR-193a, which is embedded in a CpG island, was down-regulated in the IGF-1-stimulated group and negatively correlated with c-kit expression, whereas c-kit-positive CSCs infected with lentivirus carrying micro-RNA193a displayed reduced c-kit expression, migration and proliferation. Conclusions IGF-1 upregulated c-kit expression in c-kit-positive CSCs resulting in enhanced CSC proliferation and migration by activating the PI3K/AKT/DNMT signaling pathway to epigenetically silence miR-193a, which negatively modifies the c-kit expression level. Electronic supplementary material The online version of this article (10.1186/s13287-017-0762-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yuning Sun
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, DingjiaQiao No. 87, Hunan Road, Nanjing, 210009, Jiangsu, China
| | - Rongfeng Xu
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, DingjiaQiao No. 87, Hunan Road, Nanjing, 210009, Jiangsu, China
| | - Jia Huang
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, DingjiaQiao No. 87, Hunan Road, Nanjing, 210009, Jiangsu, China
| | - Yuyu Yao
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, DingjiaQiao No. 87, Hunan Road, Nanjing, 210009, Jiangsu, China
| | - Xiaodong Pan
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, DingjiaQiao No. 87, Hunan Road, Nanjing, 210009, Jiangsu, China
| | - Zhongpu Chen
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, DingjiaQiao No. 87, Hunan Road, Nanjing, 210009, Jiangsu, China.
| | - Genshan Ma
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, DingjiaQiao No. 87, Hunan Road, Nanjing, 210009, Jiangsu, China.
| |
Collapse
|
155
|
Hervouet E, Peixoto P, Delage-Mourroux R, Boyer-Guittaut M, Cartron PF. Specific or not specific recruitment of DNMTs for DNA methylation, an epigenetic dilemma. Clin Epigenetics 2018; 10:17. [PMID: 29449903 PMCID: PMC5807744 DOI: 10.1186/s13148-018-0450-y] [Citation(s) in RCA: 160] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 01/30/2018] [Indexed: 11/28/2022] Open
Abstract
Our current view of DNA methylation processes is strongly moving: First, even if it was generally admitted that DNMT3A and DNMT3B are associated with de novo methylation and DNMT1 is associated with inheritance DNA methylation, these distinctions are now not so clear. Secondly, since one decade, many partners of DNMTs have been involved in both the regulation of DNA methylation activity and DNMT recruitment on DNA. The high diversity of interactions and the combination of these interactions let us to subclass the different DNMT-including complexes. For example, the DNMT3L/DNMT3A complex is mainly related to de novo DNA methylation in embryonic states, whereas the DNMT1/PCNA/UHRF1 complex is required for maintaining global DNA methylation following DNA replication. On the opposite to these unspecific DNA methylation machineries (no preferential DNA sequence), some recently identified DNMT-including complexes are recruited on specific DNA sequences. The coexistence of both types of DNA methylation (un/specific) suggests a close cooperation and an orchestration between these systems to maintain genome and epigenome integrities. Deregulation of these systems can lead to pathologic disorders.
Collapse
Affiliation(s)
- Eric Hervouet
- INSERM unit 1098, University of Bourgogne Franche-Comté, Besançon, France.,EPIGENExp (EPIgenetics and GENe EXPression Technical Platform), Besançon, France
| | - Paul Peixoto
- INSERM unit 1098, University of Bourgogne Franche-Comté, Besançon, France.,EPIGENExp (EPIgenetics and GENe EXPression Technical Platform), Besançon, France
| | | | | | - Pierre-François Cartron
- 3INSERM unit S1232, University of Nantes, Nantes, France.,4Institut de cancérologie de l'Ouest, Nantes, France.,REpiCGO (Cancéropole Grand-Ouest), Nantes, France.,EpiSAVMEN Networks, Nantes, Région Pays de la Loire France
| |
Collapse
|
156
|
Epigenetics and MicroRNAs in Cancer. Int J Mol Sci 2018; 19:ijms19020459. [PMID: 29401683 PMCID: PMC5855681 DOI: 10.3390/ijms19020459] [Citation(s) in RCA: 101] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 01/29/2018] [Accepted: 01/30/2018] [Indexed: 02/08/2023] Open
Abstract
The ability to reprogram the transcriptional circuitry by remodeling the three-dimensional structure of the genome is exploited by cancer cells to promote tumorigenesis. This reprogramming occurs because of hereditable chromatin chemical modifications and the consequent formation of RNA-protein-DNA complexes that represent the principal actors of the epigenetic phenomena. In this regard, the deregulation of a transcribed non-coding RNA may be both cause and consequence of a cancer-related epigenetic alteration. This review summarizes recent findings that implicate microRNAs in the aberrant epigenetic regulation of cancer cells.
Collapse
|
157
|
Trino S, Lamorte D, Caivano A, Laurenzana I, Tagliaferri D, Falco G, Del Vecchio L, Musto P, De Luca L. MicroRNAs as New Biomarkers for Diagnosis and Prognosis, and as Potential Therapeutic Targets in Acute Myeloid Leukemia. Int J Mol Sci 2018; 19:ijms19020460. [PMID: 29401684 PMCID: PMC5855682 DOI: 10.3390/ijms19020460] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 01/12/2018] [Accepted: 01/12/2018] [Indexed: 02/07/2023] Open
Abstract
Acute myeloid leukemias (AML) are clonal disorders of hematopoietic progenitor cells which are characterized by relevant heterogeneity in terms of phenotypic, genotypic, and clinical features. Among the genetic aberrations that control disease development there are microRNAs (miRNAs). miRNAs are small non-coding RNAs that regulate, at post-transcriptional level, translation and stability of mRNAs. It is now established that deregulated miRNA expression is a prominent feature in AML. Functional studies have shown that miRNAs play an important role in AML pathogenesis and miRNA expression signatures are associated with chemotherapy response and clinical outcome. In this review we summarized miRNA signature in AML with different cytogenetic, molecular and clinical characteristics. Moreover, we reviewed the miRNA regulatory network in AML pathogenesis and we discussed the potential use of cellular and circulating miRNAs as biomarkers for diagnosis and prognosis and as therapeutic targets.
Collapse
MESH Headings
- Animals
- Antagomirs/genetics
- Antagomirs/metabolism
- Antagomirs/therapeutic use
- Biomarkers, Tumor/agonists
- Biomarkers, Tumor/antagonists & inhibitors
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Chromosome Aberrations
- Extracellular Vesicles/metabolism
- Extracellular Vesicles/pathology
- Gene Expression Regulation, Leukemic
- Humans
- Leukemia, Myeloid, Acute/diagnosis
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/therapy
- Mice
- MicroRNAs/agonists
- MicroRNAs/antagonists & inhibitors
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Molecular Targeted Therapy
- Oligoribonucleotides/genetics
- Oligoribonucleotides/metabolism
- Oligoribonucleotides/therapeutic use
- Oncogene Proteins, Fusion/antagonists & inhibitors
- Oncogene Proteins, Fusion/genetics
- Oncogene Proteins, Fusion/metabolism
- Prognosis
- Signal Transduction
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Stefania Trino
- Laboratory of Preclinical and Translational Research, IRCCS-Referral Cancer Center of Basilicata (CROB), 85028 Rionero in Vulture, Italy.
| | - Daniela Lamorte
- Laboratory of Preclinical and Translational Research, IRCCS-Referral Cancer Center of Basilicata (CROB), 85028 Rionero in Vulture, Italy.
| | - Antonella Caivano
- Laboratory of Preclinical and Translational Research, IRCCS-Referral Cancer Center of Basilicata (CROB), 85028 Rionero in Vulture, Italy.
| | - Ilaria Laurenzana
- Laboratory of Preclinical and Translational Research, IRCCS-Referral Cancer Center of Basilicata (CROB), 85028 Rionero in Vulture, Italy.
| | - Daniela Tagliaferri
- Biogem Scarl, Istituto di Ricerche Genetiche 'Gaetano Salvatore', 83031 Ariano Irpino, Italy.
| | - Geppino Falco
- Biogem Scarl, Istituto di Ricerche Genetiche 'Gaetano Salvatore', 83031 Ariano Irpino, Italy.
- Department of Biology, University of Naples Federico II, 80147 Naples, Italy.
| | - Luigi Del Vecchio
- CEINGE Biotecnologie Avanzate s.c.a r.l., 80147 Naples, Italy.
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, 80138 Naples, Italy.
| | - Pellegrino Musto
- Scientific Direction, IRCCS-Referral Cancer Center of Basilicata (CROB), 85028 Rionero in Vulture, Potenza, Italy.
| | - Luciana De Luca
- Laboratory of Preclinical and Translational Research, IRCCS-Referral Cancer Center of Basilicata (CROB), 85028 Rionero in Vulture, Italy.
| |
Collapse
|
158
|
Pan Y, Liu G, Zhou F, Su B, Li Y. DNA methylation profiles in cancer diagnosis and therapeutics. Clin Exp Med 2018; 18:1-14. [PMID: 28752221 DOI: 10.1007/s10238-017-0467-0] [Citation(s) in RCA: 267] [Impact Index Per Article: 38.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Accepted: 06/16/2017] [Indexed: 12/12/2022]
Abstract
Cancer initiation and proliferation is regulated by both epigenetic and genetic events with epigenetic modifications being increasingly identified as important targets for cancer research. DNA methylation catalyzed by DNA methyltransferases (DNMTs) is one of the essential epigenetic mechanisms that control cell proliferation, apoptosis, differentiation, cell cycle, and transformation in eukaryotes. Recent progress in epigenetics revealed a deeper understanding of the mechanisms of tumorigenesis and provided biomarkers for early detection, diagnosis, and prognosis in cancer patients. Although DNA methylation biomarker possesses potential contributing to precision medicine, there are still limitations to be overcome before it reaches clinical setting. Hence, the current status of DNA methylation biomarkers was reviewed and the future use in clinic was also predicted.
Collapse
Affiliation(s)
- Yunbao Pan
- Department of Laboratory Medicine, Zhongnan Hospital, Wuhan University, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, Guangdong, China
| | - Guohong Liu
- School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou, 510275, Guangdong, China
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, 6767 Bertner Ave, Houston, TX, 77030, USA
| | - Fuling Zhou
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| | - Bojin Su
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, 6767 Bertner Ave, Houston, TX, 77030, USA.
| | - Yirong Li
- Department of Laboratory Medicine, Zhongnan Hospital, Wuhan University, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China.
| |
Collapse
|
159
|
Wang LH, Huang J, Wu CR, Huang LY, Cui J, Xing ZZ, Zhao CY. Downregulation of miR‑29b targets DNMT3b to suppress cellular apoptosis and enhance proliferation in pancreatic cancer. Mol Med Rep 2018; 17:2113-2120. [PMID: 29207141 PMCID: PMC5783451 DOI: 10.3892/mmr.2017.8145] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 05/19/2017] [Indexed: 12/16/2022] Open
Abstract
As one of the most aggressive types of tumor, pancreatic cancer is a principal cause of tumor‑associated mortality. Negative associations between microRNA‑29 (miR‑29) and DNA methyltransferases (DNMT) 3a and 3b have been demonstrated to be associated with the carcinogenesis of a number of types of cancer; however, this has not been completely elucidated in pancreatic cancer. In the present study, pancreatic cancer tissues (n=15) and corresponding paracancerous tissues (n=15) were obtained and the results of reverse transcription‑quantitative polymerase chain reaction analysis indicated decreased expression of miR‑29b and enhanced mRNA expression of DNMT3b in pancreatic cancer tissues, compared with the corresponding paracancerous tissues. Increased protein expression of DNMT3b was demonstrated by western blotting and immunohistochemistry. In addition, the negative association between miR‑29b and DNMT3b was noted in pancreatic cancer tissues, and luciferase reporter assays confirmed that miR‑29b was able to directly target DNMT3b in vitro. Notably, miR‑29b overexpression was able to decrease cell viability and to promote the apoptosis by targeting DNMT3b, and the knockdown of DNMT3b exhibited consistent results in vitro and in vivo. The results of the present study suggested that miR‑29b, as a tumor suppressor, may be a novel target for the development of treatments for pancreatic cancer.
Collapse
Affiliation(s)
- Li-Hua Wang
- Department of Gastroenterology, Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, P.R. China
| | - Ju Huang
- Department of Queen Mary University, Medical College of Nanchang University, Nanchang, Jiangxi 330038, P.R. China
| | - Cheng-Rong Wu
- Department of Gastroenterology, Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, P.R. China
| | - Liu-Ye Huang
- Department of Gastroenterology, Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, P.R. China
| | - Jun Cui
- Department of Gastroenterology, Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, P.R. China
| | - Zhi-Zhi Xing
- Department of Gastroenterology, Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, P.R. China
| | - Chun-Yu Zhao
- Department of Gastroenterology, Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, P.R. China
| |
Collapse
|
160
|
Anthelmintic niclosamide suppresses transcription of BCR-ABL fusion oncogene via disabling Sp1 and induces apoptosis in imatinib-resistant CML cells harboring T315I mutant. Cell Death Dis 2018; 9:68. [PMID: 29358661 PMCID: PMC5833368 DOI: 10.1038/s41419-017-0075-7] [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: 06/14/2017] [Revised: 09/13/2017] [Accepted: 09/25/2017] [Indexed: 12/12/2022]
Abstract
Tyrosine kinase BCR-ABL fusion protein is the driver in patients with chronic myeloid leukemia (CML). The gate-keeper mutation T315I is the most challenging mutant due to its resistance to most tyrosine kinase inhibitors (TKIs). The third generation TKI ponatinib is the only effective TKI to treat CML patients harboring T315I-BCR-ABL mutation, but with high rate of major arterial thrombotic events. Alternative strategies to specifically target T315I-BCR-ABL are needed for the treatment of CML patients harboring such a mutation. Given that Sp1 is a fundamental transcriptional factor to positively regulate WT-BCR-ABL fusion oncogene, the purpose of this investigation was aimed at evaluating the anti-tumor activity and the underlying mechanism in terms of Sp1 regulational effect on the transcription of T315I-BCR-ABL fusion oncogene. Like in WT-BCR-ABL, we identified enrichment of Sp1 on the promoter of T315I-BCR-ABL fusion gene. Treatment of WT- and T315I-BCR-ABL-expressing CML cells by niclosamide diminished such an enrichment of Sp1, and decreased WT- and T315I-BCR-ABL transcription and its downstream signaling molecules such as STAT5 and Akt. Further, niclosamide significantly inhibited the proliferation and induced apoptosis through intrinsic pathway. The in vivo efficacy validation of p-niclosamide, a water soluble derivative of niclosamide, showed that p-niclosamide significantly inhibited the tumor burden of nude mice subcutaneously bearing T315I-BCR-ABL-expressing CML cells, and prolonged the survival of allografted leukemic mice harboring BaF3-T315I-BCR-ABL. We conclude that niclosamide is active against T315I-BCR-ABL-expressing cells, and may be a promising agent for CML patients regardless of T315I mutation status.
Collapse
|
161
|
Bai J, Zhang X, Hu K, Liu B, Wang H, Li A, Lin F, Zhang L, Sun X, Du Z, Song J. Silencing DNA methyltransferase 1 (DNMT1) inhibits proliferation, metastasis and invasion in ESCC by suppressing methylation of RASSF1A and DAPK. Oncotarget 2018; 7:44129-44141. [PMID: 27286455 PMCID: PMC5190084 DOI: 10.18632/oncotarget.9866] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 03/31/2016] [Indexed: 11/25/2022] Open
Abstract
Our previous study showed DNMT1 is up-regulated in esophageal squamous cell carcinoma (ESCC), which is associated with methylation of tumor suppressors. In the current study, we investigate the role of DNMT1 in ESCC. We found silencing DNMT1 inhibited proliferation, metastasis and invasion of three different ESCC cells, K150, K410 and K450. We also found silencing DNMT1 induced G1 arrest and cell apoptosis in K150, K410 and K450 cells. In vivo study showed silencing DNMT1 suppressed tumor growth in nude mice. In addition, silencing DNMT1 increased expression of tumor suppressor genes, RASSF1A and DAPK, in ESCC cells and ESCC xenograft in nude mice. Moreover, silencing DNMT1 decreased methylation in promoter of RASSF1A and DAPK. In conclusion, our data demonstrated that silencing DNMT1 inhibits proliferation, metastasis and invasion in ESCC by suppressing methylation of RASSF1A and DAPK.
Collapse
Affiliation(s)
- Jian Bai
- Department of Thoracic & Cardiovascular Surgery, The Second Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Xue Zhang
- Department of ICU, The Second Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Kai Hu
- Department of Thoracic & Cardiovascular Surgery, The Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Bangqing Liu
- Department of Thoracic & Cardiovascular Surgery, The Second Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Haiyong Wang
- Department of Thoracic & Cardiovascular Surgery, The Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Angui Li
- Department of Thoracic & Cardiovascular Surgery, The Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Feng Lin
- Department of Thoracic & Cardiovascular Surgery, The Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Lifei Zhang
- Department of Thoracic & Cardiovascular Surgery, The Second Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Xiaolin Sun
- Department of Thoracic & Cardiovascular Surgery, The Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Zhenzong Du
- Department of Thoracic & Cardiovascular Surgery, The Second Affiliated Hospital of Guilin Medical University, Guilin, China.,Current address: Department of Thoracic & Cardiovascular Surgery, The Second Affiliated Hospital of Guilin Medical University, Lingui District, Guilin, China
| | - Jianfei Song
- Department of Thoracic & Cardiovascular Surgery, The Second Affiliated Hospital of Guilin Medical University, Guilin, China.,Current address: Department of Thoracic & Cardiovascular Surgery, The Second Affiliated Hospital of Guilin Medical University, Lingui District, Guilin, China
| |
Collapse
|
162
|
Zhao X, Liu Y, Li Z, Zheng S, Wang Z, Li W, Bi Z, Li L, Jiang Y, Luo Y, Lin Q, Fu Z, Rufu C. Linc00511 acts as a competing endogenous RNA to regulate VEGFA expression through sponging hsa-miR-29b-3p in pancreatic ductal adenocarcinoma. J Cell Mol Med 2018; 22:655-667. [PMID: 28984028 PMCID: PMC5742682 DOI: 10.1111/jcmm.13351] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 07/19/2017] [Indexed: 01/12/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a lethal malignancy. Long non-coding RNAs (lncRNAs) are important regulators in pathological processes, yet their potential roles in PDAC are poorly understood. Here, we identify a fundamental role for a novel lincRNA, linc00511, in the progression of PDAC. Linc00511 levels in PDAC tissue specimens and cell lines were examined by quantitative real-time PCR. Corresponding adjacent non-neoplastic tissues were used as controls. The function of linc00511 in PDAC cell lines was determined by RNA interference approach in vitro and in vivo. Fluorescence in situ hybridization (FISH) was used to characterize linc00511 expression in PDAC cells. Insights of the mechanism of competitive endogenous RNAs (ceRNAs) were obtained from bioinformatic analysis, luciferase assays and RIP assays. The association between the linc00511/hsa-miR29b-3p axis and VEGFA was verified by Western blotting assay. Immunohistochemistry was performed to evaluate the expression of VEGFA in PDAC samples. The aberrant up-regulation of linc00511 was detected in PDAC cell lines and patient specimens compared with controls. An increase in linc00511 expression indicates the adverse clinical pathological characteristics and poor prognosis. Functionally, linc00511 depletion in PDAC cells decreased proliferation, migration, invasion and endothelial tube formation. Mechanistically, linc00511 could up-regulate VEGFA via its competing endogenous RNA (ceRNA) activity on hsa-miR-29b-3p. In summary, our results define an important axis controlling proliferation, invasion and tumour angiogenesis in PDAC. Linc00511 is a novel lncRNA that plays a significant regulatory role in the pathogenesis and progression of PDAC. Thus, Linc00511 represents a new prognostic biomarker to predict clinical outcome of PDAC patients after surgery and may serve as a potential therapeutic target for PDAC treatment.
Collapse
Affiliation(s)
- Xiaohui Zhao
- Department of RadiotherapySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
- Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong higher Education InstitutesSun Yat‐Sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Yimin Liu
- Department of RadiotherapySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
- Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong higher Education InstitutesSun Yat‐Sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Zhihua Li
- Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong higher Education InstitutesSun Yat‐Sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
- Department of Medical OncologySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Shangyou Zheng
- Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong higher Education InstitutesSun Yat‐Sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
- Department of Hepatobiliary SurgerySun Yat‐sen Memorial HospitalGuangzhouChina
| | - Zairui Wang
- Department of NephrologyArmed Police Corps Hospital of Guangdong ProvinceGuangdongChina
| | - Wenzhu Li
- Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong higher Education InstitutesSun Yat‐Sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
- Department of Medical OncologySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Zhuofei Bi
- Department of RadiotherapySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
- Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong higher Education InstitutesSun Yat‐Sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Liting Li
- Department of RadiotherapySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
- Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong higher Education InstitutesSun Yat‐Sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Yanhui Jiang
- Department of RadiotherapySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
- Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong higher Education InstitutesSun Yat‐Sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Yuming Luo
- Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong higher Education InstitutesSun Yat‐Sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
- Department of Hepatobiliary SurgerySun Yat‐sen Memorial HospitalGuangzhouChina
| | - Qing Lin
- Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong higher Education InstitutesSun Yat‐Sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
- Department of Hepatobiliary SurgerySun Yat‐sen Memorial HospitalGuangzhouChina
| | - Zhiqiang Fu
- Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong higher Education InstitutesSun Yat‐Sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
- Department of Hepatobiliary SurgerySun Yat‐sen Memorial HospitalGuangzhouChina
| | - Chen Rufu
- Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong higher Education InstitutesSun Yat‐Sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
- Department of Hepatobiliary SurgerySun Yat‐sen Memorial HospitalGuangzhouChina
| |
Collapse
|
163
|
Zo RB, Long Z. MiR-124-3p suppresses bladder cancer by targeting DNA methyltransferase 3B. J Cell Physiol 2018; 234:464-474. [PMID: 29893409 DOI: 10.1002/jcp.26591] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 03/09/2018] [Indexed: 12/22/2022]
Abstract
This study was aimed to uncover the effects of miR-124-3p on bladder cancer (BC) by regulating DNA methyltransferase 3B. The expressions of miR-124-3p and DNMT3B mRNA in BC tissues and cell lines were detected using RT-PCR. The expression of DNMT3B in cells was determined using Western blot and immunohistochemistry in tissues. In addition, chromogenic in situ hybridization staining was used to measure the expression of miR-124-3p in tissues. BC cells were transfected with miR-124-3p mimics, miR-124-3p inhibitors, DNMT3B siRNAs, and DNMT3B cDNAs + miR-124-3p mimics. Subsequently, cell proliferation, apoptosis, migration, and invasion were measured using CCK-8, the cytometry test, wound healing assay, and Transwell assay, respectively. Finally, the relationship between miR-124-3p and DNMT3B was confirmed using dual luciferase reporter gene assay. MiR-124-3p expression was significantly lower and the level of DNMT3B was significantly higher in BC tissues and cell lines compared with the normal controls. MiR-124-3p was verified to target DNMT3B. The transfection of miR-124-3p mimics and DNMT3B siRNAs down-regulated BC cell proliferation, migration, and invasion, as well as induced cell apoptosis; miR-124-3p inhibitors promoted BC cell proliferation, migration, invasion, and reduced cell apoptosis; and the effects of DNMT3B cDNAs can be compromised by miR-124-3p mimics. Thus, we concluded that miR-124-3p could suppress the proliferation, migration, invasion, and promote apoptosis of BC cells by targeting DNMT3B.
Collapse
Affiliation(s)
- Rabah B Zo
- Department of Urology Surgery, Shigatse People's Hospital, Shigatse, Tibet, P.R. China
| | - Ziwen Long
- Department of Urology Surgery, Shigatse People's Hospital, Shigatse, Tibet, P.R. China
- Department of Gastric Cancer and Soft-Tissue Sarcoma Sugery, Fudan University Shanghai Cancer Center, Shanghai, P.R. China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| |
Collapse
|
164
|
Butrym A, Rybka J, Baczyńska D, Poręba R, Kuliczkowski K, Mazur G. Clinical response to azacitidine therapy depends on microRNA-29c (miR-29c) expression in older acute myeloid leukemia (AML) patients. Oncotarget 2017; 7:30250-7. [PMID: 26862847 PMCID: PMC5058678 DOI: 10.18632/oncotarget.7172] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 01/23/2016] [Indexed: 11/25/2022] Open
Abstract
Acute myeloid leukemia (AML) is a heterogeneous disease with different clinical course and prognosis. microRNA-29 (miR-29) family of non-coding small RNAs can play an important role in pathogenesis of AML, but also can influence response to therapy. The purpose of the study was to evaluate miR-29c expression in AML patients in relationship to clinical parameters and response to chemotherapy, including azacitidine.
Collapse
Affiliation(s)
- Aleksandra Butrym
- Department of Hematology, Blood Neoplasms and Bone Marrow Transplantation, Wroclaw Medical University, Wroclaw, Poland.,Department of Physiology, Wroclaw Medical University, Wroclaw, Poland
| | - Justyna Rybka
- Department of Hematology, Blood Neoplasms and Bone Marrow Transplantation, Wroclaw Medical University, Wroclaw, Poland
| | - Dagmara Baczyńska
- Department of Forensic Medicine, Molecular Techniques Unit, Wroclaw Medical University, Wroclaw, Poland
| | - Rafał Poręba
- Department of Internal, Occupational Diseases, Hypertension and Clinical Oncology, Wroclaw Medical University, Wroclaw, Poland
| | - Kazimierz Kuliczkowski
- Department of Hematology, Blood Neoplasms and Bone Marrow Transplantation, Wroclaw Medical University, Wroclaw, Poland
| | - Grzegorz Mazur
- Department of Internal, Occupational Diseases, Hypertension and Clinical Oncology, Wroclaw Medical University, Wroclaw, Poland
| |
Collapse
|
165
|
Yan F, Shen N, Pang J, Zhao N, Deng B, Li B, Yang Y, Yang P, Molina JR, Liu S. A regulatory circuit composed of DNA methyltransferases and receptor tyrosine kinases controls lung cancer cell aggressiveness. Oncogene 2017; 36:6919-6928. [PMID: 28869603 PMCID: PMC5730463 DOI: 10.1038/onc.2017.305] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 06/21/2017] [Accepted: 07/28/2017] [Indexed: 12/12/2022]
Abstract
Overexpression of DNMT1 and KIT is prevalent in lung cancer, yet the underlying molecular mechanisms are poorly understood. While the deregulated activation of DNMT1 or KIT has been implicated in lung cancer pathogenesis, whether and how DNMT1 and KIT orchestrate lung tumorigenesis are unclear. Here, using human lung cancer tissue microarrays and fresh frozen tissues, we found that the overexpression of DNMT1 is positively correlated with the upregulation of KIT in tumor tissues. We demonstrated that DNMT1 and KIT form a positive regulatory loop, in which ectopic DNMT1 expression increases, whereas targeted DNMT1 depletion abrogates KIT signaling cascade through Sp1/miR-29b network. Conversely, an increase of KIT levels augments, but a reduction of KIT expression ablates DNMT1 transcription by STAT3 pathway leading to in-parallel modification of the DNA methylation profiles. We provided evidence that KIT inactivation induces global DNA hypomethylation, restores the expression of tumor suppressor p15INK4B through promoter demethylation; in turn, DNMT1 dysfunction impairs KIT kinase signaling. Functionally, KIT and DNMT1 co-expression promotes, whereas dual inactivation of them suppresses, lung cancer cell proliferation and metastatic growth in vitro and in vivo, in a synergistic manner. These findings demonstrate the regulatory and functional interplay between DNA methylation and tyrosine kinase signaling in propelling tumorigenesis, providing a widely applicable approach for targeting lung cancer.
Collapse
Affiliation(s)
- Fei Yan
- The Hormel Institute, University of Minnesota, 801 16th Avenue NE, Austin, MN 55912, USA
| | - Na Shen
- The Hormel Institute, University of Minnesota, 801 16th Avenue NE, Austin, MN 55912, USA
| | - Jiuxia Pang
- The Hormel Institute, University of Minnesota, 801 16th Avenue NE, Austin, MN 55912, USA
| | - Na Zhao
- The Hormel Institute, University of Minnesota, 801 16th Avenue NE, Austin, MN 55912, USA
| | - Bo Deng
- Division of Epidemiology, Mayo Clinic, 200 1st Street SW, Rochester, MN 55905, USA
| | - Bing Li
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY 40202, USA
| | - Yanan Yang
- Division of Epidemiology, Mayo Clinic, 200 1st Street SW, Rochester, MN 55905, USA
| | - Ping Yang
- Division of Epidemiology, Mayo Clinic, 200 1st Street SW, Rochester, MN 55905, USA
| | - Julian R. Molina
- Department of Medical Oncology, Mayo Clinic, 200 1st Street SW, Rochester, MN 55905, USA
| | - Shujun Liu
- The Hormel Institute, University of Minnesota, 801 16th Avenue NE, Austin, MN 55912, USA
| |
Collapse
|
166
|
Kuc C, Richard DJ, Johnson S, Bragg L, Servos MR, Doxey AC, Craig PM. Rainbow trout exposed to benzo[a]pyrene yields conserved microRNA binding sites in DNA methyltransferases across 500 million years of evolution. Sci Rep 2017; 7:16843. [PMID: 29203905 PMCID: PMC5715007 DOI: 10.1038/s41598-017-17236-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 11/22/2017] [Indexed: 12/24/2022] Open
Abstract
The objective of this study was to examine the regulation of DNA methylation following acute (24 h) and prolonged (14 d) exposure to low (1 ng/L) and high (10 ng/L) benzo[a]pyrene. However, with the recent release of the rainbow trout genome, we were able to conduct a more detailed analysis regarding the regulation of the enzymes involved in DNA methylation; DNA methyltransferases (DNMTs). Bioinformatic approaches were used to identify candidate microRNA (miRNA) that potentially bind to the DNMT1 and DNMT3a 3′UTR. Results indicated a significant decrease in global methylation in both liver and muscle, with an associated decrease in DNA methyltransferase activity and DNMT3a transcript abundance. There was a significant increase in one specific candidate miRNA (miR29a) that was predicted to bind to DNMT3a. Taking a comparative genomics approach, the binding sites of miR29a to the DNMT3a 3′UTR was compared across species, spanning fish to mammals, and revealed a highly conserved binding motif that has been maintained since the vertebrate ancestor, approximately 500 million years ago. This research establishes that miRNA act as an essential mediator between the environment and DNA methylation patterns via DNMTs, which is further confirmed by a genomic regulatory mechanism that has been deeply conserved throughout evolution.
Collapse
Affiliation(s)
- Christopher Kuc
- Department of Biology, University of Waterloo, Waterloo, ON, Canada.,Molecular and Cellular Biology, University of Guelph, Guelph, ON, Canada
| | - Daniel J Richard
- Department of Biology, University of Waterloo, Waterloo, ON, Canada
| | - Samantha Johnson
- Department of Biology, University of Waterloo, Waterloo, ON, Canada.,Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Leslie Bragg
- Department of Biology, University of Waterloo, Waterloo, ON, Canada
| | - Mark R Servos
- Department of Biology, University of Waterloo, Waterloo, ON, Canada
| | - Andrew C Doxey
- Department of Biology, University of Waterloo, Waterloo, ON, Canada
| | - Paul M Craig
- Department of Biology, University of Waterloo, Waterloo, ON, Canada.
| |
Collapse
|
167
|
Diminished microRNA-29b level is associated with BRD4-mediated activation of oncogenes in cutaneous T-cell lymphoma. Blood 2017; 131:771-781. [PMID: 29180399 DOI: 10.1182/blood-2017-09-805663] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 11/15/2017] [Indexed: 02/07/2023] Open
Abstract
MicroRNA (miRNA) dysregulation is a hallmark of cutaneous T-cell lymphoma (CTCL), an often-fatal malignancy of skin-homing CD4+ T cells for which there are few effective therapies. The role of microRNAs (miRs) in controlling epigenetic modifier-dependent transcriptional regulation in CTCL is unknown. In this study, we characterize a novel miR dysregulation that contributes to overexpression of the epigenetic reader bromodomain-containing protein 4 (BRD4). We used patient CD4+ T cells to show diminished levels of miR-29b compared with healthy donor cells. Patient cells and miR-29b-/- mouse cells revealed an inverse relationship between miR-29b and BRD4, the latter of which is overexpressed in these cells. Chromatin immunoprecipitation and sequencing analysis revealed increased genome-wide BRD4 occupancy at promoter and enhancer regions in CD4+ T cells from CTCL patients. The cumulative result of BRD4 binding was increased expression of tumor-associated genes such as NOTCH1 and RBPJ, as well as the interleukin-15 (IL-15) receptor complex, the latter enhancing IL-15 autocrine signaling. Furthermore, we confirm the in vivo relevance of this pathway in our IL-15 transgenic mouse model of CTCL by showing that interference with BRD4-mediated pathogenesis, either by restoring miR-29b levels via bortezomib treatment or by directly inhibiting BRD4 binding via JQ1 treatment, prevents progression of CTCL. We describe a novel oncogenic pathway featuring IL-15, miR-29b, and BRD4 in CTCL and suggest targeting of these components as a potentially effective therapy for CTCL patients.
Collapse
|
168
|
Ferrosenescence: The iron age of neurodegeneration? Mech Ageing Dev 2017; 174:63-75. [PMID: 29180225 DOI: 10.1016/j.mad.2017.11.012] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 11/13/2017] [Accepted: 11/15/2017] [Indexed: 12/15/2022]
Abstract
Aging has been associated with iron retention in many cell types, including the neurons, promoting neurodegeneration by ferroptosis. Excess intracellular iron accelerates aging by damaging the DNA and blocking genomic repair systems, a process we define as ferrosenescence. Novel neuroimaging and proteomic techniques have pinpointed indicators of both iron retention and ferrosenescence, allowing for their early correction, potentially bringing prevention of neurodegenerative disorders within reach. In this review, we take a closer look at the early markers of iron dyshomeostasis in neurodegenerative disorders, focusing on preventive strategies based on nutritional and microbiome manipulations.
Collapse
|
169
|
Luo Z, Wu G, Zhang D, Liu J, Ran R. microRNA‑625 targets Yes‑associated protein 1 to suppress cell proliferation and invasion of osteosarcoma. Mol Med Rep 2017; 17:2005-2011. [PMID: 29257207 DOI: 10.3892/mmr.2017.8079] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2017] [Accepted: 07/20/2017] [Indexed: 11/06/2022] Open
Abstract
Osteosarcoma (OS) is the leading cause of cancer‑associated mortality in adolescents and children. MicroRNAs (miRNAs) have critical roles in cancer, particularly in the initiation and progression of OS. Therefore, OS‑associated miRNAs should be identified for use as therapeutic targets for treatment of OS. This study aimed to investigate the expression pattern, potential roles and underlying mechanism of microRNA‑625 (miR‑625) in OS. miR‑625 was markedly downregulated in OS tissues and cell lines compared with that in associated adjacent non‑tumor tissues and human normal osteoblasts, respectively. The enforced expression of miR‑625 using miRNA mimics significantly reduced the proliferation and invasion of OS cells. Bioinformatics analysis and luciferase reporter assays indicated that miR‑625 targeted the 3'‑untranslated region of Yes‑associated protein 1 (YAP‑1). Furthermore, upregulation of miR‑625 reduced endogenous YAP1 expression at the mRNA and protein levels. The upregulated expression of YAP1 in OS tissues was inversely correlated with miR‑625 expression. YAP1 restoration using a recombinant plasmid rescued the miR‑625‑mediated tumor‑suppressive effects in OS cells. In conclusion, miR‑625 attenuated the cell proliferation and invasion of OS by suppressing YAP1. Thus, miR‑625 may be a potential target for OS therapy.
Collapse
Affiliation(s)
- Zheng Luo
- Department of Spinal Surgery, The Central Hospital of Enshi Autonomous Prefecture, Enshi, Hubei 445000, P.R. China
| | - Gang Wu
- Department of Inspection, The Central Hospital of Enshi Autonomous Prefecture, Enshi, Hubei 445000, P.R. China
| | - Daiyang Zhang
- Department of Spinal Surgery, The Central Hospital of Enshi Autonomous Prefecture, Enshi, Hubei 445000, P.R. China
| | - Jun Liu
- Department of Spinal Surgery, The Central Hospital of Enshi Autonomous Prefecture, Enshi, Hubei 445000, P.R. China
| | - Renguo Ran
- Department of Spinal Surgery, The Central Hospital of Enshi Autonomous Prefecture, Enshi, Hubei 445000, P.R. China
| |
Collapse
|
170
|
Abstract
Glioblastoma is the most aggressive brain tumor and, even with the current multimodal therapy, is an invariably lethal cancer with a life expectancy that depends on the tumor subtype but, even in the most favorable cases, rarely exceeds 2 years. Epigenetic factors play an important role in gliomagenesis, are strong predictors of outcome, and are important determinants for the resistance to radio- and chemotherapy. The latest addition to the epigenetic machinery is the noncoding RNA (ncRNA), that is, RNA molecules that are not translated into a protein and that exert their function by base pairing with other nucleic acids in a reversible and nonmutational mode. MicroRNAs (miRNA) are a class of ncRNA of about 22 bp that regulate gene expression by binding to complementary sequences in the mRNA and silence its translation into proteins. MicroRNAs reversibly regulate transcription through nonmutational mechanisms; accordingly, they can be considered as epigenetic effectors. In this review, we will discuss the role of miRNA in glioma focusing on their role in drug resistance and on their potential applications in the therapy of this tumor.
Collapse
|
171
|
Renaud L, Silveira WAD, Hazard ES, Simpson J, Falcinelli S, Chung D, Carnevali O, Hardiman G. The Plasticizer Bisphenol A Perturbs the Hepatic Epigenome: A Systems Level Analysis of the miRNome. Genes (Basel) 2017; 8:genes8100269. [PMID: 29027980 PMCID: PMC5664119 DOI: 10.3390/genes8100269] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 09/18/2017] [Accepted: 10/04/2017] [Indexed: 02/07/2023] Open
Abstract
Ubiquitous exposure to bisphenol A (BPA), an endocrine disruptor (ED), has raised concerns for both human and ecosystem health. Epigenetic factors, including microRNAs (miRNAs), are key regulators of gene expression during cancer. The effect of BPA exposure on the zebrafish epigenome remains poorly characterized. Zebrafish represents an excellent model to study cancer as the organism develops a disease that resembles human cancer. Using zebrafish as a systems toxicology model, we hypothesized that chronic BPA-exposure impacts the miRNome in adult zebrafish and establishes an epigenome more susceptible to cancer development. After a 3 week exposure to 100 nM BPA, RNA from the liver was extracted to perform high throughput mRNA and miRNA sequencing. Differential expression (DE) analyses comparing BPA-exposed to control specimens were performed using established bioinformatics pipelines. In the BPA-exposed liver, 6188 mRNAs and 15 miRNAs were differently expressed (q ≤ 0.1). By analyzing human orthologs of the DE zebrafish genes, signatures associated with non-alcoholic fatty liver disease (NAFLD), oxidative phosphorylation, mitochondrial dysfunction and cell cycle were uncovered. Chronic exposure to BPA has a significant impact on the liver miRNome and transcriptome in adult zebrafish with the potential to cause adverse health outcomes including cancer.
Collapse
Affiliation(s)
- Ludivine Renaud
- Division of Nephrology, Department of Medicine, Medical University of South Carolina (MUSC),Charleston, SC 29425, USA.
- Laboratory for Marine Systems Biology, Hollings Marine Laboratory, Charleston, SC 29412, USA.
| | - Willian A da Silveira
- Center for Genomic Medicine, Bioinformatics, Medical University of South Carolina (MUSC), Charleston, SC 29425, USA.
| | - E Starr Hazard
- Center for Genomic Medicine, Bioinformatics, Medical University of South Carolina (MUSC), Charleston, SC 29425, USA.
- Library Science and Informatics, Medical University of South Carolina (MUSC), Charleston, SC 29425, USA.
| | - Jonathan Simpson
- Center for Genomic Medicine, Bioinformatics, Medical University of South Carolina (MUSC), Charleston, SC 29425, USA.
| | - Silvia Falcinelli
- Dipartimento Scienze della Vita e dell'Ambiente, Universita Politecnica delle Marche, 60131 Ancona, Italy.
| | - Dongjun Chung
- Department of Public Health Sciences, Medical University of South Carolina (MUSC), Charleston, SC 29425, USA.
| | - Oliana Carnevali
- Dipartimento Scienze della Vita e dell'Ambiente, Universita Politecnica delle Marche, 60131 Ancona, Italy.
| | - Gary Hardiman
- Division of Nephrology, Department of Medicine, Medical University of South Carolina (MUSC),Charleston, SC 29425, USA.
- Laboratory for Marine Systems Biology, Hollings Marine Laboratory, Charleston, SC 29412, USA.
- Center for Genomic Medicine, Bioinformatics, Medical University of South Carolina (MUSC), Charleston, SC 29425, USA.
- Department of Medicine, University of California, La Jolla, CA 92093, USA.
- Department of Public Health Sciences, Medical University of South Carolina (MUSC), Charleston, SC 29425, USA.
| |
Collapse
|
172
|
Chuang TD, Khorram O. Glucocorticoids regulate MiR-29c levels in vascular smooth muscle cells through transcriptional and epigenetic mechanisms. Life Sci 2017; 186:87-91. [DOI: 10.1016/j.lfs.2017.08.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 08/02/2017] [Accepted: 08/07/2017] [Indexed: 11/28/2022]
|
173
|
Poole CJ, Zheng W, Lodh A, Yevtodiyenko A, Liefwalker D, Li H, Felsher DW, van Riggelen J. DNMT3B overexpression contributes to aberrant DNA methylation and MYC-driven tumor maintenance in T-ALL and Burkitt's lymphoma. Oncotarget 2017; 8:76898-76920. [PMID: 29100357 PMCID: PMC5652751 DOI: 10.18632/oncotarget.20176] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 07/18/2017] [Indexed: 12/26/2022] Open
Abstract
Aberrant DNA methylation is a hallmark of cancer. However, our understanding of how tumor cell-specific DNA methylation patterns are established and maintained is limited. Here, we report that in T-cell acute lymphoblastic leukemia (T-ALL) and Burkitt's lymphoma the MYC oncogene causes overexpression of DNA methyltransferase (DNMT) 1 and 3B, which contributes to tumor maintenance. By utilizing a tetracycline-regulated MYC transgene in a mouse T-ALL (EμSRα-tTA;tet-o-MYC) and human Burkitt's lymphoma (P493-6) model, we demonstrated that DNMT1 and DNMT3B expression depend on high MYC levels, and that their transcription decreased upon MYC-inactivation. Chromatin immunoprecipitation indicated that MYC binds to the DNMT1 and DNMT3B promoters, implicating a direct transcriptional regulation. Hence, shRNA-mediated knock-down of endogenous MYC in human T-ALL and Burkitt's lymphoma cell lines downregulated DNMT3B expression. Knock-down and pharmacologic inhibition of DNMT3B in T-ALL reduced cell proliferation associated with genome-wide changes in DNA methylation, indicating a tumor promoter function during tumor maintenance. We provide novel evidence that MYC directly deregulates the expression of both de novo and maintenance DNMTs, showing that MYC controls DNA methylation in a genome-wide fashion. Our finding that a coordinated interplay between the components of the DNA methylating machinery contributes to MYC-driven tumor maintenance highlights the potential of specific DNMTs for targeted therapies.
Collapse
Affiliation(s)
- Candace J. Poole
- Augusta University, Department of Biochemistry and Molecular Biology, Augusta, GA 30912, USA
| | - Wenli Zheng
- Augusta University, Department of Biochemistry and Molecular Biology, Augusta, GA 30912, USA
| | - Atul Lodh
- Augusta University, Department of Biochemistry and Molecular Biology, Augusta, GA 30912, USA
| | - Aleksey Yevtodiyenko
- Stanford University School of Medicine, Division of Oncology, Departments of Medicine and Pathology, Stanford, CA 94305, USA
| | - Daniel Liefwalker
- Stanford University School of Medicine, Division of Oncology, Departments of Medicine and Pathology, Stanford, CA 94305, USA
| | - Honglin Li
- Augusta University, Department of Biochemistry and Molecular Biology, Augusta, GA 30912, USA
| | - Dean W. Felsher
- Stanford University School of Medicine, Division of Oncology, Departments of Medicine and Pathology, Stanford, CA 94305, USA
| | - Jan van Riggelen
- Augusta University, Department of Biochemistry and Molecular Biology, Augusta, GA 30912, USA
| |
Collapse
|
174
|
Swathy B, Banerjee M. Haloperidol induces pharmacoepigenetic response by modulating miRNA expression, global DNA methylation and expression profiles of methylation maintenance genes and genes involved in neurotransmission in neuronal cells. PLoS One 2017; 12:e0184209. [PMID: 28886082 PMCID: PMC5590913 DOI: 10.1371/journal.pone.0184209] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 08/21/2017] [Indexed: 12/22/2022] Open
Abstract
Introduction Haloperidol has been extensively used in various psychiatric conditions. It has also been reported to induce severe side effects. We aimed to evaluate whether haloperidol can influence host methylome, and if so what are the possible mechanisms for it in neuronal cells. Impact on host methylome and miRNAs can have wide spread alterations in gene expression, which might possibly help in understanding how haloperidol may impact treatment response or induce side effects. Methods SK-N-SH, a neuroblasoma cell line was treated with haloperidol at 10μm concentration for 24 hours and global DNA methylation was evaluated. Methylation at global level is maintained by methylation maintenance machinery and certain miRNAs. Therefore, the expression of methylation maintenance genes and their putative miRNA expression profiles were assessed. These global methylation alterations could result in gene expression changes. Therefore genes expressions for neurotransmitter receptors, regulators, ion channels and transporters were determined. Subsequently, we were also keen to identify a strong candidate miRNA based on biological and in-silico approach which can reflect on the pharmacoepigenetic trait of haloperidol and can also target the altered neuroscience panel of genes used in the study. Results Haloperidol induced increase in global DNA methylation which was found to be associated with corresponding increase in expression of various epigenetic modifiers that include DNMT1, DNMT3A, DNMT3B and MBD2. The expression of miR-29b that is known to putatively regulate the global methylation by modulating the expression of epigenetic modifiers was observed to be down regulated by haloperidol. In addition to miR-29b, miR-22 was also found to be downregulated by haloperidol treatment. Both these miRNA are known to putatively target several genes associated with various epigenetic modifiers, pharmacogenes and neurotransmission. Interestingly some of these putative target genes involved in neurotransmission were observed to be upregulated while CHRM2 gene expression was down regulated. Conclusions Haloperidol can influence methylation traits thereby inducing a pharmacoepigenomic response, which seems to be regulated by DNMTs and their putative miRNA expression. Increased methylation seems to influence CHRM2 gene expression while microRNA could influence neurotransmission, pharmacogene expression and methylation events. Altered expression of various therapeutically relevant genes and miRNA expression, could account for their role in therapeutic response or side effects.
Collapse
Affiliation(s)
- Babu Swathy
- Human Molecular Genetics Laboratory, Rajiv Gandhi Centre for Biotechnology, Trivandrum, Kerala, India
| | - Moinak Banerjee
- Human Molecular Genetics Laboratory, Rajiv Gandhi Centre for Biotechnology, Trivandrum, Kerala, India
- * E-mail: ,
| |
Collapse
|
175
|
Ding L, Wang L, Guo F. microRNA‑188 acts as a tumour suppressor in glioma by directly targeting the IGF2BP2 gene. Mol Med Rep 2017; 16:7124-7130. [PMID: 28901413 DOI: 10.3892/mmr.2017.7433] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2016] [Accepted: 06/09/2017] [Indexed: 11/06/2022] Open
Abstract
Glioma is the most common and aggressive human brain tumour and accounts for ~35‑61% of intracranial tumours. Despite considerable advances in treatments for glioma, the prognosis for patients with this disease remains unsatisfactory. MicroRNAs (miRNAs of miRs) are small regulatory RNA molecules that have been identified as being involved in the initiation and progression of human cancers, and represent novel therapeutic targets for anticancer treatments. The dysregulation of miR‑188 has been reported in various kinds of human cancer. However, its expression pattern, biological roles and potential mechanism in glioma remain unknown. Expression levels of miR‑188 in glioma tissues and cell lines were detected through reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR). Cell Counting Kit-8 assays and migration and invasion assays were used to explore the effects of miR‑188 on the proliferation, migration and invasion of glioma cells, respectively. Bioinformatics analysis and luciferase reporter assays were performed to examine insulin‑like growth factor 2 mRNA-binding protein 2 (IGF2BP2) as a target gene of miR‑188. RT‑qPCR and Spearman's correlation analysis were then performed to measure IGF2BP2 mRNA expression in clinical glioma tissues and its correlation with miR‑188 expression. The regulatory effect of miR‑188 on IGF2BP2 expression was also investigated through RT‑qPCR and western blotting analysis. Finally, the biological roles of IGF2BP2 in glioma cells were assessed. miR‑188 levels were significantly reduced in glioma tissues and cell lines compared with adjacent normal tissues and normal human astrocytes, respectively. In addition, miR‑188 overexpression suppressed cell proliferation, migration and invasion of glioma. The present study identified IGF2BP2 as a direct target of miR‑188 in glioma, and IGF2BP2 under‑expression served tumour‑suppressive roles in glioma growth and metastasis. Thus, miR‑188 had a similar role in glioma by inhibiting the action of its downstream target, IGF2BP2. Therefore, miR‑188 may be a potential therapeutic target for the prevention and treatment of patients with glioma.
Collapse
Affiliation(s)
- Li Ding
- Department of Neurosurgery, Linyi People's Hospital, Linyi, Shandong 276000, P.R. China
| | - Ling Wang
- Department of Neurosurgery, Linyi People's Hospital, Linyi, Shandong 276000, P.R. China
| | - Feng Guo
- Department of Neurosurgery, Linyi People's Hospital, Linyi, Shandong 276000, P.R. China
| |
Collapse
|
176
|
Liu Y, Wei B, Zhang X, Xu D, Wang B, Yin G, Gu D, Li Y, Kong D. Identification of potential therapeutic target genes and miRNAs for primary myelofibrosis with microarray analysis. Exp Ther Med 2017; 14:2743-2750. [PMID: 28966666 PMCID: PMC5615205 DOI: 10.3892/etm.2017.4912] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 11/10/2016] [Indexed: 12/19/2022] Open
Abstract
The aim of the present study was to identify potential therapeutic target genes and miRNAs for primary myelofibrosis (PMF). The dataset GSE53482 was downloaded from the Gene Expression Omnibus database. The differentially expressed genes (DEGs) and differentially expressed miRNAs (DEMs) of peripheral blood (PB) cluster of differentiation (CD)34+ cells from PMF patients (PB-PMF group) and peripheral blood CD34+ cells from healthy individuals (PB-control group) were analyzed using the Linear Models for Microarray Data package in R. The Kyoto Encyclopedia of Genes and Genomes was used for pathway enrichment analysis. MiRNA-gene joint enrichment analysis was performed by ENViz and a miRNAs-gene regulatory network was constructed. A total of 1,182 DEGs (773 upregulated and 109 downregulated) and 48 DEMs (28 upregulated and 20 downregulated) were identified. According to the pathway enrichment analysis, a number of DEGs were enriched in metabolic pathways, including IDH1 and DNMT1. Other DEGs were enriched in the citrate cycle (tricarboxylic acid cycle; IDH1 and IDH3A) and certain DEGs were enriched in pyrimidine metabolism, including CARD8. For downregulated genes, certain DEGs were enriched in the spliceosome, including SF3B1 and CDC40. Furthermore, hsa-miR-127-3p, hsa-miR-140-3p and hsa-miR345 were associated with cell cycle-related biological processes, signal transduction and cell surface receptor signaling pathway. The DEM-DEG regulatory network indicated that hsa-miR-543 regulated 113 genes, including CARD8 and TIFA. The present study identified a number of genes, including IDH1, DNMT1, SF3B1 and CARD8, and miRNAs, including hsa-miR-127-3p and hsa-miR-140-3p, which may be therapeutic targets in the treatment of PMF.
Collapse
Affiliation(s)
- Yong Liu
- Department of Orthopaedics, Jilin Oilfield General Hospital, Songyuan, Jilin 131200, P.R. China
| | - Bo Wei
- Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Xuebing Zhang
- Department of Orthopaedics, Jilin Oilfield General Hospital, Songyuan, Jilin 131200, P.R. China
| | - Dehui Xu
- Department of Orthopaedics, Jilin Oilfield General Hospital, Songyuan, Jilin 131200, P.R. China
| | - Bo Wang
- Department of Orthopaedics, Jilin Oilfield General Hospital, Songyuan, Jilin 131200, P.R. China
| | - Guochao Yin
- Department of Orthopaedics, Jilin Oilfield General Hospital, Songyuan, Jilin 131200, P.R. China
| | - Dawer Gu
- Department of Orthopaedics, Jilin Oilfield General Hospital, Songyuan, Jilin 131200, P.R. China
| | - Yuxiang Li
- Department of Orthopaedics, Jilin Oilfield General Hospital, Songyuan, Jilin 131200, P.R. China
| | - Daliang Kong
- Department of Orthopaedics, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| |
Collapse
|
177
|
Zeng K, Wang Y, Yang N, Wang D, Li S, Ming J, Wang J, Yu X, Song Y, Zhou X, Deng B, Wu X, Huang L, Yang Y. Resveratrol Inhibits Diabetic-Induced Müller Cells Apoptosis through MicroRNA-29b/Specificity Protein 1 Pathway. Mol Neurobiol 2017; 54:4000-4014. [PMID: 27311771 DOI: 10.1007/s12035-016-9972-5] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 06/08/2016] [Indexed: 02/04/2023]
Abstract
The aim of this study was to evaluate the anti-apoptosis effects of resveratrol (RSV) on diabetic rats retinal Müller cells in vivo and in vitro and to further investigate the roles of microRNA-29b (miR-29b)/specificity protein 1 (SP1) in the anti-apoptosis mechanism of RSV. Retina was obtained from normal and diabetic rats with or without RSV (5 and 10 mg/kg/day) treatments at 1-7 months. TdT-mediated dUTP-biotin nick end labeling (TUNEL) and Annexin V/PI staining were used to detect apoptosis. Immunofluorescence was used to assess distribution of SP1 in retina. MiR-29b and SP1 messenger RNA (mRNA) expression was evaluated by quantitative real-time polymerase chain reaction (qRT-PCR). SP1, Bax, and bcl-2 protein expression was evaluated by western blotting. Caspase-3 activity was detected by assay kit. Our study showed that the TUNEL-positive cells were mainly localized in the inner nuclear layer (INL) of retina and RSV administration effectively suppressed streptozotocin (STZ)-induced apoptosis of retinal cells in INL in vivo (P < 0.001). Our study also showed that RSV administration effectively suppressed high glucose (HG)-induced retinal Müller cells' apoptosis in vitro (P < 0.001). Furthermore, our study revealed that the diabetes-induced downregulated expression of miR-29b and upregulated expression of SP1 could be rescued by RSV in vivo and in vitro (P < 0.05). The anti-apoptosis effect and downregulated SP1 expression effect of RSV was prevented by miR-29b inhibitor (P < 0.05). MiR-29b mimic increased the above-mentioned effects of RSV (P < 0.001). These findings indicate that RSV is a potential therapeutic option for diabetic retinopathy (DR) and that miR-29b/SP1 pathway play roles in the anti-apoptosis mechanism of RSV.
Collapse
Affiliation(s)
- Kaihong Zeng
- Department of Clinical Nutrition, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, 610072, People's Republic of China.
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, People's Republic of China.
| | - Yuan Wang
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, People's Republic of China
| | - Na Yang
- Institute of Laboratory Animals, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, 610072, People's Republic of China
| | - Duozi Wang
- Department of Neurology, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, 610072, People's Republic of China
| | - Suping Li
- Department of Neurology, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, 610072, People's Republic of China
| | - Jian Ming
- School of Food Science, Southwest University, Chongqing, 400715, People's Republic of China
| | - Jing Wang
- School of Food Science, Southwest University, Chongqing, 400715, People's Republic of China
| | - Xuemei Yu
- School of Food Science, Southwest University, Chongqing, 400715, People's Republic of China
| | - Yi Song
- School of Food Science, Southwest University, Chongqing, 400715, People's Republic of China
| | - Xue Zhou
- School of Food Science, Southwest University, Chongqing, 400715, People's Republic of China
| | - Bo Deng
- School of Food Science, Southwest University, Chongqing, 400715, People's Republic of China
| | - Xiaona Wu
- School of Food Science, Southwest University, Chongqing, 400715, People's Republic of China
| | - Lujiao Huang
- School of Food Science, Southwest University, Chongqing, 400715, People's Republic of China
| | - Yongtao Yang
- School of Food Science, Southwest University, Chongqing, 400715, People's Republic of China
| |
Collapse
|
178
|
Sun J, Tian X, Zhang J, Huang Y, Lin X, Chen L, Zhang S. Regulation of human glioma cell apoptosis and invasion by miR-152-3p through targeting DNMT1 and regulating NF2 : MiR-152-3p regulate glioma cell apoptosis and invasion. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2017; 36:100. [PMID: 28764788 PMCID: PMC5539621 DOI: 10.1186/s13046-017-0567-4] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2017] [Accepted: 07/11/2017] [Indexed: 01/09/2023]
Abstract
Background MiRNAs are involved in aberrant DNA methylation through regulation of DNA methyltransferases (DNMTs) in the pathogenesis and progression of glioblastomas (GBM). MiR-152-3p was down-expressed in human malignancies, and served as a tumor suppressor. Neurofibromatosis type 2 (NF2) was significantly decreased in GBM tissues with a high level of methylation. However, the link between miR-152-3p, DNMT1 and methylation of NF2 in GBM is not clearly established. This study was conducted to detect the mechanism between miR-152-3p, DNMT1 and NF2 in GBM. Methods The levels of DNMT1 and NF2 expression were studied by qRT-PCR, Western blot, immunofluorescence, and immumohistochemical staining. Methylation in the promoter region of NF2 was detected by methylation-specific PCR and bisulfate genomic sequencing PCR. Cell proliferation was examined by Cell-Counting Kit-8 and 5-ethynyl-2′-deoxyuridine assay, and cell invasion was evaluated by transwell assay. Flow cytomery and Hoechst staining were used to analyze cell apoptosis. A dual luciferase system was used to confirm the relationship between miR-152-3p and DNMT1. Results Methylation of NF2 and DNMT1 was markedly increased, and miR-152-3p was downregulated in GBM tissues and glioma cells. Both knockdown of DNMT1 and overexpression miR-152-3p showed that demethylation activated the expression of NF2. Furthermore, miR-152-3p directly targeted DNMT1. Both miR-152-3p overexpression and DNMT1 knockdown significantly induced cell apoptosis and inhibited invasive activity. This was also observed after NF2 overexpression. Conclusions These results indicated that miR-152-3p can inhibit glioma cell proliferation and invasion activities by decreasing DNMT1. The restoration of miR-152-3p may have therapeutic application in the treatment of GBM. Electronic supplementary material The online version of this article (doi:10.1186/s13046-017-0567-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jin Sun
- Department of Neurosurgery, Zhujiang Hospital Southern Medical University, National Key Clinical Specialty, Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Guangzhou, Guangdong, 510282, China.,Department of Neurosurgery, Zhongshan Hospital, Xiamen University, Xiamen, Fujian, 361004, China
| | - Xinhua Tian
- Department of Neurosurgery, Zhongshan Hospital, Xiamen University, Xiamen, Fujian, 361004, China
| | - Junqing Zhang
- Department of Neurosurgery, Zhongshan Hospital, Xiamen University, Xiamen, Fujian, 361004, China
| | - Yanlin Huang
- Department of Neurosurgery, Zhongshan Hospital, Xiamen University, Xiamen, Fujian, 361004, China
| | - Xiaoning Lin
- Department of Neurosurgery, Zhongshan Hospital, Xiamen University, Xiamen, Fujian, 361004, China
| | - Luyue Chen
- Department of Neurosurgery, Zhongshan Hospital, Xiamen University, Xiamen, Fujian, 361004, China
| | - Shizhong Zhang
- Department of Neurosurgery, Zhujiang Hospital Southern Medical University, National Key Clinical Specialty, Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Guangzhou, Guangdong, 510282, China.
| |
Collapse
|
179
|
Nabavinia MS, Gholoobi A, Charbgoo F, Nabavinia M, Ramezani M, Abnous K. Anti-MUC1 aptamer: A potential opportunity for cancer treatment. Med Res Rev 2017; 37:1518-1539. [PMID: 28759115 DOI: 10.1002/med.21462] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 06/19/2017] [Accepted: 06/30/2017] [Indexed: 01/10/2023]
Abstract
Mucin 1 (MUC1) is a protein usually found on the apical surface of most normal secretory epithelial cells. However, in most adenocarcinomas, MUC1 is overexpressed, so that it not only appears over the entire cell surface, but is also shed as MUC1 fragments into the blood stream. These phenomena pinpoint MUC1 as a potential target for the diagnosis and treatment of cancer; consequently, interest has increased in MUC1 as a molecular target for overcoming cancer therapy challenges. MUC1 currently ranks second among 75 antigen candidates for cancer vaccines, and different antibodies or aptamers against MUC1 protein are proving useful for tracing cancer cells in the emerging field of targeted delivery. The unique properties of MUC1 aptamers as novel targeting agents, and the revolutionary role that MUC1 now plays in cancer therapy, are the focus of this review. Recent advancements in MUC1-targeted cancer therapy are also assessed.
Collapse
Affiliation(s)
- Maryam Sadat Nabavinia
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Aida Gholoobi
- Department of Modern Sciences and Technologies, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fahimeh Charbgoo
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.,Pharmaceutical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Mohammad Ramezani
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.,Nanotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Khalil Abnous
- Pharmaceutical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
180
|
MicroRNAs and acute myeloid leukemia: therapeutic implications and emerging concepts. Blood 2017; 130:1290-1301. [PMID: 28751524 DOI: 10.1182/blood-2016-10-697698] [Citation(s) in RCA: 127] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 07/24/2017] [Indexed: 02/08/2023] Open
Abstract
Acute myeloid leukemia (AML) is a deadly hematologic malignancy characterized by the uncontrolled growth of immature myeloid cells. Over the past several decades, we have learned a tremendous amount regarding the genetic aberrations that govern disease development in AML. Among these are genes that encode noncoding RNAs, including the microRNA (miRNA) family. miRNAs are evolutionarily conserved small noncoding RNAs that display important physiological effects through their posttranscriptional regulation of messenger RNA targets. Over the past decade, studies have identified miRNAs as playing a role in nearly all aspects of AML disease development, including cellular proliferation, survival, and differentiation. These observations have led to the study of miRNAs as biomarkers of disease, and efforts to therapeutically manipulate miRNAs to improve disease outcome in AML are ongoing. Although much has been learned regarding the importance of miRNAs in AML disease initiation and progression, there are many unanswered questions and emerging facets of miRNA biology that add complexity to their roles in AML. Moving forward, answers to these questions will provide a greater level of understanding of miRNA biology and critical insights into the many translational applications for these small regulatory RNAs in AML.
Collapse
|
181
|
Shen N, Yan F, Pang J, Zhao N, Gangat N, Wu L, Bode AM, Al-Kali A, Litzow MR, Liu S. Inactivation of Receptor Tyrosine Kinases Reverts Aberrant DNA Methylation in Acute Myeloid Leukemia. Clin Cancer Res 2017; 23:6254-6266. [PMID: 28720666 DOI: 10.1158/1078-0432.ccr-17-0235] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 06/18/2017] [Accepted: 07/12/2017] [Indexed: 01/05/2023]
Abstract
Purpose: Receptor tyrosine kinases (RTKs) are frequently deregulated in leukemia, yet the biological consequences of this deregulation remain elusive. The mechanisms underlying aberrant methylation, a hallmark of leukemia, are not fully understood. Here we investigated the role of RTKs in methylation abnormalities and characterized the hypomethylating activities of RTK inhibitors.Experimental Design: Whether and how RTKs regulate expression of DNA methyltransferases (DNMTs), tumor suppressor genes (TSGs) as well as global and gene-specific DNA methylation were examined. The pharmacologic activities and mechanisms of actions of RTK inhibitors in vitro, ex vivo, in mice, and in nilotinib-treated leukemia patients were determined.Results: Upregulation of RTKs paralleled DNMT overexpression in leukemia cell lines and patient blasts. Knockdown of RTKs disrupted, whereas enforced expression increased DNMT expression and DNA methylation. Treatment with the RTK inhibitor, nilotinib, resulted in a reduction of Sp1-dependent DNMT1 expression, the diminution of global DNA methylation, and the upregulation of the p15INK4B gene through promoter hypomethylation in AML cell lines and patient blasts. This led to disruption of AML cell clonogenicity and promotion of cellular apoptosis without obvious changes in cell cycle. Importantly, nilotinib administration in mice and human patients with AML impaired expression of DNMTs followed by DNA hypomethylation, TSG re-expression, and leukemia regression.Conclusions: Our findings demonstrate RTKs as novel regulators of DNMT-dependent DNA methylation and define DNA methylation status in AML cells as a pharmacodynamic marker for their response to RTK-based therapy, providing new therapeutic avenues for RTK inhibitors in overcoming epigenetic abnormalities in leukemia. Clin Cancer Res; 23(20); 6254-66. ©2017 AACR.
Collapse
Affiliation(s)
- Na Shen
- The Hormel Institute, University of Minnesota, Austin, Minnesota
| | - Fei Yan
- The Hormel Institute, University of Minnesota, Austin, Minnesota
| | - Jiuxia Pang
- The Hormel Institute, University of Minnesota, Austin, Minnesota
| | - Na Zhao
- The Hormel Institute, University of Minnesota, Austin, Minnesota
| | - Naseema Gangat
- Division of Hematology, Mayo Clinic, Rochester, Minnesota
| | - Laichu Wu
- Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, Ohio
| | - Ann M Bode
- The Hormel Institute, University of Minnesota, Austin, Minnesota
| | - Aref Al-Kali
- Division of Hematology, Mayo Clinic, Rochester, Minnesota
| | - Mark R Litzow
- Division of Hematology, Mayo Clinic, Rochester, Minnesota.
| | - Shujun Liu
- The Hormel Institute, University of Minnesota, Austin, Minnesota.
| |
Collapse
|
182
|
Abstract
The development of intrinsic or acquired resistance to chemotherapeutic agents used in the treatment of various human cancers is a major obstacle for the successful abolishment of cancer. The accumulated efforts in the understanding the exact mechanisms of development of multidrug resistance (MDR) have led to the introduction of several unique and common mechanisms. Recent studies demonstrate the regulatory role of small noncoding RNA or miRNA in the several parts of cancer biology. Practically all aspects of cell physiology under normal and disease conditions are reported to be controlled by miRNAs. In this review, we discuss how the miRNA profile is changed upon MDR development and the pivotal regulatory role played by miRNAs in overcoming resistance to chemotherapeutic agents. It is hoped that further studies will support the use of these differentially expressed miRNAs as prognostic and predictive markers, as well as novel therapeutic targets to overcome resistance in ovarian cancer.
Collapse
Affiliation(s)
- Aynaz Mihanfar
- Women's Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Faculty of Medicine, Department of Biochemistry, Urmia University of Medical Sciences, Urmia, Iran
| | - Amir Fattahi
- Faculty of Advanced Medical Sciences, Department of Reproductive Biology, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Obstetrics and Gynecology, Erlangen University Hospital, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Hamid Reza Nejabati
- Women's Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
183
|
Martiáñez Canales T, de Leeuw DC, Vermue E, Ossenkoppele GJ, Smit L. Specific Depletion of Leukemic Stem Cells: Can MicroRNAs Make the Difference? Cancers (Basel) 2017; 9:cancers9070074. [PMID: 28665351 PMCID: PMC5532610 DOI: 10.3390/cancers9070074] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 06/15/2017] [Accepted: 06/20/2017] [Indexed: 01/22/2023] Open
Abstract
For over 40 years the standard treatment for acute myeloid leukemia (AML) patients has been a combination of chemotherapy consisting of cytarabine and an anthracycline such as daunorubicin. This standard treatment results in complete remission (CR) in the majority of AML patients. However, despite these high CR rates, only 30–40% (<60 years) and 10–20% (>60 years) of patients survive five years after diagnosis. The main cause of this treatment failure is insufficient eradication of a subpopulation of chemotherapy resistant leukemic cells with stem cell-like properties, often referred to as “leukemic stem cells” (LSCs). LSCs co-exist in the bone marrow of the AML patient with residual healthy hematopoietic stem cells (HSCs), which are needed to reconstitute the blood after therapy. To prevent relapse, development of additional therapies targeting LSCs, while sparing HSCs, is essential. As LSCs are rare, heterogeneous and dynamic, these cells are extremely difficult to target by single gene therapies. Modulation of miRNAs and consequently the regulation of hundreds of their targets may be the key to successful elimination of resistant LSCs, either by inducing apoptosis or by sensitizing them for chemotherapy. To address the need for specific targeting of LSCs, miRNA expression patterns in highly enriched HSCs, LSCs, and leukemic progenitors, all derived from the same patients’ bone marrow, were determined and differentially expressed miRNAs between LSCs and HSCs and between LSCs and leukemic progenitors were identified. Several of these miRNAs are specifically expressed in LSCs and/or HSCs and associated with AML prognosis and treatment outcome. In this review, we will focus on the expression and function of miRNAs expressed in normal and leukemic stem cells that are residing within the AML bone marrow. Moreover, we will review their possible prospective as specific targets for anti-LSC therapy.
Collapse
Affiliation(s)
- Tania Martiáñez Canales
- Department of Hematology, VU University Medical Center, Cancer Center Amsterdam, Boelelaan 1117, 1081 HV Amsterdam, The Netherlands.
| | - David C de Leeuw
- Department of Hematology, VU University Medical Center, Cancer Center Amsterdam, Boelelaan 1117, 1081 HV Amsterdam, The Netherlands.
| | - Eline Vermue
- Department of Hematology, VU University Medical Center, Cancer Center Amsterdam, Boelelaan 1117, 1081 HV Amsterdam, The Netherlands.
| | - Gert J Ossenkoppele
- Department of Hematology, VU University Medical Center, Cancer Center Amsterdam, Boelelaan 1117, 1081 HV Amsterdam, The Netherlands.
| | - Linda Smit
- Department of Hematology, VU University Medical Center, Cancer Center Amsterdam, Boelelaan 1117, 1081 HV Amsterdam, The Netherlands.
| |
Collapse
|
184
|
Sheervalilou R, Shirvaliloo S, Fekri Aval S, Khamaneh AM, Sharifi A, Ansarin K, Zarghami N. A new insight on reciprocal relationship between microRNA expression and epigenetic modifications in human lung cancer. Tumour Biol 2017; 39:1010428317695032. [PMID: 28468581 DOI: 10.1177/1010428317695032] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Lung cancer stands among the leading causes of cancer-related death in the world. Although the molecular network implicated in lung cancer development is extensively revealed, the mortality rate is only slightly improved. MicroRNAs are small, endogenous single-stranded evolutionary conserved non-coding RNAs which involve in a wide variety of biological processes including cell growth, proliferation, metabolism, and differentiation. MicroRNAs, as novel biomarkers, have multiple functions in normal lung tissue development, and aberrant expression profiles of certain microRNAs could induce lung tumorigenesis. Similar to that of protein-coding genes, microRNA expression and function are regulated by multiple factors as well as the epigenetic network including DNA methylation and histone modification mechanisms. Furthermore, microRNAs can themselves regulate key enzymes which drive epigenetic modifications and have a pivotal effect on the cell biology. In this review, we will look into the regulatory loop linkage between microRNA expression and epigenetic modifications, and then, we will discuss the effects of epigenetics on the miRNome, as well as the role of epi-microRNAs in controlling the epigenome in human lung cancer. Better knowledge of reciprocal connection between microRNAs and epigenome will help to develop novel microRNA-orientated diagnostic, prognostic and therapeutic strategies related to human lung cancer in future.
Collapse
Affiliation(s)
- Roghayeh Sheervalilou
- 1 Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.,2 Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,3 Students' Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sakine Shirvaliloo
- 4 Department of Medical Physics, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Sedigheh Fekri Aval
- 2 Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,3 Students' Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.,5 Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Mahdi Khamaneh
- 1 Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.,2 Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Akbar Sharifi
- 2 Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Khalil Ansarin
- 2 Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nosratollah Zarghami
- 2 Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,5 Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
185
|
Pang J, Shen N, Yan F, Zhao N, Dou L, Wu LC, Seiler CL, Yu L, Yang K, Bachanova V, Weaver E, Tretyakova NY, Liu S. Thymoquinone exerts potent growth-suppressive activity on leukemia through DNA hypermethylation reversal in leukemia cells. Oncotarget 2017; 8:34453-34467. [PMID: 28415607 PMCID: PMC5470982 DOI: 10.18632/oncotarget.16431] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 03/14/2017] [Indexed: 01/27/2023] Open
Abstract
Thymoquinone (TQ), a bioactive constituent of the volatile oil of Monarda fistulosa and Nigella sativa, possesses cancer-specific growth inhibitory effects, but the underlying molecular mechanisms remain largely elusive. We propose that TQ curbs cancer cell growth through dysfunction of DNA methyltransferase 1 (DNMT1). Molecular docking analysis revealed that TQ might interact with the catalytic pocket of DNMT1 and compete with co-factor SAM/SAH for DNMT1 inhibition. In vitro inhibitory assays showed that TQ decreases DNMT1 methylation activity in a dose-dependent manner with an apparent IC50 of 30 nM. Further, exposure of leukemia cell lines and patient primary cells to TQ resulted in DNMT1 downregulation, mechanistically, through dissociation of Sp1/NFkB complex from DNMT1 promoter. This led to a reduction of DNA methylation, a decrease of colony formation and an increase of cell apoptosis via the activation of caspases. In addition, we developed and validated a sensitive and specific LC-MS/MS method and successfully detected a dynamic change of TQ in mouse plasma after administration of TQ through the tail vein, and determined a tolerable dose of TQ to be 15 mg/kg in mouse. TQ administration into leukemia-bearing mice induced leukemia regression, as indicated by the reversed splenomegaly and the inhibited leukemia cell growth in lungs and livers. Our study for the first time demonstrates that DNMT1-dependent DNA methylation mediates the anticancer actions of TQ, opening a window to develop TQ as a novel DNA hypomethylating agent for leukemia therapy.
Collapse
Affiliation(s)
- Jiuxia Pang
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Na Shen
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Fei Yan
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Na Zhao
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Liping Dou
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
- Department of Hematology, Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing 100853, China
| | - Lai-Chu Wu
- Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH 43021, USA
| | - Christopher L. Seiler
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Li Yu
- Department of Hematology, Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing 100853, China
| | - Ke Yang
- Chongqing Engineering Research Center of Stem Cell Therapy, The Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Veronika Bachanova
- Division of Hematology, Oncology and Transplantation, Minneapolis, MN 55455, USA
| | - Eric Weaver
- Prairie Pharms LLC, Nora Springs, IA 50458, USA
| | - Natalia Y. Tretyakova
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Shujun Liu
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| |
Collapse
|
186
|
A Macro View of MicroRNAs: The Discovery of MicroRNAs and Their Role in Hematopoiesis and Hematologic Disease. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2017; 334:99-175. [PMID: 28838543 DOI: 10.1016/bs.ircmb.2017.03.007] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
MicroRNAs (MiRNAs) are a class of endogenously encoded ~22 nucleotide, noncoding, single-stranded RNAs that contribute to development, body planning, stem cell differentiation, and tissue identity through posttranscriptional regulation and degradation of transcripts. Given their importance, it is predictable that dysregulation of MiRNAs, which target a wide variety of transcripts, can result in malignant transformation. In this review, we explore the discovery of MiRNAs, their mechanism of action, and the tools that aid in their discovery and study. Strikingly, many of the studies that have expanded our understanding of the contributions of MiRNAs to normal physiology and in the development of diseases have come from studies in the hematopoietic system and hematologic malignancies, with some of the earliest identified functions for mammalian MiRNAs coming from observations made in leukemias. So, with a special focus on the hematologic system, we will discuss how MiRNAs contribute to differentiation of stem cells and how dysregulation of MiRNAs contributes to the development of malignancy, by providing examples of specific MiRNAs that function as oncogenes or tumor suppressors, as well as of defects in MiRNA processing. Finally, we will discuss the promise of MiRNA-based therapeutics and challenges for the future study of disease-causing MiRNAs.
Collapse
|
187
|
Bertero T, Rezzonico R, Pottier N, Mari B. Impact of MicroRNAs in the Cellular Response to Hypoxia. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2017; 333:91-158. [PMID: 28729029 DOI: 10.1016/bs.ircmb.2017.03.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
In mammalian cells, hypoxia, or inadequate oxygen availability, regulates the expression of a specific set of MicroRNAs (MiRNAs), termed "hypoxamiRs." Over the past 10 years, the appreciation of the importance of hypoxamiRs in regulating the cellular adaptation to hypoxia has grown dramatically. At the cellular level, each hypoxamiR, including the master hypoxamiR MiR-210, can simultaneously regulate expression of multiple target genes in order to fine-tune the adaptive response of cells to hypoxia. This review addresses the complex molecular regulation of MiRNAs in both physiological and pathological conditions of low oxygen adaptation and the multiple functions of hypoxamiRs in various hypoxia-associated biological processes, including apoptosis, survival, proliferation, angiogenesis, inflammation, and metabolism. From a clinical perspective, we also discuss the potential use of hypoxamiRs as new biomarkers and/or therapeutic targets in cancer and aging-associated diseases including cardiovascular and fibroproliferative disorders.
Collapse
Affiliation(s)
- Thomas Bertero
- Université Côte d'Azur, CNRS, INSERM, IRCAN, FHU-OncoAge, Nice, France
| | - Roger Rezzonico
- Université Côte d'Azur, CNRS, IPMC, FHU-OncoAge, Sophia-Antipolis, France
| | | | - Bernard Mari
- Université Côte d'Azur, CNRS, IPMC, FHU-OncoAge, Sophia-Antipolis, France.
| |
Collapse
|
188
|
circRNA_100290 plays a role in oral cancer by functioning as a sponge of the miR-29 family. Oncogene 2017; 36:4551-4561. [PMID: 28368401 PMCID: PMC5558096 DOI: 10.1038/onc.2017.89] [Citation(s) in RCA: 329] [Impact Index Per Article: 41.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 02/24/2017] [Accepted: 02/26/2017] [Indexed: 01/17/2023]
Abstract
Circular RNAs (circRNAs) represent a class of non-coding RNAs that are widely expressed in mammals. However, it is largely unknown about the function of human circRNAs and the roles of circRNAs in human oral squamous cell carcinomas (OSCC). Here we performed a comprehensive study of circRNAs in human OSCC using circRNA and mRNA microarrays, and identified many circRNAs that are differentially expressed between OSCC tissue and paired non-cancerous matched tissue. We further found a circRNA termed circRNA_100290 that served as a critical regulator in OSCC development. We discovered that circRNA_100290 was upregulated and co-expressed with CDK6 in OSCC tissue. Knockdown of circRNA_100290 decreased expression of CDK6 and inhibited proliferation of OSCC cell lines in vitro and in vivo. Via luciferase reporter assays, circRNA_100290 was observed to directly bind to miR-29 family members. Further EGFP/RFP reporter assays showed that CDK6 was the direct target of miR-29b. Taken together, we conclude that circRNA_100290 may function as a competing endogenous RNA to regulate CDK6 expression through sponging up miR-29b family members. Taken together, it indicates that circRNAs may exert regulatory functions in OSCC and may be a potential target for OSCC therapy.
Collapse
|
189
|
Hedrich CM, Mäbert K, Rauen T, Tsokos GC. DNA methylation in systemic lupus erythematosus. Epigenomics 2017; 9:505-525. [PMID: 27885845 PMCID: PMC6040049 DOI: 10.2217/epi-2016-0096] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 10/12/2016] [Indexed: 12/18/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a systemic autoimmune disease facilitated by aberrant immune responses directed against cells and tissues, resulting in inflammation and organ damage. In the majority of patients, genetic predisposition is accompanied by additional factors conferring disease expression. While the exact molecular mechanisms remain elusive, epigenetic alterations in immune cells have been demonstrated to play a key role in disease pathogenesis through the dysregulation of gene expression. Since epigenetic marks are dynamic, allowing cells and tissues to differentiate and adjust, they can be influenced by environmental factors and also be targeted in therapeutic interventions. Here, we summarize reports on DNA methylation patterns in SLE, underlying molecular defects and their effect on immune cell function. We discuss the potential of DNA methylation as biomarker or therapeutic target in SLE.
Collapse
Affiliation(s)
- Christian M Hedrich
- Pediatric Rheumatology & Immunology, Klinik und Poliklinik für Kinder- und Jugendmedizin, Universitätsklinikum Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Katrin Mäbert
- Pediatric Rheumatology & Immunology, Klinik und Poliklinik für Kinder- und Jugendmedizin, Universitätsklinikum Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Thomas Rauen
- Department of Nephrology & Clinical Immunology, RWTH University Hospital, Aachen, Germany
| | - George C Tsokos
- Division of Rheumatology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
190
|
Özbayer C, Üstüner D, Ak GA, Saydam F, Metintaş M, Değirmenci İ. Akciğer Kanserli Hastalarda Plazma DNA Metiltransferaz ve Metil-CpG’ye Bağlanan Protein Seviyelerinin Değerlendirilmesi. DICLE MEDICAL JOURNAL 2017. [DOI: 10.5798/dicletip.298604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
191
|
Liu L, Bi N, Wu L, Ding X, Men Y, Zhou W, Li L, Zhang W, Shi S, Song Y, Wang L. MicroRNA-29c functions as a tumor suppressor by targeting VEGFA in lung adenocarcinoma. Mol Cancer 2017; 16:50. [PMID: 28241836 PMCID: PMC5330136 DOI: 10.1186/s12943-017-0620-0] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 02/22/2017] [Indexed: 12/21/2022] Open
Abstract
Background Lung adenocarcinoma (LAD) is considered to be a highly aggressive disease with heterogeneous prognosis and the molecular mechanisms underlying tumor progression remain elusive. Growing evidence demonstrates that the dysregulation of microRNAs (miRNAs) plays an important role in various tumor processes. The aim of this study is to discover prognostic miRNA and investigate its role involved in progression of LAD. Methods Prognosis related miRNA was detected by miRNA microarray using formalin-fixed paraffin-embedded (FFPE) specimens from 87 patients with IIIA-N2 LAD. The cell proliferation was evaluated by Cell Titer 96 AQueous One Solution Cell Proliferation Assay (MTS), and the migration/invasion was evaluated by transwell assay. The bioinformatics methods and luciferase reporter assay were applied to detect the relationship between miRNA and its target. The mRNA and protein levels of miRNA target were determined by quantitative real time polymerase chain reaction (qRT-PCR) analysis, western blot and enzyme-linked immunosorbent assay (ELISA). Changes of angiogenesis induced by miRNA was evaluated by human umbilical vein endothelial cell (HUVEC) tube formation assay. Immunohistochemistry (IHC) analysis was performed in FFPE specimens of patients to evaluate the correlation between miR-29c with microvessel density (MVD) and vascular endothelial growth factor A (VEGFA) expression. Results MiR-29c expression downregulation was significantly associated with unfavorable prognosis in IIIA-N2 LAD. MiR-29c inhibited cell proliferation, migration and invasion in cell lines. Integrated analysis revealed that VEGFA was a direct target of miR-29c. MiR-29c reduced the capability of tumor cells to promote HUVEC tube formation. The compromised cell proliferation, migration/invasion and angiogenesis induced by miR-29c mimic transfection were reversed by transfection of VEGFA expression plasmid. Furthermore, the correlation of miR-29c with MVD and VEGFA was confirmed in patients’ samples. Conclusions MiR-29c acts as a tumor suppressor by targeting VEGFA and may represent a promising prognostic biomarker as well as a potential therapeutic target for LAD.
Collapse
Affiliation(s)
- Lipin Liu
- Department of Radiation Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Nan Bi
- Department of Radiation Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Lihong Wu
- Department of Radiation Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xiao Ding
- Cancer Center, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021, China
| | - Yu Men
- Department of Radiation Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Wei Zhou
- The State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Lin Li
- Department of Pathology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Weimin Zhang
- The State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Susheng Shi
- Department of Pathology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yongmei Song
- The State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Luhua Wang
- Department of Radiation Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
192
|
Lu H, Lei X, Liu J, Klaassen C. Regulation of hepatic microRNA expression by hepatocyte nuclear factor 4 alpha. World J Hepatol 2017; 9:191-208. [PMID: 28217257 PMCID: PMC5295159 DOI: 10.4254/wjh.v9.i4.191] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Revised: 10/02/2016] [Accepted: 12/02/2016] [Indexed: 02/06/2023] Open
Abstract
AIM To uncover the role of hepatocyte nuclear factor 4 alpha (HNF4α) in regulating hepatic expression of microRNAs.
METHODS Microarray and real-time PCR were used to determine hepatic expression of microRNAs in young-adult mice lacking Hnf4α expression in liver (Hnf4α-LivKO). Integrative genomics viewer software was used to analyze the public chromatin immunoprecipitation-sequencing datasets for DNA-binding of HNF4α, RNA polymerase-II, and histone modifications to loci of microRNAs in mouse liver and human hepatoma cells. Dual-luciferase reporter assay was conducted to determine effects of HNF4α on the promoters of mouse and human microRNAs as well as effects of microRNAs on the untranslated regions (3’UTR) of two genes in human hepatoma cells.
RESULTS Microarray data indicated that most microRNAs remained unaltered by Hnf4α deficiency in Hnf4α-LivKO mice. However, certain liver-predominant microRNAs were down-regulated similarly in young-adult male and female Hnf4α-LivKO mice. The down-regulation of miR-101, miR-192, miR-193a, miR-194, miR-215, miR-802, and miR-122 as well as induction of miR-34 and miR-29 in male Hnf4α-LivKO mice were confirmed by real-time PCR. Analysis of public chromatin immunoprecipitation-sequencing data indicates that HNF4α directly binds to the promoters of miR-101, miR-122, miR-194-2/miR-192 and miR-193, which is associated with histone marks of active transcription. Luciferase reporter assay showed that HNF4α markedly activated the promoters of mouse and human miR-101b/miR-101-2 and the miR-194/miR-192 cluster. Additionally, miR-192 and miR-194 significantly decreased activities of luciferase reporters for the 3’UTR of histone H3F3 and chromodomain helicase DNA binding protein 1 (CHD1), respectively, suggesting that miR-192 and miR-194 might be important in chromosome remodeling through directly targeting H3F3 and CHD1.
CONCLUSION HNF4α is essential for hepatic basal expression of a group of liver-enriched microRNAs, including miR-101, miR-192, miR-193a, miR-194 and miR-802, through which HNF4α may play a major role in the post-transcriptional regulation of gene expression and maintenance of the epigenome in liver.
Collapse
|
193
|
Abstract
DNA methylation and specifically the DNA methyltransferase enzyme DNMT3A are involved in the pathogenesis of a variety of hematological diseases and in regulating the function of immune cells. Although altered DNA methylation patterns and mutations in DNMT3A correlate with mast cell proliferative disorders in humans, the role of DNA methylation in mast cell biology is not understood. By using mast cells lacking Dnmt3a, we found that this enzyme is involved in restraining mast cell responses to acute and chronic stimuli, both in vitro and in vivo. The exacerbated mast cell responses observed in the absence of Dnmt3a were recapitulated or enhanced by treatment with the demethylating agent 5-aza-2'-deoxycytidine as well as by down-modulation of Dnmt1 expression, further supporting the role of DNA methylation in regulating mast cell activation. Mechanistically, these effects were in part mediated by the dysregulated expression of the scaffold protein IQGAP2, which is characterized by the ability to regulate a wide variety of biological processes. Altogether, our data demonstrate that DNMT3A and DNA methylation are key modulators of mast cell responsiveness to acute and chronic stimulation.
Collapse
|
194
|
To KKW, Leung WW, Ng SSM. A novel miR-203-DNMT3b-ABCG2 regulatory pathway predisposing colorectal cancer development. Mol Carcinog 2017; 56:464-477. [PMID: 27253631 DOI: 10.1002/mc.22508] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2016] [Revised: 05/02/2016] [Accepted: 05/31/2016] [Indexed: 12/31/2022]
Abstract
Colorectal cancer (CRC) is a major cause of mortality and morbidity worldwide. The majority of studies to date focused on genetic mutations and epigenetic changes that drive the CRC carcinogenesis process. Xenobiotic transporters play an important role in safeguarding our body from external toxic substances. These transporters lining the gastrointestinal tract protect us from dietary carcinogens. This study aimed to investigate the downregulation of an efflux transporter ABCG2 in CRC versus normal colon mucosa, so as to shed light on its relevance to CRC initiation and progression. We found that ABCG2 expression is at least 50-fold lower in adenomatous polyps and colon carcinoma specimens obtained from CRC patients than in their matched pair of adjacent normal colon mucosa. The underlying mechanism(s) for ABCG2 under-expression in CRC is currently not known. To this end, aberrant promoter methylation of ABCG2 has been reported to cause its repression in a few cancer types including renal carcinoma and multiple myeloma. In this study, miR-203 was found to be downregulated in all polyps and CRC specimens, relative to adjacent normal colon mucosa. We demonstrated that the de novo DNA methyltransferase DNMT3b is a direct target of miR-203. Importantly, by relieving the repression on DNMT3b, the lower expression of miR-203 in CRC caused ABCG2 promoter methylation and remarkable lower ABCG2 expression in colon cancer cell lines and the patient CRC specimens. The restoration of ABCG2 function via modulating this new microRNA-methylation mechanism in precancerous cells may represent an attractive strategy to delay the carcinogenesis process. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Kenneth K W To
- Faculty of Medicine, School of Pharmacy, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Wing W Leung
- Faculty of Medicine, Department of Surgery, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Simon S M Ng
- Faculty of Medicine, Department of Surgery, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
195
|
Targeting MicroRNAs in Cancer Gene Therapy. Genes (Basel) 2017; 8:genes8010021. [PMID: 28075356 PMCID: PMC5295016 DOI: 10.3390/genes8010021] [Citation(s) in RCA: 127] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 12/28/2016] [Accepted: 12/30/2016] [Indexed: 12/15/2022] Open
Abstract
MicroRNAs (miRNAs) are a kind of conserved small non-coding RNAs that participate in regulating gene expression by targeting multiple molecules. Early studies have shown that the expression of miRNAs changes significantly in different tumor tissues and cancer cell lines. It is well acknowledged that such variation is involved in almost all biological processes, including cell proliferation, mobility, survival and differentiation. Increasing experimental data indicate that miRNA dysregulation is a biomarker of several pathological conditions including cancer, and that miRNA can exert a causal role, as oncogenes or tumor suppressor genes, in different steps of the tumorigenic process. Anticancer therapies based on miRNAs are currently being developed with a goal to improve outcomes of cancer treatment. In our present study, we review the function of miRNAs in tumorigenesis and development, and discuss the latest clinical applications and strategies of therapy targeting miRNAs in cancer.
Collapse
|
196
|
Yu F, Chen B, Dong P, Zheng J. HOTAIR Epigenetically Modulates PTEN Expression via MicroRNA-29b: A Novel Mechanism in Regulation of Liver Fibrosis. Mol Ther 2017; 25:205-217. [PMID: 28129115 PMCID: PMC5363197 DOI: 10.1016/j.ymthe.2016.10.015] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 10/27/2016] [Accepted: 10/31/2016] [Indexed: 02/06/2023] Open
Abstract
Homeobox transcript antisense RNA (HOTAIR), as a long intergenic non-coding RNA (lincRNA), is upregulated in various cancers and involved in diverse cellular functions. However, its role in liver fibrosis is unclear. In this study, HOTAIR expression was upregulated in hepatic stellate cells (HSCs) in vivo and in vitro during liver fibrosis. HOTAIR knockdown suppressed HSC activation including α-smooth muscle actin (α-SMA) and typeIcollagen in vitro and in vivo. Both HSC proliferation and cell cycle were inhibited by HOTAIR knockdown. Notably, inhibition of HOTAIR led to an increase in PTEN, associated with the loss of DNA methylation. miR-29b-mediated control of PTEN methylation was involved in the effects of HOTAIR knockdown. HOTAIR was confirmed a target of miR-29b and lack of the miR-29b binding site in HOTAIR prevented the suppression of miR-29b, suggesting HOTAIR contributes to PTEN expression downregulation via sponging miR-29b. Interestingly, increased HOTAIR was also observed in hepatocytes during liver fibrosis. Loss of HOTAIR additionally led to the increase in PTEN and the reduction in typeIcollagen in hepatocytes. Collectively, we demonstrate that HOTAIR downregulates miR-29b expression and attenuates its control on epigenetic regulation, leading to enhanced PTEN methylation, which contributes to the progression of liver fibrosis.
Collapse
Affiliation(s)
- Fujun Yu
- Department of Infectious Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China; Department of Gastroenterology, Songjiang Hospital Affiliated Shanghai First People's Hospital, Shanghai Jiao Tong University, Shanghai 201600, China; Department of Gastroenterology, Shanghai Songjiang Hospital Affiliated to Nanjing Medical University, Nanjing 210029, China
| | - Bicheng Chen
- Key Laboratory of Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Peihong Dong
- Department of Infectious Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China.
| | - Jianjian Zheng
- Key Laboratory of Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China.
| |
Collapse
|
197
|
Soci UPR, Melo SFS, Gomes JLP, Silveira AC, Nóbrega C, de Oliveira EM. Exercise Training and Epigenetic Regulation: Multilevel Modification and Regulation of Gene Expression. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1000:281-322. [PMID: 29098627 DOI: 10.1007/978-981-10-4304-8_16] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Exercise training elicits acute and adaptive long term changes in human physiology that mediate the improvement of performance and health state. The responses are integrative and orchestrated by several mechanisms, as gene expression. Gene expression is essential to construct the adaptation of the biological system to exercise training, since there are molecular processes mediating oxidative and non-oxidative metabolism, angiogenesis, cardiac and skeletal myofiber hypertrophy, and other processes that leads to a greater physiological status. Epigenetic is the field that studies about gene expression changes heritable by meiosis and mitosis, by changes in chromatin and DNA conformation, but not in DNA sequence, that studies the regulation on gene expression that is independent of genotype. The field approaches mechanisms of DNA and chromatin conformational changes that inhibit or increase gene expression and determine tissue specific pattern. The three major studied epigenetic mechanisms are DNA methylation, Histone modification, and regulation of noncoding RNA-associated genes. This review elucidates these mechanisms, focusing on the relationship between them and their relationship with exercise training, physical performance and the enhancement of health status. On this chapter, we clarified the relationship of epigenetic modulations and their intimal relationship with acute and chronic effect of exercise training, concentrating our effort on skeletal muscle, heart and vascular responses, that are the most responsive systems against to exercise training and play crucial role on physical performance and improvement of health state.
Collapse
Affiliation(s)
| | | | | | | | - Clara Nóbrega
- School of Physical Education and Sport, University of Sao Paulo, Sao Paulo, SP, Brazil
| | | |
Collapse
|
198
|
Abstract
MicroRNAs (miRNAs) are small noncoding RNAs that regulate gene expression mainly at the posttranscriptional level. Similar to protein-coding genes, their expression is also controlled by genetic and epigenetic mechanisms. Disruption of these control processes leads to abnormal expression of miRNAs in cancer. In this chapter, we discuss the supportive links between miRNAs and epigenetics in the context of carcinogenesis. miRNAs can be epigenetically regulated by DNA methylation and/or specific histone modifications. However, they can themselves (epi-miRNAs) repress key enzymes that drive epigenetic remodeling and also bind to complementary sequences in gene promoters, recruiting specific protein complexes that modulate chromatin structure and gene expression. All these issues affect the transcriptional landscape of cells. Most important, in the cancer clinical scenario, knowledge about miRNAs epigenetic dysregulation can not only be beneficial as a prognostic biomarker, but can also help in the design of new therapeutic approaches.
Collapse
Affiliation(s)
- Catia Moutinho
- Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Catalonia, Spain
| | - Manel Esteller
- Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Catalonia, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Catalonia, Spain; School of Medicine and Health Sciences, University of Barcelona (UB), Catalonia, Spain.
| |
Collapse
|
199
|
Zebisch A, Hatzl S, Pichler M, Wölfler A, Sill H. Therapeutic Resistance in Acute Myeloid Leukemia: The Role of Non-Coding RNAs. Int J Mol Sci 2016; 17:2080. [PMID: 27973410 PMCID: PMC5187880 DOI: 10.3390/ijms17122080] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 12/01/2016] [Accepted: 12/05/2016] [Indexed: 01/12/2023] Open
Abstract
Acute myeloid leukemia (AML) is caused by malignant transformation of hematopoietic stem or progenitor cells and displays the most frequent acute leukemia in adults. Although some patients can be cured with high dose chemotherapy and allogeneic hematopoietic stem cell transplantation, the majority still succumbs to chemoresistant disease. Micro-RNAs (miRNAs) and long non-coding RNAs (lncRNAs) are non-coding RNA fragments and act as key players in the regulation of both physiologic and pathologic gene expression profiles. Aberrant expression of various non-coding RNAs proved to be of seminal importance in the pathogenesis of AML, as well in the development of resistance to chemotherapy. In this review, we discuss the role of miRNAs and lncRNAs with respect to sensitivity and resistance to treatment regimens currently used in AML and provide an outlook on potential therapeutic targets emerging thereof.
Collapse
Affiliation(s)
- Armin Zebisch
- Division of Hematology, Medical University of Graz, 8036 Graz, Austria.
| | - Stefan Hatzl
- Division of Hematology, Medical University of Graz, 8036 Graz, Austria.
| | - Martin Pichler
- Division of Oncology, Medical University of Graz, 8036 Graz, Austria.
| | - Albert Wölfler
- Division of Hematology, Medical University of Graz, 8036 Graz, Austria.
| | - Heinz Sill
- Division of Hematology, Medical University of Graz, 8036 Graz, Austria.
| |
Collapse
|
200
|
Twist1 induces the expression of microRNA-29 to suppress SIN3A in head and neck cancer cells. JOURNAL OF CANCER RESEARCH AND PRACTICE 2016. [DOI: 10.1016/j.jcrpr.2016.03.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|