1
|
Maries L, Moatar AI, Chis AR, Marian C, Luca CT, Sirbu IO, Gaiță D. Plasma hsa- miR-22-3p Might Serve as an Early Predictor of Ventricular Function Recovery after ST-Elevation Acute Myocardial Infarction. Biomedicines 2023; 11:2289. [PMID: 37626785 PMCID: PMC10452683 DOI: 10.3390/biomedicines11082289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/07/2023] [Accepted: 08/10/2023] [Indexed: 08/27/2023] Open
Abstract
Left ventricle remodeling (LVR) after acute myocardial infarction (aMI) leads to impairment of both systolic and diastolic function, a major contributor to heart failure (HF). Despite extensive research, predicting post-aMI LVR and HF is still a challenge. Several circulant microRNAs have been proposed as LVR predictors; however, their clinical value is controversial. Here, we used real-time quantitative polymerase chain reaction (qRT-PCR) to quantify hsa-miR-22-3p (miR-22) plasma levels on the first day of hospital admission of ST-elevation aMI (STEMI) patients. We analyzed miR-22 correlation to the patients' clinical and paraclinical variables and evaluated its ability to discriminate between post-aMI LVR and non-LVR. We show that miR-22 is an excellent aMI discriminator and can distinguish between LVR and non-LVR patients. The discriminative performance of miR-22 significantly improves the predictive power of a multiple logistic regression model based on four continuous variables (baseline ejection fraction and end-diastolic volume, CK-MB, and troponin). Furthermore, we found that diabetes mellitus, hematocrit level, and the number of erythrocytes significantly influence its levels. These data suggest that miR-22 might be used as a predictor of ventricular function recovery in STEMI patients.
Collapse
Affiliation(s)
- Liana Maries
- Biochemistry Department, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (L.M.); (A.I.M.); (A.R.C.); (C.M.)
- Doctoral School, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Alexandra Ioana Moatar
- Biochemistry Department, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (L.M.); (A.I.M.); (A.R.C.); (C.M.)
- Doctoral School, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
- Center for Complex Network Science, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Aimee Rodica Chis
- Biochemistry Department, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (L.M.); (A.I.M.); (A.R.C.); (C.M.)
- Center for Complex Network Science, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Catalin Marian
- Biochemistry Department, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (L.M.); (A.I.M.); (A.R.C.); (C.M.)
- Center for Complex Network Science, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Constantin Tudor Luca
- Cardiology Department, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (C.T.L.); (D.G.)
- Research Center of the Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania
| | - Ioan-Ovidiu Sirbu
- Biochemistry Department, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (L.M.); (A.I.M.); (A.R.C.); (C.M.)
- Center for Complex Network Science, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Dan Gaiță
- Cardiology Department, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (C.T.L.); (D.G.)
- Research Center of the Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania
| |
Collapse
|
2
|
Deng P, Hu H. HSP90-Dependent Upregulation of EZH2 Promotes Hypoxia/Reoxygenation-Induced Pyroptosis by Inhibiting miR-22 in Endothelial Cells. J Inflamm Res 2023; 16:2615-2630. [PMID: 37360624 PMCID: PMC10289174 DOI: 10.2147/jir.s403531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 06/15/2023] [Indexed: 06/28/2023] Open
Abstract
Objective Endothelial cell pyroptosis induced by hypoxia/reoxygenation (H/R) plays a key role in the pathogenesis of myocardial infarction (MI). However, the underlying mechanism is not clearly elucidated. Methods Human umbilical vein endothelial cells (HUVECs) exposed to H/R acted as in vitro model to investigate the mechanism of H/R-induced endothelial cell pyroptosis. CCK-8 assays were performed to investigate the viability of HUVECs. Calcein-AM/PI staining was carried out to quantify the death of HUVECs. The expression level of miR-22 was measured by RT-qPCR. The protein expression levels of zeste 2 polycomb repressive complex 2 subunit (EZH2), NLRP3, cleaved caspase-1 (c-caspase-1), GSDMD-N and heat shock protein 90 (HSP90) were measured by Western blot. Levels of IL-1β and IL-18 in culture medium were detected by ELISA. The intracellular localization of EZH2 was detected by immunofluorescence staining. Chromatin immunoprecipitation (ChIP) assay was used to detect the enrichment of EZH2 and H3K27me3 in the miR-22 promoter region. The binding between miR-22 and NLRP3 in HUVECs was confirmed by the dual luciferase assay. Reciprocal coimmunoprecipitation was conducted to detect the direct interaction between HSP90 and EZH2. Results H/R increased EZH2 expression, and the EZH2 siRNA could inhibit H/R-induced pyroptosis in HUVECs. H/R reduced miR-22 expression, which was reversed by EZH2 siRNA. Silencing of miR-22 by its inhibitor reversed EZH2 siRNA-induced pyroptosis inhibition in H/R-exposed HUVECs. Upregulation of miR-22 by its mimic suppressed EZH2 overexpression-enhanced pyroptosis in H/R-exposed HUVECs. ChIP assay confirmed that EZH2 bound to the miR-22 promoter region and repressed miR-22 expression through H3K27me3. Furthermore, luciferase reporter assay indicated that NLRP3 was a direct target of miR- 22 in HUVECs. Finally, HSP90 siRNA inhibited H/R-induced EZH2 expression, miR-22 downregulation, and pyroptosis in HUVECs. Conclusion H/R induces pyroptosis via the HSP90/EZH2/miR-22/NLRP3 signaling axis in endothelial cells.
Collapse
Affiliation(s)
- Paihe Deng
- Clinical Laboratory Medicine Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210011, People’s Republic of China
| | - Huimin Hu
- Clinical Laboratory Medicine Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210011, People’s Republic of China
| |
Collapse
|
3
|
Koufaris C, Papandreou ME, Ellis JK, Nicolaidou V, Keun HC. miR-22-enriched breast cancer cells display repressed glycolytic metabolism, increased glycogen synthesis, and reduced survival in low glucose conditions. Mol Biol Rep 2023; 50:5185-5193. [PMID: 37119413 DOI: 10.1007/s11033-023-08458-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 04/12/2023] [Indexed: 05/01/2023]
Abstract
BACKGROUND Breast cancer (BC) is the second leading cause of cancer-related mortality among women. Beyond the established tumourigenic role of genetic mutations, metabolic reprogramming is another key cancer hallmark. Glucose metabolism in particular is known to be prominently altered in tumours, in order to support biomass accumulation and cancer cell survival. The tumor suppressor microRNA (miRNA) miR-22 has been previously associated with a plethora of BC phenotypes such as growth, invasion-metastasis, and regulation of metabolic phenotypes such as lipid and folate metabolism. In this study, we aimed to investigate the role of miR-22 in the regulation of glucose metabolism in BC cells. METHODS AND RESULTS Here we examined how miR-22 affects glucose metabolism in the MCF-7 BC cells. We found that over-expression of miR-22 caused a reduced glycolytic rate in these cells. Moreover, the miRNA also rendered MCF-7 cells more sensitive to lower glucose levels. We next unbiasedly screened the transcript levels of 84 genes relevant to glucose metabolism using the Human Glucose RT2 Profiler PCR Array. Interestingly, the strongest effect identified by this screen was the upregulation of genes involved in glycogen synthesis and the repression of gene involved in glycogen catabolism. Examination of publicly available transcriptomic datasets confirmed the correlations between expression of miR-22 and these glycogen metabolism genes in BC cells. CONCLUSION This study has generated evidence for a regulatory role of miR-22 in glucose and glycogen metabolism, expanding the involvement of this miRNA in BC metabolic reprogramming.
Collapse
Affiliation(s)
- Costas Koufaris
- Department of Biological Sciences, University of Cyprus, Nicosia, Cyprus.
| | | | - James K Ellis
- Department of Surgery & Cancer, Imperial College London, London, UK
| | - Vicky Nicolaidou
- Department of Life Sciences, University of Nicosia, Nicosia, Cyprus
- Non-Coding RNA Research Laboratory, School of Life and Health Sciences, University of Nicosia, Nicosia, Cyprus
| | - Hector C Keun
- Department of Surgery & Cancer, Imperial College London, London, UK
| |
Collapse
|
4
|
Wang J, Zeng J, Yin G, Deng Z, Wang L, Liu J, Yao K, Long Z, Jiang X, Tan J. Long non-coding RNA FABP5P3/ miR-22 axis improves TGFβ1-induced fatty acid oxidation deregulation and fibrotic changes in proximal tubular epithelial cells of renal fibrosis. Cell Cycle 2023; 22:433-449. [PMID: 36196456 PMCID: PMC9879175 DOI: 10.1080/15384101.2022.2122286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Severe hydronephrosis increases the risk of urinary tract infection and irretrievable renal fibrosis. While TGFβ1-mediated fibrotic changes in proximal tubular epithelial cells and fatty acid oxidation (FAO) deregulation contribute to renal fibrosis and hydronephrosis. Firstly, a few elements were analyzed in this paper, including differentially-expressed long non-coding RNAs (lncRNAs), and miRNAs correlated to CPT1A, RXRA, and NCOA1. This paper investigated TGFβ1 effects on lncRNA FABP5P3, CPT1A, RXRA, and NCOA1 expression and fibrotic changes in HK-2 cells and FABP5P3 overexpression effects on TGFβ1-induced changes. Moreover, this paper predicted and proved that miR-22 binding to lncRNA FABP5P3, 3'UTR of CPT1A, RXRA, and NCOA1 was validated. The dynamic effects of the FABP5P3/miR-22 axis on TGFβ1-induced changes were investigated. A Renal fibrosis model was established in unilateral ureteral obstruction (UUO) mice, and FABP5P3 effects were investigated. Eventually, this paper concluded that TGFβ1 inhibited lncRNA FABP5P3, CPT1A, RXRA, and NCOA1 expression, induced fibrotic changes in HK-2 cells, and induced metabolic reprogramming within HK-2 cells, especially lower FAO. FABP5P3 overexpression partially reversed TGFβ1-induced changes. miR-22 targeted lncRNA FABP5P3, CPT1A, RXRA, and NCOA1. LncRNA FABP5P3 counteracted miR-22 inhibition of CPT1A, NCOA1, and RXRA through competitive binding. TGFβ1 stimulation induced the activation of TGFβ/SMAD and JAG/Notch signaling pathways; Nocth2 knockdown reversed TGFβ1 suppression on lncRNA FABP5P3. FABP5P3 overexpression attenuated renal fibrosis in unilateral ureteral obstruction mice. The LncRNA FABP5P3/miR-22 axis might be a potent target for improving the FAO deregulation and fibrotic changes in proximal TECs under TGFβ1 stimulation.
Collapse
Affiliation(s)
- Jingrong Wang
- Department of Urology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Jia Zeng
- Department of Urology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Guangmin Yin
- Department of Urology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Zhijun Deng
- Department of Urology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Long Wang
- Department of Urology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Jianye Liu
- Department of Urology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Kun Yao
- Department of Urology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Zhi Long
- Department of Urology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Xianzhen Jiang
- Department of Urology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Jing Tan
- Department of Urology, The Third Xiangya Hospital of Central South University, Changsha, China,CONTACT Jing Tan Department of Urology, The Third Xiangya Hospital of Central South University, No.138 Tongzipo Road, Changsha, China
| |
Collapse
|
5
|
Hosseinpour-Soleimani F, Khamisipour G, Derakhshan Z, Ahmadi B. Expression analysis of circulating miR-22, miR-122, miR-217 and miR-367 as promising biomarkers of acute lymphoblastic leukemia. Mol Biol Rep 2023; 50:255-65. [PMID: 36327023 DOI: 10.1007/s11033-022-08016-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 10/07/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND The role of serum-based biomarkers such as microRNAs in cancer diagnosis has been extensively established. This study aimed to determine the expression levels of bioinformatically selected miRNAs and whether they can be used as biomarkers or a new therapeutic target in patients with acute lymphoblastic leukemia (ALL). MATERIALS AND METHODS The expression levels of serum miR-22, miR-122, miR-217, and miR-367 in 21 ALL patients and 21 healthy controls were measured using quantitative real-time PCR. The receiver operating characteristic (ROC) curve and the associated area under the curve (AUC) was used to assess candidate miRNAs' diagnostic value as a biomarker. RESULTS The results showed that miR-217 was markedly decreased in patients with ALL compared to controls. Moreover, miR-22, miR-122, and miR-367 were found to be upregulated. Furthermore, ROC analysis showed that serum miR-217 and miR-367 could differentiate ALL patients from healthy individuals, while miR-22 has approximate discriminatory power that requires further investigation. CONCLUSION These results provide promising preliminary evidence that circulating miR-217 and miR-367 could be considered potent diagnostic biomarkers and therapeutic goals in this disease.
Collapse
|
6
|
Mukhopadhyay P, Smolenkova I, Seelan RS, Pisano MM, Greene RM. Spatiotemporal Expression and Functional Analysis of miRNA-22 in the Developing Secondary Palate. Cleft Palate Craniofac J 2023; 60:27-38. [PMID: 34730446 DOI: 10.1177/10556656211054004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVE Normal development of the embryonic orofacial region requires precise spatiotemporal coordination between numerous genes. MicroRNAs represent small, single-stranded, non-coding molecules that regulate gene expression. This study examines the role of microRNA-22 (miR-22) in murine orofacial ontogeny. METHODS Spatiotemporal and differential expression of miR-22 (mmu-miR-22-3p) within the developing secondary palate was determined by in situ hybridization and quantitative real-time PCR, respectively. Bioinformatic approaches were used to predict potential mRNA targets of miR-22 and analyze their association with cellular functions indispensable for normal orofacial ontogeny. An in vitro palate organ culture system was used to assess the role of miR-22 in secondary palate development. RESULTS There was a progressive increase in miR-22 expression from GD12.5 to GD14.5 in palatal processes. On GD12.5 and GD13.5, miR-22 was expressed in the future oral, nasal, and medial edge epithelia. On GD14.5, miR-22 expression was observed in the residual midline epithelial seam (MES), the nasal epithelium and the mesenchyme, but not in the oral epithelium. Inhibition of miR-22 activity in palate organ cultures resulted in failure of MES removal. Bioinformatic analyses revealed potential mRNA targets of miR-22 that may play significant roles in regulating apoptosis, migration, and/or convergence/extrusion, developmental processes that modulate MES removal during palatogenesis. CONCLUSIONS Results from the current study suggest a key role for miR-22 in the removal of the MES during palatogenesis and that miR-22 may represent a potential contributor to the etiology of cleft palate.
Collapse
Affiliation(s)
- Partha Mukhopadhyay
- Department of Oral Immunology and Infectious Diseases, Division of Craniofacial Development and Anomalies, School of Dentistry, 5170University of Louisville, Louisville, KY 40202
| | - Irina Smolenkova
- Department of Oral Immunology and Infectious Diseases, Division of Craniofacial Development and Anomalies, School of Dentistry, 5170University of Louisville, Louisville, KY 40202
| | - Ratnam S Seelan
- Department of Oral Immunology and Infectious Diseases, Division of Craniofacial Development and Anomalies, School of Dentistry, 5170University of Louisville, Louisville, KY 40202
| | - M Michele Pisano
- Department of Oral Immunology and Infectious Diseases, Division of Craniofacial Development and Anomalies, School of Dentistry, 5170University of Louisville, Louisville, KY 40202
| | - Robert M Greene
- Department of Oral Immunology and Infectious Diseases, Division of Craniofacial Development and Anomalies, School of Dentistry, 5170University of Louisville, Louisville, KY 40202
| |
Collapse
|
7
|
Centomo ML, Vitiello M, Poliseno L, Pandolfi PP. An Immunocompetent Environment Unravels the Proto-Oncogenic Role of miR-22. Cancers (Basel) 2022; 14:cancers14246255. [PMID: 36551740 PMCID: PMC9776418 DOI: 10.3390/cancers14246255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/09/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
MiR-22 was first identified as a proto-oncogenic microRNA (miRNA) due to its ability to post-transcriptionally suppress the expression of the potent PTEN (Phosphatase And Tensin Homolog) tumor suppressor gene. miR-22 tumorigenic role in cancer was subsequently supported by its ability to positively trigger lipogenesis, anabolic metabolism, and epithelial-mesenchymal transition (EMT) towards the metastatic spread. However, during the following years, the picture was complicated by the identification of targets that support a tumor-suppressive role in certain tissues or cell types. Indeed, many papers have been published where in vitro cellular assays and in vivo immunodeficient or immunosuppressed xenograft models are used. However, here we show that all the studies performed in vivo, in immunocompetent transgenic and knock-out animal models, unanimously support a proto-oncogenic role for miR-22. Since miR-22 is actively secreted from and readily exchanged between normal and tumoral cells, a functional immune dimension at play could well represent the divider that allows reconciling these contradictory findings. In addition to a critical review of this vast literature, here we provide further proof of the oncogenic role of miR-22 through the analysis of its genomic locus vis a vis the genetic landscape of human cancer.
Collapse
Affiliation(s)
- Maria Laura Centomo
- Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Turin, Italy
- William N. Pennington Cancer Institute, Renown Health, Nevada System of Higher Education, Reno, NV 89502, USA
- Center for Genomic Medicine, Desert Research Institute, Reno, NV 89512, USA
| | - Marianna Vitiello
- Institute of Clinical Physiology, National Research Council, Via Moruzzi 1, 56124 Pisa, Italy
- Oncogenomics Unit, Core Research Laboratory, ISPRO, Via Moruzzi 1, 56124 Pisa, Italy
| | - Laura Poliseno
- Institute of Clinical Physiology, National Research Council, Via Moruzzi 1, 56124 Pisa, Italy
- Oncogenomics Unit, Core Research Laboratory, ISPRO, Via Moruzzi 1, 56124 Pisa, Italy
- Correspondence: (L.P.); (P.P.P.); Tel.: +39-050-315-2780 (L.P.); +1-775-982-6210 (P.P.P.); Fax: +39-050-315-3327 (L.P.); +1-775-982-4288 (P.P.P.)
| | - Pier Paolo Pandolfi
- Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Turin, Italy
- William N. Pennington Cancer Institute, Renown Health, Nevada System of Higher Education, Reno, NV 89502, USA
- Center for Genomic Medicine, Desert Research Institute, Reno, NV 89512, USA
- Correspondence: (L.P.); (P.P.P.); Tel.: +39-050-315-2780 (L.P.); +1-775-982-6210 (P.P.P.); Fax: +39-050-315-3327 (L.P.); +1-775-982-4288 (P.P.P.)
| |
Collapse
|
8
|
Jiang W, Sun S, Wang D, Qiu J, Song Y, Zhang Q, He W, Song B, Zhang Y, Wang S. MicroRNA-22 suppresses NLRP3/CASP1 inflammasome pathway-mediated proinflammatory cytokine production by targeting the HIF-1α and NLRP3 in human dental pulp fibroblasts. Int Endod J 2022; 55:1225-1240. [PMID: 35979583 DOI: 10.1111/iej.13814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 08/10/2022] [Accepted: 08/11/2022] [Indexed: 01/07/2023]
Abstract
AIM To investigate the synergetic regulatory effect of miR-22 on HIF-1α and NLRP3, subsequently regulating the production of the NLRP3/CASP1 inflammasome pathway-mediated proinflammatory cytokines IL-1β and IL-18 in human dental pulp fibroblasts (HDPFs) during the progression of pulpitis. METHODOLOGY Fluorescence in situ hybridization (FISH) and immunofluorescence (IF) were performed to determine the localization of miR-22-3p, NLRP3 and HIF-1α in human dental pulp tissues (HDPTs). The miR-22 mimics and inhibitor or plasmid of NLRP3 or HIF-1α were used to upregulate or downregulate miR-22 or NLRP3 or HIF-1α in HDPFs, respectively. Computational prediction via TargetScan 5.1 and a luciferase reporter assay were conducted to confirm target association. The mRNA and protein expression of HIF-1α, NLRP3, caspase-1, IL-1β and IL-18 were determined by qRT-PCR and western blotting, respectively. The release of IL-1β and IL-18 was analysed by ELISA. The significance of the differences between the experimental and control groups was determined by one-way analysis of variance, p < .05 indicated statistical significance. RESULTS A decrease in miR-22 and an increase in HIF-1α and NLRP3 in HDPTs occurred during the transformation of reversible pulpitis into irreversible pulpitis compared with that in the healthy pulp tissues (p < .05). In the normal HDPTs, miR-22-3p was extensively expressed in dental pulp cells. HIF-1α and NLRP3 were mainly expressed in the odontoblasts and vascular endothelial cells. Whereas in the inflamed HDPTs, the odontoblast layers were disrupted. HDPFs were positive for miR-22-3p, HIF-1α and NLRP3. Computational prediction via TargetScan 5.1 and luciferase reporter assays confirmed that both NLRP3 and HIF-1α were direct targets of miR-22 in HDPFs. The miR-22 inhibitor further promoted the activation of NLRP3/CASP1 inflammasome pathway induced by ATP plus LPS and hypoxia (p < .05). In contrast, the miR-22 mimic significantly inhibited the NLRP3/CASP1 inflammasome pathway activation induced by ATP plus LPS and hypoxia (p < .05). CONCLUSION MiR-22, as a synergetic negative regulator, is involved in controlling the secretion of proinflammatory cytokines mediated by the NLRP3/CASP1 inflammasome pathway by targeting NLRP3 and HIF-1α. These results provide a novel function and mechanism of miR-22-HIF-1α-NLRP3 signalling in the control of proinflammatory cytokine secretion, thus indicating a potential therapeutic strategy for future endodontic treatment.
Collapse
Affiliation(s)
- Wenkai Jiang
- Department of Operative Dentistry & Endodontics, School of Stomatology, State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Fourth Military Medical University, Xi'an, China.,School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Cardiff, UK
| | - Shukai Sun
- Department of Pediatric Dentistry, School of Stomatology, State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Clinical Research Center for Oral Diseases, Fourth Military Medical University, Xi'an, China
| | - Diya Wang
- Department of Occupational and Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, China
| | - Jun Qiu
- Department of Operative Dentistry & Endodontics, School of Stomatology, State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Fourth Military Medical University, Xi'an, China
| | - Ya Song
- Department of Operative Dentistry & Endodontics, School of Stomatology, State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Fourth Military Medical University, Xi'an, China
| | - Qianxia Zhang
- Department of Operative Dentistry & Endodontics, School of Stomatology, State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Fourth Military Medical University, Xi'an, China
| | - Wenxi He
- Department of Operative Dentistry & Endodontics, School of Stomatology, State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Fourth Military Medical University, Xi'an, China
| | - Bing Song
- Department of Operative Dentistry & Endodontics, School of Stomatology, State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Fourth Military Medical University, Xi'an, China.,School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Cardiff, UK
| | - Yaqing Zhang
- Department of Operative Dentistry & Endodontics, School of Stomatology, State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Fourth Military Medical University, Xi'an, China
| | - Shengchao Wang
- Department of Operative Dentistry & Endodontics, School of Stomatology, State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
9
|
Hu Y, Fan Y, Zhang C, Wang C. Palmitic acid inhibits vascular smooth muscle cell switch to synthetic phenotype via upregulation of miR-22 expression. Aging (Albany NY) 2022; 14:8046-8060. [PMID: 36227173 PMCID: PMC9596196 DOI: 10.18632/aging.204334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 10/03/2022] [Indexed: 11/25/2022]
Abstract
Synthetic phenotype switch of vascular smooth muscle cells (VSMCs) has been shown to play key roles in vascular diseases. Mounting evidence has shown that fatty acid metabolism is highly associated with vascular diseases. However, how fatty acids regulate VSMC phenotype is poorly understood. Hence, the effects of palmitic acid (PA) on VSMC phenotype were determined in this study. The effect of the PA on VSMCs was measured by live/dead and EdU assays, as well as flow cytometry. Migration ability of VSMCs was evaluated using transwell assay. The underlying targets of miR-22 were predicted using bioinformatics online tools, and confirmed by luciferase reporter assay. The RNA and protein expression of certain gene was detected by qRT-PCR or western blot. PA inhibited VSMC switch to synthetic phenotype, as manifested by inhibiting VSMC proliferation, migration, and synthesis. PA upregulated miR-22 in VSMCs, and miR-22 mimics exerted similar effects as PA treatment, inhibiting VSMC switch to synthetic phenotype. Inhibition of miR-22 using miR-22 inhibitor blocked the impacts of PA on VSMC phenotype modulation, suggesting that PA modulated VSMC phenotype through upregulation of miR-22 expression. We found that ecotropic virus integration site 1 protein homolog (EVI1) was the target of miR-22 in regulation of VSMC phenotype. Overexpression of miR-22 or/and PA treatment attenuated the inhibition of EVI1 on switch of VSMCs. These findings suggested that PA inhibits VSMC switch to synthetic phenotype through upregulation of miR-22 thereby inhibiting EVI1, and correcting the dysregulation of miR-22/EVI1 or PA metabolism is a potential treatment to vascular diseases.
Collapse
Affiliation(s)
- Yanchao Hu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Shaanxi, Xi'an 710004, China
| | - Yajie Fan
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Shaanxi, Xi'an 710004, China
| | - Chunyan Zhang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Shaanxi, Xi'an 710004, China
| | - Congxia Wang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Shaanxi, Xi'an 710004, China
| |
Collapse
|
10
|
Li W, Yuan X, He X, Yang L, Wu Y, Deng X, Zeng Y, Hu K, Tang B. The downregulation of miR-22 and miR-372 may contribute to gestational diabetes mellitus through regulating glucose metabolism via the PI3K/AKT/GLUT4 pathway. J Clin Lab Anal 2022; 36:e24557. [PMID: 35712865 PMCID: PMC9279990 DOI: 10.1002/jcla.24557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/07/2022] [Accepted: 05/25/2022] [Indexed: 11/27/2022] Open
Abstract
Background Identifying effective regulatory mechanisms will be significant for Gestational diabetes mellitus (GDM) diagnosis and treatment. Methods The expressions of miR‐22 and miR‐372 in placenta tissues from 75 pregnant women with GDM and 75 matched healthy controls and HRT8/SVneo cells (a model of insulin resistance) were analyzed by qPCR. The expressions of PI3K, AKT, IRS, and GLUT4 in high glucose‐treated HRT8/SVneo cells transfected with miR‐22 or miR‐372 mimics or inhibitors was assessed by Western blot. A luciferase gene reporter assay was employed to verify miRNAs' target genes. Results The expressions of miR‐22 and miR‐372 in placental tissues from GDM patients and HRT8/SVneo cells were significantly decreased compared with the respective controls. The GLUT4 expression was significantly decreased in the placenta tissues of GDM and HRT8/SVneo cells with high glucose transfected with miR‐22 and miR‐372 inhibitors. We confirmed that SLC2A4, the gene encoding GLUT4, was a direct target of miR‐22 and miR‐372. In this study, we report that the lower expressions of miR‐22 and miR‐372 in placental tissue from GDM patients. Conclusion Our results further suggested that the downregulations of miR‐22 and miR‐372 may contribute to GDM through regulating the PI3K/GLUT4 pathway.
Collapse
Affiliation(s)
- Wei Li
- Department of Endocrinology, Armed Police Corps Hospital of Guangdong Province, Guangzhou, China
| | - Xianlin Yuan
- Department of Food and Biological Engineering, Guangdong Industry Technical College, Guangzhou, China
| | - Xin He
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China
| | - Li Yang
- Department of Endocrinology, Armed Police Corps Hospital of Guangdong Province, Guangzhou, China
| | - Yingyuan Wu
- Department of Obstetrics and Gynecology, Armed Police Corps Hospital of Guangdong Province, Guangzhou, China
| | - Xiaofeng Deng
- Department of Central Sterile Supply, Armed Police Corps Hospital of Guangdong Province, Guangzhou, China
| | - Yiwen Zeng
- Department of Endocrinology, Armed Police Corps Hospital of Guangdong Province, Guangzhou, China
| | - Kesheng Hu
- Department of Clinical Laboratory, Armed Police Corps Hospital of Guangdong Province, Guangzhou, China
| | - Bo Tang
- Department of Clinical Pharmacy, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
11
|
Gu Y, Pais G, Becker V, Körbel C, Ampofo E, Ebert E, Hohneck J, Ludwig N, Meese E, Bohle RM, Zhao Y, Menger MD, Laschke MW. Suppression of endothelial miR-22 mediates non-small cell lung cancer cell-induced angiogenesis. Mol Ther Nucleic Acids 2021; 26:849-864. [PMID: 34729252 PMCID: PMC8536510 DOI: 10.1016/j.omtn.2021.10.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 09/06/2021] [Accepted: 10/01/2021] [Indexed: 11/16/2022]
Abstract
MicroRNAs (miRNAs) expressed in endothelial cells (ECs) are powerful regulators of angiogenesis, which is essential for tumor growth and metastasis. Here, we demonstrated that miR-22 is preferentially and highly expressed in ECs, while its endothelial level is significantly downregulated in human non-small cell lung cancer (NSCLC) tissues when compared to matched nontumor lung tissues. This reduction of endothelial miR-22 is possibly induced by NSCLC cell-secreted interleukin-1β and subsequently activated transcription factor nuclear factor-κB. Endothelial miR-22 functions as a potent angiogenesis inhibitor that inhibits all of the key angiogenic activities of ECs and consequently NSCLC growth through directly targeting sirtuin 1 and fibroblast growth factor receptor 1 in ECs, leading to inactivation of AKT/mammalian target of rapamycin signaling. These findings provide insight into the molecular mechanisms of NSCLC angiogenesis and indicate that endothelial miR-22 represents a potential target for the future antiangiogenic treatment of NSCLC.
Collapse
Affiliation(s)
- Yuan Gu
- Institute for Clinical and Experimental Surgery, Saarland University, 66421 Homburg/Saar, Germany
| | - Gianni Pais
- Institute for Clinical and Experimental Surgery, Saarland University, 66421 Homburg/Saar, Germany
| | - Vivien Becker
- Institute for Clinical and Experimental Surgery, Saarland University, 66421 Homburg/Saar, Germany
| | - Christina Körbel
- Institute for Clinical and Experimental Surgery, Saarland University, 66421 Homburg/Saar, Germany
| | - Emmanuel Ampofo
- Institute for Clinical and Experimental Surgery, Saarland University, 66421 Homburg/Saar, Germany
| | - Elke Ebert
- Institute of Pathology, Saarland University, 66421 Homburg/Saar, Germany
| | - Johannes Hohneck
- Institute of Pathology, Saarland University, 66421 Homburg/Saar, Germany
| | - Nicole Ludwig
- Institute of Human Genetics, Saarland University, 66421 Homburg/Saar, Germany
| | - Eckart Meese
- Institute of Human Genetics, Saarland University, 66421 Homburg/Saar, Germany
| | - Rainer M Bohle
- Institute of Pathology, Saarland University, 66421 Homburg/Saar, Germany
| | - Yingjun Zhao
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Michael D Menger
- Institute for Clinical and Experimental Surgery, Saarland University, 66421 Homburg/Saar, Germany
| | - Matthias W Laschke
- Institute for Clinical and Experimental Surgery, Saarland University, 66421 Homburg/Saar, Germany
| |
Collapse
|
12
|
Wu Z, Geng J, Bai Y, Qi Y, Chang C, Jiao Y, Guo Z. MicroRNA-22 inhibition promotes the development of atherosclerosis via targeting interferon regulator factor 5. Exp Cell Res 2021; 409:112922. [PMID: 34780785 DOI: 10.1016/j.yexcr.2021.112922] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 10/15/2021] [Accepted: 11/11/2021] [Indexed: 12/12/2022]
Abstract
Atherosclerosis is generally accepted as a chronic inflammatory disease and is the most important pathological process underlying the cardiovascular diseases. MiR-22 exerts an important role in tumorgenesis, obesity and NAFLD development, as well as cardiovascular diseases. However, a certain role of miR-22 in the pathogenesis of atherosclerosis remains undetermined. Here, we showed that miR-22 exhibited a negative association with the deteriorated atherosclerotic plaque and showed significant downregulated expression in macrophages. Next, treatment of ApoE deficiency (ApoE-/-) mice with miR-22 inhibitors which were then subjected to high fat diet (HFD) for 12 weeks were performed to investigate the function of miR-22 on atherogenesis. The results exhibited that miR-22 inhibition dramatically promoted atherosclerotic plaques but attenuated plaque stabilization which were accompanied by decreased smooth muscle cell and collagen content, but increased macrophage infiltration and lipid accumulation. More importantly, the in vivo and in vitro experiments suggested that miR-22 inhibition accelerated inflammatory response and foam cell formation. Mechanistically, we demonstrated interferon regulator factor 5 (IRF5) was an important target of miR-22 and it was required for the regulation of inflammation mediated by miR-22 inhibition. Collectively, these evidences revealed that miR-22 inhibition promoted the atherosclerosis progression through activation of IRF5.
Collapse
Affiliation(s)
- Zhenhua Wu
- Department of Cardiac Surgery, Tianjin Chest Hospital, Tianjin, 300222, China; Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300073, China
| | - Jie Geng
- Cardiac Intensive Care Unit, Tianjin Chest Hospital, Tianjin, 300222, China
| | - Yunpeng Bai
- Department of Cardiac Surgery, Tianjin Chest Hospital, Tianjin, 300222, China
| | - Yujuan Qi
- Department of Cardiac Surgery, Tianjin Chest Hospital, Tianjin, 300222, China
| | - Chao Chang
- Department of Cardiac Surgery, Tianjin Chest Hospital, Tianjin, 300222, China
| | - Yan Jiao
- Department of Cardiac Surgery, Tianjin Chest Hospital, Tianjin, 300222, China
| | - Zhigang Guo
- Department of Cardiac Surgery, Tianjin Chest Hospital, Tianjin, 300222, China.
| |
Collapse
|
13
|
Wang XS, Li LC, Zhang X, Gao J. Lipoxin A 4 methyl ester protects PC12 cells from ketamine-induced neurotoxicity via the miR-22/BAG5 pathway. Hum Exp Toxicol 2021; 40:S519-S529. [PMID: 34670429 DOI: 10.1177/09603271211051602] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Ketamine is an anesthetic that induces neurotoxicity when administered at high doses. In this work, we explored the protective effects of lipoxin A4 methyl ester (LXA4 ME) against ketamine-induced neurotoxicity and the underlying protective mechanism in pheochromocytoma (PC12) cells. METHODS PC12 cells were treated with 50 μM of ketamine and different LXA4 ME concentrations of LXA4 ME (5-50 nM) for 24 h, and their viability, apoptosis, and oxidative status were assessed. RESULTS Quantitative real-time polymerase chain reaction experiments showed that ketamine downregulated miR-22 expression and upregulated Bcl-2-associated athanogene 5 (BAG5) in PC12 cells in a concentration-dependent manner. LXA4 ME induced the opposite effects, thus attenuating ketamine-induced neurotoxicity. Further in vitro assays showed that miR-22 directly targeted BAG5, thus promoting cell viability by suppressing cell apoptosis and oxidative stress. Under expression miR-22 or upregulation of BAG5 antagonized the effects of LXA4 ME. CONCLUSION LXA4 ME can protect PC12 cells from ketamine-induced neurotoxicity by activating the miR-22/BAG5 signaling pathway. Thus, LXA4 ME can be used as a protective drug against ketamine-induced neural damage.
Collapse
Affiliation(s)
- Xue-Song Wang
- Department of Anesthesiology, 74731Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| | - Long-Cheng Li
- Department of Anesthesiology, 74731Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| | - Xue Zhang
- Department of Anesthesiology, 74731Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| | - Jin Gao
- Department of Anesthesiology, 74731Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| |
Collapse
|
14
|
Zhang S, Zhang D, Yi C, Wang Y, Wang H, Wang J. [Retracted] MicroRNA‑22 functions as a tumor suppressor by targeting SIRT1 in renal cell carcinoma. Oncol Rep 2021; 46:243. [PMID: 34558635 PMCID: PMC8482452 DOI: 10.3892/or.2021.8194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 07/30/2015] [Indexed: 11/10/2022] Open
Affiliation(s)
- Shoulin Zhang
- Internal Medicine Department, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin 130021, P.R. China
| | - Dongmei Zhang
- Scientific Research Office, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin 130021, P.R. China
| | - Chunguang Yi
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun Jingyue National High-Tech Industrial Development Zone, Changchun, Jilin 130117, P.R. China
| | - Yinping Wang
- Internal Medicine Department, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin 130021, P.R. China
| | - Hongan Wang
- Internal Medicine Department, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin 130021, P.R. China
| | - Jian Wang
- Internal Medicine Department, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
15
|
Caracciolo D, Riillo C, Juli G, Scionti F, Todoerti K, Polerà N, Grillone K, Fiorillo L, Arbitrio M, Di Martino MT, Neri A, Tagliaferri P, Tassone P. miR-22 Modulates Lenalidomide Activity by Counteracting MYC Addiction in Multiple Myeloma. Cancers (Basel) 2021; 13:4365. [PMID: 34503175 DOI: 10.3390/cancers13174365] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 08/21/2021] [Accepted: 08/24/2021] [Indexed: 11/25/2022] Open
Abstract
Simple Summary MYC-driven deregulation of microRNAs represents a critical event in human malignancies, including multiple myeloma (MM). Although the introduction of new therapeutic strategies has prolonged survival of patients, MM remains an incurable disease, often due to the onset of drug resistance. MYC hyperactivation is involved in the development of resistance to immunomodulatory imide drugs (IMiDs), but the mechanism is still unclear. Here, we report that MYC represses the transcription of tumor suppressor miR-22 in MM, and that low miR-22 expression is associated with IMiD resistance in MM patients. By in silico and in vitro analysis, we show that miR-22 mimics affect MYC signaling, leading to MM cell death in MYC proficient cells. Furthermore, we demonstrate here that lenalidomide treatment enhances miR-22 activity by reducing the MYC inhibitory effect, and that the combination of lenalidomide with miR-22 mimics restores drug sensitivity, leading to synergistic anti-MM activity. Abstract Background: MYC is a master regulator of multiple myeloma (MM) by orchestrating several pro-tumoral pathways, including reprograming of the miRNA transcriptome. MYC is also involved in the acquirement of resistance to anti-MM drugs, including immunomodulatory imide drugs (IMiDs). Methods: In silico analysis was performed on MM proprietary and on public MMRF-CoMMpass datasets. Western blot and chromatin immunoprecipitation (ChIP) experiments were performed to validate miR-22 repression induced by MYC. Cell viability and apoptosis assays were used to evaluate lenalidomide sensitization after miR-22 overexpression. Results: We found an inverse correlation between MYC and miR-22 expression, which is associated with poor outcome in IMiD-treated MM patients. Mechanistically, we showed that MYC represses transcription of miR-22, which, in turn, targets MYC, thus establishing a feed-forward loop. Interestingly, we found that IMiD lenalidomide increases miR-22 expression by reducing MYC repression and, most importantly, that the combination of lenalidomide with miR-22 mimics results in a synergistic direct and NK-mediated cytotoxic activity. Conclusions: Taken together, our findings indicate that: (1) low miR-22 expression could represent a potential predictive biomarker of poor lenalidomide response in MM patients; and (2) miR-22 reduces MYC oncogenic activity, thus triggering a novel synthetic lethality loop, which sensitizes MM cells to lenalidomide.
Collapse
|
16
|
Yun J, Ji YS, Jang GH, Lim SH, Kim SH, Kim CK, Bae SB, Won JH, Park SK. TET2 Mutation and High miR-22 Expression as Biomarkers to Predict Clinical Outcome in Myelodysplastic Syndrome Patients Treated with Hypomethylating Therapy. Curr Issues Mol Biol 2021; 43:917-31. [PMID: 34449560 DOI: 10.3390/cimb43020065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/23/2021] [Accepted: 07/29/2021] [Indexed: 11/16/2022] Open
Abstract
Tet methylcytosine dioxygenase 2 (TET2) is one of the most frequently mutated genes in myelodysplastic syndrome (MDS). TET2 is known to involve a demethylation process, and the loss of TET2 is thought to cause DNA hypermethylation. Loss of TET2 function is known to be caused by genetic mutations and miRNA, such as miR-22. We analyzed 41 MDS patients receiving hypomethylating therapy (HMT) to assess whether TET2 mutation status and miR-22 expression status were associated with their clinical characteristics and treatment outcomes. Responsiveness to HMT was not affected by both TET2 mutation (odds ratio (OR) 0.900, p = 0.909) and high miR-22 expression (OR 1.548, p = 0.631). There was a tendency for TET2 mutation to be associated with lower-risk disease based on IPSS (Gamma = -0.674, p = 0.073), lower leukemic transformation (OR 0.170, p = 0.040) and longer survival (Hazard ratio 0.354, p = 0.059). Although high miR-22 expression also showed a similar tendency, this tendency was weaker than that of TET2 mutation. In summary, the loss of TET2 function, including both TET2 mutation and high miR-22 expression, was not a good biomarker for predicting the response to HMT but may be associated with lower-risk disease based on IPSS, lower leukemic transformation and longer survival.
Collapse
|
17
|
Rinaldi F, Marchesi F, Palombi F, Pelosi A, Di Pace AL, Sacconi A, Terrenato I, Annibali O, Tomarchio V, Marino M, Cantonetti M, Vaccarini S, Papa E, Moretta L, Bertoni F, Mengarelli A, Regazzo G, Rizzo MG. MiR-22, a serum predictor of poor outcome and therapy response in diffuse large B-cell lymphoma patients. Br J Haematol 2021; 195:399-404. [PMID: 34318932 DOI: 10.1111/bjh.17734] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 07/13/2021] [Indexed: 11/30/2022]
Abstract
Diffuse large B-cell lymphoma (DLBCL) is an aggressive, heterogeneous neoplasm where prognostication and therapeutic decision are challenging. The available prognostic tools are not able to identify all patients refractory to treatment. MicroRNAs, small RNAs frequently deregulated in cancer, stably circulate in biofluids, representing interesting candidates for non-invasive biomarkers. Here we validated serum miR-22, an evolutionarily conserved microRNA, as a prognostic/predictive biomarker in DLBCL. Moreover, we found that its expression and release from DLBCL cells are related to therapy response and adversely affect cell proliferation. These results suggest that miR-22 is a promising complementary or even independent non-invasive biomarker for DLBCL management.
Collapse
Affiliation(s)
- Federica Rinaldi
- Department of Research, Advanced Diagnostics and Technological Innovation, Oncogenomic and Epigenetic Unit, Translational Research Area, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Francesco Marchesi
- Department of Clinical and Experimental Oncology, Hematology and Stem Cell Transplant Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Francesca Palombi
- Department of Clinical and Experimental Oncology, Hematology and Stem Cell Transplant Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Andrea Pelosi
- Immunology Area, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | | | - Andrea Sacconi
- Department of Research, Advanced Diagnostics and Technological Innovation, Clinical Trial Center, Biostatistics and Bioinformatics Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Irene Terrenato
- Department of Research, Advanced Diagnostics and Technological Innovation, Clinical Trial Center, Biostatistics and Bioinformatics Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Ombretta Annibali
- Hematology and Stem Cell Transplantation Unit, University Campus Bio-Medico, Rome, Italy
| | - Valeria Tomarchio
- Hematology and Stem Cell Transplantation Unit, University Campus Bio-Medico, Rome, Italy
| | - Mirella Marino
- Department of Research, Advanced Diagnostics and Technological Innovation, Pathology Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Maria Cantonetti
- Hematology Unit of Lymphoproliferative Disorders, Policlinico Tor Vergata (PTV) Foundation, Rome, Italy
| | - Sara Vaccarini
- Hematology Unit of Lymphoproliferative Disorders, Policlinico Tor Vergata (PTV) Foundation, Rome, Italy
| | - Elena Papa
- Department of Clinical and Experimental Oncology, Hematology and Stem Cell Transplant Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Lorenzo Moretta
- Immunology Area, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Francesco Bertoni
- Faculty of Biomedical Sciences, Institute of Oncology Research, USI, Bellinzona, Switzerland.,Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - Andrea Mengarelli
- Department of Clinical and Experimental Oncology, Hematology and Stem Cell Transplant Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Giulia Regazzo
- Department of Research, Advanced Diagnostics and Technological Innovation, Oncogenomic and Epigenetic Unit, Translational Research Area, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Maria G Rizzo
- Department of Research, Advanced Diagnostics and Technological Innovation, Oncogenomic and Epigenetic Unit, Translational Research Area, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| |
Collapse
|
18
|
Chi K, Geng X, Liu C, Zhang Y, Cui J, Cai G, Chen X, Wang F, Hong Q. LncRNA-HOTAIR promotes endothelial cell pyroptosis by regulating the miR-22/NLRP3 axis in hyperuricaemia. J Cell Mol Med 2021; 25:8504-8521. [PMID: 34296520 PMCID: PMC8419175 DOI: 10.1111/jcmm.16812] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 07/06/2021] [Accepted: 07/09/2021] [Indexed: 12/20/2022] Open
Abstract
Long non‐coding RNA (lncRNA) plays an important role in the renal inflammatory response caused by hyperuricaemia. However, the underlying molecular mechanisms through which lncRNA is involved in endothelial injury induced by hyperuricaemia remain unclear. In this study, we investigated the regulatory role of lncRNA‐HOTAIR in high concentration of uric acid (HUA)–induced renal injury. We established hyperuricaemia mouse model and an in vitro uric acid (UA)–induced human umbilical vein endothelial cell (HUVEC) injury model. In HUA‐treated HUVECs and hyperuricaemia mice, we observed increased HOTAIR and decreased miR‐22 expression. The expression of pyroptosis‐associated protein (NLRP3, Caspase‐1, GSDMD‐N, GSDMD‐FL) was increased. The release of LDH, IL‐1β and IL‐18 in cell supernatants and the sera of model mice was also increased. The proliferation of HUVECs stimulated by HUA was significantly inhibited, and the number of TUNEL‐positive cells in hyperuricaemia mouse kidney was increased. Bioinformatics analysis and luciferase reporter and RIP assays confirmed that HOTAIR promoted NLRP3 inflammasome activation by competitively binding miR‐22. In gain‐ or loss‐of‐function experiments, we found that HOTAIR and NLRP3 overexpression or miR‐22 knock down activated the NLRP3 inflammasome and promoted pyroptosis in HUA‐treated HUVECs, while NLRP3 and HOTAIR knockdown or a miR‐22 mimic exerted the opposite effects. Furthermore, in vivo experiments validated that HOTAIR knockdown alleviated renal inflammation in hyperuricaemia mice. In conclusion, we demonstrated that in hyperuricaemia, lncRNA‐HOTAIR promotes endothelial cell pyroptosis by competitively binding miR‐22 to regulate NLRP3 expression.
Collapse
Affiliation(s)
- Kun Chi
- Department of Nephrology, Chinese PLA General Hospital, Medical School of Chinese PLA, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Beijing, China
| | - Xiaodong Geng
- Department of Nephrology, Chinese PLA General Hospital, Medical School of Chinese PLA, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Beijing, China.,Beidaihe Rehabilitation and Recuperation Center, Chinese People's Liberation Army Joint Logistics Support Force Qinhuangdao, Qinhuangdao, China
| | - Chao Liu
- Department of Nephrology, Chinese PLA General Hospital, Medical School of Chinese PLA, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Beijing, China
| | - Yang Zhang
- Department of Nephrology, Chinese PLA General Hospital, Medical School of Chinese PLA, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Beijing, China
| | - Jie Cui
- Beidaihe Rehabilitation and Recuperation Center, Chinese People's Liberation Army Joint Logistics Support Force Qinhuangdao, Qinhuangdao, China
| | - GuangYan Cai
- Department of Nephrology, Chinese PLA General Hospital, Medical School of Chinese PLA, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Beijing, China
| | - Xiangmei Chen
- Department of Nephrology, Chinese PLA General Hospital, Medical School of Chinese PLA, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Beijing, China
| | - Fangfang Wang
- Department of Cardiology and Institute of Vascular Medicine, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing Key Laboratory of Cardiovascular Receptors Research., Peking University Third Hospital, Beijing, 100191, China
| | - Quan Hong
- Department of Nephrology, Chinese PLA General Hospital, Medical School of Chinese PLA, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Beijing, China
| |
Collapse
|
19
|
Zhang L, Yang P, Wang J, Liu Q, Wang T, Wang Y, Lin F. MiR-22 regulated T cell differentiation and hepatocellular carcinoma growth by directly targeting Jarid2. Am J Cancer Res 2021; 11:2159-2173. [PMID: 34094675 PMCID: PMC8167680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 12/21/2020] [Indexed: 06/12/2023] Open
Abstract
MiR-22 has been demonstrated to inhibits tumor growth in several cancers. However, its function in the tumor microenvironment is still unclear, especially for T cell differentiation. Here, miR-22 expression in the circulating T cells from hepatocellular carcinoma (HCC) patients and healthy controls was analyzed with quantitative polymerase chain reaction (qPCR). Diethylnitrosamine (DEN)/phenobarbital (PB)-mediated primary HCC and Hepa1-6 subcutaneous tumor mouse models were established and subjected to lenti-miR-22 injection. Mice immunoreconstituted with miR-22-overexpressing T cells were employed to investigate the antitumor effect of miR-22 in mice. Luciferase assay, immunofluorescent staining, in vitro Th17 cell differentiation assay, and rescue experiments were employed to investigate the mechanism underlying the miR-22-mediated regulation of Th17 cell differentiation and liver tumor growth. Results confirmed the dramatic downregulation of miR-22 expression in malignant tissues and circulating T cells from patients with HCC. MiR-22 expression correlated with good prognosis of patients. Overexpression of miR-22 impaired the DEN/PB-induced primary HCC formation and the growth of Hepa1-6 subcutaneous tumors by promoting Th17 differentiation. Injection of miR-22-overexpressing T cells in irradiated mice resulted in the inhibition of Hepa1-6 subcutaneous tumor growth via Th17 differentiation promotion. MiR-22 could directly bind to Jarid2, which played an important role during the miR-22-mediated regulation of Th17 differentiation. Taken together, our study expands the understanding of miR-22 function and provides a therapy target for HCC.
Collapse
Affiliation(s)
- Lian Zhang
- Department of Oncology, Affiliated Eighth People's Hospital, Jiangsu University Shanghai, China
| | - Ping Yang
- Department of Oncology, Affiliated Eighth People's Hospital, Jiangsu University Shanghai, China
| | - Jing Wang
- Department of Oncology, Affiliated Eighth People's Hospital, Jiangsu University Shanghai, China
| | - Qi Liu
- Department of Oncology, Affiliated Eighth People's Hospital, Jiangsu University Shanghai, China
| | - Tian Wang
- Department of Oncology, Affiliated Eighth People's Hospital, Jiangsu University Shanghai, China
| | - Yaling Wang
- Department of Oncology, Affiliated Eighth People's Hospital, Jiangsu University Shanghai, China
| | - Feng Lin
- Department of Oncology, Affiliated Eighth People's Hospital, Jiangsu University Shanghai, China
| |
Collapse
|
20
|
Nejati K, Alivand M, Arabzadeh A. MicroRNA-22 in female malignancies: Focusing on breast, cervical, and ovarian cancers. Pathol Res Pract 2021; 223:153452. [PMID: 33993061 DOI: 10.1016/j.prp.2021.153452] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 04/15/2021] [Accepted: 04/16/2021] [Indexed: 12/11/2022]
Abstract
MicroRNAs (miRNAs), a novelty-defined class of regulatory genes, have revolutionized principles of classical bimolecular. These RNAs regulate the expression of a gene through inhibition of translational initiation or targeting mRNAs for degradation. MiRNAs act in several biological operations, including proliferation, differentiation, and cell death, and their expression is often abnormal in human diseases such as cancer. In recent years, miR-22 has attracted much attention from researchers. Its expression is downregulated in female malignancies such as breast, cervical, and ovarian cancers, exhibiting that miR-22 plays a tumor-suppressive function in these cancers. Also, different reports exist about the involvement of miR-22 in non-tumor diseases. In the present review, we report the results of performed studies on the potential roles of miR-22 in female malignancies with a focus on breast, cervical, and ovarian cancers. Also, we summary its predicted target genes in various cancers. In conclusion, it is effective for researchers to understand the role of miR-22 in different cellular operations.
Collapse
Affiliation(s)
- Kazem Nejati
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - MohammadReza Alivand
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - AmirAhmad Arabzadeh
- Department of Surgery, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran.
| |
Collapse
|
21
|
Wang R, Xu Y, Niu X, Fang Y, Guo D, Chen J, Zhu H, Dong J, Zhao R, Wang Y, Qi B, Ren G, Li X, Liu L, Zhang M. MiR-22 Inhibition Alleviates Cardiac Dysfunction in Doxorubicin-Induced Cardiomyopathy by Targeting the sirt1/PGC-1α Pathway. Front Physiol 2021; 12:646903. [PMID: 33868015 PMCID: PMC8047466 DOI: 10.3389/fphys.2021.646903] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 02/22/2021] [Indexed: 11/13/2022] Open
Abstract
Doxorubicin (DOX) cardiotoxicity is a life-threatening side effect that leads to a poor prognosis in patients receiving chemotherapy. We investigated the role of miR-22 in doxorubicin-induced cardiomyopathy and the underlying mechanism in vivo and in vitro. Specifically, we designed loss-of-function and gain-of-function experiments to identify the role of miR-22 in doxorubicin-induced cardiomyopathy. Our data suggested that inhibiting miR-22 alleviated cardiac fibrosis and cardiac dysfunction induced by doxorubicin. In addition, inhibiting miR-22 mitigated mitochondrial dysfunction through the sirt1/PGC-1α pathway. Knocking out miR-22 enhanced mitochondrial biogenesis, as evidenced by increased PGC-1α, TFAM, and NRF-1 expression in vivo. Furthermore, knocking out miR-22 rescued mitophagy, which was confirmed by increased expression of PINK1 and parkin and by the colocalization of LC3 and mitochondria. These protective effects were abolished by overexpressing miR-22. In conclusion, miR-22 may represent a new target to alleviate cardiac dysfunction in doxorubicin-induced cardiomyopathy and improve prognosis in patients receiving chemotherapy.
Collapse
Affiliation(s)
- Runze Wang
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China.,Department of Hematology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Yuerong Xu
- Department of Orthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Xiaolin Niu
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Yexian Fang
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Dong Guo
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Jiangwei Chen
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Hanzhao Zhu
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Jiaying Dong
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Ran Zhao
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Ying Wang
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Bingchao Qi
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Gaotong Ren
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Xue Li
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Li Liu
- Department of Hematology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Mingming Zhang
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| |
Collapse
|
22
|
Wang R, Xu Y, Zhang W, Fang Y, Yang T, Zeng D, Wei T, Liu J, Zhou H, Li Y, Huang ZP, Zhang M. Inhibiting miR-22 Alleviates Cardiac Dysfunction by Regulating Sirt1 in Septic Cardiomyopathy. Front Cell Dev Biol 2021; 9:650666. [PMID: 33869205 PMCID: PMC8047209 DOI: 10.3389/fcell.2021.650666] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 03/05/2021] [Indexed: 01/20/2023] Open
Abstract
High morbidity and mortality are the most typical characteristics of septic cardiomyopathy. We aimed to reveal the role of miR-22 in septic cardiomyopathy and to explore the underlying mechanisms. miR-22 cardiac-specific knockout (miR-22cKO) mice and miR-22 cardiac-specific transgenic (miR-22cOE) mice were subjected to a cecal ligation and puncture (CLP) operation, while a sham operation was used in the control group. The echocardiogram results suggested that miR-22cKO CLP mice cardiac dysfunction was alleviated. The serum LDH and CK-MB were reduced in the miR-22cKO CLP mice. As expected, there was reduced apoptosis, increased autophagy and alleviated mitochondrial dysfunction in the miR-22cKO CLP mice, while it had contrary role in the miR-22cOE group. Inhibiting miR-22 promoted autophagy by increasing the LC3II/GAPDH ratio and decreasing the p62 level. Additionally, culturing primary cardiomyocytes with lipopolysaccharide (LPS) simulated sepsis-induced cardiomyopathy in vitro. Inhibiting miR-22 promoted autophagic flux confirmed by an increased LC3II/GAPDH ratio and reduced p62 protein level under bafilomycin A1 conditions. Knocking out miR-22 may exert a cardioprotective effect on sepsis by increasing autophagy and decreasing apoptosis via sirt1. Our results revealed that targeting miR-22 may become a new strategy for septic cardiomyopathy treatment.
Collapse
Affiliation(s)
- Runze Wang
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China.,Department of Hematology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Yuerong Xu
- Department of Orthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Wei Zhang
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Yexian Fang
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Tiqun Yang
- Department of Cardiology, Center for Translational Medicine, The First Affiliated Hospital, Institute of Precision Medicine, Sun Yat-sen University, Guangzhou, China.,NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Di Zeng
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Ting Wei
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Jing Liu
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Haijia Zhou
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Yan Li
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Zhan-Peng Huang
- Department of Cardiology, Center for Translational Medicine, The First Affiliated Hospital, Institute of Precision Medicine, Sun Yat-sen University, Guangzhou, China.,NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Mingming Zhang
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| |
Collapse
|
23
|
Zhu C, He X, Chen K, Huang Z, Yao A, Tian X, You Y, Zeng M. LncRNA NBR2 aggravates hepatoblastoma cell malignancy and promotes cell proliferation under glucose starvation through the miR-22/TCF7 axis. Cell Cycle 2021; 20:575-590. [PMID: 33651649 DOI: 10.1080/15384101.2021.1885236] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Hepatoblastoma (HB) is the most commonly seen pediatric liver malignancy. With frequent mutations in CTNNB1 gene that encodes β-catenin, hepatoblastoma has been considered as a Wnt/β-catenin-activated malignant tumor. Altered glucose metabolism upon nutrient deprivation (glucose starvation) might also be a critical event in hepatoblastoma carcinogenesis. The present study provides a lncRNA NBR2/miR-22/TCF7 axis modulating proliferation, invasion, migration, and apoptosis of hepatoblastoma cells upon glucose starvation through Wnt and downstream TCF7 signaling pathways. The expression of NBR2 is significantly increased within hepatoblastoma tissue samples; moreover, under incubation with 0 mM glucose (glucose starvation), NBR2 expression is significantly upregulated. NBR2 silencing not only inhibited hepatoblastoma cell viability, invasion, and migration under normal culture condition but also promoted the cell apoptosis under glucose starvation. NBR2 silencing in hepatoblastoma cells also decreased TCF7 mRNA expression and TCF7 protein levels, as well as the protein levels of the cell cycle, glucose entrapment, and EMT markers. miR-22 is directly bound to both NBR2 and TCF7; lncRNA NBR2 counteracted miR-22-mediated repression on TCF7 via acting as a ceRNA. The effects of NBR2 silencing on TCF7 expression, hepatoblastoma cell phenotype, and cell cycle, glucose entrapment, and EMT markers were all significantly reversed by miR-22 inhibition. In conclusion, lncRNA NBR2 aggravates hepatoblastoma cell malignancy through competing with TCF7 for miR-22 binding, therefore counteracting miR-22-mediated repression on TCF7. LncRNA NBR2 might be a promising target to inhibit hepatoblastoma cell proliferation under glucose starvation.
Collapse
Affiliation(s)
- Chengguang Zhu
- Department of Pediatric Hematology and Oncology, Children's Medical Center of Hunan Provincial People's Hospital (The First-Affiliated Hospital of Hunan Normal University), Changsha, China
| | - Xiangling He
- Department of Pediatric Hematology and Oncology, Children's Medical Center of Hunan Provincial People's Hospital (The First-Affiliated Hospital of Hunan Normal University), Changsha, China
| | - Keke Chen
- Department of Pediatric Hematology and Oncology, Children's Medical Center of Hunan Provincial People's Hospital (The First-Affiliated Hospital of Hunan Normal University), Changsha, China
| | - Zhijun Huang
- Department of Pediatric Hematology and Oncology, Children's Medical Center of Hunan Provincial People's Hospital (The First-Affiliated Hospital of Hunan Normal University), Changsha, China
| | - Anqi Yao
- Department of Pediatric Hematology and Oncology, Children's Medical Center of Hunan Provincial People's Hospital (The First-Affiliated Hospital of Hunan Normal University), Changsha, China
| | - Xin Tian
- Department of Pediatric Hematology and Oncology, Children's Medical Center of Hunan Provincial People's Hospital (The First-Affiliated Hospital of Hunan Normal University), Changsha, China
| | - Yalan You
- Department of Pediatric Hematology and Oncology, Children's Medical Center of Hunan Provincial People's Hospital (The First-Affiliated Hospital of Hunan Normal University), Changsha, China
| | - Minhui Zeng
- Department of Pediatric Hematology and Oncology, Children's Medical Center of Hunan Provincial People's Hospital (The First-Affiliated Hospital of Hunan Normal University), Changsha, China
| |
Collapse
|
24
|
Vesuna F, Lisok A, van Diest P, Raman V. Twist activates miR-22 to suppress estrogen receptor alpha in breast cancer. Mol Cell Biochem 2021; 476:2295-306. [PMID: 33582945 DOI: 10.1007/s11010-021-04065-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 01/11/2021] [Indexed: 12/16/2022]
Abstract
TWIST1 (Twist) is a basic helix-loop-helix transcription factor that is overexpressed in many cancers and promotes tumor cell invasion, metastasis, and recurrence. In this study, we demonstrate that Twist upregulates expression of microRNA 22 (miR-22) which, in turn, downregulates estrogen receptor alpha (ER) expression in breast cancer. Initial analysis of miR-22 and Twist expression in a panel of breast cancer cell lines showed a direct correlation between Twist and miR-22 levels with miR-22 being highly expressed in ER negative cell lines. Overexpressing Twist caused increased miR-22 levels while downregulating it led to decreased miR-22 expression. To characterize the upstream promoter region of miR-22, we utilized rapid amplification of cDNA ends and identified the transcription start site and the putative promoter region of miR-22. Mechanistically, we determined that Twist, in combination with HDAC1 and DNMT3B, transcriptionally upregulates miR-22 expression by binding to E-boxes in the proximal miR-22 promoter. We also established that miR-22 causes an increase in growth in 3D but not 2D cultures. Importantly, we observed a direct correlation between increased breast cancer grade and Twist and miR-22 expression. We also identified two potential miR-22 binding sites in the 3'-UTR region of ER and confirmed by promoter assays that miR-22 regulates ER expression by binding to both target sites. These results reveal a novel pathway of ER suppression by Twist through miR-22 activation that could potentially promote the ER negative phenotype in breast cancers.
Collapse
|
25
|
Lou P, Bi X, Tian Y, Li G, Kang Q, Lv C, Song Y, Xu J, Sheng X, Yang X, Liu R, Meng Q, Ren F, Plikus MV, Liang B, Zhang B, Guo H, Yu Z. MiR-22 modulates brown adipocyte thermogenesis by synergistically activating the glycolytic and mTORC1 signaling pathways. Am J Cancer Res 2021; 11:3607-3623. [PMID: 33664851 PMCID: PMC7914365 DOI: 10.7150/thno.50900] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 12/08/2020] [Indexed: 12/17/2022] Open
Abstract
Background: Brown adipose tissue (BAT) dissipates chemical energy as heat and has the potential to be a protective strategy to prevent obesity. microRNAs (miRNAs) are emerging as important posttranscriptional factors affecting the thermogenic function of BAT. However, the regulatory mechanism underlying miRNA-mediated energy metabolism in BAT is not fully understood. Here, we explored the roles of miR-22 in BAT thermogenesis and energy metabolism. Methods: Using global and conditional knockout mice as in vivo models and primary brown adipocytes as an in vitro system, we investigated the function of miR-22 in BAT thermogenesis in vivo and in vitro. Results: miR-22 expression was upregulated in BAT in response to cold exposure and during brown preadipocyte differentiation. Both global and conditional knockout mice displayed BAT whitening, impaired cold tolerance, and decreased BAT thermogenesis. Moreover, we found that miR-22 deficiency impaired BAT glycolytic capacity, which is critical for thermogenesis. The mechanistic results revealed that miR-22 activated the mTORC1 signaling pathway by directly suppressing Tsc1 and concomitantly directly suppressing Hif1an, an inhibitor of Hif1α, which promotes glycolysis and maintains thermogenesis. Conclusions: Our findings identify miR-22 as a critical regulator in the control of thermogenesis in BAT and as a potential therapeutic target for human metabolic disorders.
Collapse
|
26
|
Gorur A, Bayraktar R, Ivan C, Mokhlis HA, Bayraktar E, Kahraman N, Karakas D, Karamil S, Kabil NN, Kanlikilicer P, Aslan B, Tamer L, Wang Z, Cristini V, Lopez-Berestein G, Calin G, Ozpolat B. ncRNA therapy with miRNA-22-3p suppresses the growth of triple-negative breast cancer. Mol Ther Nucleic Acids 2021; 23:930-943. [PMID: 33614241 PMCID: PMC7868999 DOI: 10.1016/j.omtn.2021.01.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 01/14/2021] [Indexed: 12/14/2022]
Abstract
Deregulation of noncoding RNAs, including microRNAs (miRs), is implicated in the pathogenesis of many human cancers, including breast cancer. Through extensive analysis of The Cancer Genome Atlas, we found that expression of miR-22-3p is markedly lower in triple-negative breast cancer (TNBC) than in normal breast tissue. The restoration of miR-22-3p expression led to significant inhibition of TNBC cell proliferation, colony formation, migration, and invasion. We demonstrated that miR-22-3p reduces eukaryotic elongation factor 2 kinase (eEF2K) expression by directly binding to the 3' untranslated region of eEF2K mRNA. Inhibition of EF2K expression recapitulated the effects of miR-22-3p on TNBC cell proliferation, motility, invasion, and suppression of phosphatidylinositol 3-kinase/Akt and Src signaling. Systemic administration of miR-22-3p in single-lipid nanoparticles significantly suppressed tumor growth in orthotopic MDA-MB-231 and MDA-MB-436 TNBC models. Evaluation of the tumor response, following miR-22-3p therapy in these models using a novel mathematical model factoring in various in vivo parameters, demonstrated that the therapy is highly effective against TNBC. These findings suggest that miR-22-3p functions as a tumor suppressor by targeting clinically significant oncogenic pathways and that miR-22-3p loss contributes to TNBC growth and progression. The restoration of miR-22-3p expression is a potential novel noncoding RNA-based therapy for TNBC.
Collapse
Affiliation(s)
- Aysegul Gorur
- Department of Experimental Therapeutics, Unit 1950, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA.,Department of Biochemistry, School of Medicine, Mersin University, Mersin, Turkey
| | - Recep Bayraktar
- Department of Experimental Therapeutics, Unit 1950, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Cristina Ivan
- Department of Experimental Therapeutics, Unit 1950, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA.,Center for RNA Interference and Non-Coding RNAs, Unit 2080, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Hamada Ahmed Mokhlis
- Department of Experimental Therapeutics, Unit 1950, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA.,Department of Pharmacology and Toxicology, Faculty of Pharmacy, The University of Al-Azhar, Cairo, Egypt
| | - Emine Bayraktar
- Department of Experimental Therapeutics, Unit 1950, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Nermin Kahraman
- Department of Experimental Therapeutics, Unit 1950, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Didem Karakas
- Department of Experimental Therapeutics, Unit 1950, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Selda Karamil
- Department of Experimental Therapeutics, Unit 1950, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Nashwa N Kabil
- Department of Experimental Therapeutics, Unit 1950, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Pinar Kanlikilicer
- Department of Experimental Therapeutics, Unit 1950, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Burcu Aslan
- Department of Experimental Therapeutics, Unit 1950, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Lulufer Tamer
- Department of Biochemistry, School of Medicine, Mersin University, Mersin, Turkey
| | - Zhihui Wang
- Mathematics in Medicine, Houston Methodist Research Institute, 6565 Fannin Street, Houston, TX 77030, USA
| | - Vittorio Cristini
- Mathematics in Medicine, Houston Methodist Research Institute, 6565 Fannin Street, Houston, TX 77030, USA
| | - Gabriel Lopez-Berestein
- Department of Experimental Therapeutics, Unit 1950, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA.,Center for RNA Interference and Non-Coding RNAs, Unit 2080, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - George Calin
- Department of Experimental Therapeutics, Unit 1950, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA.,Center for RNA Interference and Non-Coding RNAs, Unit 2080, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Bulent Ozpolat
- Department of Experimental Therapeutics, Unit 1950, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA.,Center for RNA Interference and Non-Coding RNAs, Unit 2080, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| |
Collapse
|
27
|
Zhou L, Li FF, Wang SM. Circ-ITCH restrains the expression of MMP-2, MMP-9 and TNF-α in diabetic retinopathy by inhibiting miR-22. Exp Mol Pathol 2021; 118:104594. [PMID: 33309614 DOI: 10.1016/j.yexmp.2020.104594] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 11/10/2020] [Accepted: 12/06/2020] [Indexed: 02/07/2023]
Abstract
Diabetic retinopathy (DR), the most frequent complication of diabetes mellitus, is the principal cause of acquired blindness worldwide. Although the roles of circRNAs have been extensively explored, the detailed physiological and pathological functions of circRNAs in DR are less understood. Here, we studied the biological effects of circ-ITCH in diabetic retinal pigment epithelial cells (RPEs) and explored the underlying mechanisms. As our results shown, the RNA expression of circ-ITCH was significantly lower in RPEs isolated from diabetic rats than they were in those isolated from normal rats. While diabetes induced an increase in MMP-2, MMP-9 and TNF-α in RPEs, circ-ITCH overexpression exerted an inhibitory on these increases and knockdown of circ-ITCH reversed the inhibitory. In addition, increased expression of miR-22 in RPEs correlated with diabetes and downregulation of circ-ITCH. Remarkably, in the presence of miR-22 mimics, the effects of circ-ITCH on the MMP-2 and MMP-9 were both antagonized. Collectively, our data supports a cellular signaling cascade in which circ-ITCH-inhibited miR-22 activity modulates the expression of MMP-2, MMP-9 and TNF-α in DR.
Collapse
|
28
|
Zhang M, Li Y, Xie H, Chen J, Liu S. Curcumin inhibits proliferation, migration and neointimal formation of vascular smooth muscle via activating miR-22. Pharm Biol 2020; 58:610-619. [PMID: 32631202 PMCID: PMC8641690 DOI: 10.1080/13880209.2020.1781904] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 05/15/2020] [Accepted: 06/08/2020] [Indexed: 06/11/2023]
Abstract
Context: Curcumin has antitumor, antioxidative, anti-inflammatory, and anti-proliferative properties.Objective: To investigate the role of miR-22 during curcumin-induced changes in vascular smooth muscle cells (VSMC) and neointima formation in balloon-injured rat abdominal aorta.Materials and methods: Sprague-Dawley rats were randomised to the sham-operated (n = 10), operated control (injured, n = 10), and curcumin treatment (n = 10) groups. miR-22 expression was determined by real-time PCR. SP1 was assessed by western blot and real-time PCR. Rat aortic smooth muscle A7r5 cells were used to determine VSMC proliferation and migration, which were measured by the MTS, EdU staining, Transwell, and wound healing assays.Results: miR-22 levels declined following arterial balloon injury in vivo (48% at 3d, p < 0.05) and serum stimulation in vitro (45% at 24 h, p < 0.01). Functional studies revealed that miR-22 negatively regulated the proliferation and migration of VSMCs by directly targeting the SP1 transcription factor in VSMCs. Curcumin increased the expression of miR-22 (81%, p < 0.05) and decreased the protein expression of SP1 in VSMCs (25%, p < 0.05). miR-22 inhibition was found to attenuate the effects of curcumin on VSMC functions. Curcumin increased miR-22 (46%, p < 0.01), decreased the SP1 protein (19%, p < 0.05), and inhibited vascular neointimal area (48%, p < 0.01) in vivo.Discussion: The miR-22/SP1 pathway is involved in the protective role of curcumin during arterial balloon injury, but the mechanisms remain unclear.Conclusion: miR-22 is involved in the inhibitory effects of curcumin on VSMCs' proliferation, migration and neointima hyperplasia after arterial balloon injury in rats. Curcumin could be used to prevent neointimal hyperplasia after angioplasty.
Collapse
Affiliation(s)
- Minghua Zhang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
- Cardiovascular Department, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yuntian Li
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
- Cardiovascular Center, 305 Hospital of Chinese People′s Liberation Army, Beijing, China
| | - Hui Xie
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Jing Chen
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Shiming Liu
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
29
|
Yang Z, Qin W, Huo J, Zhuo Q, Wang J, Wang L. MiR-22 modulates the expression of lipogenesis-related genes and promotes hepatic steatosis in vitro. FEBS Open Bio 2020; 11:322-332. [PMID: 33159388 PMCID: PMC7780092 DOI: 10.1002/2211-5463.13026] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 10/15/2020] [Accepted: 11/04/2020] [Indexed: 12/11/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is highly correlated with obesity, and lifestyle changes to reduce weight remain the main therapeutic approach. The noncoding RNA miR-22 has previously been reported to be highly abundant in the sera of NAFLD patients. In addition, miR-22 directly targets peroxisome proliferative-activated receptor, Pgc-1α, peroxisome proliferator-activated receptor α, and sirtuin 1 (Sirt1), which are important factors involved in fatty acid metabolism. Given that miR-22 directly targets genes involved in the control of metabolism and obesity, we investigated whether miR-22 contributes to metabolic alterations induced by obesity. We observed increased expression of miR-22, decreased expression of Sirt1, and alterations in the expression of adipogenesis-related genes in a mouse model of obesity and a human hepatocyte cell line. We identified that miR-22 and the 3'-UTR of Sirt1 are complementary. Mutation of the complementary fragment abolishes the ability of miR-22 to regulate the Sirt1 gene. Furthermore, treatment of hepatic steatosis cells with miR-22 mimics or inhibitors showed that miR-22 can promote hepatic steatosis, and miR-22 inhibitors effectively reduced triglyceride levels without affecting cell activity. Finally, we validated that miR-22 has similar effects on downstream lipid metabolism-related genes. Our data reveal the pathways and mechanisms through which miR-22 regulates lipid metabolism and suggest that miR-22 inhibitors may have potential as candidate drugs for NAFLD and obesity.
Collapse
|