1
|
Javed MJ, Howard RM, Li H, Carrasco L, Dirain MLS, Su G, Cai G, Upchurch GR, Jiang Z. Gasdermin D deficiency attenuates development of ascending aortic dissections in a novel mouse model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.22.609270. [PMID: 39229014 PMCID: PMC11370574 DOI: 10.1101/2024.08.22.609270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Background Thoracic aortic dissection (TAD) is a silent killer. Approximately two-thirds of the cases occur in the ascending aorta (i.e. type A dissection) and majority of them are unrelated to genetic mutations. However, animal models of spontaneous type A dissection are not widely available. In the present study, a novel mouse TAD model was created. Further, the role of gasdermin D (GSDMD) in TAD development was evaluated. Methods TADs were created by treating ascending aorta of adult mice (C57BL/6J) with active elastase (40.0 U/ml) and β-aminopropionitrile (Act E+BAPN). The temporal progress of the TAD pathology was rigorously characterized by histological evaluation and scanning electron microscopy, while potential mechanisms explored with bulk RNA sequencing of specimens collected at multiple timepoints. With this novel TAD model, further experiments were performed with Gsdmd -/- mice to evaluate its impact on TAD formation. Results The ascending aorta challenged with Act E+BAPN developed pathology characterized by an early onset of intimomedial tears (complete penetration) and intramural hematoma, followed by progressive medial loss and aortic dilation. Ingenuity Pathway Analysis and functional annotation of differentially expressed genes suggested that a unique inflammatory micro-environment, rather than general inflammation, promoted the onset of TADs by specifically recruiting neutrophils to the aortic wall, while the pathology at the advanced stage was driven by T-cell mediated immune injury. Gsdmd -/- attenuated medial loss, adventitial fibrosis, and dilation of TADs. This protective effect was associated with a reduced number of TUNEL (terminal deoxynucleotidyl transferase dUTP nick end labeling) positive cells and T-cells in TADs. Conclusions A novel mouse TAD model was created in the ascending aorta. It produces a unique microenvironment to activate different immune cell subsets, promoting onset and subsequent remodeling of TADs. Consistently, Gsdmd -/- attenuates TAD development, with modulation of cell death and T-cell response likely acting as the underlying mechanism.
Collapse
|
2
|
Gunnersen S, Shim JT, Liu F, Tietge UJ, Sørensen CB, Bentzon JF. Conditional deletion of Ccl2 in smooth muscle cells does not reduce early atherosclerosis in mice. ATHEROSCLEROSIS PLUS 2024; 55:12-20. [PMID: 38234375 PMCID: PMC10792688 DOI: 10.1016/j.athplu.2023.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 11/29/2023] [Accepted: 12/18/2023] [Indexed: 01/19/2024]
Abstract
Background and aims C-C motif chemokine ligand 2 (CCL2) is a pro-inflammatory chemokine important for monocyte recruitment to the arterial wall and atherosclerotic plaques. Global knockout of Ccl2 reduces plaque formation and macrophage content in mice, but the importance of different plaque cell types in mediating this effect has not been resolved. Smooth muscle cells (SMCs) can adopt a potentially pro-inflammatory function with expression of CCL2. The present study aimed to test the hypothesis that SMC-secreted CCL2 is involved in early atherogenesis in mice. Methods SMC-restricted Cre recombinase was activated at 6 weeks of age in mice with homozygous floxed or wildtype Ccl2 alleles. Separate experiments in mice lacking the Cre recombinase transgene were conducted to control for genetic background effects. Hypercholesterolemia and atherosclerosis were induced by a tail vein injection of recombinant adeno-associated virus (rAAV) encoding proprotein convertase subtilisin/kexin type 9 (PCSK9) and a high-fat diet for 12 weeks. Results Unexpectedly, mice with SMC-specific Ccl2 deletion developed higher levels of plasma cholesterol and larger atherosclerotic plaques with more macrophages compared with wild-type littermates. When total cholesterol levels were incorporated into the statistical analysis, none of the effects on plaque development between groups remained significant. Importantly, changes in plasma cholesterol and atherosclerosis remained in mice lacking Cre recombinase indicating that they were not caused by SMC-specific CCL2 deletion but by effects of the floxed allele or passenger genes. Conclusions SMC-specific deficiency of Ccl2 does not significantly affect early plaque development in hypercholesterolemic mice.
Collapse
Affiliation(s)
- Stine Gunnersen
- Atherosclerosis Research Unit, Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 11, 8200 Aarhus N, Denmark
- Department of Cardiology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200 Aarhus N, Denmark
| | - Jeong Tangkjær Shim
- Atherosclerosis Research Unit, Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 11, 8200 Aarhus N, Denmark
- Department of Cardiology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200 Aarhus N, Denmark
| | - Fan Liu
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet, 14183 Stockholm, Sweden
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
| | - Uwe J.F. Tietge
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet, 14183 Stockholm, Sweden
- Clinical Chemistry, Karolinska University Laboratory, Karolinska University Hospital, SE-14186 Stockholm, Sweden
| | - Charlotte Brandt Sørensen
- Atherosclerosis Research Unit, Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 11, 8200 Aarhus N, Denmark
- Department of Cardiology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200 Aarhus N, Denmark
| | - Jacob Fog Bentzon
- Atherosclerosis Research Unit, Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 11, 8200 Aarhus N, Denmark
- Department of Cardiology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200 Aarhus N, Denmark
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Calle de Melchor Fernández Almagro, 3, 28029 Madrid, Spain
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Palle Juul-Jensens Boulevard 11, 8200 Aarhus N, Denmark
| |
Collapse
|
3
|
Lin A, Brittan M, Baker AH, Dimmeler S, Fisher EA, Sluimer JC, Misra A. Clonal Expansion in Cardiovascular Pathology. JACC Basic Transl Sci 2024; 9:120-144. [PMID: 38362345 PMCID: PMC10864919 DOI: 10.1016/j.jacbts.2023.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 04/13/2023] [Accepted: 04/14/2023] [Indexed: 02/17/2024]
Abstract
Clonal expansion refers to the proliferation and selection of advantageous "clones" that are better suited for survival in a Darwinian manner. In recent years, we have greatly enhanced our understanding of cell clonality in the cardiovascular context. However, our knowledge of the underlying mechanisms behind this clonal selection is still severely limited. There is a transpiring pattern of clonal expansion of smooth muscle cells and endothelial cells-and, in some cases, macrophages-in numerous cardiovascular diseases irrespective of their differing microenvironments. These findings indirectly suggest the possible existence of stem-like vascular cells which are primed to respond during disease. Subsequent clones may undergo further phenotypic changes to adopt either protective or detrimental roles. By investigating these clone-forming vascular cells, we may be able to harness this inherent clonal nature for future therapeutic intervention. This review comprehensively discusses what is currently known about clonal expansion across the cardiovascular field. Comparisons of the clonal nature of vascular cells in atherosclerosis (including clonal hematopoiesis of indeterminate potential), pulmonary hypertension, aneurysm, blood vessel injury, ischemia- and tumor-induced angiogenesis, and cerebral cavernous malformations are evaluated. Finally, we discuss the potential clinical implications of these findings and propose that proper understanding and specific targeting of these clonal cells may provide unique therapeutic options for the treatment of these cardiovascular conditions.
Collapse
Affiliation(s)
- Alexander Lin
- Atherosclerosis and Vascular Remodeling Group, Heart Research Institute, Sydney, New South Wales, Australia
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Sydney, New South Wales, Australia
| | - Mairi Brittan
- Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Andrew H. Baker
- Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
- CARIM School for Cardiovascular Sciences, Department of Pathology, Maastricht University Medical Center (MUMC), Maastricht, the Netherlands
| | - Stefanie Dimmeler
- Institute for Cardiovascular Regeneration, Goethe University Frankfurt, Frankfurt, Germany
- German Center for Cardiovascular Research (DZHK), partner site Frankfurt Rhine-Main, Berlin, Germany
- Cardiopulmonary Institute, Goethe University Frankfurt, Frankfurt, Germany
| | - Edward A. Fisher
- Department of Medicine/Division of Cardiology, New York University Grossman School of Medicine, New York, New York, USA
- Cardiovascular Research Center, New York University Grossman School of Medicine, New York, New York, USA
| | - Judith C. Sluimer
- Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
- CARIM School for Cardiovascular Sciences, Department of Pathology, Maastricht University Medical Center (MUMC), Maastricht, the Netherlands
| | - Ashish Misra
- Atherosclerosis and Vascular Remodeling Group, Heart Research Institute, Sydney, New South Wales, Australia
- Heart Research Institute, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
4
|
Abstract
The medial layer of the arterial wall is composed mainly of vascular smooth muscle cells (VSMCs). Under physiological conditions, VSMCs assume a contractile phenotype, and their primary function is to regulate vascular tone. In contrast with terminally differentiated cells, VSMCs possess phenotypic plasticity, capable of transitioning into other cellular phenotypes in response to changes in the vascular environment. Recent research has shown that VSMC phenotypic switching participates in the pathogenesis of atherosclerosis, where the various types of dedifferentiated VSMCs accumulate in the atherosclerotic lesion and participate in the associated vascular remodeling by secreting extracellular matrix proteins and proteases. This review article discusses the 9 VSMC phenotypes that have been reported in atherosclerotic lesions and classifies them into differentiated VSMCs, intermediately dedifferentiated VSMCs, and dedifferentiated VSMCs. It also provides an overview of several methodologies that have been developed for studying VSMC phenotypic switching and discusses their respective advantages and limitations.
Collapse
Affiliation(s)
- Runji Chen
- Shantou University Medical CollegeShantouChina
| | - David G. McVey
- Department of Cardiovascular SciencesUniversity of LeicesterLeicesterUnited Kingdom
| | - Daifei Shen
- Research Center for Translational MedicineThe Second Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | | | - Shu Ye
- Shantou University Medical CollegeShantouChina
- Department of Cardiovascular SciencesUniversity of LeicesterLeicesterUnited Kingdom
- Cardiovascular‐Metabolic Disease Translational Research ProgrammeNational University of SingaporeSingapore
| |
Collapse
|
5
|
Gomez D. Nobody Is Perfect: Cre Drivers Deserve Careful Consideration. Arterioscler Thromb Vasc Biol 2023; 43:2071-2074. [PMID: 37650324 PMCID: PMC11507126 DOI: 10.1161/atvbaha.123.319683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Affiliation(s)
- Delphine Gomez
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh; Pittsburgh, PA, USA
- Division of Cardiology, Department of Medicine, University of Pittsburgh; Pittsburgh, PA, USA
| |
Collapse
|
6
|
Li J, Xu H, Hung A, Javed MJ, Upchurch GR, Jiang Z. Commonly Used Myh11-CreERT2Strain Carries a Y-Linked Functional Wild-Type Tlr7 Allele. Arterioscler Thromb Vasc Biol 2023; 43:2068-2070. [PMID: 37317852 PMCID: PMC10538427 DOI: 10.1161/atvbaha.122.318919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Affiliation(s)
- Jie Li
- Division of Vascular Surgery and Endovascular Therapy, University of Florida College of Medicine, Gainesville
| | - Haiyan Xu
- Division of Vascular Surgery and Endovascular Therapy, University of Florida College of Medicine, Gainesville
| | - Alex Hung
- Division of Vascular Surgery and Endovascular Therapy, University of Florida College of Medicine, Gainesville
| | - Muhammad Javad Javed
- Division of Vascular Surgery and Endovascular Therapy, University of Florida College of Medicine, Gainesville
| | - Gilbert R Upchurch
- Division of Vascular Surgery and Endovascular Therapy, University of Florida College of Medicine, Gainesville
| | - Zhihua Jiang
- Division of Vascular Surgery and Endovascular Therapy, University of Florida College of Medicine, Gainesville
| |
Collapse
|
7
|
Zhao Y, Zhao G, Chang Z, Zhu T, Zhao Y, Lu H, Xue C, Saunders TL, Guo Y, Chang L, Chen YE, Zhang J. Generating endogenous Myh11-driven Cre mice for sex-independent gene deletion in smooth muscle cells. JCI Insight 2023; 8:e171661. [PMID: 37289544 PMCID: PMC10443793 DOI: 10.1172/jci.insight.171661] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 06/05/2023] [Indexed: 06/10/2023] Open
Abstract
Specific and efficient smooth muscle cell-targeted (SMC-targeted) gene deletion is typically achieved by pairing SMMHC-CreERT2-Tg mice with mice carrying the loxP-flanked gene. However, the transgene, CreERT2, is not controlled by the endogenous Myh11 gene promoter, and the codon-modified iCreERT2 exhibits significant tamoxifen-independent leakage. Furthermore, because the Cre-bearing bacterial artificial chromosome (BAC) is inserted onto the Y chromosome, the SMMHC-CreERT2-Tg mice strain can only exhibit gene deletions in male mice. Additionally, there is a lack of Myh11-driven constitutive Cre mice when tamoxifen usage is a concern. We used CRISPR/Cas9-mediated homologous recombination between a donor vector carrying the CreNLSP2A or CreERT2-P2A sequence and homologous arm surrounding the translation start site of the Myh11 gene to generate Cre-knockin mice. The P2A sequence enables the simultaneous translation of Cre and endogenous proteins. Using reporter mice, we assessed Cre-mediated recombination efficiency, specificity, tamoxifen-dependent controllability, and functionality in both sexes. Both constitutive (Myh11-CreNLSP2A) and inducible (Myh11-CreERT2-P2A) Cre mice demonstrated efficient, SMC-specific, sex-independent Cre recombinase activity without confounding endogenous gene expression. Combined with recently generated BAC transgenic Myh11-CreERT2-RAD mice and the Itga8-CreERT2 mouse models, our models will help expand the research toolbox, facilitating unbiased and comprehensive research in SMCs and SMC-dependent cardiovascular diseases.
Collapse
Affiliation(s)
- Yang Zhao
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
- Department of Pharmacology and
| | - Guizhen Zhao
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Ziyi Chang
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Tianqing Zhu
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Ying Zhao
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Haocheng Lu
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Chao Xue
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Thomas L. Saunders
- Transgenic Animal Model Core, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Yanhong Guo
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Lin Chang
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Y. Eugene Chen
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Jifeng Zhang
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| |
Collapse
|
8
|
Zhang C, Li Y, Chakraborty A, Li Y, Rebello KR, Ren P, Luo W, Zhang L, Lu HS, Cassis LA, Coselli JS, Daugherty A, LeMaire SA, Shen YH. Aortic Stress Activates an Adaptive Program in Thoracic Aortic Smooth Muscle Cells That Maintains Aortic Strength and Protects Against Aneurysm and Dissection in Mice. Arterioscler Thromb Vasc Biol 2023; 43:234-252. [PMID: 36579645 PMCID: PMC9877188 DOI: 10.1161/atvbaha.122.318135] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 12/08/2022] [Indexed: 12/30/2022]
Abstract
BACKGROUND When aortic cells are under stress, such as increased hemodynamic pressure, they adapt to the environment by modifying their functions, allowing the aorta to maintain its strength. To understand the regulation of this adaptive response, we examined transcriptomic and epigenomic programs in aortic smooth muscle cells (SMCs) during the adaptive response to AngII (angiotensin II) infusion and determined its importance in protecting against aortic aneurysm and dissection (AAD). METHODS We performed single-cell RNA sequencing and single-cell sequencing assay for transposase-accessible chromatin (scATAC-seq) analyses in a mouse model of sporadic AAD induced by AngII infusion. We also examined the direct effects of YAP (yes-associated protein) on the SMC adaptive response in vitro. The role of YAP in AAD development was further evaluated in AngII-infused mice with SMC-specific Yap deletion. RESULTS In wild-type mice, AngII infusion increased medial thickness in the thoracic aorta. Single-cell RNA sequencing analysis revealed an adaptive response in thoracic SMCs characterized by upregulated genes with roles in wound healing, elastin and collagen production, proliferation, migration, cytoskeleton organization, cell-matrix focal adhesion, and PI3K-PKB/Akt (phosphoinositide-3-kinase-protein kinase B/Akt) and TGF-β (transforming growth factor beta) signaling. ScATAC-seq analysis showed increased chromatin accessibility at regulatory regions of adaptive genes and revealed the mechanical sensor YAP/transcriptional enhanced associate domains as a top candidate transcription complex driving the expression of these genes (eg, Lox, Col5a2, Tgfb2). In cultured human aortic SMCs, cyclic stretch activated YAP, which directly bound to adaptive gene regulatory regions (eg, Lox) and increased their transcript abundance. SMC-specific Yap deletion in mice compromised this adaptive response in SMCs, leading to an increased AAD incidence. CONCLUSIONS Aortic stress triggers the systemic epigenetic induction of an adaptive response (eg, wound healing, proliferation, matrix organization) in thoracic aortic SMCs that depends on functional biomechanical signal transduction (eg, YAP signaling). Our study highlights the importance of the adaptive response in maintaining aortic homeostasis and preventing AAD in mice.
Collapse
Affiliation(s)
- Chen Zhang
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX (C.Z., Y.L., A.C., Y.L., K.R.R., P.R., W.L., L.Z., J.S.C., S.A.L., Y.H.S.)
- Texas Heart Institute, Houston (C.Z., Y.L., A.C., Y.L., K.R.R., P.R., W.L., L.Z., J.S.C., S.A.L.)
| | - Yanming Li
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX (C.Z., Y.L., A.C., Y.L., K.R.R., P.R., W.L., L.Z., J.S.C., S.A.L., Y.H.S.)
- Texas Heart Institute, Houston (C.Z., Y.L., A.C., Y.L., K.R.R., P.R., W.L., L.Z., J.S.C., S.A.L.)
| | - Abhijit Chakraborty
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX (C.Z., Y.L., A.C., Y.L., K.R.R., P.R., W.L., L.Z., J.S.C., S.A.L., Y.H.S.)
- Texas Heart Institute, Houston (C.Z., Y.L., A.C., Y.L., K.R.R., P.R., W.L., L.Z., J.S.C., S.A.L.)
| | - Yang Li
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX (C.Z., Y.L., A.C., Y.L., K.R.R., P.R., W.L., L.Z., J.S.C., S.A.L., Y.H.S.)
- Texas Heart Institute, Houston (C.Z., Y.L., A.C., Y.L., K.R.R., P.R., W.L., L.Z., J.S.C., S.A.L.)
| | - Kimberly R Rebello
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX (C.Z., Y.L., A.C., Y.L., K.R.R., P.R., W.L., L.Z., J.S.C., S.A.L., Y.H.S.)
- Texas Heart Institute, Houston (C.Z., Y.L., A.C., Y.L., K.R.R., P.R., W.L., L.Z., J.S.C., S.A.L.)
| | - Pingping Ren
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX (C.Z., Y.L., A.C., Y.L., K.R.R., P.R., W.L., L.Z., J.S.C., S.A.L., Y.H.S.)
- Texas Heart Institute, Houston (C.Z., Y.L., A.C., Y.L., K.R.R., P.R., W.L., L.Z., J.S.C., S.A.L.)
| | - Wei Luo
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX (C.Z., Y.L., A.C., Y.L., K.R.R., P.R., W.L., L.Z., J.S.C., S.A.L., Y.H.S.)
- Texas Heart Institute, Houston (C.Z., Y.L., A.C., Y.L., K.R.R., P.R., W.L., L.Z., J.S.C., S.A.L.)
| | - Lin Zhang
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX (C.Z., Y.L., A.C., Y.L., K.R.R., P.R., W.L., L.Z., J.S.C., S.A.L., Y.H.S.)
- Texas Heart Institute, Houston (C.Z., Y.L., A.C., Y.L., K.R.R., P.R., W.L., L.Z., J.S.C., S.A.L.)
| | - Hong S Lu
- Saha Cardiovascular Research Center (H.S.L., A.D.), University of Kentucky, Lexington
- Department of Physiology (H.S.L., A.D.), University of Kentucky, Lexington
| | - Lisa A Cassis
- Department of Pharmacology and Nutritional Sciences (L.A.C.), University of Kentucky, Lexington
| | - Joseph S Coselli
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX (C.Z., Y.L., A.C., Y.L., K.R.R., P.R., W.L., L.Z., J.S.C., S.A.L., Y.H.S.)
- Texas Heart Institute, Houston (C.Z., Y.L., A.C., Y.L., K.R.R., P.R., W.L., L.Z., J.S.C., S.A.L.)
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX (J.S.C., S.A.L., Y.S.)
| | - Alan Daugherty
- Saha Cardiovascular Research Center (H.S.L., A.D.), University of Kentucky, Lexington
- Department of Physiology (H.S.L., A.D.), University of Kentucky, Lexington
| | - Scott A LeMaire
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX (C.Z., Y.L., A.C., Y.L., K.R.R., P.R., W.L., L.Z., J.S.C., S.A.L., Y.H.S.)
- Texas Heart Institute, Houston (C.Z., Y.L., A.C., Y.L., K.R.R., P.R., W.L., L.Z., J.S.C., S.A.L.)
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX (J.S.C., S.A.L., Y.S.)
| | - Ying H Shen
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX (C.Z., Y.L., A.C., Y.L., K.R.R., P.R., W.L., L.Z., J.S.C., S.A.L., Y.H.S.)
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX (J.S.C., S.A.L., Y.S.)
| |
Collapse
|
9
|
Deaton RA, Bulut G, Serbulea V, Salamon A, Shankman LS, Nguyen AT, Owens GK. A New Autosomal Myh11-CreERT2 Smooth Muscle Cell Lineage Tracing and Gene Knockout Mouse Model-Brief Report. Arterioscler Thromb Vasc Biol 2023; 43:203-211. [PMID: 36519470 PMCID: PMC9877184 DOI: 10.1161/atvbaha.122.318160] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 12/01/2022] [Indexed: 12/23/2022]
Abstract
BACKGROUND The Myh11 promoter is extensively used as a smooth muscle cell (SMC) Cre-driver and is regarded as the most restrictive and specific promoter available to study SMCs. Unfortunately, in the existing Myh11-CreERT2 mouse, the transgene was inserted on the Y chromosome precluding the study of female mice. Given the importance of including sex as a biological variable and that numerous SMC-based diseases have a sex-dependent bias, the field has been tremendously limited by the lack of a model to study both sexes. Here, we describe a new autosomal Myh11-CreERT2 mouse (referred to as Myh11-CreERT2-RAD), which allows for SMC-specific lineage tracing and gene knockout studies in vivo using both male and female mice. METHODS A Myh11-CreERT2-RAD transgenic C57BL/6 mouse line was generated using bacterial artificial chromosome clone RP23-151J22 modified to contain a Cre-ERT2 after the Myh11 start codon. Myh11-CreERT2-RAD mice were crossed with 2 different fluorescent reporter mice and tested for SMC-specific labeling by flow cytometric and immunofluorescence analyses. RESULTS Myh11-CreERT2-RAD transgene insertion was determined to be on mouse chromosome 2. Myh11-CreERT2-RAD fluorescent reporter mice showed Cre-dependent, tamoxifen-inducible labeling of SMCs equivalent to the widely used Myh11-CreERT2 mice. Labeling was equivalent in both male and female Cre+ mice and was limited to vascular and visceral SMCs and pericytes in various tissues as assessed by immunofluorescence. CONCLUSIONS We generated and validated the function of an autosomal Myh11-CreERT2-RAD mouse that can be used to assess sex as a biological variable with respect to the normal and pathophysiological functions of SMCs.
Collapse
Affiliation(s)
- Rebecca A. Deaton
- Robert M. Berne Cardiovascular Research Center, University of Virginia-School of Medicine, 415 Lane Road, Suite 1010, Charlottesville, VA, 22908
| | - Gamze Bulut
- College of William and Mary, Williamsburg, Virginia 23185
| | - Vlad Serbulea
- Robert M. Berne Cardiovascular Research Center, University of Virginia-School of Medicine, 415 Lane Road, Suite 1010, Charlottesville, VA, 22908
| | - Anita Salamon
- Robert M. Berne Cardiovascular Research Center, University of Virginia-School of Medicine, 415 Lane Road, Suite 1010, Charlottesville, VA, 22908
| | - Laura S. Shankman
- Robert M. Berne Cardiovascular Research Center, University of Virginia-School of Medicine, 415 Lane Road, Suite 1010, Charlottesville, VA, 22908
| | | | - Gary K. Owens
- Robert M. Berne Cardiovascular Research Center, University of Virginia-School of Medicine, 415 Lane Road, Suite 1010, Charlottesville, VA, 22908
| |
Collapse
|
10
|
Warthi G, Faulkner JL, Doja J, Ghanam AR, Gao P, Yang AC, Slivano OJ, Barris CT, Kress TC, Zawieja SD, Griffin SH, Xie X, Ashworth A, Christie CK, Bryant WB, Kumar A, Davis MJ, Long X, Gan L, de Chantemèle EJB, Lyu Q, Miano JM. Generation and Comparative Analysis of an Itga8-CreER T2 Mouse with Preferential Activity in Vascular Smooth Muscle Cells. NATURE CARDIOVASCULAR RESEARCH 2022; 1:1084-1100. [PMID: 36424917 PMCID: PMC9681021 DOI: 10.1038/s44161-022-00162-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 09/29/2022] [Indexed: 11/12/2022]
Abstract
All current smooth muscle cell (SMC) Cre mice similarly recombine floxed alleles in vascular and visceral SMCs. Here, we present an Itga8-CreER T2 knock-in mouse and compare its activity with a Myh11-CreER T2 mouse. Both Cre drivers demonstrate equivalent recombination in vascular SMCs. However, Myh11-CreER T2 mice, but not Itga8-CreER T2 mice, display high activity in visceral SMC-containing tissues such as intestine, show early tamoxifen-independent activity, and produce high levels of CreERT2 protein. Whereas Myh11-CreER T2 -mediated knockout of serum response factor (Srf) causes a lethal intestinal phenotype precluding analysis of the vasculature, loss of Srf with Itga8-CreER T2 (Srf Itga8 ) yields viable mice with no evidence of intestinal pathology. Male and female Srf Itga8 mice exhibit vascular contractile incompetence, and angiotensin II causes elevated blood pressure in wild type, but not Srf Itga8 , male mice. These findings establish the Itga8-CreER T2 mouse as an alternative to existing SMC Cre mice for unfettered phenotyping of vascular SMCs following selective gene loss.
Collapse
Affiliation(s)
- Ganesh Warthi
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| | - Jessica L. Faulkner
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| | - Jaser Doja
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| | - Amr R. Ghanam
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| | - Pan Gao
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| | - Allison C. Yang
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| | - Orazio J. Slivano
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| | - Candee T. Barris
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| | - Taylor C. Kress
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| | - Scott D. Zawieja
- Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, MO 65212
| | - Susan H. Griffin
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| | - Xiaoling Xie
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| | - Alan Ashworth
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, 94158
| | - Christine K. Christie
- Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642
| | - William B. Bryant
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| | - Ajay Kumar
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| | - Michael J. Davis
- Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, MO 65212
| | - Xiaochun Long
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| | - Lin Gan
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| | | | - Qing Lyu
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
- Biomedical and Health Institute, Chongqing Institute of Green and Intelligence Technology, Chongqing, China 400714
- Chongqing General Hospital, Chongqing, China 401147
| | - Joseph M. Miano
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| |
Collapse
|
11
|
Gogiraju R, Gachkar S, Velmeden D, Bochenek ML, Zifkos K, Hubert A, Münzel T, Offermanns S, Schäfer K. Protein Tyrosine Phosphatase 1B Deficiency in Vascular Smooth Muscle Cells Promotes Perivascular Fibrosis following Arterial Injury. Thromb Haemost 2022; 122:1814-1826. [PMID: 36075234 PMCID: PMC9512587 DOI: 10.1055/s-0042-1755329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Background
Smooth muscle cell (SMC) phenotype switching plays a central role during vascular remodeling. Growth factor receptors are negatively regulated by protein tyrosine phosphatases (PTPs), including its prototype PTP1B. Here, we examine how reduction of PTP1B in SMCs affects the vascular remodeling response to injury.
Methods
Mice with inducible PTP1B deletion in SMCs (SMC.PTP1B-KO) were generated by crossing mice expressing Cre.ER
T2
recombinase under the
Myh11
promoter with PTP1B
flox/flox
mice and subjected to FeCl
3
carotid artery injury.
Results
Genetic deletion of PTP1B in SMCs resulted in adventitia enlargement, perivascular SMA
+
and PDGFRβ
+
myofibroblast expansion, and collagen accumulation following vascular injury. Lineage tracing confirmed the appearance of
Myh11
-Cre reporter cells in the remodeling adventitia, and SCA1
+
CD45
-
vascular progenitor cells increased. Elevated mRNA expression of transforming growth factor β (TGFβ) signaling components or enzymes involved in extracellular matrix remodeling and TGFβ liberation was seen in injured SMC.PTP1B-KO mouse carotid arteries, and mRNA transcript levels of contractile SMC marker genes were reduced already at baseline. Mechanistically, Cre recombinase (mice) or siRNA (cells)-mediated downregulation of PTP1B or inhibition of ERK1/2 signaling in SMCs resulted in nuclear accumulation of KLF4, a central transcriptional repressor of SMC differentiation, whereas phosphorylation and nuclear translocation of SMAD2 and SMAD3 were reduced. SMAD2 siRNA transfection increased protein levels of PDGFRβ and MYH10 while reducing ERK1/2 phosphorylation, thus phenocopying genetic PTP1B deletion.
Conclusion
Chronic reduction of PTP1B in SMCs promotes dedifferentiation, perivascular fibrosis, and adverse remodeling following vascular injury by mechanisms involving an ERK1/2 phosphorylation-driven shift from SMAD2 to KLF4-regulated gene transcription.
Collapse
Affiliation(s)
- Rajinikanth Gogiraju
- Department of Cardiology, Cardiology I, University Medical Center Mainz, Mainz, Germany
| | - Sogol Gachkar
- Department of Cardiology, Cardiology I, University Medical Center Mainz, Mainz, Germany
| | - David Velmeden
- Department of Cardiology, Cardiology I, University Medical Center Mainz, Mainz, Germany
| | - Magdalena L Bochenek
- Department of Cardiology, Cardiology I, University Medical Center Mainz, Mainz, Germany.,Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
| | - Konstantinos Zifkos
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
| | - Astrid Hubert
- Department of Cardiology, Cardiology I, University Medical Center Mainz, Mainz, Germany
| | - Thomas Münzel
- Department of Cardiology, Cardiology I, University Medical Center Mainz, Mainz, Germany.,German Center for Cardiovascular Research (DZHK), Rhine-Main Site, Mainz, Germany
| | - Stefan Offermanns
- Department of Pharmacology, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany.,Centre for Molecular Medicine, Medical Faculty, JW Goethe University Frankfurt, Frankfurt, Germany.,Cardiopulmonary Institute (CPI), Frankfurt, Germany.,German Center for Cardiovascular Research (DZHK e.V.), Rhine-Main Site, Frankfurt and Bad Nauheim, Germany
| | - Katrin Schäfer
- Department of Cardiology, Cardiology I, University Medical Center Mainz, Mainz, Germany.,German Center for Cardiovascular Research (DZHK), Rhine-Main Site, Mainz, Germany
| |
Collapse
|
12
|
Zou F, Li Y, Zhang S, Zhang J. DP1 (Prostaglandin D 2 Receptor 1) Activation Protects Against Vascular Remodeling and Vascular Smooth Muscle Cell Transition to Myofibroblasts in Angiotensin II-Induced Hypertension in Mice. Hypertension 2022; 79:1203-1215. [PMID: 35354317 DOI: 10.1161/hypertensionaha.121.17584] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Vascular smooth muscle cell (VSMC) phenotype transition plays an essential role in vascular remodeling. PGD2 (Prostaglandin D2) is involved in cardiovascular inflammation. In this study, we aimed to investigates the role of DP1 (PGD2 receptor 1) on VSMC phenotype transition in vascular remodeling after Ang II (angiotensin II) infusion in mice. METHODS VSMC-specific DP1 knockout mice and DP1flox/flox mice were infused with Ang II for 28 days and systolic blood pressure was measured by noninvasive tail-cuff system. The arterial samples were applied to an unbiased proteome analysis. DP1f/f Myh11 (myosin heavy chain 11) CREERT2 R26mTmG/+ mice were generated for VSMC lineage tracing. Multiple genetic and pharmacological approaches were used to investigate DP1-mediated signaling in phenotypic transition of VSMCs in response to Ang II administration. RESULTS DP1 knockout promoted vascular media thickness and increased systolic blood pressure after Ang II infusion by impairing Epac (exchange protein directly activated by cAMP)-1-mediated Rap-1 (Ras-related protein 1) activation. The DP1 agonist facilitated the interaction of myocardin-related transcription factor A and G-actin, which subsequently inhibited the VSMC transition to myofibroblasts through the suppression of RhoA (Ras homolog family member A)/ROCK-1 (Rho associated coiled-coil containing protein kinase 1) activity. Moreover, Epac-1 overexpression by lentivirus blocked the progression of vascular fibrosis in DP1 deficient mice in response to Ang II infusion. CONCLUSIONS Our finding revealed a protective role of DP1 in VSMC switch to myofibroblasts by impairing the phosphorylation of MRTF (myocardin-related transcription factor)-A by ROCK-1 through Epac-1/Rap-1/RhoA pathway and thus inhibited the expression of collagen I, fibronectin, ED-A (extra domain A) fibronectin, and vinculin. Thus, DP1 activation has therapeutic potential for vascular fibrosis in hypertension.
Collapse
Affiliation(s)
- Fangdi Zou
- Department of Pharmacology, School of Basic Medical Sciences (F.Z., Y.L., S.Z., J.Z.), Tianjin Medical University, China.,School of Pharmacy (F.Z.), Tianjin Medical University, China
| | - Yong Li
- Department of Pharmacology, School of Basic Medical Sciences (F.Z., Y.L., S.Z., J.Z.), Tianjin Medical University, China
| | - Shijie Zhang
- Department of Pharmacology, School of Basic Medical Sciences (F.Z., Y.L., S.Z., J.Z.), Tianjin Medical University, China
| | - Jian Zhang
- Department of Pharmacology, School of Basic Medical Sciences (F.Z., Y.L., S.Z., J.Z.), Tianjin Medical University, China.,School of Pharmacy, East China University of Science and Technology, Shanghai, China (J.Z.)
| |
Collapse
|
13
|
Qi X, Wang F, Chun C, Saldarriaga L, Jiang Z, Pruitt EY, Arnaoutakis GJ, Upchurch GR, Jiang Z. A validated mouse model capable of recapitulating the protective effects of female sex hormones on ascending aortic aneurysms and dissections (AADs). Physiol Rep 2020; 8:e14631. [PMID: 33242364 PMCID: PMC7690909 DOI: 10.14814/phy2.14631] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 09/28/2020] [Accepted: 10/07/2020] [Indexed: 12/19/2022] Open
Abstract
Fewer females develop AADs (ascending aortic aneurysms and dissections) and the reasons for this protection remain poorly understood. The present study seeks to develop a mouse model that may be utilized to address this sexual dimorphism. Adult normolipidemic mice were challenged with BAPN (β-aminopropionitrile), AngII (angiotensin II), or BAPN + AngII. An initial protocol optimization found that 0.2% BAPN in drinking water plus AngII-infusion at 1,000 ng kg-1 min-1 produced favorable rates of AAD rupture (~50%) and dilation (~40%) in 28 days. Using these dosages, further experiments revealed that BAPN is toxic to naïve mature aortas and it acted synergistically with AngII to promote aortic tears and dissections. BAPN + AngII provoked early infiltration of myeloid cells and subsequent recruitment of lymphoid cells to the aortic wall. AADs established with BAPN + AngII, but not AngII alone, continued to expand after the cessation of AngII-infusion. This indefinite growth precipitated a 61% increase in the AAD diameter in 56 days. More importantly, with the optimized protocol, significant differences in AAD dilation (p = .012) and medial degeneration (p = .036) were detected between male and female mice. Treatment of ovariectomized mice with estradiol protected AAD formation (p = .014). In summary, this study developed a powerful mouse AAD model that can be used to study the sexual dimorphism in AAD formation.
Collapse
Affiliation(s)
- Xiaoyan Qi
- Division of Vascular Surgery and Endovascular TherapyUniversity of Florida College of MedicineGainesvilleFLUSA
- Institute of Cardiovascular DiseaseUniversity of South ChinaHengyangChina
| | - Fen Wang
- Division of Vascular Surgery and Endovascular TherapyUniversity of Florida College of MedicineGainesvilleFLUSA
| | - Changzoon Chun
- Division of Vascular Surgery and Endovascular TherapyUniversity of Florida College of MedicineGainesvilleFLUSA
| | - Lennon Saldarriaga
- Division of Vascular Surgery and Endovascular TherapyUniversity of Florida College of MedicineGainesvilleFLUSA
| | - Zhisheng Jiang
- Institute of Cardiovascular DiseaseUniversity of South ChinaHengyangChina
| | - Eric Y. Pruitt
- Division of Vascular Surgery and Endovascular TherapyUniversity of Florida College of MedicineGainesvilleFLUSA
| | - George J. Arnaoutakis
- Division of Vascular Surgery and Endovascular TherapyUniversity of Florida College of MedicineGainesvilleFLUSA
- Division of Thoracic and Cardiovascular SurgeryUniversity of Florida College of MedicineGainesvilleFLUSA
| | - Gilbert R. Upchurch
- Division of Vascular Surgery and Endovascular TherapyUniversity of Florida College of MedicineGainesvilleFLUSA
| | - Zhihua Jiang
- Division of Vascular Surgery and Endovascular TherapyUniversity of Florida College of MedicineGainesvilleFLUSA
| |
Collapse
|
14
|
Chakraborty R, Saddouk FZ, Carrao AC, Krause DS, Greif DM, Martin KA. Promoters to Study Vascular Smooth Muscle. Arterioscler Thromb Vasc Biol 2020; 39:603-612. [PMID: 30727757 DOI: 10.1161/atvbaha.119.312449] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Smooth muscle cells (SMCs) are a critical component of blood vessel walls that provide structural support, regulate vascular tone, and allow for vascular remodeling. These cells also exhibit a remarkable plasticity that contributes to vascular growth and repair but also to cardiovascular pathologies, including atherosclerosis, intimal hyperplasia and restenosis, aneurysm, and transplant vasculopathy. Mouse models have been an important tool for the study of SMC functions. The development of smooth muscle-expressing Cre-driver lines has allowed for exciting discoveries, including recent advances revealing the diversity of phenotypes derived from mature SMC transdifferentiation in vivo using inducible CreER T2 lines. We review SMC-targeting Cre lines driven by the Myh11, Tagln, and Acta2 promoters, including important technical considerations associated with these models. Limitations that can complicate study of the vasculature include expression in visceral SMCs leading to confounding phenotypes, and expression in multiple nonsmooth muscle cell types, such as Acta2-Cre expression in myofibroblasts. Notably, the frequently employed Tagln/ SM22α- Cre driver expresses in the embryonic heart but can also confer expression in nonmuscular cells including perivascular adipocytes and their precursors, myeloid cells, and platelets, with important implications for interpretation of cardiovascular phenotypes. With new Cre-driver lines under development and the increasing use of fate mapping methods, we are entering an exciting new era in SMC research.
Collapse
Affiliation(s)
- Raja Chakraborty
- From the Department of Medicine, Section of Cardiovascular Medicine (R.C., F.Z.S., A.C.C., D.M.G., K.A.M.)
| | - Fatima Zahra Saddouk
- From the Department of Medicine, Section of Cardiovascular Medicine (R.C., F.Z.S., A.C.C., D.M.G., K.A.M.).,Department of Genetics (F.Z.S., D.M.G.)
| | - Ana Catarina Carrao
- From the Department of Medicine, Section of Cardiovascular Medicine (R.C., F.Z.S., A.C.C., D.M.G., K.A.M.)
| | - Diane S Krause
- Departments of Laboratory Medicine, Cell Biology, and Pathology (D.S.K.)
| | - Daniel M Greif
- From the Department of Medicine, Section of Cardiovascular Medicine (R.C., F.Z.S., A.C.C., D.M.G., K.A.M.).,Department of Genetics (F.Z.S., D.M.G.)
| | - Kathleen A Martin
- From the Department of Medicine, Section of Cardiovascular Medicine (R.C., F.Z.S., A.C.C., D.M.G., K.A.M.).,Department of Pharmacology (K.A.M.), Yale University School of Medicine, New Haven, CT
| |
Collapse
|
15
|
Zhou G, Liao M, Wang F, Qi X, Yang P, Berceli SA, Sharma AK, Upchurch GR, Jiang Z. Cyclophilin A contributes to aortopathy induced by postnatal loss of smooth muscle TGFBR1. FASEB J 2019; 33:11396-11410. [PMID: 31311317 PMCID: PMC6766662 DOI: 10.1096/fj.201900601rr] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 06/25/2019] [Indexed: 12/12/2022]
Abstract
Recent recognition that TGF-β signaling disruption is involved in the development of aortic aneurysms has led to renewed investigations into the role of TGF-β biology in the aortic wall. We previously found that the type I receptor of TGF-β (TGFBR2) receptor contributes to formation of ascending aortic aneurysms and dissections (AADs) induced by smooth muscle cell (SMC)-specific, postnatal deletion of Tgfbr1 (Tgfbr1iko). Here, we aimed to decipher the mechanistic signaling pathway underlying the pathogenic effects of TGFBR2 in this context. Gene expression profiling demonstrated that Tgfbr1iko triggers an acute inflammatory response in developing AADs, and Tgfbr1iko SMCs express an inflammatory phenotype in culture. Comparative proteomics profiling and mass spectrometry revealed that Tgfbr1iko SMCs respond to TGF-β1 stimulation via robust up-regulation of cyclophilin A (CypA). This up-regulation is abrogated by inhibition of TGFBR2 kinase activity, small interfering RNA silencing of Tgfbr2 expression, or inhibition of SMAD3 activation. In mice, Tgfbr1iko rapidly promotes CypA production in SMCs of developing AADs, whereas treatment with a CypA inhibitor attenuates aortic dilation by 56% (P = 0.003) and ameliorates aneurysmal degeneration (P = 0.016). These protective effects are associated with reduced aneurysm-promoting inflammation. Collectively, these results suggest a novel mechanism, wherein loss of type I receptor of TGF-β triggers promiscuous, proinflammatory TGFBR2 signaling in SMCs, thereby promoting AAD formation.-Zhou, G., Liao, M., Wang, F., Qi, X., Yang, P., Berceli, S. A., Sharma, A. K., Upchurch, G. R., Jr., Jiang, Z. Cyclophilin A contributes to aortopathy induced by postnatal loss of smooth muscle TGFBR1.
Collapse
Affiliation(s)
- Guannan Zhou
- Division of Vascular Surgery and Endovascular Therapy, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Mingmei Liao
- Division of Vascular Surgery and Endovascular Therapy, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Fen Wang
- Division of Vascular Surgery and Endovascular Therapy, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Xiaoyan Qi
- Division of Vascular Surgery and Endovascular Therapy, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Pu Yang
- Division of Vascular Surgery and Endovascular Therapy, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Scott A. Berceli
- Division of Vascular Surgery and Endovascular Therapy, University of Florida College of Medicine, Gainesville, Florida, USA
- Malcom Randall Veterans Affairs Medical Center, Gainesville, Florida, USA
| | - Ashish K. Sharma
- Division of Vascular Surgery and Endovascular Therapy, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Gilbert R. Upchurch
- Division of Vascular Surgery and Endovascular Therapy, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Zhihua Jiang
- Division of Vascular Surgery and Endovascular Therapy, University of Florida College of Medicine, Gainesville, Florida, USA
| |
Collapse
|
16
|
Peterson SM, Turner JE, Harrington A, Davis-Knowlton J, Lindner V, Gridley T, Vary CPH, Liaw L. Notch2 and Proteomic Signatures in Mouse Neointimal Lesion Formation. Arterioscler Thromb Vasc Biol 2018; 38:1576-1593. [PMID: 29853569 PMCID: PMC6023756 DOI: 10.1161/atvbaha.118.311092] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Accepted: 05/16/2018] [Indexed: 12/29/2022]
Abstract
Supplemental Digital Content is available in the text. Objective— Vascular remodeling is associated with complex molecular changes, including increased Notch2, which promotes quiescence in human smooth muscle cells. We used unbiased protein profiling to understand molecular signatures related to neointimal lesion formation in the presence or absence of Notch2 and to test the hypothesis that loss of Notch2 would increase neointimal lesion formation because of a hyperproliferative injury response. Approach and Results— Murine carotid arteries isolated at 6 or 14 days after ligation injury were analyzed by mass spectrometry using a data-independent acquisition strategy in comparison to uninjured or sham injured arteries. We used a tamoxifen-inducible, cell-specific Cre recombinase strain to delete the Notch2 gene in smooth muscle cells. Vessel morphometric analysis and immunohistochemical staining were used to characterize lesion formation, assess vascular smooth muscle cell proliferation, and validate proteomic findings. Loss of Notch2 in smooth muscle cells leads to protein profile changes in the vessel wall during remodeling but does not alter overall lesion morphology or cell proliferation. Loss of smooth muscle Notch2 also decreases the expression of enhancer of rudimentary homolog, plectin, and annexin A2 in vascular remodeling. Conclusions— We identified unique protein signatures that represent temporal changes in the vessel wall during neointimal lesion formation in the presence and absence of Notch2. Overall lesion formation was not affected with loss of smooth muscle Notch2, suggesting compensatory pathways. We also validated the regulation of known injury- or Notch-related targets identified in other vascular contexts, providing additional insight into conserved pathways involved in vascular remodeling.
Collapse
Affiliation(s)
- Sarah M Peterson
- From the Maine Medical Center Research Institute, Scarborough (S.M.P., J.E.T., A.H., J.D.-K., V.L., T.G., C.P.H.V., L.L.).,University of Maine Graduate School of Biomedical Science and Engineering, Orono (S.M.P., V.L., T.G., C.P.H.V., L.L.)
| | - Jacqueline E Turner
- From the Maine Medical Center Research Institute, Scarborough (S.M.P., J.E.T., A.H., J.D.-K., V.L., T.G., C.P.H.V., L.L.)
| | - Anne Harrington
- From the Maine Medical Center Research Institute, Scarborough (S.M.P., J.E.T., A.H., J.D.-K., V.L., T.G., C.P.H.V., L.L.)
| | - Jessica Davis-Knowlton
- From the Maine Medical Center Research Institute, Scarborough (S.M.P., J.E.T., A.H., J.D.-K., V.L., T.G., C.P.H.V., L.L.).,Tufts Sackler School of Graduate Biomedical Sciences, Boston, MA (J.D.-K., V.L., T.G., C.P.H.V., L.L.)
| | - Volkhard Lindner
- From the Maine Medical Center Research Institute, Scarborough (S.M.P., J.E.T., A.H., J.D.-K., V.L., T.G., C.P.H.V., L.L.).,University of Maine Graduate School of Biomedical Science and Engineering, Orono (S.M.P., V.L., T.G., C.P.H.V., L.L.).,Tufts Sackler School of Graduate Biomedical Sciences, Boston, MA (J.D.-K., V.L., T.G., C.P.H.V., L.L.)
| | - Thomas Gridley
- From the Maine Medical Center Research Institute, Scarborough (S.M.P., J.E.T., A.H., J.D.-K., V.L., T.G., C.P.H.V., L.L.).,University of Maine Graduate School of Biomedical Science and Engineering, Orono (S.M.P., V.L., T.G., C.P.H.V., L.L.).,Tufts Sackler School of Graduate Biomedical Sciences, Boston, MA (J.D.-K., V.L., T.G., C.P.H.V., L.L.)
| | - Calvin P H Vary
- From the Maine Medical Center Research Institute, Scarborough (S.M.P., J.E.T., A.H., J.D.-K., V.L., T.G., C.P.H.V., L.L.).,University of Maine Graduate School of Biomedical Science and Engineering, Orono (S.M.P., V.L., T.G., C.P.H.V., L.L.).,Tufts Sackler School of Graduate Biomedical Sciences, Boston, MA (J.D.-K., V.L., T.G., C.P.H.V., L.L.)
| | - Lucy Liaw
- From the Maine Medical Center Research Institute, Scarborough (S.M.P., J.E.T., A.H., J.D.-K., V.L., T.G., C.P.H.V., L.L.) .,University of Maine Graduate School of Biomedical Science and Engineering, Orono (S.M.P., V.L., T.G., C.P.H.V., L.L.).,Tufts Sackler School of Graduate Biomedical Sciences, Boston, MA (J.D.-K., V.L., T.G., C.P.H.V., L.L.)
| |
Collapse
|