1
|
Toft K, Honoré ML, Ripley N, Nielsen MK, Mardahl M, Fromm B, Hedberg-Alm Y, Tydén E, Nielsen LN, Nejsum P, Thamsborg SM, Cirera S, Pihl TH. Profiling host- and parasite-derived miRNAs associated with Strongylus vulgaris infection in horses. Vet Parasitol 2025; 334:110379. [PMID: 39721258 DOI: 10.1016/j.vetpar.2024.110379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 12/11/2024] [Accepted: 12/16/2024] [Indexed: 12/28/2024]
Abstract
The equine bloodworm, Strongylus vulgaris, is a common and highly pathogenic parasite in horses due to its migratory life cycle involving the intestinal arteries. Current diagnostic techniques cannot detect the prepatent migrating stages of S. vulgaris, highlighting the need for new biomarkers. Parasites release microRNAs (miRNAs) into their environment, which could potentially be detectable in host blood samples. Additionally, host miRNA expression patterns may change in response to infection. This study aimed to identify miRNAs associated with S. vulgaris infection by profiling the horse's miRNA response in the larval predilection site, the Cranial Mesenteric Artery (CMA) and examining the circulating parasite and horse-derived miRNAs in plasma of S. vulgaris-infected horses. Plasma samples were collected from 27 horses naturally infected with S. vulgaris and 28 uninfected horses. Arterial tissue samples from the CMA and Aorta were collected from a subset (n = 12) of the infected horses. Small RNA sequencing (small RNAseq) of a subset of the plasma samples (n = 12) identified miRNAs of interest, followed by quantitative real-time PCR (qPCR) evaluation of selected miRNAs in plasma from a larger cohort of horses. Small RNAseq detected 138 parasite-derived and 533 horse-derived miRNAs in the plasma samples. No difference in parasite-derived miRNA abundance was found between the infected and uninfected horses, but 140 horse-derived miRNAs were significantly differentially abundant between the two groups. When evaluated by qPCR, none of the selected parasite-derived miRNAs were detectable in plasma, but seven horse-derived miRNAs were confirmed differentially abundant in plasma between the two groups. Seven horse-derived miRNAs were differentially expressed in CMA tissue affected by migrating S. vulgaris compared with unaffected aortic tissue, with Eca-Mir-223-3p (Log2FC: 4.74) and Eca-Mir-140-3p (Log2FC: -3.64) being most differentially expressed. A receiver operating characteristic curve analysis suggested that Eca-Mir-486-5p and Eca-Mir-140-3p had the best diagnostic performance for distinguishing between infected and uninfected horses, with areas under the curve (AUC) of 0.78 and 0.77, respectively. Notably, Eca-Mir-140-3p was associated with age, and correcting for interaction with age increased the AUC to 0.96. In conclusion, several horse-derived miRNAs were associated with S. vulgaris infection and could differentiate between infected and uninfected horses based on their plasma abundance. However, the levels of these miRNAs were influenced by other factors (i.e age, breed), complicating their use as biomarkers. Parasite-derived miRNA abundance did not differ between S. vulgaris infected horses and those infected with other parasites using small RNAseq and were below detection limits of qPCR.
Collapse
Affiliation(s)
- Katrine Toft
- Department of Veterinary Clinical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Marie Louise Honoré
- Department of Veterinary Clinical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Nichol Ripley
- M.H. Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, KY, USA
| | - Martin K Nielsen
- M.H. Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, KY, USA
| | | | - Bastian Fromm
- The Arctic University Museum of Norway, UiT, the Arctic University of NorwayTromsø, Norway
| | - Ylva Hedberg-Alm
- Department of Biomedical Science and Veterinary Public Health, Parasitology Unit, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Eva Tydén
- Department of Biomedical Science and Veterinary Public Health, Parasitology Unit, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Lise N Nielsen
- Department of Veterinary Clinical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Peter Nejsum
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Stig Milan Thamsborg
- Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Susanna Cirera
- Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Tina Holberg Pihl
- Department of Veterinary Clinical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
2
|
Mani AM, Lamin V, Peach RC, Friesen EH, Wong T, Singh MV, Dokun AO. miRNA-6236 Regulation of Postischemic Skeletal Muscle Angiogenesis. J Am Heart Assoc 2024:e035923. [PMID: 39604034 DOI: 10.1161/jaha.124.035923] [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: 04/05/2024] [Accepted: 10/22/2024] [Indexed: 11/29/2024]
Abstract
BACKGROUND Peripheral arterial disease affects >200 million people worldwide and is characterized by impaired blood flow to the lower extremities. There are no effective medical treatments available. Using the mouse hind-limb ischemia model and miRNA sequencing, we identified a novel miRNA, miR-6236, whose expression significantly elevated in ischemic mouse limbs compared with nonischemic limbs. The role of miR-6236 in general or in postischemic angiogenesis is not known. Here we describe its role using in vivo and in vitro models of peripheral arterial disease. METHODS AND RESULTS In primary mouse and human endothelial cells, we studied the effect of simulated ischemia on miR-6236 expression and assessed its role in cell viability, apoptosis, migration, and tube formation during ischemia. Furthermore, we developed miR-6236 null mice and tested its role in postischemic perfusion recovery using the hind-limb ischemia model. Lastly, using bioinformatics and gene expression analysis, we identified putative angiogenic miR-6236 targets. In vitro simulated ischemia-enhanced miR-6236 expression in mouse and human endothelial cells, whereas its inhibition improved viability, migration, tube formation, and reduced apoptosis. In vivo ischemic mouse skeletal muscle tissue showed higher miR-6236 expression compared with nonischemic muscles. Loss of miR-6236 improved impaired postischemic perfusion recovery and poor angiogenesis associated with streptozotocin-induced diabetes in mice. Six of the 8 miR-6236 predicted angiogenic target mRNAs showed expression consistent with regulation by miR-6236 in ischemic skeletal muscle. CONCLUSIONS Our results show for the first time that miR-6236 plays a key role in regulating postischemic perfusion recovery and angiogenesis.
Collapse
Affiliation(s)
- Arul M Mani
- Division of Endocrinology and Metabolism, Carver College of Medicine University of Iowa Iowa City IA USA
| | - Victor Lamin
- Division of Endocrinology and Metabolism, Carver College of Medicine University of Iowa Iowa City IA USA
| | - Ronan C Peach
- Division of Endocrinology and Metabolism, Carver College of Medicine University of Iowa Iowa City IA USA
| | - Eli H Friesen
- Division of Endocrinology and Metabolism, Carver College of Medicine University of Iowa Iowa City IA USA
| | - Thomas Wong
- Division of Endocrinology and Metabolism, Carver College of Medicine University of Iowa Iowa City IA USA
| | - Madhu V Singh
- Division of Endocrinology and Metabolism, Carver College of Medicine University of Iowa Iowa City IA USA
| | - Ayotunde O Dokun
- Division of Endocrinology and Metabolism, Carver College of Medicine University of Iowa Iowa City IA USA
- Fraternal Order of Eagles Diabetes Research Centre, Carver College of Medicine University of Iowa Iowa City IA USA
| |
Collapse
|
3
|
Wu S, Cheng G, Lu W, Xu Y. Postoperative serum mir-28-5p level has predictive value for the prognosis after endovascular abdominal aortic aneurysm repair. J Cardiothorac Surg 2024; 19:267. [PMID: 38664759 PMCID: PMC11044549 DOI: 10.1186/s13019-024-02758-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 03/30/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND We explored the clinical significance of miR-28-5p pre- and post-endovascular abdominal aortic aneurysm repair (EVAR) in abdominal aortic aneurysm (AAA) patients. METHODS Subjects included AAA patients receiving EVAR and non-AAA people without statistical differences from AAA patient in comorbidities/Framingham risk score. Fasting elbow venous blood (4 mL) was collected in the morning of the day of EVAR surgery and in the morning of 3 months post-EVAR. Pre-/post-EVAR serum miR-28-5p expression, AAA maximum diameter alterations, CD3+/CD4+/CD8+/TC/TG pre-/post-EVAR, and the correlations between miR-28-5p and AAA maximum diameter were investigated. Prediction of miR-28-5p on post-EVAR mortality, prognosis, and independent factors of post-EVAR death were analyzed using receiver operating characteristic curve (ROC)/Kaplan-Meier curve/univariable and multivariable Cox regression. According to the cut-off value of ROC curve for postoperative miR-28-5p was the cut-off value, and the patients were classified into the miR-28-5p high- and low-expression groups. The survival or death of both groups were compared after 48-month follow-up. RESULTS Serum miR-28-5p levels in AAA patients dropped post-EVAR. AAA patients showed notable differences in CD3+/CD4+/CD8+/TC/TG levels pre-/post-EVAR. The miR-28-5p low-expression group exhibited higher CD3+/CD4+ and lower CD8+/TC/TG levels. We observed a positive correlation between post-EVAR miR-28-5p and AAA maximum diameter and between the pre-/post-EVAR miR-28-5p fold change and the AAA maximum diameter change. Postoperative miR-28-5p demonstrated good predictive value for postoperative death. Hypertension, Framingham risk score, TC, TG, and miR-28-5p were independent influencing factors of post-EVAR death. CONCLUSION EVAR decreased serum miR-28-5p expression in AAA patients. Post-operative miR-28-5p level and pre-/post-operative fold change level are positively-correlated with AAA diameter.
Collapse
Affiliation(s)
- Senyan Wu
- Department of Vascular Surgery, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, 100 Minjiang Avenue, Kecheng District, Quzhou, Zhejiang, 324000, China
| | - Guobing Cheng
- Department of Vascular Surgery, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, 100 Minjiang Avenue, Kecheng District, Quzhou, Zhejiang, 324000, China.
| | - Wei Lu
- Department of Vascular Surgery, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, 100 Minjiang Avenue, Kecheng District, Quzhou, Zhejiang, 324000, China
| | - Youyao Xu
- Department of Vascular Surgery, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, 100 Minjiang Avenue, Kecheng District, Quzhou, Zhejiang, 324000, China
| |
Collapse
|
4
|
Thanigaimani S, Iyer V, Bingley J, Browne D, Phie J, Doolan D, Golledge J. Association Between Serum MicroRNAs and Abdominal Aortic Aneurysm Diagnosis and Growth. Eur J Vasc Endovasc Surg 2023; 65:573-581. [PMID: 36596338 DOI: 10.1016/j.ejvs.2022.12.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 12/13/2022] [Accepted: 12/24/2022] [Indexed: 01/01/2023]
Abstract
OBJECTIVE This study aimed to examine the association between serum microRNAs (miRNAs) and diagnosis and growth of abdominal aortic aneurysm (AAA), and to test their diagnostic and prognostic value. METHODS The expression levels of 800 miRNA tags were assessed in 108 patients with AAA, 12 age and sex matched healthy controls (HCs), and 12 patients with peripheral artery disease (PAD) using NanoString technology. Findings were assessed in an independent sample of 66 patients with AAA and 29 age and sex matched HCs by reverse transcriptase polymerase chain reaction. AAA growth was assessed by a median of three (interquartile range [IQR] 2, 3) repeat ultrasound scans over a median follow up of 1.1 (IQR 1.0, 2.0) years. The association between the miRNA and AAA diagnosis and growth was examined by regression and linear mixed effects analyses. The diagnostic and prognostic potential of the miRNAs were examined using area under the receiver operator characteristic curve (AUC), net re-classification index (NRI), and Cox hazard analyses. RESULTS In comparison with HCs, a model combining clinical risk factors, let-7b-5p and miR-548n had an AUC of 98.0% (95% confidence interval [CI] 95.6 - 100.0; p = .003) for diagnosing AAA, which was a significant improvement over clinical risk factors alone (NRI 1.74; 95% CI 1.61 - 1.87; p < .001). Compared with PAD, a model combining clinical risk factors and miR-548n had an AUC of 99.6% (95% CI 98.9 - 100.0, p = .037) for diagnosing AAA, which was a significant improvement over clinical risk factors alone (NRI 1.79, 95% CI 1.68 - 1.91; p < .001). In the longitudinal cohort, none of the miRNAs were able to predict the likelihood of reaching surgical threshold diameter better than clinical risk factors alone. CONCLUSION Serum let-7b-5p and miR548n significantly improved the ability to diagnose AAA. None of the miRNAs had independent prognosis value in predicting AAA growth.
Collapse
Affiliation(s)
- Shivshankar Thanigaimani
- Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, Queensland, Australia; Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns and Townsville, Queensland, Australia
| | - Vikram Iyer
- Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, Queensland, Australia; Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns and Townsville, Queensland, Australia; Department of Vascular Surgery, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - John Bingley
- Department of Vascular Surgery, Mater Hospital, Brisbane
| | - Daniel Browne
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns and Townsville, Queensland, Australia
| | - James Phie
- Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, Queensland, Australia; Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns and Townsville, Queensland, Australia
| | - Denise Doolan
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns and Townsville, Queensland, Australia
| | - Jonathan Golledge
- Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, Queensland, Australia; Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns and Townsville, Queensland, Australia; The Department of Vascular and Endovascular Surgery, Townsville University Hospital, Townsville, Queensland, Australia.
| |
Collapse
|
5
|
Tasopoulou KM, Argiriou C, Tsaroucha AK, Georgiadis GS. Circulating miRNAs as biomarkers for diagnosis, surveillance and post-operative follow-up of abdominal aortic aneurysms. Ann Vasc Surg 2023:S0890-5096(23)00144-9. [PMID: 36921794 DOI: 10.1016/j.avsg.2023.02.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/21/2023] [Accepted: 02/23/2023] [Indexed: 03/15/2023]
Abstract
OBJECTIVE To provide a summary of the current state of research in English medical literature on circulating miRNAs, as biomarkers for AAA. Additionally, for the most commonly mentioned circulating miRNAs in the literature, to attempt a documentation of the biological mechanisms underlying their role in AAA development. METHODS A literature search was undertaken in the MEDLINE database. Only reports that involved peripheral blood samples (whole blood, plasma, serum) were included. The following terms were used in combination: microrna, mirna, abdominal aortic aneurysm, human, circulating, plasma, serum, endovascular and EVAR. RESULTS A total of 25 reports, published from 2012 to 2022 were included with a total of 1259 patients with AAA, predominantly men (N= 1040, 90%). Six of these reports recruited healthy donors who underwent ultrasound screening for AAA as control samples. The majority of studies were undertaken in plasma samples and the most preferred microRNA profiling method was Real - Time quantitative polymerase chain reaction (qRT-PCR). The following nine miRNAs (out of a total of 76) were studied in more than two references: miR-145, miR-24, miR-33, miR-125, let-7, miR-15, miR-191, miR-29 and miR-133. CONCLUSION The nine miRNAs described in this study, are implicated in known pathogenetic mechanisms of AAA such as atherosclerosis, vascular smooth muscle cell phenotype switch and apoptosis, vascular inflammation, extracellular matrix degradation and lipid metabolism. Identifying disease-specific miRNAs, in combination with other clinical parameters, as indicators of AAA, is crucial for early diagnosis as well as follow-up of AAAs. For future research on miRNAs as AAA biomarkers, strict case and control group definitions, sample acquisition protocols, and miRNA expression profiling techniques are warranted.
Collapse
Affiliation(s)
- Kalliopi-Maria Tasopoulou
- Department of Vascular Surgery, Medical School, Democritus University of Thrace, University General Hospital of Evros, Alexandroupolis, Greece.
| | - Christos Argiriou
- Department of Vascular Surgery, Medical School, Democritus University of Thrace, University General Hospital of Evros, Alexandroupolis, Greece
| | - Alexandra K Tsaroucha
- Laboratory of Experimental Surgery and Surgical Research, School of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - George S Georgiadis
- Department of Vascular Surgery, Medical School, Democritus University of Thrace, University General Hospital of Evros, Alexandroupolis, Greece
| |
Collapse
|
6
|
Škrlec I. Circadian system microRNAs - Role in the development of cardiovascular diseases. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 137:225-267. [PMID: 37709378 DOI: 10.1016/bs.apcsb.2023.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
Abstract
Circadian rhythm regulates numerous physiological processes, and disruption of the circadian clock can lead to cardiovascular disease. Cardiovascular disease is the leading cause of morbidity and mortality worldwide. Small non-coding RNAs, microRNAs (miRNAs), are involved in regulating gene expression, both those important for the cardiovascular system and key circadian clock genes. Epigenetic mechanisms based on miRNAs are essential for fine-tuning circadian physiology. Indeed, some miRNAs depend on circadian periodicity, others are under the influence of light, and still others are under the influence of core clock genes. Dysregulation of miRNAs involved in circadian rhythm modulation has been associated with inflammatory conditions of the endothelium and atherosclerosis, which can lead to coronary heart disease and myocardial infarction. Epigenetic processes are reversible through their association with environmental factors, enabling innovative preventive and therapeutic strategies for cardiovascular disease. Here, is a review of recent findings on how miRNAs modulate circadian rhythm desynchronization in cardiovascular disease. In the era of personalized medicine, the possibility of treatment with miRNA antagomirs should be time-dependent to correspond to chronotherapy and achieve the most significant efficacy.
Collapse
Affiliation(s)
- Ivana Škrlec
- Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia.
| |
Collapse
|
7
|
Nopp S, van der Bent ML, Kraemmer D, Königsbrügge O, Wojta J, Pabinger I, Ay C, Nossent AY. Circulatory miR-411-5p as a Novel Prognostic Biomarker for Major Adverse Cardiovascular Events in Patients with Atrial Fibrillation. Int J Mol Sci 2023; 24:3861. [PMID: 36835272 PMCID: PMC9964230 DOI: 10.3390/ijms24043861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/12/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023] Open
Abstract
The risk stratification of patients with atrial fibrillation (AF) for subsequent cardiovascular events could help in guiding prevention strategies. In this study, we aimed at investigating circulating microRNAs as prognostic biomarkers for major adverse cardiovascular events (MACE) in AF patients. We conducted a three-stage nested case-control study within the framework of a prospective registry, including 347 AF patients. First, total small RNA-sequencing was performed in 26 patients (13 cases with MACE) and the differential expression of microRNAs was analyzed. Seven candidate microRNAs with promising results in a subgroup analysis on cardiovascular death were selected and measured via using RT-qPCR in 97 patients (42 cases with cardiovascular death). To further validate our findings and investigate broader clinical applicability, we analyzed the same microRNAs in a subsequent nested case-control study of 102 patients (37 cases with early MACE) by using Cox regression. In the microRNA discovery cohort (n = 26), we detected 184 well-expressed microRNAs in circulation without overt differential expression between the cases and controls. A subgroup analysis on cardiovascular death revealed 26 microRNAs that were differentially expressed at a significance level < 0.05 (three of which with an FDR-adjusted p-value <0.05). We, therefore, proceeded with a nested case-control approach (n = 97) focusing on patients with cardiovascular death and selected, in total, seven microRNAs for further RT-qPCR analysis. One microRNA, miR-411-5p, was significantly associated with cardiovascular death (adjusted HR (95% CI): 1.95 (1.04-3.67)). Further validation (n = 102) in patients who developed early MACE showed similar results (adjusted HR (95% CI) 2.35 (1.17-4.73)). In conclusion, circulating miR-411-5p could be a valuable prognostic biomarker for MACE in AF patients.
Collapse
Affiliation(s)
- Stephan Nopp
- Clinical Division of Haematology and Haemostaseology, Department of Medicine I, Medical University of Vienna, 1090 Vienna, Austria
| | - M. Leontien van der Bent
- Department of Surgery and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2333 Leiden, The Netherlands
| | - Daniel Kraemmer
- Clinical Division of Haematology and Haemostaseology, Department of Medicine I, Medical University of Vienna, 1090 Vienna, Austria
| | - Oliver Königsbrügge
- Clinical Division of Haematology and Haemostaseology, Department of Medicine I, Medical University of Vienna, 1090 Vienna, Austria
| | - Johann Wojta
- Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria
- Ludwig Boltzmann Institute for Cardiovascular Research, 1090 Vienna, Austria
| | - Ingrid Pabinger
- Clinical Division of Haematology and Haemostaseology, Department of Medicine I, Medical University of Vienna, 1090 Vienna, Austria
| | - Cihan Ay
- Clinical Division of Haematology and Haemostaseology, Department of Medicine I, Medical University of Vienna, 1090 Vienna, Austria
| | - Anne Yaël Nossent
- Department of Surgery and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2333 Leiden, The Netherlands
- Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria
- Department of Laboratory Medicine, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
8
|
Teixeira AR, Ferreira VV, Pereira-da-Silva T, Ferreira RC. The role of miRNAs in the diagnosis of stable atherosclerosis of different arterial territories: A critical review. Front Cardiovasc Med 2022; 9:1040971. [PMID: 36505351 PMCID: PMC9733725 DOI: 10.3389/fcvm.2022.1040971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 10/20/2022] [Indexed: 11/26/2022] Open
Abstract
Atherosclerotic disease is a major cause of morbidity and mortality worldwide. Atherosclerosis may be present in different arterial territories and as a single- or multi-territorial disease. The different phenotypes of atherosclerosis are attributable only in part to acquired cardiovascular risk factors and genetic Mendelian inheritance. miRNAs, which regulate the gene expression at the post-transcriptional level, may also contribute to such heterogeneity. Numerous miRNAs participate in the pathophysiology of atherosclerosis by modulating endothelial function, smooth vascular cell function, vascular inflammation, and cholesterol homeostasis in the vessel, among other biological processes. Moreover, miRNAs are present in peripheral blood with high stability and have the potential to be used as non-invasive biomarkers for the diagnosis of atherosclerosis. However, the circulating miRNA profile may vary according to the involved arterial territory, considering that atherosclerosis expression, including the associated molecular phenotype, varies according to the affected arterial territory. In this review, we discuss the specific circulating miRNA profiles associated with atherosclerosis of different arterial territories, the common circulating miRNA profile of stable atherosclerosis irrespective of the involved arterial territory, and the circulating miRNA signature of multi-territorial atherosclerosis. miRNAs may consist of a simple non-invasive method for discriminating atherosclerosis of different arterial sites. The limitations of miRNA profiling for such clinical application are also discussed.
Collapse
Affiliation(s)
- Ana Rita Teixeira
- Department of Cardiology, Hospital de Santa Marta, Centro Hospitalar Universitário de Lisboa Central, Lisbon, Portugal
- *Correspondence: Ana Rita Teixeira
| | - Vera Vaz Ferreira
- Department of Cardiology, Hospital de Santa Marta, Centro Hospitalar Universitário de Lisboa Central, Lisbon, Portugal
| | - Tiago Pereira-da-Silva
- Department of Cardiology, Hospital de Santa Marta, Centro Hospitalar Universitário de Lisboa Central, Lisbon, Portugal
- NOVA Medical School | Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Rui Cruz Ferreira
- Department of Cardiology, Hospital de Santa Marta, Centro Hospitalar Universitário de Lisboa Central, Lisbon, Portugal
| |
Collapse
|
9
|
Yang L, Sui HG, Wang MM, Li JY, He XF, Li JY, Wang XZ. MiR-30c-1-3p targets matrix metalloproteinase 9 involved in the rupture of abdominal aortic aneurysms. J Mol Med (Berl) 2022; 100:1209-1221. [PMID: 35840740 PMCID: PMC9329399 DOI: 10.1007/s00109-022-02230-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 06/23/2022] [Accepted: 06/27/2022] [Indexed: 11/26/2022]
Abstract
Abstract Abdominal aortic aneurysm (AAA) can be fatal if ruptured, but there is no predictive biomarker. Our aim was to evaluate the prognostic potential of microRNAs (miRNAs/miRs) in an AAA mouse model and patients with unruptured AAA (URAAA) and ruptured AAA (RAAA). Among the 64 miRNAs differentially expressed in mice with AAA compared to control, miR-30c-1-3p, miR-432-3p, miR-3154, and miR-379-5p had high homology with human miRNAs. MiR-30c-1-3p plasma levels were significantly lower in patients with RAAA than in those with URAAA or control and tended to negatively correlate with the maximum aortic diameter (r = −0.3153, P = 0.06109). MiR-30c-1-3p targeted matrix metalloproteinase (MMP)-9 mRNA through the coding region and downregulated its expression in vitro. MMP-9 plasma concentrations were significantly higher in the RAAA group than in the URAAA group (P < 0.001) and were negatively associated with miR-30c-1-3p levels (r = −0.3671, P = 0.01981) and positively–with the maximal aortic diameter (r = 0.6251, P < 0.0001). The optimal cutoff values for MMP-9 expression and the maximal aortic diameter were 461.08 ng/ml and 55.95 mm, with areas under the curve of 0.816 and 0.844, respectively. Our results indicate that plasma levels of miR-30c-1-3p and MMP-9 may be candidate biomarkers of AAA progression. Key messages Downregulation of miR-30c-1-3p expression and upregulation of its potential target MMP-9 are predictors of the devastation of AAA. Downregulation of miR-30c-1-3p expression and its downstream impact on MMP-9 have a potential on predicting the development and rupture of AAA.
Supplementary Information The online version contains supplementary material available at 10.1007/s00109-022-02230-2.
Collapse
Affiliation(s)
- Lin Yang
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, Liaoning, 110167, China
- Department of Cardiology and Cardiovascular Research Institute, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, 110016, Liaoning, China
| | - Hong-Gang Sui
- Department of Cardiology and Cardiovascular Research Institute, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, 110016, Liaoning, China
| | - Meng-Meng Wang
- Department of Cardiology and Cardiovascular Research Institute, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, 110016, Liaoning, China
| | - Jia-Yin Li
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, Liaoning, 110167, China
- Department of Cardiology and Cardiovascular Research Institute, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, 110016, Liaoning, China
| | - Xiao-Feng He
- Department of Cardiology and Cardiovascular Research Institute, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, 110016, Liaoning, China
| | - Jing-Yuan Li
- Department of Cardiology and Cardiovascular Research Institute, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, 110016, Liaoning, China
| | - Xiao-Zeng Wang
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, Liaoning, 110167, China.
- Department of Cardiology and Cardiovascular Research Institute, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, 110016, Liaoning, China.
| |
Collapse
|
10
|
Han J, Luo L, Marcelina O, Kasim V, Wu S. Therapeutic angiogenesis-based strategy for peripheral artery disease. Theranostics 2022; 12:5015-5033. [PMID: 35836800 PMCID: PMC9274744 DOI: 10.7150/thno.74785] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 06/14/2022] [Indexed: 01/12/2023] Open
Abstract
Peripheral artery disease (PAD) poses a great challenge to society, with a growing prevalence in the upcoming years. Patients in the severe stages of PAD are prone to amputation and death, leading to poor quality of life and a great socioeconomic burden. Furthermore, PAD is one of the major complications of diabetic patients, who have higher risk to develop critical limb ischemia, the most severe manifestation of PAD, and thus have a poor prognosis. Hence, there is an urgent need to develop an effective therapeutic strategy to treat this disease. Therapeutic angiogenesis has raised concerns for more than two decades as a potential strategy for treating PAD, especially in patients without option for surgery-based therapies. Since the discovery of gene-based therapy for therapeutic angiogenesis, several approaches have been developed, including cell-, protein-, and small molecule drug-based therapeutic strategies, some of which have progressed into the clinical trial phase. Despite its promising potential, efforts are still needed to improve the efficacy of this strategy, reduce its cost, and promote its worldwide application. In this review, we highlight the current progress of therapeutic angiogenesis and the issues that need to be overcome prior to its clinical application.
Collapse
Affiliation(s)
- Jingxuan Han
- The Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China.,State and Local Joint Engineering Laboratory for Vascular Implants, Chongqing 400044, China
| | - Lailiu Luo
- The Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China.,State and Local Joint Engineering Laboratory for Vascular Implants, Chongqing 400044, China
| | - Olivia Marcelina
- The Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China.,State and Local Joint Engineering Laboratory for Vascular Implants, Chongqing 400044, China
| | - Vivi Kasim
- The Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China.,State and Local Joint Engineering Laboratory for Vascular Implants, Chongqing 400044, China.,The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China.,✉ Corresponding authors: Vivi Kasim, College of Bioengineering, Chongqing University, Chongqing, China; Phone: +86-23-65112672, Fax: +86-23-65111802, ; Shourong Wu, College of Bioengineering, Chongqing University, Chongqing, China; Phone: +86-23-65111632, Fax: +86-23-65111802,
| | - Shourong Wu
- The Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China.,State and Local Joint Engineering Laboratory for Vascular Implants, Chongqing 400044, China.,The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China.,✉ Corresponding authors: Vivi Kasim, College of Bioengineering, Chongqing University, Chongqing, China; Phone: +86-23-65112672, Fax: +86-23-65111802, ; Shourong Wu, College of Bioengineering, Chongqing University, Chongqing, China; Phone: +86-23-65111632, Fax: +86-23-65111802,
| |
Collapse
|
11
|
Abdallah HY, Hassan R, Fareed A, Abdelgawad M, Mostafa SA, Mohammed EAM. Identification of a circulating microRNAs biomarker panel for non-invasive diagnosis of coronary artery disease: case-control study. BMC Cardiovasc Disord 2022; 22:286. [PMID: 35751015 PMCID: PMC9233383 DOI: 10.1186/s12872-022-02711-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 06/09/2022] [Indexed: 12/07/2022] Open
Abstract
Background Circulating microRNAs (miRNAs) are considered a hot spot of research that can be employed for monitoring and/or diagnostic purposes in coronary artery disease (CAD). Since different disease features might be reflected on altered profiles or plasma miRNAs concentrations, a combination of miRNAs can provide more reliable non-invasive biomarkers for CAD. Subjects and methods We investigated a panel of 14-miRNAs selected using bioinformatics databases and current literature searching for miRNAs involved in CAD using quantitative real-time PCR technique in 73 CAD patients compared to 73 controls followed by function and pathway enrichment analysis for the 14-miRNAs. Results Our results revealed three out of the 14 circulating miRNAs understudy; miRNAs miR133a, miR155 and miR208a were downregulated. While 11 miRNAs were up-regulated in a descending order from highest fold change to lowest: miR-182, miR-145, miR-21, miR-126, miR-200b, miR-146A, miR-205, miR-135b, miR-196b, miR-140b and, miR-223. The ROC curve analysis indicated that miR-145, miR-182, miR-133a and, miR-205 were excellent biomarkers with the highest AUCs as biomarkers in CAD. All miRNAs under study except miR-208 revealed a statistically significant relation with dyslipidemia. MiR-126 and miR-155 showed significance with BMI grade, while only miR-133a showed significance with the obese patients in general. MiR-135b and miR-140b showed a significant correlation with the Wall Motion Severity Index. Pathway enrichment analysis for the miRNAS understudy revealed pathways relevant to the fatty acid biosynthesis, ECM-receptor interaction, proteoglycans in cancer, and adherens junction. Conclusion The results of this study identified a differentially expressed circulating miRNAs signature that can discriminate CAD patients from normal subjects. These results provide new insights into the significant role of miRNAs expression associated with CAD pathogenesis. Supplementary Information The online version contains supplementary material available at 10.1186/s12872-022-02711-9.
Collapse
Affiliation(s)
- Hoda Y Abdallah
- Medical Genetics Unit, Department of Histology and Cell Biology, Faculty of Medicine, Suez Canal University, Ismailia, 41522, Egypt. .,Center of Excellence in Molecular & Cellular Medicine, Faculty of Medicine, Suez Canal University, Ismailia, Egypt.
| | - Ranya Hassan
- Department of Clinical Pathology, Faculty of Medicine, Suez Canal University, Ismailia, 41522, Egypt
| | - Ahmed Fareed
- Department of Cardiology, Faculty of Medicine, Suez Canal University, Ismailia, 41522, Egypt
| | - Mai Abdelgawad
- Biotechnology and Life Sciences Department, Faculty of Postgraduate Studies for Advanced Sciences (PSAS), Beni-Suef University, Beni-Suef, 62511, Egypt
| | - Sally Abdallah Mostafa
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Eman Abdel-Moemen Mohammed
- Medical Genetics Unit, Department of Histology and Cell Biology, Faculty of Medicine, Suez Canal University, Ismailia, 41522, Egypt.,Center of Excellence in Molecular & Cellular Medicine, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
12
|
Hasemaki N, Andreou NP, Legaki E, Katsargyris A, Gazouli M, Klonaris C. Association of miRNA-145 Single Nucleotide Polymorphisms in Abdominal Aortic Aneurysms. In Vivo 2022; 36:1120-1125. [PMID: 35478113 PMCID: PMC9087113 DOI: 10.21873/invivo.12810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/19/2022] [Accepted: 02/21/2022] [Indexed: 11/10/2022]
Abstract
BACKGROUND/AIM MicroRNAs (miRNAs) are non-coding RNA molecules that exert post-transcriptional gene expression regulation in response to cellular or environmental changes. Genetic variation affects their synthesis and cellular actions, and single nucleotide polymorphisms (SNPs) are one example of genetic variants studied in relation to various diseases. Literature indicates that the differentially expressed miRNA-145 in patients' serum is an essential biomarker for abdominal aortic aneurysm (AAA). However, the correlation between specific miR-145 genetic polymorphisms with AAA susceptibility is inadequately studied. MATERIALS AND METHODS Eighty-seven AAA patients and 122 healthy controls were recruited. Peripheral blood samples were genotyped for miRNA-145 SNPs; rs55945735, rs73798217 and rs353291. RESULTS The GG genotype of the rs55945735 polymorphism (p=0.047) and the AG genotype of the rs353291 polymorphism (p=0.036) were overrepresented in AAA patients compared to healthy individuals, revealing an association with susceptibility to AAA development. CONCLUSION SNPs rs55945735 and rs353291 are associated with AAA susceptibility.
Collapse
Affiliation(s)
- Natasha Hasemaki
- First Department of Surgery, Vascular Unit, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
| | - Nikolaos-Panagiotis Andreou
- Department of Basic Medical Science, Laboratory of Biology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Evangelia Legaki
- Department of Basic Medical Science, Laboratory of Biology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Athanasios Katsargyris
- First Department of Surgery, Vascular Unit, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
- Department of Vascular and Endovascular Surgery, General Hospital Nuremberg, Paracelsus Medical University, Nuremberg, Germany
| | - Maria Gazouli
- Department of Basic Medical Science, Laboratory of Biology, Medical School, National and Kapodistrian University of Athens, Athens, Greece;
| | - Christos Klonaris
- First Department of Surgery, Vascular Unit, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
| |
Collapse
|
13
|
Ziegler L, Hedin U, Gottsäter A. Circulating Biomarkers in Lower Extremity Artery Disease. Eur Cardiol 2022; 17:e09. [PMID: 35401792 PMCID: PMC8978021 DOI: 10.15420/ecr.2021.58] [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: 12/20/2021] [Accepted: 01/31/2022] [Indexed: 12/24/2022] Open
Abstract
Lower extremity artery disease (LEAD), a chronic condition with disturbed lower extremity circulation due to narrowing of the arteries, is predominantly caused by atherosclerosis and is associated with the presence of cardiovascular risk factors and an increased risk of cardiovascular events. LEAD is prevalent among older individuals and predicted to rise with the ageing population. In progressive disease, the patient experiences symptoms of ischaemia when walking and, in advanced critical limb-threatening ischaemia, even at rest. However, LEAD is asymptomatic in most patients, delaying diagnosis and treatment. In this setting, circulating biomarkers may facilitate earlier diagnosis in selected individuals. This review provides a broad overview of the circulating biomarkers investigated to date in relation to LEAD and discusses their usefulness in clinical practice.
Collapse
Affiliation(s)
- Louise Ziegler
- Division of Internal Medicine, Department of Clinical Sciences, Karolinska Institute, Danderyd Hospital, Stockholm, Sweden
| | - Ulf Hedin
- Vascular Surgery Division, Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
| | - Anders Gottsäter
- Department of Medicine, Lund University, Malmö, Sweden; Department of Medicine, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
14
|
Zalewski DP, Ruszel KP, Stępniewski A, Gałkowski D, Feldo M, Kocki J, Bogucka-Kocka A. Relationships between Indicators of Lower Extremity Artery Disease and miRNA Expression in Peripheral Blood Mononuclear Cells. J Clin Med 2022; 11:1619. [PMID: 35329950 PMCID: PMC8948757 DOI: 10.3390/jcm11061619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 03/11/2022] [Accepted: 03/12/2022] [Indexed: 12/24/2022] Open
Abstract
Lower extremity artery disease (LEAD) is an underdiagnosed and globally underestimated vascular disease caused by the progressive and chronic formation of atherosclerotic plaques in the arteries of the lower limbs. Much evidence indicates that the abnormal course of pathophysiological processes underlying LEAD development is associated with altered miRNA modulatory function. In the presented study, relationships between miRNA expression and clinical indicators of this disease (ABI, claudication distance, length of arterial occlusion, Rutherford category, and plaque localization) were identified. MiRNA expression profiles were obtained using next-generation sequencing in peripheral blood mononuclear cells (PBMCs) of 40 LEAD patients. Correlation analysis performed using the Spearman rank correlation test revealed miRNAs related to ABI, claudication distance, and length of arterial occlusion. In the DESeq2 analysis, five miRNAs were found to be dysregulated in patients with Rutherford category 3 compared to patients with Rutherford category 2. No miRNAs were found to be differentially expressed between patients with different plaque localizations. Functional analysis performed using the miRNet 2.0 website tool determined associations of selected miRNAs with processes underlying vascular pathology, such as vascular smooth muscle cell differentiation, endothelial cell apoptosis, response to hypoxia, inflammation, lipid metabolism, and circadian rhythm. The most enriched functional terms for genes targeted by associated miRNAs were linked to regulation of the cell cycle, regulation of the transcription process, and nuclear cellular compartment. In conclusion, dysregulations of miRNA expression in PBMCs of patients with LEAD are indicative of the disease and could potentially be used in the prediction of LEAD progression.
Collapse
Affiliation(s)
- Daniel P. Zalewski
- Chair and Department of Biology and Genetics, Medical University of Lublin, 4a Chodźki St., 20-093 Lublin, Poland;
| | - Karol P. Ruszel
- Department of Clinical Genetics, Chair of Medical Genetics, Medical University of Lublin, 11 Radziwiłłowska St., 20-080 Lublin, Poland; (K.P.R.); (J.K.)
| | - Andrzej Stępniewski
- Ecotech Complex Analytical and Programme Centre for Advanced Environmentally Friendly Technologies, University of Marie Curie-Skłodowska, 39 Głęboka St., 20-612 Lublin, Poland;
| | - Dariusz Gałkowski
- Department of Pathology and Laboratory Medicine, Rutgers-Robert Wood Johnson Medical School, One Robert Wood Johnson Place, New Brunswick, NJ 08903-0019, USA;
| | - Marcin Feldo
- Chair and Department of Vascular Surgery and Angiology, Medical University of Lublin, 11 Staszica St., 20-081 Lublin, Poland;
| | - Janusz Kocki
- Department of Clinical Genetics, Chair of Medical Genetics, Medical University of Lublin, 11 Radziwiłłowska St., 20-080 Lublin, Poland; (K.P.R.); (J.K.)
| | - Anna Bogucka-Kocka
- Chair and Department of Biology and Genetics, Medical University of Lublin, 4a Chodźki St., 20-093 Lublin, Poland;
| |
Collapse
|
15
|
Ring A, Ismaeel A, Wechsler M, Fletcher E, Papoutsi E, Miserlis D, Koutakis P. MicroRNAs in peripheral artery disease: potential biomarkers and pathophysiological mechanisms. Ther Adv Cardiovasc Dis 2022; 16:17539447221096940. [PMID: 35583375 PMCID: PMC9121511 DOI: 10.1177/17539447221096940] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 04/07/2022] [Indexed: 11/29/2022] Open
Abstract
Peripheral artery disease (PAD) is a disease of atherosclerosis in the lower extremities. PAD carries a massive burden worldwide, while diagnosis and treatment options are often lacking. One of the key points of research in recent years is the involvement of microRNAs (miRNAs), which are short 20-25 nucleotide single-stranded RNAs that can act as negative regulators of post-transcriptional gene expression. Many of these miRNAs have been discovered to be misregulated in PAD patients, suggesting a potential utility as biomarkers for PAD diagnosis. miRNAs have also been shown to play an important role in many different pathophysiological aspects involved in the initiation and progression of the disease including angiogenesis, hypoxia, inflammation, as well as other cellular functions like cell proliferation and migration. The research on miRNAs in PAD has the potential to lead to a whole new class of diagnostic tools and treatments.
Collapse
Affiliation(s)
- Andrew Ring
- Department of Biology, Baylor University, Waco,
TX, USA
| | - Ahmed Ismaeel
- Department of Biology, Baylor University, Waco,
TX, USA
| | - Marissa Wechsler
- Department of Biomedical Engineering and
Chemical Engineering, The University of Texas at San Antonio, San Antonio,
TX, USA
| | - Emma Fletcher
- Department of Biology, Baylor University, Waco,
TX, USA
| | | | - Dimitrios Miserlis
- Department of Surgery, The University of Texas
Health Science Center at San Antonio, San Antonio, TX, USA
| | - Panagiotis Koutakis
- Department of Biology, Baylor University, B.207
Baylor Science Building, One Bear Place #97388, Waco, TX 76798-7388,
USA
| |
Collapse
|
16
|
Pereira-da-Silva T, Napoleão P, Costa MC, Gabriel AF, Selas M, Silva F, Enguita FJ, Cruz Ferreira R, Mota Carmo M. Association between miR-146a and Tumor Necrosis Factor Alpha (TNF-α) in Stable Coronary Artery Disease. ACTA ACUST UNITED AC 2021; 57:medicina57060575. [PMID: 34199767 PMCID: PMC8230353 DOI: 10.3390/medicina57060575] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 06/02/2021] [Indexed: 12/12/2022]
Abstract
Background and Objectives: Tumor necrosis factor alpha (TNF-α) is proatherogenic and associated with the risk of acute ischemic events, although the mechanisms that regulate TNF-α expression in stable coronary artery disease (SCAD) are not fully understood. We investigated whether metabolic, inflammatory, and epigenetic (microRNA (miRNA)) markers are associated with TNF-α expression in SCAD. Materials and Methods: Patients with SCAD were prospectively recruited and their metabolic and inflammatory profiles were assessed. TNF-α levels were assessed using an enzyme-linked immunosorbent assay. The relative expression of six circulating miRNAs associated with the regulation of inflammation and/or atherosclerosis was determined. Results: Of the 24 included patients with the mean age of 65 (9) years, 88% were male, and 54% were diabetic. The TNF-α levels were (median (interquartile range)) 1.0 (0.7–1.1) pg/mL. The percentage of glycosylated hemoglobin (r = 0.418, p = 0.042), serum triglyceride levels (r = 0.429, p = 0.037), and C-reactive protein levels (r = 0.407, p = 0.048) were positively correlated with TNF-α levels. Of the candidate miRNAs, miR-146a expression levels were negatively correlated with TNF-α levels (as indicated by r = 0.500, p = 0.035 for correlation between delta cycle threshold (ΔCt) miR-146a and TNF-α levels). In multivariate analysis, serum triglyceride levels and miR-146a expression levels were independently associated with TNF-α levels. miR-146 expression levels were not associated with metabolic or other inflammatory parameters and were negatively correlated with the number of coronary vessels with obstructive disease (as indicated by r = 0.556, p = 0.017 for correlation between ΔCt miR-146a and number of diseased vessels). Conclusions: miR-146a expression levels were negatively correlated with TNF-α levels in patients with SCAD, irrespective of other metabolic or inflammatory markers, and with the severity of coronary artery disease. The results add to the knowledge on the role of miR-146a in TNF-α-based inflammation in SCAD and support future research on the potential therapeutic use of miR-146a in such a clinical scenario.
Collapse
Affiliation(s)
- Tiago Pereira-da-Silva
- Department of Cardiology, Hospital de Santa Marta, Centro Hospitalar Universitário de Lisboa Central, 1169-024 Lisbon, Portugal; (M.S.); (F.S.); (R.C.F.)
- NOVA Doctoral School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1169-056 Lisbon, Portugal
- Correspondence: ; Tel.: +351-919908505
| | - Patrícia Napoleão
- Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, 1649-028 Lisbon, Portugal; (P.N.); (M.C.C.); (A.F.G.); (F.J.E.)
| | - Marina C. Costa
- Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, 1649-028 Lisbon, Portugal; (P.N.); (M.C.C.); (A.F.G.); (F.J.E.)
- Cardiomics Unit, Centro Cardiovascular da Universidade de Lisboa, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - André F. Gabriel
- Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, 1649-028 Lisbon, Portugal; (P.N.); (M.C.C.); (A.F.G.); (F.J.E.)
- Cardiomics Unit, Centro Cardiovascular da Universidade de Lisboa, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Mafalda Selas
- Department of Cardiology, Hospital de Santa Marta, Centro Hospitalar Universitário de Lisboa Central, 1169-024 Lisbon, Portugal; (M.S.); (F.S.); (R.C.F.)
| | - Filipa Silva
- Department of Cardiology, Hospital de Santa Marta, Centro Hospitalar Universitário de Lisboa Central, 1169-024 Lisbon, Portugal; (M.S.); (F.S.); (R.C.F.)
| | - Francisco J. Enguita
- Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, 1649-028 Lisbon, Portugal; (P.N.); (M.C.C.); (A.F.G.); (F.J.E.)
- Cardiomics Unit, Centro Cardiovascular da Universidade de Lisboa, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Rui Cruz Ferreira
- Department of Cardiology, Hospital de Santa Marta, Centro Hospitalar Universitário de Lisboa Central, 1169-024 Lisbon, Portugal; (M.S.); (F.S.); (R.C.F.)
| | - Miguel Mota Carmo
- Chronic Diseases Research Center (CEDOC), NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1150-082 Lisbon, Portugal;
| |
Collapse
|
17
|
Newman JD, Cornwell MG, Zhou H, Rockman C, Heguy A, Suarez Y, Cheng HS, Feinberg MW, Hochman JS, Ruggles KV, Berger JS. Gene Expression Signature in Patients With Symptomatic Peripheral Artery Disease. Arterioscler Thromb Vasc Biol 2021; 41:1521-1533. [PMID: 33657880 PMCID: PMC8048111 DOI: 10.1161/atvbaha.120.315857] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 02/09/2021] [Indexed: 12/31/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Jonathan D. Newman
- Department of Medicine, Division of Cardiology and the Center for the Prevention of Cardiovascular Disease
| | - MacIntosh G. Cornwell
- Department of Medicine, Division of Translational Medicine
- Institute of Systems Genetics
| | - Hua Zhou
- Applied Bioinformatics Laboratories
| | - Caron Rockman
- Department of Surgery, Division of Vascular Surgery, New York University School of Medicine, New York, NY, 10016
| | - Adriana Heguy
- Department of Pathology, NYU School of Medicine
- Genome Technology Center, Division of Advanced Research Technologies, NYU School of Medicine
| | - Yajaira Suarez
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520
| | - Henry S. Cheng
- Department of Surgery, Division of Vascular Surgery, New York University School of Medicine, New York, NY, 10016
| | - Mark W. Feinberg
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital/Harvard Medical School, Boston, MA 02115
| | - Judith S. Hochman
- Department of Medicine, Division of Cardiology and the Center for the Prevention of Cardiovascular Disease
| | - Kelly V. Ruggles
- Department of Medicine, Division of Translational Medicine
- Institute of Systems Genetics
| | - Jeffrey S. Berger
- Department of Medicine, Division of Cardiology and the Center for the Prevention of Cardiovascular Disease
- Department of Surgery, Division of Vascular Surgery, New York University School of Medicine, New York, NY, 10016
| |
Collapse
|
18
|
Yu Q, Li Q, Yang X, Liu Q, Deng J, Zhao Y, Hu R, Dai M. Dexmedetomidine suppresses the development of abdominal aortic aneurysm by downregulating the mircoRNA‑21/PDCD 4 axis. Int J Mol Med 2021; 47:90. [PMID: 33786608 PMCID: PMC8029612 DOI: 10.3892/ijmm.2021.4923] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 01/27/2021] [Indexed: 12/12/2022] Open
Abstract
Abdominal aortic aneurysm (AAA) is a pathological state with permanent dilation, which indicates a fatal potential for aortic rupture. It has been reported that dexmedetomidine (Dex) and microRNA (miR)-21 are involved in the progression of AAA. Thus, the present study aimed to investigate the joint effects of these factors on AAA treatment. For this purpose, rat models of AAA were established with enzyme perfusion and the rats were then injected with Dex. Alterations in the abdominal aorta in rats with AAA were recorded. miR-21 expression in the rats with AAA was determined. Inflammatory factor expression was detected by western blot analysis. Subsequently, a dual-luciferase reporter gene assay was performed to verify the targeting association between miR-21 and programmed cell death protein 4 (PDCD4). Additionally, AAA-related indices and inflammatory responses were examined by an injection of a combination of antagomiR (ant)-miR-21 and Dex or lentivirus-PDCD4-short hairpin RNA. It was found that Dex markedly alleviated the development of AAA and downregulated the expression of inflammatory factors and matrix metalloproteinase in rats with AAA. The high expression of miR-21, which targets PDCD4, was observed in the rats with AAA. However, ant-miR-21 induced AAA development and inflammatory responses. Additionally, the inhibition of PDCD4 reduced AAA development and inflammatory responses. On the whole, the present study demonstrates that Dex inhibits AAA development by downregulating the miR-21/PCDP4 axis. The findings of the present study may provide novel insight for the treatment of AAA. These findings may provide a reference for the future treatment of AAA and may provide theoretical guidance for the early prevention and development of AAA.
Collapse
Affiliation(s)
- Qi Yu
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Qianqian Li
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Xinglong Yang
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Qiang Liu
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Jun Deng
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yanping Zhao
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Ruilin Hu
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Min Dai
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
19
|
Pereira-da-Silva T, Napoleão P, Costa MC, Gabriel AF, Selas M, Silva F, Enguita FJ, Ferreira RC, Carmo MM. Cigarette Smoking, miR-27b Downregulation, and Peripheral Artery Disease: Insights into the Mechanisms of Smoking Toxicity. J Clin Med 2021; 10:jcm10040890. [PMID: 33671744 PMCID: PMC7926909 DOI: 10.3390/jcm10040890] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 02/14/2021] [Accepted: 02/17/2021] [Indexed: 12/26/2022] Open
Abstract
Cigarette smoking is a risk factor for the development of peripheral artery disease (PAD), although the proatherosclerotic mediators of cigarette smoking are not entirely known. We explored whether circulating microRNAs (miRNAs) are dysregulated in cigarette smokers and associated with the presence of PAD. Ninety-four participants were recruited, including 58 individuals without and 36 with PAD, 51 never smokers, 28 prior smokers, and 15 active smokers. The relative expression of six circulating miRNAs with distinct biological roles (miR-21, miR-27b, miR-29a, miR-126, miR-146, and miR-218) was assessed. Cigarette smoking was associated with the presence of PAD in multivariate analysis. Active smokers, but not prior smokers, presented miR-27b downregulation and higher leukocyte, neutrophil, and lymphocyte counts; miR-27b expression levels were independently associated with active smoking. Considering the metabolic and/or inflammatory abnormalities induced by cigarette smoking, miR-27b was independently associated with the presence of PAD and downregulated in patients with more extensive PAD. In conclusion, the atheroprotective miR-27b was downregulated in active smokers, but not in prior smokers, and miR-27b expression was independently associated with the presence of PAD. These unreported data suggest that the proatherogenic properties of cigarette smoking are mediated by a downregulation of miR-27b, which may be attenuated by smoking cessation.
Collapse
Affiliation(s)
- Tiago Pereira-da-Silva
- Department of Cardiology, Hospital de Santa Marta, Centro Hospitalar Universitário de Lisboa Central, 1169-024 Lisbon, Portugal; (M.S.); (F.S.); (R.C.F.)
- NOVA Doctoral School, NOVA Medical School|Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1169-056 Lisbon, Portugal
- Correspondence: ; Tel.: +351-919908505
| | - Patrícia Napoleão
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal; (P.N.); (M.C.C.); (A.F.G.); (F.J.E.)
| | - Marina C. Costa
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal; (P.N.); (M.C.C.); (A.F.G.); (F.J.E.)
- Cardiomics Unit, Centro Cardiovascular da Universidade de Lisboa, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - André F. Gabriel
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal; (P.N.); (M.C.C.); (A.F.G.); (F.J.E.)
- Cardiomics Unit, Centro Cardiovascular da Universidade de Lisboa, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Mafalda Selas
- Department of Cardiology, Hospital de Santa Marta, Centro Hospitalar Universitário de Lisboa Central, 1169-024 Lisbon, Portugal; (M.S.); (F.S.); (R.C.F.)
| | - Filipa Silva
- Department of Cardiology, Hospital de Santa Marta, Centro Hospitalar Universitário de Lisboa Central, 1169-024 Lisbon, Portugal; (M.S.); (F.S.); (R.C.F.)
| | - Francisco J. Enguita
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal; (P.N.); (M.C.C.); (A.F.G.); (F.J.E.)
- Cardiomics Unit, Centro Cardiovascular da Universidade de Lisboa, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Rui Cruz Ferreira
- Department of Cardiology, Hospital de Santa Marta, Centro Hospitalar Universitário de Lisboa Central, 1169-024 Lisbon, Portugal; (M.S.); (F.S.); (R.C.F.)
| | - Miguel Mota Carmo
- Chronic Diseases Research Center (CEDOC), NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1150-082 Lisbon, Portugal;
| |
Collapse
|
20
|
Pereira-da-Silva T, Napoleão P, Costa MC, Gabriel AF, Selas M, Silva F, Enguita FJ, Ferreira RC, Carmo MM. Circulating miRNAs Are Associated with the Systemic Extent of Atherosclerosis: Novel Observations for miR-27b and miR-146. Diagnostics (Basel) 2021; 11:318. [PMID: 33669374 PMCID: PMC7920287 DOI: 10.3390/diagnostics11020318] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 02/01/2021] [Accepted: 02/12/2021] [Indexed: 12/18/2022] Open
Abstract
The mechanisms that regulate the systemic extent of atherosclerosis are not fully understood. We investigated whether the expression of circulating miRNAs is associated with the extent of stable atherosclerosis to a single territory or multiple territories (polyvascular) and with the severity of atherosclerosis in each territory. Ninety-four participants were prospectively recruited and divided into five age- and sex-matched groups: presenting no atherosclerosis, isolated coronary atherosclerosis, coronary and lower extremity atherosclerosis, coronary and carotid atherosclerosis, and atherosclerosis of the coronary, lower extremity, and carotid territories. The expression of six circulating miRNAs with distinct biological roles was assessed. The expression of miR-27b and miR-146 differed across groups (p < 0.05), showing a decrease in the presence of atherosclerosis, particularly in the three territories. miR-27b and miR-146 expression decreased in association with a higher severity of coronary, lower extremity, and carotid atherosclerosis. Polyvascular atherosclerosis involving the three territories was independently associated with a decreased miR-27b and miR-146 expression. Both miRNAs presented an area under the curve of ≥0.75 for predicting polyvascular atherosclerosis involving the three territories. To conclude, miR-27b and miR-146 were associated with the presence of severe polyvascular atherosclerosis and with the atherosclerosis severity in each territory. Both are potential biomarkers of severe systemic atherosclerosis.
Collapse
Affiliation(s)
- Tiago Pereira-da-Silva
- Department of Cardiology, Hospital de Santa Marta, Centro Hospitalar Universitário de Lisboa Central, 1169-024 Lisbon, Portugal; (M.S.); (F.S.); (R.C.F.)
- NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1169-056 Lisbon, Portugal
| | - Patrícia Napoleão
- Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, 1649-028 Lisbon, Portugal; (P.N.); (M.C.C.); (A.F.G.); (F.J.E.)
| | - Marina C. Costa
- Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, 1649-028 Lisbon, Portugal; (P.N.); (M.C.C.); (A.F.G.); (F.J.E.)
- Cardiomics Unit, Centro Cardiovascular da Universidade de Lisboa, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - André F. Gabriel
- Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, 1649-028 Lisbon, Portugal; (P.N.); (M.C.C.); (A.F.G.); (F.J.E.)
- Cardiomics Unit, Centro Cardiovascular da Universidade de Lisboa, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Mafalda Selas
- Department of Cardiology, Hospital de Santa Marta, Centro Hospitalar Universitário de Lisboa Central, 1169-024 Lisbon, Portugal; (M.S.); (F.S.); (R.C.F.)
| | - Filipa Silva
- Department of Cardiology, Hospital de Santa Marta, Centro Hospitalar Universitário de Lisboa Central, 1169-024 Lisbon, Portugal; (M.S.); (F.S.); (R.C.F.)
| | - Francisco J. Enguita
- Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, 1649-028 Lisbon, Portugal; (P.N.); (M.C.C.); (A.F.G.); (F.J.E.)
- Cardiomics Unit, Centro Cardiovascular da Universidade de Lisboa, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Rui Cruz Ferreira
- Department of Cardiology, Hospital de Santa Marta, Centro Hospitalar Universitário de Lisboa Central, 1169-024 Lisbon, Portugal; (M.S.); (F.S.); (R.C.F.)
| | - Miguel Mota Carmo
- Chronic Diseases Research Center (CEDOC), NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1150-082 Lisbon, Portugal;
| |
Collapse
|
21
|
Identification of Novel microRNA Profiles Dysregulated in Plasma and Tissue of Abdominal Aortic Aneurysm Patients. Int J Mol Sci 2020; 21:ijms21134600. [PMID: 32605321 PMCID: PMC7370113 DOI: 10.3390/ijms21134600] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/23/2020] [Accepted: 06/24/2020] [Indexed: 12/19/2022] Open
Abstract
microRNAs (miRNAs) are small RNAs that regulate different biological processes. Our objective was to identify miRNAs dysregulated in plasma and tissue of patients with abdominal aortic aneurysm (AAA) and explore new potential targets involved in AAA. Fifty-seven subjects were recruited for a plasma study (30 AAA patients, 16 healthy volunteers and 11 patients with atherosclerosis). The expression level of 179 miRNAs was screened in plasma from a subset of samples, and dysregulated miRNAs were validated in the entire study population. Dysregulated miRNAs were also quantified in aortic tissue of 21 AAA patients and 8 organ donors. Applying a gene set enrichment analysis, an interaction map of dysregulated miRNAs and their targets was built, and selected targets were quantified in tissue samples. miR-27b-3p and miR-221-3p were overexpressed in plasma of AAA patients compared with healthy controls, 1.6 times and 1.9 times, respectively. In AAA tissue, six miRNAs (miR-1, miR-27b-3p, miR-29b-3p, miR-133a-3p, miR-133b, and miR-195-5p) were underexpressed from 1.6 to 4.8 times and four miRNAs (miR-146a-5p, miR-21-5p, miR-144-3p, and miR-103a-3p) were overexpressed from 1.3 to 7.2 times. Thrombospondin-2, a target of miR-195-5p, was increased in AAA tissue and negatively correlated with the expression of miR-195-5p, suggesting their involvement in a common regulatory mechanism.
Collapse
|
22
|
Zalewski DP, Ruszel KP, Stępniewski A, Gałkowski D, Bogucki J, Komsta Ł, Kołodziej P, Chmiel P, Zubilewicz T, Feldo M, Kocki J, Bogucka-Kocka A. Dysregulation of microRNA Modulatory Network in Abdominal Aortic Aneurysm. J Clin Med 2020; 9:jcm9061974. [PMID: 32599769 PMCID: PMC7355415 DOI: 10.3390/jcm9061974] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 06/13/2020] [Accepted: 06/22/2020] [Indexed: 12/16/2022] Open
Abstract
Abdominal artery aneurysm (AAA) refers to abdominal aortic dilatation of 3 cm or greater. AAA is frequently underdiagnosed due to often asymptomatic character of the disease, leading to elevated mortality due to aneurysm rupture. MiRNA constitute a pool of small RNAs controlling gene expression and is involved in many pathologic conditions in human. Targeted panel detecting altered expression of miRNA and genes involved in AAA would improve early diagnosis of this disease. In the presented study, we selected and analyzed miRNA and gene expression signatures in AAA patients. Next, generation sequencing was applied to obtain miRNA and gene-wide expression profiles from peripheral blood mononuclear cells in individuals with AAA and healthy controls. Differential expression analysis was performed using DESeq2 and uninformative variable elimination by partial least squares (UVE-PLS) methods. A total of 31 miRNAs and 51 genes were selected as the most promising biomarkers of AAA. Receiver operating characteristics (ROC) analysis showed good diagnostic ability of proposed biomarkers. Genes regulated by selected miRNAs were determined in silico and associated with functional terms closely related to cardiovascular and neurological diseases. Proposed biomarkers may be used for new diagnostic and therapeutic approaches in management of AAA. The findings will also contribute to the pool of knowledge about miRNA-dependent regulatory mechanisms involved in pathology of that disease.
Collapse
Affiliation(s)
- Daniel P. Zalewski
- Chair and Department of Biology and Genetics, Medical University of Lublin, 4a Chodźki St., 20-093 Lublin, Poland; (D.P.Z.); (P.C.)
| | - Karol P. Ruszel
- Chair of Medical Genetics, Department of Clinical Genetics, Medical University of Lublin, 11 Radziwiłłowska St., 20-080 Lublin, Poland; (K.P.R.); (J.B.); (J.K.)
| | - Andrzej Stępniewski
- Ecotech Complex Analytical and Programme Centre for Advanced Environmentally Friendly Technologies, University of Marie Curie-Skłodowska, 39 Głęboka St., 20-612 Lublin, Poland;
| | - Dariusz Gałkowski
- Department of Pathology and Laboratory Medicine, Rutgers - Robert Wood Johnson Medical School, One Robert Wood Johnson Place, New Brunswick, NJ 08903-0019, USA;
| | - Jacek Bogucki
- Chair of Medical Genetics, Department of Clinical Genetics, Medical University of Lublin, 11 Radziwiłłowska St., 20-080 Lublin, Poland; (K.P.R.); (J.B.); (J.K.)
| | - Łukasz Komsta
- Chair and Department of Medicinal Chemistry, Medical University of Lublin, 4 Jaczewskiego St., 20-090 Lublin, Poland;
| | - Przemysław Kołodziej
- Laboratory of Diagnostic Parasitology, Chair and Department of Biology and Genetics, Medical University of Lublin, 4a Chodźki St., 20-093 Lublin, Poland;
| | - Paulina Chmiel
- Chair and Department of Biology and Genetics, Medical University of Lublin, 4a Chodźki St., 20-093 Lublin, Poland; (D.P.Z.); (P.C.)
| | - Tomasz Zubilewicz
- Chair and Department of Vascular Surgery and Angiology, Medical University of Lublin, 11 Staszica St., 20-081 Lublin, Poland; (T.Z.); (M.F.)
| | - Marcin Feldo
- Chair and Department of Vascular Surgery and Angiology, Medical University of Lublin, 11 Staszica St., 20-081 Lublin, Poland; (T.Z.); (M.F.)
| | - Janusz Kocki
- Chair of Medical Genetics, Department of Clinical Genetics, Medical University of Lublin, 11 Radziwiłłowska St., 20-080 Lublin, Poland; (K.P.R.); (J.B.); (J.K.)
| | - Anna Bogucka-Kocka
- Chair and Department of Biology and Genetics, Medical University of Lublin, 4a Chodźki St., 20-093 Lublin, Poland; (D.P.Z.); (P.C.)
- Correspondence: ; Tel.: +48-81-448-7232
| |
Collapse
|
23
|
MiR-144-5p limits experimental abdominal aortic aneurysm formation by mitigating M1 macrophage-associated inflammation: Suppression of TLR2 and OLR1. J Mol Cell Cardiol 2020; 143:1-14. [PMID: 32278833 DOI: 10.1016/j.yjmcc.2020.04.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 04/03/2020] [Accepted: 04/05/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND It has been noted that dysregulation of microRNAs (miRNAs) contributes to the formation of abdominal aortic aneurysm (AAA), a vascular disease associated with progressive aortic dilatation and degradation, and pathological infiltration and activation of inflammatory cells, such as macrophages. Our microarray data revealing that miR-144-5p was the top 1 downregulated miRNA in mouse AAA tissues as compared to normal aortas motivated us to explore its role in AAA development. METHODS We profiled miRNA and mRNA expression in Angiotensin II (Ang II)- (n = 3) and saline-infused abdominal aortas (n = 4) via Agilent microarrays, and further validated the data with real-time QPCR. In vivo, miR-144-5p or control agomirs were given to Apoe-/- mice with Ang II infusion-induced AAA. In vitro, mouse RAW 264.7 macrophages and human THP-1 macrophage-like cells were transfected with miR-144-5p or control agomirs/antagomirs, and oxidized Low Density Lipoprotein (ox-LDL) was used to stimulate M1 macrophage polarization. RESULTS Based on the microarray and real-time QPCR validation data, we identified miR-144-5p as a novel downregulated miRNA in AAA tissues. Overexpression of miR-144-5p by utilizing its specific agomirs in vivo significantly attenuated Ang II-induced aortic dilatation and elastic degradation in Apoe-/- mice and improved their survival. AAA incidence was reduced by miR-144-5p as well. MiR-144-5p polarized macrophages to M2 type in Ang II-infused aortas. Further, the expression levels of two predictive targets for miR-144-5p, Toll Like Receptor 2 (TLR2) and ox-LDL Receptor 1 (OLR1), were higher in AAA specimens, and negatively correlated to miR-144-5p (Pearson correlation coefficient r < -0.9, P < .01). These two molecules were then confirmed as novel miR-144-5p targets via dual-luciferase assay. MiR-144-5p agomirs suppressed ox-LDL-induced upregulation of M1 macrophage markers, including interleukin 1β (IL1β), tumor necrosis factor α (TNFα), prostaglandin-endoperoxide synthase 2 (PTGS2) and nitric oxide synthase 2 (NOS2), in macrophages probably by targeting TLR2. MiR-144-5p also inhibited the signaling transduction of pathways downstream to TLR2 and OLR1, including NF-κB and ERK1/2 pathways, whose abnormal activation contributed AAA formation. CONCLUSION Our work suggests miR-144-5p as a novel regulator for AAA pathology. Management of miR-144-5p and its targets TLR2 and OLR1 provides therapeutic potential for limiting AAA formation.
Collapse
|
24
|
Zhao F, Chen T, Jiang N. CDR1as/miR-7/CKAP4 axis contributes to the pathogenesis of abdominal aortic aneurysm by regulating the proliferation and apoptosis of primary vascular smooth muscle cells. Exp Ther Med 2020; 19:3760-3766. [PMID: 32346440 PMCID: PMC7185088 DOI: 10.3892/etm.2020.8622] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Accepted: 02/13/2020] [Indexed: 01/08/2023] Open
Abstract
Abdominal aortic aneurysm (AAA) is characterized as dilation of the aortic wall. Dysregulation of vascular smooth muscle cells (VSMCs) can contribute to the development of this phenotype. Circular RNAs and microRNAs (miRNAs) can regulate the proliferation and apoptosis of VSMCs. This present study aimed to identify the mechanisms of action behind the regulation of cerebellar degeneration-related protein 1 antisense RNA (CDR1as)/miRNA (miR)-7 in VSMCs. The expression levels of miR-7 were upregulated, whereas the levels of CDR1as and cytoskeleton-associated protein 4 (CKAP4) were downregulated in aortic specimens obtained from 10 patients who underwent surgery for AAA compared with aortic specimens from 10 control patients who underwent coronary artery bypass surgery. The molecular mechanism of action of CDR1as/miR-7 was investigated in primary VSMCs. The results of Cell Counting kit-8 and cell growth curve assays revealed that overexpression of CDR1as and knockdown of miR-7, increased VSMC proliferation, whereas knockdown of CDR1as and overexpression of miR-7 suppressed VSMC proliferation. In addition, overexpression of CDR1as and knockdown of miR-7, suppressed apoptosis in VSMCs, indicated by the decreased levels of reactive oxygen species (ROS) and lactate dehydrogenase (LDH) activity, whereas knockdown of CDR1as and overexpression of miR-7 exhibited the opposite effects. The results of luciferase reporter and biotin pull-down assays confirmed that CDR1as directly bound to miR-7 and suppressed its expression. Additionally, the CDR1as-induced proliferation and suppressed apoptosis was reversed by the overexpression of miR-7. Furthermore, luciferase reporter, reverse transcription-quantitative PCR and western blot assays revealed that miR-7 directly targeted CKAP4 and suppressed its expression. Additionally, the miR-7-suppressed proliferation and increased ROS and LDH activity were reversed by the overexpression of CKAP4. CDR1as also decreased caspase 3/7 activity, which was reversed by miR-7 mimics. miR-7 increased the activity of caspase 3/7, which was again reversed by the overexpression of CKAP4. Therefore, CDR1as, miR-7 and CKAP4 may act in the same pathway to regulate VSMC proliferation and apoptosis.
Collapse
Affiliation(s)
- Feng Zhao
- Department of Cardiac Surgery, Tianjin Chest Hospital, Tianjin 300222, P.R. China
| | - Tongyun Chen
- Department of Cardiac Surgery, Tianjin Chest Hospital, Tianjin 300222, P.R. China
| | - Nan Jiang
- Department of Cardiac Surgery, Tianjin Chest Hospital, Tianjin 300222, P.R. China
| |
Collapse
|
25
|
Schiano C, Benincasa G, Franzese M, Della Mura N, Pane K, Salvatore M, Napoli C. Epigenetic-sensitive pathways in personalized therapy of major cardiovascular diseases. Pharmacol Ther 2020; 210:107514. [PMID: 32105674 DOI: 10.1016/j.pharmthera.2020.107514] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The complex pathobiology underlying cardiovascular diseases (CVDs) has yet to be explained. Aberrant epigenetic changes may result from alterations in enzymatic activities, which are responsible for putting in and/or out the covalent groups, altering the epigenome and then modulating gene expression. The identification of novel individual epigenetic-sensitive trajectories at single cell level might provide additional opportunities to establish predictive, diagnostic and prognostic biomarkers as well as drug targets in CVDs. To date, most of studies investigated DNA methylation mechanism and miRNA regulation as epigenetics marks. During atherogenesis, big epigenetic changes in DNA methylation and different ncRNAs, such as miR-93, miR-340, miR-433, miR-765, CHROME, were identified into endothelial cells, smooth muscle cells, and macrophages. During man development, lipid metabolism, inflammation and homocysteine homeostasis, alter vascular transcriptional mechanism of fundamental genes such as ABCA1, SREBP2, NOS, HIF1. At histone level, increased HDAC9 was associated with matrix metalloproteinase 1 (MMP1) and MMP2 expression in pro-inflammatory macrophages of human carotid plaque other than to have a positive effect on toll like receptor signaling and innate immunity. HDAC9 deficiency promoted inflammation resolution and reverse cholesterol transport, which might block atherosclerosis progression and promote lesion regression. Here, we describe main human epigenetic mechanisms involved in atherosclerosis, coronary heart disease, ischemic stroke, peripheral artery disease; cardiomyopathy and heart failure. Different epigenetics mechanisms are activated, such as regulation by circular RNAs, as MICRA, and epitranscriptomics at RNA level. Moreover, in order to open new frontiers for precision medicine and personalized therapy, we offer a panoramic view on the most innovative bioinformatic tools designed to identify putative genes and molecular networks underlying CVDs in man.
Collapse
Affiliation(s)
- Concetta Schiano
- Clinical Department of Internal Medicine and Specialistics, Department of Advanced Clinical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy.
| | - Giuditta Benincasa
- Clinical Department of Internal Medicine and Specialistics, Department of Advanced Clinical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | | | | | | | | | - Claudio Napoli
- Clinical Department of Internal Medicine and Specialistics, Department of Advanced Clinical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy; IRCCS SDN, Naples, Italy
| |
Collapse
|
26
|
Cooper DM, Radom-Aizik S. Exercise-associated prevention of adult cardiovascular disease in children and adolescents: monocytes, molecular mechanisms, and a call for discovery. Pediatr Res 2020; 87:309-318. [PMID: 31649340 PMCID: PMC11177628 DOI: 10.1038/s41390-019-0581-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 08/07/2019] [Accepted: 08/15/2019] [Indexed: 12/28/2022]
Abstract
Atherosclerosis originates in childhood and adolescence. The goal of this review is to highlight how exercise and physical activity during childhood and adolescence, critical periods of growth and development, can prevent adult cardiovascular disease (CVD), particularly through molecular mechanisms of monocytes, a key cell of the innate immune system. Monocytes are heterogeneous and pluripotential cells that can, paradoxically, play a role in both the instigation and prevention of atherosclerosis. Recent discoveries in young adults reveal that brief exercise affects monocyte gene pathways promoting a cell phenotype that patrols the vascular system and repairs injuries. Concurrently, exercise inhibits pro-inflammatory monocytes, cells that contribute to vascular damage and plaque formation. Because CVD is typically asymptomatic in youth, minimally invasive techniques must be honed to study the subtle anatomic and physiologic evidence of vascular dysfunction. Exercise gas exchange and heart rate measures can be combined with ultrasound assessments of vascular anatomy and reactivity, and near-infrared spectroscopy to quantify impaired O2 transport that is often hidden at rest. Combined with functional, transcriptomic, and epigenetic monocyte expression and measures of monocyte-endothelium interaction, molecular mechanisms of early CVD can be formulated, and then translated into effective physical activity-based strategies in youth to prevent adult-onset CVD.
Collapse
Affiliation(s)
- Dan M Cooper
- Pediatric Exercise and Genomics Research Center, University of California Irvine School of Medicine, Pediatrics, Irvine, CA, USA.
| | - Shlomit Radom-Aizik
- Pediatric Exercise and Genomics Research Center, University of California Irvine School of Medicine, Pediatrics, Irvine, CA, USA
| |
Collapse
|
27
|
Zhuo LA, Wen YT, Wang Y, Liang ZF, Wu G, Nong MD, Miao L. LncRNA SNHG8 is identified as a key regulator of acute myocardial infarction by RNA-seq analysis. Lipids Health Dis 2019; 18:201. [PMID: 31739782 PMCID: PMC6862811 DOI: 10.1186/s12944-019-1142-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 10/27/2019] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Long noncoding RNAs (lncRNAs) are involved in numerous physiological functions. However, their mechanisms in acute myocardial infarction (AMI) are not well understood. METHODS We performed an RNA-seq analysis to explore the molecular mechanism of AMI by constructing a lncRNA-miRNA-mRNA axis based on the ceRNA hypothesis. The target microRNA data were used to design a global AMI triple network. Thereafter, a functional enrichment analysis and clustering topological analyses were conducted by using the triple network. The expression of lncRNA SNHG8, SOCS3 and ICAM1 was measured by qRT-PCR. The prognostic values of lncRNA SNHG8, SOCS3 and ICAM1 were evaluated using a receiver operating characteristic (ROC) curve. RESULTS An AMI lncRNA-miRNA-mRNA network was constructed that included two mRNAs, one miRNA and one lncRNA. After RT-PCR validation of lncRNA SNHG8, SOCS3 and ICAM1 between the AMI and normal samples, only lncRNA SNHG8 had significant diagnostic value for further analysis. The ROC curve showed that SNHG8 presented an AUC of 0.850, while the AUC of SOCS3 was 0.633 and that of ICAM1 was 0.594. After a pairwise comparison, we found that SNHG8 was statistically significant (P SNHG8-ICAM1 = 0.002; P SNHG8-SOCS3 = 0.031). The results of a functional enrichment analysis of the interacting genes and microRNAs showed that the shared lncRNA SNHG8 may be a new factor in AMI. CONCLUSIONS Our investigation of the lncRNA-miRNA-mRNA regulatory networks in AMI revealed a novel lncRNA, lncRNA SNHG8, as a risk factor for AMI and expanded our understanding of the mechanisms involved in the pathogenesis of AMI.
Collapse
Affiliation(s)
- Liu-An Zhuo
- Department of Cardiology, Institute of Cardiovascular Diseases, the Liu Zhou People's Hospital, 8 Wenchang Road, Liuzhou, 545006, Guangxi, China
| | - Yi-Tao Wen
- Department of Cardiology, Institute of Cardiovascular Diseases, the Liu Zhou People's Hospital, 8 Wenchang Road, Liuzhou, 545006, Guangxi, China
| | - Yong Wang
- Department of Cardiology, Institute of Cardiovascular Diseases, the Liu Zhou People's Hospital, 8 Wenchang Road, Liuzhou, 545006, Guangxi, China
| | - Zhi-Fang Liang
- Department of Cardiology, Institute of Cardiovascular Diseases, the Liu Zhou People's Hospital, 8 Wenchang Road, Liuzhou, 545006, Guangxi, China
| | - Gang Wu
- Department of Cardiology, Institute of Cardiovascular Diseases, the Liu Zhou People's Hospital, 8 Wenchang Road, Liuzhou, 545006, Guangxi, China
| | - Mei-Dan Nong
- Department of Cardiology, Institute of Cardiovascular Diseases, the Liu Zhou People's Hospital, 8 Wenchang Road, Liuzhou, 545006, Guangxi, China
| | - Liu Miao
- Department of Cardiology, Institute of Cardiovascular Diseases, the Liu Zhou People's Hospital, 8 Wenchang Road, Liuzhou, 545006, Guangxi, China.
| |
Collapse
|
28
|
Let-7f: A New Potential Circulating Biomarker Identified by miRNA Profiling of Cells Isolated from Human Abdominal Aortic Aneurysm. Int J Mol Sci 2019; 20:ijms20215499. [PMID: 31694153 PMCID: PMC6862487 DOI: 10.3390/ijms20215499] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/29/2019] [Accepted: 10/30/2019] [Indexed: 01/28/2023] Open
Abstract
Abdominal aortic aneurysm (AAA) is a progressive vascular disease responsible for 1–4% of the deaths in elderly men. This study aimed to characterize specific microRNA (miRNA) expression in aneurysmal smooth muscle cells (SMCs) and macrophages in order to identify circulating miRNAs associated with AAA. We screened 850 miRNAs in aneurysmal SMCs, M1 and M2 macrophages, and in control SMCs isolated by micro-dissection from aortic biopsies using microarray analysis. In all, 92 miRNAs were detected and 10 miRNAs were selected for validation by qRT-PCR in isolated cells (n = 5), whole control and aneurysmal aorta biopsies (n = 13), and plasma from patients (n = 24) undergoing AAA (over 50 mm) repair matched to patients (n = 18) with peripheral arterial disease (PAD) with atherosclerosis but not AAA. Seven miRNAs were modulated similarly in all aneurysmal cells. The Let-7f was downregulated in aneurysmal cells compared to control SMCs with a significant lower expression in M1 compared to M2 macrophages (0.1 fold, p = 0.03), correlated with a significant downregulation in whole aneurysmal aorta compared to control aorta (0.2 fold, p = 0.03). Significant levels of circulating let-7f (p = 0.048) were found in AAA patients compared to PAD patients with no significant correlation with aortic diameter (R2 = 0.03). Our study underlines the utility of profiling isolated aneurysmal cells to identify other miRNAs for which the modulation of expression might be masked when the whole aorta is used. The results highlight let-7f as a new potential biomarker for AAA.
Collapse
|
29
|
Lareyre F, Clément M, Moratal C, Loyer X, Jean-Baptiste E, Hassen-Khodja R, Chinetti G, Mallat Z, Raffort J. Differential micro-RNA expression in diabetic patients with abdominal aortic aneurysm. Biochimie 2019; 162:1-7. [DOI: 10.1016/j.biochi.2019.03.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 03/18/2019] [Indexed: 12/29/2022]
|
30
|
Comparison of Cardiac miRNA Transcriptomes Induced by Diabetes and Rapamycin Treatment and Identification of a Rapamycin-Associated Cardiac MicroRNA Signature. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:8364608. [PMID: 30647817 PMCID: PMC6311877 DOI: 10.1155/2018/8364608] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 08/16/2018] [Accepted: 08/29/2018] [Indexed: 02/07/2023]
Abstract
Rapamycin (Rap), an inhibitor of mTORC1, reduces obesity and improves lifespan in mice. However, hyperglycemia and lipid disorders are adverse side effects in patients receiving Rap treatment. We previously reported that diabetes induces pansuppression of cardiac cytokines in Zucker obese rats (ZO-C). Rap treatment (750 μg/kg/day for 12 weeks) reduced their obesity and cardiac fibrosis significantly; however, it increased their hyperglycemia and did not improve their cardiac diastolic parameters. Moreover, Rap treatment of healthy Zucker lean rats (ZL-C) induced cardiac fibrosis. Rap-induced changes in ZL-C's cardiac cytokine profile shared similarities with that of diabetes-induced ZO-C. Therefore, we hypothesized that the cardiac microRNA transcriptome induced by diabetes and Rap treatment could share similarities. Here, we compared the cardiac miRNA transcriptome of ZL-C to ZO-C, Rap-treated ZL (ZL-Rap), and ZO (ZO-Rap). We report that 80% of diabetes-induced miRNA transcriptome (40 differentially expressed miRNAs by minimum 1.5-fold in ZO-C versus ZL-C; p ≤ 0.05) is similar to 47% of Rap-induced miRNA transcriptome in ZL (68 differentially expressed miRNAs by minimum 1.5-fold in ZL-Rap versus ZL-C; p ≤ 0.05). This remarkable similarity between diabetes-induced and Rap-induced cardiac microRNA transcriptome underscores the role of miRNAs in Rap-induced insulin resistance. We also show that Rap treatment altered the expression of the same 17 miRNAs in ZL and ZO hearts indicating that these 17 miRNAs comprise a unique Rap-induced cardiac miRNA signature. Interestingly, only four miRNAs were significantly differentially expressed between ZO-C and ZO-Rap, indicating that, unlike the nondiabetic heart, Rap did not substantially change the miRNA transcriptome in the diabetic heart. In silico analyses showed that (a) mRNA-miRNA interactions exist between differentially expressed cardiac cytokines and miRNAs, (b) human orthologs of rat miRNAs that are strongly correlated with cardiac fibrosis may modulate profibrotic TGF-β signaling, and (c) changes in miRNA transcriptome caused by diabetes or Rap treatment include cardioprotective miRNAs indicating a concurrent activation of an adaptive mechanism to protect the heart in conditions that exacerbate diabetes.
Collapse
|
31
|
Johnson JL. Elucidating the contributory role of microRNA to cardiovascular diseases (a review). Vascul Pharmacol 2018; 114:31-48. [PMID: 30389614 PMCID: PMC6445803 DOI: 10.1016/j.vph.2018.10.010] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 07/13/2018] [Accepted: 10/28/2018] [Indexed: 02/07/2023]
Abstract
Cardiovascular diseases encompassing atherosclerosis, aortic aneurysms, restenosis, and pulmonary arterial hypertension, remain the leading cause of morbidity and mortality worldwide. In response to a range of stimuli, the dynamic interplay between biochemical and biomechanical mechanisms affect the behaviour and function of multiple cell types, driving the development and progression of cardiovascular diseases. Accumulating evidence has highlighted microRNAs (miRs) as significant regulators and micro-managers of key cellular and molecular pathophysiological processes involved in predominant cardiovascular diseases, including cell mitosis, motility and viability, lipid metabolism, generation of inflammatory mediators, and dysregulated proteolysis. Human pathological and clinical studies have aimed to identify select microRNA which may serve as biomarkers of disease and their progression, which are discussed within this review. In addition, I provide comprehensive coverage of in vivo investigations elucidating the modulation of distinct microRNA on the pathophysiology of atherosclerosis, abdominal aortic aneurysms, restenosis, and pulmonary arterial hypertension. Collectively, clinical and animal studies have begun to unravel the complex and often diverse effects microRNAs and their targets impart during the development of cardiovascular diseases and revealed promising therapeutic strategies through which modulation of microRNA function may be applied clinically.
Collapse
Affiliation(s)
- Jason L Johnson
- Laboratory of Cardiovascular Pathology, Bristol Medical School, University of Bristol, UK.
| |
Collapse
|
32
|
Sun L, Li W, Lei F, Li X. The regulatory role of microRNAs in angiogenesis-related diseases. J Cell Mol Med 2018; 22:4568-4587. [PMID: 29956461 PMCID: PMC6156236 DOI: 10.1111/jcmm.13700] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 04/17/2018] [Indexed: 02/06/2023] Open
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs that regulate gene expression at a post-transcriptional level via either the degradation or translational repression of a target mRNA. They play an irreplaceable role in angiogenesis by regulating the proliferation, differentiation, apoptosis, migration and tube formation of angiogenesis-related cells, which are indispensable for multitudinous physiological and pathological processes, especially for the occurrence and development of vascular diseases. Imbalance between the regulation of miRNAs and angiogenesis may cause many diseases such as cancer, cardiovascular disease, aneurysm, Kawasaki disease, aortic dissection, phlebothrombosis and diabetic microvascular complication. Therefore, it is important to explore the essential role of miRNAs in angiogenesis, which might help to uncover new and effective therapeutic strategies for vascular diseases. This review focuses on the interactions between miRNAs and angiogenesis, and miRNA-based biomarkers in the diagnosis, treatment and prognosis of angiogenesis-related diseases, providing an update on the understanding of the clinical value of miRNAs in targeting angiogenesis.
Collapse
Affiliation(s)
- Li‐Li Sun
- Department of Vascular Surgerythe Affiliated Drum Tower HospitalNanjing University Medical SchoolNanjingChina
- Department of Vascular Surgerythe Second Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Wen‐Dong Li
- Department of Vascular Surgerythe Affiliated Drum Tower HospitalNanjing University Medical SchoolNanjingChina
| | - Feng‐Rui Lei
- Department of Vascular Surgerythe Second Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Xiao‐Qiang Li
- Department of Vascular Surgerythe Affiliated Drum Tower HospitalNanjing University Medical SchoolNanjingChina
| |
Collapse
|
33
|
Lopez S, Bermudez B, Montserrat-de la Paz S, Abia R, Muriana FJ. A microRNA expression signature of the postprandial state in response to a high-saturated-fat challenge. J Nutr Biochem 2018; 57:45-55. [DOI: 10.1016/j.jnutbio.2018.03.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 01/23/2018] [Accepted: 03/07/2018] [Indexed: 12/13/2022]
|
34
|
Tenorio EJR, Braga AFF, Tirapelli DPDC, Ribeiro MS, Piccinato CE, Joviliano EE. Expression in Whole Blood Samples of miRNA-191 and miRNA-455-3p in Patients with AAA and Their Relationship to Clinical Outcomes after Endovascular Repair. Ann Vasc Surg 2018. [DOI: 10.1016/j.avsg.2018.01.086] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
35
|
Plana E, Gálvez L, Medina P, Navarro S, Miralles M. Estudio de selección de microRNA como posibles biomarcadores de aneurisma de aorta abdominal. ANGIOLOGIA 2018. [DOI: 10.1016/j.angio.2017.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
36
|
Abstract
PURPOSE OF REVIEW In this paper, we review the progress made thus far in research related to the genetics of peripheral arterial disease (PAD) by detailing efforts to date in heritability, linkage analyses, and candidate gene studies. We further summarize more contemporary genome-wide association studies (GWAS) and epigenetic studies of PAD. Finally, we review current challenges and future avenues of advanced research in PAD genetics including whole genome sequencing studies. RECENT FINDINGS Studies have estimated the heritability of PAD to be moderate, though the contribution to this heritability that is independent of traditional cardiovascular risk factors remains unclear. Recent efforts have identified SNPs associated with PAD in GWAS analyses, but these have yet to be replicated in independent studies. Much remains to be discovered in the field of PAD genetics. An improved understanding of the genetic foundation for PAD will allow for earlier diagnosis of disease and a more complete pathophysiological understanding of the mechanisms of the disease leading to novel therapeutic interventions. Future avenues for success will likely arise from very large-scale GWAS, whole genome sequencing, and epigenetic studies involving very well-characterized cohorts.
Collapse
Affiliation(s)
- Nathan Belkin
- Division of Vascular and Endovascular Surgery, Hospital of the University of Pennsylvania, 3400 Spruce Street, 4 Maloney, Philadelphia, PA, 19104, USA
| | - Scott M Damrauer
- Division of Vascular and Endovascular Surgery, Hospital of the University of Pennsylvania, 3400 Spruce Street, 4 Maloney, Philadelphia, PA, 19104, USA. .,Department of Surgery, Corporal Michael Crescenz VA Medical Center, 3900 Woodland Ave., Philadelphia, PA, 19104, USA.
| |
Collapse
|
37
|
Chan CYT, Cheng SWK. Elevated homocysteine in human abdominal aortic aneurysmal tissues. Vasc Med 2017; 22:370-377. [DOI: 10.1177/1358863x17718260] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
An abnormally high level of homocysteine (Hcy) has been consistently observed in the blood of abdominal aortic aneurysm (AAA) patients. However, the expression of Hcy in human AAA tissues has not been investigated. In this study, the expression of Hcy in aneurysmal tissues from AAA patients ( n=30) was compared with non-aneurysmal tissues from organ donors ( n=31) by dot blotting and immunohistochemistry. A significantly higher expression of Hcy was observed in AAA than control tissues ( p<0.001). Furthermore, the associations of methylenetetrahydrofolate reductase (MTHFR) C677T polymorphism, detected by polymerase chain reaction-restriction fragment length polymorphism, with both AAA and tissue Hcy expression were evaluated. Our results showed MTHFR C677T polymorphism was not significantly associated with AAA or tissue Hcy expression. Lastly, the expression of Hcy in vascular smooth muscle cells (VSMCs), which were isolated from human aortic tissues by explant culture, and their release to cultured media was investigated by dot blotting. The AAA VSMCs expressed and released a significantly higher level of Hcy than the control VSMCs ( p<0.001). In summary, our novel findings showed Hcy expression was abnormally elevated in human AAA tissues, which may not be dependent on MTHFR C677T polymorphism.
Collapse
Affiliation(s)
- Crystal Yin Tung Chan
- Division of Vascular Surgery, Department of Surgery, University of Hong Kong Medical Centre, Queen Mary Hospital, Pokfulam, Hong Kong
| | - Stephen Wing Keung Cheng
- Division of Vascular Surgery, Department of Surgery, University of Hong Kong Medical Centre, Queen Mary Hospital, Pokfulam, Hong Kong
| |
Collapse
|
38
|
Cao X, Cai Z, Liu J, Zhao Y, Wang X, Li X, Xia H. miRNA‑504 inhibits p53‑dependent vascular smooth muscle cell apoptosis and may prevent aneurysm formation. Mol Med Rep 2017; 16:2570-2578. [PMID: 28677789 PMCID: PMC5548046 DOI: 10.3892/mmr.2017.6873] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 03/23/2017] [Indexed: 12/11/2022] Open
Abstract
Abdominal aortic aneurysm (AAA) is a common disease that is associated with the proliferation and apoptosis of vascular smooth muscle cells (VSMCs). VSMCs are regulated by microRNAs (miRNA). The aim of the present study was to identify miRNA sequences that regulate aortic SMCs during AAA. miRNA-504 was identified using a miRNA PCR array and by reverse transcription-quantitative polymerase chain reaction analysis, and its expression levels were observed to be downregulated in the aortic cells derived from patients with AAA when compared with controls. Transfection of SMCs with pMSCV-miRNA-504 vector was performed, and cell proliferation and the expression levels of proliferating cell nuclear antigen (PCNA), replication factor C subunit 4 (RFC4), B-cell lymphoma-2 (Bcl-2) and caspase-3/9 were measured by western blotting. The mechanisms underlying the effects of miRNA-504 was then analyzed. The results demonstrated that overexpression of miRNA-504 significantly upregulated the expression levels of PCNA, RFC4 and Bcl-2, while caspase-3/9 expression was significantly inhibited when compared with non-targeting controls. In addition, miRNA-504 overexpression was observed to promote the proliferation of SMCs. The expression level of the tumor suppressor, p53, which is known to be a direct target of miRNA-504, was inhibited following transfection of SMCs with pMSCV-miRNA-504. In addition, the expression of the downstream targets of p53, p21 and Bcl-like protein-4, were significantly reduced following overexpression of miRNA-504. These results revealed the anti-apoptotic role of miRNA-504 in SMCs derived from patients with AAA via direct targeting of p53.
Collapse
Affiliation(s)
- Xue Cao
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150006, P.R. China
| | - Zhenguo Cai
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150006, P.R. China
| | - Junyan Liu
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150006, P.R. China
| | - Yanru Zhao
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150006, P.R. China
| | - Xin Wang
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150006, P.R. China
| | - Xueqi Li
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150006, P.R. China
| | - Hongyuan Xia
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150006, P.R. China
| |
Collapse
|
39
|
Iyer V, Rowbotham S, Biros E, Bingley J, Golledge J. A systematic review investigating the association of microRNAs with human abdominal aortic aneurysms. Atherosclerosis 2017; 261:78-89. [DOI: 10.1016/j.atherosclerosis.2017.03.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 03/02/2017] [Accepted: 03/06/2017] [Indexed: 12/24/2022]
|
40
|
Aquila G, Fortini C, Pannuti A, Delbue S, Pannella M, Morelli MB, Caliceti C, Castriota F, de Mattei M, Ongaro A, Pellati A, Ferrante P, Miele L, Tavazzi L, Ferrari R, Rizzo P, Cremonesi A. Distinct gene expression profiles associated with Notch ligands Delta-like 4 and Jagged1 in plaque material from peripheral artery disease patients: a pilot study. J Transl Med 2017; 15:98. [PMID: 28472949 PMCID: PMC5418727 DOI: 10.1186/s12967-017-1199-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 04/25/2017] [Indexed: 12/13/2022] Open
Abstract
Background The lack of early diagnosis, progression markers and effective pharmacological treatment has dramatic unfavourable effects on clinical outcomes in patients with peripheral artery disease (PAD). Addressing these issues will require dissecting the molecular mechanisms underlying this disease. We sought to characterize the Notch signaling and atherosclerosis relevant markers in lesions from femoral arteries of symptomatic PAD patients. Methods Plaque material from the common femoral, superficial femoral or popliteal arteries of 20 patients was removed by directional atherectomy. RNA was obtained from 9 out of 20 samples and analysed by quantitative reverse transcriptase-polymerase chain reaction (qRT-PCR). Results We detected expression of Notch ligands Delta-like 4 (Dll4) and Jagged1 (Jag1), of Notch target genes Hes1, Hey1, Hey2, HeyL and of markers of plaque inflammation and stability such as vascular cell adhesion molecule 1 (VCAM1), smooth muscle 22 (SM22), cyclooxygenase 2 (COX2), Bcl2, CD68 and miRNAs 21-5p, 125a-5p, 126-5p,146-5p, 155-5p, 424-5p. We found an “inflamed plaque” gene expression profile characterized by high Dll4 associated to medium/high CD68, COX2, VCAM1, Hes1, miR126-5p, miR146a-5p, miR155-5p, miR424-5p and low Jag1, SM22, Bcl2, Hey2, HeyL, miR125a-5p (2/9 patients) and a “stable plaque” profile characterized by high Jag1 associated to medium/high Hey2, HeyL, SM22, Bcl2, miR125a and low Dll4, CD68, COX2, VCAM1, miR126-5p, miR146a-5p, miR155-5p, miR424-5p (3/9 patients). The remaining patients (4/9) showed a plaque profile with intermediate characteristics. Conclusions This study reveals the existence of a gene signature associated to Notch activation by specific ligands that could be predictive of PAD progression. Electronic supplementary material The online version of this article (doi:10.1186/s12967-017-1199-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Giorgio Aquila
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Cinzia Fortini
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Antonio Pannuti
- Department of Genetics and Stanley Scott Cancer Center, Louisiana State University Health Sciences Center and Louisiana Cancer Research Consortium, New Orleans, LA, USA
| | - Serena Delbue
- Department of Biomedical, Surgical and Dental SciencesUniversity of Milan, Milan, Italy
| | - Micaela Pannella
- GoldyneSavad Institute of Gene Therapy, Hadassah-Hebrew University Medical Center, 91120, Jerusalem, Israel
| | | | - Cristiana Caliceti
- Department of Chemistry "G. Ciamician", University of Bologna, Bologna, Italy
| | - Fausto Castriota
- Maria Cecilia Hospital, GVM Care & Research, E.S. Health Science Foundation, Cotignola, Italy
| | - Monica de Mattei
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Via Fossato di Mortara 64/B, 44121, Ferrara, Italy
| | - Alessia Ongaro
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Via Fossato di Mortara 64/B, 44121, Ferrara, Italy
| | - Agnese Pellati
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Via Fossato di Mortara 64/B, 44121, Ferrara, Italy
| | - Pasquale Ferrante
- Department of Biomedical, Surgical and Dental SciencesUniversity of Milan, Milan, Italy
| | - Lucio Miele
- Department of Genetics and Stanley Scott Cancer Center, Louisiana State University Health Sciences Center and Louisiana Cancer Research Consortium, New Orleans, LA, USA
| | - Luigi Tavazzi
- Maria Cecilia Hospital, GVM Care & Research, E.S. Health Science Foundation, Cotignola, Italy
| | - Roberto Ferrari
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Paola Rizzo
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Via Fossato di Mortara 64/B, 44121, Ferrara, Italy.
| | - Alberto Cremonesi
- Maria Cecilia Hospital, GVM Care & Research, E.S. Health Science Foundation, Cotignola, Italy
| |
Collapse
|
41
|
Meeuwsen JAL, van ´t Hof FNG, van Rheenen W, Rinkel GJE, Veldink JH, Ruigrok YM. Circulating microRNAs in patients with intracranial aneurysms. PLoS One 2017; 12:e0176558. [PMID: 28459827 PMCID: PMC5411042 DOI: 10.1371/journal.pone.0176558] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 04/12/2017] [Indexed: 12/31/2022] Open
Abstract
Introduction We compared circulating microRNA (miRNA) levels in plasma of patients with intracranial aneurysms (IA) to those of controls as a first step towards finding potential biomarkers for individuals at high risk of IA development and its subsequent rupture. Patients and methods Using a PCR array we measured 370 miRNAs in plasma of 15 patients with prior aneurysmal subarachnoid hemorrhage (aSAH), of whom 11 had an additional unruptured IA (UIA), and of 15 controls. MiRNAs with a difference in levels with an absolute fold change (FC) > 1.2 and p<0.01 were further tested using real-time (RT) PCR in an additional independent set of 15 aSAH patients, 15 untreated UIA patients and 15 controls for replication (absolute FC >1.2 and p<0.05). We used receiver operating characteristic (ROC) curves to illustrate the diagnostic potential of these miRNAs. Results Three of five miRNAs with a difference in levels in the PCR array study were replicated with miRNA-183-5p decreased in all patients (FC = -2.2, p = 1.7x10-3), miRNA-200a-3p increased in aSAH patients (FC = 1.8, p = 2.8x10-2) and miRNA-let7b-5p decreased in UIA patients (FC = -1.7, p = 1.27x10-3) as compared to controls. In distinguishing aSAH patients from controls, the area under the ROC curve (AUC) was 0.80 (95% confidence interval (95% CI) 0.63–0.97) for miRNA-183-5p, and 0.74 (95% CI 0.55–0.94) for miRNA-200a-3p. In distinguishing untreated UIA patients from controls, AUC was 0.83 (95% CI 0.69–0.98) for miRNA-183-5p and 0.92 (95% CI 0.81–1) for miRNA-let-7b. Discussion/Conclusions We identified three specific circulating miRNAs that are able to discriminate between IA patients and controls. Follow-up studies should assess if these miRNAs may be used biomarkers for identifying individuals at high risk of IA development and its subsequent rupture.
Collapse
Affiliation(s)
- John A. L. Meeuwsen
- Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, The Netherlands
| | - Femke N. G. van ´t Hof
- Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, The Netherlands
| | - Wouter van Rheenen
- Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, The Netherlands
| | - Gabriel J. E. Rinkel
- Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, The Netherlands
| | - Jan H. Veldink
- Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, The Netherlands
| | - Ynte M. Ruigrok
- Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, The Netherlands
- * E-mail:
| |
Collapse
|
42
|
Wang X, Suofu Y, Akpinar B, Baranov SV, Kim J, Carlisle DL, Zhang Y, Friedlander RM. Systemic antimiR-337-3p delivery inhibits cerebral ischemia-mediated injury. Neurobiol Dis 2017; 105:156-163. [PMID: 28461247 DOI: 10.1016/j.nbd.2017.04.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 04/20/2017] [Accepted: 04/27/2017] [Indexed: 11/15/2022] Open
Abstract
Modulation of miRNA expression has been shown to be beneficial in the context of multiple diseases. The purpose of this study was to determine if an inhibitor of miR-337-3p is neuroprotective for hypoxic injury after tail vein injection. We evaluated miR-337-3p expression levels and in brain tissue in vivo before and after permanent middle cerebral artery occlusion (pMCAO) in mice. Subsequently, a custom locked nucleic acid (LNA) antimir-337-3p oligonucleotide was developed and tested in vitro after induction of oxygen glucose-deprivation (OGD) and in vivo by injection into the mouse tail vein for 3 consecutive days before pMCAO. Ischemic lesion volume was measured by TTC staining. We show that systemically administered LNA antimir-337-3p crosses the blood brain-brain-barrier (BBB), penetrates into neurosn, downregulates endogenous miR-337-3p expression and reduces ischemic brain injury. The findings support the use of similar antimir-LNA constructs as novel therapies in neurological disease.
Collapse
Affiliation(s)
- Xiaomin Wang
- Department of Neurological Surgery, Neuroapoptosis Laboratory, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, United States
| | - Yalikun Suofu
- Department of Neurological Surgery, Neuroapoptosis Laboratory, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, United States
| | - Berkcan Akpinar
- Department of Neurological Surgery, Neuroapoptosis Laboratory, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, United States
| | - Sergei V Baranov
- Department of Neurological Surgery, Neuroapoptosis Laboratory, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, United States
| | - Jinho Kim
- Department of Neurological Surgery, Neuroapoptosis Laboratory, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, United States
| | - Diane L Carlisle
- Department of Neurological Surgery, Neuroapoptosis Laboratory, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, United States
| | - Yu Zhang
- Department of Neurological Surgery, Neuroapoptosis Laboratory, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, United States.
| | - Robert M Friedlander
- Department of Neurological Surgery, Neuroapoptosis Laboratory, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, United States.
| |
Collapse
|
43
|
Circulating microRNAs signature correlates with positive [ 18F]fluorodeoxyglucose-positron emission tomography in patients with abdominal aortic aneurysm. J Vasc Surg 2017; 67:585-595.e3. [PMID: 28431866 DOI: 10.1016/j.jvs.2016.12.112] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 12/12/2016] [Indexed: 12/12/2022]
Abstract
BACKGROUND Prediction of abdominal aortic aneurysm (AAA) rupture is a challenging issue. Small noncoding microRNAs (miRNAs) are potent regulators of gene expression and are considered as valuable circulating biomarkers. Recently, [18F]fluorodeoxyglucose (FDG) uptake detected by positron emission tomography (PET) in AAA was correlated with cellular and molecular alterations involved in wall instability and its potential rupture. Our study aimed at identifying circulating miRNAs correlated with a positive PET that could help discriminate patients at high risk of rupture. METHODS The level of 372 miRNAs was evaluated by polymerase chain reaction array in plasma from 35 AAA patients displaying no FDG uptake (A0) and 22 patients with a positive PET uptake (A+). The modulated miRNAs were validated by quantitative polymerase chain reaction and measured in aneurysmal tissues from both groups of patients. RESULTS Six circulating miRNAs were found significantly modulated in A+ vs A0 patients. They were significantly correlated not only between them but also with the intensity of FDG uptake. Two of them correlated also with the AAA diameter. These miRNAs displayed significant discriminating power between the A+ and A0 groups as determined by receiver operating characteristic curves. Three downregulated circulating miRNAs (miR-99b-5p, miR-125b-5p, and miR-204-5p) were also significantly reduced in the aneurysmal tissue, specifically in the FDG-uptake site, compared with a negative zone in the same aneurysm and with A0 aneurysms. They were further significantly inversely correlated with the expression, at the positive uptake site, of some of their potential gene targets, most notably matrix metalloproteinase 13. CONCLUSIONS Six miRNAs were identified as potential new circulating biomarkers of PET+ AAA. Three of these were similarly modulated in the metabolically active aneurysmal wall and might be directly involved in AAA instability.
Collapse
|
44
|
Genetic and Epigenetic Regulation of Aortic Aneurysms. BIOMED RESEARCH INTERNATIONAL 2017; 2017:7268521. [PMID: 28116311 PMCID: PMC5237727 DOI: 10.1155/2017/7268521] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 12/15/2016] [Indexed: 02/07/2023]
Abstract
Aneurysms are characterized by structural deterioration of the vascular wall leading to progressive dilatation and, potentially, rupture of the aorta. While aortic aneurysms often remain clinically silent, the morbidity and mortality associated with aneurysm expansion and rupture are considerable. Over 13,000 deaths annually in the United States are attributable to aortic aneurysm rupture with less than 1 in 3 persons with aortic aneurysm rupture surviving to surgical intervention. Environmental and epidemiologic risk factors including smoking, male gender, hypertension, older age, dyslipidemia, atherosclerosis, and family history are highly associated with abdominal aortic aneurysms, while heritable genetic mutations are commonly associated with aneurysms of the thoracic aorta. Similar to other forms of cardiovascular disease, family history, genetic variation, and heritable mutations modify the risk of aortic aneurysm formation and provide mechanistic insight into the pathogenesis of human aortic aneurysms. This review will examine the relationship between heritable genetic and epigenetic influences on thoracic and abdominal aortic aneurysm formation and rupture.
Collapse
|
45
|
Carvalho LSF. Can microRNAs improve prediction of abdominal aortic aneurysm growth? Atherosclerosis 2016; 256:131-133. [PMID: 28007302 DOI: 10.1016/j.atherosclerosis.2016.12.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 12/01/2016] [Indexed: 01/20/2023]
Affiliation(s)
- Luiz Sérgio F Carvalho
- Laboratory of Atherosclerosis and Vascular Biology, State University of Campinas, (UNICAMP), Campinas, SP, Brazil; Escola Superior de Ciências da Saúde, Brasília, DF, Brazil.
| |
Collapse
|
46
|
Wanhainen A, Mani K, Vorkapic E, De Basso R, Björck M, Länne T, Wågsäter D. Screening of circulating microRNA biomarkers for prevalence of abdominal aortic aneurysm and aneurysm growth. Atherosclerosis 2016; 256:82-88. [PMID: 27993388 DOI: 10.1016/j.atherosclerosis.2016.11.007] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 11/04/2016] [Accepted: 11/08/2016] [Indexed: 10/20/2022]
Abstract
BACKGROUND AND AIMS MicroRNA (miR) are important regulators of gene expression and biological processes and have recently been suggested as possible biomarkers for abdominal aortic aneurysm (AAA) disease. The aim of the present study was to assess the role of miR as biomarkers for initiation and progression of AAA disease, through evaluation of a wide range of miRs in a large population-based cohort, with AAA patients with linked clinical data regarding risk factors, AAA size and growth, as well as controls. METHODS The expression of the 172 most commonly expressed miRs in plasma was analyzed by real-time PCR in samples from 169 screening-detected AAA patients and 48 age-matched controls. RESULTS For 103 miRs, there was a significant difference in expression between AAA and controls. Of these, 20 miRs were differently expressed between fast and slow growing aneurysms. These miRs target genes known to be involved in AAA disease as well as novel genes and pathways. By combining the top altered miRs together with clinical variables, strong predictive values, determining growth of AAA, were obtained (area under curve = 0.86, p < 0.001). CONCLUSIONS This large cohort study identified several novel miRs with altered expression in AAA patients when compared to controls. Assessment of miR expression may offer an opportunity to predict disease progression and aneurysm growth.
Collapse
Affiliation(s)
- Anders Wanhainen
- Department of Surgical Sciences, Section of Vascular Surgery, Uppsala University, Uppsala, Sweden
| | - Kevin Mani
- Department of Surgical Sciences, Section of Vascular Surgery, Uppsala University, Uppsala, Sweden
| | - Emina Vorkapic
- Division of Drug Research, Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| | - Rachel De Basso
- Division of Medical Diagnostics, Department of Clinical Physiology, Region Jönköping County, Jönköping, Sweden; Department of Natural Science and Biomedicine, School of Health and Welfare, Jönköping University, Jönköping, Sweden
| | - Martin Björck
- Department of Surgical Sciences, Section of Vascular Surgery, Uppsala University, Uppsala, Sweden
| | - Toste Länne
- Division of Cardiovascular Medicine, Department of Medical and Health Sciences, Faculty of Health Sciences, Linköping University, Linköping, Sweden
| | - Dick Wågsäter
- Division of Drug Research, Department of Medical and Health Sciences, Linköping University, Linköping, Sweden.
| |
Collapse
|
47
|
Abstract
The term epigenetics is usually used to describe inheritable changes in gene function which do not involve changes in the DNA sequence. These typically include non-coding RNAs, DNA methylation and histone modifications. Smoking and older age are recognised risk factors for peripheral artery diseases, such as occlusive lower limb artery disease and abdominal aortic aneurysm, and have been implicated in promoting epigenetic changes. This brief review describes studies that have associated epigenetic factors with peripheral artery diseases and investigations which have examined the effect of epigenetic modifications on the outcome of peripheral artery diseases in mouse models. Investigations have largely focused on microRNAs and have identified a number of circulating microRNAs associated with human peripheral artery diseases. Upregulating or antagonising a number of microRNAs has also been reported to limit aortic aneurysm development and hind limb ischemia in mouse models. The importance of DNA methylation and histone modifications in peripheral artery disease has been relatively little studied. Whether circulating microRNAs can be used to assist identification of patients with peripheral artery diseases and be modified in order to improve the outcome of peripheral artery disease will require further investigation.
Collapse
Affiliation(s)
- Jonathan Golledge
- The Vascular Biology Unit, Queensland Research Centre for Peripheral Vascular Disease, College of Medicine & Dentistry, James Cook University, Townsville, QLD, 4811, Australia.
- Department of Vascular and Endovascular Surgery, The Townsville Hospital, Townsville, QLD, Australia.
| | - Erik Biros
- The Vascular Biology Unit, Queensland Research Centre for Peripheral Vascular Disease, College of Medicine & Dentistry, James Cook University, Townsville, QLD, 4811, Australia
| | - John Bingley
- Vascular Surgery Unit, Mater Hospital Brisbane, South Brisbane, QLD, Australia
- Department of Surgery, University of Queensland, Brisbane, Australia
| | - Vikram Iyer
- The Vascular Biology Unit, Queensland Research Centre for Peripheral Vascular Disease, College of Medicine & Dentistry, James Cook University, Townsville, QLD, 4811, Australia
- Department of Vascular and Endovascular Surgery, The Townsville Hospital, Townsville, QLD, Australia
- Department of Surgery, University of Queensland, Brisbane, Australia
| | - Smriti M Krishna
- The Vascular Biology Unit, Queensland Research Centre for Peripheral Vascular Disease, College of Medicine & Dentistry, James Cook University, Townsville, QLD, 4811, Australia
| |
Collapse
|
48
|
Busch A, Eken SM, Maegdefessel L. Prospective and therapeutic screening value of non-coding RNA as biomarkers in cardiovascular disease. ANNALS OF TRANSLATIONAL MEDICINE 2016; 4:236. [PMID: 27429962 DOI: 10.21037/atm.2016.06.06] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Non-coding RNA (ncRNA) is a class of genetic, epigenetic and translational regulators, containing short and long transcripts with intriguing abilities for use as biomarkers due to their superordinate role in disease development. In the past five years many of these have been investigated in cardiovascular diseases (CVD), mainly myocardial infarction (MI) and heart failure. To extend this view, we summarize the existing data about ncRNA as biomarker in the whole entity of CVDs by literature-based review and comparison of the identified candidates. The myomirs miRNA-1, -133a/b, -208a, -499 with well-defined cellular functions have proven equal to classic protein biomarkers for disease detection in MI. Other microRNAs (miRNAs) were reproducibly found to correlate with disease, disease severity and outcome in heart failure, stroke, coronary artery disease (CAD) and aortic aneurysm. An additional utilization has been discovered for therapeutic monitoring. The function of long non-coding transcripts is only about to be unraveled, yet shows great potential for outcome prediction. ncRNA biomarkers have a distinct role if no alternative test is available or has is performing poorly. With increasing mechanistic understanding, circulating miRNA and long non-coding transcripts will provide useful disease information with high predictive power.
Collapse
Affiliation(s)
- Albert Busch
- Cardiovascular Medicine Unit, Department of Medicine, Karolinska Institute, Center for Molecular Medicine, Stockholm, Sweden
| | - Suzanne M Eken
- Cardiovascular Medicine Unit, Department of Medicine, Karolinska Institute, Center for Molecular Medicine, Stockholm, Sweden
| | - Lars Maegdefessel
- Cardiovascular Medicine Unit, Department of Medicine, Karolinska Institute, Center for Molecular Medicine, Stockholm, Sweden
| |
Collapse
|
49
|
The Discovery of Novel Genomic, Transcriptomic, and Proteomic Biomarkers in Cardiovascular and Peripheral Vascular Disease: The State of the Art. BIOMED RESEARCH INTERNATIONAL 2016; 2016:7829174. [PMID: 27298828 PMCID: PMC4889798 DOI: 10.1155/2016/7829174] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 04/26/2016] [Accepted: 05/05/2016] [Indexed: 12/14/2022]
Abstract
Cardiovascular disease (CD) and peripheral vascular disease (PVD) are leading causes of mortality and morbidity in western countries and also responsible of a huge burden in terms of disability, functional decline, and healthcare costs. Biomarkers are measurable biological elements that reflect particular physiological or pathological states or predisposition towards diseases and they are currently widely studied in medicine and especially in CD. In this context, biomarkers can also be used to assess the severity or the evolution of several diseases, as well as the effectiveness of particular therapies. Genomics, transcriptomics, and proteomics have opened new windows on disease phenomena and may permit in the next future an effective development of novel diagnostic and prognostic medicine in order to better prevent or treat CD. This review will consider the current evidence of novel biomarkers with clear implications in the improvement of risk assessment, prevention strategies, and medical decision making in the field of CD.
Collapse
|
50
|
Raffort J, Lareyre F, Clement M, Mallat Z. Micro-RNAs in abdominal aortic aneurysms: insights from animal models and relevance to human disease. Cardiovasc Res 2016; 110:165-77. [PMID: 26965051 DOI: 10.1093/cvr/cvw046] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 01/28/2016] [Indexed: 01/09/2023] Open
Abstract
Abdominal aortic aneurysm (AAA) is a major health concern and may be associated with high rates of mortality linked to acute complications. Diagnosis and treatment are, respectively, based on imaging and surgical techniques. Drug-based therapies are still mostly ineffective, which highlight a real unmet need. Major pathophysiological mechanisms leading to aneurysm formation involve inflammatory processes, degradation of the extracellular matrix, and loss of smooth muscle cells. However, the precise cellular and molecular pathways are still poorly understood. Recently, microRNAs have emerged as major intracellular players in a wide range of biological processes, and their stability in extracellular medium within microvesicles has led to propose them as mediators of intercellular crosstalk and as potential biomarkers and therapeutic targets in a variety of disease settings. To date, several studies have been performed to address the involvement of micro-RNAs (miRs) in aneurysm formation and complications. Here, we discuss the roles and implications of miRs in animal models and their relevance to human AAA.
Collapse
Affiliation(s)
- Juliette Raffort
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge CB20 SZ, UK University of Nice-Sophia Antipolis, Medical School, Nice 06107, France INSERM U1081, CNRS UMR7284, IRCAN, Nice, France Clinical Chemistry Laboratory, University Hospital of Nice, Nice, France
| | - Fabien Lareyre
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge CB20 SZ, UK University of Nice-Sophia Antipolis, Medical School, Nice 06107, France INSERM U1081, CNRS UMR7284, IRCAN, Nice, France Department of Vascular Surgery, University Hospital of Nice, Nice, France
| | - Marc Clement
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge CB20 SZ, UK
| | - Ziad Mallat
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge CB20 SZ, UK Institut National de la Santé et de la Recherche Médicale (Inserm), Unit 970, Paris Cardiovascular Research Center, Paris 75015, France
| |
Collapse
|