1
|
Zeng X, Li L, Tong L. Therapeutic Effects of Proanthocyanidins on Diabetic Erectile Dysfunction in Rats. Int J Mol Sci 2024; 25:11004. [PMID: 39456785 PMCID: PMC11506934 DOI: 10.3390/ijms252011004] [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: 09/15/2024] [Revised: 10/11/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
The rising occurrence of erectile dysfunction related to diabetes mellitus (DMED) has led to the creation of new medications. Proanthocyanidins (PROs) is a potential agent for DMED. In this study, the DMED rat model was established using streptozotocin (STZ) and erectile function was assessed using apomorphine (APO) in rats. Following this, the rats were subjected to oral treatment with PRO. Then, we evaluated the influence of PROs on DMED rats. The findings suggest that PROs significantly enhance erectile function in DMED rats. PROs modulated glucose and lipid metabolism in DMED rats by decreasing blood glucose and lipid levels while increasing liver glycogen and serum insulin levels. Furthermore, PROs enhanced vascular endothelial function in DMED rats by augmenting nitric oxide (NO) levels and reducing the levels of endothelin-1 (ET-1) and lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1). Additionally, PROs have been shown to elevate testosterone (T) levels, mitigate pathological testicular damage, and enhance sperm concentration and survival rates. Finally, the core targets were screened using network pharmacology, followed by validation through molecular docking, enzyme-linked immunoassay (ELISA), and real-time PCR methodologies. Our findings imply that PROs may treat DMED by elevating AKT1 levels while concurrently diminishing CASP3 levels, thereby effectively regulating the PI3K-Akt signaling pathway. Overall, these results support using PROs as a potential candidate for the treatment of DMED.
Collapse
Affiliation(s)
- Xiaoyan Zeng
- Qinghai University, Xining 810000, China; (X.Z.); (L.L.)
- Qinghai Provincial Key Laboratory of Traditional Chinese Medicine Research for Glucolipid Metabolic Diseases, Xining 810000, China
| | - Lanlan Li
- Qinghai University, Xining 810000, China; (X.Z.); (L.L.)
- Qinghai Provincial Key Laboratory of Traditional Chinese Medicine Research for Glucolipid Metabolic Diseases, Xining 810000, China
| | - Li Tong
- Qinghai University, Xining 810000, China; (X.Z.); (L.L.)
- Qinghai Provincial Key Laboratory of Traditional Chinese Medicine Research for Glucolipid Metabolic Diseases, Xining 810000, China
| |
Collapse
|
2
|
Nam U, Kim J, Yi HG, Jeon JS. Investigation of the Dysfunction Caused by High Glucose, Advanced Glycation End Products, and Interleukin-1 Beta and the Effects of Therapeutic Agents on the Microphysiological Artery Model. Adv Healthc Mater 2024; 13:e2302682. [PMID: 38575148 DOI: 10.1002/adhm.202302682] [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: 08/15/2023] [Revised: 03/31/2024] [Indexed: 04/06/2024]
Abstract
Diabetes mellitus (DM) has substantial global implications and contributes to vascular inflammation and the onset of atherosclerotic cardiovascular diseases. However, translating the findings from animal models to humans has inherent limitations, necessitating a novel platform. Therefore, herein, an arterial model is established using a microphysiological system. This model successfully replicates the stratified characteristics of human arteries by integrating collagen, endothelial cells (ECs), and vascular smooth muscle cells (VSMCs). Perfusion via a peristaltic pump shows dynamic characteristics distinct from those of static culture models. High glucose, advanced glycation end products (AGEs), and interleukin-1 beta are employed to stimulate diabetic conditions, resulting in notable cellular changes and different levels of cytokines and nitric oxide. Additionally, the interactions between the disease models and oxidized low-density lipoproteins (LDL) are examined. Finally, the potential therapeutic effects of metformin, atorvastatin, and diphenyleneiodonium are investigated. Metformin and diphenyleneiodonium mitigate high-glucose- and AGE-associated pathological changes, whereas atorvastatin affects only the morphology of ECs. Altogether, the arterial model represents a pivotal advancement, offering a robust and insightful platform for investigating cardiovascular diseases and their corresponding drug development.
Collapse
Affiliation(s)
- Ungsig Nam
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
- Center for Scientific Instrumentation, Korea Basic Science Institute (KBSI), Daejeon, 34133, Republic of Korea
| | - Jaesang Kim
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Hee-Gyeong Yi
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
- Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Jessie S Jeon
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| |
Collapse
|
3
|
Viggiano D. Mechanisms of Diabetic Nephropathy Not Mediated by Hyperglycemia. J Clin Med 2023; 12:6848. [PMID: 37959313 PMCID: PMC10650633 DOI: 10.3390/jcm12216848] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 10/27/2023] [Accepted: 10/27/2023] [Indexed: 11/15/2023] Open
Abstract
Diabetes mellitus (DM) is characterized by the appearance of progressive kidney damage, which may progress to end-stage kidney disease. The control of hyperglycemia is usually not sufficient to halt this progression. The kidney damage is quantitatively and qualitatively different in the two forms of diabetes; the typical nodular fibrosis (Kimmelstiel Wilson nodules) appears mostly in type 1 DM, whereas glomerulomegaly is primarily present in type 2 obese DM. An analysis of the different metabolites and hormones in type 1 and type 2 DM and their differential pharmacological treatments might be helpful to advance the hypotheses on the different histopathological patterns of the kidneys and their responses to sodium/glucose transporter type 2 inhibitors (SGLT2i).
Collapse
Affiliation(s)
- Davide Viggiano
- Department of Translational Medical Sciences, University of Campania, 80131 Naples, Italy
| |
Collapse
|
4
|
Bertelli PM, Pedrini E, Hughes D, McDonnell S, Pathak V, Peixoto E, Guduric-Fuchs J, Stitt AW, Medina RJ. Long term high glucose exposure induces premature senescence in retinal endothelial cells. Front Physiol 2022; 13:929118. [PMID: 36091370 PMCID: PMC9459081 DOI: 10.3389/fphys.2022.929118] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 07/22/2022] [Indexed: 01/10/2023] Open
Abstract
Purpose: Features of cellular senescence have been described in diabetic retinal vasculature. The aim of this study was to investigate how the high glucose microenvironment impacts on the senescence program of retinal endothelial cells. Methods: Human retinal microvascular endothelial cells were cultured under control and high glucose conditions of 5 mM and 25 mM D-glucose, respectively. Isomeric l-glucose was used as the osmotic control. Cells were counted using CASY technology until they reached their Hayflick limit. Senescence-associated β-Galactosidase was used to identify senescent cells. Endothelial cell functionality was evaluated by the clonogenic, 3D tube formation, and barrier formation assays. Cell metabolism was characterized using the Seahorse Bioanalyzer. Gene expression analysis was performed by bulk RNA sequencing. Retinal tissues from db/db and db/+ mice were evaluated for the presence of senescent cells. Publicly available scRNA-sequencing data for retinas from Akimba and control mice was used for gene set enrichment analysis. Results: Long term exposure to 25 mM D-Glucose accelerated the establishment of cellular senescence in human retinal endothelial cells when compared to 5 mM D-glucose and osmotic controls. This was shown from 4 weeks, by a significant slower growth, higher percentages of cells positive for senescence-associated β-galactosidase, an increase in cell size, and lower expression of pRb and HMGB2. These senescence features were associated with decreased clonogenic capacity, diminished tubulogenicity, and impaired barrier function. Long term high glucose-cultured cells exhibited diminished glycolysis, with lower protein expression of GLUT1, GLUT3, and PFKFB3. Transcriptomic analysis, after 4 weeks of culture, identified downregulation of ALDOC, PFKL, and TPI1, in cells cultured with 25 mM D-glucose when compared to controls. The retina from db/db mice showed a significant increase in acellular capillaries associated with a significant decrease in vascular density in the intermediate and deep retinal plexuses, when compared to db/+ mice. Senescent endothelial cells within the db/db retinal vasculature were identified by senescence-associated β-galactosidase staining. Analysis of single cell transcriptomics data for the Akimba mouse retina highlighted an enrichment of senescence and senescence-associated secretory phenotype gene signatures when compared to control mice. Conclusion: A diabetic-like microenvironment of 25 mM D-glucose was sufficient to accelerate the establishment of cellular senescence in human retinal microvascular endothelial cells.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Reinhold J. Medina
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Sciences, Faculty of Medicine, Health, and Life Sciences, Queen’s University Belfast, Belfast, United Kingdom
| |
Collapse
|
5
|
Multi-omics study identifies novel signatures of DNA/RNA, amino acid, peptide, and lipid metabolism by simulated diabetes on coronary endothelial cells. Sci Rep 2022; 12:12027. [PMID: 35835939 PMCID: PMC9283518 DOI: 10.1038/s41598-022-16300-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 07/07/2022] [Indexed: 12/14/2022] Open
Abstract
Coronary artery endothelial cells (CAEC) exert an important role in the development of cardiovascular disease. Dysfunction of CAEC is associated with cardiovascular disease in subjects with type 2 diabetes mellitus (T2DM). However, comprehensive studies of the effects that a diabetic environment exerts on this cellular type are scarce. The present study characterized the molecular perturbations occurring on cultured bovine CAEC subjected to a prolonged diabetic environment (high glucose and high insulin). Changes at the metabolite and peptide level were assessed by Liquid Chromatography–Mass Spectrometry (LC–MS2) and chemoinformatics. The results were integrated with published LC–MS2-based quantitative proteomics on the same in vitro model. Our findings were consistent with reports on other endothelial cell types and identified novel signatures of DNA/RNA, amino acid, peptide, and lipid metabolism in cells under a diabetic environment. Manual data inspection revealed disturbances on tryptophan catabolism and biosynthesis of phenylalanine-based, glutathione-based, and proline-based peptide metabolites. Fluorescence microscopy detected an increase in binucleation in cells under treatment that also occurred when human CAEC were used. This multi-omics study identified particular molecular perturbations in an induced diabetic environment that could help unravel the mechanisms underlying the development of cardiovascular disease in subjects with T2DM.
Collapse
|
6
|
Tong KL, Tan KE, Lim YY, Tien XY, Wong PF. CircRNA-miRNA interactions in atherogenesis. Mol Cell Biochem 2022; 477:2703-2733. [PMID: 35604519 DOI: 10.1007/s11010-022-04455-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 04/27/2022] [Indexed: 11/30/2022]
Abstract
Atherosclerosis is the major cause of coronary artery disease (CAD) which includes unstable angina, myocardial infarction, and heart failure. The onset of atherogenesis, a process of atherosclerotic lesion formation in the intima of arteries, is driven by lipid accumulation, a vicious cycle of reactive oxygen species (ROS)-induced oxidative stress and inflammatory reactions leading to endothelial cell (EC) dysfunction, vascular smooth muscle cell (VSMC) activation, and foam cell formation which further fuel plaque formation and destabilization. In recent years, there is a surge in the number of publications reporting the involvement of circular RNAs (circRNAs) in the pathogenesis of cardiovascular diseases, cancers, and metabolic syndromes. These studies have advanced our understanding on the biological functions of circRNAs. One of the most common mechanism of action of circRNAs reported is the sponging of microRNAs (miRNAs) by binding to the miRNAs response element (MRE), thereby indirectly increases the transcription of their target messenger RNAs (mRNAs). Individual networks of circRNA-miRNA-mRNA associated with atherogenesis have been extensively reported, however, there is a need to connect these findings for a complete overview. This review aims to provide an update on atherogenesis-related circRNAs and analyze the circRNA-miRNA-mRNA interactions in atherogenesis. The atherogenic mechanisms and clinical relevance of each atherogenesis-related circRNA were systematically discussed for better understanding of the knowledge gap in this area.
Collapse
Affiliation(s)
- Kind-Leng Tong
- Department of Pharmacology, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Ke-En Tan
- Institute of Biological Sciences, Faculty of Science, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Yat-Yuen Lim
- Institute of Biological Sciences, Faculty of Science, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Xin-Yi Tien
- Department of Pharmacology, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Pooi-Fong Wong
- Department of Pharmacology, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia.
| |
Collapse
|
7
|
Vascular Protective Effect and Its Possible Mechanism of Action on Selected Active Phytocompounds: A Review. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:3311228. [PMID: 35469164 PMCID: PMC9034927 DOI: 10.1155/2022/3311228] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 03/22/2022] [Accepted: 03/30/2022] [Indexed: 12/16/2022]
Abstract
Vascular endothelial dysfunction is characterized by an imbalance of vasodilation and vasoconstriction, deficiency of nitric oxide (NO) bioavailability and elevated reactive oxygen species (ROS), and proinflammatory factors. This dysfunction is a key to the early pathological development of major cardiovascular diseases including hypertension, atherosclerosis, and diabetes. Therefore, modulation of the vascular endothelium is considered an important therapeutic strategy to maintain the health of the cardiovascular system. Epidemiological studies have shown that regular consumption of medicinal plants, fruits, and vegetables promotes vascular health, lowering the risk of cardiovascular diseases. This is mainly attributed to the phytochemical compounds contained in these resources. Various databases, including Google Scholar, MEDLINE, PubMed, and the Directory of Open Access Journals, were searched to identify studies demonstrating the vascular protective effects of phytochemical compounds. The literature had revealed abundant data on phytochemical compounds protecting and improving the vascular system. Of the numerous compounds reported, curcumin, resveratrol, cyanidin-3-glucoside, berberine, epigallocatechin-3-gallate, and quercetin are discussed in this review to provide recent information on their vascular protective mechanisms in vivo and in vitro. Phytochemical compounds are promising therapeutic agents for vascular dysfunction due to their antioxidative mechanisms. However, future human studies will be necessary to confirm the clinical effects of these vascular protective mechanisms.
Collapse
|
8
|
Abstract
Regulatory RNAs like microRNAs (miRNAs) and long non-coding RNAs (lncRNAs) control vascular and immune cells' phenotype and thus play a crucial role in atherosclerosis. Moreover, the mutual interactions between miRNAs and lncRNAs link both types of regulatory RNAs in a functional network that affects lesion formation. In this review, we deduce novel concepts of atherosclerosis from the analysis of the current data on regulatory RNAs' role in endothelial cells (ECs) and macrophages. In contrast to arterial ECs, which adopt a stable phenotype by adaptation to high shear stress, macrophages are highly plastic and quickly change their activation status. At predilection sites of atherosclerosis, such as arterial bifurcations, ECs are exposed to disturbed laminar flow, which generates a dysadaptive stress response mediated by miRNAs. Whereas the highly abundant miR-126-5p promotes regenerative proliferation of dysadapted ECs, miR-103-3p stimulates inflammatory activation and impairs endothelial regeneration by aberrant proliferation and micronuclei formation. In macrophages, miRNAs are essential in regulating energy and lipid metabolism, which affects inflammatory activation and foam cell formation.Moreover, lipopolysaccharide-induced miR-155 and miR-146 shape inflammatory macrophage activation through their oppositional effects on NF-kB. Most lncRNAs are not conserved between species, except a small group of very long lncRNAs, such as MALAT1, which blocks numerous miRNAs by providing non-functional binding sites. In summary, regulatory RNAs' roles are highly context-dependent, and therapeutic approaches that target specific functional interactions of miRNAs appear promising against cardiovascular diseases.
Collapse
Affiliation(s)
- Andreas Schober
- Institute for Cardiovascular Prevention, University Hospital, Ludwig-Maximilians-University, Munich, Germany.
| | - Saffiyeh Saboor Maleki
- Institute for Cardiovascular Prevention, University Hospital, Ludwig-Maximilians-University, Munich, Germany
| | - Maliheh Nazari-Jahantigh
- Institute for Cardiovascular Prevention, University Hospital, Ludwig-Maximilians-University, Munich, Germany
| |
Collapse
|
9
|
Xiao M, Bai S, Chen J, Li Y, Zhang S, Hu Z. CDKN2B-AS1 participates in high glucose-induced apoptosis and fibrosis via NOTCH2 through functioning as a miR-98-5p decoy in human podocytes and renal tubular cells. Diabetol Metab Syndr 2021; 13:107. [PMID: 34649592 PMCID: PMC8518318 DOI: 10.1186/s13098-021-00725-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Accepted: 09/24/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Diabetic nephropathy (DN) is the most common causes of end-stage renal disease. Long non-coding RNA cyclin-dependent kinase inhibitor 2B antisense RNA 1 (CDKN2B-AS1) is connected with the development of DN, but the role of CDKN2B-AS1 in DN has not been entirely elucidated. METHODS Quantitative real-time polymerase chain reaction (qRT-PCR) was carried out to measure CDKN2B-AS1 and miR-98-5p levels. Cell viability, proliferation, and apoptosis were analyzed with 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide (MTT) or flow cytometry assays. Protein levels were measured by western blotting. The relationship between CDKN2B-AS1 or notch homolog 2 (NOTCH2) and miR-98-5p was verified via dual-luciferase reporter assay. RESULTS CDKN2B-AS1 and NOTCH2 were upregulated in the serum of DN patients and high glucose-disposed human podocytes (HPCs) and human renal tubular cells (HK-2), whereas miR-98-5p was downregulated. High glucose repressed viability and accelerated apoptosis of HPCs and HK-2 cells. CDKN2B-AS1 knockdown impaired high glucose-induced apoptosis and fibrosis of HPCs and HK-2 cells. Mechanistically, CDKN2B-AS1 sponged miR-98-5p to regulate NOTCH2 expression. Also, CDKN2B-AS1 inhibition-mediated effects on apoptosis and fibrosis of high glucose-disposed HPCs and HK-2 cells were weakened by miR-98-5p inhibitor. Also, NOTCH2 knockdown partly reversed miR-98-5p inhibitor-mediated impacts on apoptosis and fibrosis of high glucose-disposed HPCs and HK-2 cells. CONCLUSION High glucose-induced CDKN2B-AS1 promoted apoptosis and fibrosis via the TGF-β1 signaling mediated by the miR-98-5p/NOTCH2 axis in HPCs and HK-2 cells.
Collapse
Affiliation(s)
- Min Xiao
- Department of Nephrology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhua West Road, Jinan, 250012, Shandong, China
| | - Shoujun Bai
- Department of Nephrology, Qingpu Branch of Zhongshan Hospital Affiliated To Fudan University, Shanghai, 201700, China
| | - Jing Chen
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China
| | - Yaxi Li
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77004, USA
| | - Shu Zhang
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77004, USA
| | - Zhao Hu
- Department of Nephrology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhua West Road, Jinan, 250012, Shandong, China.
| |
Collapse
|
10
|
Zhang Q, Yan YF, Lv Q, Li YJ, Wang RR, Sun GB, Pan L, Hu JX, Xie N, Zhang C, Tian BC, Jiao F, Xu S, Wang PY, Xie SY. miR-4293 upregulates lncRNA WFDC21P by suppressing mRNA-decapping enzyme 2 to promote lung carcinoma proliferation. Cell Death Dis 2021; 12:735. [PMID: 34301920 PMCID: PMC8302752 DOI: 10.1038/s41419-021-04021-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 07/10/2021] [Accepted: 07/12/2021] [Indexed: 12/19/2022]
Abstract
Non-coding RNAs (ncRNAs) involve in diverse biological processes by post-transcriptional regulation of gene expression. Emerging evidence shows that miRNA-4293 plays a significant role in the development of non-small cell lung cancer. However, the oncogenic functions of miR-4293 have not been studied. Our results demonstrated that miR-4293 expression is markedly enhanced in lung carcinoma tissue and cells. Moreover, miR-4293 promotes tumor cell proliferation and metastasis but suppresses apoptosis. Mechanistic investigations identified mRNA-decapping enzyme 2 (DCP2) as a target of miR-4293 and its expression is suppressed by miR-4293. DCP2 can directly or indirectly bind to WFDC21P and downregulates its expression. Consequently, miR-4293 can further promote WFDC21P expression by regulating DCP2. With a positive correlation to miR-4293 expression, WFDC21P also plays an oncogenic role in lung carcinoma. Furthermore, knockdown of WFDC21P results in functional attenuation of miR-4293 on tumor promotion. In vivo xenograft growth is also promoted by both miR-4293 and WFDC21P. Overall, our results establish oncogenic roles for both miR-4293 and WFDC21P and demonstrate that interactions between miRNAs and lncRNAs through DCP2 are important in the regulation of carcinoma pathogenesis. These results provided a valuable theoretical basis for the discovery of lung carcinoma therapeutic targets and diagnostic markers based on miR-4293 and WFDC21P.
Collapse
MESH Headings
- Adult
- Aged
- Animals
- Apoptosis/genetics
- Base Sequence
- Carcinogenesis/genetics
- Carcinogenesis/pathology
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/pathology
- Cell Line, Tumor
- Cell Movement/genetics
- Cell Proliferation/genetics
- Female
- Gene Expression Regulation, Neoplastic
- Gene Knockdown Techniques
- Humans
- Lung Neoplasms/genetics
- Lung Neoplasms/pathology
- Male
- Mice, Inbred BALB C
- Mice, Nude
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Middle Aged
- Models, Biological
- Protein Binding
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- STAT3 Transcription Factor/metabolism
- Up-Regulation/genetics
- Mice
Collapse
Affiliation(s)
- Qian Zhang
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, YanTai, ShanDong, P. R. China
| | - Yun-Fei Yan
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, YanTai, ShanDong, P. R. China
| | - Qing Lv
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, YanTai, ShanDong, P. R. China
| | - You-Jie Li
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, YanTai, ShanDong, P. R. China
| | - Ran-Ran Wang
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, YanTai, ShanDong, P. R. China
| | - Guang-Bin Sun
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, YanTai, ShanDong, P. R. China
| | - Li Pan
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, YanTai, ShanDong, P. R. China
| | - Jin-Xia Hu
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, YanTai, ShanDong, P. R. China
| | - Ning Xie
- Department of Chest Surgery, YanTaiShan Hospital, YanTai, ShanDong, P. R. China
| | - Can Zhang
- Genetics and Aging Research Unit, Mass General Institute for Neurodegenerative Diseases (MIND), Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Bao-Cheng Tian
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, YanTai, ShanDong, P. R. China
| | - Fei Jiao
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, YanTai, ShanDong, P. R. China
| | - Sen Xu
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, YanTai, ShanDong, P. R. China
| | - Ping-Yu Wang
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, YanTai, ShanDong, P. R. China.
| | - Shu-Yang Xie
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, YanTai, ShanDong, P. R. China.
| |
Collapse
|
11
|
Circulating miR-206 and Wnt-signaling are associated with cardiovascular complications and a history of preeclampsia in women. Clin Sci (Lond) 2020; 134:87-101. [PMID: 31899480 PMCID: PMC8299351 DOI: 10.1042/cs20190920] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 12/17/2019] [Accepted: 01/03/2020] [Indexed: 02/06/2023]
Abstract
Women with a history of preeclampsia (PE) have increased risk of cardiovascular disease (CVD) later in life. However, the molecular determinants underlying this risk remain unclear. We sought to understand how circulating miRNA levels are affected by prior PE, and related to biological pathways underpinning cardiovascular disease. RNA sequencing was used to profile plasma levels of 2578 miRNAs in a retrospective study of women with a history of PE or normotensive pregnancy, in two independent cohorts with either acute coronary syndrome (ACS) (n = 17–18/group) or no ACS (n = 20/group). Differential miRNA alterations were assessed in relation to a history of PE (within each cohort) or ACS (across cohorts), and compared with miRNAs previously reported to be altered during PE. A history of PE was associated with altered levels of 30 and 20 miRNAs in the ACS and non-ACS cohorts, respectively, whereas ACS exposure was associated with alterations in 259 miRNAs. MiR-206 was identified at the intersection of all comparisons relating to past/current PE and ACS exposure, and has previously been implicated in atherogenic activities related to hepatocytes, vascular smooth muscle cells and macrophages. Integration of all differentially altered miRNAs with their predicted and experimentally validated targets in silico revealed a number of highly targeted genes with potential atherogenic functions (including NFAT5, CCND2 and SMAD2), and one significantly enriched KEGG biological pathway (Wnt signaling) that was shared between all exposure groups. The present study provides novel insights into miRNAs, target genes and biological pathways that may underlie the long-term cardiovascular sequelae of PE.
Collapse
|
12
|
Reiterer M, Branco CM. Endothelial cells and organ function: applications and implications of understanding unique and reciprocal remodelling. FEBS J 2019; 287:1088-1100. [PMID: 31736207 PMCID: PMC7155104 DOI: 10.1111/febs.15143] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 10/21/2019] [Accepted: 11/15/2019] [Indexed: 12/16/2022]
Abstract
The microvasculature is a heterogeneous, dynamic and versatile component of the systemic circulation, with a unique ability to locally self-regulate and to respond to organ demand and environmental stimuli. Endothelial cells from different organs display considerable variation, but it is currently unclear to what extent functional properties of organ-specific endothelial cells are intrinsic, acquired and/or reprogrammable. Vascular function is a fundamental pillar of homeostasis, and dysfunction results in systemic consequences for the organism. Additionally, vascular failure can occur downstream of organ disease or environmental stress, often driving an exacerbation of symptoms and pathologies originally independent of the local circulation. The understanding of the molecular mechanisms underlying endothelial physiology and metabolism holds the promise to inform and improve diagnosis, prognosis and treatment options for a myriad of conditions as unrelated as cancer, neurodegeneration or pulmonary hypertension, and likely everything in between, if we consider that also treatments for such conditions are primarily distributed via the bloodstream. However, studying endothelial function has its challenges: the origin, isolation, culture conditions and preconditioning stimuli make this an extremely variable cell type to study and difficult to source. Animal models exist but are neither trivial to generate, nor necessarily adequately translatable to human disease. In this article, we aim to illustrate the breadth of microvascular functions in different environments, highlighting current and pioneering studies that have advanced our insight into the importance of the integrity of this tissue, as well as the limitations posed by its heterogeneity and plasticity.
Collapse
Affiliation(s)
- Moritz Reiterer
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, UK.,Department of Physiology, Development and Neuroscience, University of Cambridge, UK
| | - Cristina M Branco
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, UK
| |
Collapse
|
13
|
Rong ZH, Chang NB, Yao QP, Li T, Zhu XL, Cao Y, Jiang MJ, Cheng YS, Jiang R, Jiang J. Suppression of circDcbld1 Alleviates Intimal Hyperplasia in Rat Carotid Artery by Targeting miR-145-3p/Neuropilin-1. MOLECULAR THERAPY-NUCLEIC ACIDS 2019; 18:999-1008. [PMID: 31778958 PMCID: PMC6889766 DOI: 10.1016/j.omtn.2019.10.023] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Revised: 10/22/2019] [Accepted: 10/23/2019] [Indexed: 12/21/2022]
Abstract
We replicated the rat common carotid artery (CCA) intima hyperplasia model and found the expression of a circular RNA, circRNA_009723 (circDcbld1), was markedly increased in the CCA with intimal hyperplasia. In vitro, the suppression of circDcbld1 in rat vascular smooth muscle cells (VSMCs) led the increase of contractile smooth muscle cell markers and the decrease of cell migration. In vivo, the injection of chemically modified circDcbld1 small interfering RNA (siRNA) lessened the formation of neointima in rat CCA after balloon injury. Further experiments proved that circDcbld1, as a competing endogenous RNA, interacted with miR-145-3p and upregulated the level of neuropilin-1 (Nrp1), thereby regulating the migration of VSMCs. In this study, we demonstrated a new mechanism by which circular RNA promotes intimal hyperplasia. We deem that intervention in the circDcbld1-miR-145-3p/Nrp1 pathway might be a feasible approach to alleviate the post-injury intimal hyperplasia.
Collapse
Affiliation(s)
- Zhi-Hua Rong
- Department of Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Neng-Bin Chang
- Department of Anatomy, Southwest Medical University, Luzhou, China
| | - Qing-Ping Yao
- School of Life Sciences and Biotechnology, Institute of Mechanobiology and Medical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Tao Li
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease/Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Xiao-Ling Zhu
- Department of Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yu Cao
- Department of Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Mei-Jun Jiang
- School of Clinical Medicine, Southwest Medical University, Luzhou, China
| | - Yan-Shuang Cheng
- School of Clinical Medicine, Southwest Medical University, Luzhou, China
| | - Rui Jiang
- Department of Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China; Nephropathy Clinical Medical Research Center of Sichuan Province, Luzhou, China
| | - Jun Jiang
- Department of Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China; Key Laboratory of Medical Electrophysiology, Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease/Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China.
| |
Collapse
|
14
|
Comparative Transcriptomics of Ex Vivo, Patient-Derived Endothelial Cells Reveals Novel Pathways Associated With Type 2 Diabetes Mellitus. JACC Basic Transl Sci 2019; 4:567-574. [PMID: 31768474 PMCID: PMC6872769 DOI: 10.1016/j.jacbts.2019.05.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 05/23/2019] [Accepted: 05/23/2019] [Indexed: 12/18/2022]
Abstract
Endothelial cells can be harvested directly from humans, rapidly sorted and subjected to RNA-sequencing to study global gene expression. In endothelial cells isolated from patients with type 2 diabetes mellitus, pathways involved in TGF-β and Cyclin-D1 signaling were positively enriched while androgen signaling and oxidative phosphorylation were negatively enriched compared to healthy individuals. Patient-derived endothelial cells can be used to discover and validate disease-associated pathways.
In this study low-input RNA-sequencing was used to annotate the molecular identity of endothelial cells isolated and immunopurified with CD144 microbeads. Using this technique, comparative gene expression profiling from healthy subjects and patients with type 2 diabetes mellitus identified both known and novel pathways linked with EC dysfunction. Modeling of diabetes by treating cultured ECs with high glucose identified shared changes in gene expression in diabetic cells. Overall, the data demonstrate how purified ECs from patients can be used to generate new hypotheses about mechanisms of human vascular disease.
Collapse
Key Words
- BSA, bovine serum albumin
- EC, endothelial cell
- EDTA, ethylenediamine tetra-acetic acid
- FACS, fluorescence activated cell sorting
- FDR, false discovery rate
- GSEA, gene set enrichment analysis
- HUVEC, human umbilical vein endothelial cell
- IV, intravenous
- PBS, phosphate buffered saline
- Seq, sequencing
- T2DM, type 2 diabetes mellitus
- TGFβ, transforming growth factor beta
- VEGF, vascular endothelial growth factor
- VUMC, Vanderbilt University Medical Center
- WBC, white blood cell
- ddCt, delta-delta cycle threshold
- diabetes mellitus
- endothelial cell dysfunction
- endothelial cells
- gene expression
- qPCR, quantitative polymerase chain reaction
Collapse
|
15
|
Bencsik P, Kiss K, Ágg B, Baán JA, Ágoston G, Varga A, Gömöri K, Mendler L, Faragó N, Zvara Á, Sántha P, Puskás LG, Jancsó G, Ferdinandy P. Sensory Neuropathy Affects Cardiac miRNA Expression Network Targeting IGF-1, SLC2a-12, EIF-4e, and ULK-2 mRNAs. Int J Mol Sci 2019; 20:ijms20040991. [PMID: 30823517 PMCID: PMC6412859 DOI: 10.3390/ijms20040991] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 02/14/2019] [Accepted: 02/19/2019] [Indexed: 12/20/2022] Open
Abstract
Background: Here we examined myocardial microRNA (miRNA) expression profile in a sensory neuropathy model with cardiac diastolic dysfunction and aimed to identify key mRNA molecular targets of the differentially expressed miRNAs that may contribute to cardiac dysfunction. Methods: Male Wistar rats were treated with vehicle or capsaicin for 3 days to induce systemic sensory neuropathy. Seven days later, diastolic dysfunction was detected by echocardiography, and miRNAs were isolated from the whole ventricles. Results: Out of 711 known miRNAs measured by miRNA microarray, the expression of 257 miRNAs was detected in the heart. As compared to vehicle-treated hearts, miR-344b, miR-466b, miR-98, let-7a, miR-1, miR-206, and miR-34b were downregulated, while miR-181a was upregulated as validated also by quantitative real time polymerase chain reaction (qRT-PCR). By an in silico network analysis, we identified common mRNA targets (insulin-like growth factor 1 (IGF-1), solute carrier family 2 facilitated glucose transporter member 12 (SLC2a-12), eukaryotic translation initiation factor 4e (EIF-4e), and Unc-51 like autophagy activating kinase 2 (ULK-2)) targeted by at least three altered miRNAs. Predicted upregulation of these mRNA targets were validated by qRT-PCR. Conclusion: This is the first demonstration that sensory neuropathy affects cardiac miRNA expression network targeting IGF-1, SLC2a-12, EIF-4e, and ULK-2, which may contribute to cardiac diastolic dysfunction. These results further support the need for unbiased omics approach followed by in silico prediction and validation of molecular targets to reveal novel pathomechanisms.
Collapse
Affiliation(s)
- Péter Bencsik
- Cardiovascular Research Group, Department of Biochemistry, University of Szeged, Dóm tér 9, H-6720 Szeged, Hungary.
- Pharmahungary Group, Graphisoft Park, Záhony utca 7, H-1031 Budapest, Hungary.
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Dóm tér 12, H-6720 Szeged, Hungary.
| | - Krisztina Kiss
- Cardiovascular Research Group, Department of Biochemistry, University of Szeged, Dóm tér 9, H-6720 Szeged, Hungary.
| | - Bence Ágg
- Pharmahungary Group, Graphisoft Park, Záhony utca 7, H-1031 Budapest, Hungary.
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Nagyvárad tér 4, H-1085 Budapest, Hungary.
- Heart and Vascular Center, Semmelweis University, Városmajor utca 68, H-1122 Budapest, Hungary.
| | - Júlia A Baán
- Muscle Adaptation Group, Department of Biochemistry, University of Szeged, Dóm tér 9, H-6720 Szeged, Hungary.
| | - Gergely Ágoston
- Institute of Family Medicine, University of Szeged, Tisza Lajos krt. 109., H-6720 Szeged, Hungary.
| | - Albert Varga
- Institute of Family Medicine, University of Szeged, Tisza Lajos krt. 109., H-6720 Szeged, Hungary.
| | - Kamilla Gömöri
- Pharmahungary Group, Graphisoft Park, Záhony utca 7, H-1031 Budapest, Hungary.
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Dóm tér 12, H-6720 Szeged, Hungary.
| | - Luca Mendler
- Muscle Adaptation Group, Department of Biochemistry, University of Szeged, Dóm tér 9, H-6720 Szeged, Hungary.
- Institute of Biochemistry II, Goethe University Medical School, University Hospital Building 75, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany.
| | - Nóra Faragó
- Institute of Genetics, Biological Research Center, Hungarian Academy of Sciences, Temesvári körút 62, H-6726 Szeged, Hungary.
| | - Ágnes Zvara
- Institute of Genetics, Biological Research Center, Hungarian Academy of Sciences, Temesvári körút 62, H-6726 Szeged, Hungary.
| | - Péter Sántha
- Department of Physiology, University of Szeged, Dóm tér 10, H-6720 Szeged, Hungary.
| | - László G Puskás
- Institute of Genetics, Biological Research Center, Hungarian Academy of Sciences, Temesvári körút 62, H-6726 Szeged, Hungary.
| | - Gábor Jancsó
- Department of Physiology, University of Szeged, Dóm tér 10, H-6720 Szeged, Hungary.
| | - Péter Ferdinandy
- Pharmahungary Group, Graphisoft Park, Záhony utca 7, H-1031 Budapest, Hungary.
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Nagyvárad tér 4, H-1085 Budapest, Hungary.
| |
Collapse
|
16
|
Huang R, Hu Z, Cao Y, Li H, Zhang H, Su W, Xu Y, Liang L, Melgiri ND, Jiang L. MiR-652-3p inhibition enhances endothelial repair and reduces atherosclerosis by promoting Cyclin D2 expression. EBioMedicine 2019; 40:685-694. [PMID: 30674440 PMCID: PMC6413686 DOI: 10.1016/j.ebiom.2019.01.032] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Revised: 01/12/2019] [Accepted: 01/14/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Atherosclerosis is a hyperlipidemia-induced condition affecting the arterial wall that damages healthy endothelial cell (EC) function, leading to enhanced risk of atherothrombotic events. Certain microRNAs regulate EC dysfunction in response to hyperlipidemia and may be suitable therapeutic targets to combat atherosclerosis. METHODS miRNA expression in human ECs was analyzed under various conditions to identify key microRNAs. High-cholesterol diet (HCD)-fed Mir652-/-Apoe-/- (Mir652-/-) mice and matching Mir652+/+Apoe-/- (Mir652+/+) mice were subjected to carotid injury to analyze the effects of miR-652 knockdown on endothelial repair. In silico analysis followed by in vitro and in vivo experiments were applied to identify miR-652's target gene Ccnd2 and investigate the pair's effects on ECs. miR-652-5p and miR-652-3p antagomir therapies were tested in Mir652+/+ mice under normal and HCD diet to assess their effect on endothelial repair. FINDINGS miR-652-3p, which is upregulated in human and murine atherosclerotic plaques, suppresses expression of the endothelial repair gene Ccnd2, thereby enhancing atherosclerotic lesion formation. Post-denudation recovery of ECs was promoted in Mir652-/- mice due to enhanced EC proliferation attributable to de-repression of miR-652-3p's (but not miR-652-5p's) regulation of Ccnd2 expression. Under hyperlipidemic conditions at non-predilection sites, miR-652-3p produces anti-proliferative effects in ECs, such that Mir652-/- mice display reduced atherosclerotic progression. In contrast, neither miR-652-3p nor Ccnd2 displayed significant effects on the endothelium at predilection sites or under disturbed flow conditions. Administration of a miR-652-3p antagomir rescued the proliferation of ECs in vivo, thereby limiting atherosclerotic development. INTERPRETATION miR-652-3p blockade may be a potential therapeutic strategy against atherosclerosis.
Collapse
Affiliation(s)
- Rongzhong Huang
- Department of Cardiothoracic Surgery, The First People's Hospital of Yunnan Province, Kunming, Yunnan Province, China
| | - Zicheng Hu
- Department of Neurology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Yu Cao
- Department of Cardiothoracic Surgery, The First People's Hospital of Yunnan Province, Kunming, Yunnan Province, China
| | - Hongrong Li
- Department of Cardiothoracic Surgery, The First People's Hospital of Yunnan Province, Kunming, Yunnan Province, China
| | - Hong Zhang
- Department of Cardiology, The First People's Hospital of Yunnan Province, Kunming, Yunnan Province, China
| | - Wenhua Su
- Department of Cardiology, The First People's Hospital of Yunnan Province, Kunming, Yunnan Province, China
| | - Yu Xu
- Statistical Laboratory, Chuangxu Institute of Lifescience, Chongqing, China
| | - Liwen Liang
- Department of Cardiology, The First People's Hospital of Yunnan Province, Kunming, Yunnan Province, China
| | - N D Melgiri
- Impactys Foundation for Biomedical Research, San Diego, CA, USA.
| | - Lihong Jiang
- Department of Cardiothoracic Surgery, The First People's Hospital of Yunnan Province, Kunming, Yunnan Province, China.
| |
Collapse
|
17
|
Ma Y, Mao Y, An Y, Tian T, Zhang H, Yan J, Zhu Z, Yang CJ. Target-responsive DNA hydrogel for non-enzymatic and visual detection of glucose. Analyst 2019; 143:1679-1684. [PMID: 29512663 DOI: 10.1039/c8an00010g] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
We have successfully developed a target-responsive aptamer cross-linked hydrogel for the visual detection of glucose, an important biomedical analyte. In this work, the glucose-responsive hydrogel was prepared using the target aptamer and its two short complementary DNA strands grafted onto a linear polyacrylamide chain as cross-linkers. Gold nanoparticles (AuNPs) modified with thiol-PEG were encapsulated in the gel and used as the output signal for visible detection. The complex of glucose and its ligand of boronic acid derivatives (Shinkai's receptor) can bind with the aptamer to disrupt the hydrogel, leading to the release of AuNPs with a distinct red colour in the supernatant. By this method glucose can be detected with the naked eye, and the sensor has a detection limit of 0.44 mM in buffer with the help of UV-Vis spectrophotometry. Furthermore, glucose spiked in 50% urine and 30% serum could also be detected respectively with the naked eye, and glucose was quantitatively detected in 50% urine. The hydrogel system provides a non-enzymatic and visual method for glucose detection, and offers promising applications in biotechnology and biomedicine.
Collapse
Affiliation(s)
- Yanli Ma
- MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Collaborative Innovation Center of Chemistry for Energy Materials, Key Laboratory for Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China.
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Zhu Y, Xu J, Liang W, Li J, Feng L, Zheng P, Ji T, Bai S. miR-98-5p Alleviated Epithelial-to-Mesenchymal Transition and Renal Fibrosis via Targeting Hmga2 in Diabetic Nephropathy. Int J Endocrinol 2019; 2019:4946181. [PMID: 31885559 PMCID: PMC6925681 DOI: 10.1155/2019/4946181] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 08/01/2019] [Accepted: 09/17/2019] [Indexed: 12/13/2022] Open
Abstract
Recently, microRNAs have been recognized as crucial regulators of diabetic nephropathy (DN) development. Epithelial-to-mesenchymal transition (EMT) can play a significant role in tubulointerstitial fibrosis, and it is a hallmark of diabetic nephropathy progression. Nevertheless, the function of miR-98-5p in the modulation of EMT and renal fibrosis during DN remains barely investigated. Hence, identifying the mechanisms of miR-98-5p in regulating EMT and fibrosis is of huge significance. In our present research, decreased miR-98-5p was demonstrated in db/db mice and mice mesangial cells treated with the high dose of glucose. Meanwhile, activated EMT and increased fibrosis was accompanied with the decrease of miR-98-5p in vitro and in vivo. Additionally, to further find out the roles of miR-98-5p in DN development, overexpression of miR-98-5p was applied. Firstly, in vivo investigation exhibited that elevation of miR-98-5p restrained proteinuria, serum creatinine, BUN, the EMT process, and fibrosis. Furthermore, high glucose was able to promote mice mesangial cell proliferation, EMT process, and induced renal fibrosis, which could be prevented by overexpression of miR-98-5p. Moreover, high mobility group A (HMGA2) can exhibit an important role in diverse biological processes. Here, HMGA2 was investigated as a target of miR-98-5p currently. Luciferase reporter assay was conducted and the correlation of miR-98-5p and HMGA2 was validated. Moreover, it was displayed that HMGA2 was remarkably elevated in db/db mice and mice mesangial cells. Furthermore, miR-98-5p strongly depressed HMGA2 protein and mRNA levels in mice mesangial cells. Overall, these revealed miR-98-5p could suppress the EMT process and renal fibrosis through targeting HMGA2 in DN.
Collapse
Affiliation(s)
- Yingchun Zhu
- Department of Nephrology, Qingpu Branch of Zhongshan Hospital Affiliated to Fudan University, 1158 Gongyuan East Road, Qingpu District, Shanghai 201700, China
| | - Jiang Xu
- Department of Rehabilitation, Huai'an Second People's Hospital, The Affiliated Hospital of Xuzhou Medical University, Huai'an, China
| | - Wenxing Liang
- Department of Nephrology, Qingpu Branch of Zhongshan Hospital Affiliated to Fudan University, 1158 Gongyuan East Road, Qingpu District, Shanghai 201700, China
| | - Ji Li
- Department of Nephrology, Qingpu Branch of Zhongshan Hospital Affiliated to Fudan University, 1158 Gongyuan East Road, Qingpu District, Shanghai 201700, China
| | - Linhong Feng
- Department of Nephrology, Qingpu Branch of Zhongshan Hospital Affiliated to Fudan University, 1158 Gongyuan East Road, Qingpu District, Shanghai 201700, China
| | - PengXi Zheng
- Department of Nephrology, Qingpu Branch of Zhongshan Hospital Affiliated to Fudan University, 1158 Gongyuan East Road, Qingpu District, Shanghai 201700, China
| | - Tingting Ji
- Department of Nephrology, Qingpu Branch of Zhongshan Hospital Affiliated to Fudan University, 1158 Gongyuan East Road, Qingpu District, Shanghai 201700, China
| | - Shoujun Bai
- Department of Nephrology, Qingpu Branch of Zhongshan Hospital Affiliated to Fudan University, 1158 Gongyuan East Road, Qingpu District, Shanghai 201700, China
| |
Collapse
|
19
|
Huang Z, Li N, Shan Y, Liang C. Hsa-miRNA-29a protects against high glucose-induced damage in human umbilical vein endothelial cells. J Cell Biochem 2018; 120:5860-5868. [PMID: 30520097 DOI: 10.1002/jcb.27871] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 09/19/2018] [Indexed: 12/30/2022]
Abstract
Sustained exposure to high glucose (HG) results in dysfunction of vascular endothelial cells. Hence, diabetic patients often suffer from secondary vascular damages, such as vascular sclerosis and thrombogenesis, which may eventually cause cardiovascular problems. Thus, elucidating how HG results in vascular endothelial cell damage and finding an approach for prevention are important to prevent and treat vascular damages in diabetic patients. In the current study, we first showed that 72-hour exposure to HG-decreased hsa-miRNA-29a and increased the expression of Bcl-2 associated X protein (Bax), which subsequently inhibited Bcl-2 and promoted the expression of apoptotic protease activating factor-1 and activation of caspase-3, thus directly triggering the mitochondrial apoptotic pathway in human umbilical vein endothelial cells (HUVECs). Study of the underlying mechanism showed that hsa-miRNA-29a/Bax plays an essential role in the decreased proliferation and increased apoptosis of HUVECs induced by HG, and overexpression of hsa-miRNA-29a effectively inhibits HG-induced apoptosis and restores the proliferation and tube formation of HUVECs exposed to HG by inhibiting its target gene Bax. In short, our study demonstrates that hsa-miRNA-29a is a promising target for the prevention and treatment of vascular injury in diabetic patients.
Collapse
Affiliation(s)
- Zhigang Huang
- Department of Cardiology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Na Li
- Department of Cardiology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Yi Shan
- Department of Emergency and ICU, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Chun Liang
- Department of Cardiology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| |
Collapse
|
20
|
Gestational diabetes alters microRNA signatures in human feto-placental endothelial cells depending on fetal sex. Clin Sci (Lond) 2018; 132:2437-2449. [DOI: 10.1042/cs20180825] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 10/24/2018] [Accepted: 11/01/2018] [Indexed: 12/12/2022]
Abstract
MicroRNAs (miRNAs), small non-coding RNAs, have emerged as important, epigenetic regulators of endothelial function. Metabolic disturbances such as diabetes alter miRNA expression. In adults, the miRNA transcriptome as well as endothelial function differ between the sexes. Here, we hypothesized that metabolic disturbances associated with gestational diabetes (GDM) alter miRNA signatures in feto-placental endothelial cells (fpEC), dependent on fetal sex. We isolated human primary fpEC after normal and GDM-complicated pregnancies with male and female neonates and screened for differential miRNA expression using next-generation miRNA sequencing. To test for miRNAs commonly regulated in fpEC of female and male progeny, data were stratified for fetal sex and maternal body mass index (BMI). Analyses were also performed separately for female and male fpEC, again accounting for maternal BMI as covariate. Potential biological pathways regulated by the altered set of miRNAs were determined using mirPath software. Maternal GDM altered 26 miRNA signatures when male and female fpEC were analyzed together. Separate analysis of male versus female fpEC revealed 22 GDM affected miRNAs in the females and only 4 in the males, without overlap. Biological functions potentially modulated by the affected miRNAs related to ‘Protein Processing in Endoplasmic Reticulum’ and ‘Proteoglycans in Cancer’. Maternal GDM alters miRNA signatures in fpEC, and biological functions affected by these miRNAs relate to well-known adverse functional consequences of diabetes on endothelium. GDM effects were highly dependent on fetal sex with miRNA signatures in female fpEC being more susceptible to metabolic derangements of GDM than miRNAs in male fpEC.
Collapse
|
21
|
Xie L, Xu J. Role of MiR-98 and Its Underlying Mechanisms in Systemic Lupus Erythematosus. J Rheumatol 2018; 45:1397-1405. [DOI: 10.3899/jrheum.171290] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/30/2018] [Indexed: 12/12/2022]
Abstract
Objective.T-lymphocyte apoptosis plays a critical role in the pathogenesis of systemic lupus erythematosus (SLE). However, the underlying regulatory mechanisms of apoptosis in SLE remain unclear. The aim of this study was to explore the role of miR-98 in SLE and its underlying mechanisms.Methods.Western blotting and quantitative reverse transcription PCR (qRT-PCR) were used to analyze miR-98 and Fas expression. Luciferase reporter assays were performed to identify miR-98 targets. To modify miRNA levels, miR-98 mimics and inhibitor were transfected into cells. A lentiviral construct was used to overexpress the level of Fas in SLE CD4+ T cells. Gene and protein expression were determined by qRT-PCR and Western blotting. Apoptosis levels were evaluated by annexin V staining and flow cytometry.Results.Compared to those of healthy donors, miR-98 was downregulated in SLE CD4+ T cells, whereas Fas mRNA and protein expression were upregulated. Upregulation of miR-98 by mimic transfection protected Jurkat cells against Fas-mediated apoptosis at both mRNA and protein levels, while miR-98 inhibitor induced the completely opposite effect. Luciferase reporter assays demonstrated that miR-98 directly targeted Fas mRNA. Further, miR-98 inhibitor induced apoptosis in primary healthy CD4+ T cells through the Fas-caspase axis, while upregulation of miR-98 in SLE CD4+ T cells led to the opposite effect.Conclusion.The current study revealed that downregulation of miR-98 induces apoptosis by modulating the Fas-mediated apoptotic signaling pathway in SLE CD4+ T cells. These results suggest that miR-98 might serve as a potential target for SLE treatment.
Collapse
|
22
|
Xu YX, Huang C, Liu M, Chen N, Chen W, Yang C, Zhao Y, Li X, Duan J, Liu S, Yang S. Survivin regulated by autophagy mediates hyperglycemia-induced vascular endothelial cell dysfunction. Exp Cell Res 2018; 364:152-159. [DOI: 10.1016/j.yexcr.2018.01.037] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 12/21/2017] [Accepted: 01/28/2018] [Indexed: 12/11/2022]
|
23
|
Wang TY, Zhang J, Zhu J, Lian HY, Yuan HJ, Gao M, Luo MJ, Tan JH. Expression profiles and function analysis of microRNAs in postovulatory aging mouse oocytes. Aging (Albany NY) 2018; 9:1186-1201. [PMID: 28394765 PMCID: PMC5425121 DOI: 10.18632/aging.101219] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 03/28/2017] [Indexed: 01/07/2023]
Abstract
In this study, microRNA (miRNA) profiles in postovulatory aging mouse oocytes were analyzed by microarray screening and RT-qPCR. Hierarchical cluster analysis on the microarray data and KEGG pathway enrichment analysis on the mRNAs targeted by differentially expressed (DE) miRNAs between two adjacent egg-ages suggest that while only a mild alteration in miRNA expression occurred from 13 to 18 h, a great change took place from 18 to 24 h post hCG injection. Theoretical exploration on functions of the predicted target genes suggest that KEGG pathways enriched by 13-18 h DE miRNAs are correlated with early events of oocyte aging while pathways most enriched by 18-24 h or 24-30 h DE miRNAs are correlated with the late symptoms of aged oocytes. Experimental verification on functions of the key proteins predicted by the KEGG analysis and injection of miR-98 mimics or inhibitors further confirmed that miRNAs played stimulatory/inhibitory roles in postovulatory oocyte aging. In conclusion, marked changes in miRNA expression are associated with significant alterations in function and morphology of postovulatory aging oocytes.
Collapse
Affiliation(s)
- Tian-Yang Wang
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai-an City 271018, P. R. China
| | - Jie Zhang
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai-an City 271018, P. R. China
| | - Jiang Zhu
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai-an City 271018, P. R. China
| | - Hua-Yu Lian
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai-an City 271018, P. R. China
| | - Hong-Jie Yuan
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai-an City 271018, P. R. China
| | - Min Gao
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai-an City 271018, P. R. China
| | - Ming-Jiu Luo
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai-an City 271018, P. R. China
| | - Jing-He Tan
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai-an City 271018, P. R. China
| |
Collapse
|
24
|
Florijn BW, Bijkerk R, van der Veer EP, van Zonneveld AJ. Gender and cardiovascular disease: are sex-biased microRNA networks a driving force behind heart failure with preserved ejection fraction in women? Cardiovasc Res 2017; 114:210-225. [DOI: 10.1093/cvr/cvx223] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 11/23/2017] [Indexed: 01/08/2023] Open
Abstract
AbstractCardiovascular disease (CVD) is the primary cause of death among men and women worldwide. Nevertheless, our comprehension of how CVD progresses in women and elicits clinical outcomes is lacking, leading CVD to be under-diagnosed and under-treated in women. A clear example of this differential presentation of CVD pathophysiologies in females is the strikingly higher prevalence of heart failure with preserved ejection fraction (HFpEF). Women with a history of pre-eclampsia or those who present with co-morbidities such as obesity, hypertension, and diabetes mellitus are at increased risk of developing HFpEF. Long understood to be a critical CVD risk factor, our understanding of how gender differentially affects the development of CVD has been greatly expanded by extensive genomic and transcriptomic studies. These studies uncovered a pivotal role for differential microRNA (miRNA) expression in response to systemic inflammation, where their co-ordinated expression forms a post-transcriptional regulatory network that instigates microcirculation defects. Importantly, the potential sex-biased expression of the given miRNAs may explain sex-specific cardiovascular pathophysiologies in women, such as HFpEF. Sex-biased miRNAs are regulated by oestrogen (E2) in their transcription and processing or are expressed from loci on the X-chromosome due to incomplete X-chromosome inactivation. Interestingly, while E2-induced miRNAs predominantly appear to serve protective functions, it could be argued that many X-linked miRNAs have been found to challenge microvascular and myocardial integrity. Therefore, menopausal E2 deficiency, resulting in protective miRNA loss, and the augmentation of X-linked miRNA expression, may well contribute to the molecular mechanisms that underlie the female-specific cardiovascular aetiology in HFpEF.
Collapse
Affiliation(s)
- Barend W Florijn
- Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Albinusdreef 2, 2300 RC Leiden, The Netherlands
- Department of Internal Medicine (Nephrology), Leiden University Medical Center, Albinusdreef 2, 2300 RC Leiden, The Netherlands
| | - Roel Bijkerk
- Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Albinusdreef 2, 2300 RC Leiden, The Netherlands
- Department of Internal Medicine (Nephrology), Leiden University Medical Center, Albinusdreef 2, 2300 RC Leiden, The Netherlands
| | - Eric P van der Veer
- Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Albinusdreef 2, 2300 RC Leiden, The Netherlands
- Department of Internal Medicine (Nephrology), Leiden University Medical Center, Albinusdreef 2, 2300 RC Leiden, The Netherlands
| | - Anton Jan van Zonneveld
- Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Albinusdreef 2, 2300 RC Leiden, The Netherlands
- Department of Internal Medicine (Nephrology), Leiden University Medical Center, Albinusdreef 2, 2300 RC Leiden, The Netherlands
| |
Collapse
|
25
|
Liao B, Chen R, Lin F, Mai A, Chen J, Li H, Xu Z, Dong S. Long noncoding RNA HOTTIP promotes endothelial cell proliferation and migration via activation of the Wnt/β‐catenin pathway. J Cell Biochem 2017; 119:2797-2805. [DOI: 10.1002/jcb.26448] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 10/17/2017] [Indexed: 01/17/2023]
Affiliation(s)
- Bihong Liao
- Department of Cardiology, Shenzhen People's HospitalSecond Clinical Medical College of Jinan UniversityShenzhenGuangdong ProvinceChina
| | - Ruimian Chen
- Department of Cardiology, Shenzhen People's HospitalSecond Clinical Medical College of Jinan UniversityShenzhenGuangdong ProvinceChina
| | - Feng Lin
- Department of Cardiology, Shenzhen People's HospitalSecond Clinical Medical College of Jinan UniversityShenzhenGuangdong ProvinceChina
| | - Aihuan Mai
- Department of Cardiology, Shenzhen People's HospitalSecond Clinical Medical College of Jinan UniversityShenzhenGuangdong ProvinceChina
| | - Jie Chen
- Department of Cardiology, Shenzhen People's HospitalSecond Clinical Medical College of Jinan UniversityShenzhenGuangdong ProvinceChina
| | - Huimin Li
- Department of Cardiology, Shenzhen People's HospitalSecond Clinical Medical College of Jinan UniversityShenzhenGuangdong ProvinceChina
| | - Zhenglei Xu
- Department of Gastroenterology, Shenzhen People's HospitalSecond Clinical Medical College of Jinan UniversityShenzhenGuangdong ProvinceChina
| | - Shaohong Dong
- Department of Cardiology, Shenzhen People's HospitalSecond Clinical Medical College of Jinan UniversityShenzhenGuangdong ProvinceChina
| |
Collapse
|
26
|
Takagi K, Yamakuchi M, Matsuyama T, Kondo K, Uchida A, Misono S, Hashiguchi T, Inoue H. IL-13 enhances mesenchymal transition of pulmonary artery endothelial cells via down-regulation of miR-424/503 in vitro. Cell Signal 2017; 42:270-280. [PMID: 29102771 DOI: 10.1016/j.cellsig.2017.10.019] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 10/06/2017] [Accepted: 10/23/2017] [Indexed: 12/14/2022]
Abstract
Pulmonary arterial hypertension (PAH) has a major effect on life expectancy with functional degeneracy of the lungs and right heart. Interleukin-13 (IL-13), one of the type 2 cytokines mainly associated with allergic diseases, has recently been reported to be associated with Schistosomiasis-associated PAH which shares pathological features with other forms of PAH, such as idiopathic PAH and connective tissue disease-associated PAH. But a direct pathological role of IL-13 in the development of PAH has not been explored. We examined the effects of recombinant human IL-13 on the function of primary human pulmonary artery endothelial cells (HPAECs) to examine how IL-13 influences exacerbation of PAH. IL-13 increased the expression of Rictor, which is a key molecule of mammalian target of rapamycin complex 2. Treatment of IL-13 induced HPAEC migration via Rictor. Rictor was directly regulated by both miR-424 and 503 (miR-424/503). Therefore, IL-13 increases Rictor level by regulating miR-424/503, causing the increase of HPAEC migration. Since enhancement of HPAEC migration in the lung is thought to be associated with PAH, these data suggest that IL-13 takes some roles in exacerbating PAH.
Collapse
Affiliation(s)
- Koichi Takagi
- Department of Pulmonary Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1, Sakuragaoka, Kagoshima 890-8520, Japan; Department of Laboratory and Vascular Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1, Sakuragaoka, Kagoshima 890-8520, Japan
| | - Munekazu Yamakuchi
- Department of Laboratory and Vascular Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1, Sakuragaoka, Kagoshima 890-8520, Japan.
| | - Takahiro Matsuyama
- Department of Pulmonary Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1, Sakuragaoka, Kagoshima 890-8520, Japan
| | - Kiyotaka Kondo
- Department of Pulmonary Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1, Sakuragaoka, Kagoshima 890-8520, Japan
| | - Akifumi Uchida
- Department of Pulmonary Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1, Sakuragaoka, Kagoshima 890-8520, Japan
| | - Shunsuke Misono
- Department of Pulmonary Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1, Sakuragaoka, Kagoshima 890-8520, Japan
| | - Teruto Hashiguchi
- Department of Laboratory and Vascular Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1, Sakuragaoka, Kagoshima 890-8520, Japan
| | - Hiromasa Inoue
- Department of Pulmonary Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1, Sakuragaoka, Kagoshima 890-8520, Japan
| |
Collapse
|
27
|
Feng L, Ma X, Wang J, Tian Q. Up-regulation of 14-3-3β plays a role in intimal hyperplasia following carotid artery injury in diabetic Sprague Dawley rats by promoting endothelial cell migration and proliferation. Biochem Biophys Res Commun 2017; 490:1237-1243. [DOI: 10.1016/j.bbrc.2017.06.199] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2017] [Accepted: 06/30/2017] [Indexed: 12/16/2022]
|
28
|
McKee TJ, Komarova SV. Is it time to reinvent basic cell culture medium? Am J Physiol Cell Physiol 2017; 312:C624-C626. [PMID: 28228375 DOI: 10.1152/ajpcell.00336.2016] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 02/22/2017] [Accepted: 02/22/2017] [Indexed: 11/22/2022]
Affiliation(s)
- Turney J McKee
- Faculty of Dentistry, McGill University, Shriners Hospital for Children - Canada, Montreal, Quebec, Canada
| | - Svetlana V Komarova
- Faculty of Dentistry, McGill University, Shriners Hospital for Children - Canada, Montreal, Quebec, Canada
| |
Collapse
|
29
|
Feng J, Huang P, Wu FY. Gold–platinum bimetallic nanoclusters with enhanced peroxidase-like activity and their integrated agarose hydrogel-based sensing platform for the colorimetric analysis of glucose levels in serum. Analyst 2017; 142:4106-4115. [DOI: 10.1039/c7an01343d] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Bimetallic Au–PtNCs with enhanced peroxidase-like activity were integrated into agarose hydrogels for direct visualization of glucose in human serum.
Collapse
Affiliation(s)
- Jiayu Feng
- College of Chemistry
- Nanchang University
- Nanchang 330031
- China
| | | | - Fang-Ying Wu
- College of Chemistry
- Nanchang University
- Nanchang 330031
- China
| |
Collapse
|
30
|
Liao LX, Zhao MB, Dong X, Jiang Y, Zeng KW, Tu PF. TDB protects vascular endothelial cells against oxygen-glucose deprivation/reperfusion-induced injury by targeting miR-34a to increase Bcl-2 expression. Sci Rep 2016; 6:37959. [PMID: 27885275 PMCID: PMC5122842 DOI: 10.1038/srep37959] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 11/03/2016] [Indexed: 12/30/2022] Open
Abstract
Prolonged ischemia can result in apoptotic death of vascular endothelial cells and lead to ischemic vascular diseases including vascular dementia, arteriosclerosis and brain oedema. Finding protective strategies to prevent this is therefore an urgent mission. Recent studies have shown that dysregulation of microRNAs (miRNAs) can lead to imbalance of Bcl-2 family proteins and mitochondrial dysfunction, leading to further damage of vascular cells under ischemic conditions. However, whether miRNAs can be used as a drug target for treating vascular diseases is not fully understood. In this study, we observed that the natural product 2,4,5-trihydroxybenzaldehyde (TDB) could effectively inhibit vascular cell apoptosis following oxygen-glucose deprivation/reperfusion (OGD/R) by maintaining mitochondrial membrane potential (MMP) and suppressing activation of the mitochondria-dependent caspase-9/3 apoptosis pathway. Furthermore, we identified miR-34a, a crucial negative regulator of Bcl-2, as a target for the protective effect of TDB on vascular cells. TDB-induced suppression of miR-34a resulted in a significant upregulation of Bcl-2 protein, MMP maintenance, and the survival of vascular cells following OGD/R. Our findings suggest that targeting miR-34a with the natural product TDB may provide a novel strategy for the treatment of ischemic vascular injuries, and demonstrate the therapeutic potential in targeting miRNAs using appropriate small molecules.
Collapse
Affiliation(s)
- Li-Xi Liao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Ming-Bo Zhao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xin Dong
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Yong Jiang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Ke-Wu Zeng
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Peng-Fei Tu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| |
Collapse
|