1
|
Wu X, Yuan C, Pan J, Zhou Y, Pan X, Kang J, Ren L, Gong L, Li Y. CXCL9, IL2RB, and SPP1, potential diagnostic biomarkers in the co-morbidity pattern of atherosclerosis and non-alcoholic steatohepatitis. Sci Rep 2024; 14:16364. [PMID: 39013959 PMCID: PMC11252365 DOI: 10.1038/s41598-024-66287-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 07/01/2024] [Indexed: 07/18/2024] Open
Abstract
Non-alcoholic steatohepatitis (NASH) is a hepatocyte inflammation based on hepatocellular steatosis, yet there is no effective drug treatment. Atherosclerosis (AS) is caused by lipid deposition in the endothelium, which can lead to various cardiovascular diseases. NASH and AS share common risk factors, and NASH can also elevate the risk of AS, causing a higher morbidity and mortality rate for atherosclerotic heart disease. Therefore, timely detection and diagnosis of NASH and AS are particularly important. In this study, differential gene expression analysis and weighted gene co-expression network analysis were performed on the AS (GSE100927) and NASH (GSE89632) datasets to obtain common crosstalk genes, respectively. Then, candidate Hub genes were screened using four topological algorithms and externally validated in the GSE43292 and GSE63067 datasets to obtain Hub genes. Furthermore, immune infiltration analysis and gene set variation analysis were performed on the Hub genes to explore the underlying mechanisms. The DGIbd database was used to screen candidate drugs for AS and NASH. Finally, a NASH model was constructed using free fatty acid-induced human L02 cells, an AS model was constructed using lipopolysaccharide-induced HUVECs, and a co-morbidity model was constructed using L02 cells and HUVECs to verify Hub gene expression. The result showed that a total of 113 genes common to both AS and NASH were identified as crosstalk genes, and enrichment analysis indicated that these genes were mainly involved in the regulation of immune and metabolism-related pathways. 28 candidate Hub genes were screened according to four topological algorithms, and CXCL9, IL2RB, and SPP1 were identified as Hub genes after in vitro experiments and external dataset validation. The ROC curves and SVM modeling demonstrated the good diagnostic efficacy of these three Hub genes. In addition, the Hub genes are strongly associated with immune cell infiltration, especially macrophages and γ-δ T cell infiltration. Finally, five potential therapeutic drugs were identified. has-miR-185 and hsa-miR-335 were closely related to AS and NASH. This study demonstrates that CXCL9, IL2RB, and SPP1 may serve as potential biomarkers for the diagnosis of the co-morbidity patterns of AS and NASH and as potential targets for drug therapy.
Collapse
Affiliation(s)
- Xize Wu
- Liaoning University of Traditional Chinese Medicine, No. 79 Chongshan East Road, Huanggu District, Shenyang, 110847, Liaoning, China
- Nantong Hospital of Traditional Chinese Medicine, Nantong Hospital Affiliated to Nanjing University of Chinese Medicine, Nantong, 226000, Jiangsu, China
| | - Changbin Yuan
- Liaoning University of Traditional Chinese Medicine, No. 79 Chongshan East Road, Huanggu District, Shenyang, 110847, Liaoning, China
| | - Jiaxiang Pan
- The Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, 110032, Liaoning, China
| | - Yi Zhou
- Liaoning University of Traditional Chinese Medicine, No. 79 Chongshan East Road, Huanggu District, Shenyang, 110847, Liaoning, China
| | - Xue Pan
- Liaoning University of Traditional Chinese Medicine, No. 79 Chongshan East Road, Huanggu District, Shenyang, 110847, Liaoning, China
- Dazhou Vocational College of Chinese Medicine, Dazhou, 635000, Sichuan, China
| | - Jian Kang
- Liaoning University of Traditional Chinese Medicine, No. 79 Chongshan East Road, Huanggu District, Shenyang, 110847, Liaoning, China
| | - Lihong Ren
- Nantong Hospital of Traditional Chinese Medicine, Nantong Hospital Affiliated to Nanjing University of Chinese Medicine, Nantong, 226000, Jiangsu, China.
| | - Lihong Gong
- Liaoning University of Traditional Chinese Medicine, No. 79 Chongshan East Road, Huanggu District, Shenyang, 110847, Liaoning, China.
- The Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, 110032, Liaoning, China.
- Liaoning Provincial Key Laboratory of TCM Geriatric Cardio-Cerebrovascular Diseases, Shenyang, 110847, Liaoning, China.
| | - Yue Li
- The Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, 110032, Liaoning, China.
- Liaoning Provincial Key Laboratory of TCM Geriatric Cardio-Cerebrovascular Diseases, Shenyang, 110847, Liaoning, China.
| |
Collapse
|
2
|
Manwani B, Brathaban N, Baqai A, Munshi Y, Ahnstedt HW, Zhang M, Arkelius K, Llera T, Amorim E, Elahi FM, Singhal NS. Small RNA signatures of acute ischemic stroke in L1CAM positive extracellular vesicles. Sci Rep 2024; 14:13560. [PMID: 38866905 PMCID: PMC11169361 DOI: 10.1038/s41598-024-63633-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 05/30/2024] [Indexed: 06/14/2024] Open
Abstract
L1CAM-positive extracellular vesicles (L1EV) are an emerging biomarker that may better reflect ongoing neuronal damage than other blood-based biomarkers. The physiological roles and regulation of L1EVs and their small RNA cargoes following stroke is unknown. We sought to characterize L1EV small RNAs following stroke and assess L1EV RNA signatures for diagnosing stroke using weighted gene co-expression network analysis and random forest (RF) machine learning algorithms. Interestingly, small RNA sequencing of plasma L1EVs from patients with stroke and control patients (n = 28) identified micro(mi)RNAs known to be enriched in the brain. Weighted gene co-expression network analysis (WGCNA) revealed small RNA transcript modules correlated to diagnosis, initial NIH stroke scale, and age. L1EV RNA signatures associated with the diagnosis of AIS were derived from WGCNA and RF classification. These small RNA signatures demonstrated a high degree of accuracy in the diagnosis of AIS with an area under the curve (AUC) of the signatures ranging from 0.833 to 0.932. Further work is necessary to understand the role of small RNA L1EV cargoes in the response to brain injury, however, this study supports the utility of L1EV small RNA signatures as a biomarker of stroke.
Collapse
Affiliation(s)
- Bharti Manwani
- Department of Neurology, University of Texas Health Science Center, Houston, TX, 77030, USA
| | - Nivetha Brathaban
- Department of Neurology, University of California-San Francisco, San Francisco, CA, 94158, USA
| | - Abiya Baqai
- Department of Neurology, University of California-San Francisco, San Francisco, CA, 94158, USA
| | - Yashee Munshi
- Department of Neurology, University of Texas Health Science Center, Houston, TX, 77030, USA
| | - Hilda W Ahnstedt
- Department of Neurology, University of Texas Health Science Center, Houston, TX, 77030, USA
| | - Mengqi Zhang
- Department of Neurology, University of California-San Francisco, San Francisco, CA, 94158, USA
| | - Kajsa Arkelius
- Department of Neurology, University of California-San Francisco, San Francisco, CA, 94158, USA
| | - Ted Llera
- Department of Neurology, University of California-San Francisco, San Francisco, CA, 94158, USA
| | - Edilberto Amorim
- Department of Neurology, University of California-San Francisco, San Francisco, CA, 94158, USA
| | - Fanny M Elahi
- Department of Neurology, University of California-San Francisco, San Francisco, CA, 94158, USA
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- James J. Peters Department of Veterans Affairs Medical Center, Bronx, NY, USA
| | - Neel S Singhal
- Department of Neurology, University of California-San Francisco, San Francisco, CA, 94158, USA.
- Neurology Service, San Francisco Veterans Affairs Medical Center, San Francisco, CA, 94150, USA.
| |
Collapse
|
3
|
Wei Q, Xiao Y, Du L, Li Y. Advances in Nanoparticles in the Prevention and Treatment of Myocardial Infarction. Molecules 2024; 29:2415. [PMID: 38893291 PMCID: PMC11173599 DOI: 10.3390/molecules29112415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/17/2024] [Accepted: 05/17/2024] [Indexed: 06/21/2024] Open
Abstract
Myocardial infarction (MI) is one of the most prevalent types of cardiovascular disease. During MI, myocardial cells become ischemic and necrotic due to inadequate blood perfusion, leading to irreversible damage to the heart. Despite the development of therapeutic strategies for the prevention and treatment of MI, their effects are still unsatisfactory. Nanoparticles represent a new strategy for the pre-treatment and treatment of MI, and novel multifunctional nanoparticles with preventive and therapeutic capabilities hold promise for the prevention and treatment of this disease. This review summarizes the common types and properties of nanoparticles, and focuses on the research progress of nanoparticles for the prevention and treatment of MI.
Collapse
Affiliation(s)
| | | | | | - Ya Li
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China; (Q.W.); (Y.X.); (L.D.)
| |
Collapse
|
4
|
Hu HL, Zheng HX, Yuan N, Zhai CL, Chen H, Pan HH, Qian G. CircUsp9x/miR-599/stim1 axis regulates proliferation and migration in vascular smooth muscle cells induced by oxidized-low density lipoprotein. Clin Exp Hypertens 2023; 45:2280758. [PMID: 37963203 DOI: 10.1080/10641963.2023.2280758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 11/03/2023] [Indexed: 11/16/2023]
Abstract
Circular RNAs (circRNAs) regulate the function of vascular smooth muscle cells (VSMCs) in atherosclerosis (AS) progression. We aimed to explore the role of circUSP9X in oxidized low-density lipoprotein (ox-LDL)-induced VSMCs. Cell proliferation was assessed using cell counting kit-8 and EDU assays. Cell migration was evaluated using Transwell and wound healing assays. The interaction between circUSP9X or STIM1 and miR-599 was analyzed using dual-luciferase reporter and RNA pull-down assays. Their levels were examined using quantitative real-time PCR. CircUSP9X and STIM1 expression was increased, whereas miR-599 expression was reduced in the serum of patients with AS and ox-LDL-stimulated VSMCs. Overexpression of circUSP9X facilitated the proliferation and migration of VSMCs induced by ox-LDL. CircUSP9X sponged miR-599, which targeted STIM1. MiR-599 reversed the effects induced by circUSP9X, and STIM1 reversed the effects induced by miR-599. Taken together, CircUSP9X promoted proliferation and migration in ox-LDL-treated VSMCs via the miR-599/STIM1 axis, providing a theoretical basis for the role of circUSP9X/miR-599/STIM1 axis in AS.
Collapse
Affiliation(s)
- Hui-Lin Hu
- Jiaxing University Master Degree Cultivation Base, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Cardiology, The First Hospital of Jiaxing Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Hui-Xiu Zheng
- Jiaxing University Master Degree Cultivation Base, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Cardiology, The First Hospital of Jiaxing Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Na Yuan
- Department of Cardiology, The First Hospital of Jiaxing Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Chang-Lin Zhai
- Jiaxing University Master Degree Cultivation Base, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Cardiology, The First Hospital of Jiaxing Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Hao Chen
- Department of Cardiology, The First Hospital of Jiaxing Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Hai-Hua Pan
- Department of Cardiology, The First Hospital of Jiaxing Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Gang Qian
- Jiaxing University Master Degree Cultivation Base, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Cardiology, The First Hospital of Jiaxing Affiliated Hospital of Jiaxing University, Jiaxing, China
| |
Collapse
|
5
|
Wan X, Zhang H, Tian J, Hao P, Liu L, Zhou Y, Zhang J, Song X, Ge C. The Chains of Ferroptosis Interact in the Whole Progression of Atherosclerosis. J Inflamm Res 2023; 16:4575-4592. [PMID: 37868832 PMCID: PMC10588755 DOI: 10.2147/jir.s430885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 10/03/2023] [Indexed: 10/24/2023] Open
Abstract
Atherosclerosis (AS), a category of cardiovascular disease (CVD) that can cause other more severe disabilities, increasingly jeopardizes human health. Owing to its imperceptible and chronic symptoms, it is hard to determine the pathogenesis and precise therapeutics for AS. A novel type of programmed cell death called ferroptosis was discovered in recent years that is distinctively different from other traditional cell death pathways in morphological and biochemical aspects. Characterized by iron overload, redox disequilibrium, and accumulation of lipid hydroperoxides (L-OOH), ferroptosis influences endothelial cells, vascular smooth muscle cells (VSMCs), and macrophages, as well as inflammation, partaking in the pathology of many cardiovascular diseases such as atherosclerosis, stroke, ischemia-reperfusion injury, and heart failure. The mechanisms behind ferroptosis are so sophisticated and interwoven that many molecules involved in this procedure are unknown. This review systematically depicts the initiation and modulation of ferroptosis and summarizes the contribution of ferroptosis to AS, which may open a feasible approach for target treatment in the alleviation of AS progression.
Collapse
Affiliation(s)
- Xueqi Wan
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Huan Zhang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Jinfan Tian
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Peng Hao
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Libo Liu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Yuquan Zhou
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Jing Zhang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Xiantao Song
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Changjiang Ge
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, People’s Republic of China
| |
Collapse
|
6
|
Pordel S, Khorrami M, Saadatpour F, Rezaee D, Cho WC, Jahani S, Aghaei-Zarch SM, Hashemi E, Najafi S. The role of microRNA-185 in the pathogenesis of human diseases: A focus on cancer. Pathol Res Pract 2023; 249:154729. [PMID: 37639952 DOI: 10.1016/j.prp.2023.154729] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 07/29/2023] [Indexed: 08/31/2023]
Abstract
MicroRNAs (miRNAs) are a widely-studied class of non-coding RNAs characterized by their short length (18-25 nucleotides). The precise functions of miRNAs are not well-elucidated; however, an increasing number of studies suggest their involvement in various physiologic processes and deregulation in pathologic conditions. miRNA-185 (miR-185) is among the mostly-studied miRNAs in human diseases, which is found to play putative roles in conditions like metabolic disorders, asthma, frailty, schizophrenia, and hepatitis. Notably, many cancer studies report the downregulation of miR-185 in cell lines, tumor tissues, and plasma specimens of patients, while it demonstrates a suppressing role on the malignant properties of cancer cells in vitro and in vivo. Accordingly, miR-185 can be considered a tumor suppressor miRNA in human malignancies, while a few studies also report inconsistent findings. Being suggested as a prognostic/diagnostic biomarker, mi-185 is also found to offer clinical potentials, particularly for early diagnosis and prediction of the prognosis of cancer patients. In this review, we have outlined the studies that have evaluated the functions and clinical significance of miR-185 in different human diseases with a particular focus on cancer.
Collapse
Affiliation(s)
- Safoora Pordel
- Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Immunology and Allergy, The Persian Gulf Tropical Medicine Research Center, The Persian Gulf Biomedical Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Motahare Khorrami
- Immunology Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Saadatpour
- Pharmaceutical Biotechnology Lab, Department of Microbiology, School of Biology and Center of Excellence in Phylogeny of Living Organisms, College of Science, University of Tehran, Tehran, Iran
| | - Delsuz Rezaee
- School of Allied Medical Sciences, Ilam University of Medical Sciences, Ilam, Iran
| | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, 30 Gascoigne Road, Hong Kong, China
| | | | - Seyed Mohsen Aghaei-Zarch
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Elham Hashemi
- Department of Anatomical Sciences, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Sajad Najafi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
7
|
Choi YA, Jeong A, Woo CH, Cha SC, Park DY, Sagong M. Aqueous microRNA profiling in age-related macular degeneration and polypoidal choroidal vasculopathy by next-generation sequencing. Sci Rep 2023; 13:1274. [PMID: 36690666 PMCID: PMC9870898 DOI: 10.1038/s41598-023-28385-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 01/18/2023] [Indexed: 01/24/2023] Open
Abstract
Although many studies demonstrated the differences of clinical features, natural course, and response to treatment between typical age-related macular degeneration (AMD) and polypoidal choroidal vasculopathy (PCV), differential microRNAs (miRNAs) expression in the aqueous humor (AH) between them has not been reported yet. We investigated the roles of miRNAs in the AH of patients with typical AMD and PCV using next-generation sequencing (NGS) and quantitative PCR (qPCR). Target genes and predicted pathways of miRNAs were investigated via pathway enrichment analysis using the Kyoto Encyclopedia of Genes and Genomes database. A total of 161 miRNAs from eyes with typical AMD and 185 miRNAs from eyes with PCV were differentially expressed. 33 miRNAs were commonly upregulated, and 77 miRNAs were commonly downregulated in both typical AMD and PCV groups. Among them, hsa-miR-140-5p, hsa-miR-374c-3p, and hsa-miR-200a-5p were differentially expressed and were predicted to regulate proteoglycans in cancer, p53 signaling pathway, Hippo signaling pathway, and adherens junction. The differential expression profiles and target gene regulation networks of AH miRNAs may contribute to the development of different pathological phenotypes in typical AMD and PCV. The results of this study provide novel insights into the pathogenesis, associated prognostic biomarkers, and therapeutic targets in AMD and PCV.
Collapse
Affiliation(s)
- Yeong A Choi
- Department of Ophthalmology, Yeungnam University College of Medicine, #170 Hyunchungro, Nam-Gu, Daegu, 42415, South Korea
- Yeungnam Eye Center, Yeungnam University Hospital, Daegu, South Korea
| | - Areum Jeong
- Department of Ophthalmology, Yeungnam University College of Medicine, #170 Hyunchungro, Nam-Gu, Daegu, 42415, South Korea
- Yeungnam Eye Center, Yeungnam University Hospital, Daegu, South Korea
| | - Chang-Hoon Woo
- Department of Pharmacology, Yeungnam University College of Medicine, Daegu, South Korea
| | - Soon Cheol Cha
- Department of Ophthalmology, Yeungnam University College of Medicine, #170 Hyunchungro, Nam-Gu, Daegu, 42415, South Korea
- Yeungnam Eye Center, Yeungnam University Hospital, Daegu, South Korea
| | - Do Young Park
- Department of Ophthalmology, Yeungnam University College of Medicine, #170 Hyunchungro, Nam-Gu, Daegu, 42415, South Korea
- Yeungnam Eye Center, Yeungnam University Hospital, Daegu, South Korea
| | - Min Sagong
- Department of Ophthalmology, Yeungnam University College of Medicine, #170 Hyunchungro, Nam-Gu, Daegu, 42415, South Korea.
- Yeungnam Eye Center, Yeungnam University Hospital, Daegu, South Korea.
| |
Collapse
|
8
|
Chen D, Zhu C, Ye S, Yang Q. Curcumin ameliorates oxidized low-density lipoprotein (ox-LDL)-caused damage in human umbilical vein endothelial cells (HUVECs) through the miR-599/MYD88/NF-κB axis. Toxicol In Vitro 2022; 85:105481. [PMID: 36156291 DOI: 10.1016/j.tiv.2022.105481] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 09/16/2022] [Accepted: 09/19/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND The role of curcumin in multiple human diseases was widely reported, including arteriosclerosis (AS). We aimed to investigate the correlation between curcumin and AS-related microRNAs (miRNAs) to find out more underlying mechanism of curcumin used in AS. METHODS Cell proliferation and apoptosis were determined using CCK-8 assay, EdU staining assay, flow cytometry, and western blot for the detection of PCNA and Bax protein expression in human umbilical vein endothelial cells (HUVECs). Inflammation response was evaluated using ELISA kits, and oxidative stress was evaluated by detecting SOD activity and MDA level using the matched commercial kits. RT-qPCR analysis was applied for miR-599 and MYD88 mRNA level measurement. RESULTS Curcumin treatment and miR-599 overexpression could promote cell proliferation, and inhibit cell apoptosis, inflammation response and oxidative stress, thereby alleviating ox-LDL-induced cell damage in HUVECs. Mir-599 was lowly expressed and MYD88 was highly expressed in AS patients and AS cell model. Curcumin could modulate miR-599 to exert the protective effect on ox-LDL-caused cell damage, and miR-599 directly targeted MYD88 to alleviate ox-LDL-caused cell damage in HUVECs. Curcumin targeted miR-599 to regulate MYD88 expression, thereby inactivating the NF-κB pathway in AS cell model. CONCLUSION Our findings illustrated that curcumin exhibited anti-AS effect through the miR-599/MYD88 axis and thereby inhibiting the NF-κB pathway.
Collapse
Affiliation(s)
- Decai Chen
- Department of Vascular Surgery, Nanyang First People's Hospital affiliated to Henan University, Nanyang 473012, China
| | - Chongmei Zhu
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Shouwan Ye
- Department of Thyroid and Breast Surgery, Nanyang First People's Hospital affiliated to Henan University, Nanyang 473012, China
| | - Qiong Yang
- General Surgery, Cancer Center, Department of Breast Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou 310014, China.
| |
Collapse
|
9
|
Du N, Li M, Yang D. Hsa_circRNA_102541 regulates the development of atherosclerosis by targeting miR-296-5p/PLK1 pathway. Ir J Med Sci 2022; 191:1153-1159. [PMID: 34251586 DOI: 10.1007/s11845-021-02708-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 06/26/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND Cardiovascular disorders pose great threat to public health. As a common type of cardiovascular disease, atherosclerosis is characterized by high morbidity and mortality/recurrence rate. However, the pathogenesis of atherosclerosis is complex and not fully understood. The aim of this study was to investigate the influences of hsa_circRNA_102541 (circ_102541) on proliferation and apoptosis of HUVEC cells and to identify the underlying mechanisms. METHODS RT-PCR was used to determine the expression levels of circ_102541, miR-296-5p, and PLK1 in atherosclerosis and healthy blood samples. Following the transfection with sh-circ_102541, LV-circ_102541, miR-296-5p mimics, miR-296-5p inhibitors, and si-PLK1, cell proliferation was evaluated using CCK8 assay; cell apoptosis was determined by flow cytometry; dual luciferase assay was performed to examine the interaction between abovementioned molecules. The levels of associated markers including PCNA and caspase-3 were assessed by western blotting and RT-qPCR. RESULTS The expression of circRNA_102541 and PLK1 were significantly elevated in atherosclerosis specimens, where the level of miR-296-5p was reduced. Furthermore, circRNA_102541 could bind miR-296-5p and subsequently target PLK1. Following treatment with sh-circRNA_102541 or miR-296-5p mimics, proliferative ability and levels of PCNA were remarkably reduced in HUVEC cells, while apoptosis was significantly enhanced. Co-transfection with miR-296-5p mimics abrogated the effects induced by the overexpressed circ_102541. Additionally, treatment with si-PLK1 attenuated the biological behavior changes caused by miR-296-5p inhibitors in HUVEC cells. Moreover, transfection with LV-PLK1 reversed the effects triggered by miR-296-5p mimics. CONCLUSION Taken together, circRNA_102541 was upregulated in atherosclerosis, and knockdown of circRNA_102541 suppressed cell proliferation while promoted apoptosis of HUVEC cells via miR-296-5p/PLK1. This novel pathway may serve essential roles on the development of atherosclerosis, and circRNA_102541 could be a promising therapeutic candidate for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Na Du
- Department of Cardiology, the First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, 121001, People's Republic of China
| | - Mingjin Li
- Liaoning Jinqiu Hospital, Shenyang, Liaoning, 110015, People's Republic of China
| | - Dan Yang
- Department of Dermatology, the First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, 121001, People's Republic of China.
| |
Collapse
|
10
|
Jiang Q, Li Y, Wu Q, Huang L, Xu J, Zeng Q. Pathogenic role of microRNAs in atherosclerotic ischemic stroke: Implications for diagnosis and therapy. Genes Dis 2022; 9:682-696. [PMID: 35782982 PMCID: PMC9243347 DOI: 10.1016/j.gendis.2021.01.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 12/16/2020] [Accepted: 01/04/2021] [Indexed: 12/15/2022] Open
Abstract
Ischemic stroke resulting from atherosclerosis (particularly in the carotid artery) is one of the major subtypes of stroke and has a high incidence of death. Disordered lipid homeostasis, lipid deposition, local macrophage infiltration, smooth muscle cell proliferation, and plaque rupture are the main pathological processes of atherosclerotic ischemic stroke. Hepatocytes, macrophages, endothelial cells and vascular smooth muscle cells are the main cell types participating in these processes. By inhibiting the expression of the target genes in these cells, microRNAs play a key role in regulating lipid disorders and atherosclerotic ischemic stroke. In this article, we listed the microRNAs implicated in the pathology of atherosclerotic ischemic stroke and aimed to explain their pro- or antiatherosclerotic roles. Our article provides an update on the potential diagnostic use of miRNAs for detecting growing plaques and impending clinical events. Finally, we provide a perspective on the therapeutic use of local microRNA delivery and discuss the challenges for this potential therapy.
Collapse
|
11
|
Role of microRNA-185 in the FoxO1-CYP7A1 mediated regulation of bile acid and cholesterol metabolism: A novel target for drug discovery? Atherosclerosis 2022; 348:53-55. [DOI: 10.1016/j.atherosclerosis.2022.03.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 03/23/2022] [Indexed: 12/31/2022]
|
12
|
MicroRNA-185 modulates CYP7A1 mediated cholesterol-bile acid metabolism through post-transcriptional and post-translational regulation of FoxO1. Atherosclerosis 2022; 348:56-67. [DOI: 10.1016/j.atherosclerosis.2022.03.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 02/22/2022] [Accepted: 03/03/2022] [Indexed: 12/22/2022]
|
13
|
Loscalzo G, Scheel J, Ibañez-Cabellos JS, García-Lopez E, Gupta S, García-Gimenez JL, Mena-Mollá S, Perales-Marín A, Morales-Roselló J. Overexpression of microRNAs miR-25-3p, miR-185-5p and miR-132-3p in Late Onset Fetal Growth Restriction, Validation of Results and Study of the Biochemical Pathways Involved. Int J Mol Sci 2021; 23:ijms23010293. [PMID: 35008715 PMCID: PMC8745308 DOI: 10.3390/ijms23010293] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/21/2021] [Accepted: 12/24/2021] [Indexed: 11/27/2022] Open
Abstract
In a prospective study, 48 fetuses were evaluated with Doppler ultrasound after 34 weeks and classified, according to the cerebroplacental ratio (CPR) and estimated fetal weight (EFW), into fetuses with normal growth and fetuses with late-onset fetal growth restriction (LO-FGR). Overexpression of miRNAs from neonatal cord blood belonging to LO-FGR fetuses, was validated by real-time PCR. In addition, functional characterization of overexpressed miRNAs was performed by analyzing overrepresented pathways, gene ontologies, and prioritization of synergistically working miRNAs. Three miRNAs: miR-25-3p, miR-185-5p and miR-132-3p, were significantly overexpressed in cord blood of LO-FGR fetuses. Pathway and gene ontology analysis revealed over-representation of certain molecular pathways associated with cardiac development and neuron death. In addition, prioritization of synergistically working miRNAs highlighted the importance of miR-185-5p and miR-25-3p in cholesterol efflux and starvation responses associated with LO-FGR phenotypes. Evaluation of miR-25-3p; miR-132-3p and miR-185-5p might serve as molecular biomarkers for the diagnosis and management of LO-FGR; improving the understanding of its influence on adult disease.
Collapse
Affiliation(s)
- Gabriela Loscalzo
- Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain; (A.P.-M.); (J.M.-R.)
- Department of Obstetrics and Gynecology, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain
- Correspondence: (G.L.); (J.S.)
| | - Julia Scheel
- Department of Systems Biology and Bioinformatics, University Rostock, 18055 Rostock, Germany;
- Correspondence: (G.L.); (J.S.)
| | - José Santiago Ibañez-Cabellos
- EpiDisease S.L, Parc Científic, University of Valencia, 46980 Paterna, Spain; (J.S.I.-C.); (E.G.-L.); (J.L.G.-G.); (S.M.-M.)
- Consortium Center for Biomedical Network Research on Rare Diseases (CIBERER), Carrer d’Alvaro de Bazan, 10, 46010 Valencia, Spain
| | - Eva García-Lopez
- EpiDisease S.L, Parc Científic, University of Valencia, 46980 Paterna, Spain; (J.S.I.-C.); (E.G.-L.); (J.L.G.-G.); (S.M.-M.)
| | - Shailendra Gupta
- Department of Systems Biology and Bioinformatics, University Rostock, 18055 Rostock, Germany;
| | - José Luis García-Gimenez
- EpiDisease S.L, Parc Científic, University of Valencia, 46980 Paterna, Spain; (J.S.I.-C.); (E.G.-L.); (J.L.G.-G.); (S.M.-M.)
- Consortium Center for Biomedical Network Research on Rare Diseases (CIBERER), Carrer d’Alvaro de Bazan, 10, 46010 Valencia, Spain
- Institute of Health Carlos III, Biomedical Research Institute INCLIVA, 46010 Valencia, Spain
- Department of Physiology, School of Medicine and Dentistry, University of Valencia, 46010 Valencia, Spain
| | - Salvador Mena-Mollá
- EpiDisease S.L, Parc Científic, University of Valencia, 46980 Paterna, Spain; (J.S.I.-C.); (E.G.-L.); (J.L.G.-G.); (S.M.-M.)
- Institute of Health Carlos III, Biomedical Research Institute INCLIVA, 46010 Valencia, Spain
- Department of Physiology, School of Medicine and Dentistry, University of Valencia, 46010 Valencia, Spain
| | - Alfredo Perales-Marín
- Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain; (A.P.-M.); (J.M.-R.)
- Department of Obstetrics and Gynecology, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain
- Department of Pediatrics, Obstetrics and Gynecology, School of Medicine and Dentistry, University of Valencia, 46010 Valencia, Spain
| | - José Morales-Roselló
- Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain; (A.P.-M.); (J.M.-R.)
- Department of Obstetrics and Gynecology, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain
- Department of Pediatrics, Obstetrics and Gynecology, School of Medicine and Dentistry, University of Valencia, 46010 Valencia, Spain
| |
Collapse
|
14
|
Chen C, Matye D, Wang Y, Li T. Liver-specific microRNA-185 knockout promotes cholesterol dysregulation in mice. LIVER RESEARCH 2021; 5:232-238. [PMID: 35173984 PMCID: PMC8846416 DOI: 10.1016/j.livres.2020.09.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND The liver plays a key role in regulating whole body cholesterol homeostasis. Hepatic cholesterol accumulation causes liver injury in fatty liver disease and hypercholesterolemia increases the risk of cardiovascular disease. MicroRNAs (miRNAs, miRs) have been shown to regulate various pathways in cholesterol metabolism. Recently, miR-185 has been shown to regulate sterol regulatory element-binding protein 2 (SREBP2) and low-density lipoprotein receptor (LDLR) to modulate cholesterol synthesis and uptake. MATERIALS AND METHODS The role of miR-185 in regulating diet-induced metabolic disorders were studied in liver-specific miRNA-185 knockout (L-miR-185 KO) mice. RESULTS L-miR-185 KO mice developed worsened hepatic steatosis upon high fat high cholesterol Western diet feeding with accumulation of triglyceride and cholesterol in the liver. In addition, L-miR-185 KO mice developed hypercholesterolemia upon Western diet feeding. Gene expression analysis showed that L-miR-185 KO mice did not show increased hepatic mRNA expression of SREBP2 or its targets LDLR and HMG-CoA reductase (HMGCR). Although expression of miR-185 mimic inhibited the mRNA of SREBP2, HMGCR and LDLR in HepG2 cells, miR-185 inhibitor did not increase the mRNA of SREBP2, HMGCR or LDLR in HepG2 cells. CONCLUSIONS In conclusion, we reported that L-miR-185 KO mice were more sensitive to Western diet induced hepatic steatosis and hypercholesterolemia. The molecular mechanisms underlying these metabolic changes remain to be investigated in future studies.
Collapse
|
15
|
Ge P, Gao M, Du J, Yu J, Zhang L. Downregulation of microRNA-512-3p enhances the viability and suppresses the apoptosis of vascular endothelial cells, alleviates autophagy and endoplasmic reticulum stress as well as represses atherosclerotic lesions in atherosclerosis by adjusting spliced/unspliced ratio of X-box binding protein 1 (XBP-1S/XBP-1U). Bioengineered 2021; 12:12469-12481. [PMID: 34783632 PMCID: PMC8810154 DOI: 10.1080/21655979.2021.2006862] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2022] Open
Abstract
AS is an important pathological basis of cardiovascular disease. It has been reported that miRNAs are involved in almost all steps of AS, including the injury and dysfunction of endothelial cells and vascular smooth muscle cells. This work was designed to elucidate the biological functions of miR-512-3p in the pathological process of AS and probe into the underlying molecular mechanism. In the present work, ox-LDL-treated HUVECs served as the in vitro model of AS and ApoE-/- mice were nourished with a high-fat diet to establish an in vivo model of AS. Proliferation, apoptosis and migration of HUVECs were evaluated by performing CCK-8, TUNEL staining, western blot and transwell assays. Immunofluorescence examined LC3 expression and levels of autophagy-related and ER stress-related proteins were determined by western blot assay. In addition, starBase predicted the complementary binding sites of XBP-1 to miR-512-3p and luciferase reporter assay confirmed the interaction between miR-512-3p and XBP-1. Moreover, H&E staining was employed to evaluate atherosclerotic lesions in AS model mice. Results revealed that ox-LDL treatment decreased the proliferative and migrative activities and promoted the apoptosis of HUVECs as well as induced autophagy and ER stress, which were abrogated by miR-512-3p silencing. Importantly, ox-LDL treatment elevated miR-512-3p expression and XBP-1 was a direct target of miR-512-3p. Mechanistically, knockdown of miR-512-3p enhanced the viability, suppressed the apoptosis and promoted the migration of ox-LDL-treated HUVECs, alleviated atherosclerotic lesions in AS model mice as well as repressed autophagy and ER stress by targeting XBP-1 to manipulate the ratio of XBP-1S/XBP-1U.
Collapse
Affiliation(s)
- Peipei Ge
- Department of Cardiology, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, 264000, Shandong Province, People's Republic of China
| | - Mingxiao Gao
- Department of Cardiology, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, 264000, Shandong Province, People's Republic of China
| | - Juan Du
- Department of Cardiology, Rizhao Hospital of Traditional Chinese Medicine, Rizhao, 276800, Shandong Province, People's Republic of China
| | - Jingbin Yu
- Department of Cardiology, Zibo Central Hospital, No54 Gongqingtuan West Road, Zibo, 255036, Shandong Province, People's Republic of China
| | - Lei Zhang
- Department of Cardiology, Zibo Central Hospital, No54 Gongqingtuan West Road, Zibo, 255036, Shandong Province, People's Republic of China
| |
Collapse
|
16
|
Peng H, Sun J, Li Y, Zhang Y, Zhong Y. Circ-USP9X Inhibition Reduces Oxidized Low-density Lipoprotein-induced Endothelial Cell Injury via the microRNA 599/Chloride Intracellular Channel 4 Axis. J Cardiovasc Pharmacol 2021; 78:560-571. [PMID: 34269702 DOI: 10.1097/fjc.0000000000001104] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 05/29/2021] [Indexed: 11/27/2022]
Abstract
ABSTRACT Atherosclerosis (AS) is the common pathological basis of cardiovascular disease. Circular RNA circ-USP9X (hsa_circ_0090231) has been discovered to be upregulated in oxidized low-density lipoprotein (ox-LDL)-induced human umbilical vein endothelial cells (HUVECs), but the role of circ-USP9X in ox-LDL-induced endothelial cell injury is indistinct. The purpose of the research was to investigate the role and regulatory mechanism of circ-USP9X in ox-LDL--induced endothelial cell injury. Expression of circ-USP9X was examined by quantitative real-time polymerase chain reaction. Loss-of-function experiments were performed to assess the impacts of circ-USP9X inhibition on viability, cell cycle progression, apoptosis, and tube formation, inflammation, and oxidative stress of ox-LDL-induced HUVEC. The regulatory mechanism of circ-USP9X predicted by bioinformatics analysis and verified by dual-luciferase reporter or RNA immunoprecipitation assays. We observed that circ-USP9X was upregulated in AS patients' serum and ox-LDL-induced HUVEC. Inhibition of circ-USP9X elevated viability, promoted cell cycle progression and angiopoiesis, and decreased apoptosis, inflammation, and oxidative stress of ox-LDL-induced HUVEC. Mechanically, circ-USP9X regulated chloride intracellular channel 4 (CLIC4) messenger RNA expression by sponging microRNA (miR)-599. Furthermore, miR-599 inhibitor overturned circ-USP9X silencing-mediated influence on ox-LDL-induced HUVEC injury. Also, CLIC4 overexpression reversed miR-599 elevation-mediated effect on ox-LDL-induced HUVEC injury. In conclusion, circ-USP9X silencing decreased ox-LDL-induced endothelial cell injury via the miR-599/CLIC4 axis, which offered a novel molecular mechanism to comprehend the pathology of AS.
Collapse
Affiliation(s)
- Huaiyu Peng
- Department of Vascular Intervention, The Ninth Hospital of Xi'an, Xi'an City, Shanxi Province, China ; and
| | - Jihu Sun
- Department of Oncology, Xi'an Hospital of Traditional Chinese Medicine, Xi'an City, Shanxi Province, China
| | - Yi Li
- Department of Vascular Intervention, The Ninth Hospital of Xi'an, Xi'an City, Shanxi Province, China ; and
| | - Ye Zhang
- Department of Vascular Intervention, The Ninth Hospital of Xi'an, Xi'an City, Shanxi Province, China ; and
| | - Yongjin Zhong
- Department of Vascular Intervention, The Ninth Hospital of Xi'an, Xi'an City, Shanxi Province, China ; and
| |
Collapse
|
17
|
Cross-Talk between Mechanosensitive Ion Channels and Calcium Regulatory Proteins in Cardiovascular Health and Disease. Int J Mol Sci 2021; 22:ijms22168782. [PMID: 34445487 PMCID: PMC8395829 DOI: 10.3390/ijms22168782] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 08/13/2021] [Accepted: 08/14/2021] [Indexed: 12/12/2022] Open
Abstract
Mechanosensitive ion channels are widely expressed in the cardiovascular system. They translate mechanical forces including shear stress and stretch into biological signals. The most prominent biological signal through which the cardiovascular physiological activity is initiated or maintained are intracellular calcium ions (Ca2+). Growing evidence show that the Ca2+ entry mediated by mechanosensitive ion channels is also precisely regulated by a variety of key proteins which are distributed in the cell membrane or endoplasmic reticulum. Recent studies have revealed that mechanosensitive ion channels can even physically interact with Ca2+ regulatory proteins and these interactions have wide implications for physiology and pathophysiology. Therefore, this paper reviews the cross-talk between mechanosensitive ion channels and some key Ca2+ regulatory proteins in the maintenance of calcium homeostasis and its relevance to cardiovascular health and disease.
Collapse
|
18
|
Circular RNA circUBR4 regulates ox-LDL-induced proliferation and migration of vascular smooth muscle cells through miR-185-5p/FRS2 axis. Mol Cell Biochem 2021; 476:3899-3910. [PMID: 34159479 DOI: 10.1007/s11010-021-04207-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 06/09/2021] [Indexed: 10/21/2022]
Abstract
Circular RNAs (circRNAs) have been reported to play vital roles in atherosclerosis. However, the precise roles of circUBR4 in atherosclerosis remain unclear. The purpose of this study is to investigate the regulatory roles of circUBR4 in atherosclerosis. The expression levels of circUBR4, miR-185-5p, and Fibroblast growth factor receptor substrate 2 (FRS2) were analyzed by real-time quantitative polymerase chain reaction (RT-qPCR) assay. Human vascular smooth muscle cells (VSMCs) were treated with oxidized low-density lipoprotein (ox-LDL) to mimic atherosclerosis condition in vitro. Cell proliferation was determined by 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyl-2H-tetrazol-3-ium bromide (MTT), colony-forming, and 5-ethynyl-2'-deoxyuridine (EdU) assays. Wound healing and transwell assays were used to assess cell migration. The interaction relationship between miR-185-5p and circUBR4 or FRS2 was confirmed by dual-luciferase reporter and RNA pull-down assays. CircUBR4 was overexpressed in atherosclerosis patients and VSMCs treated with ox-LDL, and the knockdown of circUBR4 abolished ox-LDL-induced enhanced effects on the proliferation and migration of VSMCs. MiR-185-5p, interacted with FRS2, was a target of circUBR4 in VSMCs. The silencing of miR-185-5p reversed the effects caused by circUBR4 knockdown on ox-LDL-induced VSMCs. In addition, overexpression of miR-185-5p suppressed the proliferation and migration of VSMCs by targeting FRS2. CircUBR4 contributed to ox-LDL-induced VSMC proliferation and migration through up-regulating FRS2 via miR-185-5p.
Collapse
|
19
|
Lin F, Zhang S, Liu X, Wu M. RETRACTED: Mouse bone marrow derived mesenchymal stem cells-secreted exosomal microRNA-125b-5p suppresses atherosclerotic plaque formation via inhibiting Map4k4. Life Sci 2021; 274:119249. [PMID: 33652034 DOI: 10.1016/j.lfs.2021.119249] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 01/16/2021] [Accepted: 01/25/2021] [Indexed: 02/08/2023]
Abstract
This article has been retracted: please see Elsevier Policy on Article Withdrawal (http://www.elsevier.com/locate/withdrawalpolicy). This article has been retracted at the request of the Editor-in-Chief. Concern was raised about the reliability of the Western blot results in Figs. 2D and 4E, which appear to have the same eyebrow shaped phenotype as many other publications tabulated here (https://docs.google.com/spreadsheets/d/149EjFXVxpwkBXYJOnOHb6RhAqT4a2llhj9LM60MBffM/edit#gid=0). The journal requested the corresponding author comment on these concerns and provide the raw data. However the authors were not able to satisfactorily fulfil this request and therefore the Editor-in-Chief decided to retract the article.
Collapse
Affiliation(s)
- Feng Lin
- Department of Cardiology, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen 518000, Guangdong, China.
| | - Suihao Zhang
- Department of Cardiology, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen 518000, Guangdong, China
| | - Xia Liu
- Department of Cardiology, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen 518000, Guangdong, China
| | - Meishan Wu
- Department of Cardiology, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen 518000, Guangdong, China
| |
Collapse
|
20
|
Ma G, Bi S, Zhang P. Long non-coding RNA MIAT regulates ox-LDL-induced cell proliferation, migration and invasion by miR-641/STIM1 axis in human vascular smooth muscle cells. BMC Cardiovasc Disord 2021; 21:248. [PMID: 34016053 PMCID: PMC8139145 DOI: 10.1186/s12872-021-02048-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 05/05/2021] [Indexed: 11/16/2022] Open
Abstract
Background Atherosclerosis (AS) is a primary cause of coronary heart and vascular diseases. Long non-coding RNAs (lncRNAs) are indicated to regulate AS progression. This study aimed to reveal the biological roles of lncRNA myocardial infarction associated transcript (MIAT) in oxidized low-density lipoprotein (ox-LDL)-induced human vascular smooth muscle cells (VSMCs). Methods The RNA levels of MIAT, microRNA-641 (miR-641) and stromal interaction molecule 1 (STIM1) were detected by quantitative real-time polymerase chain reaction (qRT-PCR). The protein levels were determined by western blot analysis. Cell proliferation was assessed by cell colony formation and DNA content quantitation assays. Cell migration and invasion were demonstrated by wound-healing and transwell assays. The putative binding relationships between miR-641 and MIAT or STIM1 were predicted by starbase online database, and identified by dual-luciferase reporter and RNA immunoprecipitation assays. Results MIAT and STIM1 expression were substantially upregulated, whereas miR-641 expression was downregulated in ox-LDL-induced VSMCs compared with control groups. Functionally, MIAT silencing attenuated ox-LDL-induced cell proliferation, migration and invasion in VSMCs; however, these effects were impaired by miR-641 inhibitor. STIM1 overexpression also restrained miR-641-mediated impacts on cell proliferation and metastasis under ox-LDL. Mechanistically, MIAT acted as a sponge for miR-641, and miR-641 was associated with STIM1. Conclusions MIAT silencing hindered ox-LDL-induced cell proliferation, migration and invasion by downregulating STIM1 expression through binding to miR-641 in VSMCs. The mechanism provided us with a new target for AS therapy. Supplementary Information The online version contains supplementary material available at 10.1186/s12872-021-02048-9.
Collapse
Affiliation(s)
- Gang Ma
- Deptment of Cardiac Surgury, Zibo Central Hospital, Zibo, 255036, Shandong, People's Republic of China
| | - Shuting Bi
- Deptment of Cardiac Surgury, Zibo Central Hospital, Zibo, 255036, Shandong, People's Republic of China
| | - Pengfei Zhang
- Department of Cardiac Surgery, Central Hospital Affiliated to Shandong First Medical University, No.105, Jiefang Road, Jinan, 250013, Shandong, People's Republic of China. .,Department of Cardiac Surgery, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong, People's Republic of China.
| |
Collapse
|
21
|
Wang W, Wang P, Xie K, Luo R, Gao X, Yan Z, Huang X, Yang Q, Gun S. ssc-miR-185 targets cell division cycle 42 and promotes the proliferation of intestinal porcine epithelial cell. Anim Biosci 2021; 34:801-810. [PMID: 33152231 PMCID: PMC8100468 DOI: 10.5713/ajas.20.0325] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 07/30/2020] [Accepted: 09/02/2020] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVE microRNAs (miRNAs) can play a role in a variety of physiological and pathological processes, and their role is achieved by regulating the expression of target genes. Our previous high-throughput sequencing found that ssc-miR-185 plays an important regulatory role in piglet diarrhea, but its specific target genes and functions in intestinal porcine epithelial cell (IPEC-J2) are still unclear. We intended to verify the target relationship between porcine miR-185 and cell division cycle 42 (CDC42) gene in IPEC-J2 and to explore the effect of miR-185 on the proliferation of IPEC-J2 cells. METHODS The TargetScan, miRDB, and miRanda software were used to predict the target genes of porcine miR-185, and CDC42 was selected as a candidate target gene. The CDC423' UTR-wild type (WT) and CDC42-3'UTR-mutant type (MUT) segments were successfully cloned into pmirGLO luciferase vector, and the luciferase activity was detected after co-transfection with miR-185 mimics and pmirGLO-CDC42-3'UTR. The expression level of CDC42 was analyzed using quantitative polymerase chain reaction and Western blot. The proliferation of IPEC-J2 was detected using cell counting kit-8 (CCK-8), methylthiazolyldiphenyltetrazolium bromide (MTT), and 5-ethynyl-2'-deoxyuridine (EdU) assays. RESULTS Double enzyme digestion and sequencing confirmed that CDC42-3'UTR-WT and CDC42-3'UTR-MUT were successfully cloned into pmirGLO luciferase reporter vector, and the luciferase activity was significantly reduced after co-transfection with miR-185 mimics and CDC42-3'UTR-WT. Further we found that the mRNA and protein expression level of CDC42 were down-regulated after transfection with miR-185 mimics, while the opposite trend was observed after transfection with miR-185 inhibitor (p<0.01). In addition, the CCK-8, MTT, and EdU results demonstrated that miR-185 promotes IPEC-J2 cells proliferation by targeting CDC42. CONCLUSION These findings indicate that porcine miR-185 can directly target CDC42 and promote the proliferation of IPEC-J2 cells. However, the detailed regulatory mechanism of miR-185/CDC42 axis in piglets' resistance to diarrhea is yet to be elucidated in further investigation.
Collapse
Affiliation(s)
- Wei Wang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu 730070, China
| | - Pengfei Wang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu 730070, China
| | - Kaihui Xie
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu 730070, China
| | - Ruirui Luo
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu 730070, China
| | - Xiaoli Gao
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu 730070, China
| | - Zunqiang Yan
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu 730070, China
| | - Xiaoyu Huang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu 730070, China
| | - Qiaoli Yang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu 730070, China
| | - Shuangbao Gun
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu 730070, China
- Gansu Research Center for Swine Production Engineering and Technology, Lanzhou, Gansu 730070, China
| |
Collapse
|
22
|
LncRNA XIST Promotes Atherosclerosis by Regulating miR-599/TLR4 Axis. Inflammation 2021; 44:965-973. [PMID: 33566259 DOI: 10.1007/s10753-020-01391-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 11/18/2020] [Accepted: 11/30/2020] [Indexed: 12/15/2022]
Abstract
Long noncoding RNAs (lncRNAs) have been reported to be implicated in various biological and pathological processes. However, the function and mechanism of XIST in vascular smooth muscle cells (VSMCs) remains unknown. The levels of XIST, miR-599, and TLR4 were tested by RT-qPCR. VSMCs and human mononuclear cells (U937) treated with ox-LDL were used as atherosclerosis (AS) cell models. The CCK-8 assay was adopted to detect cell viability. Cell apoptosis was examined by the TUNEL assay. A dual-luciferase reporter assay was employed to investigate the interaction between miR-599 and XIST or TLR4. In this research, we uncovered that the XIST level was elevated in the serum of AS patients and ox-LDL-treated AS cell models. Functional analysis revealed that XIST depletion restrained cell proliferation, while induced the apoptosis in AS cell models. Besides, miR-599 was verified to be a direct downstream target of XIST and miR-599 inhibitor reversed the effects of XIST knockdown on AS progression. Finally, we demonstrated that XIST increased TLR4 expression by serving as a ceRNA of miR-599. All these findings manifested the role of the XIST/miR-599/TLR4 axis in AS development.
Collapse
|
23
|
Liu L, Zhang J, Liu Y. MicroRNA-1323 serves as a biomarker in gestational diabetes mellitus and aggravates high glucose-induced inhibition of trophoblast cell viability by suppressing TP53INP1. Exp Ther Med 2021; 21:230. [PMID: 33603839 PMCID: PMC7851622 DOI: 10.3892/etm.2021.9661] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 12/14/2020] [Indexed: 12/21/2022] Open
Abstract
Gestational diabetes mellitus (GDM) leads to poor pregnancy outcomes, and microRNAs (miRNAs/miRs) have been suggested to be associated with GDM, but the pathological mechanisms remain unclear. The present study aimed to investigate the diagnostic value of miR-1323 in GDM patients and its effects on trophoblast cell viability. Additionally, the present study investigated the correlation between miR-1323 and TP53INP1 to understand the pathological mechanism of GDM progression. Reverse transcription-quantitative polymerase chain reaction was used to detect the miR-1323 expression and TP53INP1 mRNA expression. The diagnostic value of serum miR-1323 was evaluated by receiver operating characteristic analysis. HTR-8/SVneo and BeWo cells were treated with high glucose (HG) to construct cell models of GDM, and trophoblast cell viability was assessed using an MTT assay. The protein expression of TP53INP1 was detected by western blot analysis. The correlation between miR-1323 and TP53INP1 was investigated by luciferase reporter assay. The miR-1323 expression was increased in patients with GDM, which had relatively high diagnostic accuracy for GDM screening and was positively correlated with fasting blood glucose in patients GDM. HG upregulated the miR-1323 expression and inhibited trophoblast cell viability. Overexpression of miR-1323 significantly inhibited the viability of HG-induced trophoblast cells. TP53INP1, a target gene of miR-1323, was negatively correlated with miR-1323. TP53INP1 overexpression reversed the inhibitory effect of miR-1323 overexpression on the viability of HG-treated trophoblast cells. Increased levels of serum miR-1323 may be a diagnostic biomarker for GDM. Additionally, miR-1323 may inhibit trophoblast cell viability by inhibiting TP53INP1, suggesting that it may be a potential therapeutic target for GDM.
Collapse
Affiliation(s)
- Lijun Liu
- Department of Gynecology, Weifang Maternal and Child Health Hospital, Weifang, Shandong 261011, P.R. China
| | - Jun Zhang
- Department of Pharmacy, Weifang Maternal and Child Health Hospital, Weifang, Shandong 261011, P.R. China
| | - Yujuan Liu
- Department of Central Supply Room, Weifang Maternal and Child Health Hospital, Weifang, Shandong 261011, P.R. China
| |
Collapse
|
24
|
Liu X, Pan Z. Store-Operated Calcium Entry in the Cardiovascular System. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1349:303-333. [DOI: 10.1007/978-981-16-4254-8_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
25
|
2,3,7,8-Tetrachlorodibenzo- p-dioxin (TCDD) and Polychlorinated Biphenyl Coexposure Alters the Expression Profile of MicroRNAs in the Liver Associated with Atherosclerosis. BIOMED RESEARCH INTERNATIONAL 2020; 2020:2652756. [PMID: 32855961 PMCID: PMC7443005 DOI: 10.1155/2020/2652756] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 07/14/2020] [Accepted: 07/20/2020] [Indexed: 12/12/2022]
Abstract
MicroRNAs (miRNAs) are a class of small RNAs that regulate gene expression. 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) and polychlorinated biphenyls (PCBs) are persistent organic pollutants that exist as complex mixtures in vivo. When humans are simultaneously exposed to these compounds, the development of atherosclerosis is known to be enhanced. However, the roles of miRNA in TCDD- and PCB-induced atherosclerosis are largely unknown. Therefore, the present study is aimed at elucidating the possible dysregulation of miRNAs in atherogenesis induced by coexposure to TCDD and PCBs. Eight-week-old male ApoE−/− mice were coexposed to TCDD (15 μg/kg) and Aroclor1254 (55 mg/kg, a representative mixture of PCBs) by intraperitoneal injection four times over a 6-week period. Microarray analysis of miRNAs and mRNAs in the liver of ApoE−/− mice with or without TCDD and Aroclor1254 coexposure was performed. We discovered that 68 miRNAs and 1312 mRNAs exhibited significant expression changes in response to TCDD and PCB coexposure and revealed that both changed miRNAs and mRNAs are involved in cardiovascular disease processes. An integrated miRNA-mRNA approach indicated that miRNA-26a-5p, miRNA-193a-3p, and miRNA-30c-5p participated in specific TCDD and Aroclor1254 coresponsive networks which are relevant to the cardiovascular system development and function network. Furthermore, our results also indicated that miRNA-130a-3p and miRNA-376a-3p were novel players in the regulation of TCDD- and Aroclor1254-induced atherosclerosis pathways. In summary, our finding provided new insights into the mechanism of atherosclerosis in response to TCDD and PCB coexposure.
Collapse
|
26
|
Huang Z, Li P, Wu L, Zhang D, Du B, Liang C, Gao L, Zhang Y, Yao R. Hsa_circ_0029589 knockdown inhibits the proliferation, migration and invasion of vascular smooth muscle cells via regulating miR-214-3p and STIM1. Life Sci 2020; 259:118251. [PMID: 32795540 DOI: 10.1016/j.lfs.2020.118251] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 07/30/2020] [Accepted: 08/07/2020] [Indexed: 12/13/2022]
Abstract
AIMS Circular RNAs (circRNAs) are relevant to atherosclerosis progression. However, the role and mechanism of circRNA hsa_circ_0029589 (circ_0029589) in atherosclerosis are not fully understood. This research aims to explore the function and mechanism of circ_0029589 in oxidized low-density lipoprotein (ox-LDL)-caused vascular smooth muscle cells (VSMCs) injury in vitro. MAIN METHODS VSMCs were challenged via ox-LDL to mimic atherosclerosis-like injury in vitro. Circ_0029589, microRNA-214-3p (miR-214-3p) and stromal interaction molecule 1 (STIM1) abundances were detected via quantitative reverse transcription polymerase chain reaction or western blot. Cell proliferation was investigated via cell viability, cycle, apoptosis and proliferation-associated protein levels. Cell migration and invasion were assessed via transwell analysis. The relationship between miR-214-3p and circ_0029589 or STIM1 was tested via dual-luciferase reporter analysis and RNA immunoprecipitation. KEY FINDINGS Circ_0029589 level was enhanced in ox-LDL-challenged VSMCs. Circ_0029589 interference constrained cell proliferation, migration and invasion in ox-LDL-challenged VSMCs. miR-214-3p was targeted by circ_0029589 and miR-214-3p knockdown weakened the suppressive function of circ_0029589 silence on VSMCs proliferation, migration and invasion. STIM1 was targeted via miR-214-3p and miR-214-3p could suppress VSMCs proliferation, migration and invasion via decreasing STIM1. Moreover, circ_0029589 modulated STIM1 level by miR-214-3p. SIGNIFICANCE Circ_0029589 knockdown repressed proliferation, migration and invasion of VSMCs challenged via ox-LDL by regulating miR-214-3p and STIM1, indicating that circ_0029589 might play important role in atherosclerosis.
Collapse
Affiliation(s)
- Zhen Huang
- Cardiovascular Hospital, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Penglei Li
- Department of Vasculocardiology, People's Hospital of Zhongmu, Zhengzhou, Henan, China
| | - Leiming Wu
- Cardiovascular Hospital, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Dianhong Zhang
- Cardiovascular Hospital, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Binbin Du
- Cardiovascular Hospital, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Cui Liang
- Cardiovascular Hospital, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Lu Gao
- Cardiovascular Hospital, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yanzhou Zhang
- Cardiovascular Hospital, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Rui Yao
- Cardiovascular Hospital, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
27
|
Li Y, Zhong W, Zhu M, Li M, Yang Z. miR-185 inhibits prostate cancer angiogenesis induced by the nodal/ALK4 pathway. BMC Urol 2020; 20:49. [PMID: 32366240 PMCID: PMC7197131 DOI: 10.1186/s12894-020-00617-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 04/21/2020] [Indexed: 12/12/2022] Open
Abstract
Background Inhibition of angiogenesis in prostatic cancer could be a brand-new method to suppress tumour progression. Nodal/ALK4 has been associated with vascularization in many cancers. However, the relationship between and role of Nodal/ALK4 and miR-185 in human prostatic cancer is still unknown. Methods Prostatic cancer DU145 cells and LNCaP cells were used to investigate the angiogenic effect induced by Nodal and the anti-angiogenic roles of miR-185. Colony formation assay, MTT assay, transwell assay and tube formation assay were used to explore cell proliferation, migration and tube-forming ability, respectively. A luciferase reporter assay confirmed the binding relationship between miR-185 and ALK4. The expression levels of miR-185, ALK4 and VEGF were detected by qRT-PCR and Western blotting. The effects of miR-185 and Nodal in prostate cancer were also investigated in animal experiments. Results VEGF expression was increased in DU145 cells and LNCaP cells after Nodal incubation, and Nodal activated the proliferation ability of prostatic cancer cells and the migration and tube-forming ability of human umbilical vein endothelial cells (HUVECs), which were all inhibited by treatment with the Nodal inhibitor SB431524. Bioinformatics analysis and luciferase assay were used to verify miR-185 as a target of ALK4. Prostatic cancer cell proliferation was inhibited by overexpression of miR-185, which was shown to regulate the migration and angiogenesis of HUVECs by targeting ALK4 for suppression. miR-185 also showed a significant inverse correlation with Nodal treatment and reversed the angiogenic effects induced by Nodal. More importantly, for the first time, xenograft experiments indicated that overexpression of miR-185 suppressed tumour development. Conclusion The Nodal/ALK4 pathway is important in the angiogenesis of prostate cancer and can be inhibited by targeting miR-185 to downregulate ALK4. These findings provide a new perspective on the mechanism of prostate cancer formation.
Collapse
Affiliation(s)
- Youkong Li
- Department of Urology, Jingzhou Central Hospital and The Second Clinical Medical College, Yangtze University, No.60 Jingzhong Road, Jingzhou District, Jingzhou, 434020, Hubei Province, People's Republic of China.
| | - Wen Zhong
- Department of Endocrine, Jingzhou Central Hospital and The Second Clinical Medical College, Yangtze University, Jingzhou, 434020, Hubei Province, People's Republic of China
| | - Min Zhu
- Department of Urology, Jingzhou Central Hospital and The Second Clinical Medical College, Yangtze University, No.60 Jingzhong Road, Jingzhou District, Jingzhou, 434020, Hubei Province, People's Republic of China
| | - Mengbo Li
- Department of Urology, Jingzhou Central Hospital and The Second Clinical Medical College, Yangtze University, No.60 Jingzhong Road, Jingzhou District, Jingzhou, 434020, Hubei Province, People's Republic of China
| | - Zhenwei Yang
- Department of Urology, Jingzhou Central Hospital and The Second Clinical Medical College, Yangtze University, No.60 Jingzhong Road, Jingzhou District, Jingzhou, 434020, Hubei Province, People's Republic of China
| |
Collapse
|
28
|
Li Y, Zhou J, Zhang O, Wu X, Guan X, Xue Y, Li S, Zhuang X, Zhou B, Miao G, Zhang L. RETRACTED: Bone marrow mesenchymal stem cells-derived exosomal microRNA-185 represses ventricular remolding of mice with myocardial infarction by inhibiting SOCS2. Int Immunopharmacol 2020; 80:106156. [PMID: 31945609 DOI: 10.1016/j.intimp.2019.106156] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 12/09/2019] [Accepted: 12/25/2019] [Indexed: 02/06/2023]
Abstract
This article has been retracted: please see Elsevier Policy on Article Withdrawal (http://www.elsevier.com/locate/withdrawalpolicy). This article has been retracted at the request of the Editor-in-Chief. Concern was raised about the integrity of the images in Figures 5B and 7C, which appear to contain suspected duplications, as detailed here: https://pubpeer.com/publications/C968FDCECE2069D7FF43B346B261ED and here: https://docs.google.com/spreadsheets/d/1r0MyIYpagBc58BRF9c3luWNlCX8VUvUuPyYYXzxWvgY/edit#gid=262337249. Numerous additional suspected image duplications were detected within Figures 5 and 6. Most of these image duplications involve either pasting portions of one image into another, or rotating/flipping the image. The journal requested the corresponding author comment on these concerns and provide the raw data. The authors did not respond to this request and therefore the Editor-in-Chief decided to retract the article.
Collapse
Affiliation(s)
- Yanbing Li
- Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100043 China
| | - Jie Zhou
- Department of Cardiology, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing 102218, China
| | - Ou Zhang
- Department of Cardiology, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing 102218, China
| | - Xuejiao Wu
- Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100043 China
| | - Xiaonan Guan
- Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100043 China
| | - Yajun Xue
- Department of Cardiology, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing 102218, China
| | - Siyuan Li
- School of Clinical Medicine, Tsinghua University, China
| | | | - Boda Zhou
- Department of Cardiology, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing 102218, China
| | - Guobin Miao
- Department of Cardiology, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing 102218, China.
| | - Lin Zhang
- Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100043 China.
| |
Collapse
|
29
|
Fu W, Liu Z, Zhang J, Shi Y, Zhao R, Zhao H. Effect of microRNA-144-5p on the proliferation, invasion and migration of human umbilical vein endothelial cells by targeting SMAD1. Exp Ther Med 2019; 19:165-171. [PMID: 31853287 PMCID: PMC6909792 DOI: 10.3892/etm.2019.8194] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 08/21/2019] [Indexed: 12/28/2022] Open
Abstract
Atherosclerosis is a multifactorial chronic disease that is a major cause of death and injury worldwide. Apoptosis of endothelial cells (ECs) serves an important role in the occurrence and development of atherosclerosis. MicroRNAs (miRNAs) serve a key role in atherosclerosis though regulating the function of ECs. At present, the role of miRNA-144-5p (miR-144-5p) in atherosclerosis is unclear. The aim of this study was to investigate the effect of miR-144-5p on atherosclerosis in oxidized low-density lipoprotein (ox-LDL)-stimulated human umbilical vein endothelial cells (HUVECs). Results from the present study demonstrated that miR-144-5p overexpression could inhibit proliferation and induce apoptosis in HUVECs. To further study the biological function of miR-144-5p, the effects of modulating miR-144-5p expression on the invasion and migration of HUVECs were also examined. The results demonstrated that miR-144-5p upregulation suppressed HUVEC migration and invasion. TargetScan and dual luciferase reporter assay results demonstrated that SMAD1 was a direct target gene of miR-144-5p. miR-144-5p upregulation inhibited the expression of phosphorylated-SMAD1/5/8 in the SMAD pathway. In conclusion, the data indicated that miR-144-5p serves an important role in the development of atherosclerosis through regulating the function of HUVECs by targeting SMAD1.
Collapse
Affiliation(s)
- Wei Fu
- Department of Cardiology, The Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| | - Zidong Liu
- Department of Cardiology, The Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| | - Jing Zhang
- Department of Cardiology, The Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| | - Yuxue Shi
- Department of Cardiology, The Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| | - Ruiyao Zhao
- Department of Cardiology, The Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| | - Heng Zhao
- Department of Cardiology, The Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| |
Collapse
|
30
|
Shiou YL, Lin HT, Ke LY, Wu BN, Shin SJ, Chen CH, Tsai WC, Chu CS, Lee HC. Very Low-Density Lipoproteins of Metabolic Syndrome Modulates STIM1, Suppresses Store-Operated Calcium Entry, and Deranges Myofilament Proteins in Atrial Myocytes. J Clin Med 2019; 8:jcm8060881. [PMID: 31226824 PMCID: PMC6617489 DOI: 10.3390/jcm8060881] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 06/14/2019] [Accepted: 06/15/2019] [Indexed: 01/16/2023] Open
Abstract
Individuals with metabolic syndrome (MetS) are at high risk for atrial myopathy and atrial fibrillation. Very low-density lipoproteins (VLDLs) of MetS (MetS-VLDLs) are cytotoxic to atrial myocytes in vivo and in vitro. The calcineurin-nuclear factor of activated T-cells (NFAT) pathway, which is regulated by stromal interaction molecule 1 (STIM1)/ calcium release-activated calcium channel protein 1 (Orai1)-mediated store-operated Ca2+ entry (SOCE), is a pivotal mediator of adaptive cardiac hypertrophy. We hypothesized that MetS-VLDLs could affect SOCE and the calcineurin-NFAT pathway. Normal-VLDL and MetS-VLDL samples were isolated from the peripheral blood of healthy volunteers and individuals with MetS. VLDLs were applied to HL-1 atrial myocytes for 18 h and were also injected into wild-type C57BL/6 male mouse tails three times per week for six weeks. After the sarcoplasmic reticulum (SR) Ca2+ store was depleted, SOCE was triggered upon reperfusion with 1.8 mM of Ca2+. SOCE was attenuated by MetS-VLDLs, along with reduced transcriptional and membranous expression of STIM1 (P = 0.025), and enhanced modification of O-GlcNAcylation on STIM1 protein, while Orai1 was unaltered. The nuclear translocation and activity of calcineurin were both reduced (P < 0.05), along with the alteration of myofilament proteins in atrial tissues. These changes were absent in normal-VLDL-treated cells. Our results demonstrated that MetS-VLDLs suppressed SOCE by modulating STIM1 at the transcriptional, translational, and post-translational levels, resulting in the inhibition of the calcineurin-NFAT pathway, which resulted in the alteration of myofilament protein expression and sarcomere derangement in atrial tissues. These findings may help explain atrial myopathy in MetS. We suggest a therapeutic target on VLDLs to prevent atrial fibrillation, especially for individuals with MetS.
Collapse
Affiliation(s)
- Yi-Lin Shiou
- Center for Lipid Biosciences, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan.
- Lipid Science and Aging Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Hsin-Ting Lin
- Center for Lipid Biosciences, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan.
| | - Liang-Yin Ke
- Center for Lipid Biosciences, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan.
- Lipid Science and Aging Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Bin-Nan Wu
- Department of Pharmacology, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Shyi-Jang Shin
- Department of Internal Medicine, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Chu-Huang Chen
- Center for Lipid Biosciences, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan.
- Lipid Science and Aging Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Wei-Chung Tsai
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Department of Internal Medicine, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Chih-Sheng Chu
- Center for Lipid Biosciences, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan.
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Department of Internal Medicine, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Hsiang-Chun Lee
- Center for Lipid Biosciences, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan.
- Lipid Science and Aging Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Department of Internal Medicine, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Institute/Center of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung 807, Taiwan.
| |
Collapse
|