1
|
Martin-Blazquez A, Martin-Lorenzo M, Santiago-Hernandez A, Heredero A, Donado A, Lopez JA, Anfaiha-Sanchez M, Ruiz-Jimenez R, Esteban V, Vazquez J, Aldamiz-Echevarria G, Alvarez-Llamas G. Analysis of Vascular Smooth Muscle Cells from Thoracic Aortic Aneurysms Reveals DNA Damage and Cell Cycle Arrest as Hallmarks in Bicuspid Aortic Valve Patients. J Proteome Res 2024; 23:3012-3024. [PMID: 38594816 PMCID: PMC11301675 DOI: 10.1021/acs.jproteome.3c00649] [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: 10/04/2023] [Revised: 02/26/2024] [Accepted: 03/24/2024] [Indexed: 04/11/2024]
Abstract
Thoracic aortic aneurysm (TAA) is mainly sporadic and with higher incidence in the presence of a bicuspid aortic valve (BAV) for unknown reasons. The lack of drug therapy to delay TAA progression lies in the limited knowledge of pathophysiology. We aimed to identify the molecular hallmarks that differentiate the aortic dilatation associated with BAV and tricuspid aortic valve (TAV). Aortic vascular smooth muscle cells (VSMCs) isolated from sporadic TAA patients with BAV or TAV were analyzed by mass spectrometry. DNA oxidative damage assay and cell cycle profiling were performed in three independent cohorts supporting proteomics data. The alteration of secreted proteins was confirmed in plasma. Stress phenotype, oxidative stress, and enhanced DNA damage response (increased S-phase arrest and apoptosis) were found in BAV-TAA patients. The increased levels of plasma C1QTNF5, LAMA2, THSB3, and FAP confirm the enhanced stress in BAV-TAA. Plasma FAP and BGN point to an increased inflammatory condition in TAV. The arterial wall of BAV patients shows a limited capacity to counteract drivers of sporadic TAA. The molecular pathways identified support the need of differential molecular diagnosis and therapeutic approaches for BAV and TAV patients, showing specific markers in plasma which may serve to monitor therapy efficacy.
Collapse
Affiliation(s)
- Ariadna Martin-Blazquez
- Immunology
Department, IIS-Fundación Jiménez
Díaz, Fundación Jiménez Díaz Hospital-UAM, 28040 Madrid, Spain
| | - Marta Martin-Lorenzo
- Immunology
Department, IIS-Fundación Jiménez
Díaz, Fundación Jiménez Díaz Hospital-UAM, 28040 Madrid, Spain
| | | | - Angeles Heredero
- Cardiac
Surgery Service, Fundación Jiménez
Díaz Hospital-UAM, 28040 Madrid, Spain
| | - Alicia Donado
- Cardiac
Surgery Service, Fundación Jiménez
Díaz Hospital-UAM, 28040 Madrid, Spain
| | - Juan A Lopez
- Laboratory
of Cardiovascular Proteomics, Centro Nacional
de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
- CIBER
de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Miriam Anfaiha-Sanchez
- Immunology
Department, IIS-Fundación Jiménez
Díaz, Fundación Jiménez Díaz Hospital-UAM, 28040 Madrid, Spain
| | - Rocio Ruiz-Jimenez
- Immunology
Department, IIS-Fundación Jiménez
Díaz, Fundación Jiménez Díaz Hospital-UAM, 28040 Madrid, Spain
| | - Vanesa Esteban
- Department
of Allergy and Immunology, IIS-Fundación
Jiménez Díaz, Fundación Jiménez Díaz
Hospital-UAM, 28040 Madrid, Spain
- Faculty
of Medicine and Biomedicine, Alfonso X El
Sabio University, 28691 Madrid, Spain
| | - Jesus Vazquez
- Laboratory
of Cardiovascular Proteomics, Centro Nacional
de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
- CIBER
de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | | | - Gloria Alvarez-Llamas
- Immunology
Department, IIS-Fundación Jiménez
Díaz, Fundación Jiménez Díaz Hospital-UAM, 28040 Madrid, Spain
- RICORS2040, Fundación Jiménez Díaz, 28040 Madrid, Spain
- Department
of Biochemistry and Molecular Biology, Complutense
University, 28040 Madrid, Spain
| |
Collapse
|
2
|
Rega S, Farina F, Bouhuis S, de Donato S, Chiesa M, Poggio P, Cavallotti L, Bonalumi G, Giambuzzi I, Pompilio G, Perrucci GL. Multi-omics in thoracic aortic aneurysm: the complex road to the simplification. Cell Biosci 2023; 13:131. [PMID: 37475058 DOI: 10.1186/s13578-023-01080-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 07/05/2023] [Indexed: 07/22/2023] Open
Abstract
BACKGROUND Thoracic aortic aneurysm (TAA) is a serious condition that affects the aorta, characterized by the dilation of its first segment. The causes of TAA (e.g., age, hypertension, genetic syndromes) are heterogeneous and contribute to the weakening of the aortic wall. This complexity makes treating this life-threatening aortopathy challenging, as there are currently no etiological therapy available, and pharmacological strategies, aimed at avoiding surgical aortic replacement, are merely palliative. Recent studies on novel therapies for TAA have focused on identifying biological targets and etiological mechanisms of the disease by using advanced -omics techniques, including epigenomics, transcriptomics, proteomics, and metabolomics approaches. METHODS This review presents the latest findings from -omics approaches and underscores the importance of integrating multi-omics data to gain more comprehensive understanding of TAA. RESULTS Literature suggests that the alterations in TAA mediators frequently involve members of pro-fibrotic process (i.e., TGF-β signaling pathways) or proteins associated with cell/extracellular structures (e.g., aggrecans). Further analyses often reported the importance in TAA of processes as inflammation (PCR, CD3, leukotriene compounds), oxidative stress (chromatin OXPHOS, fatty acids), mitochondrial respiration and glycolysis/gluconeogenesis (e.g., PPARs and HIF1a). Of note, more recent metabolomics studies added novel molecular markers to the list of TAA-specific detrimental mediators (proteoglycans). CONCLUSION It is increasingly clear that integrating data from different -omics branches, along with clinical data, is essential as well as complicated both to reveal hidden relevant information and to address complex diseases such as TAA. Importantly, recent progresses in metabolomics highlighted novel potential and unprecedented marks in TAA diagnosis and therapy.
Collapse
Affiliation(s)
- Sara Rega
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino IRCCS, Milan, Italy
- Unit for the Study of Aortic, Valvular and Coronary Pathologies, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Floriana Farina
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximillians-Universität (LMU) München, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Silvia Bouhuis
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Silvia de Donato
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Mattia Chiesa
- Bioinformatics and Artificial Intelligence Facility, Centro Cardiologico Monzino IRCCS, Milan, Italy
- Department of Electronics, Information and Biomedical Engineering, Politecnico Di Milano, Milan, Italy
| | - Paolo Poggio
- Unit for the Study of Aortic, Valvular and Coronary Pathologies, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Laura Cavallotti
- Department of Cardiovascular Surgery, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Giorgia Bonalumi
- Department of Cardiovascular Surgery, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Ilaria Giambuzzi
- Department of Cardiovascular Surgery, Centro Cardiologico Monzino IRCCS, Milan, Italy
- Department of Clinical Sciences and Community Health, Università Degli Studi Di Milano, Milan, Italy
| | - Giulio Pompilio
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino IRCCS, Milan, Italy
- Department of Cardiovascular Surgery, Centro Cardiologico Monzino IRCCS, Milan, Italy
- Department of Biomedical, Surgical and Dental Sciences, Università Degli Studi Di Milano, Milan, Italy
| | - Gianluca L Perrucci
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino IRCCS, Milan, Italy.
| |
Collapse
|
3
|
Michel JB, Lagrange J, Regnault V, Lacolley P. Conductance Artery Wall Layers and Their Respective Roles in the Clearance Functions. Arterioscler Thromb Vasc Biol 2022; 42:e253-e272. [PMID: 35924557 DOI: 10.1161/atvbaha.122.317759] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Evolutionary organization of the arterial wall into layers occurred concomitantly with the emergence of a highly muscularized, pressurized arterial system that facilitates outward hydraulic conductance and mass transport of soluble substances across the arterial wall. Although colliding circulating cells disperse potential energy within the arterial wall, the different layers counteract this effect: (1) the endothelium ensures a partial barrier function; (2) the media comprises smooth muscle cells capable of endocytosis/phagocytosis; (3) the outer adventitia and perivascular adipocytic tissue are the final receptacles of convected substances. While the endothelium forms a physical and a biochemical barrier, the medial layer is avascular, relying on the specific permeability properties of the endothelium for metabolic support. Different components of the media interact with convected molecules: medial smooth muscle cells take up numerous molecules via scavenger receptors and are capable of phagocytosis of macro/micro particles. The outer layers-the highly microvascularized innervated adventitia and perivascular adipose tissue-are also involved in the clearance functions of the media: the adventitia is the seat of immune response development, inward angiogenesis, macromolecular lymphatic drainage, and neuronal stimulation. Consequently, the clearance functions of the arterial wall are physiologically essential, but also may favor the development of arterial wall pathologies. This review describes how the walls of large conductance arteries have acquired physiological clearance functions, how this is determined by the attributes of the endothelial barrier, governed by endocytic and phagocytic capacities of smooth muscle cells, impacting adventitial functions, and the role of these clearance functions in arterial wall diseases.
Collapse
|
4
|
Venisse L, François D, Madjène C, Brouwers E, de Raucourt E, Boulaftali Y, Declerck P, Arocas V, Bouton M. Novel ELISA for the specific detection of protease NEXIN-1 in human biological samples. Res Pract Thromb Haemost 2022; 6:e12756. [PMID: 35865733 PMCID: PMC9294866 DOI: 10.1002/rth2.12756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 04/19/2022] [Accepted: 05/03/2022] [Indexed: 12/03/2022] Open
Abstract
Introduction Serpin E2 or protease nexin-1 (PN-1) is a glycoprotein belonging to the serpin superfamily, whose function is closely linked to its ability to inhibit thrombin and proteases of the plasminergic system. Objectives In the absence of specific quantitative methods, an ELISA for the quantification of human PN-1 was characterized and used in biological fluids. Methods The ELISA for human PN-1 was developed using two monoclonal antibodies raised against human recombinant PN-1. PN-1 was quantified in plasma, serum, platelet secretion from controls and patients with hemophilia A and in conditioned medium of aortic tissue. Results A linear dose-response curve was observed between 2 and 35 ng/mL human PN-1. Intra- and interassay coefficients of variation were 6.2% and 11.1%, respectively. Assay recoveries of PN-1 added to biological samples were ≈95% in plasma, ≈97% in platelet reaction buffer, and ≈93% in RPMI cell culture medium. Levels of PN-1 secreted from activated human platelets from controls was similar to that of patients with hemophilia A. PN-1 could be detected in conditioned media of aneurysmal aorta but not in that of control aorta. Conclusion This is the first fully characterized ELISA for human serpin E2 level in biological fluids. It may constitute a relevant novel tool for further investigations on the pathophysiological role of serpin E2 in a variety of clinical studies.
Collapse
Affiliation(s)
- Laurence Venisse
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM U1148‐LVTSParisFrance
| | - Déborah François
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM U1148‐LVTSParisFrance
| | - Célina Madjène
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM U1148‐LVTSParisFrance
| | - Els Brouwers
- Laboratory for Therapeutic and Diagnostic Antibodies, Department of Pharmaceutical and Pharmacological SciencesKU LeuvenLeuvenBelgium
| | - Emmanuelle de Raucourt
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM U1148‐LVTSParisFrance
- Département d'HématologieHôpital BeaujonClichyFrance
- Centre de Traitement de l'Hémophilie Hôpital MignotLe ChesnayFrance
| | - Yacine Boulaftali
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM U1148‐LVTSParisFrance
| | - Paul Declerck
- Laboratory for Therapeutic and Diagnostic Antibodies, Department of Pharmaceutical and Pharmacological SciencesKU LeuvenLeuvenBelgium
| | - Véronique Arocas
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM U1148‐LVTSParisFrance
| | | |
Collapse
|
5
|
Jauhiainen S, Kiema M, Hedman M, Laakkonen JP. Large Vessel Cell Heterogeneity and Plasticity: Focus in Aortic Aneurysms. Arterioscler Thromb Vasc Biol 2022; 42:811-818. [PMID: 35587695 DOI: 10.1161/atvbaha.121.316237] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Smooth muscle cells and endothelial cells have a remarkable level of plasticity in vascular pathologies. In thoracic and abdominal aortic aneurysms, smooth muscle cells have been suggested to undergo phenotypic switching and to contribute to degradation of the aortic wall structure in response to, for example, inflammatory mediators, dysregulation of growth factor signaling or oxidative stress. Recently, endothelial-to-mesenchymal transition, and a clonal expansion of degradative smooth muscle cells and immune cells, as well as mesenchymal stem-like cells have been suggested to contribute to the progression of aortic aneurysms. What are the factors driving the aortic cell phenotype changes and how vascular flow, known to affect aortic wall structure and to be altered in aortic aneurysms, could affect aortic cell remodeling? In this review, we summarize the current literature on aortic cell heterogeneity and phenotypic switching in relation to changes in vascular flow and aortic wall structure in aortic aneurysms in clinical samples with special focus on smooth muscle and endothelial cells. The differences between thoracic and abdominal aortic aneurysms are discussed.
Collapse
Affiliation(s)
- Suvi Jauhiainen
- A.I. Virtanen Institute for Molecular Sciences (S.J., M.K., J.P.L.), University of Eastern Finland, Kuopio
| | - Miika Kiema
- A.I. Virtanen Institute for Molecular Sciences (S.J., M.K., J.P.L.), University of Eastern Finland, Kuopio
| | - Marja Hedman
- Institute of Clinical Medicine (M.H.), University of Eastern Finland, Kuopio
- Department of Clinical Radiology, Kuopio University Hospital, Finland (M.H.)
- Department of Heart and Thoracic Surgery, Kuopio University Hospital, Heart Center, Kuopio, Finland (M.H.)
| | - Johanna P Laakkonen
- A.I. Virtanen Institute for Molecular Sciences (S.J., M.K., J.P.L.), University of Eastern Finland, Kuopio
| |
Collapse
|
6
|
Rombouts KB, van Merrienboer TAR, Ket JCF, Bogunovic N, van der Velden J, Yeung KK. The role of vascular smooth muscle cells in the development of aortic aneurysms and dissections. Eur J Clin Invest 2022; 52:e13697. [PMID: 34698377 PMCID: PMC9285394 DOI: 10.1111/eci.13697] [Citation(s) in RCA: 103] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 09/12/2021] [Accepted: 10/11/2021] [Indexed: 12/30/2022]
Abstract
BACKGROUND Aortic aneurysms (AA) are pathological dilations of the aorta, associated with an overall mortality rate up to 90% in case of rupture. In addition to dilation, the aortic layers can separate by a tear within the layers, defined as aortic dissections (AD). Vascular smooth muscle cells (vSMC) are the predominant cell type within the aortic wall and dysregulation of vSMC functions contributes to AA and AD development and progression. However, since the exact underlying mechanism is poorly understood, finding potential therapeutic targets for AA and AD is challenging and surgery remains the only treatment option. METHODS In this review, we summarize current knowledge about vSMC functions within the aortic wall and give an overview of how vSMC functions are altered in AA and AD pathogenesis, organized per anatomical location (abdominal or thoracic aorta). RESULTS Important functions of vSMC in healthy or diseased conditions are apoptosis, phenotypic switch, extracellular matrix regeneration and degradation, proliferation and contractility. Stressors within the aortic wall, including inflammatory cell infiltration and (epi)genetic changes, modulate vSMC functions and cause disturbance of processes within vSMC, such as changes in TGF-β signalling and regulatory RNA expression. CONCLUSION This review underscores a central role of vSMC dysfunction in abdominal and thoracic AA and AD development and progression. Further research focused on vSMC dysfunction in the aortic wall is necessary to find potential targets for noninvasive AA and AD treatment options.
Collapse
Affiliation(s)
- Karlijn B Rombouts
- Department of Surgery, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center and AMC, Amsterdam, The Netherlands.,Department of Physiology, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center, Amsterdam, The Netherlands
| | - Tara A R van Merrienboer
- Department of Surgery, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center and AMC, Amsterdam, The Netherlands.,Department of Physiology, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center, Amsterdam, The Netherlands
| | | | - Natalija Bogunovic
- Department of Surgery, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center and AMC, Amsterdam, The Netherlands.,Department of Physiology, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center, Amsterdam, The Netherlands.,Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jolanda van der Velden
- Department of Physiology, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center, Amsterdam, The Netherlands
| | - Kak Khee Yeung
- Department of Surgery, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center and AMC, Amsterdam, The Netherlands.,Department of Physiology, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
7
|
Chuang HW, Hsia KT, Liao JB, Yeh CC, Kuo WT, Yang YF. SERPINE2 Overexpression Is Associated with Poor Prognosis of Urothelial Carcinoma. Diagnostics (Basel) 2021; 11:diagnostics11101928. [PMID: 34679626 PMCID: PMC8535068 DOI: 10.3390/diagnostics11101928] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/07/2021] [Accepted: 10/15/2021] [Indexed: 01/17/2023] Open
Abstract
Recent studies have reported that SERPINE2 contributes to the development of various cancers. However, its association with urothelial carcinoma (UC) remains unclear. In this study, data on urinary bladder UC (UBUC) cases from The Cancer Genome Atlas (TCGA) database were used to investigate the prognostic value of SERPINE2 mRNA expression. Then, SERPINE2 expression was analyzed with tissue microarrays constructed from 117 upper tract UC (UTUC) and 84 UBUC tissue specimens using immunohistochemical staining. Results were compared to clinicopathologic data by multivariate analysis. In the TCGA database, high SERPINE2 mRNA expression indicated a poor prognosis in patients with UBUC. Furthermore, Mann-Whitney U test showed that high SERPINE2 immunoexpression was significantly associated with adverse pathologic parameters including invasion, high grade, coexistence of UC in situ, and advanced pT stage (all p < 0.05, except for a marginal association with high-grade UBUC, p = 0.066). Kaplan-Meier analysis revealed that high SERPINE2 expression was associated with worse overall survival (OS; UTUC, p = 0.003; UBUC, p = 0.014) and disease-free survival (UTUC, p = 0.031; UBUC, p = 0.033). Moreover, multivariate analysis identified high SERPINE2 expression as an independent prognostic factor for OS (UTUC, p = 0.002; UBUC, p = 0.024). Taken together, our findings demonstrated that increased SERPINE2 expression is associated with adverse pathologic features and may serve as a prognostic biomarker for UC.
Collapse
Affiliation(s)
- Hao-Wen Chuang
- Department of Pathology and Laboratory Medicine, Kaohsiung Veterans General Hospital, Kaohsiung 813414, Taiwan; (H.-W.C.); (J.-B.L.); (C.-C.Y.)
- Institute of Oral Biology, School of Dentistry, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan;
| | - Kan-Tai Hsia
- Institute of Oral Biology, School of Dentistry, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan;
| | - Jia-Bin Liao
- Department of Pathology and Laboratory Medicine, Kaohsiung Veterans General Hospital, Kaohsiung 813414, Taiwan; (H.-W.C.); (J.-B.L.); (C.-C.Y.)
| | - Chih-Ching Yeh
- Department of Pathology and Laboratory Medicine, Kaohsiung Veterans General Hospital, Kaohsiung 813414, Taiwan; (H.-W.C.); (J.-B.L.); (C.-C.Y.)
- Department of Nursing, Shu-Zen Junior College of Medicine and Management, Kaohsiung 82144, Taiwan
| | - Wei-Ting Kuo
- Division of Urology, Department of Surgery, Kaohsiung Veterans General Hospital, Kaohsiung 813414, Taiwan
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
- Correspondence: (W.-T.K.); (Y.-F.Y.)
| | - Yi-Fang Yang
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung 813414, Taiwan
- Correspondence: (W.-T.K.); (Y.-F.Y.)
| |
Collapse
|
8
|
Xie X, Shirasu T, Guo LW, Kent KC. Smad2 inhibition of MET transcription potentiates human vascular smooth muscle cell apoptosis. ATHEROSCLEROSIS PLUS 2021; 44:31-42. [PMID: 35445204 PMCID: PMC9017589 DOI: 10.1016/j.athplu.2021.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
Background: Vascular smooth muscle cell (SMC) apoptosis is involved in major cardiovascular diseases. Smad2 is a transcription factor implicated in aortic aneurysm. The molecular mediators of Smad2-driven SMC apoptosis are not well defined. Here we have identified a Smad2-directed mechanism involving MET and FAS, both encoding cell membrane signaling receptors. Methods and results: Guided by microarray analysis in human primary aortic SMCs, loss/gain-of-function (siRNA/overexpression) indicated that Smad2 negatively and positively regulated, respectively, the gene expression of Met which was identified herein as anti-apoptotic and that of Fas, a known pro-apoptotic factor. While co-immunoprecipitation suggested a physical association of Smad2 with p53, chromatin immunoprecipitation followed by quantitative PCR revealed their co-occupancy in the same region of the MET promoter. Activating p53 with nutlin3a further potentiated the suppression of MET promoter-dependent luciferase activity and the exacerbation of SMC apoptosis that were caused by Smad2 overexpression. These results indicated that Smad2 in SMCs repressed the transcription of MET by cooperating with p53, and that Smad2 also activated FAS, a target gene of its transcription factor activity. Conclusions: Our study suggests a pro-apoptotic mechanism in human SMCs, whereby Smad2 negatively and positively regulates MET and FAS, genes encoding anti-apoptotic and pro-apoptotic factors, respectively.
Collapse
Affiliation(s)
- Xiujie Xie
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Takuro Shirasu
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Lian-Wang Guo
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA.,Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, 22908, USA
| | - K Craig Kent
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| |
Collapse
|
9
|
Rahimnejad M, Nasrollahi Boroujeni N, Jahangiri S, Rabiee N, Rabiee M, Makvandi P, Akhavan O, Varma RS. Prevascularized Micro-/Nano-Sized Spheroid/Bead Aggregates for Vascular Tissue Engineering. NANO-MICRO LETTERS 2021; 13:182. [PMID: 34409511 PMCID: PMC8374027 DOI: 10.1007/s40820-021-00697-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 07/13/2021] [Indexed: 05/02/2023]
Abstract
Efficient strategies to promote microvascularization in vascular tissue engineering, a central priority in regenerative medicine, are still scarce; nano- and micro-sized aggregates and spheres or beads harboring primitive microvascular beds are promising methods in vascular tissue engineering. Capillaries are the smallest type and in numerous blood vessels, which are distributed densely in cardiovascular system. To mimic this microvascular network, specific cell components and proangiogenic factors are required. Herein, advanced biofabrication methods in microvascular engineering, including extrusion-based and droplet-based bioprinting, Kenzan, and biogripper approaches, are deliberated with emphasis on the newest works in prevascular nano- and micro-sized aggregates and microspheres/microbeads.
Collapse
Affiliation(s)
- Maedeh Rahimnejad
- Biomedical Engineering Institute, School of Medicine, Université de Montréal, Montreal, Canada
- Research Centre, Centre Hospitalier de L'Université de Montréal (CRCHUM), Montreal, Canada
| | | | - Sepideh Jahangiri
- Research Centre, Centre Hospitalier de L'Université de Montréal (CRCHUM), Montreal, Canada
- Department of Biomedical Sciences, Faculty of Medicine, Université de Montréal, Montreal, Canada
| | - Navid Rabiee
- Department of Physics, Sharif University of Technology, P.O. Box 11155-9161, Tehran, Iran.
| | - Mohammad Rabiee
- Biomaterial Group, Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
| | - Pooyan Makvandi
- Centre for Materials Interfaces, Istituto Italiano Di Tecnologia, viale Rinaldo Piaggio 34, 56 025, Pontedera, Pisa, Italy
| | - Omid Akhavan
- Department of Physics, Sharif University of Technology, P.O. Box 11155-9161, Tehran, Iran.
| | - Rajender S Varma
- Regional Centre of Advanced Technologies and Materials, Czech Advanced Technology and Research Institute, Palacky University, Šlechtitelů 27, 783 71, Olomouc, Czech Republic.
| |
Collapse
|
10
|
Madjene C, Boutigny A, Bouton MC, Arocas V, Richard B. Protease Nexin-1 in the Cardiovascular System: Wherefore Art Thou? Front Cardiovasc Med 2021; 8:652852. [PMID: 33869311 PMCID: PMC8044347 DOI: 10.3389/fcvm.2021.652852] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 03/10/2021] [Indexed: 01/26/2023] Open
Abstract
The balance between proteases and protease inhibitors plays a critical role in tissue remodeling during cardiovascular diseases. Different serine protease inhibitors termed serpins, which are expressed in the cardiovascular system, can exert a fine-tuned regulation of protease activities. Among them, protease nexin-1 (PN-1, encoded by SERPINE2) is a very powerful thrombin inhibitor and can also inactivate plasminogen activators and plasmin. Studies have shown that this serpin is expressed by all cell subpopulations in the vascular wall and by circulating cells but is barely detectable in plasma in the free form. PN-1 present in platelet granules and released upon activation has been shown to present strong antithrombotic and antifibrinolytic properties. PN-1 has a broad spectrum of action related to both hemostatic and blood vessel wall protease activities. Different studies showed that PN-1 is not only an important protector of vascular cells against protease activities but also a significant actor in the clearance of the complexes it forms with its targets. In this context, PN-1 overexpression has been observed in the pathophysiology of thoracic aortic aneurysms (TAA) and during the development of atherosclerosis in humans. Similarly, in the heart, PN-1 has been shown to be overexpressed in a mouse model of heart failure and to be involved in cardiac fibrosis. Overall, PN-1 appears to serve as a "hand brake" for protease activities during cardiovascular remodeling. This review will thus highlight the role of PN-1 in the cardiovascular system and deliver a comprehensive assessment of its position among serpins.
Collapse
Affiliation(s)
- Celina Madjene
- LVTS, INSERM, U1148, Paris, France.,Université de Paris, Paris, France.,X. Bichat Hospital, Paris, France
| | - Alexandre Boutigny
- LVTS, INSERM, U1148, Paris, France.,Université de Paris, Paris, France.,X. Bichat Hospital, Paris, France
| | - Marie-Christine Bouton
- LVTS, INSERM, U1148, Paris, France.,Université de Paris, Paris, France.,X. Bichat Hospital, Paris, France
| | - Veronique Arocas
- LVTS, INSERM, U1148, Paris, France.,Université de Paris, Paris, France.,X. Bichat Hospital, Paris, France
| | - Benjamin Richard
- LVTS, INSERM, U1148, Paris, France.,X. Bichat Hospital, Paris, France.,Université Sorbonne Paris Nord, Villetaneuse, France
| |
Collapse
|
11
|
Creamer TJ, Bramel EE, MacFarlane EG. Insights on the Pathogenesis of Aneurysm through the Study of Hereditary Aortopathies. Genes (Basel) 2021; 12:183. [PMID: 33514025 PMCID: PMC7912671 DOI: 10.3390/genes12020183] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/20/2021] [Accepted: 01/22/2021] [Indexed: 12/15/2022] Open
Abstract
Thoracic aortic aneurysms (TAA) are permanent and localized dilations of the aorta that predispose patients to a life-threatening risk of aortic dissection or rupture. The identification of pathogenic variants that cause hereditary forms of TAA has delineated fundamental molecular processes required to maintain aortic homeostasis. Vascular smooth muscle cells (VSMCs) elaborate and remodel the extracellular matrix (ECM) in response to mechanical and biochemical cues from their environment. Causal variants for hereditary forms of aneurysm compromise the function of gene products involved in the transmission or interpretation of these signals, initiating processes that eventually lead to degeneration and mechanical failure of the vessel. These include mutations that interfere with transduction of stimuli from the matrix to the actin-myosin cytoskeleton through integrins, and those that impair signaling pathways activated by transforming growth factor-β (TGF-β). In this review, we summarize the features of the healthy aortic wall, the major pathways involved in the modulation of VSMC phenotypes, and the basic molecular functions impaired by TAA-associated mutations. We also discuss how the heterogeneity and balance of adaptive and maladaptive responses to the initial genetic insult might contribute to disease.
Collapse
Affiliation(s)
- Tyler J. Creamer
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (T.J.C.); (E.E.B.)
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Emily E. Bramel
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (T.J.C.); (E.E.B.)
- Predoctoral Training in Human Genetics and Molecular Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Elena Gallo MacFarlane
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (T.J.C.); (E.E.B.)
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
12
|
Michel JB. Phylogenic Determinants of Cardiovascular Frailty, Focus on Hemodynamics and Arterial Smooth Muscle Cells. Physiol Rev 2020; 100:1779-1837. [DOI: 10.1152/physrev.00022.2019] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The evolution of the circulatory system from invertebrates to mammals has involved the passage from an open system to a closed in-parallel system via a closed in-series system, accompanying the increasing complexity and efficiency of life’s biological functions. The archaic heart enables pulsatile motion waves of hemolymph in invertebrates, and the in-series circulation in fish occurs with only an endothelium, whereas mural smooth muscle cells appear later. The present review focuses on evolution of the circulatory system. In particular, we address how and why this evolution took place from a closed, flowing, longitudinal conductance at low pressure to a flowing, highly pressurized and bifurcating arterial compartment. However, although arterial pressure was the latest acquired hemodynamic variable, the general teleonomy of the evolution of species is the differentiation of individual organ function, supported by specific fueling allowing and favoring partial metabolic autonomy. This was achieved via the establishment of an active contractile tone in resistance arteries, which permitted the regulation of blood supply to specific organ activities via its localized function-dependent inhibition (active vasodilation). The global resistance to viscous blood flow is the peripheral increase in frictional forces caused by the tonic change in arterial and arteriolar radius, which backscatter as systemic arterial blood pressure. Consequently, the arterial pressure gradient from circulating blood to the adventitial interstitium generates the unidirectional outward radial advective conductance of plasma solutes across the wall of conductance arteries. This hemodynamic evolution was accompanied by important changes in arterial wall structure, supported by smooth muscle cell functional plasticity, including contractility, matrix synthesis and proliferation, endocytosis and phagocytosis, etc. These adaptive phenotypic shifts are due to epigenetic regulation, mainly related to mechanotransduction. These paradigms actively participate in cardio-arterial pathologies such as atheroma, valve disease, heart failure, aneurysms, hypertension, and physiological aging.
Collapse
|
13
|
Michel JB, Jondeau G, Milewicz DM. From genetics to response to injury: vascular smooth muscle cells in aneurysms and dissections of the ascending aorta. Cardiovasc Res 2019; 114:578-589. [PMID: 29360940 DOI: 10.1093/cvr/cvy006] [Citation(s) in RCA: 117] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 01/16/2018] [Indexed: 12/20/2022] Open
Abstract
Vascular smooth muscle cells (vSMCs) play a crucial role in both the pathogenesis of Aneurysms and Dissections of the ascending thoracic aorta (TAAD) in humans and in the associated adaptive compensatory responses, since thrombosis and inflammatory processes are absent in the majority of cases. Aneurysms and dissections share numerous characteristics, including aetiologies and histopathological alterations: vSMC disappearance, medial areas of mucoid degeneration, and extracellular matrix (ECM) breakdown. Three aetiologies predominate in TAAD in humans: (i) genetic causes in heritable familial forms, (ii) an association with bicuspid aortic valves, and (iii) a sporadic degenerative form linked to the aortic aging process. Genetic forms include mutations in vSMC genes encoding for molecules of the ECM or the TGF-β pathways, or participating in vSMC tone. On the other hand, aneurysms and dissections, whatever their aetiologies, are characterized by an increase in wall permeability leading to transmural advection of plasma proteins which could interact with vSMCs and ECM components. In this context, blood-borne plasminogen appears to play an important role, because its outward convection through the wall is increased in TAAD, and it could be converted to active plasmin at the vSMC membrane. Active plasmin can induce vSMC disappearance, proteolysis of adhesive proteins, activation of MMPs and release of TGF-β from its ECM storage sites. Conversely, vSMCs could respond to aneurysmal biomechanical and proteolytic injury by an epigenetic phenotypic switch, including constitutional overexpression and nuclear translocation of Smad2 and an increase in antiprotease and ECM protein synthesis. In contrast, such an epigenetic phenomenon is not observed in dissections. In this context, dysfunction of proteins involved in vSMC tone are interesting to study, particularly in interaction with plasma protein transport through the wall and TGF-β activation, to establish the relationship between these dysfunctions and ECM proteolysis.
Collapse
Affiliation(s)
- Jean-Baptiste Michel
- UMR 1148, Laboratory for Translational Vascular Science, Inserm and Paris 7- Denis Diderot University, Xavier Bichat Hospital, 75018 Paris, France
| | - Guillaume Jondeau
- UMR 1148, Laboratory for Translational Vascular Science, Inserm and Paris 7- Denis Diderot University, Xavier Bichat Hospital, 75018 Paris, France.,Cardiology Department, National Reference Center for Marfan Syndrome and Related Diseases, APHP Hopital Bichat, 75018 Paris
| | - Dianna M Milewicz
- Division of Medical Genetics, Department of Internal Medicine, University of Texas Medical School at Houston, Houston, TX 77030, USA
| |
Collapse
|
14
|
Messner B, Bernhard D. Bicuspid aortic valve-associated aortopathy: Where do we stand? J Mol Cell Cardiol 2019; 133:76-85. [PMID: 31152748 DOI: 10.1016/j.yjmcc.2019.05.023] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 05/23/2019] [Accepted: 05/28/2019] [Indexed: 01/30/2023]
Abstract
Herein we summarize the current knowledge on the bicuspid aortic valve (BAV)-associated aortopathy regarding clinical presentation and disease sub-classification, genetic background, hemodynamics, histopathology, cells and signaling, animal models, and biomarkers. Despite enormous efforts in research in all of the above areas, important issues remain unknown: (i) what is the ontogenetic basis of BAV development? (ii) how can we explain the diversity of BAV and associated aortopathy phenotypes? (iii) what are the signaling processes in aortopathy pathogenesis and how can we interfere with these processes? Despite undoubtedly great progress that has been made in the understanding of BAV-associated aortopathy, so far researchers have put together a heap of Lego bricks, but at present it is unclear if the bricks are compatible, how they fit together, and which parts are missing to build the true model of the BAV aorta. A joint approach is needed to accelerate research progress.
Collapse
Affiliation(s)
- Barbara Messner
- Cardiac Surgery Research Laboratory, Department of Surgery, Medical University of Vienna, Vienna, Austria
| | - David Bernhard
- Center for Medical Research, Medical Faculty, Johannes Kepler University Linz, Linz, Austria.
| |
Collapse
|
15
|
Boileau A, Lindsay ME, Michel JB, Devaux Y. Epigenetics in Ascending Thoracic Aortic Aneurysm and Dissection. AORTA (STAMFORD, CONN.) 2018; 6:1-12. [PMID: 30079931 PMCID: PMC6136679 DOI: 10.1055/s-0038-1639610] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Thoracic aortic aneurysm (TAA) is an asymptomatic and progressive dilatation of the thoracic aorta. Ascending aortic dissection (AAD) is an acute intraparietal tear, occurring or not on a pre-existing dilatation. AAD is a condition associated with a poor prognosis and a high mortality rate. TAA and AAD share common etiology as monogenic diseases linked to transforming growth factor β signaling pathway, extracellular matrix defect, or smooth muscle cell protein mutations. They feature a complex pathogenesis including loss of smooth muscle cells, altered phenotype, and extracellular matrix degradation in aortic media layer. A better knowledge of the mechanisms responsible for TAA progression and AAD occurrence is needed to improve healthcare, nowadays mainly consisting of aortic open surgery or endovascular replacement. Recent breakthrough discoveries allowed a deeper characterization of the mechanisms of gene regulation. Since alteration in gene expression has been linked to TAA and AAD, it is conceivable that a better knowledge of the causes of this alteration may lead to novel theranostic approaches. In this review article, the authors will focus on epigenetic regulation of gene expression, including the role of histone methylation and acetylation, deoxyribonucleic acid methylation, and noncoding ribonucleic acids in the pathogenesis of TAA and AAD. They will provide a translational perspective, presenting recent data that motivate the evaluation of the potential of epigenetics to diagnose TAA and prevent AAD.
Collapse
Affiliation(s)
- Adeline Boileau
- Cardiovascular Research Unit, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Mark E. Lindsay
- Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jean-Baptiste Michel
- UMRS 1148, INSERM, Paris 7-Denis Diderot University, Hôpital Xavier Bichat, Paris, France
| | - Yvan Devaux
- Cardiovascular Research Unit, Luxembourg Institute of Health, Luxembourg, Luxembourg
| |
Collapse
|
16
|
Boukais K, Borges LF, Venisse L, Touat Z, François D, Arocas V, Jondeau G, Declerck P, Bouton MC, Michel JB. Clearance of plasmin-PN-1 complexes by vascular smooth muscle cells in human aneurysm of the ascending aorta. Cardiovasc Pathol 2017; 32:15-25. [PMID: 29149696 DOI: 10.1016/j.carpath.2017.10.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Revised: 09/06/2017] [Accepted: 10/18/2017] [Indexed: 12/20/2022] Open
Abstract
Plasminogen is a circulating zymogen which enters the arterial wall by radial, transmural hydraulic conductance, where it is converted to plasmin by tissue plasminogen activator t-PA on an activation platform involving S100A4 on the vascular smooth muscle cell (vSMC) membrane. Plasmin is involved in the progression of human thoracic aneurysm of the ascending aorta (TAA). vSMCs protect the TAA wall from plasmin-induced proteolytic injury by expressing high levels of antiproteases. Protease nexin-1 (PN-1) is a tissue antiprotease belonging to the serpin superfamily, expressed in the vascular wall, and is able to form a covalent complex with plasmin. LDL receptor-related protein-1 (LRP-1) is a scavenger receptor implicated in protease-antiprotease complex internalization. In this study, we investigated whether PN-1 and LRP-1 are involved in the inhibition and clearance of plasminogen by the SMCs of human TAA. We demonstrated an overexpression of S100A4, PN-1, and LRP-1 in the medial layer of human TAA. Plasminogen activation taking place in the media of TAA was revealed by immunohistochemical staining and plasmin activity analyses. We showed by cell biology studies that plasmin-PN-1 complexes are internalized via LRP-1 in vSMCs from healthy and TAA media. Thus, two complementary mechanisms are involved in the protective role of PN-1 in human TAA: one involving plasmin inhibition and the other involving tissue clearance of plasmin-PN1 complexes via the scavenger receptor LRP-1.
Collapse
Affiliation(s)
- Kamel Boukais
- UMR 1148, Laboratory for Vascular Translational Science, Inserm; Paris7 Denis Diderot University
| | - Luciano F Borges
- Heart Institute (InCor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil; Department of Biological science, Federal University of São Paulo, São Paulo, Brazil
| | - Laurence Venisse
- UMR 1148, Laboratory for Vascular Translational Science, Inserm; Paris7 Denis Diderot University
| | - Ziad Touat
- UMR 1148, Laboratory for Vascular Translational Science, Inserm
| | - Déborah François
- UMR 1148, Laboratory for Vascular Translational Science, Inserm; Paris7 Denis Diderot University
| | - Véronique Arocas
- UMR 1148, Laboratory for Vascular Translational Science, Inserm; Paris7 Denis Diderot University
| | - Guillaume Jondeau
- UMR 1148, Laboratory for Vascular Translational Science, Inserm; Centre national de Référence pour le Syndrome de Marfan et apparentés, Hôpital Xavier Bichat
| | - Paul Declerck
- Laboratory for Therapeutic and Diagnostic Antibodies, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Marie-Christine Bouton
- UMR 1148, Laboratory for Vascular Translational Science, Inserm; Paris7 Denis Diderot University
| | - Jean-Baptiste Michel
- UMR 1148, Laboratory for Vascular Translational Science, Inserm; Paris7 Denis Diderot University.
| |
Collapse
|
17
|
Shen YH, LeMaire SA. Molecular pathogenesis of genetic and sporadic aortic aneurysms and dissections. Curr Probl Surg 2017; 54:95-155. [PMID: 28521856 DOI: 10.1067/j.cpsurg.2017.01.001] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 01/16/2017] [Indexed: 12/20/2022]
Affiliation(s)
- Ying H Shen
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX; Department of Cardiovascular Surgery, Texas Heart Institute, Houston, TX; Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX.
| | - Scott A LeMaire
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX; Department of Cardiovascular Surgery, Texas Heart Institute, Houston, TX; Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX; Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX.
| |
Collapse
|
18
|
Li X, Zhao D, Guo Z, Li T, Qili M, Xu B, Qian M, Liang H, E X, Chege Gitau S, Wang L, Huangfu L, Wu Q, Xu C, Shan H. Overexpression of SerpinE2/protease nexin-1 Contribute to Pathological Cardiac Fibrosis via increasing Collagen Deposition. Sci Rep 2016; 6:37635. [PMID: 27876880 PMCID: PMC5120308 DOI: 10.1038/srep37635] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 11/01/2016] [Indexed: 01/01/2023] Open
Abstract
Although increases in cardiovascular load (pressure overload) are known to elicit ventricular remodeling including cardiomyocyte hypertrophy and interstitial fibrosis, the molecular mechanisms of pressure overload or AngII -induced cardiac interstitial fibrosis remain elusive. In this study, serpinE2/protease nexin-1 was over-expressed in a cardiac fibrosis model induced by pressure-overloaded via transverse aortic constriction (TAC) in mouse. Knockdown of serpinE2 attenuates cardiac fibrosis in a mouse model of TAC. At meantime, the results showed that serpinE2 significantly were increased with collagen accumulations induced by AngII or TGF-β stimulation in vitro. Intriguingly, extracellular collagen in myocardial fibroblast was reduced by knockdown of serpinE2 compared with the control in vitro. In stark contrast, the addition of exogenous PN-1 up-regulated the content of collagen in myocardial fibroblast. The MEK1/2- ERK1/2 signaling probably promoted the expression of serpinE2 via transcription factors Elk1 in myocardial fibroblast. In conclusion, stress-induced the ERK1/2 signaling pathway activation up-regulated serpinE2 expression, consequently led accumulation of collagen protein, and contributed to cardiac fibrosis.
Collapse
Affiliation(s)
- Xuelian Li
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Dandan Zhao
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Zhenfeng Guo
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China.,The second Clinical Medical School of Inner Mongolia University for Nationalities, Inner Mongolia Forestry General Hospital, Inner Mongolia, China
| | - Tianshi Li
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Muge Qili
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Bozhi Xu
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Ming Qian
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Haihai Liang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Xiaoqiang E
- Department of Orthopaedics, the First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Samuel Chege Gitau
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China.,Department of Pharmacy and Complementary Medicine, School of Health Sciences, Kenyatta University, Nairobi, Kenya
| | - Lu Wang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Longtao Huangfu
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Qiuxia Wu
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Chaoqian Xu
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Hongli Shan
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| |
Collapse
|
19
|
Smooth muscle cell-specific Tgfbr1 deficiency promotes aortic aneurysm formation by stimulating multiple signaling events. Sci Rep 2016; 6:35444. [PMID: 27739498 PMCID: PMC5064316 DOI: 10.1038/srep35444] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 09/29/2016] [Indexed: 12/21/2022] Open
Abstract
Transforming growth factor (TGF)-β signaling disorder has emerged as a common molecular signature for aortic aneurysm development. The timing of postnatal maturation plays a key role in dictating the biological outcome of TGF-β signaling disorders in the aortic wall. In this study, we investigated the impact of deficiency of TGFβ receptors on the structural homeostasis of mature aortas. We used an inducible Cre-loxP system driven by a Myh11 promoter to delete Tgfbr1, Tgfbr2, or both in smooth muscle cells (SMCs) of adult mice. TGFBR1 deficiency resulted in rapid and severe aneurysmal degeneration, with 100% penetrance of ascending thoracic aortas, whereas TGFBR2 deletion only caused mild aortic pathology with low (26%) lesion prevalence. Removal of TGFBR2 attenuated the aortic pathology caused by TGFBR1 deletion and correlated with a reduction of early ERK phosphorylation. In addition, the production of angiotensin (Ang)-converting enzyme was upregulated in TGFBR1 deficient aortas at the early stage of aneurysmal degeneration. Inhibition of ERK phosphorylation or blockade of AngII type I receptor AT1R prevented aneurysmal degeneration of TGFBR1 deficient aortas. In conclusion, loss of SMC-Tgfbr1 triggers multiple deleterious pathways, including abnormal TGFBR2, ERK, and AngII/AT1R signals that disrupt aortic wall homeostasis to cause aortic aneurysm formation.
Collapse
|
20
|
Boukais K, Bayles R, Borges LDF, Louedec L, Boulaftali Y, Ho-Tin-Noé B, Arocas V, Bouton MC, Michel JB. Uptake of Plasmin-PN-1 Complexes in Early Human Atheroma. Front Physiol 2016; 7:273. [PMID: 27445860 PMCID: PMC4927630 DOI: 10.3389/fphys.2016.00273] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 06/16/2016] [Indexed: 12/16/2022] Open
Abstract
Zymogens are delivered to the arterial wall by radial transmural convection. Plasminogen can be activated within the arterial wall to produce plasmin, which is involved in evolution of the atherosclerotic plaque. Vascular smooth muscle cells (vSMCs) protect the vessels from proteolytic injury due to atherosclerosis development by highly expressing endocytic LDL receptor-related protein-1 (LRP-1), and by producing anti-proteases, such as Protease Nexin-1 (PN-1). PN-1 is able to form covalent complexes with plasmin. We hypothesized that plasmin-PN-1 complexes could be internalized via LRP-1 by vSMCs during the early stages of human atheroma. LRP-1 is also responsible for the capture of aggregated LDL in human atheroma. Plasmin activity and immunohistochemical analyses of early human atheroma showed that the plasminergic system is activated within the arterial wall, where intimal foam cells, including vSMCs and platelets, are the major sites of PN-1 accumulation. Both PN-1 and LRP-1 are overexpressed in early atheroma at both messenger and protein levels. Cell biology studies demonstrated an increased expression of PN-1 and tissue plasminogen activator by vSMCs in response to LDL. Plasmin-PN-1 complexes are internalized via LRP-1 in vSMCs, whereas plasmin alone is not. Tissue PN-1 interacts with plasmin in early human atheroma via two complementary mechanisms: plasmin inhibition and tissue uptake of plasmin-PN-1 complexes via LRP-1 in vSMCs. Despite this potential protective effect, plasminogen activation by vSMCs remains abnormally elevated in the intima in early stages of human atheroma.
Collapse
Affiliation(s)
- Kamel Boukais
- UMR 1148, Laboratory for Vascular Translational Science, Institut National de la Santé et de la Recherche MédicaleParis, France; Paris7 Denis Diderot UniversityParis, France
| | - Richard Bayles
- UMR 1148, Laboratory for Vascular Translational Science, Institut National de la Santé et de la Recherche MédicaleParis, France; Department of Physiology and Pharmacology, Oregon Health and Science UniversityPortland, OR, USA
| | - Luciano de Figueiredo Borges
- Departement of Biological Science, Federal University of São PauloSão Paulo, Brazil; Heart Institute (InCor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São PauloSão Paulo, Brazil
| | - Liliane Louedec
- UMR 1148, Laboratory for Vascular Translational Science, Institut National de la Santé et de la Recherche MédicaleParis, France; Paris7 Denis Diderot UniversityParis, France
| | - Yacine Boulaftali
- UMR 1148, Laboratory for Vascular Translational Science, Institut National de la Santé et de la Recherche MédicaleParis, France; Paris7 Denis Diderot UniversityParis, France
| | - Benoit Ho-Tin-Noé
- UMR 1148, Laboratory for Vascular Translational Science, Institut National de la Santé et de la Recherche MédicaleParis, France; Paris7 Denis Diderot UniversityParis, France
| | - Véronique Arocas
- UMR 1148, Laboratory for Vascular Translational Science, Institut National de la Santé et de la Recherche MédicaleParis, France; Paris7 Denis Diderot UniversityParis, France
| | - Marie-Christine Bouton
- UMR 1148, Laboratory for Vascular Translational Science, Institut National de la Santé et de la Recherche MédicaleParis, France; Paris7 Denis Diderot UniversityParis, France
| | - Jean-Baptiste Michel
- UMR 1148, Laboratory for Vascular Translational Science, Institut National de la Santé et de la Recherche MédicaleParis, France; Paris7 Denis Diderot UniversityParis, France
| |
Collapse
|
21
|
Solleti SK, Srisuma S, Bhattacharya S, Rangel-Moreno J, Bijli KM, Randall TD, Rahman A, Mariani TJ. Serpine2 deficiency results in lung lymphocyte accumulation and bronchus-associated lymphoid tissue formation. FASEB J 2016; 30:2615-26. [PMID: 27059719 DOI: 10.1096/fj.201500159r] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 03/28/2016] [Indexed: 11/11/2022]
Abstract
Serine proteinase inhibitor, clade E, member 2 (SERPINE2), is a cell- and extracellular matrix-associated inhibitor of thrombin. Although SERPINE2 is a candidate susceptibility gene for chronic obstructive pulmonary disease, the physiologic role of this protease inhibitor in lung development and homeostasis is unknown. We observed spontaneous monocytic-cell infiltration in the lungs of Serpine2-deficient (SE2(-/-)) mice, beginning at or before the time of lung maturity, which resulted in lesions that resembled bronchus-associated lymphoid tissue (BALT). The initiation of lymphocyte accumulation in the lungs of SE2(-/-) mice involved the excessive expression of chemokines, cytokines, and adhesion molecules that are essential for BALT induction, organization, and maintenance. BALT-like lesion formation in the lungs of SE2(-/-) mice was also associated with a significant increase in the activation of thrombin, a recognized target of SE2, and excess stimulation of NF-κB, a major regulator of chemokine expression and inflammation. Finally, systemic delivery of thrombin rapidly stimulated lung chemokine expression in vivo These data uncover a novel mechanism whereby loss of serine protease inhibition leads to lung lymphocyte accumulation.-Solleti, S. K., Srisuma, S., Bhattacharya, S., Rangel-Moreno, J., Bijli, K. M., Randall, T. D., Rahman, A., Mariani, T. J. Serpine2 deficiency results in lung lymphocyte accumulation and bronchus-associated lymphoid tissue formation.
Collapse
Affiliation(s)
- Siva Kumar Solleti
- Division of Neonatology, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, USA
| | - Sorachai Srisuma
- Department of Physiology, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Soumyaroop Bhattacharya
- Division of Neonatology, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, USA
| | - Javier Rangel-Moreno
- Division of Allergy, Immunology, and Rheumatology, University of Rochester Medical Center, Rochester, New York, USA
| | - Kaiser M Bijli
- Division of Neonatology, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, USA; Division of Pulmonary, Allergy, and Critical Care Medicine, Emory University/Atlanta Veterans Affairs Medical Center, Atlanta, Georgia, USA
| | - Troy D Randall
- Division of Allergy, Immunology, and Rheumatology, University of Rochester Medical Center, Rochester, New York, USA; Division of Clinical Immunology and Rheumatology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Arshad Rahman
- Division of Neonatology, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, USA
| | - Thomas J Mariani
- Division of Neonatology, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, USA;
| |
Collapse
|
22
|
Abstract
PURPOSE OF REVIEW It is well established that blocking the renin-angiotensin-aldosterone system (RAAS) is effective for the treatment of cardiovascular and renal complications in hypertension and diabetes mellitus. Although the induction of transforming growth factor beta1 (TGFbeta1) by components of the RAAS mediates the hypertrophic and fibrogenic changes in cardiovascular-renal complications, it is still controversial as to whether TGFbeta1 can be a target to prevent such complications. Here, we review recent findings on the role of TGFbeta1 in fluid homeostasis, focusing on the relationship with aldosterone. RECENT FINDINGS TGFbeta1 suppresses the adrenal production of aldosterone and renal tubular sodium reabsorption. We have generated mice with TGFbeta1 mRNA expression graded in five steps, from 10 to 300% of normal, and found that blood pressure and plasma volume are negatively regulated by TGFbeta1. Notably, the 10% hypomorph exhibits primary aldosteronism and sodium and water retention due to markedly impaired urinary excretion of water and electrolytes. SUMMARY These results identify TGFbeta signalling as an important counterregulatory system against aldosterone. Understanding the molecular mechanisms for the suppressive effects of TGFbeta1 on adrenocortical and renal function may further our understanding of primary aldosteronism, as well as assist in the development of novel therapeutic strategies for hypertension.
Collapse
|
23
|
Abstract
PURPOSE OF REVIEW A lot of new data have been obtained in familial thoracic aortic aneurysms, including description of new entities and better understanding of pathophysiology. The aim of this review is to put them in perspective. RECENT FINDINGS The new data have been collected, put together, and allowed a new classification scheme to be proposed by the Montalcino Aortic Consortium on the basis of the role of proteins coded by the culprit gene (either protein of the extracellular matrix or protein of the transforming growth factor-beta pathway, or protein of the contractile apparatus of the smooth muscle cell). These groups of diseases include aortic aneurysm, but the extent of extra-aortic vascular risk and the presence of extra-aortic (skeletal, ophthalmologic, neurological, or immunological) features vary according to the gene involved. This understanding also sheds light on the therapeutic benefits that can be foreseen for new molecules, or old molecules used in a newer way. SUMMARY Classification of familial forms of thoracic aortic aneurysm should allow a better understanding of these diseases and therefore standardization of initial evaluation of the patients (vascular evaluation limited or not to the aorta, and extravascular evaluation, including or not skeleton, eyes, neurology, digestive tract, and immunological diseases) and individualization of therapy (adapted to both the genotype and the phenotype).
Collapse
|
24
|
Rabkin SW. Accentuating and Opposing Factors Leading to Development of Thoracic Aortic Aneurysms Not Due to Genetic or Inherited Conditions. Front Cardiovasc Med 2015; 2:21. [PMID: 26664893 PMCID: PMC4671360 DOI: 10.3389/fcvm.2015.00021] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 04/15/2015] [Indexed: 12/12/2022] Open
Abstract
Understanding and unraveling the pathophysiology of thoracic aortic aneurysm (TAA), a vascular disease with a potentially high-mortality rate, is one of the next frontiers in vascular biology. The processes leading to the formation of TAA, of unknown cause, so-called degenerative TAA, are complex. This review advances the concept of promoters and inhibitors of the development of degenerative TAA. Promoters of TAA development include age, blood pressure elevation, increased pulse pressure, neurohumeral factors increasing blood pressure, inflammation specifically IFN-γ, IL-1 β, IL-6, TNF-α, and S100 A12; the coagulation system specifically plasmin, platelets, and thrombin as well as matrix metalloproteinases (MMPs). SMAD-2 signaling and specific microRNAs modulate TAA development. The major inhibitors or factors opposing TAA development are the constituents of the aortic wall (elastic lamellae, collagen, fibulins, fibronectin, proteoglycans, and vascular smooth muscle cells), which maintain normal aortic dimensions in the face of aortic wall stress, specific tissue MMP inhibitors, plasminogen activator inhibitor-1, protease nexin-1, and Syndecans. Increases in promoters and reductions in inhibitors expand the thoracic aorta leading to TAA formation.
Collapse
Affiliation(s)
- Simon W Rabkin
- Division of Cardiology, Department of Medicine, University of British Columbia , Vancouver, BC , Canada
| |
Collapse
|
25
|
Emrich FC, Okamura H, Dalal AR, Penov K, Merk DR, Raaz U, Hennigs JK, Chin JT, Miller MO, Pedroza AJ, Craig JK, Koyano TK, Blankenberg FG, Connolly AJ, Mohr FW, Alvira CM, Rabinovitch M, Fischbein MP. Enhanced Caspase Activity Contributes to Aortic Wall Remodeling and Early Aneurysm Development in a Murine Model of Marfan Syndrome. Arterioscler Thromb Vasc Biol 2015; 35:146-54. [DOI: 10.1161/atvbaha.114.304364] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Objective—
Rupture and dissection of aortic root aneurysms remain the leading causes of death in patients with the Marfan syndrome, a hereditary connective tissue disorder that affects 1 in 5000 individuals worldwide. In the present study, we use a Marfan mouse model (
Fbn1
C1039G/+
) to investigate the biological importance of apoptosis during aneurysm development in Marfan syndrome.
Approach and Results—
Using in vivo single-photon emission computed tomographic-imaging and ex vivo autoradiography for Tc99m-annexin, we discovered increased apoptosis in the
Fbn1
C1039G/+
ascending aorta during early aneurysm development peaking at 4 weeks. Immunofluorescence colocalization studies identified smooth muscle cells (SMCs) as the apoptotic cell population. As biological proof of concept that early aortic wall apoptosis plays a role in aneurysm development in Marfan syndrome,
Fbn1
C1039G/+
mice were treated daily from 2 to 6 weeks with either (1) a pan-caspase inhibitor, Q-V
D
-OPh (20 mg/kg), or (2) vehicle control intraperitoneally. Q-V
D
-OPh treatment led to a significant reduction in aneurysm size and decreased extracellular matrix degradation in the aortic wall compared with control mice. In vitro studies using
Fbn1
C1039G/+
ascending SMCs showed that apoptotic SMCs have increased elastolytic potential compared with viable cells, mostly because of caspase activity. Moreover, in vitro (1) cell membrane isolation, (2) immunofluorescence staining, and (3) scanning electron microscopy studies illustrate that caspases are expressed on the exterior cell surface of apoptotic SMCs.
Conclusions—
Caspase inhibition attenuates aneurysm development in an
Fbn1
C1039G/+
Marfan mouse model. Mechanistically, during apoptosis, caspases are expressed on the cell surface of SMCs and likely contribute to elastin degradation and aneurysm development in Marfan syndrome.
Collapse
Affiliation(s)
- Fabian C. Emrich
- From the Department of Cardiothoracic Surgery (F.C.E., H.O., A.R.D., K.P., D.R.M., J.T.C., M.O.M., A.J.P., J.K.C., T.K.K, M.P.F.), Department of Cardiovascular Medicine (U.R.), Department of Pediatrics (J.K.H., C.M.A, M.R.), Department of Radiology (F.G.B.), and Department of Pathology (A.J.C.), Stanford University, CA; Department of Cardiothoracic Surgery, Heart Center, Leipzig University, Leipzig, Germany (F.C.E., K.P., D.R.M., F.W.M.); and Department of Cardiovascular Surgery, Saitama Medical
| | - Homare Okamura
- From the Department of Cardiothoracic Surgery (F.C.E., H.O., A.R.D., K.P., D.R.M., J.T.C., M.O.M., A.J.P., J.K.C., T.K.K, M.P.F.), Department of Cardiovascular Medicine (U.R.), Department of Pediatrics (J.K.H., C.M.A, M.R.), Department of Radiology (F.G.B.), and Department of Pathology (A.J.C.), Stanford University, CA; Department of Cardiothoracic Surgery, Heart Center, Leipzig University, Leipzig, Germany (F.C.E., K.P., D.R.M., F.W.M.); and Department of Cardiovascular Surgery, Saitama Medical
| | - Alex R. Dalal
- From the Department of Cardiothoracic Surgery (F.C.E., H.O., A.R.D., K.P., D.R.M., J.T.C., M.O.M., A.J.P., J.K.C., T.K.K, M.P.F.), Department of Cardiovascular Medicine (U.R.), Department of Pediatrics (J.K.H., C.M.A, M.R.), Department of Radiology (F.G.B.), and Department of Pathology (A.J.C.), Stanford University, CA; Department of Cardiothoracic Surgery, Heart Center, Leipzig University, Leipzig, Germany (F.C.E., K.P., D.R.M., F.W.M.); and Department of Cardiovascular Surgery, Saitama Medical
| | - Kiril Penov
- From the Department of Cardiothoracic Surgery (F.C.E., H.O., A.R.D., K.P., D.R.M., J.T.C., M.O.M., A.J.P., J.K.C., T.K.K, M.P.F.), Department of Cardiovascular Medicine (U.R.), Department of Pediatrics (J.K.H., C.M.A, M.R.), Department of Radiology (F.G.B.), and Department of Pathology (A.J.C.), Stanford University, CA; Department of Cardiothoracic Surgery, Heart Center, Leipzig University, Leipzig, Germany (F.C.E., K.P., D.R.M., F.W.M.); and Department of Cardiovascular Surgery, Saitama Medical
| | - Denis R. Merk
- From the Department of Cardiothoracic Surgery (F.C.E., H.O., A.R.D., K.P., D.R.M., J.T.C., M.O.M., A.J.P., J.K.C., T.K.K, M.P.F.), Department of Cardiovascular Medicine (U.R.), Department of Pediatrics (J.K.H., C.M.A, M.R.), Department of Radiology (F.G.B.), and Department of Pathology (A.J.C.), Stanford University, CA; Department of Cardiothoracic Surgery, Heart Center, Leipzig University, Leipzig, Germany (F.C.E., K.P., D.R.M., F.W.M.); and Department of Cardiovascular Surgery, Saitama Medical
| | - Uwe Raaz
- From the Department of Cardiothoracic Surgery (F.C.E., H.O., A.R.D., K.P., D.R.M., J.T.C., M.O.M., A.J.P., J.K.C., T.K.K, M.P.F.), Department of Cardiovascular Medicine (U.R.), Department of Pediatrics (J.K.H., C.M.A, M.R.), Department of Radiology (F.G.B.), and Department of Pathology (A.J.C.), Stanford University, CA; Department of Cardiothoracic Surgery, Heart Center, Leipzig University, Leipzig, Germany (F.C.E., K.P., D.R.M., F.W.M.); and Department of Cardiovascular Surgery, Saitama Medical
| | - Jan K. Hennigs
- From the Department of Cardiothoracic Surgery (F.C.E., H.O., A.R.D., K.P., D.R.M., J.T.C., M.O.M., A.J.P., J.K.C., T.K.K, M.P.F.), Department of Cardiovascular Medicine (U.R.), Department of Pediatrics (J.K.H., C.M.A, M.R.), Department of Radiology (F.G.B.), and Department of Pathology (A.J.C.), Stanford University, CA; Department of Cardiothoracic Surgery, Heart Center, Leipzig University, Leipzig, Germany (F.C.E., K.P., D.R.M., F.W.M.); and Department of Cardiovascular Surgery, Saitama Medical
| | - Jocelyn T. Chin
- From the Department of Cardiothoracic Surgery (F.C.E., H.O., A.R.D., K.P., D.R.M., J.T.C., M.O.M., A.J.P., J.K.C., T.K.K, M.P.F.), Department of Cardiovascular Medicine (U.R.), Department of Pediatrics (J.K.H., C.M.A, M.R.), Department of Radiology (F.G.B.), and Department of Pathology (A.J.C.), Stanford University, CA; Department of Cardiothoracic Surgery, Heart Center, Leipzig University, Leipzig, Germany (F.C.E., K.P., D.R.M., F.W.M.); and Department of Cardiovascular Surgery, Saitama Medical
| | - Miquell O. Miller
- From the Department of Cardiothoracic Surgery (F.C.E., H.O., A.R.D., K.P., D.R.M., J.T.C., M.O.M., A.J.P., J.K.C., T.K.K, M.P.F.), Department of Cardiovascular Medicine (U.R.), Department of Pediatrics (J.K.H., C.M.A, M.R.), Department of Radiology (F.G.B.), and Department of Pathology (A.J.C.), Stanford University, CA; Department of Cardiothoracic Surgery, Heart Center, Leipzig University, Leipzig, Germany (F.C.E., K.P., D.R.M., F.W.M.); and Department of Cardiovascular Surgery, Saitama Medical
| | - Albert J. Pedroza
- From the Department of Cardiothoracic Surgery (F.C.E., H.O., A.R.D., K.P., D.R.M., J.T.C., M.O.M., A.J.P., J.K.C., T.K.K, M.P.F.), Department of Cardiovascular Medicine (U.R.), Department of Pediatrics (J.K.H., C.M.A, M.R.), Department of Radiology (F.G.B.), and Department of Pathology (A.J.C.), Stanford University, CA; Department of Cardiothoracic Surgery, Heart Center, Leipzig University, Leipzig, Germany (F.C.E., K.P., D.R.M., F.W.M.); and Department of Cardiovascular Surgery, Saitama Medical
| | - Juliana K. Craig
- From the Department of Cardiothoracic Surgery (F.C.E., H.O., A.R.D., K.P., D.R.M., J.T.C., M.O.M., A.J.P., J.K.C., T.K.K, M.P.F.), Department of Cardiovascular Medicine (U.R.), Department of Pediatrics (J.K.H., C.M.A, M.R.), Department of Radiology (F.G.B.), and Department of Pathology (A.J.C.), Stanford University, CA; Department of Cardiothoracic Surgery, Heart Center, Leipzig University, Leipzig, Germany (F.C.E., K.P., D.R.M., F.W.M.); and Department of Cardiovascular Surgery, Saitama Medical
| | - Tiffany K. Koyano
- From the Department of Cardiothoracic Surgery (F.C.E., H.O., A.R.D., K.P., D.R.M., J.T.C., M.O.M., A.J.P., J.K.C., T.K.K, M.P.F.), Department of Cardiovascular Medicine (U.R.), Department of Pediatrics (J.K.H., C.M.A, M.R.), Department of Radiology (F.G.B.), and Department of Pathology (A.J.C.), Stanford University, CA; Department of Cardiothoracic Surgery, Heart Center, Leipzig University, Leipzig, Germany (F.C.E., K.P., D.R.M., F.W.M.); and Department of Cardiovascular Surgery, Saitama Medical
| | - Francis G. Blankenberg
- From the Department of Cardiothoracic Surgery (F.C.E., H.O., A.R.D., K.P., D.R.M., J.T.C., M.O.M., A.J.P., J.K.C., T.K.K, M.P.F.), Department of Cardiovascular Medicine (U.R.), Department of Pediatrics (J.K.H., C.M.A, M.R.), Department of Radiology (F.G.B.), and Department of Pathology (A.J.C.), Stanford University, CA; Department of Cardiothoracic Surgery, Heart Center, Leipzig University, Leipzig, Germany (F.C.E., K.P., D.R.M., F.W.M.); and Department of Cardiovascular Surgery, Saitama Medical
| | - Andrew J. Connolly
- From the Department of Cardiothoracic Surgery (F.C.E., H.O., A.R.D., K.P., D.R.M., J.T.C., M.O.M., A.J.P., J.K.C., T.K.K, M.P.F.), Department of Cardiovascular Medicine (U.R.), Department of Pediatrics (J.K.H., C.M.A, M.R.), Department of Radiology (F.G.B.), and Department of Pathology (A.J.C.), Stanford University, CA; Department of Cardiothoracic Surgery, Heart Center, Leipzig University, Leipzig, Germany (F.C.E., K.P., D.R.M., F.W.M.); and Department of Cardiovascular Surgery, Saitama Medical
| | - Friedrich W. Mohr
- From the Department of Cardiothoracic Surgery (F.C.E., H.O., A.R.D., K.P., D.R.M., J.T.C., M.O.M., A.J.P., J.K.C., T.K.K, M.P.F.), Department of Cardiovascular Medicine (U.R.), Department of Pediatrics (J.K.H., C.M.A, M.R.), Department of Radiology (F.G.B.), and Department of Pathology (A.J.C.), Stanford University, CA; Department of Cardiothoracic Surgery, Heart Center, Leipzig University, Leipzig, Germany (F.C.E., K.P., D.R.M., F.W.M.); and Department of Cardiovascular Surgery, Saitama Medical
| | - Cristina M. Alvira
- From the Department of Cardiothoracic Surgery (F.C.E., H.O., A.R.D., K.P., D.R.M., J.T.C., M.O.M., A.J.P., J.K.C., T.K.K, M.P.F.), Department of Cardiovascular Medicine (U.R.), Department of Pediatrics (J.K.H., C.M.A, M.R.), Department of Radiology (F.G.B.), and Department of Pathology (A.J.C.), Stanford University, CA; Department of Cardiothoracic Surgery, Heart Center, Leipzig University, Leipzig, Germany (F.C.E., K.P., D.R.M., F.W.M.); and Department of Cardiovascular Surgery, Saitama Medical
| | - Marlene Rabinovitch
- From the Department of Cardiothoracic Surgery (F.C.E., H.O., A.R.D., K.P., D.R.M., J.T.C., M.O.M., A.J.P., J.K.C., T.K.K, M.P.F.), Department of Cardiovascular Medicine (U.R.), Department of Pediatrics (J.K.H., C.M.A, M.R.), Department of Radiology (F.G.B.), and Department of Pathology (A.J.C.), Stanford University, CA; Department of Cardiothoracic Surgery, Heart Center, Leipzig University, Leipzig, Germany (F.C.E., K.P., D.R.M., F.W.M.); and Department of Cardiovascular Surgery, Saitama Medical
| | - Michael P. Fischbein
- From the Department of Cardiothoracic Surgery (F.C.E., H.O., A.R.D., K.P., D.R.M., J.T.C., M.O.M., A.J.P., J.K.C., T.K.K, M.P.F.), Department of Cardiovascular Medicine (U.R.), Department of Pediatrics (J.K.H., C.M.A, M.R.), Department of Radiology (F.G.B.), and Department of Pathology (A.J.C.), Stanford University, CA; Department of Cardiothoracic Surgery, Heart Center, Leipzig University, Leipzig, Germany (F.C.E., K.P., D.R.M., F.W.M.); and Department of Cardiovascular Surgery, Saitama Medical
| |
Collapse
|
26
|
Increased expression of protease nexin-1 in fibroblasts during idiopathic pulmonary fibrosis regulates thrombin activity and fibronectin expression. J Transl Med 2014; 94:1237-46. [PMID: 25199049 DOI: 10.1038/labinvest.2014.111] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Revised: 07/03/2014] [Accepted: 07/29/2014] [Indexed: 11/08/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic diffuse lung disease characterized by an accumulation of excess fibrous material in the lung. Protease nexin-1 (PN-1) is a tissue serpin produced by many cell types, including lung fibroblasts. PN-1 is capable of regulating proteases of both coagulation and fibrinolysis systems, by inhibiting, respectively, thrombin and plasminergic enzymes. PN-1 is thus a good candidate for regulating tissue remodeling occurring during IPF. We demonstrated a significant increase of PN-1 expression in lung tissue extracts, lung fibroblasts and bronchoalveolar lavage fluids of patients with IPF. The increase of PN-1 expression was reproduced after stimulation of control lung fibroblasts by transforming growth factor-β, a major pro-fibrotic cytokine involved in IPF. Another serpin, plasminogen activator inhibitor-1 (PAI-1) is also overexpressed in fibrotic fibroblasts. Unlike PAI-1, cell-bound PN-1 as well as secreted PN-1 from IPF and stimulated fibroblasts were shown to inhibit efficiently thrombin activity, indicating that both serpins should exhibit complementary roles in IPF pathogenesis, via their different preferential antiprotease activities. Moreover, we observed that overexpression of PN-1 induced by transfection of control fibroblasts led to increased fibronectin expression, whereas PN-1 silencing induced in fibrotic fibroblasts led to decreased fibronectin expression. Overexpression of PN-1 lacking either its antiprotease activity or its binding capacity to glycosaminoglycans had no effect on fibronectin expression. These novel findings suggest that modulation of PN-1 expression in lung fibroblasts may also have a role in the development of IPF by directly influencing the expression of extracellular matrix proteins. Our data provide new insights into the role of PN-1 in the poorly understood pathological processes involved in IPF and could therefore give rise to new therapeutic approaches.
Collapse
|
27
|
Kessler K, Borges LF, Ho-Tin-Noé B, Jondeau G, Michel JB, Vranckx R. Angiogenesis and remodelling in human thoracic aortic aneurysms. Cardiovasc Res 2014; 104:147-59. [PMID: 25139748 DOI: 10.1093/cvr/cvu196] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
AIMS Human thoracic aneurysm of the ascending aorta (TAA) is a chronic disease characterized by dilatation of the aortic wall, which can progress to vessel dissection and rupture. TAA has several aetiologies, but all forms present common features, including tissue remodelling. Here, we determined and characterized the angiogenic process associated with TAA and its relation with wall remodelling. METHODS AND RESULTS Immunostaining for blood vessels showed an increased density of microvessels originating from the adventitia in the external medial layer of TAA compared with healthy aortas. Proteomic array analysis of 55 angiogenic factors in medial and adventitial layers showed different expression profiles in both tissue compartments between aneurysmal and healthy aortas. Quantification by ELISA confirmed that all forms of TAA contained higher levels of several pro- and anti-angiogenic factors, including angiopoietin-1 and -2, fibroblast growth factor-acidic, and thrombospondin-1, than that of healthy aortas. However, all groups showed comparable levels of vascular endothelial growth factor-A. Quantitative RT-PCR demonstrated that angiopoietins were overexpressed in TAA media. Immunostaining and electron microscopy revealed that neovessels had defective endothelial junctions and poor mural cell coverage. This incomplete structure was associated with the accumulation of plasminogen and albumin in the media of TAA. CONCLUSION We describe, for the first time, leaky neovessel formation in TAA media in association with an imbalance of angiogenic factor levels. Although the initiating mechanisms of neo-angiogenesis in TAA and the potential aetiology-related differences remain to be determined, our results suggest that neo-angiogenesis could participate in TAA wall remodelling and weakening through deposition of blood-borne zymogens.
Collapse
Affiliation(s)
- Ketty Kessler
- Univ Paris Diderot, Sorbonne Paris Cité, LVTS, UMR-S1148, F-75018 Paris, France INSERM Unit 1148, Hôpital Xavier Bichat, Secteur Claude Bernard, 46 rue Henri Huchard, FR-75877 Paris cedex 18, France
| | - Luciano F Borges
- Heart Institute (InCor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil Department of Morphology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Benoît Ho-Tin-Noé
- Univ Paris Diderot, Sorbonne Paris Cité, LVTS, UMR-S1148, F-75018 Paris, France
| | - Guillaume Jondeau
- Centre National de Référence pour le syndrome de Marfan et apparentés, Hôpital Xavier Bichat, Paris, France
| | - Jean-Baptiste Michel
- Univ Paris Diderot, Sorbonne Paris Cité, LVTS, UMR-S1148, F-75018 Paris, France INSERM Unit 1148, Hôpital Xavier Bichat, Secteur Claude Bernard, 46 rue Henri Huchard, FR-75877 Paris cedex 18, France
| | - Roger Vranckx
- Univ Paris Diderot, Sorbonne Paris Cité, LVTS, UMR-S1148, F-75018 Paris, France
| |
Collapse
|
28
|
Affiliation(s)
- Alan Daugherty
- From the Saha Cardiovascular Research Center, University of Kentucky, Lexington (A.D.); and Department of Surgery and Cancer, Imperial College, London, United Kingdom (J.T.P.)
| | | |
Collapse
|