1
|
Chiarelli N, Cinquina V, Martini P, Bertini V, Zoppi N, Venturini M, Ritelli M, Colombi M. Deciphering disease signatures and molecular targets in vascular Ehlers-Danlos syndrome through transcriptome and miRNome sequencing of dermal fibroblasts. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166915. [PMID: 37827202 DOI: 10.1016/j.bbadis.2023.166915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 09/28/2023] [Accepted: 10/02/2023] [Indexed: 10/14/2023]
Abstract
Vascular Ehlers-Danlos syndrome (vEDS) is a severe connective tissue disorder caused by dominant mutations in the COL3A1 gene encoding type III collagen (COLLIII). COLLIII is primarily found in blood vessels and hollow organs, and its deficiency leads to soft connective tissues fragility, resulting in life-threatening arterial and organ ruptures. There are no current targeted therapies available. Although the disease usually results from COLLIII misfolding due to triple helix structure disruption, the underlying pathomechanisms are largely unknown. To address this knowledge gap, we performed a comprehensive transcriptome analysis using RNA- and miRNA-seq on a large cohort of dermal fibroblasts from vEDS patients and healthy donors. Our investigation revealed an intricate interplay between proteostasis abnormalities, inefficient endoplasmic reticulum stress response, and compromised autophagy, which may significantly impact the molecular pathology. We also present the first detailed miRNAs expression profile in patient cells, demonstrating that several aberrantly expressed miRNAs can disrupt critical cellular functions involved in vEDS pathophysiology, such as autophagy, proteostasis, and mTOR signaling. Target prediction and regulatory networks analyses suggested potential interactions among miRNAs, lncRNAs, and candidate target genes linked to extracellular matrix organization and autophagy-lysosome pathway. Our results highlight the importance of understanding the functional role of ncRNAs in vEDS pathogenesis, shedding light on possible miRNAs and lncRNAs signatures and their functional implications for dysregulated pathways related to disease. Deciphering this complex molecular network of RNA interactions may yield additional evidence for potential disease biomolecules and targets, assisting in the design of effective patient treatment strategies.
Collapse
Affiliation(s)
- Nicola Chiarelli
- Division of Biology and Genetics, Department of Molecular and Translational Medicine, University of Brescia, 25121 Brescia, Italy.
| | - Valeria Cinquina
- Division of Biology and Genetics, Department of Molecular and Translational Medicine, University of Brescia, 25121 Brescia, Italy
| | - Paolo Martini
- Division of Biology and Genetics, Department of Molecular and Translational Medicine, University of Brescia, 25121 Brescia, Italy
| | - Valeria Bertini
- Division of Biology and Genetics, Department of Molecular and Translational Medicine, University of Brescia, 25121 Brescia, Italy
| | - Nicoletta Zoppi
- Division of Biology and Genetics, Department of Molecular and Translational Medicine, University of Brescia, 25121 Brescia, Italy
| | - Marina Venturini
- Division of Dermatology, Department of Clinical and Experimental Sciences, Spedali Civili University Hospital Brescia, 25121 Brescia, Italy
| | - Marco Ritelli
- Division of Biology and Genetics, Department of Molecular and Translational Medicine, University of Brescia, 25121 Brescia, Italy
| | - Marina Colombi
- Division of Biology and Genetics, Department of Molecular and Translational Medicine, University of Brescia, 25121 Brescia, Italy
| |
Collapse
|
2
|
Kallenbach K, Remes A, Müller OJ, Arif R, Zaradzki M, Wagner AH. Translational Medicine: Towards Gene Therapy of Marfan Syndrome. J Clin Med 2022; 11:jcm11143934. [PMID: 35887698 PMCID: PMC9319421 DOI: 10.3390/jcm11143934] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/28/2022] [Accepted: 07/01/2022] [Indexed: 12/12/2022] Open
Abstract
Marfan syndrome (MFS) is one of the most common inherited disorders of connective tissue caused by mutations of the fibrillin-1 gene (FBN1). Vascular abnormalities, such as the enlargement of the aorta with the risk of life-threatening rupture are frequently observed. However, current treatment is limited and therapeutic options focus solely on symptomatic therapy. Gene therapy focuses on genetically modifying cells to produce a therapeutic effect and may be a promising treatment option for MFS. Here, we first provide an overview of the historical background and characterization of MFS. Subsequently, we summarise current gene therapy options and possible translational concepts for this inherited disorder that affects connective tissue.
Collapse
Affiliation(s)
- Klaus Kallenbach
- Institute for Cardiac Surgery and Interventional Cardiology (INCCI), Department of Cardiac Surgery, 1210 Luxembourg, Luxembourg;
- VASCERN HTAD European Reference Center, 1210 Luxembourg, Luxembourg
| | - Anca Remes
- Department of Internal Medicine III, University of Kiel and University Hospital Schleswig-Holstein, 24105 Kiel, Germany; (A.R.); (O.J.M.)
- German Centre for Cardiovascular Research, Partner Site Hamburg/Kiel/Lübeck, 20251 Hamburg, Germany
| | - Oliver J. Müller
- Department of Internal Medicine III, University of Kiel and University Hospital Schleswig-Holstein, 24105 Kiel, Germany; (A.R.); (O.J.M.)
- German Centre for Cardiovascular Research, Partner Site Hamburg/Kiel/Lübeck, 20251 Hamburg, Germany
| | - Rawa Arif
- Department of Cardiac Surgery, University Hospital Heidelberg, 69120 Heidelberg, Germany; (R.A.); (M.Z.)
| | - Marcin Zaradzki
- Department of Cardiac Surgery, University Hospital Heidelberg, 69120 Heidelberg, Germany; (R.A.); (M.Z.)
| | - Andreas H. Wagner
- Department of Cardiovascular Physiology, Heidelberg University, Im Neuenheimer Feld 326, 69120 Heidelberg, Germany
- Correspondence: ; Tel.: +49-6221-544062; Fax: +49-6221-544038
| |
Collapse
|
3
|
Oller J, Gabandé-Rodríguez E, Ruiz-Rodríguez MJ, Desdín-Micó G, Aranda JF, Rodrigues-Diez R, Ballesteros-Martínez C, Blanco EM, Roldan-Montero R, Acuña P, Forteza Gil A, Martín-López CE, Nistal JF, Lino Cardenas CL, Lindsay ME, Martín-Ventura JL, Briones AM, Miguel Redondo J, Mittelbrunn M. Extracellular Tuning of Mitochondrial Respiration Leads to Aortic Aneurysm. Circulation 2021; 143:2091-2109. [PMID: 33709773 PMCID: PMC8140666 DOI: 10.1161/circulationaha.120.051171] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 02/26/2021] [Indexed: 12/19/2022]
Abstract
BACKGROUND Marfan syndrome (MFS) is an autosomal dominant disorder of the connective tissue caused by mutations in the FBN1 (fibrillin-1) gene encoding a large glycoprotein in the extracellular matrix called fibrillin-1. The major complication of this connective disorder is the risk to develop thoracic aortic aneurysm. To date, no effective pharmacologic therapies have been identified for the management of thoracic aortic disease and the only options capable of preventing aneurysm rupture are endovascular repair or open surgery. Here, we have studied the role of mitochondrial dysfunction in the progression of thoracic aortic aneurysm and mitochondrial boosting strategies as a potential treatment to managing aortic aneurysms. METHODS Combining transcriptomics and metabolic analysis of aortas from an MFS mouse model (Fbn1c1039g/+) and MFS patients, we have identified mitochondrial dysfunction alongside with mtDNA depletion as a new hallmark of aortic aneurysm disease in MFS. To demonstrate the importance of mitochondrial decline in the development of aneurysms, we generated a conditional mouse model with mitochondrial dysfunction specifically in vascular smooth muscle cells (VSMC) by conditional depleting Tfam (mitochondrial transcription factor A; Myh11-CreERT2Tfamflox/flox mice). We used a mouse model of MFS to test for drugs that can revert aortic disease by enhancing Tfam levels and mitochondrial respiration. RESULTS The main canonical pathways highlighted in the transcriptomic analysis in aortas from Fbn1c1039g/+ mice were those related to metabolic function, such as mitochondrial dysfunction. Mitochondrial complexes, whose transcription depends on Tfam and mitochondrial DNA content, were reduced in aortas from young Fbn1c1039g/+ mice. In vitro experiments in Fbn1-silenced VSMCs presented increased lactate production and decreased oxygen consumption. Similar results were found in MFS patients. VSMCs seeded in matrices produced by Fbn1-deficient VSMCs undergo mitochondrial dysfunction. Conditional Tfam-deficient VSMC mice lose their contractile capacity, showed aortic aneurysms, and died prematurely. Restoring mitochondrial metabolism with the NAD precursor nicotinamide riboside rapidly reverses aortic aneurysm in Fbn1c1039g/+ mice. CONCLUSIONS Mitochondrial function of VSMCs is controlled by the extracellular matrix and drives the development of aortic aneurysm in Marfan syndrome. Targeting vascular metabolism is a new available therapeutic strategy for managing aortic aneurysms associated with genetic disorders.
Collapse
Affiliation(s)
- Jorge Oller
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas Universidad Autónoma de Madrid, Spain (J.O., E.G-R., G.D-M., J.F.A., E.M.B., P.A., M.M.)
- Instituto de Investigación Sanitaria del Hospital 12 de Octubre (i+12), Madrid, Spain (J.O., E.G-R., G.D-M., J.F.A., E.M.B., M.M.)
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Spain (J.O., R.R-D., R.R-M., A.M.B., J.M.R.)
| | - Enrique Gabandé-Rodríguez
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas Universidad Autónoma de Madrid, Spain (J.O., E.G-R., G.D-M., J.F.A., E.M.B., P.A., M.M.)
- Instituto de Investigación Sanitaria del Hospital 12 de Octubre (i+12), Madrid, Spain (J.O., E.G-R., G.D-M., J.F.A., E.M.B., M.M.)
| | | | - Gabriela Desdín-Micó
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas Universidad Autónoma de Madrid, Spain (J.O., E.G-R., G.D-M., J.F.A., E.M.B., P.A., M.M.)
- Instituto de Investigación Sanitaria del Hospital 12 de Octubre (i+12), Madrid, Spain (J.O., E.G-R., G.D-M., J.F.A., E.M.B., M.M.)
| | - Juan Francisco Aranda
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas Universidad Autónoma de Madrid, Spain (J.O., E.G-R., G.D-M., J.F.A., E.M.B., P.A., M.M.)
- Instituto de Investigación Sanitaria del Hospital 12 de Octubre (i+12), Madrid, Spain (J.O., E.G-R., G.D-M., J.F.A., E.M.B., M.M.)
| | - Raquel Rodrigues-Diez
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Spain (J.O., R.R-D., R.R-M., A.M.B., J.M.R.)
- Departamento de Farmacología, Universidad Autónoma de Madrid, Instituto de Investigación Hospital La Paz, Spain (R.R-D., C.B-M., A.M.B.)
| | - Constanza Ballesteros-Martínez
- Departamento de Farmacología, Universidad Autónoma de Madrid, Instituto de Investigación Hospital La Paz, Spain (R.R-D., C.B-M., A.M.B.)
| | - Eva María Blanco
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas Universidad Autónoma de Madrid, Spain (J.O., E.G-R., G.D-M., J.F.A., E.M.B., P.A., M.M.)
- Instituto de Investigación Sanitaria del Hospital 12 de Octubre (i+12), Madrid, Spain (J.O., E.G-R., G.D-M., J.F.A., E.M.B., M.M.)
| | - Raquel Roldan-Montero
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Spain (J.O., R.R-D., R.R-M., A.M.B., J.M.R.)
- Instituto de Investigación Sanitaria-Fundación Jimenez Diaz, Madrid, Spain (R.R-M. J.L.M-V.)
- Hospital Universitario Puerta de Hierro, Madrid, Spain. (R.R-M., J.L.M-V.)
| | - Pedro Acuña
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas Universidad Autónoma de Madrid, Spain (J.O., E.G-R., G.D-M., J.F.A., E.M.B., P.A., M.M.)
| | - Alberto Forteza Gil
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas Universidad Autónoma de Madrid, Spain (J.O., E.G-R., G.D-M., J.F.A., E.M.B., P.A., M.M.)
- Instituto de Investigación Sanitaria del Hospital 12 de Octubre (i+12), Madrid, Spain (J.O., E.G-R., G.D-M., J.F.A., E.M.B., M.M.)
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Spain (J.O., R.R-D., R.R-M., A.M.B., J.M.R.)
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain (M.J.R-R., J.M.R.)
- Departamento de Farmacología, Universidad Autónoma de Madrid, Instituto de Investigación Hospital La Paz, Spain (R.R-D., C.B-M., A.M.B.)
- Instituto de Investigación Sanitaria-Fundación Jimenez Diaz, Madrid, Spain (R.R-M. J.L.M-V.)
- Hospital Universitario Puerta de Hierro, Madrid, Spain. (R.R-M., J.L.M-V.)
- Cardiovascular Surgery, Hospital Universitario Marqués de Valdecilla, IDIVAL, Universidad de Cantabria, Santander, Spain. (J.F.N.)
- Massachusetts General Hospital Thoracic Aortic Center, Boston (C.L.L.C., M.E.L.)
| | - Carlos E. Martín-López
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas Universidad Autónoma de Madrid, Spain (J.O., E.G-R., G.D-M., J.F.A., E.M.B., P.A., M.M.)
- Instituto de Investigación Sanitaria del Hospital 12 de Octubre (i+12), Madrid, Spain (J.O., E.G-R., G.D-M., J.F.A., E.M.B., M.M.)
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Spain (J.O., R.R-D., R.R-M., A.M.B., J.M.R.)
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain (M.J.R-R., J.M.R.)
- Departamento de Farmacología, Universidad Autónoma de Madrid, Instituto de Investigación Hospital La Paz, Spain (R.R-D., C.B-M., A.M.B.)
- Instituto de Investigación Sanitaria-Fundación Jimenez Diaz, Madrid, Spain (R.R-M. J.L.M-V.)
- Hospital Universitario Puerta de Hierro, Madrid, Spain. (R.R-M., J.L.M-V.)
- Cardiovascular Surgery, Hospital Universitario Marqués de Valdecilla, IDIVAL, Universidad de Cantabria, Santander, Spain. (J.F.N.)
- Massachusetts General Hospital Thoracic Aortic Center, Boston (C.L.L.C., M.E.L.)
| | - J. Francisco Nistal
- Cardiovascular Surgery, Hospital Universitario Marqués de Valdecilla, IDIVAL, Universidad de Cantabria, Santander, Spain. (J.F.N.)
| | | | - Mark Evan Lindsay
- Massachusetts General Hospital Thoracic Aortic Center, Boston (C.L.L.C., M.E.L.)
| | - José Luís Martín-Ventura
- Instituto de Investigación Sanitaria-Fundación Jimenez Diaz, Madrid, Spain (R.R-M. J.L.M-V.)
- Hospital Universitario Puerta de Hierro, Madrid, Spain. (R.R-M., J.L.M-V.)
| | - Ana M. Briones
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Spain (J.O., R.R-D., R.R-M., A.M.B., J.M.R.)
- Departamento de Farmacología, Universidad Autónoma de Madrid, Instituto de Investigación Hospital La Paz, Spain (R.R-D., C.B-M., A.M.B.)
| | - Juan Miguel Redondo
- Instituto de Investigación Sanitaria del Hospital 12 de Octubre (i+12), Madrid, Spain (J.O., E.G-R., G.D-M., J.F.A., E.M.B., M.M.)
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Spain (J.O., R.R-D., R.R-M., A.M.B., J.M.R.)
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain (M.J.R-R., J.M.R.)
| | - María Mittelbrunn
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas Universidad Autónoma de Madrid, Spain (J.O., E.G-R., G.D-M., J.F.A., E.M.B., P.A., M.M.)
| |
Collapse
|
4
|
Schellinger IN, Dannert AR, Mattern K, Raaz U, Tsao PS. Unresolved Issues in RNA Therapeutics in Vascular Diseases With a Focus on Aneurysm Disease. Front Cardiovasc Med 2021; 8:571076. [PMID: 33937351 PMCID: PMC8081859 DOI: 10.3389/fcvm.2021.571076] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 02/23/2021] [Indexed: 12/20/2022] Open
Abstract
New technologies have greatly shaped the scientific and medical landscape within the last years. The unprecedented expansion of data and information on RNA biology has led to the discovery of new RNA classes with unique functions and unexpected modifications. Today, the biggest challenge is to transfer the large number of findings in basic RNA biology into corresponding clinical RNA-based therapeutics. Lately, this research begins to yield positive outcomes. RNA drugs advance to the final phases of clinical trials or even receive FDA approval. Furthermore, the introduction of the RNA-guided gene-editing technology CRISPR and advances in the delivery of messenger RNAs have triggered a major progression in the field of RNA-therapeutics. Especially short interfering RNAs and antisense oligonucleotides are promising examples for novel categories of therapeutics. However, several issues need to be addressed including intracellular delivery, toxicity, and immune responses before utilizing RNAs in a clinical setting. In this review, we provide an overview on opportunities and challenges for clinical translation of RNA-based therapeutics, with an emphasis on advances in novel delivery technologies and abdominal aortic aneurysm disease where non-coding RNAs have been shown to play a crucial regulatory role.
Collapse
Affiliation(s)
- Isabel N Schellinger
- Department of Cardiology and Pneumology, Heart Center at the University Medical Center Göttingen, Göttingen, Germany.,German Center for Cardiovascular Research (DZHK) e.V., Partner Site Göttingen, Göttingen, Germany.,Department for Endocrinology, Nephrology and Rheumatology, University Medical Center Leipzig, University of Leipzig, Leipzig, Germany.,Department for Angiology, University Medical Center Leipzig, University of Leipzig, Leipzig, Germany
| | - Angelika R Dannert
- Department of Cardiology and Pneumology, Heart Center at the University Medical Center Göttingen, Göttingen, Germany
| | - Karin Mattern
- Department of Cardiology and Pneumology, Heart Center at the University Medical Center Göttingen, Göttingen, Germany.,German Center for Cardiovascular Research (DZHK) e.V., Partner Site Göttingen, Göttingen, Germany
| | - Uwe Raaz
- Department of Cardiology and Pneumology, Heart Center at the University Medical Center Göttingen, Göttingen, Germany.,German Center for Cardiovascular Research (DZHK) e.V., Partner Site Göttingen, Göttingen, Germany
| | - Philip S Tsao
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, United States.,Veteran Affairs (VA) Palo Alto Health Care System, Palo Alto, CA, United States
| |
Collapse
|
5
|
Specific miRNA and Gene Deregulation Characterize the Increased Angiogenic Remodeling of Thoracic Aneurysmatic Aortopathy in Marfan Syndrome. Int J Mol Sci 2020; 21:ijms21186886. [PMID: 32961817 PMCID: PMC7555983 DOI: 10.3390/ijms21186886] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/17/2020] [Accepted: 09/19/2020] [Indexed: 12/11/2022] Open
Abstract
Marfan syndrome (MFS) is a connective tissue disease caused by mutations in the FBN1 gene, leading to alterations in the extracellular matrix microfibril assembly and the early formation of thoracic aorta aneurysms (TAAs). Non-genetic TAAs share many clinico-pathological aspects with MFS and deregulation of some microRNAs (miRNAs) has been demonstrated to be involved in the progression of TAA. In this study, 40 patients undergoing elective ascending aorta surgery were enrolled to compare TAA histomorphological features, miRNA profile and related target genes in order to find specific alterations that may explain the earlier and more severe clinical outcomes in MFS patients. Histomorphological, ultrastructural and in vitro studies were performed in order to compare aortic wall features of MFS and non-MFS TAA. MFS displayed greater glycosaminoglycan accumulation and loss/fragmentation of elastic fibers compared to non-MFS TAA. Immunohistochemistry revealed increased CD133+ angiogenic remodeling, greater MMP-2 expression, inflammation and smooth muscle cell (SMC) turnover in MFS TAA. Cultured SMCs from MFS confirmed higher turnover and α-smooth muscle actin expression compared with non-MFS TAA. Moreover, twenty-five miRNAs, including miR-26a, miR-29, miR-143 and miR-145, were found to be downregulated and only miR-632 was upregulated in MFS TAA in vivo. Bioinformatics analysis revealed that some deregulated miRNAs in MFS TAA are implicated in cell proliferation, extracellular matrix structure/function and TGFβ signaling. Finally, gene analysis showed 28 upregulated and seven downregulated genes in MFS TAA, some of them belonging to the CDH1/APC and CCNA2/TP53 signaling pathways. Specific miRNA and gene deregulation characterized the aortopathy of MFS and this was associated with increased angiogenic remodeling, likely favoring the early and more severe clinical outcomes, compared to non-MFS TAA. Our findings provide new insights concerning the pathogenetic mechanisms of MFS TAA; further investigation is needed to confirm if these newly identified specific deregulated miRNAs may represent potential therapeutic targets to counteract the rapid progression of MFS aortopathy.
Collapse
|
6
|
Sato T, Arakawa M, Tashima Y, Tsuboi E, Burdon G, Trojan J, Koyano T, Youn YN, Penov K, Pedroza AJ, Shabazzi M, Palmon I, Nguyen MN, Connolly AJ, Yamaguchi A, Fischbein MP. Statins Reduce Thoracic Aortic Aneurysm Growth in Marfan Syndrome Mice via Inhibition of the Ras-Induced ERK (Extracellular Signal-Regulated Kinase) Signaling Pathway. J Am Heart Assoc 2019; 7:e008543. [PMID: 30571378 PMCID: PMC6404178 DOI: 10.1161/jaha.118.008543] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Background Statins reduce aneurysm growth in mouse models of Marfan syndrome, although the mechanism is unknown. In addition to reducing cholesterol, statins block farnesylation and geranylgeranylation, which participate in membrane‐bound G‐protein signaling, including Ras. We dissected the prenylation pathway to define the effect of statins on aneurysm reduction. Methods and Results Fbn1C1039G/+ mice were treated with (1) pravastatin (HMG‐CoA [3‐hydroxy‐3‐methylglutaryl coenzyme A] reductase inhibitor), (2) manumycin A (MA; FPT inhibitor), (3) perillyl alcohol (GGPT1 and ‐2 inhibitor), or (4) vehicle control from age 4 to 8 weeks and euthanized at 12 weeks. Histological characterization was performed. Protein analysis was completed on aortic specimens to measure ERK (extracellular signal‐regulated kinase) signaling. In vitro Fbn1C1039G/+ aortic smooth muscle cells were utilized to measure Ras‐dependent ERK signaling and MMP (matrix metalloproteinase) activity. Pravastatin and MA significantly reduced aneurysm growth compared with vehicle control (n=8 per group). In contrast, PA did not significantly decrease aneurysm size. Histology illustrated reduced elastin breakdown in MA‐treated mice compared with vehicle control (n=5 per group). Although elevated in control Marfan mice, both phosphorylated c‐Raf and phosphorylated ERK1/2 were significantly reduced in MA‐treated mice (4–5 per group). In vitro smooth muscle cell studies confirmed phosphorylated cRaf and phosphorylated ERK1/2 signaling was elevated in Fbn1C1039G/+ smooth muscle cells (n=5 per group). Fbn1C1039G/+ smooth muscle cell Ras‐dependent ERK signaling and MMP activity were reduced following MA treatment (n=5 per group). Corroborating in vitro findings, MMP activity was also decreased in pravastatin‐treated mice. Conclusions Aneurysm reduction in Fbn1C1039G/+ mice following pravastatin and MA treatment was associated with a decrease in Ras‐dependent ERK signaling. MMP activity can be reduced by diminishing Ras signaling.
Collapse
Affiliation(s)
- Tetsuya Sato
- 1 Department of Cardiothoracic Surgery Stanford University Stanford CA.,2 Department of Cardiovascular Surgery Jichi Medical University Saitama Medical Center Saitama Japan
| | - Mamoru Arakawa
- 1 Department of Cardiothoracic Surgery Stanford University Stanford CA.,2 Department of Cardiovascular Surgery Jichi Medical University Saitama Medical Center Saitama Japan
| | - Yasushi Tashima
- 1 Department of Cardiothoracic Surgery Stanford University Stanford CA.,2 Department of Cardiovascular Surgery Jichi Medical University Saitama Medical Center Saitama Japan
| | - Eitoshi Tsuboi
- 1 Department of Cardiothoracic Surgery Stanford University Stanford CA.,4 Department of Cardiovascular Surgery Iwaki Kyoritsu General Hospital Fukushima Japan
| | - Grayson Burdon
- 1 Department of Cardiothoracic Surgery Stanford University Stanford CA
| | - Jeffrey Trojan
- 1 Department of Cardiothoracic Surgery Stanford University Stanford CA
| | - Tiffany Koyano
- 1 Department of Cardiothoracic Surgery Stanford University Stanford CA
| | - Young-Nam Youn
- 1 Department of Cardiothoracic Surgery Stanford University Stanford CA.,3 Division of Cardiovascular Surgery Severance Cardiovascular Hospital Yonsei University College of Medicine Seoul Korea
| | - Kiril Penov
- 1 Department of Cardiothoracic Surgery Stanford University Stanford CA.,5 Department of Cardiac Surgery Heart Center Leipzig University of Leipzig Germany
| | - Albert J Pedroza
- 1 Department of Cardiothoracic Surgery Stanford University Stanford CA
| | - Mohammad Shabazzi
- 1 Department of Cardiothoracic Surgery Stanford University Stanford CA
| | - Itai Palmon
- 1 Department of Cardiothoracic Surgery Stanford University Stanford CA
| | - Marie Noel Nguyen
- 1 Department of Cardiothoracic Surgery Stanford University Stanford CA
| | | | - Atsushi Yamaguchi
- 2 Department of Cardiovascular Surgery Jichi Medical University Saitama Medical Center Saitama Japan
| | | |
Collapse
|
7
|
Su Y, Li Q, Zheng Z, Wei X, Hou P. Integrative bioinformatics analysis of miRNA and mRNA expression profiles and identification of associated miRNA-mRNA network in aortic dissection. Medicine (Baltimore) 2019; 98:e16013. [PMID: 31192949 PMCID: PMC6587623 DOI: 10.1097/md.0000000000016013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Aortic dissection (AD) is one of the most lethal cardiovascular diseases. The aim of this study was to identify core genes and pathways revealing pathogenesis in AD. METHODS We screened differentially expressed mRNAs and miRNAs using mRNA and miRNA expression profile data of AD from Gene Expression Omnibus. Then functional and pathway enrichment analyses of differential expression genes (DEGs) was performed utilizing the database for annotation, visualization, and integrated discovery (DAVID). Target genes with differential expression miRNAs (DEMIs) were predicted using the miRWalk database, and the intersection between these predictions and DEGs was selected as differentially expressed miRNA-target genes. In addition, a protein-protein interaction (PPI) network and miRNA-mRNA regulatory network were constructed. RESULTS In total, 130 DEGs and 47 DEMIs were identified from mRNA and miRNA microarray, respectively, and 45 DEGs were DEMI-target genes. The PPI and miRNA-mRNA network included 79 node genes and 74 node genes, respectively, while 23 hub genes and 2 hub miRNAs were identified. The DEGs, PPI and modules differential expression miRNA-target genes were all mainly enriched in cell cycle, cell proliferation and cell apoptosis signaling pathways. CONCLUSION Taken above, the study reveals some candidate genes and pathways potentially involving molecular mechanisms of AD. These findings provide a new insight for research and treatment of AD.
Collapse
|
8
|
Wagner AH, Zaradzki M, Arif R, Remes A, Müller OJ, Kallenbach K. Marfan syndrome: A therapeutic challenge for long-term care. Biochem Pharmacol 2019; 164:53-63. [PMID: 30926475 DOI: 10.1016/j.bcp.2019.03.034] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 03/25/2019] [Indexed: 12/14/2022]
Abstract
Marfan syndrome (MFS) is an autosomal dominant genetic disorder caused by mutations in the fibrillin-1 gene. Acute aortic dissection is the leading cause of death in patients suffering from MFS and consequence of medial degeneration and aneurysm formation. In addition to its structural function in the formation of elastic fibers, fibrillin has a major role in keeping maintaining transforming growth factor β (TGF-β) in an inactive form. Dysfunctional fibrillin increases TGF-β bioavailability and concentration in the extracellular matrix, leading to activation of proinflammatory transcription factors. In turn, these events cause increased expression of matrix metalloproteinases and cytokines that control the migration and infiltration of inflammatory cells into the aorta. Moreover, TGF-β causes accumulation of reactive oxygen species leading to further degradation of elastin fibers. All these processes result in medial elastolysis, which increases the risk of vascular complications. Although MFS is a hereditary disease, symptoms and traits are usually not noticeable at birth. During childhood or adolescence affected individuals present with severe tissue weaknesses, especially in the aorta, heart, eyes, and skeleton. Considering this, even young patients should avoid activities that exert additional stress and pressure on the aorta and the cardiovascular system. Thus, if the diagnosis is made and prophylactic treatment is initiated in a timely fashion, MFS and its preliminary pathophysiologic vascular remodeling can be successfully ameliorated reducing the risk of life-threatening complications. This commentary focuses on new research opportunities and molecular findings on MFS, discusses future challenges and possible long-term therapies.
Collapse
Affiliation(s)
- A H Wagner
- Institute of Physiology and Pathophysiology, Division of Cardiovascular Physiology, Heidelberg University, Germany.
| | - M Zaradzki
- Department of Cardiac Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - R Arif
- Department of Cardiac Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - A Remes
- Department of Internal Medicine III, University Hospital Kiel, Kiel, Germany
| | - O J Müller
- Department of Internal Medicine III, University Hospital Kiel, Kiel, Germany
| | - K Kallenbach
- INCCI HaerzZenter, Department of Cardiac Surgery, Luxembourg, Luxembourg
| |
Collapse
|
9
|
Abstract
Abdominal aortic aneurysm (AAA) is a local dilatation of the abdominal aortic vessel wall and is among the most challenging cardiovascular diseases as without urgent surgical intervention, ruptured AAA has a mortality rate of >80%. Most patients present acutely after aneurysm rupture or dissection from a previously asymptomatic condition and are managed by either surgery or endovascular repair. Patients usually are old and have other concurrent diseases and conditions, such as diabetes mellitus, obesity, and hypercholesterolemia making surgical intervention more difficult. Collectively, these issues have driven the search for alternative methods of diagnosing, monitoring, and treating AAA using therapeutics and less invasive approaches. Noncoding RNAs-short noncoding RNAs (microRNAs) and long-noncoding RNAs-are emerging as new fundamental regulators of gene expression. Researchers and clinicians are aiming at targeting these microRNAs and long noncoding RNAs and exploit their potential as clinical biomarkers and new therapeutic targets for AAAs. While the role of miRNAs in AAA is established, studies on long-noncoding RNAs are only beginning to emerge, suggesting their important yet unexplored role in vascular physiology and disease. Here, we review the role of noncoding RNAs and their target genes focusing on their role in AAA. We also discuss the animal models used for mechanistic understanding of AAA. Furthermore, we discuss the potential role of microRNAs and long noncoding RNAs as clinical biomarkers and therapeutics.
Collapse
Affiliation(s)
- Sandeep Kumar
- Wallace H. Coulter Department of Biomedical Engineering,
Emory University and Georgia Institute of Technology, Atlanta, GA, USA
| | - Reinier A. Boon
- Institute for Cardiovascular Regeneration, Center of
Molecular Medicine, Goethe University, Frankfurt, Germany
- Department of Physiology, Amsterdam Cardiovascular
Sciences, VU University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, The
Netherlands
- German Center of Cardiovascular Research DZHK, Frankfurt,
Germany
| | - Lars Maegdefessel
- Department of Medicine, Karolinska Institute, Stockholm,
Sweden
- Department of Vascular and Endovascular Surgery, Technical
University Munich, Munich, Germany
- German Center for Cardiovascular Research DZHK, Munich,
Germany
| | - Stefanie Dimmeler
- Institute for Cardiovascular Regeneration, Center of
Molecular Medicine, Goethe University, Frankfurt, Germany
- German Center of Cardiovascular Research DZHK, Frankfurt,
Germany
- Corresponding authors: Hanjoong Jo, PhD, John and Jan Portman
Professor, Wallace H. Coulter Department of Biomedical Engineering, Emory
University and Georgia Institute of Technology, 1760 Haygood Drive, Atlanta, GA
30322, , Stefanie Dimmeler, PhD, Institute for
Cardiovascular Regeneration, Centre of Molecular Medicine, Goethe University
Frankfurt, Theodor Stern Kai 7, 60590, Frankfurt, Germany,
| | - Hanjoong Jo
- Wallace H. Coulter Department of Biomedical Engineering,
Emory University and Georgia Institute of Technology, Atlanta, GA, USA
- Division of Cardiology, Emory University, Atlanta, GA,
USA
- Corresponding authors: Hanjoong Jo, PhD, John and Jan Portman
Professor, Wallace H. Coulter Department of Biomedical Engineering, Emory
University and Georgia Institute of Technology, 1760 Haygood Drive, Atlanta, GA
30322, , Stefanie Dimmeler, PhD, Institute for
Cardiovascular Regeneration, Centre of Molecular Medicine, Goethe University
Frankfurt, Theodor Stern Kai 7, 60590, Frankfurt, Germany,
| |
Collapse
|
10
|
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
|
11
|
Portelli SS, Robertson EN, Malecki C, Liddy KA, Hambly BD, Jeremy RW. Epigenetic influences on genetically triggered thoracic aortic aneurysm. Biophys Rev 2018; 10:1241-1256. [PMID: 30267337 PMCID: PMC6233334 DOI: 10.1007/s12551-018-0460-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Accepted: 09/17/2018] [Indexed: 12/14/2022] Open
Abstract
Genetically triggered thoracic aortic aneurysms (TAAs) account for 30% of all TAAs and can result in early morbidity and mortality in affected individuals. Epigenetic factors are now recognised to influence the phenotype of many genetically triggered conditions and have become an area of interest because of the potential for therapeutic manipulation. Major epigenetic modulators include DNA methylation, histone modification and non-coding RNA. This review examines epigenetic modulators that have been significantly associated with genetically triggered TAAs and their potential utility for translation to clinical practice.
Collapse
Affiliation(s)
- Stefanie S Portelli
- Discipline of Pathology and Bosch Institute, The University of Sydney, Sydney, NSW, 2006, Australia.
| | - Elizabeth N Robertson
- Discipline of Pathology and Bosch Institute, The University of Sydney, Sydney, NSW, 2006, Australia
- Cardiology Department, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Cassandra Malecki
- Discipline of Pathology and Bosch Institute, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Kiersten A Liddy
- Discipline of Pathology and Bosch Institute, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Brett D Hambly
- Discipline of Pathology and Bosch Institute, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Richmond W Jeremy
- Discipline of Pathology and Bosch Institute, The University of Sydney, Sydney, NSW, 2006, Australia
- Cardiology Department, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| |
Collapse
|
12
|
Takeda N, Hara H, Fujiwara T, Kanaya T, Maemura S, Komuro I. TGF-β Signaling-Related Genes and Thoracic Aortic Aneurysms and Dissections. Int J Mol Sci 2018; 19:ijms19072125. [PMID: 30037098 PMCID: PMC6073540 DOI: 10.3390/ijms19072125] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 07/13/2018] [Accepted: 07/16/2018] [Indexed: 12/29/2022] Open
Abstract
Transforming growth factor-β (TGF)-β signaling plays a crucial role in the development and maintenance of various organs, including the vasculature. Accordingly, the mutations in TGF-β signaling pathway-related genes cause heritable disorders of the connective tissue, such as Marfan syndrome (MFS), Loeys-Dietz syndrome (LDS), and Shprintzen-Goldberg syndrome (SGS), and these syndromes may affect skeletal, ocular, pulmonary, and cardiovascular systems. Aortic root aneurysms are common problems that can result in aortic dissection or rupture, which is the leading cause of sudden death in the natural history of MFS and LDS, and recent improvements in surgical treatment have improved life expectancy. However, there is currently no genotype-specific medical treatment. Accumulating evidence suggest that not only structural weakness of connective tissue but also increased TGF-β signaling contributes to the complicated pathogenesis of aortic aneurysm formation, but a comprehensive understanding of governing molecular mechanisms remains lacking. Inhibition of angiotensin II receptor signaling and endothelial dysfunction have gained attention as a possible MFS treatment strategy, but interactions with TGF-β signaling remain elusive. Heterozygous loss-of-function mutations in TGF-β receptors 1 and 2 (TGFBR1 and TGFBR2) cause LDS, but TGF-β signaling is activated in the aorta (referred to as the TGF-β paradox) by mechanisms yet to be elucidated. In this review, we present and discuss the current understanding of molecular mechanisms responsible for aortopathies of MFS and related disorders.
Collapse
Affiliation(s)
- Norifumi Takeda
- Department of Cardiovascular Medicine, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan.
| | - Hironori Hara
- Department of Cardiovascular Medicine, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan.
| | - Takayuki Fujiwara
- Department of Cardiovascular Medicine, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan.
| | - Tsubasa Kanaya
- Department of Cardiovascular Medicine, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan.
| | - Sonoko Maemura
- Department of Cardiovascular Medicine, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan.
| | - Issei Komuro
- Department of Cardiovascular Medicine, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan.
| |
Collapse
|
13
|
Parker SJ, Stotland A, MacFarlane E, Wilson N, Orosco A, Venkatraman V, Madrid K, Gottlieb R, Dietz HC, Van Eyk JE. Proteomics reveals Rictor as a noncanonical TGF-β signaling target during aneurysm progression in Marfan mice. Am J Physiol Heart Circ Physiol 2018; 315:H1112-H1126. [PMID: 30004239 DOI: 10.1152/ajpheart.00089.2018] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The objective of the present study was to 1) analyze the ascending aortic proteome within a mouse model of Marfan syndrome (MFS; Fbn1C1041G/+) at early and late stages of aneurysm and 2) subsequently test a novel hypothesis formulated on the basis of this unbiased proteomic screen that links changes in integrin composition to transforming growth factor (TGF)-β-dependent activation of the rapamycin-independent component of mammalian target of rapamycin (Rictor) signaling pathway. Ingenuity Pathway Analysis of over 1,000 proteins quantified from the in vivo MFS mouse aorta by data-independent acquisition mass spectrometry revealed a predicted upstream regulator, Rictor, that was selectively activated in aged MFS mice. We validated this pattern of Rictor activation in vivo by Western blot analysis for phosphorylation on Thr1135 in a separate cohort of mice and showed in vitro that TGF-β activates Rictor in an integrin-linked kinase-dependent manner in cultured aortic vascular smooth muscle cells. Expression of β3-integrin was upregulated in the aged MFS aorta relative to young MFS mice and wild-type mice. We showed that β3-integrin expression and activation modulated TGF-β-induced Rictor phosphorylation in vitro, and this signaling effect was associated with an altered vascular smooth muscle cell proliferative-migratory and metabolic in vitro phenotype that parallels the in vivo aneurysm phenotype in MFS. These results reveal that Rictor is a novel, context-dependent, noncanonical TGF-β signaling effector with potential pathogenic implications in aortic aneurysm. NEW & NOTEWORTHY We present the most comprehensive quantitative analysis of the ascending aortic aneurysm proteome in Marfan syndrome to date resulting in novel and potentially wide-reaching findings that expression and signaling by β3-integrin constitute a modulator of transforming growth factor-β-induced rapamycin-independent component of mammalian target of rapamycin (Rictor) signaling and physiology in aortic vascular smooth muscle cells.
Collapse
Affiliation(s)
- Sarah J Parker
- Advanced Clinical Biosystems Research Institute, The Smidt Heart Institute, Cedars-Sinai Medical Center , Los Angeles, California.,Institute for Genetic Medicine, Johns Hopkins University , Baltimore, Maryland
| | - Aleksandr Stotland
- Molecular Cardiobiology, The Heart Institute, Cedars-Sinai Medical Center , Los Angeles, California
| | - Elena MacFarlane
- Institute for Genetic Medicine, Johns Hopkins University , Baltimore, Maryland
| | - Nicole Wilson
- Institute for Genetic Medicine, Johns Hopkins University , Baltimore, Maryland
| | - Amanda Orosco
- Advanced Clinical Biosystems Research Institute, The Smidt Heart Institute, Cedars-Sinai Medical Center , Los Angeles, California
| | - Vidya Venkatraman
- Advanced Clinical Biosystems Research Institute, The Smidt Heart Institute, Cedars-Sinai Medical Center , Los Angeles, California.,Institute for Genetic Medicine, Johns Hopkins University , Baltimore, Maryland
| | - Kyle Madrid
- Biomedical Sciences, Cedars-Sinai Medical Center , Los Angeles, California
| | - Roberta Gottlieb
- Molecular Cardiobiology, The Heart Institute, Cedars-Sinai Medical Center , Los Angeles, California
| | - Harry C Dietz
- Institute for Genetic Medicine, Johns Hopkins University , Baltimore, Maryland.,Howard Hughes Medical Institute , Chevy Chase, Maryland
| | - Jennifer E Van Eyk
- Advanced Clinical Biosystems Research Institute, The Smidt Heart Institute, Cedars-Sinai Medical Center , Los Angeles, California.,Institute for Genetic Medicine, Johns Hopkins University , Baltimore, Maryland
| |
Collapse
|
14
|
Qi YF, Shu C, Xiao ZX, Luo MY, Fang K, Guo YY, Zhang WB, Yue J. Post-Transcriptional Control of Tropoelastin in Aortic Smooth Muscle Cells Affects Aortic Dissection Onset. Mol Cells 2018; 41:198-206. [PMID: 29487278 PMCID: PMC5881093 DOI: 10.14348/molcells.2018.2193] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Revised: 12/15/2017] [Accepted: 12/21/2017] [Indexed: 11/27/2022] Open
Abstract
Aortic dissection (AD) is a catastrophic disease with high mortality and morbidity, characterized with fragmentation of elastin and loss of smooth muscle cells. Although AD has been largely attributable to polymorphisms defect in the elastin-coding gene, tropoelastin (TE), other undermined factors also appear to play roles in AD onset. Here, we investigated the effects of post-transcriptional control of TE by microRNAs (miRNAs) on elastin levels in aortic smooth muscle cells (ASMC). We found that miR-144-3p is a miRNA that targets TE mRNA in both human and mouse. Bioinformatics analyses and dual luciferase reporter assay showed that miR-144-3p inhibited protein translation of TE, through binding to the 3'-UTR of the TE mRNA. Interestingly, higher miR-144-3p levels and lower TE were detected in the ASMC obtained from AD patients, compared to those from non-AD controls. In a mouse model for human AD, infusion of adeno-associated viruses (serotype 6) carrying antisense for miR-144-3p (as-miR-144-3p) under CAG promoter significantly reduced the incidence and severity of AD, seemingly through enhancement of TE levels in ASMC. Thus, our data suggest an essential role of miR-144-3p on the pathogenesis of AD.
Collapse
Affiliation(s)
- You-Fei Qi
- Department of Vascular Surgery, the Second Xiang-ya Hospital, Central South University, Changsha 410011,
China
- Department of Vascular Surgery, Hainan General Hospital, Haikou 570311,
China
| | - Chang Shu
- Department of Vascular Surgery, the Second Xiang-ya Hospital, Central South University, Changsha 410011,
China
- Center of Vascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037,
China
| | - Zhan-Xiang Xiao
- Department of Vascular Surgery, Hainan General Hospital, Haikou 570311,
China
| | - Ming-Yao Luo
- Center of Vascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037,
China
| | - Kun Fang
- Center of Vascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037,
China
| | - Yuan-Yuan Guo
- Department of Vascular Surgery, Fuwai Yunnan Cardiovascular Hospital, Kunming 650032,
China
| | - Wen-Bo Zhang
- Department of Vascular Surgery, Hainan General Hospital, Haikou 570311,
China
| | - Jie Yue
- Department of Vascular Surgery, Hainan General Hospital, Haikou 570311,
China
| |
Collapse
|
15
|
Ramirez F, Caescu C, Wondimu E, Galatioto J. Marfan syndrome; A connective tissue disease at the crossroads of mechanotransduction, TGFβ signaling and cell stemness. Matrix Biol 2017; 71-72:82-89. [PMID: 28782645 DOI: 10.1016/j.matbio.2017.07.004] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 07/27/2017] [Accepted: 07/27/2017] [Indexed: 12/16/2022]
Abstract
Mutations in fibrillin-1 cause Marfan syndrome (MFS), the most common heritable disorder of connective tissue. Fibrillin-1 assemblies (microfibrils and elastic fibers) represent a unique dual-function component of the architectural matrix. The first role is structural for they endow tissues with tensile strength and elasticity, transmit forces across them and demarcate functionally discrete areas within them. The second role is instructive in that these macroaggregates modulate a large variety of sub-cellular processes by interacting with mechanosensors, and integrin and syndecan receptors, and by modulating the bioavailability of local TGFβ signals. The multifunctional, tissue-specific nature of fibrillin-1 assemblies is reflected in the variety of clinical manifestations and disease mechanisms associated with the MFS phenotype. Characterization of mice with ubiquitous or cell type-restricted fibrillin-1 deficiency has unraveled some pathophysiological mechanisms associated with the MFS phenotype, such as altered mechanotransduction in the heart, dysregulated TGFβ signaling in the ascending aorta and perturbed stem cell fate in the bone marrow. In each case, potential druggable targets have also been identified. However, the finding that distinct disease mechanisms underlie different organ abnormalities strongly argues for developing multi-drug strategies to mitigate or even prevent both life-threatening and morbid manifestations in pediatric and adult MFS patients.
Collapse
Affiliation(s)
- Francesco Ramirez
- Department of Pharmacological Sciences and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States.
| | - Cristina Caescu
- Department of Pharmacological Sciences and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - Elisabeth Wondimu
- Department of Pharmacological Sciences and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - Josephine Galatioto
- Department of Pharmacological Sciences and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| |
Collapse
|
16
|
Okamura H, Emrich F, Trojan J, Chiu P, Dalal AR, Arakawa M, Sato T, Penov K, Koyano T, Pedroza A, Connolly AJ, Rabinovitch M, Alvira C, Fischbein MP. Long-term miR-29b suppression reduces aneurysm formation in a Marfan mouse model. Physiol Rep 2017; 5:5/8/e13257. [PMID: 28455451 PMCID: PMC5408287 DOI: 10.14814/phy2.13257] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 03/23/2017] [Indexed: 11/24/2022] Open
Abstract
Aortic root aneurysm formation and subsequent dissection and/or rupture remain the leading cause of death in patients with Marfan syndrome. Our laboratory has reported that miR‐29b participates in aortic root/ascending aorta extracellular matrix remodeling during early aneurysm formation in Fbn1C1039G/+ Marfan mice. Herein, we sought to determine whether miR‐29b suppression can reduce aneurysm formation long‐term. Fbn1C1039G/+ Marfan mice were treated with retro‐orbital LNA‐anti‐miR‐29b inhibitor or scrambled‐control‐miR before aneurysms develop either (1) a single dose prenatally (pregnant Fbn1C1039G/+ mice at 14.5 days post‐coitum) (n = 8–10, each group) or (2) postnatally every other week, from 2 to 22 weeks of age, and sacrificed at 24 weeks (n = 8–10, each group). To determine if miR‐29b blockade was beneficial even after aneurysms develop, a third group of animals were treated every other week, starting at 8 weeks of age, until sacrificed (n = 4–6, each group). miR‐29b inhibition resulted in aneurysm reduction, increased elastogenesis, decreased matrix metalloproteinase activity and decreased elastin breakdown. Prenatal LNA‐anti‐miR‐29b inhibitor treatment decreased aneurysm formation up to age 32 weeks, whereas postnatal treatment was effective up to 16 weeks. miR‐29b blockade did not slow aortic growth once aneurysms already developed. Systemic miR‐29b inhibition significantly reduces aneurysm development long‐term in a Marfan mouse model. Drug administration during aortic wall embryologic development appears fundamental. miR‐29b suppression could be a potential therapeutic target for reducing aneurysm formation in Marfan syndrome patients.
Collapse
Affiliation(s)
- Homare Okamura
- Department of Cardiothoracic Surgery, Stanford University, Stanford, California
| | - Fabian Emrich
- Department of Cardiothoracic Surgery, Stanford University, Stanford, California
| | - Jeffrey Trojan
- Department of Cardiothoracic Surgery, Stanford University, Stanford, California
| | - Peter Chiu
- Department of Cardiothoracic Surgery, Stanford University, Stanford, California
| | - Alex R Dalal
- Department of Cardiothoracic Surgery, Stanford University, Stanford, California
| | - Mamoru Arakawa
- Department of Cardiothoracic Surgery, Stanford University, Stanford, California
| | - Tetsuya Sato
- Department of Cardiothoracic Surgery, Stanford University, Stanford, California
| | - Kiril Penov
- Department of Cardiothoracic Surgery, Stanford University, Stanford, California
| | - Tiffany Koyano
- Department of Cardiothoracic Surgery, Stanford University, Stanford, California
| | - Albert Pedroza
- Department of Cardiothoracic Surgery, Stanford University, Stanford, California
| | | | | | - Cristina Alvira
- Department of Pediatrics, Stanford University, Stanford, California
| | - Michael P Fischbein
- Department of Cardiothoracic Surgery, Stanford University, Stanford, California
| |
Collapse
|