1
|
Hossain MR, Tareq MMI, Biswas P, Tauhida SJ, Bibi S, Zilani MNH, Albadrani GM, Al‐Ghadi MQ, Abdel‐Daim MM, Hasan MN. Identification of molecular targets and small drug candidates for Huntington's disease via bioinformatics and a network-based screening approach. J Cell Mol Med 2024; 28:e18588. [PMID: 39153206 PMCID: PMC11330274 DOI: 10.1111/jcmm.18588] [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: 05/18/2024] [Revised: 07/07/2024] [Accepted: 07/23/2024] [Indexed: 08/19/2024] Open
Abstract
Huntington's disease (HD) is a gradually severe neurodegenerative ailment characterised by an increase of a specific trinucleotide repeat sequence (cytosine-adenine-guanine, CAG). It is passed down as a dominant characteristic that worsens over time, creating a significant risk. Despite being monogenetic, the underlying mechanisms as well as biomarkers remain poorly understood. Furthermore, early detection of HD is challenging, and the available diagnostic procedures have low precision and accuracy. The research was conducted to provide knowledge of the biomarkers, pathways and therapeutic targets involved in the molecular processes of HD using informatic based analysis and applying network-based systems biology approaches. The gene expression profile datasets GSE97100 and GSE74201 relevant to HD were studied. As a consequence, 46 differentially expressed genes (DEGs) were identified. 10 hub genes (TPM1, EIF2S3, CCN2, ACTN1, ACTG2, CCN1, CSRP1, EIF1AX, BEX2 and TCEAL5) were further differentiated in the protein-protein interaction (PPI) network. These hub genes were typically down-regulated. Additionally, DEGs-transcription factors (TFs) connections (e.g. GATA2, YY1 and FOXC1), DEG-microRNA (miRNA) interactions (e.g. hsa-miR-124-3p and has-miR-26b-5p) were also comprehensively forecast. Additionally, related gene ontology concepts (e.g. sequence-specific DNA binding and TF activity) connected to DEGs in HD were identified using gene set enrichment analysis (GSEA). Finally, in silico drug design was employed to find candidate drugs for the treatment HD, and while the possible modest therapeutic compounds (e.g. cortistatin A, 13,16-Epoxy-25-hydroxy-17-cheilanthen-19,25-olide, Hecogenin) against HD were expected. Consequently, the results from this study may give researchers useful resources for the experimental validation of Huntington's diagnosis and therapeutic approaches.
Collapse
Affiliation(s)
- Md Ridoy Hossain
- Laboratory of Pharmaceutical Biotechnology and Bioinformatics, Department of Genetic Engineering and BiotechnologyJashore University of Science and TechnologyJessoreBangladesh
| | - Md. Mohaimenul Islam Tareq
- Laboratory of Pharmaceutical Biotechnology and Bioinformatics, Department of Genetic Engineering and BiotechnologyJashore University of Science and TechnologyJessoreBangladesh
| | - Partha Biswas
- Laboratory of Pharmaceutical Biotechnology and Bioinformatics, Department of Genetic Engineering and BiotechnologyJashore University of Science and TechnologyJessoreBangladesh
| | - Sadia Jannat Tauhida
- Laboratory of Pharmaceutical Biotechnology and Bioinformatics, Department of Genetic Engineering and BiotechnologyJashore University of Science and TechnologyJessoreBangladesh
| | - Shabana Bibi
- Department of BiosciencesShifa Tameer‐e‐Millat UniversityIslamabadPakistan
- Department of Health SciencesNovel Global Community Educational FoundationHebershamNew South WalesAustralia
| | | | - Ghadeer M. Albadrani
- Department of Biology, College of SciencePrincess Nourah bint Abdulrahman UniversityRiyadhSaudi Arabia
| | - Muath Q. Al‐Ghadi
- Department of Zoology, College of ScienceKing Saud UniversityRiyadhSaudi Arabia
| | - Mohamed M. Abdel‐Daim
- Department of Pharmaceutical Sciences, Pharmacy ProgramBatterjee Medical CollegeJeddahSaudi Arabia
- Pharmacology Department, Faculty of Veterinary MedicineSuez Canal UniversityIsmailiaEgypt
| | - Md. Nazmul Hasan
- Laboratory of Pharmaceutical Biotechnology and Bioinformatics, Department of Genetic Engineering and BiotechnologyJashore University of Science and TechnologyJessoreBangladesh
| |
Collapse
|
2
|
Kucher AN, Koroleva IA, Nazarenko MS. Pathogenetic Significance of Long Non-Coding RNAs in the Development of Thoracic and Abdominal Aortic Aneurysms. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:130-147. [PMID: 38467550 DOI: 10.1134/s0006297924010085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 11/30/2023] [Accepted: 12/01/2023] [Indexed: 03/13/2024]
Abstract
Aortic aneurysm (AA) is a life-threatening condition with a high prevalence and risk of severe complications. The aim of this review was to summarize the data on the role of long non-coding RNAs (lncRNAs) in the development of AAs of various location. Within less than a decade of studies on the role of lncRNAs in AA, using experimental and bioinformatic approaches, scientists have obtained the data confirming the involvement of these molecules in metabolic pathways and pathogenetic mechanisms critical for the aneurysm development. Regardless of the location of pathological process (thoracic or abdominal aorta), AA was found to be associated with changes in the expression of various lncRNAs in the tissue of the affected vessels. The consistency of changes in the expression level of lncRNA, mRNA and microRNA in aortic tissues during AA development has been recordedand regulatory networks implicated in the AA pathogenesis in which lncRNAs act as competing endogenous RNAs (ceRNA networks) have been identified. It was found that the same lncRNA can be involved in different ceRNA networks and regulate different biochemical and cellular events; on the other hand, the same pathological process can be controlled by different lncRNAs. Despite some similarities in pathogenesis and overlapping of involved lncRNAs, the ceRNA networks described for abdominal and thoracic AA are different. Interactions between lncRNAs and other molecules, including those participating in epigenetic processes, have also been identified as potentially relevant to the AA pathogenesis. The expression levels of some lncRNAs were found to correlate with clinically significant aortic features and biochemical parameters. Identification of regulatory RNAs functionally significant in the aneurysm development is important for clarification of disease pathogenesis and will provide a basis for early diagnostics and development of new preventive and therapeutic drugs.
Collapse
Affiliation(s)
- Aksana N Kucher
- Research Institute of Medical Genetics, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, 634050, Russia
| | - Iuliia A Koroleva
- Research Institute of Medical Genetics, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, 634050, Russia
| | - Maria S Nazarenko
- Research Institute of Medical Genetics, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, 634050, Russia.
| |
Collapse
|
3
|
Wang H, Wang L, Tan Y, Fang C, Li C, Zhang L. Identification of miRNAs Involved in Intracranial Aneurysm Rupture in Cigarette-Smoking Patients. Neurol Ther 2023; 12:2101-2119. [PMID: 37792217 PMCID: PMC10630182 DOI: 10.1007/s40120-023-00547-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 09/13/2023] [Indexed: 10/05/2023] Open
Abstract
INTRODUCTION Smoking is an independent risk factor for the formation and rupture of intracranial aneurysms (IA). However, the underlying mechanism remains unclear. METHODS In this study, we performed miRNA sequencing on plasma from 10 smoking patients with IA, 10 non-smoking patients with IA, and 10 healthy controls. The differentially expressed miRNAs (DE miRNAs) between smoking and non-smoking patients with IA were identified. Functional and pathway enrichment analysis is employed to investigate the potential functions of those DE miRNA target genes. The correlations with the clinical parameters were assessed using receiver operating characteristic curve (ROC) analysis. RESULTS In total, we identified 428 DE miRNAs. Functional enrichment analysis showed the target genes were significantly enriched in biological aspects related to cell characteristics, such as cell cycle, cell differentiation, and cell migration. Pathway analysis showed DE miRNAs mainly enriched in the PI3K-Akt signaling pathway, Focal adhesion, and JAK-STAT signaling pathway. The expressions of miR-574-5p, miR-151a-3p, and miR-652-3p correlated well with aneurysm parameters. The AUC of miR-574-5p, miR-151a-3p, and miR-652-3p were 97%, 92%, and 99%, respectively. CONCLUSION Our study indicated that smoking significantly altered the plasma miRNA profile in patients with IA. The expression of miR-574-5p, miR-151a-3p, and miR-652-3p correlated with aneurysm parameters, which may play a significant role in the formation and rupture of IA.
Collapse
Affiliation(s)
- Hanbin Wang
- Department of Neurosurgery, Affiliated Hospital of Hebei University, Hebei University, Baoding, 071000, Hebei, China
| | - Luxuan Wang
- Department of Neurological Function Examination, Affiliated Hospital of Hebei University, Hebei University, Baoding, 071000, Hebei, China
| | - Yanli Tan
- Department of Pathology, Affiliated Hospital of Hebei University, Hebei University, Baoding, 071000, Hebei, China
| | - Chuan Fang
- Department of Neurosurgery, Affiliated Hospital of Hebei University, Hebei University, Baoding, 071000, Hebei, China.
- Postdoctoral Research Station of Neurosurgery, Affiliated Hospital of Hebei University, Hebei University, Baoding, 071000, Hebei, China.
- Hebei Key Laboratory of Precise Diagnosis and Treatment of Glioma, Baoding, China.
| | - Chunhui Li
- Department of Neurosurgery, Affiliated Hospital of Hebei University, Hebei University, Baoding, 071000, Hebei, China.
| | - Lijian Zhang
- Department of Neurosurgery, Affiliated Hospital of Hebei University, Hebei University, Baoding, 071000, Hebei, China.
- Postdoctoral Research Station of Neurosurgery, Affiliated Hospital of Hebei University, Hebei University, Baoding, 071000, Hebei, China.
| |
Collapse
|
4
|
Dong CX, Malecki C, Robertson E, Hambly B, Jeremy R. Molecular Mechanisms in Genetic Aortopathy-Signaling Pathways and Potential Interventions. Int J Mol Sci 2023; 24:ijms24021795. [PMID: 36675309 PMCID: PMC9865322 DOI: 10.3390/ijms24021795] [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: 11/30/2022] [Revised: 01/02/2023] [Accepted: 01/11/2023] [Indexed: 01/18/2023] Open
Abstract
Thoracic aortic disease affects people of all ages and the majority of those aged <60 years have an underlying genetic cause. There is presently no effective medical therapy for thoracic aneurysm and surgery remains the principal intervention. Unlike abdominal aortic aneurysm, for which the inflammatory/atherosclerotic pathogenesis is well established, the mechanism of thoracic aneurysm is less understood. This paper examines the key cell signaling systems responsible for the growth and development of the aorta, homeostasis of endothelial and vascular smooth muscle cells and interactions between pathways. The evidence supporting a role for individual signaling pathways in pathogenesis of thoracic aortic aneurysm is examined and potential novel therapeutic approaches are reviewed. Several key signaling pathways, notably TGF-β, WNT, NOTCH, PI3K/AKT and ANGII contribute to growth, proliferation, cell phenotype and survival for both vascular smooth muscle and endothelial cells. There is crosstalk between pathways, and between vascular smooth muscle and endothelial cells, with both synergistic and antagonistic interactions. A common feature of the activation of each is response to injury or abnormal cell stress. Considerable experimental evidence supports a contribution of each of these pathways to aneurysm formation. Although human information is less, there is sufficient data to implicate each pathway in the pathogenesis of human thoracic aneurysm. As some pathways i.e., WNT and NOTCH, play key roles in tissue growth and organogenesis in early life, it is possible that dysregulation of these pathways results in an abnormal aortic architecture even in infancy, thereby setting the stage for aneurysm development in later life. Given the fine tuning of these signaling systems, functional polymorphisms in key signaling elements may set up a future risk of thoracic aneurysm. Multiple novel therapeutic agents have been developed, targeting cell signaling pathways, predominantly in cancer medicine. Future investigations addressing cell specific targeting, reduced toxicity and also less intense treatment effects may hold promise for effective new medical treatments of thoracic aortic aneurysm.
Collapse
Affiliation(s)
- Charlotte Xue Dong
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
| | - Cassandra Malecki
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
- The Baird Institute, Camperdown, NSW 2042, Australia
| | - Elizabeth Robertson
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
| | - Brett Hambly
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
| | - Richmond Jeremy
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
- The Baird Institute, Camperdown, NSW 2042, Australia
- Correspondence:
| |
Collapse
|
5
|
Zhou H, Wang L, Liu S, Wang W. The role of phosphoinositide 3-kinases in immune-inflammatory responses: potential therapeutic targets for abdominal aortic aneurysm. Cell Cycle 2022; 21:2339-2364. [PMID: 35792922 DOI: 10.1080/15384101.2022.2094577] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The pathogenesis of abdominal aortic aneurysm (AAA) includes inflammatory responses, matrix metalloproteinases (MMPs) degradation, VSMC apoptosis, oxidative stress, and angiogenesis, among which the inflammatory response plays a key role. At present, surgery is the only curing treatment, and no effective drug can delay AAA progression in clinical practice. Therefore, searching for a signaling pathway related to the immune-inflammatory response is an essential direction for developing drugs targeting AAA. Recent studies have confirmed that the PI3K family plays an important role in many inflammatory diseases and is involved in regulating various cellular functions, especially in the immune-inflammatory response. This review focuses on the role of each isoform of PI3K in each stage of AAA immune-inflammatory response, making available explorations for a deeper understanding of the mechanism of inflammation and immune response during the formation and development of AAA.
Collapse
Affiliation(s)
- Haiyang Zhou
- Department of General &vascular Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Lei Wang
- Department of General &vascular Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Shuai Liu
- Department of General &vascular Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Wei Wang
- Department of General &vascular Surgery, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
6
|
Lim MS, Portelli SS, Padang R, Bannon PG, Hambly BD, Jeremy RW, Celermajer DS, Robertson EN. Novel insights into bicuspid aortic valve (BAV) aortopathy: Long non-coding RNAs TUG1 and MIAT are differentially expressed in BAV ascending aortas. Cardiovasc Pathol 2022; 60:107433. [PMID: 35588998 DOI: 10.1016/j.carpath.2022.107433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 05/10/2022] [Accepted: 05/10/2022] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND Whilst a combination of genetically mediated vulnerability and haemodynamic insult is suspected to contribute to bicuspid aortic valve (BAV) aortopathy, the underlying pathophysiological mechanisms are poorly understood. METHODS Utilising RT-qPCR, we compared the expression of 28 potentially relevant long non-coding RNA (lncRNA) in aortic tissue from BAV patients undergoing aortic surgery for aortopathy, to healthy controls. Relative lncRNA expression was measured using ΔΔCT, with fold-change calculated as RQ=2-ΔΔCT. RESULTS When comparing samples from BAV patients (n=29, males n=25; median age 58 years, Q1-Q3 51-65, maximum aortic dimension 50±5mm) with healthy controls (n=7; males n=4, p=0.12; median age 39 years, Q1-Q3 18-47, p=0.001), there were two differentially expressed lncRNA: TUG1 expression was significantly lower in BAV aortic tissue (RQ 0.59, 95% CI 0.50-0.69, p=0.02), whilst MIAT expression was significantly higher (RQ 2.87, 95% CI 1.96-4.20, p=0.01). Sensitivity analysis including only patients with normal BAV function showed similar trends of differential expression of TUG1 (RQ 0.69, 95% CI 0.50-0.90, p=0.29) and MIAT (RQ 2.55, 95% CI 1.21-5.36, p=0.29) compared to controls. CONCLUSIONS LncRNA TUG1 and MIAT are differentially expressed in BAV aortopathy compared to healthy controls, independent of BAV haemodynamics. Aberrant lncRNA expression may be involved in the pathogenesis of BAV aortopathy.
Collapse
Affiliation(s)
- M S Lim
- The University of Sydney, Sydney Medical School, Faculty of Medicine and Health, Camperdown, New South Wales, Australia; Royal Prince Alfred Hospital, Department of Cardiology, Camperdown, New South Wales, Australia.
| | - S S Portelli
- The University of Sydney, Sydney Medical School, Faculty of Medicine and Health, Camperdown, New South Wales, Australia
| | - R Padang
- The University of Sydney, Sydney Medical School, Faculty of Medicine and Health, Camperdown, New South Wales, Australia; Royal Prince Alfred Hospital, Department of Cardiology, Camperdown, New South Wales, Australia
| | - P G Bannon
- The University of Sydney, Sydney Medical School, Faculty of Medicine and Health, Camperdown, New South Wales, Australia; Royal Prince Alfred Hospital, Department of Cardiothoracic Surgery, Camperdown, New South Wales, Australia
| | - B D Hambly
- The University of Sydney, Sydney Medical School, Faculty of Medicine and Health, Camperdown, New South Wales, Australia
| | - R W Jeremy
- The University of Sydney, Sydney Medical School, Faculty of Medicine and Health, Camperdown, New South Wales, Australia; Royal Prince Alfred Hospital, Department of Cardiology, Camperdown, New South Wales, Australia
| | - D S Celermajer
- The University of Sydney, Sydney Medical School, Faculty of Medicine and Health, Camperdown, New South Wales, Australia; Royal Prince Alfred Hospital, Department of Cardiology, Camperdown, New South Wales, Australia; Heart Research Institute, Sydney, Australia
| | - E N Robertson
- The University of Sydney, Sydney Medical School, Faculty of Medicine and Health, Camperdown, New South Wales, Australia; Royal Prince Alfred Hospital, Department of Cardiology, Camperdown, New South Wales, Australia
| |
Collapse
|
7
|
Rombouts KB, van Merrienboer TAR, Ket JCF, Bogunovic N, van der Velden J, Yeung KK. The role of vascular smooth muscle cells in the development of aortic aneurysms and dissections. Eur J Clin Invest 2022; 52:e13697. [PMID: 34698377 PMCID: PMC9285394 DOI: 10.1111/eci.13697] [Citation(s) in RCA: 89] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 09/12/2021] [Accepted: 10/11/2021] [Indexed: 12/30/2022]
Abstract
BACKGROUND Aortic aneurysms (AA) are pathological dilations of the aorta, associated with an overall mortality rate up to 90% in case of rupture. In addition to dilation, the aortic layers can separate by a tear within the layers, defined as aortic dissections (AD). Vascular smooth muscle cells (vSMC) are the predominant cell type within the aortic wall and dysregulation of vSMC functions contributes to AA and AD development and progression. However, since the exact underlying mechanism is poorly understood, finding potential therapeutic targets for AA and AD is challenging and surgery remains the only treatment option. METHODS In this review, we summarize current knowledge about vSMC functions within the aortic wall and give an overview of how vSMC functions are altered in AA and AD pathogenesis, organized per anatomical location (abdominal or thoracic aorta). RESULTS Important functions of vSMC in healthy or diseased conditions are apoptosis, phenotypic switch, extracellular matrix regeneration and degradation, proliferation and contractility. Stressors within the aortic wall, including inflammatory cell infiltration and (epi)genetic changes, modulate vSMC functions and cause disturbance of processes within vSMC, such as changes in TGF-β signalling and regulatory RNA expression. CONCLUSION This review underscores a central role of vSMC dysfunction in abdominal and thoracic AA and AD development and progression. Further research focused on vSMC dysfunction in the aortic wall is necessary to find potential targets for noninvasive AA and AD treatment options.
Collapse
Affiliation(s)
- Karlijn B Rombouts
- Department of Surgery, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center and AMC, Amsterdam, The Netherlands.,Department of Physiology, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center, Amsterdam, The Netherlands
| | - Tara A R van Merrienboer
- Department of Surgery, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center and AMC, Amsterdam, The Netherlands.,Department of Physiology, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center, Amsterdam, The Netherlands
| | | | - Natalija Bogunovic
- Department of Surgery, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center and AMC, Amsterdam, The Netherlands.,Department of Physiology, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center, Amsterdam, The Netherlands.,Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jolanda van der Velden
- Department of Physiology, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center, Amsterdam, The Netherlands
| | - Kak Khee Yeung
- Department of Surgery, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center and AMC, Amsterdam, The Netherlands.,Department of Physiology, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
8
|
Elwazir MY, Hussein MH, Toraih EA, Al Ageeli E, Esmaeel SE, Fawzy MS, Faisal S. Association of Angio-LncRNAs MIAT rs1061540/MALAT1 rs3200401 Molecular Variants with Gensini Score in Coronary Artery Disease Patients Undergoing Angiography. Biomolecules 2022; 12:biom12010137. [PMID: 35053285 PMCID: PMC8773982 DOI: 10.3390/biom12010137] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 01/09/2022] [Accepted: 01/13/2022] [Indexed: 02/05/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) have emerged as essential biomolecules with variable diagnostic and/or prognostic utility in several diseases, including coronary artery disease (CAD). We aimed for the first time to investigate the potential association of five angiogenesis-related lncRNAs (PUNISHER, SENCR, MIAT, MALAT1, and GATA6-AS) variants with CAD susceptibility and/or severity. TaqMan Real-Time genotyping for PUNISHER rs12318065A/C, SENCR rs12420823C/T, MIAT rs1061540C/T, MALAT1 rs3200401T/C, and GATA6-AS1 rs73390820A/G were run on the extracted genomic DNA from 100 unrelated patients with stable CAD undergoing diagnostic coronary angiography and from 100 controls. After adjusting covariates, the studied variants showed no association with disease susceptibility; however, MIAT*T/T genotype was associated with a more severe Gensini score. In contrast, MALAT1*T/C heterozygosity was associated with a lower score. The lipid profile, and to a lesser extent smoking status, male sex, weight, hypertension, and MALAT1 (T > C) (negative correlation), explained the variance between patients/control groups via a principal component analysis. Incorporating the principal components into a logistic regression model to predict CAD yielded a 0.92 AUC. In conclusion: MIAT rs1061540 and MALAT1 rs3200401 variants were associated with CAD severity and Gensini score in the present sample of the Egyptian population. Further large multi-center and functional analyses are needed to confirm the results and identify the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Mohamed Y. Elwazir
- Department of Cardiology, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt;
| | - Mohammad H. Hussein
- Division of Endocrine and Oncologic Surgery, Department of Surgery, School of Medicine, Tulane University, New Orleans, LA 70112, USA;
| | - Eman A. Toraih
- Division of Endocrine and Oncologic Surgery, Department of Surgery, School of Medicine, Tulane University, New Orleans, LA 70112, USA;
- Genetics Unit, Department of Histology and Cell Biology, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt
- Correspondence: (E.A.T.); (M.S.F.); Tel.: +1-346-907-4237 (E.A.T.); +20-1008584720 (M.S.F.)
| | - Essam Al Ageeli
- Department of Clinical Biochemistry (Medical Genetics), Faculty of Medicine, Jazan University, Jazan 45142, Saudi Arabia;
| | - Safya E. Esmaeel
- Department of Physiology, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt;
| | - Manal S. Fawzy
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt;
- Department of Biochemistry, Faculty of Medicine, Northern Border University, Arar 1321, Saudi Arabia
- Correspondence: (E.A.T.); (M.S.F.); Tel.: +1-346-907-4237 (E.A.T.); +20-1008584720 (M.S.F.)
| | - Salwa Faisal
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt;
| |
Collapse
|
9
|
Ye T, Feng J, Cui M, Yang J, Wan X, Xie D, Liu J. LncRNA MIAT Services as a Noninvasive Biomarker for Diagnosis and Correlated with Immune Infiltrates in Breast Cancer. Int J Womens Health 2021; 13:991-1004. [PMID: 34712062 PMCID: PMC8548061 DOI: 10.2147/ijwh.s312714] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 10/08/2021] [Indexed: 12/13/2022] Open
Abstract
Background Myocardial infarction associated transcript (MIAT) is identified as a long chain non-coding RNA (lncRNA), which was associated with myocardial infarction susceptibility. While intense efforts have been made to elucidate the relationship between MIAT and carcinogenesis, the tumor immunoreaction of MIAT remains elusive. Thus, this study aimed to investigate the role of MIAT in the immunoregulation of breast cancer (BC) and further explore the better clinical significance. Methods The differential expression of MIAT between BC and normal/adjacent tissues was compared using Wilcoxon rank sum test. The diagnostic and prognostic values of elevated MIAT expression in BC tissues were unveiled via receiver operating characteristic (ROC) analysis and KM-plotter analysis. Limma and edgeR package were used to identify differentially expressed genes (DEGs) and microRNAs (DEMs) from TCGA database respectively. A co-expression dataset was constructed to comprehensively understand the relationship between MIAT and DEGs based on the Pearson correlation coefficient. Furthermore, GO and KEGG analyses were conducted to predict the potential functions of MIAT. We next intersected immune-related genes (IRGs) from ImmPort database with MIAT-co-expressed genes to obtain MIAT-co-expressed IRGs, in order to construct MIAT-microRNA (miRNA)-mRNA network. And the correlation between MIAT and tumor-infiltrating immune cells (TICs) and immunophenoscore (IPS) analysis was analyzed by TIMER and CIBERSORT. Finally, the reverse transcription quantitative real-time polymerase chain reaction (RT-qPCR) was used to detect the expression profiles of MIAT in serum samples. Results The expression levels of MIAT were notably higher in BC than in normal or adjacent tissues. And MIAT expression could be used as a prognostic indicator of mortality risk in patients with BC in different aspects. Moreover, the enrichment analyses suggested that MIAT was strongly involved in BC immune response. In addition, TIMER database and CIBERSORT analyses indicated that MIAT was significantly correlated with 13 types of TICs (B cells, dendritic cells, neutrophils, CD8 T cells, CD4 memory resting T cells, CD4 memory activated T cells, gamma delta T cells, M1 macrophages, plasma cells, activated NK cells, monocytes, M2 macrophages, activated mast cells). Simultaneously, the IPS analysis implied that the higher the MIAT expression, the better the immunotherapy effect. The ROC curve analysis showed that the area under the curve (AUC) value of MIAT was 0.86 (sensitivity = 87.80%, specificity = 75.61%). And the high MIAT expression in serum was positive related to TNM stage (P = 0.032) and lymph node metastasis (P = 0.028). Conclusion MIAT may be a valuable noninvasive diagnostic biomarker for BC and is associated with tumor-infiltrating immune cells in tumor microenvironment, suggesting MIAT as a potential target for future treatment of BC.
Collapse
Affiliation(s)
- Ting Ye
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, People's Republic of China
| | - Jia Feng
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, People's Republic of China
| | - Meng Cui
- Department of Laboratory Medicine, The Leshan People's Hospital, Luzhou, Sichuan, 614000, People's Republic of China
| | - Jia Yang
- Department of Laboratory Medicine, The Leshan People's Hospital, Luzhou, Sichuan, 614000, People's Republic of China
| | - Xue Wan
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, People's Republic of China
| | - Dan Xie
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, People's Republic of China
| | - Jinbo Liu
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, People's Republic of China
| |
Collapse
|
10
|
Programmed cell death in aortic aneurysm and dissection: A potential therapeutic target. J Mol Cell Cardiol 2021; 163:67-80. [PMID: 34597613 DOI: 10.1016/j.yjmcc.2021.09.010] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 09/07/2021] [Accepted: 09/23/2021] [Indexed: 12/20/2022]
Abstract
Rupture of aortic aneurysm and dissection (AAD) remains a leading cause of death. Progressive smooth muscle cell (SMC) loss is a crucial feature of AAD that contributes to aortic dysfunction and degeneration, leading to aortic aneurysm, dissection, and, ultimately, rupture. Understanding the molecular mechanisms of SMC loss and identifying pathways that promote SMC death in AAD are critical for developing an effective pharmacologic therapy to prevent aortic destruction and disease progression. Cell death is controlled by programmed cell death pathways, including apoptosis, necroptosis, pyroptosis, and ferroptosis. Although these pathways share common stimuli and triggers, each type of programmed cell death has unique features and activation pathways. A growing body of evidence supports a critical role for programmed cell death in the pathogenesis of AAD, and inhibitors of various types of programmed cell death represent a promising therapeutic strategy. This review discusses the different types of programmed cell death pathways and their features, induction, contributions to AAD development, and therapeutic potential. We also highlight the clinical significance of programmed cell death for further studies.
Collapse
|
11
|
Tello-Flores VA, Beltrán-Anaya FO, Ramírez-Vargas MA, Esteban-Casales BE, Navarro-Tito N, Alarcón-Romero LDC, Luciano-Villa CA, Ramírez M, del Moral-Hernández Ó, Flores-Alfaro E. Role of Long Non-Coding RNAs and the Molecular Mechanisms Involved in Insulin Resistance. Int J Mol Sci 2021; 22:7256. [PMID: 34298896 PMCID: PMC8306787 DOI: 10.3390/ijms22147256] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 06/27/2021] [Accepted: 07/02/2021] [Indexed: 12/14/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) are single-stranded RNA biomolecules with a length of >200 nt, and they are currently considered to be master regulators of many pathological processes. Recent publications have shown that lncRNAs play important roles in the pathogenesis and progression of insulin resistance (IR) and glucose homeostasis by regulating inflammatory and lipogenic processes. lncRNAs regulate gene expression by binding to other non-coding RNAs, mRNAs, proteins, and DNA. In recent years, several mechanisms have been reported to explain the key roles of lncRNAs in the development of IR, including metastasis-associated lung adenocarcinoma transcript 1 (MALAT1), imprinted maternal-ly expressed transcript (H19), maternally expressed gene 3 (MEG3), myocardial infarction-associated transcript (MIAT), and steroid receptor RNA activator (SRA), HOX transcript antisense RNA (HOTAIR), and downregulated Expression-Related Hexose/Glucose Transport Enhancer (DREH). LncRNAs participate in the regulation of lipid and carbohydrate metabolism, the inflammatory process, and oxidative stress through different pathways, such as cyclic adenosine monophosphate/protein kinase A (cAMP/PKA), phosphatidylinositol 3-kinase/protein kinase B (PI3K/AKT), polypyrimidine tract-binding protein 1/element-binding transcription factor 1c (PTBP1/SREBP-1c), AKT/nitric oxide synthase (eNOS), AKT/forkhead box O1 (FoxO1), and tumor necrosis factor-alpha (TNF-α)/c-Jun-N-terminal kinases (JNK). On the other hand, the mechanisms linked to the molecular, cellular, and biochemical actions of lncRNAs vary according to the tissue, biological species, and the severity of IR. Therefore, it is essential to elucidate the role of lncRNAs in the insulin signaling pathway and glucose and lipid metabolism. This review analyzes the function and molecular mechanisms of lncRNAs involved in the development of IR.
Collapse
Affiliation(s)
- Vianet Argelia Tello-Flores
- Laboratorio de Epidemiología Clínica y Molecular, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39087, GRO, Mexico; (V.A.T.-F.); (F.O.B.-A.); (M.A.R.-V.); (B.E.E.-C.); (C.A.L.-V.)
| | - Fredy Omar Beltrán-Anaya
- Laboratorio de Epidemiología Clínica y Molecular, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39087, GRO, Mexico; (V.A.T.-F.); (F.O.B.-A.); (M.A.R.-V.); (B.E.E.-C.); (C.A.L.-V.)
| | - Marco Antonio Ramírez-Vargas
- Laboratorio de Epidemiología Clínica y Molecular, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39087, GRO, Mexico; (V.A.T.-F.); (F.O.B.-A.); (M.A.R.-V.); (B.E.E.-C.); (C.A.L.-V.)
| | - Brenda Ely Esteban-Casales
- Laboratorio de Epidemiología Clínica y Molecular, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39087, GRO, Mexico; (V.A.T.-F.); (F.O.B.-A.); (M.A.R.-V.); (B.E.E.-C.); (C.A.L.-V.)
| | - Napoleón Navarro-Tito
- Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39087, GRO, Mexico;
| | - Luz del Carmen Alarcón-Romero
- Laboratorio de Citopatología e Histoquímica, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39087, GRO, Mexico;
| | - Carlos Aldair Luciano-Villa
- Laboratorio de Epidemiología Clínica y Molecular, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39087, GRO, Mexico; (V.A.T.-F.); (F.O.B.-A.); (M.A.R.-V.); (B.E.E.-C.); (C.A.L.-V.)
| | - Mónica Ramírez
- CONACyT, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39087, GRO, Mexico;
| | - Óscar del Moral-Hernández
- Laboratorio de Virología, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39087, GRO, Mexico
| | - Eugenia Flores-Alfaro
- Laboratorio de Epidemiología Clínica y Molecular, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39087, GRO, Mexico; (V.A.T.-F.); (F.O.B.-A.); (M.A.R.-V.); (B.E.E.-C.); (C.A.L.-V.)
| |
Collapse
|
12
|
LncRNA Sox2ot modulates the progression of thoracic aortic aneurysm by regulating miR-330-5p/Myh11. Biosci Rep 2021; 40:225469. [PMID: 32578852 PMCID: PMC7364482 DOI: 10.1042/bsr20194040] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 04/21/2020] [Accepted: 06/10/2020] [Indexed: 12/16/2022] Open
Abstract
Thoracic aortic aneurysm (TAA) has been causing the death of elder people. Myosin heavy chain 11 (Myh11) has been reported associated with aortic aneurysm, but there is no specific study on its function on TAA. Here we aimed to explore the function of Myh11 on mouse aortic smooth muscle cells (SMCs) for studying the inner mechanism of TAA. H2O2 treatment was implemented on mouse aortic SMCs for detecting cell apoptosis. Meanwhile, functional assays were conducted to verify the function of Myh11 on mouse aortic SMCs. Also, pull-down assay, RIP assay were implemented to identify the potential RNAs for study. Quantitative real-time polymerase chain reaction (qRT-PCR) and luciferase reporter assay were implemented to identify the expression and binding relationships of RNAs. Myh11 expression was increased by treatment of H2O2. Myh11 could decrease proliferation and enhance apoptosis of mouse aortic SMCs. At the same time, mmu-miR-330-5p could bind to Myh11 and Sox2ot, forming a competing endogenous RNA (ceRNA) pathway to regulate the proliferation and apoptosis of mouse aortic SMCs. Moreover, both Sox2ot and Myh11 were proved to be up-regulated whereas miR-330-5p down-regulated in Fbn1C1039G/+ mice, the in vivo model of TAA. In a word, long noncoding RNA (lncRNA) Sox2ot modulates the progression of TAA by regulating miR-330-5p/Myh11 axis.
Collapse
|
13
|
Pan YB, Lu J, Yang B, Lenahan C, Zhang J, Shao A. Construction of competitive endogenous RNA network reveals regulatory role of long non-coding RNAs in intracranial aneurysm. BMC Neurosci 2021; 22:15. [PMID: 33750300 PMCID: PMC7945298 DOI: 10.1186/s12868-021-00622-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 02/25/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Rupture of intracranial aneurysm (IA) is the main cause of devastating subarachnoid hemorrhage, which urges our understanding of the pathogenesis and regulatory mechanisms of IA. However, the regulatory roles of long non-coding RNAs (lncRNAs) in IA is less known. RESULTS We processed the raw SRR files of 12 superficial temporal artery (STA) samples and 6 IA samples to count files. Then the differentially expressed (DE) mRNAs, miRNAs, and lncRNAs between STAs and IAs were identified. The enrichment analyses were performed using DEmRNAs. Next, a lncRNA-miRNA-mRNA regulatory network was constructed using integrated bioinformatics analysis. In summary, 341 DElncRNAs, 234 DEmiRNAs, and 2914 DEmRNAs between the STA and IA. The lncRNA-miRNA-mRNA regulatory network of IA contains 91 nodes and 146 edges. The subnetwork of hub lncRNA PVT1 was extracted. The expression level of PVT1 was positively correlated with a majority of the mRNAs in its subnetwork. Moreover, we found that several mRNAs (CCND1, HIF1A, E2F1, CDKN1A, VEGFA, COL1A1 and COL5A2) in the PVT1 subnetwork served as essential components in the PI3K-Akt signaling pathway, and that some of the non-coding RNAs (ncRNAs) (PVT1, HOTAIR, hsa-miR-17, hsa-miR-142, hsa-miR-383 and hsa-miR-193b) interacted with these mRNAs. CONCLUSION Our annotations noting ncRNA's role in the pathway may uncover novel regulatory mechanisms of ncRNAs and mRNAs in IA. These findings provide significant insights into the lncRNA regulatory network in IA.
Collapse
Affiliation(s)
- Yuan-Bo Pan
- Department of Neurosurgery, School of Medicine, Second Affiliated Hospital, Zhejiang University, NO.88 Jiefang Rd, Hangzhou, 310009, Zhejiang, China
| | - Jianan Lu
- Department of Neurosurgery, School of Medicine, Second Affiliated Hospital, Zhejiang University, NO.88 Jiefang Rd, Hangzhou, 310009, Zhejiang, China
| | - Biao Yang
- Department of Neurosurgery, Huashan Hospital of Fudan University, Shanghai, China
| | - Cameron Lenahan
- Burrell College of Osteopathic Medicine, Las Cruces, NM, 88003, USA
- Center for Neuroscience Research, School of Medicine, Loma Linda University, Loma Linda, CA, 92324, USA
| | - Jianmin Zhang
- Department of Neurosurgery, School of Medicine, Second Affiliated Hospital, Zhejiang University, NO.88 Jiefang Rd, Hangzhou, 310009, Zhejiang, China.
- Brain Research Institute, Zhejiang University, Hangzhou, Zhejiang, China.
- Collaborative Innovation Center for Brain Science, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Anwen Shao
- Department of Neurosurgery, School of Medicine, Second Affiliated Hospital, Zhejiang University, NO.88 Jiefang Rd, Hangzhou, 310009, Zhejiang, China.
| |
Collapse
|
14
|
Di Y, Wang Y, Wang X, Nie QZ. Effects of long non-coding RNA myocardial infarction-associated transcript on retinal neovascularization in a newborn mouse model of oxygen-induced retinopathy. Neural Regen Res 2021; 16:1877-1881. [PMID: 33510096 PMCID: PMC8328761 DOI: 10.4103/1673-5374.306098] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Whether long non-coding RNA myocardial infarction-associated transcript is involved in oxygen-induced retinopathy remains poorly understood. To validate this hypothesis, we established a newborn mouse model of oxygen-induced retinopathy by feeding in an oxygen concentration of 75 ± 2% from postnatal day 8 to postnatal day 12, followed by in normal air. On postnatal day 11, the mice were injected with the myocardial infarction-associated transcript siRNA plasmid via the vitreous cavity to knockdown long non-coding RNA myocardial infarction-associated transcript. Myocardial infarction-associated transcript siRNA transcription significantly inhibited myocardial infarction-associated transcript mRNA expression, reduced the phosphatidylinosital-3-kinase, phosphorylated Akt and vascular endothelial growth factor immunopositivities, protein and mRNA expression, and alleviated the pathological damage to the retina of oxygen-induced retinopathy mouse models. These findings suggest that myocardial infarction-associated transcript is likely involved in the retinal neovascularization in retinopathy of prematurity and that inhibition of myocardial infarction-associated transcript can downregulate phosphatidylinosital-3-kinase, phosphorylated Akt and vascular endothelial growth factor expression levels and inhibit neovascularization. This study was approved by the Animal Ethics Committee of Shengjing Hospital of China Medical University, China (approval No. 2016PS074K) on February 25, 2016.
Collapse
Affiliation(s)
- Yu Di
- Department of Ophthalmology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Yue Wang
- Department of Ophthalmology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Xue Wang
- Department of Ophthalmology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Qing-Zhu Nie
- Department of Ophthalmology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
| |
Collapse
|
15
|
Gareev I, Beylerli O, Aliev G, Pavlov V, Izmailov A, Zhang Y, Liang Y, Yang G. The Role of Long Non-Coding RNAs in Intracranial Aneurysms and Subarachnoid Hemorrhage. Life (Basel) 2020; 10:life10090155. [PMID: 32825276 PMCID: PMC7555693 DOI: 10.3390/life10090155] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 08/06/2020] [Accepted: 08/15/2020] [Indexed: 12/14/2022] Open
Abstract
Intracranial aneurysms (IAs) represent the most complex and relevant problem of modern neurology and neurosurgery. They serve as one of the main causes of non-traumatic subarachnoid hemorrhage (SAH), causing up to 85% of all cases of intracranial hemorrhage, which is associated with frequent disability and high mortality among patients. Unfortunately, the molecular mechanisms of the development and rupture of IAs are still under study. Long non-coding RNAs (lncRNAs) are non-coding RNAs that typically have a length of more than 200 nucleotides. It is known that lncRNAs regulate many processes, such as transcription, translation, cell differentiation, regulation of gene expression, and regulation of the cell cycle. In recent years, a lot of evidence has established their role in human diseases from oncology to cardiovascular disease. Recent studies have shown that lncRNAs may be involved in the pathogenesis of IAs. The study of lncRNAs and its targets in various pathological conditions of a person is a rapidly developing field, and it is likely that the knowledge obtained from these studies regarding the pathogenesis of intracranial aneurysms will have the potential to use lncRNAs in therapy, as well as in the diagnosis and prediction of high aneurysms risk of rupture.
Collapse
Affiliation(s)
- Ilgiz Gareev
- Bashkir State Medical University, 450008 Ufa, Republic of Bashkortostan, Russia; (I.G.); (O.B.); (V.P.)
| | - Ozal Beylerli
- Bashkir State Medical University, 450008 Ufa, Republic of Bashkortostan, Russia; (I.G.); (O.B.); (V.P.)
| | - Gjumrakch Aliev
- Sechenov First Moscow State Medical University (Sechenov University), 119146 Moscow, Russia;
- Research Institute of Human Morphology, Russian Academy of Medical Science, 117418 Moscow, Russia
- Institute of Physiologically Active Compounds, Russian Academy of Sciences, Chernogolovka, 142432 Moscow, Russia
- GALLY International Research Institute, San Antonio, TX 78229, USA
| | - Valentin Pavlov
- Bashkir State Medical University, 450008 Ufa, Republic of Bashkortostan, Russia; (I.G.); (O.B.); (V.P.)
| | - Adel Izmailov
- Regional Clinical Oncology Center, 450054 Ufa, Republic of Bashkortostan, Russia;
| | - Yiwei Zhang
- Harbin Medical University, Harbin 150081, China; or
| | - Yanchao Liang
- Department of Neurosurgery, the First Affiliated Hospital of Harbin Medical University, Harbin 150001, China;
- Institute of Brain Science, Harbin Medical University, Harbin 150001, China
| | - Guang Yang
- Department of Neurosurgery, the First Affiliated Hospital of Harbin Medical University, Harbin 150001, China;
- Institute of Brain Science, Harbin Medical University, Harbin 150001, China
- Correspondence: or ; Tel.: +86-187-4607-2927
| |
Collapse
|
16
|
Zhao M, Xu L, Qian H. Bioinformatics analysis of microRNA profiles and identification of microRNA-mRNA network and biological markers in intracranial aneurysm. Medicine (Baltimore) 2020; 99:e21186. [PMID: 32756097 PMCID: PMC7402807 DOI: 10.1097/md.0000000000021186] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Intracranial aneurysm (IA) is a kind of cerebrovascular disorder, which may result in the subarachnoid hemorrhage with high lethality and disability. The purpose of this study was to reveal the pathogenesis and identify novel biomarkers in IA.We processed the raw microRNA (miRNA) expression profile data of IA obtained from Gene Expression Omnibus. Then weighted correlation network analysis was performed to identify the hub miRNAs in IA. Target genes of hub miRNAs were predicted using multiR package. In addition, a protein-protein network as well as miRNA-mRNA network was constructed and functional and pathway enrichment analyses were done. Finally, the prediction value of hub miRNAs in IA was tested in validation set.Two modules that had relation with IA were identified and 10 hub miRNAs in each module with higher gene-module association were selected. The protein-protein network and miRNA-mRNA network contained 243 nodes and 1496 edges. Functional and pathway enrichment analyses showed that they were mainly enriched in cell cycle, cell proliferation, and PI3K/Akt signaling pathways. Besides, hsa-miR-191-3p, hsa-miR-423-5p, hsa-miR-424-5p, hsa-miR-425-3p were proven to be valuable in prediction IA occurrence.In a word, this study reveals hub miRNAs, target genes and pathways potentially participating in formation and development of IA and screens out some candidate biomarkers. Our findings provide some new perspectives for research and treatment of IA.
Collapse
Affiliation(s)
- Ming Zhao
- Department of Neurosurgery, Zhuji People's Hospital of Zhejiang Province, Zhuji Affiliated Hospital of Shaoxing University, Zhuji
| | - Longbiao Xu
- Department of Neurosurgery, Zhuji People's Hospital of Zhejiang Province, Zhuji Affiliated Hospital of Shaoxing University, Zhuji
| | - Hui Qian
- Department of Neurosurgery, Shaoxing Central Hospital, Shaoxing, Zhejiang, China
| |
Collapse
|
17
|
Zhang X, Li H, Guo X, Hu J, Li B. Long Noncoding RNA Hypoxia-Inducible Factor-1 Alpha-Antisense RNA 1 Regulates Vascular Smooth Muscle Cells to Promote the Development of Thoracic Aortic Aneurysm by Modulating Apoptotic Protease-Activating Factor 1 and Targeting let-7g. J Surg Res 2020; 255:602-611. [PMID: 32653692 DOI: 10.1016/j.jss.2020.05.063] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 04/24/2020] [Accepted: 05/03/2020] [Indexed: 12/25/2022]
Abstract
BACKGROUND Thoracic aortic aneurysm (TAA) is a severe threat that is characterized by the increased aortic diameter. The dysfunction of vascular smooth muscle cells (VSMCs) contributes to the formation of TAA. Previous research indicated that long noncoding RNAs (lncRNAs) were involved in the development of TAA. This article aimed to explore the role of lncRNA hypoxia-inducible factor-1 alpha-antisense RNA 1 (HIF1A-AS1) and potential action mechanisms in VSMCs. METHODS The expression of HIF1A-AS1, collagen I, collagen III, microRNA let-7g (let-7g) and apoptotic protease-activating factor 1 (APAF1) was detected by quantitative real-time polymerase chain reaction. Cell proliferation and cell apoptosis were assessed by Cell Counting Kit-8 and flow cytometry assays, respectively. The protein levels of proliferating cell nuclear antigen, Cleaved caspase-3 (Cleaved-cas3), B cell lymphoma/leukemia-2 (Bcl-2), Collagen I, Collagen III, and APAF1 were quantified by Western blot. The relationship between let-7g and HIF1A-AS1 or APAF1 was predicted by the online bioinformatics tool and verified by dual-luciferase reporter assay and RNA pull-down assay. RESULTS HIF1A-AS1 was upregulated in TAA tissues and was a valuable diagnostic marker of TAA. HIF1A-AS1 overexpression suppressed proliferation, induced apoptosis, and reduced the expression of extracellular matrix proteins in VSMCs. let-7 g was a target of HIF1A-AS1, and its inhibition functioned the same role as HIF1A-AS1 overexpression. APAF1 was a target of let-7g, and its knockdown played the opposite role with HIF1A-AS1 overexpression. The reintroduction of let-7g or APAF1 knockdown reversed the effects of HIF1A-AS1 overexpression in VSMCs. CONCLUSIONS HIF1A-AS1 regulated the proliferation, apoptosis ,and the activity of extracellular matrix proteins in VSMCs through modulating APAF1 by targeting let-7g, leading to the development of TAA.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Cardiac Surgery, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, China
| | - Hongwei Li
- Department of Cardiac Surgery, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, China
| | - Xiaofeng Guo
- Department of Cardiac Surgery, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, China
| | - Jiting Hu
- Department of Neonatal Intensive Care Unit, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, China
| | - Bin Li
- Department of Cardiac Surgery, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, China.
| |
Collapse
|
18
|
Loganathan TS, Sulaiman SA, Abdul Murad NA, Shah SA, Abdul Gafor AH, Jamal R, Abdullah N. Interactions Among Non-Coding RNAs in Diabetic Nephropathy. Front Pharmacol 2020; 11:191. [PMID: 32194418 PMCID: PMC7062796 DOI: 10.3389/fphar.2020.00191] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 02/10/2020] [Indexed: 12/14/2022] Open
Abstract
Diabetic Nephropathy (DN) is the most common cause of End-stage renal disease (ESRD). Although various treatments and diagnosis applications are available, DN remains a clinical and economic burden. Recent findings showed that noncoding RNAs (ncRNAs) play an important role in DN progression, potentially can be used as biomarkers and therapeutic targets. NcRNAs refers to the RNA species that do not encode for any protein, and the most known ncRNAs are the microRNAs (miRNAs), long noncoding RNAs (lncRNAs), and circular RNAs (circRNAs). Dysregulation of these ncRNAs was reported before in DN patients and animal models of DN. Importantly, there are some interactions between these ncRNAs to regulate the crucial steps in DN progression. Here, we aimed to discuss the reported ncRNAs in DN and their interactions with critical genes in DN progression. Elucidating these ncRNAs regulatory network will allow for a better understanding of the molecular mechanisms in DN and how they can act as new biomarkers for DN and also as the potential targets for treatment.
Collapse
Affiliation(s)
- Tamil Selvi Loganathan
- UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Siti Aishah Sulaiman
- UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Nor Azian Abdul Murad
- UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Shamsul Azhar Shah
- Department of Community Health, UKM Medical Centre, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Abdul Halim Abdul Gafor
- Nephrology Unit, Faculty of Medicine, UKM Medical Centre, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Rahman Jamal
- UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Noraidatulakma Abdullah
- UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
19
|
Wang Z, Zhang B, Chen Z, He Y, Ru F, Liu P, Chen X. The long noncoding RNA myocardial infarction-associated transcript modulates the epithelial-mesenchymal transition in renal interstitial fibrosis. Life Sci 2020; 241:117187. [PMID: 31863776 DOI: 10.1016/j.lfs.2019.117187] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 12/13/2019] [Accepted: 12/15/2019] [Indexed: 01/12/2023]
Abstract
AIMS Renal interstitial fibrosis (RIF) is marked by the epithelial-mesenchymal transition (EMT) and excessive extracellular matrix deposition. The long noncoding RNA myocardial infarction-associated transcript (MIAT) facilitates RIF; however, the molecular mechanism of MIAT in RIF remains unclear. Here, we explored the possible underlying mechanisms through which MIAT modulates RIF. MATERIALS AND METHODS MIAT expression in human renal fibrotic tissues and unilateral ureteral obstruction (UUO) model mice was detected by qPCR. Transforming growth factor β1 (TGF-β1) was introduced to stimulate the EMT in human renal proximal tubular epithelial (HK-2) cells. CCK8, EdU, transwell and wound healing assays were employed to measure cell viability, proliferation, and migration respectively. RNA immunoprecipitation (RIP) assays and dual luciferase reporter assays were applied to determine the relationships among MIAT, miR-145, and EIF5A2. KEY FINDINGS MIAT was upregulated in human renal fibrotic tissues and UUO model mice compared with normal tissue adjacent to renal tumors and sham operation mice, respectively. MIAT knockdown reduced cell viability, proliferation, migration, and the EMT in HK-2 cells. Additionally, MIAT served as an endogenous sponge for miR-145 in the TGF-β1-induced-EMT in HK-2 cells, as demonstrated by dual luciferase reporter assays and RIP assays. EIF5A2 was confirmed as a target of miR-145, and MIAT knockdown suppressed EIF5A2 expression by sponging miR-145. Downregulation of EIF5A2 partly reversed induction of the EMT by miR-145 inhibitor transfection. SIGNIFICANCE MIAT promoted cell viability, proliferation, migration, and the EMT via regulation of the miR-145/EIF5A2 axis. These data established a potential therapy for RIF.
Collapse
Affiliation(s)
- Zhaohui Wang
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Bo Zhang
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Zhi Chen
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Yao He
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Feng Ru
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Peihua Liu
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, PR China.
| | - Xiang Chen
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, PR China.
| |
Collapse
|
20
|
Wu ZY, Trenner M, Boon RA, Spin JM, Maegdefessel L. Long noncoding RNAs in key cellular processes involved in aortic aneurysms. Atherosclerosis 2019; 292:112-118. [PMID: 31785492 PMCID: PMC6949864 DOI: 10.1016/j.atherosclerosis.2019.11.013] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 11/05/2019] [Accepted: 11/13/2019] [Indexed: 12/22/2022]
Abstract
Aortic aneurysm (AA) is a complex and dangerous vascular disease, featuring progressive and irreversible vessel dilatation. AA is typically detected either by screening, or identified incidentally through imaging studies. To date, no effective pharmacological therapies have been identified for clinical AA management, and either endovascular repair or open surgery remains the only option capable of preventing aneurysm rupture. In recent years, multiple research groups have endeavored to both identify noncoding RNAs and to clarify their function in vascular diseases, including aneurysmal pathologies. Notably, the molecular roles of noncoding RNAs in AA development appear to vary significantly between thoracic aortic aneurysms (TAAs) and abdominal aortic aneurysms (AAAs). Some microRNAs (miRNA - a non-coding RNA subspecies) appear to contribute to AA pathophysiology, with some showing major potential for use as biomarkers or as therapeutic targets. Studies of long noncoding RNAs (lncRNAs) are more limited, and their specific contributions to disease development and progression largely remain unexplored. This review aims to summarize and discuss the most current data on lncRNAs and their mediation of AA pathophysiology. This current review covers studies that have identified long non-coding RNAs in aortic aneurysm development and progression. We separately discuss transcripts and mechanisms of importance to thoracic as well as abdominal aortic aneurysms. Functional data on lncRNAs being identified are highlighted. Some have been studied in human as well as experimental models of the disease pathology.
Collapse
Affiliation(s)
- Zhi-Yuan Wu
- Department of Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Munich, Germany
| | - Matthias Trenner
- Department of Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Reinier A Boon
- Department of Physiology, VU University Medical Center Amsterdam, Netherlands; Institute for Cardiovascular Regeneration, University Frankfurt, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhein-Main, Germany
| | - Joshua M Spin
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Lars Maegdefessel
- Department of Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Munich, Germany; Department of Medicine, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|