1
|
Terriaca S, Scioli MG, Bertoldo F, Pisano C, Nardi P, Balistreri CR, Magro D, Belmonte B, Savino L, Ferlosio A, Orlandi A. miRNA-Driven Regulation of Endothelial-to-Mesenchymal Transition Differs among Thoracic Aortic Aneurysms. Cells 2024; 13:1252. [PMID: 39120283 PMCID: PMC11312012 DOI: 10.3390/cells13151252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/16/2024] [Accepted: 07/24/2024] [Indexed: 08/10/2024] Open
Abstract
Thoracic aortic aneurysms (TAAs) represent a serious health concern, as they are associated with early aortic dissection and rupture. TAA formation is triggered by genetic conditions, in particular Marfan syndrome (MFS) and bicuspid aortic valve (BAV). During the aneurysmatic process, aortic endothelial cells can undergo endothelial-to-mesenchymal transition (End-MT) with consequent phenotypic and functional alterations. We previously documented that MFS TAA is characterized by miR-632-driven End-MT exacerbation, whereas in BAV aortopathy, the occurrence of this process remains still controversial. We investigated the End-MT process and the underlined regulatory mechanisms in BAV, TAV and MFS TAA tissues. Gene expression and immunohistochemical analysis were performed in order to analyze some important miRNAs and genes characterizing End-MT. We documented that BAV endothelium maintains the expression of the endothelial homeostasis markers, such as ERG, CD31 and miR-126-5p, while it shows lower levels of miR-632 and mesenchymal markers compared with MFS. Interestingly, we also found higher levels of miR-632 in MFS patients' blood. Our findings definitively demonstrate that the End-MT process does not characterize BAV that, among the other TAAs, better maintains the endothelial features. In addition, our results suggest miR-632 as a promising diagnostic/prognostic factor in MFS aortopathy.
Collapse
Affiliation(s)
- Sonia Terriaca
- Anatomic Pathology, Fondazione Policlinico Tor Vergata, 00133 Rome, Italy; (S.T.); (L.S.)
| | - Maria Giovanna Scioli
- Anatomic Pathology, Department of Biomedicine and Prevention, Tor Vergata University, 00133 Rome, Italy; (A.F.); (A.O.)
| | - Fabio Bertoldo
- Cardiac Surgery Unit, Department of Surgery, Tor Vergata University, 00133 Rome, Italy; (F.B.); (C.P.); (P.N.)
| | - Calogera Pisano
- Cardiac Surgery Unit, Department of Surgery, Tor Vergata University, 00133 Rome, Italy; (F.B.); (C.P.); (P.N.)
| | - Paolo Nardi
- Cardiac Surgery Unit, Department of Surgery, Tor Vergata University, 00133 Rome, Italy; (F.B.); (C.P.); (P.N.)
| | - Carmela Rita Balistreri
- Cellular and Molecular Laboratory, Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), University of Palermo, 90134 Palermo, Italy; (C.R.B.); (D.M.)
| | - Daniele Magro
- Cellular and Molecular Laboratory, Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), University of Palermo, 90134 Palermo, Italy; (C.R.B.); (D.M.)
| | - Beatrice Belmonte
- Tumor Immunology Unit, Department of Health Sciences, University of Palermo, 90134 Palermo, Italy;
- Azienda sanitaria Provinciale di Catania (ASP), 95124 Catania, Italy
| | - Luca Savino
- Anatomic Pathology, Fondazione Policlinico Tor Vergata, 00133 Rome, Italy; (S.T.); (L.S.)
| | - Amedeo Ferlosio
- Anatomic Pathology, Department of Biomedicine and Prevention, Tor Vergata University, 00133 Rome, Italy; (A.F.); (A.O.)
| | - Augusto Orlandi
- Anatomic Pathology, Department of Biomedicine and Prevention, Tor Vergata University, 00133 Rome, Italy; (A.F.); (A.O.)
- Department of Biomedical Sciences, Catholic University Our Lady of Good Counsel, 1001 Tirana, Albania
| |
Collapse
|
2
|
Udugampolage NS, Frolova S, Taurino J, Pini A, Martelli F, Voellenkle C. Coding and Non-Coding Transcriptomic Landscape of Aortic Complications in Marfan Syndrome. Int J Mol Sci 2024; 25:7367. [PMID: 39000474 PMCID: PMC11242319 DOI: 10.3390/ijms25137367] [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: 05/20/2024] [Revised: 06/21/2024] [Accepted: 06/27/2024] [Indexed: 07/16/2024] Open
Abstract
Marfan syndrome (MFS) is a rare congenital disorder of the connective tissue, leading to thoracic aortic aneurysms (TAA) and dissection, among other complications. Currently, the most efficient strategy to prevent life-threatening dissection is preventive surgery. Periodic imaging applying complex techniques is required to monitor TAA progression and to guide the timing of surgical intervention. Thus, there is an acute demand for non-invasive biomarkers for diagnosis and prognosis, as well as for innovative therapeutic targets of MFS. Unraveling the intricate pathomolecular mechanisms underlying the syndrome is vital to address these needs. High-throughput platforms are particularly well-suited for this purpose, as they enable the integration of different datasets, such as transcriptomic and epigenetic profiles. In this narrative review, we summarize relevant studies investigating changes in both the coding and non-coding transcriptome and epigenome in MFS-induced TAA. The collective findings highlight the implicated pathways, such as TGF-β signaling, extracellular matrix structure, inflammation, and mitochondrial dysfunction. Potential candidates as biomarkers, such as miR-200c, as well as therapeutic targets emerged, like Tfam, associated with mitochondrial respiration, or miR-632, stimulating endothelial-to-mesenchymal transition. While these discoveries are promising, rigorous and extensive validation in large patient cohorts is indispensable to confirm their clinical relevance and therapeutic potential.
Collapse
Affiliation(s)
| | - Svetlana Frolova
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, 20097 Milan, Italy; (S.F.); (C.V.)
- Department of Biosciences, University of Milan, 20122 Milan, Italy
| | - Jacopo Taurino
- Cardiovascular-Genetic Center, IRCCS Policlinico San Donato, 20097 Milan, Italy; (N.S.U.); (J.T.); (A.P.)
| | - Alessandro Pini
- Cardiovascular-Genetic Center, IRCCS Policlinico San Donato, 20097 Milan, Italy; (N.S.U.); (J.T.); (A.P.)
| | - Fabio Martelli
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, 20097 Milan, Italy; (S.F.); (C.V.)
| | - Christine Voellenkle
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, 20097 Milan, Italy; (S.F.); (C.V.)
| |
Collapse
|
3
|
Van Den Heuvel LJF, Peeters S, Meester JAN, Coucke PJ, Loeys BL. An exploration of alternative therapeutic targets for aortic disease in Marfan syndrome. Drug Discov Today 2024; 29:104023. [PMID: 38750929 DOI: 10.1016/j.drudis.2024.104023] [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: 02/27/2024] [Revised: 04/29/2024] [Accepted: 05/08/2024] [Indexed: 05/21/2024]
Abstract
Marfan syndrome is a rare connective tissue disorder that causes aortic dissection-related sudden death. Current conventional treatments, beta-blockers, and type 1 angiotensin II receptor blockers are prescribed to slow down aortic aneurysm progression and delay (prophylactic) aortic surgery. However, neither of these treatments ceases aortic growth completely. This review focuses on potential alternative therapeutic leads in the field, ranging from widely used medication with beneficial effects on the aorta to experimental inhibitors with the potential to stop aortic growth in Marfan syndrome. Clinical trials are warranted to uncover their full potential.
Collapse
Affiliation(s)
- Lotte J F Van Den Heuvel
- Center for Medical Genetics Antwerp, University of Antwerp, Antwerp, Belgium; Antwerp University Hospital, Edegem, Belgium; Center for Medical Genetics Ghent, Ghent University, Ghent, Belgium
| | - Silke Peeters
- Center for Medical Genetics Antwerp, University of Antwerp, Antwerp, Belgium; Antwerp University Hospital, Edegem, Belgium
| | - Josephina A N Meester
- Center for Medical Genetics Antwerp, University of Antwerp, Antwerp, Belgium; Antwerp University Hospital, Edegem, Belgium
| | - Paul J Coucke
- Center for Medical Genetics Ghent, Ghent University, Ghent, Belgium
| | - Bart L Loeys
- Center for Medical Genetics Antwerp, University of Antwerp, Antwerp, Belgium; Antwerp University Hospital, Edegem, Belgium; Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands.
| |
Collapse
|
4
|
Terriaca S, Ferlosio A, Scioli MG, Coppa F, Bertoldo F, Pisano C, Belmonte B, Balistreri CR, Orlandi A. miRNA Regulation of Cell Phenotype and Parietal Remodeling in Atherosclerotic and Non-Atherosclerotic Aortic Aneurysms: Differences and Similarities. Int J Mol Sci 2024; 25:2641. [PMID: 38473887 DOI: 10.3390/ijms25052641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 02/21/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
Aortic aneurysms are a serious health concern as their rupture leads to high morbidity and mortality. Abdominal aortic aneurysms (AAAs) and thoracic aortic aneurysms (TAAs) exhibit differences and similarities in their pathophysiological and pathogenetic features. AAA is a multifactorial disease, mainly associated with atherosclerosis, characterized by a relevant inflammatory response and calcification. TAA is rarely associated with atherosclerosis and in some cases is associated with genetic mutations such as Marfan syndrome (MFS) and bicuspid aortic valve (BAV). MFS-related and non-genetic or sporadic TAA share aortic degeneration with endothelial-to-mesenchymal transition (End-Mt) and fibrosis, whereas in BAV TAA, aortic degeneration with calcification prevails. microRNA (miRNAs) contribute to the regulation of aneurysmatic aortic remodeling. miRNAs are a class of non-coding RNAs, which post-transcriptionally regulate gene expression. In this review, we report the involvement of deregulated miRNAs in the different aortic remodeling characterizing AAAs and TAAs. In AAA, miRNA deregulation appears to be involved in parietal inflammatory response, smooth muscle cell (SMC) apoptosis and aortic wall calcification. In sporadic and MFS-related TAA, miRNA deregulation promotes End-Mt, SMC myofibroblastic phenotypic switching and fibrosis with glycosaminoglycan accumulation. In BAV TAA, miRNA deregulation sustains aortic calcification. Those differences may support the development of more personalized therapeutic approaches.
Collapse
Affiliation(s)
- Sonia Terriaca
- Anatomic Pathology, Policlinico Tor Vergata, 00133 Rome, Italy
| | - Amedeo Ferlosio
- Anatomic Pathology, Department of Biomedicine and Prevention, Tor Vergata University, 00133 Rome, Italy
| | - Maria Giovanna Scioli
- Anatomic Pathology, Department of Biomedicine and Prevention, Tor Vergata University, 00133 Rome, Italy
| | - Francesca Coppa
- Anatomic Pathology, Department of Biomedicine and Prevention, Tor Vergata University, 00133 Rome, Italy
| | - Fabio Bertoldo
- Cardiac Surgery Unit, Department of Surgery, Tor Vergata University, 00133 Rome, Italy
| | - Calogera Pisano
- Cardiac Surgery Unit, Department of Surgery, Tor Vergata University, 00133 Rome, Italy
| | - Beatrice Belmonte
- Tumor Immunology Unit, Department of Health Sciences, University of Palermo, 90134 Palermo, Italy
- Azienda sanitaria Provinciale di Catania (ASP), 95124 Catania, Italy
| | - Carmela Rita Balistreri
- Cellular and Molecular Laboratory, Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), University of Palermo, 90134 Palermo, Italy
| | - Augusto Orlandi
- Anatomic Pathology, Department of Biomedicine and Prevention, Tor Vergata University, 00133 Rome, Italy
| |
Collapse
|
5
|
Terriaca S, Monastero R, Orlandi A, Balistreri CR. The key role of miRNA in syndromic and sporadic forms of ascending aortic aneurysms as biomarkers and targets of novel therapeutic strategies. Front Genet 2024; 15:1365711. [PMID: 38450200 PMCID: PMC10915088 DOI: 10.3389/fgene.2024.1365711] [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: 01/04/2024] [Accepted: 01/30/2024] [Indexed: 03/08/2024] Open
Abstract
Increasing evidence shows that epigenetics also plays a key role in regulating the pathogenetic mechanism of all types of aortic aneurysms. It is well-known that epigenetic factors modulate gene expression. This mechanism appears to be of interest especially knowing the relevance of genetic susceptibility and genetic factors in the complex pathophysiology of aortic aneurysms, and of sporadic forms; in fact, the latter are the result of a close interaction between genetic and modifiable lifestyle factors (i.e., nutrition, smoking, infections, use of drugs, alcohol, sedentary lifestyle, etc.). Epigenetic factors include DNA methylation, post-translational histone modifications, and non-coding RNA. Here, our attention is focused on the role of miRNA in syndromic and sporadic forms of thoracic aortic aneurysms. They could be both biomarkers and targets of novel therapeutic strategies.
Collapse
Affiliation(s)
- Sonia Terriaca
- Pathological Anatomy, Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - Roberto Monastero
- Section of Neurology, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Augusto Orlandi
- Pathological Anatomy, Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - Carmela Rita Balistreri
- Cellular, Molecular, and Clinical Pathological Laboratory, Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi N D), University of Palermo, Palermo, Italy
| |
Collapse
|
6
|
Shi Y, Chen J, Cai L, Zhang X, Chen Z, Yang J, Jiang Y, Lu Y. Uncovering the Hidden World of Aqueous Humor Proteins for Discovery of Biomarkers for Marfan Syndrome. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2303161. [PMID: 38088571 PMCID: PMC10853735 DOI: 10.1002/advs.202303161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 10/23/2023] [Indexed: 12/19/2023]
Abstract
Ectopia lentis is a hallmark of Marfan syndrome (MFS), a genetic connective tissue disorder affecting 1/5000 to 1/10 000 individuals worldwide. Early detection in ophthalmology clinics and timely intervention of cardiovascular complications can be lifesaving. In this study, a modified proteomics workflow with liquid chromatography-tandem mass spectrometry (LC-MS/MS)-based data-independent acquisition (DIA) and field asymmetric ion mobility spectrometry (FAIMS) to profile the proteomes of aqueous humor (AH) and lens tissue from MFS children with ectopia lentis is utilized. Over 2300 and 2938 comparable proteins are identified in AH and the lens capsule, respectively. Functional enrichment analyses uncovered dysregulation of complement and coagulation-related pathways, collagen binding, and cell adhesion in MFS. Through weighted correlation network analysis (WGCNA) and machine learning, distinct modules associated with clinical traits are constructed and a unique biomarker panel (Q14376, Q99972, P02760, Q07507; gene names: GALE, MYOC, AMBP, DPT) is defined. These biomarkers are further validated using advanced parallel reaction monitoring (PRM) in an independent patient cohort. The results provide novel insights into the proteome characterization of ectopia lentis and offer a promising approach for developing a valuable biomarker panel to aid in the early diagnosis of Marfan syndrome via AH proteome.
Collapse
Affiliation(s)
- Yumeng Shi
- Eye Institute and Department of Ophthalmology, Eye and ENT HospitalFudan UniversityShanghai200031China
- NHC Key Laboratory of MyopiaFudan UniversityShanghai200031China
- Key Laboratory of MyopiaChinese Academy of Medical SciencesShanghai200031China
- Shanghai Key Laboratory of Visual Impairment and RestorationShanghai200031China
| | - Jiahui Chen
- Eye Institute and Department of Ophthalmology, Eye and ENT HospitalFudan UniversityShanghai200031China
- NHC Key Laboratory of MyopiaFudan UniversityShanghai200031China
- Key Laboratory of MyopiaChinese Academy of Medical SciencesShanghai200031China
- Shanghai Key Laboratory of Visual Impairment and RestorationShanghai200031China
| | - Lei Cai
- Eye Institute and Department of Ophthalmology, Eye and ENT HospitalFudan UniversityShanghai200031China
- NHC Key Laboratory of MyopiaFudan UniversityShanghai200031China
- Key Laboratory of MyopiaChinese Academy of Medical SciencesShanghai200031China
- Shanghai Key Laboratory of Visual Impairment and RestorationShanghai200031China
| | - Xueling Zhang
- Eye Institute and Department of Ophthalmology, Eye and ENT HospitalFudan UniversityShanghai200031China
- NHC Key Laboratory of MyopiaFudan UniversityShanghai200031China
- Key Laboratory of MyopiaChinese Academy of Medical SciencesShanghai200031China
- Shanghai Key Laboratory of Visual Impairment and RestorationShanghai200031China
| | - Zexu Chen
- Eye Institute and Department of Ophthalmology, Eye and ENT HospitalFudan UniversityShanghai200031China
- NHC Key Laboratory of MyopiaFudan UniversityShanghai200031China
- Key Laboratory of MyopiaChinese Academy of Medical SciencesShanghai200031China
- Shanghai Key Laboratory of Visual Impairment and RestorationShanghai200031China
| | - Jin Yang
- Eye Institute and Department of Ophthalmology, Eye and ENT HospitalFudan UniversityShanghai200031China
- NHC Key Laboratory of MyopiaFudan UniversityShanghai200031China
- Key Laboratory of MyopiaChinese Academy of Medical SciencesShanghai200031China
- Shanghai Key Laboratory of Visual Impairment and RestorationShanghai200031China
| | - Yongxiang Jiang
- Eye Institute and Department of Ophthalmology, Eye and ENT HospitalFudan UniversityShanghai200031China
- NHC Key Laboratory of MyopiaFudan UniversityShanghai200031China
- Key Laboratory of MyopiaChinese Academy of Medical SciencesShanghai200031China
- Shanghai Key Laboratory of Visual Impairment and RestorationShanghai200031China
| | - Yi Lu
- Eye Institute and Department of Ophthalmology, Eye and ENT HospitalFudan UniversityShanghai200031China
- NHC Key Laboratory of MyopiaFudan UniversityShanghai200031China
- Key Laboratory of MyopiaChinese Academy of Medical SciencesShanghai200031China
- Shanghai Key Laboratory of Visual Impairment and RestorationShanghai200031China
| |
Collapse
|
7
|
Kucher AN, Koroleva IA, Nazarenko MS. Pathogenetic Significance of Long Non-Coding RNAs in the Development of Thoracic and Abdominal Aortic Aneurysms. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:130-147. [PMID: 38467550 DOI: 10.1134/s0006297924010085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 11/30/2023] [Accepted: 12/01/2023] [Indexed: 03/13/2024]
Abstract
Aortic aneurysm (AA) is a life-threatening condition with a high prevalence and risk of severe complications. The aim of this review was to summarize the data on the role of long non-coding RNAs (lncRNAs) in the development of AAs of various location. Within less than a decade of studies on the role of lncRNAs in AA, using experimental and bioinformatic approaches, scientists have obtained the data confirming the involvement of these molecules in metabolic pathways and pathogenetic mechanisms critical for the aneurysm development. Regardless of the location of pathological process (thoracic or abdominal aorta), AA was found to be associated with changes in the expression of various lncRNAs in the tissue of the affected vessels. The consistency of changes in the expression level of lncRNA, mRNA and microRNA in aortic tissues during AA development has been recordedand regulatory networks implicated in the AA pathogenesis in which lncRNAs act as competing endogenous RNAs (ceRNA networks) have been identified. It was found that the same lncRNA can be involved in different ceRNA networks and regulate different biochemical and cellular events; on the other hand, the same pathological process can be controlled by different lncRNAs. Despite some similarities in pathogenesis and overlapping of involved lncRNAs, the ceRNA networks described for abdominal and thoracic AA are different. Interactions between lncRNAs and other molecules, including those participating in epigenetic processes, have also been identified as potentially relevant to the AA pathogenesis. The expression levels of some lncRNAs were found to correlate with clinically significant aortic features and biochemical parameters. Identification of regulatory RNAs functionally significant in the aneurysm development is important for clarification of disease pathogenesis and will provide a basis for early diagnostics and development of new preventive and therapeutic drugs.
Collapse
Affiliation(s)
- Aksana N Kucher
- Research Institute of Medical Genetics, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, 634050, Russia
| | - Iuliia A Koroleva
- Research Institute of Medical Genetics, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, 634050, Russia
| | - Maria S Nazarenko
- Research Institute of Medical Genetics, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, 634050, Russia.
| |
Collapse
|
8
|
Terriaca S, Scioli MG, Pisano C, Ruvolo G, Ferlosio A, Orlandi A. miR-632 Induces DNAJB6 Inhibition Stimulating Endothelial-to-Mesenchymal Transition and Fibrosis in Marfan Syndrome Aortopathy. Int J Mol Sci 2023; 24:15133. [PMID: 37894814 PMCID: PMC10607153 DOI: 10.3390/ijms242015133] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 09/19/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023] Open
Abstract
Marfan syndrome (MFS) is a connective tissue disorder caused by FBN1 gene mutations leading to TGF-β signaling hyperactivation, vascular wall weakness, and thoracic aortic aneurysms (TAAs). The pathogenetic mechanisms are not completely understood and patients undergo early vascular surgery to prevent TAA ruptures. We previously reported miR-632 upregulation in MFS TAA tissues compared with non-genetic TAA tissues. DNAJB6 is a gene target of miR-632 in cancer and plays a critical role in blocking epithelial-to-mesenchymal transition by inhibiting the Wnt/β catenin pathway. TGF-β signaling also activates Wnt/β catenin signaling and induces endothelial-to-mesenchymal transition (End-Mt) and fibrosis. We documented that miR-632 upregulation correlated with DNAJB6 expression in both the endothelium and the tunica media of MFS TAA (p < 0.01). Wnt/β catenin signaling, End-Mt, and fibrosis markers were also upregulated in MFS TAA tissues (p < 0.05, p < 0.01 and p < 0.001). Moreover, miR-632 overexpression inhibited DNAJB6, inducing Wnt/β catenin signaling, as well as End-Mt and fibrosis exacerbation (p < 0.05 and p < 0.01). TGF-β1 treatment also determined miR-632 upregulation (p < 0.01 and p < 0.001), with the consequent activation of the aforementioned processes. Our study provides new insights about the pathogenetic mechanisms in MFS aortopathy. Moreover, the high disease specificity of miR-632 and DNAJB6 suggests new potential prognostic factors and/or therapeutic targets in the progression of MFS aortopathy.
Collapse
Affiliation(s)
- Sonia Terriaca
- Anatomic Pathology, Department of Biomedicine and Prevention, Tor Vergata University, 00133 Rome, Italy; (S.T.); (A.F.); (A.O.)
| | - Maria Giovanna Scioli
- Anatomic Pathology, Department of Biomedicine and Prevention, Tor Vergata University, 00133 Rome, Italy; (S.T.); (A.F.); (A.O.)
| | - Calogera Pisano
- Cardiac Surgery, Department of Surgical Sciences, Tor Vergata University, 00133 Rome, Italy; (C.P.); (G.R.)
| | - Giovanni Ruvolo
- Cardiac Surgery, Department of Surgical Sciences, Tor Vergata University, 00133 Rome, Italy; (C.P.); (G.R.)
| | - Amedeo Ferlosio
- Anatomic Pathology, Department of Biomedicine and Prevention, Tor Vergata University, 00133 Rome, Italy; (S.T.); (A.F.); (A.O.)
| | - Augusto Orlandi
- Anatomic Pathology, Department of Biomedicine and Prevention, Tor Vergata University, 00133 Rome, Italy; (S.T.); (A.F.); (A.O.)
| |
Collapse
|
9
|
Hu Y, Cai Z, He B. Smooth Muscle Heterogeneity and Plasticity in Health and Aortic Aneurysmal Disease. Int J Mol Sci 2023; 24:11701. [PMID: 37511460 PMCID: PMC10380637 DOI: 10.3390/ijms241411701] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 07/16/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
Vascular smooth muscle cells (VSMCs) are the predominant cell type in the medial layer of the aorta, which plays a critical role in the maintenance of aortic wall integrity. VSMCs have been suggested to have contractile and synthetic phenotypes and undergo phenotypic switching to contribute to the deteriorating aortic wall structure. Recently, the unprecedented heterogeneity and diversity of VSMCs and their complex relationship to aortic aneurysms (AAs) have been revealed by high-resolution research methods, such as lineage tracing and single-cell RNA sequencing. The aortic wall consists of VSMCs from different embryonic origins that respond unevenly to genetic defects that directly or indirectly regulate VSMC contractile phenotype. This difference predisposes to hereditary AAs in the aortic root and ascending aorta. Several VSMC phenotypes with different functions, for example, secreting VSMCs, proliferative VSMCs, mesenchymal stem cell-like VSMCs, immune-related VSMCs, proinflammatory VSMCs, senescent VSMCs, and stressed VSMCs are identified in non-hereditary AAs. The transformation of VSMCs into different phenotypes is an adaptive response to deleterious stimuli but can also trigger pathological remodeling that exacerbates the pathogenesis and development of AAs. This review is intended to contribute to the understanding of VSMC diversity in health and aneurysmal diseases. Papers that give an update on VSMC phenotype diversity in health and aneurysmal disease are summarized and recent insights on the role of VSMCs in AAs are discussed.
Collapse
Affiliation(s)
- Yunwen Hu
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Zhaohua Cai
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Ben He
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| |
Collapse
|
10
|
Sophocleous F, De Garate E, Bigotti MG, Anwar M, Jover E, Chamorro-Jorganes A, Rajakaruna C, Mitrousi K, De Francesco V, Wilson A, Stoica S, Parry A, Benedetto U, Chivasso P, Gill F, Hamilton MCK, Bucciarelli-Ducci C, Caputo M, Emanueli C, Biglino G. A Segmental Approach from Molecular Profiling to Medical Imaging to Study Bicuspid Aortic Valve Aortopathy. Cells 2022; 11:cells11233721. [PMID: 36496981 PMCID: PMC9737804 DOI: 10.3390/cells11233721] [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: 08/25/2022] [Revised: 11/01/2022] [Accepted: 11/14/2022] [Indexed: 11/25/2022] Open
Abstract
Bicuspid aortic valve (BAV) patients develop ascending aortic (AAo) dilation. The pathogenesis of BAV aortopathy (genetic vs. haemodynamic) remains unclear. This study aims to identify regional changes around the AAo wall in BAV patients with aortopathy, integrating molecular data and clinical imaging. BAV patients with aortopathy (n = 15) were prospectively recruited to surgically collect aortic tissue and measure molecular markers across the AAo circumference. Dilated (anterior/right) vs. non-dilated (posterior/left) circumferential segments were profiled for whole-genomic microRNAs (next-generation RNA sequencing, miRCURY LNA PCR), protein content (tandem mass spectrometry), and elastin fragmentation and degeneration (histomorphometric analysis). Integrated bioinformatic analyses of RNA sequencing and proteomic datasets identified five microRNAs (miR-128-3p, miR-210-3p, miR-150-5p, miR-199b-5p, and miR-21-5p) differentially expressed across the AAo circumference. Among them, three miRNAs (miR-128-3p, miR-150-5p, and miR-199b-5p) were predicted to have an effect on eight common target genes, whose expression was dysregulated, according to proteomic analyses, and involved in the vascular-endothelial growth-factor signalling, Hippo signalling, and arachidonic acid pathways. Decreased elastic fibre levels and elastic layer thickness were observed in the dilated segments. Additionally, in a subset of patients n = 6/15, a four-dimensional cardiac magnetic resonance (CMR) scan was performed. Interestingly, an increase in wall shear stress (WSS) was observed at the anterior/right wall segments, concomitantly with the differentially expressed miRNAs and decreased elastic fibres. This study identified new miRNAs involved in the BAV aortic wall and revealed the concomitant expressional dysregulation of miRNAs, proteins, and elastic fibres on the anterior/right wall in dilated BAV patients, corresponding to regions of elevated WSS.
Collapse
Affiliation(s)
- Froso Sophocleous
- Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol BS8 1TH, UK
- Bristol Heart Institute, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol BS1 3NU, UK
| | - Estefania De Garate
- Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol BS8 1TH, UK
- Bristol Heart Institute, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol BS1 3NU, UK
| | - Maria Giulia Bigotti
- Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol BS8 1TH, UK
- School of Biochemistry, Faculty of Life Sciences, University of Bristol, Bristol BS8 1TH, UK
| | - Maryam Anwar
- National Heart and Lung Institute, Imperial College London, London SW7 2BX, UK
| | - Eva Jover
- Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol BS8 1TH, UK
- Instituto de Investigación Sanitaria de Navarra, 31008 Pamplona, Spain
| | | | - Cha Rajakaruna
- Bristol Heart Institute, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol BS1 3NU, UK
| | - Konstantina Mitrousi
- Bristol Heart Institute, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol BS1 3NU, UK
| | - Viola De Francesco
- Bristol Heart Institute, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol BS1 3NU, UK
| | - Aileen Wilson
- Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol BS8 1TH, UK
- Bristol Heart Institute, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol BS1 3NU, UK
| | - Serban Stoica
- Bristol Heart Institute, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol BS1 3NU, UK
| | - Andrew Parry
- Bristol Heart Institute, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol BS1 3NU, UK
| | - Umberto Benedetto
- Bristol Heart Institute, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol BS1 3NU, UK
| | - Pierpaolo Chivasso
- Bristol Heart Institute, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol BS1 3NU, UK
| | - Frances Gill
- Bristol Heart Institute, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol BS1 3NU, UK
| | - Mark C. K. Hamilton
- Department of Clinical Radiology, University Hospitals Bristol, Bristol Royal Infirmary, Bristol BS2 8EJ, UK
| | - Chiara Bucciarelli-Ducci
- Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol BS8 1TH, UK
- Bristol Heart Institute, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol BS1 3NU, UK
- Royal Brompton & Harefield Hospitals, Guy’s and St Thomas’ NHS Foundation Trust, London SW3 6NP, UK
| | - Massimo Caputo
- Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol BS8 1TH, UK
- Bristol Heart Institute, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol BS1 3NU, UK
| | - Costanza Emanueli
- National Heart and Lung Institute, Imperial College London, London SW7 2BX, UK
| | - Giovanni Biglino
- Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol BS8 1TH, UK
- National Heart and Lung Institute, Imperial College London, London SW7 2BX, UK
- Correspondence: ; Tel.: +44-117-342-3287
| |
Collapse
|
11
|
Pisano C, Terriaca S, Scioli MG, Nardi P, Altieri C, Orlandi A, Ruvolo G, Balistreri CR. The Endothelial Transcription Factor ERG Mediates a Differential Role in the Aneurysmatic Ascending Aorta with Bicuspid or Tricuspid Aorta Valve: A Preliminary Study. Int J Mol Sci 2022; 23:10848. [PMID: 36142762 PMCID: PMC9502538 DOI: 10.3390/ijms231810848] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 09/01/2022] [Accepted: 09/13/2022] [Indexed: 11/18/2022] Open
Abstract
The pathobiology of ascending aorta aneurysms (AAA) onset and progression is not well understood and only partially characterized. AAA are also complicated in case of bicuspid aorta valve (BAV) anatomy. There is emerging evidence about the crucial role of endothelium-related pathways, which show in AAA an altered expression and function. Here, we examined the involvement of ERG-related pathways in the differential progression of disease in aortic tissues from patients having a BAV or tricuspid aorta valve (TAV) with or without AAA. Our findings identified ERG as a novel endothelial-specific regulator of TGF-β-SMAD, Notch, and NO pathways, by modulating a differential fibrotic or calcified AAA progression in BAV and TAV aortas. We provided evidence that calcification is correlated to different ERG expression (as gene and protein), which appears to be under control of Notch signaling. The latter, when increased, associated with an early calcification in aortas with BAV valve and aneurysmatic, was demonstrated to favor the progression versus severe complications, i.e., dissection or rupture. In TAV aneurysmatic aortas, ERG appeared to modulate fibrosis. Therefore, we proposed that ERG may represent a sensitive tissue biomarker to monitor AAA progression and a target to develop therapeutic strategies and influence surgical procedures.
Collapse
Affiliation(s)
- Calogera Pisano
- Department of Cardiac Surgery, Tor Vergata University Polyclinic, 00133 Rome, Italy
| | - Sonia Terriaca
- Pathological Anatomy, Department of Biomedicine and Prevention, Tor Vergata University, 00133 Rome, Italy
| | - Maria Giovanna Scioli
- Pathological Anatomy, Department of Biomedicine and Prevention, Tor Vergata University, 00133 Rome, Italy
| | - Paolo Nardi
- Department of Cardiac Surgery, Tor Vergata University Polyclinic, 00133 Rome, Italy
| | - Claudia Altieri
- Department of Cardiac Surgery, Tor Vergata University Polyclinic, 00133 Rome, Italy
| | - Augusto Orlandi
- Pathological Anatomy, Department of Biomedicine and Prevention, Tor Vergata University, 00133 Rome, Italy
- Department of Biomedical Sciences, Catholic University of Our Lady of Good Counsel, 1001 Tirana, Albania
| | - Giovanni Ruvolo
- Department of Cardiac Surgery, Tor Vergata University Polyclinic, 00133 Rome, Italy
| | - Carmela Rita Balistreri
- Cellular and Molecular Laboratory, Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), University of Palermo, 90134 Palermo, Italy
| |
Collapse
|
12
|
Tracking an Elusive Killer: State of the Art of Molecular-Genetic Knowledge and Laboratory Role in Diagnosis and Risk Stratification of Thoracic Aortic Aneurysm and Dissection. Diagnostics (Basel) 2022; 12:diagnostics12081785. [PMID: 35892496 PMCID: PMC9329974 DOI: 10.3390/diagnostics12081785] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/12/2022] [Accepted: 07/19/2022] [Indexed: 02/08/2023] Open
Abstract
The main challenge in diagnosing and managing thoracic aortic aneurysm and dissection (TAA/D) is represented by the early detection of a disease that is both deadly and “elusive”, as it generally grows asymptomatically prior to rupture, leading to death in the majority of cases. Gender differences exist in aortic dissection in terms of incidence and treatment options. Efforts have been made to identify biomarkers that may help in early diagnosis and in detecting those patients at a higher risk of developing life-threatening complications. As soon as the hereditability of the TAA/D was demonstrated, several genetic factors were found to be associated with both the syndromic and non-syndromic forms of the disease, and they currently play a role in patient diagnosis/prognosis and management-guidance purposes. Likewise, circulating biomarker could represent a valuable resource in assisting the diagnosis, and several studies have attempted to identify specific molecules that may help with risk stratification outside the emergency department. Even if promising, those data lack specificity/sensitivity, and, in most cases, they need more testing before entering the “clinical arena”. This review summarizes the state of the art of the laboratory in TAA/D diagnostics, with particular reference to the current and future role of molecular-genetic testing.
Collapse
|
13
|
Zhang X, Yang Z, Li X, Liu X, Wang X, Qiu T, Wang Y, Li T, Li Q. Bioinformatics Analysis Reveals Cell Cycle-Related Gene Upregulation in Ascending Aortic Tissues From Murine Models. Front Genet 2022; 13:823769. [PMID: 35356426 PMCID: PMC8959095 DOI: 10.3389/fgene.2022.823769] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 02/14/2022] [Indexed: 11/13/2022] Open
Abstract
Thoracic aortic aneurysm and dissection (TAAD) is a high-risk aortic disease. Mouse models are usually used to explore the pathological progression of TAAD. In our studies, we performed bioinformatics analysis on a microarray dataset (GSE36778) and verified experiments to define the integrated hub genes of TAAD in three different mouse models. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) and protein-protein interaction (PPI) network analyses, and histological and quantitative reverse transcription-PCR (qRT-PCR) experiments were used in our study. First, differentially expressed genes (DEGs) were identified, and twelve common differentially expressed genes were found. Second, genes related to the cell cycle and inflammation were enriched by using GO and PPI. We focused on filtering and validating eighteen hub genes that were upregulated. Then, expression data from human ascending aortic tissues in the GSE153434 dataset were also used to verify our findings. These results indicated that cell cycle-related genes participate in the pathological mechanism of TAAD and provide new insight into the molecular mechanisms of TAAD.
Collapse
Affiliation(s)
- Xiaoping Zhang
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,Beijing Institute of Heart, Lung and Blood Vessel Disease, Beijing, China
| | - Zuozhen Yang
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Xiaoyan Li
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,Beijing Institute of Heart, Lung and Blood Vessel Disease, Beijing, China
| | - Xuxia Liu
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,Beijing Institute of Heart, Lung and Blood Vessel Disease, Beijing, China
| | - Xipeng Wang
- Department of Vascular Surgery, Peking University People's Hospital, Beijing, China
| | - Tao Qiu
- The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yueli Wang
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Tongxun Li
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Qingle Li
- Department of Vascular Surgery, Peking University People's Hospital, Beijing, China
| |
Collapse
|
14
|
Magouliotis DE, Fergadi MP, Christodoulidis G, Svokos AA, Svokos KA, Bareka M, Athanasiou T. In-depth bioinformatic study of the cadherin 5 interactome in patients with thoracic aortic aneurysm unveils 8 novel biomarkers. Eur J Cardiothorac Surg 2021; 61:11-18. [PMID: 34293135 DOI: 10.1093/ejcts/ezab338] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 05/25/2021] [Accepted: 06/12/2021] [Indexed: 01/23/2023] Open
Abstract
OBJECTIVES Thoracic aortic aneurysm (TAA) is characterized by the dilation of the aorta and is associated with poor prognosis if not diagnosed and treated early. In this context, the identification of biomarkers regarding the TAA diagnosis, monitoring and prognosis is crucial. The purpose of the current study was to investigate the differential gene expression profile of the cadherin 5 (CDH5 or VE-Cadherin) gene network in patients with TAA, to propose novel biomarkers. METHODS In silico techniques were used to construct the interactome of the CDH5 network, identify the differentially expressed genes (DEGs) in TAA as compared to healthy controls, and uncover the related molecular functions and the regulating miRNAs. RESULTS Transcriptomic data of one microarray dataset were included, incorporating 43 TAA and 43 control samples. Eight DEGs were identified; 7 were up-regulated and 1 was down-regulated. A molecular signature of 8 genes (CDH5; Calcitonin Receptor-Like Receptor-CALCRL; Activin A Receptor-Like Type 1-ACVRL1, Tryptophanyl-TRNA Synthetase 1-WARS; Junction Plakoglobin-JUP, Protein Tyrosine Phosphatase Receptor Type J-PTPRJ, Purinergic Receptor P2X 4-P2RX4, Kinase Insert Domain Receptor-KDR) were identified as biomarkers associated with TAA. PTPRJ was associated with excellent discrimination and calibration in predicting TAA presentation. Positive correlations were reported regarding the expression of CDH5-CALCRL, CDH5-ACVRL1, CDH5-WARS and CDH5-PTPRJ. Finally, gene set enrichment analysis indicated the molecular functions and miRNA families (hsa-miR-296-5p, hsa-miR-6836-5p, hsa-miR-6132, hsa-miR-27a-5p and hsa-miR-6773-5p) relevant to the 8 biomarkers. CONCLUSIONS These outcomes propose an 8-gene molecular panel associated with TAA.
Collapse
Affiliation(s)
- Dimitrios E Magouliotis
- Division of Surgery and Interventional Science, Faculty of Medical Sciences, UCL, London, UK
- Department of Cardiothoracic Surgery, University of Thessaly, Biopolis, Larissa, Greece
| | - Maria P Fergadi
- Department of Radiology, University of Thessaly, Biopolis, Larissa, Greece
| | | | - Alexis A Svokos
- Department of Obstetrics and Gynecology, Geisinger Medical Center, Danville, PA, USA
| | - Konstantina A Svokos
- Department of Neurosurgery, The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Metaxia Bareka
- Department of Anesthesiology, University of Thessaly, Biopolis, Larissa, Greece
| | - Thanos Athanasiou
- Department of Surgery and Cancer, Imperial College London, St Mary's Hospital, London, UK
| |
Collapse
|
15
|
Tuttolomondo D, De Filippo M, Sartorio D, Nicolini F, Niccoli G, Gaibazzi N. Peri-vascular adipose tissue attenuation on chest computed tomography in patients with Marfan Syndrome: a case series. ACTA BIO-MEDICA : ATENEI PARMENSIS 2021; 92:e2021468. [PMID: 34747387 PMCID: PMC10523050 DOI: 10.23750/abm.v92is1.11669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 04/26/2021] [Indexed: 11/23/2022]
Abstract
BACKGROUND AND AIM OF THE WORK Marfan Syndrome is a genetic disorder that determines histopathological alterations of the aortic vascular wall leading to increased inflammatory component. The peri-vascular adipose tissue attenuation is a method able to capture localized vascular inflammation by mapping spatial changes of perivascular tissue attenuation on computed tomography. METHODS We measured peri-vascular adipose tissue attenuation around the ascending aorta in three consecutive subjects with confirmed genetic diagnosis of Marfan Syndrome. All subjects received the genetic diagnosis of fibrillin-1 gene mutation as part of the family screening of patients with known Marfan Syndrome. Chest computed tomography was performed in such asymptomatic subjects after genetic confirmation of Marfan Syndrome. None of these subjects showed aortic aneurysms or suffered from chronic inflammatory/infectious disease. RESULTS In the three subjects identified with Marfan Syndrome the value of aortic peri-vascular adipose tissue attenuation measured at chest computed tomography was higher than normal and the volume of aortic peri-vascular adipose tissue was lower. CONCLUSION These preliminary observations suggest that peri-vascular adipose tissue attenuation is unexpectedly high in patients with Marfan Syndrome, notwithstanding the normal aortic diameter at the time of computed tomography. Whether this observation may find a clinical use in suspected Marfan Syndrome or in predicting aortic complications in Marfan Syndrome is worth to be assessed in prospective studies.
Collapse
Affiliation(s)
- Domenico Tuttolomondo
- Department of Cardiac Surgery, Parma University Hospital, Via Gramsci 14, 43125, Parma, Italy..
| | - Massimo De Filippo
- Department of Medicine and Surgery, Unit of Radiologic Science, Parma University Hospital, Via Gramsci 14, 43125, Parma, Italy..
| | - Daniele Sartorio
- Department of Cardiology, Parma University Hospital, Via Gramsci 14, 43125, Parma, Italy..
| | - Francesco Nicolini
- Department of Cardiac Surgery, Parma University Hospital, Via Gramsci 14, 43125, Parma, Italy..
| | - Giampaolo Niccoli
- Department of Cardiology, Parma University Hospital, Via Gramsci 14, 43125, Parma, Italy..
| | - Nicola Gaibazzi
- Department of Cardiology, Parma University Hospital, Via Gramsci 14, 43125, Parma, Italy..
| |
Collapse
|
16
|
Liu MN, Luo G, Gao WJ, Yang SJ, Zhou H. miR-29 family: A potential therapeutic target for cardiovascular disease. Pharmacol Res 2021; 166:105510. [PMID: 33610720 DOI: 10.1016/j.phrs.2021.105510] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 02/16/2021] [Indexed: 01/11/2023]
Abstract
Cardiovascular disease (CVD), including heart failure, myocardial fibrosis and myocardial infarction, etc, remains one of the leading causes of mortality worldwide. Evidence shows that miRNA plays an important role in the pathogenesis of CVD. miR-29 family is one of miRNA, and over the past decades, many studies have demonstrated that miR-29 is involved in maintaining the integrity of arteries and in the regulation of atherosclerosis, especially in the process of myocardial fibrosis. Besides, heart failure, myocardial fibrosis and myocardial infarction are inseparable from the regulatory role of miR-29. Here, we comprehensively review recent studies regarding miR-29 and CVD, illustrate the possibility of miR-29 as a potential marker for prevention, treatment and prognostic observation.
Collapse
Affiliation(s)
- Meng-Nan Liu
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, China; National Traditional Chinese Medicine Clinical Research Base and Department of Cardiovascular Medicine, Hospital (T.C.M) Affiliated to Southwest Medical University, Luzhou, Sichuan, China
| | - Gang Luo
- National Traditional Chinese Medicine Clinical Research Base and Department of Cardiovascular Medicine, Hospital (T.C.M) Affiliated to Southwest Medical University, Luzhou, Sichuan, China
| | - Wan-Jiao Gao
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, China
| | - Si-Jin Yang
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, China; National Traditional Chinese Medicine Clinical Research Base and Department of Cardiovascular Medicine, Hospital (T.C.M) Affiliated to Southwest Medical University, Luzhou, Sichuan, China.
| | - Hua Zhou
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, China.
| |
Collapse
|