1
|
Du Y, Hu P, Ding X, Wang D, Luo J, Le S, Ren L, Chen M, Ye P, Xia J. Deficiency of ATF3 facilitates both angiotensin II-induced and spontaneously formed aortic aneurysm and dissection development by activating cGAS-STING pathway. Clin Transl Med 2025; 15:e70147. [PMID: 39731276 DOI: 10.1002/ctm2.70147] [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: 07/29/2024] [Revised: 12/03/2024] [Accepted: 12/09/2024] [Indexed: 12/29/2024] Open
Abstract
BACKGROUND Sporadic aortic aneurysm and dissection (AAD) is a critical condition characterised by the progressive loss of vascular smooth muscle cells (VSMCs) and the breakdown of the extracellular matrix. However, the molecular mechanisms responsible for the phenotypic switch and loss of VSMCs in AAD are not fully understood. METHODS AND RESULTS In this study, we employed a discovery-driven, unbiased approach. This approach encourages us to explore the unknown functions of activating transcription factor 3 (ATF3) rather than merely confirming existing hypotheses, while no assumptions were made about ATF3 prior to the experiments. We ensured the unbiased nature of our assessment by conducting morphological evaluations with two independent observers in a blinded manner. We identified elevated expression of ATF3 in both human sporadic AAD tissues and mouse AAD models. VSMC-specific ATF3 conditional knockout (Atf3 cKO) mice showed notable enlargement, dissection and rupture in both thoracic and abdominal aortic regions after exposure to Ang II. Interestingly, older Atf3 cKO mice exhibited spontaneous aortic dissections and senescence of the aortic wall. Mechanistically, ATF3 deficiency led to the degradation of P21 through ubiquitination. Impaired DNA repair in VSMCs resulted in micronuclei formation in the cytoplasm, activating the cyclicGMP-AMP synthase- stimulator of interferon genes (cGAS-STING) pathway and inducing VSMC phenotypic switching and apoptosis. Finally, both pharmacological complementation of P21 function and knockdown of STING expression alleviated ATF3 deficiency-induced AAD. CONCLUSIONS Our study indicates that ATF3 is essential for genomic DNA stability in VSMCs through the P21-cGAS-STING pathway, suggesting that enhancing ATF3 expression in VSMCs could help prevent sporadic AAD. KEY POINTS ATF3 deficiency led to degradation of P21 through ubiquitination, which abolished the G1 phase arrest. VSMCs had no time window to repair the damaged DNA, leading to generation of micronuclei in cytoplasm. Cytoplasmic micronuclei facilitating the activation of cGAS-STING pathway, thus inducing the phenotypic switch and apoptosis of VSMCs.
Collapse
Affiliation(s)
- Yifan Du
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Poyi Hu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiangchao Ding
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Dashuai Wang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Henan Province, China
| | - Jingjing Luo
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sheng Le
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Lingyun Ren
- Department of Anesthesiology, Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Manhua Chen
- Department of Cardiology, Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ping Ye
- Department of Cardiology, Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiahong Xia
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
2
|
Stougiannou TM, Christodoulou KC, Karangelis D. Olfactory Receptors and Aortic Aneurysm: Review of Disease Pathways. J Clin Med 2024; 13:7778. [PMID: 39768700 PMCID: PMC11727755 DOI: 10.3390/jcm13247778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/10/2024] [Accepted: 12/17/2024] [Indexed: 01/16/2025] Open
Abstract
Aortic aneurysm, the pathological dilatation of the aorta at distinct locations, can be attributed to many different genetic and environmental factors. The resulting pathobiological disturbances generate a complex interplay of processes affecting cells and extracellular molecules of the tunica interna, media and externa. In short, aortic aneurysm can affect processes involving the extracellular matrix, lipid trafficking/atherosclerosis, vascular smooth muscle cells, inflammation, platelets and intraluminal thrombus formation, as well as various endothelial functions. Many of these processes are interconnected, potentiating one another. Newer discoveries, including the involvement of odorant olfactory receptors in these processes, have further shed light on disease initiation and pathology. Olfactory receptors are a varied group of G protein coupled-receptors responsible for the recognition of chemosensory information. Although they comprise many different subgroups, some of which are not well-characterized or identified in humans, odorant olfactory receptors, in particular, are most commonly associated with recognition of olfactory information. They can also be ectopically localized and thus carry out additional functions relevant to the tissue in which they are identified. It is thus the purpose of this narrative review to summarize and present pathobiological processes relevant to the initiation and propagation of aortic aneurysm, while also incorporating evidence associating these ectopically functioning odorant olfactory receptors with the overall pathology.
Collapse
Affiliation(s)
- Theodora M. Stougiannou
- Department of Cardiothoracic Surgery, University General Hospital, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (K.C.C.); (D.K.)
| | | | | |
Collapse
|
3
|
Ren P, Jiang B, Hassab A, Li G, Li W, Assi R, Tellides G. Heterogeneous Cardiac-Derived and Neural Crest-Derived Aortic Smooth Muscle Cells Exhibit Similar Transcriptional Changes After TGFβ Signaling Disruption. Arterioscler Thromb Vasc Biol 2024. [PMID: 39697172 DOI: 10.1161/atvbaha.124.321706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 12/05/2024] [Indexed: 12/20/2024]
Abstract
BACKGROUND Smooth muscle cells (SMCs) of cardiac and neural crest origin contribute to the developing proximal aorta and are linked to disease propensity in adults. METHODS We analyzed single-cell transcriptomes of aortic SMCs from adult mice to determine basal states and changes after disrupting TGFβ (transforming growth factor-β) signaling necessary for aortic homeostasis. RESULTS A minority of Myh11 lineage-marked SMCs differentially expressed genes suggestive of embryological origin. Additional analyses in Nkx2-5 and Wnt1 lineage-marked SMCs derived from cardiac and neural crest progenitors, respectively, showed both lineages contributed to a major common cluster and each lineage to a minor distinct cluster. Common cluster SMCs extended from root to arch, cardiac subset cluster SMCs from root to ascending, and neural crest subset cluster SMCs were restricted to the arch. The neural crest subset cluster had greater expression of a subgroup of TGFβ-dependent genes. Nonetheless, conditional deletion of TGFβ receptors resulted in similar transcriptional changes among all SMC clusters. Several disease-associated transcriptional responses were comparable among SMC clusters in a mouse model of Marfan syndrome aortopathy, while many embryological markers of murine aortic SMCs were not detected in adult human aortas. CONCLUSIONS There are multiple subtypes of cardiac-derived and neural crest-derived SMCs with shared or distinctive transcriptional profiles; neural crest subset cluster SMCs with increased expression of certain TGFβ-inducible genes are not spatially linked to the aortic root predisposed to aneurysms from aberrant TGFβ signaling; and loss of TGFβ responses after receptor deletion is uniform among SMC clusters.
Collapse
Affiliation(s)
- Pengwei Ren
- Department of Surgery (Cardiac), Yale School of Medicine, New Haven, CT. (P.R., B.J., A.H., G.L., W.L., R.A., G.T.)
| | - Bo Jiang
- Department of Surgery (Cardiac), Yale School of Medicine, New Haven, CT. (P.R., B.J., A.H., G.L., W.L., R.A., G.T.)
- Now with Department of Vascular Surgery, The First Hospital of China Medical University, Shenyang, Liaoning Province (B.J.)
| | - Abdulrahman Hassab
- Department of Surgery (Cardiac), Yale School of Medicine, New Haven, CT. (P.R., B.J., A.H., G.L., W.L., R.A., G.T.)
| | - Guangxin Li
- Department of Surgery (Cardiac), Yale School of Medicine, New Haven, CT. (P.R., B.J., A.H., G.L., W.L., R.A., G.T.)
- Now with Department of Breast and Thyroid Surgery, Peking University Shenzhen Hospital, Guangdong Province, China (G.L.)
| | - Wei Li
- Department of Surgery (Cardiac), Yale School of Medicine, New Haven, CT. (P.R., B.J., A.H., G.L., W.L., R.A., G.T.)
- Now with Department of Vascular Surgery, Peking University People's Hospital, Beijing, China (W.L.)
| | - Roland Assi
- Department of Surgery (Cardiac), Yale School of Medicine, New Haven, CT. (P.R., B.J., A.H., G.L., W.L., R.A., G.T.)
- Program in Vascular Biology and Therapeutics, Yale School of Medicine, New Haven, CT. (R.A., G.T.)
- Veterans Affairs Connecticut Healthcare System, West Haven, CT (R.A., G.T.)
| | - George Tellides
- Department of Surgery (Cardiac), Yale School of Medicine, New Haven, CT. (P.R., B.J., A.H., G.L., W.L., R.A., G.T.)
- Program in Vascular Biology and Therapeutics, Yale School of Medicine, New Haven, CT. (R.A., G.T.)
- Veterans Affairs Connecticut Healthcare System, West Haven, CT (R.A., G.T.)
| |
Collapse
|
4
|
Cao G, Zhang R, Jia X, Bo J, Li Y, Xuan X, Tian J, Hui H, Xin S, Dong H. CXCR4-targeted sensitive magnetic particle imaging for abdominal aortic aneurysm early detection and prognosis evaluation by recognizing total inflammatory cells. Cardiovasc Res 2024:cvae255. [PMID: 39658102 DOI: 10.1093/cvr/cvae255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 08/22/2024] [Accepted: 12/07/2024] [Indexed: 12/12/2024] Open
Abstract
BACKGROUND The maximum aortic diameter remains the diagnostic criteria and the indicator for prognosis prediction of abdominal aortic aneurysms (AAA). An additional imaging modality is currently needed to help evaluate the prognosis of AAA as well as early detection of AAA formation. This study evaluated the most effective inflammatory markers for AAA using single-cell sequencing and, from these, developed probes to facilitate in vivo multimodal imaging of AAA inflammation. METHODS AND RESULTS Single-cell RNA sequencing (scRNAseq) of the human aortic aneurysms, GSE155468 and GSE166676 datasets, identified CXCR4 as the most representative marker. Anti-CXCR4-PE antibody was conjugated to superparamagnetic iron oxide nanoparticles to synthesise Fe3O4-anti-CXCR4-PE probes. The biocompatibility and specificity of the probes were validated in vivo and in vitro. Magnetic particle imaging (MPI) and fluorescence imaging (FLI) were performed to assess inflammation in early and advanced AAA mouse models. CXCR4-specific receptor inhibitor, AMD3100, was used for confirming CXCR4 as an excellent target for AAA imaging and therapy.scRNAseq indicated that chemokine-related pathways were upregulated in aortic aneurysms, and CXCR4 was the chemokine receptor that markers all AAA-related immune cells and inflammatory vascular cells. Fe3O4-anti-CXCR4-PE effectively recognised immune cells and inflammatory vascular cells, as strong MPI and FLI signals corresponded to increased CXCR4, CD45, and CD68 levels which represented AAA severity and rupture risk. Importantly, Fe3O4-anti-CXCR4-PE can help identify early AAA formation when ultrasound is undiagnosable. Finally, AMD3100 treatment in AAA mouse model inhibited AAA expansion and rupture and reduced aortic wall inflammation by inhibiting accumulation of immune cells and hematopoietic stem cells. CONCLUSION CXCR4 markers immune cells and inflammatory vascular cells in AAA and is associated with AAA prognosis in a mouse model of AAA. CXCR4-targeting multimodal MPI/FLI provides a novel approach for AAA prognosis prediction and early detection.
Collapse
Affiliation(s)
- Genmao Cao
- Department of Vascular Surgery, 2nd Hospital of Shanxi Medical University, No. 382, Wuyi road, Taiyuan, China
| | - Ruijing Zhang
- Department of Nephrology, 2nd Hospital of Shanxi Medical University, No. 382, Wuyi road, Taiyuan, China
| | - Xiaohua Jia
- Key Laboratory of Molecular Imaging of Chinese Academy of Sciences, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
- Department of Ultrasound, Shuozhou Grand Hospital of Shanxi Medical University, Shuozhou 036000, China
| | - Jiang Bo
- Department of Vascular and Thyroid Surgery, The First Hospital of China Medical University, Shenyang, Liaoning Province, People's Republic of China
| | - Yaling Li
- Department of Vascular Surgery, 2nd Hospital of Shanxi Medical University, No. 382, Wuyi road, Taiyuan, China
| | - Xuezhen Xuan
- Department of Vascular Surgery, 2nd Hospital of Shanxi Medical University, No. 382, Wuyi road, Taiyuan, China
| | - Jie Tian
- Key Laboratory of Molecular Imaging of Chinese Academy of Sciences, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
- National Key Laboratory of Kidney Diseases, Beijing, 100853, China
| | - Hui Hui
- Key Laboratory of Molecular Imaging of Chinese Academy of Sciences, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
- National Key Laboratory of Kidney Diseases, Beijing, 100853, China
| | - Shijie Xin
- Department of Vascular and Thyroid Surgery, The First Hospital of China Medical University, Shenyang, Liaoning Province, People's Republic of China
| | - Honglin Dong
- Department of Vascular Surgery, 2nd Hospital of Shanxi Medical University, No. 382, Wuyi road, Taiyuan, China
| |
Collapse
|
5
|
Yu Y, Ye J, Wang R, Wang J, Wang J, Xu Q, Wang P, Wang B, Zhang Y. Research trends and hotspots of the applications of single-cell RNA sequencing in cardiovascular diseases: a bibliometric and visualized study. Ann Med Surg (Lond) 2024; 86:7164-7177. [PMID: 39649887 PMCID: PMC11623828 DOI: 10.1097/ms9.0000000000002681] [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: 06/27/2024] [Accepted: 09/17/2024] [Indexed: 12/11/2024] Open
Abstract
Background Cardiovascular diseases (CVDs) are the leading causes of death globally. The use of single-cell RNA sequencing (scRNA-seq) in CVDs has gained significant attention in recent years, and there is a growing body of literature on the subject. However, a thorough and impartial analysis of the existing state and trends of scRNA-seq in CVDs is lacking. This study aims to examine the development of scRNA-seq in CVDs using bibliometric and visualized analysis. Methods Global publications on scRNA-seq and CVDs from 2009 to 2023 were extracted from the Web of Science Core Collection (WoSCC) database. The R package "Bibliometrix", VOSviewer, and CiteSpace were employed to perform a bibliometric study. Results After applying the screening criteria and omitting documents that met exclusive criteria, this bibliometric study included 1170 papers. These were authored by 8595 scholars from 1565 organizations in 57 countries or regions and were published in 369 journals, with 51 073 co-cited references included. Publication volume, citations, and relative research interest index focusing on this field have dramatically increased since 2019. The cooperation network showed that the USA, the Chinese Academy of Medical Sciences, and Qingbo Xu were the most active countries, institutes, and authors in this field, respectively. Circulation Research was the journal with the most publications, which was confirmed to be the top core source by Bradford's law. The hotspots and emerging direction in the field manifest in (1) three CVDs (atherosclerosis, myocardial infarction, and heart failure) and (2) three cell types (macrophage, fibroblast, and smooth muscle cell). Conclusions Our study provides a systematic visualization of the research literature on scRNA-seq in CVDs and provides guidance and reference for understanding the current research status and discovering new research directions.
Collapse
Affiliation(s)
- Yue Yu
- Department of Cardiothoracic Surgery, Changzheng Hospital, Naval Medical University, Shanghai, People’s Republic of China
| | - Juan Ye
- Department of Cardiothoracic Surgery, Changzheng Hospital, Naval Medical University, Shanghai, People’s Republic of China
| | - Rubing Wang
- Department of Cardiothoracic Surgery, Changzheng Hospital, Naval Medical University, Shanghai, People’s Republic of China
| | - JingJing Wang
- Department of Cardiothoracic Surgery, Changzheng Hospital, Naval Medical University, Shanghai, People’s Republic of China
| | - Junnan Wang
- Department of Cardiothoracic Surgery, Changzheng Hospital, Naval Medical University, Shanghai, People’s Republic of China
| | - Qiumeng Xu
- Department of Orthopaedics, Changzheng Hospital, Naval Medical University, Shanghai, People’s Republic of China
| | - Pei Wang
- Department of Cardiothoracic Surgery, Changzheng Hospital, Naval Medical University, Shanghai, People’s Republic of China
| | - Bei Wang
- Department of Nursing, Changzheng Hospital, Naval Medical University, Shanghai, People’s Republic of China
| | - Yufeng Zhang
- Department of Cardiothoracic Surgery, Changzheng Hospital, Naval Medical University, Shanghai, People’s Republic of China
| |
Collapse
|
6
|
Bramel EE, Espinoza Camejo WA, Creamer TJ, Restrepo L, Saqib M, Bagirzadeh R, Zeng A, Mitchell JT, Stein-O'Brien GL, Pedroza AJ, Fischbein MP, Dietz HC, Gallo MacFarlane E. Intrinsic GATA4 expression sensitizes the aortic root to dilation in a Loeys-Dietz syndrome mouse model. NATURE CARDIOVASCULAR RESEARCH 2024; 3:1468-1481. [PMID: 39567770 PMCID: PMC11634776 DOI: 10.1038/s44161-024-00562-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 10/09/2024] [Indexed: 11/22/2024]
Abstract
Loeys-Dietz syndrome (LDS) is a connective tissue disorder caused by mutations that decrease transforming growth factor-β signaling. LDS-causing mutations increase the risk of aneurysm throughout the arterial tree, yet the aortic root is a site of heightened susceptibility. Here we investigate the heterogeneity of vascular smooth muscle cells (VSMCs) in the aorta of Tgfbr1M318R/+ LDS mice by single-cell transcriptomics to identify molecular determinants of this vulnerability. Reduced expression of components of the extracellular matrix-receptor apparatus and upregulation of stress and inflammatory pathways were observed in all LDS VSMCs. However, regardless of genotype, a subset of Gata4-expressing VSMCs predominantly located in the aortic root intrinsically displayed a less differentiated, proinflammatory profile. A similar population was also identified among aortic VSMCs in a human single-cell RNA sequencing dataset. Postnatal VSMC-specific Gata4 deletion reduced aortic root dilation in LDS mice, suggesting that this factor sensitizes the aortic root to the effects of impaired transforming growth factor-β signaling.
Collapse
MESH Headings
- Animals
- Loeys-Dietz Syndrome/genetics
- Loeys-Dietz Syndrome/pathology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Disease Models, Animal
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- GATA4 Transcription Factor/genetics
- GATA4 Transcription Factor/metabolism
- Receptor, Transforming Growth Factor-beta Type I/genetics
- Receptor, Transforming Growth Factor-beta Type I/metabolism
- Humans
- Signal Transduction
- Dilatation, Pathologic/genetics
- Male
- Aorta/metabolism
- Aorta/pathology
- Single-Cell Analysis
- Mice
- Aortic Aneurysm/genetics
- Aortic Aneurysm/metabolism
- Aortic Aneurysm/pathology
- Mice, Inbred C57BL
- Transcriptome
- Mice, Knockout
Collapse
Affiliation(s)
- Emily E Bramel
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Predoctoral Training in Human Genetics and Genomics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Wendy A Espinoza Camejo
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Predoctoral Training in Human Genetics and Genomics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Tyler J Creamer
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Leda Restrepo
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Muzna Saqib
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Rustam Bagirzadeh
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Anthony Zeng
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jacob T Mitchell
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Predoctoral Training in Human Genetics and Genomics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Genevieve L Stein-O'Brien
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Albert J Pedroza
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael P Fischbein
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Harry C Dietz
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Elena Gallo MacFarlane
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
7
|
Lin CJ, Keating C, Roth R, Caliskan Y, Nazzal M, Exil V, DiPaolo R, Verma DR, Harjai K, Zayed M, Lin CY, Mecham RP, Jain AK. Distinct Patterns of Smooth Muscle Phenotypic Modulation in Thoracic and Abdominal Aortic Aneurysms. J Cardiovasc Dev Dis 2024; 11:349. [PMID: 39590192 PMCID: PMC11594343 DOI: 10.3390/jcdd11110349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/25/2024] [Accepted: 10/29/2024] [Indexed: 11/28/2024] Open
Abstract
Thoracic and abdominal aortic aneurysms (TAAs and AAAs, respectively) share morphological features but have distinct clinical and hereditary characteristics. Studies using bulk RNA comparisons revealed distinct patterns of gene expression in human TAA and AAA tissues. However, given the summative nature of bulk RNA studies, these findings represent the totality of gene expression without regards to the differences in cellular composition. Single-cell RNA sequencing provides an opportunity to interrogate cell-type-specific transcriptomes. Single cell RNA sequencing datasets from mouse TAA (GSE153534) and AAA (GSE164678 and GSE152583) with respective controls were obtained from the Gene Expression Omnibus. Bioinformatic analysis was performed with the Seurat 4, clusterProfiler, and Connectome software packages (V1.0.1). Immunostaining was performed with standard protocols. Within normal and aneurysmal aortae, three unique populations of cells that express smooth muscle cell (SMC) markers were identified (SMC1, SMC2, and SMCmod). A greater proportion of TAA SMCs clustered as a unique population, SMCmod, relative to the AAA SMCs (38% vs. 10-12%). These cells exhibited transcriptional features distinct from other SMCs, which were characterized by Igfbp2 and Tnfrsf11b expression. Genes upregulated in TAA SMCs were enriched for the Reactome terms "extracellular matrix organization" and "insulin-like growth factor (IGF) transport and uptake by IGF binding proteins (IGFBPs)", indicating a role for Igfbp2 in TAA pathogenesis. Regulon analysis revealed transcription factors enriched in TAAs and AAAs. Validating these mouse bioinformatic findings, immunostaining demonstrated that both IGFBP2 and TNFRSF11B proteins increased in human TAAs compared to AAAs. These results highlight the unique cellular composition and transcriptional signature of SMCs in TAAs and AAAs. Future studies are needed to reveal the pathogenetic pathways of IGFBP2 and TNFRSF11B.
Collapse
Affiliation(s)
- Chien-Jung Lin
- Division of Cardiology, Department of Internal Medicine, SSM-Saint Louis University Hospital, St. Louis, MO 63110, USA
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Campbell Keating
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Robyn Roth
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Yasar Caliskan
- Division of Nephrology and Hypertension, Department of Internal Medicine, SSM-Saint Louis University Hospital, St. Louis, MO 63110, USA
| | - Mustafa Nazzal
- Department of Surgery, SSM-Saint Louis University Hospital, St. Louis, MO 63110, USA
| | - Vernat Exil
- Division of Cardiology, Department of Pediatrics, SSM-Cardinal Glennon Children’s Hospital, St. Louis, MO 63104, USA
| | - Richard DiPaolo
- Department of Molecular Microbiology and Immunology, Saint Louis University, St. Louis, MO 63104, USA
| | - Divya Ratan Verma
- Division of Cardiology, Department of Internal Medicine, SSM-Saint Louis University Hospital, St. Louis, MO 63110, USA
| | - Kishore Harjai
- Division of Cardiology, Department of Internal Medicine, SSM-Saint Louis University Hospital, St. Louis, MO 63110, USA
| | - Mohamed Zayed
- Department of Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Molecular Cell Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Biomedical Engineering, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Chieh-Yu Lin
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Robert P. Mecham
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ajay K. Jain
- Division of Gastroenterology and Hepatology, Department of Pediatrics, SSM-Cardinal Glennon Children’s Hospital, St. Louis, MO 63104, USA
| |
Collapse
|
8
|
Abstract
Aortic aneurysm is a life-threatening condition and mechanisms underlying its formation and progression are still incompletely understood. Omics approach has brought new insights to identify a broad spectrum of biomarkers and better understand cellular and molecular pathways involved. Omics generate a large amount of data and several studies have highlighted that artificial intelligence (AI) and techniques such as machine learning (ML)/deep learning (DL) can be of use in analyzing such complex datasets. However, only a few studies have so far reported the use of ML/DL for omics analysis in aortic aneurysms. The aim of this study is to summarize recent advances on the use of ML/DL for omics analysis to decipher aortic aneurysm pathophysiology and develop patient-tailored risk prediction models. In the light of current knowledge, we discuss current limits and highlight future directions in the field.
Collapse
Affiliation(s)
- Fabien Lareyre
- Department of Vascular Surgery, Hospital of Antibes Juan-les-Pins, Nice, France
- Inserm U1065, C3M, Université Côte d'Azur, Nice, France
| | - Arindam Chaudhuri
- Bedfordshire-Milton Keynes Vascular Centre, Bedfordshire Hospitals NHS Foundation Trust, Bedford, UK
| | - Bahaa Nasr
- Department of Vascular and Endovascular Surgery, Brest University Hospital, Brest, France
- INSERM UMR 1101, LaTIM, Brest, France
| | - Juliette Raffort
- Inserm U1065, C3M, Université Côte d'Azur, Nice, France
- Clinical Chemistry Laboratory, University Hospital of Nice, Nice, France
- 3IA Institute, Université Côte d'Azur, Nice, France
| |
Collapse
|
9
|
Johnson RT, Wostear F, Solanki R, Steward O, Bradford A, Morris C, Bidula S, Warren DT. A microtubule stability switch alters isolated vascular smooth muscle Ca2+ flux in response to matrix rigidity. J Cell Sci 2024; 137:jcs262310. [PMID: 39301761 PMCID: PMC11586521 DOI: 10.1242/jcs.262310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 09/15/2024] [Indexed: 09/22/2024] Open
Abstract
During ageing, the extracellular matrix of the aortic wall becomes more rigid. In response, vascular smooth muscle cells (VSMCs) generate enhanced contractile forces. Our previous findings demonstrate that VSMC volume is enhanced in response to increased matrix rigidity, but our understanding of the mechanisms regulating this process remain incomplete. In this study, we show that microtubule stability in VSMCs is reduced in response to enhanced matrix rigidity via Piezo1-mediated Ca2+ influx. Moreover, VSMC volume and Ca2+ flux is regulated by microtubule dynamics; microtubule-stabilising agents reduced both VSMC volume and Ca2+ flux on rigid hydrogels, whereas microtubule-destabilising agents increased VSMC volume and Ca2+ flux on pliable hydrogels. Finally, we show that disruption of the microtubule deacetylase HDAC6 uncoupled these processes and increased α-tubulin acetylation on K40, VSMC volume and Ca2+ flux on pliable hydrogels, but did not alter VSMC microtubule stability. These findings uncover a microtubule stability switch that controls VSMC volume by regulating Ca2+ flux. Taken together, these data demonstrate that manipulation of microtubule stability can modify VSMC response to matrix stiffness.
Collapse
Affiliation(s)
- Robert T. Johnson
- School of Chemistry, Pharmacy and Pharmacology, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK
- Biomedical Research Centre, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, Norfolk, UK
| | - Finn Wostear
- School of Chemistry, Pharmacy and Pharmacology, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK
- Biomedical Research Centre, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, Norfolk, UK
| | - Reesha Solanki
- School of Chemistry, Pharmacy and Pharmacology, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK
- Biomedical Research Centre, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, Norfolk, UK
| | - Oliver Steward
- School of Chemistry, Pharmacy and Pharmacology, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK
- Biomedical Research Centre, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, Norfolk, UK
| | - Alice Bradford
- School of Chemistry, Pharmacy and Pharmacology, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK
- Biomedical Research Centre, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, Norfolk, UK
| | | | - Stefan Bidula
- School of Chemistry, Pharmacy and Pharmacology, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK
| | - Derek T. Warren
- School of Chemistry, Pharmacy and Pharmacology, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK
- Biomedical Research Centre, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, Norfolk, UK
| |
Collapse
|
10
|
Zhao Q, Pedroza A, Sharma D, Gu W, Dalal A, Weldy C, Jackson W, Li DY, Ryan Y, Nguyen T, Shad R, Palmisano BT, Monteiro JP, Worssam M, Berezwitz A, Iyer M, Shi H, Kundu R, Limbu L, Kim JB, Kundaje A, Fischbein M, Wirka R, Quertermous T, Cheng P. A cell and transcriptome atlas of the human arterial vasculature. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.10.612293. [PMID: 39314359 PMCID: PMC11419041 DOI: 10.1101/2024.09.10.612293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Vascular beds show different propensities for different vascular pathologies, yet mechanisms explaining these fundamental differences remain unknown. We sought to build a transcriptomic, cellular, and spatial atlas of human arterial cells across multiple different arterial segments to understand this phenomenon. We found significant cell type-specific segmental heterogeneity. Determinants of arterial identity are predominantly encoded in fibroblasts and smooth muscle cells, and their differentially expressed genes are particularly enriched for vascular disease-associated loci and genes. Adventitial fibroblast-specific heterogeneity in gene expression coincides with numerous vascular disease risk genes, suggesting a previously unrecognized role for this cell type in disease risk. Adult arterial cells from different segments cluster not by anatomical proximity but by embryonic origin, with differentially regulated genes heavily influenced by developmental master regulators. Non-coding transcriptomes across arterial cells contain extensive variation in lnc-RNAs expressed in cell type- and segment-specific patterns, rivaling heterogeneity in protein coding transcriptomes, and show enrichment for non-coding genetic signals for vascular diseases.
Collapse
|
11
|
Jia K, Luo X, Yi J, Zhang C. Hormonal influence: unraveling the impact of sex hormones on vascular smooth muscle cells. Biol Res 2024; 57:61. [PMID: 39227995 PMCID: PMC11373308 DOI: 10.1186/s40659-024-00542-w] [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: 04/30/2024] [Accepted: 08/26/2024] [Indexed: 09/05/2024] Open
Abstract
Sex hormones play a pivotal role as endocrine hormones that exert profound effects on the biological characteristics and vascular function of vascular smooth muscle cells (VSMCs). By modulating intracellular signaling pathways, activating nuclear receptors, and regulating gene expression, sex hormones intricately influence the morphology, function, and physiological state of VSMCs, thereby impacting the biological properties of vascular contraction, relaxation, and growth. Increasing evidence suggests that abnormal phenotypic changes in VSMCs contribute to the initiation of vascular diseases, including atherosclerosis. Therefore, understanding the factors governing phenotypic alterations in VSMCs and elucidating the underlying mechanisms can provide crucial insights for refining interventions targeted at vascular diseases. Additionally, the varying levels of different types of sex hormones in the human body, influenced by sex and age, may also affect the phenotypic conversion of VSMCs. This review aims to explore the influence of sex hormones on the phenotypic switching of VSMCs and the development of associated vascular diseases in the human body.
Collapse
Affiliation(s)
- Keran Jia
- Department of Medical Cell Biology and Genetics, School of Basic Medical Sciences, Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Xin Luo
- Department of Medical Cell Biology and Genetics, School of Basic Medical Sciences, Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, Sichuan, 646000, China
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Jingyan Yi
- Department of Medical Cell Biology and Genetics, School of Basic Medical Sciences, Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, Sichuan, 646000, China.
| | - Chunxiang Zhang
- Department of Cardiology, The Affiliated Hospital, Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, Sichuan, 646000, China.
| |
Collapse
|
12
|
Mao A, Li Z, Shi X, Zhang K, Kan H, Geng L, He D. Complement Factor C1q Mediates Vascular Endothelial Dysfunction in STZ-Induced Diabetic Mice. Diabetes 2024; 73:1527-1536. [PMID: 38869460 DOI: 10.2337/db23-0981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 06/05/2024] [Indexed: 06/14/2024]
Abstract
Diabetes is a significant global public health issue with implications for vascular endothelial cells (ECs) dysfunction and the subsequent development and advancement of diabetes complications. This study aims to compare the cellular and molecular properties of the aorta in normal and streptozotocin (STZ)-induced diabetic mice, with a focus on elucidating potential mechanism underlying EC dysfunction. Here, we performed a single-cell RNA sequencing survey of 32,573 cells from the aorta of normal and STZ-induced diabetic mice. We found a compendium of 10 distinct cell types, mainly ECs, smooth muscle cells, fibroblast, pericyte, immune cells, and stromal cells. As the diabetes condition progressed, we observed a subpopulation of aortic ECs that exhibited significantly elevated expression of complement (C) molecule C1qa compared with their healthy counterparts. This increased expression of C1qa was found to induce reactive oxygen species (ROS) production, facilitate EC migration and increased permeability, and impair the vasodilation within the aortic segment of mice. Furthermore, AAV-Tie2-shRNA-C1qa was administered into diabetic mice by tail vein injection, showing that inhibition of C1qa in the endothelium led to a reduction in ROS production, decreased vascular permeability, and improved vasodilation. Collectively, these findings highlight the crucial involvement of C1qa in endothelial dysfunction associated with diabetes. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Aiqin Mao
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Zicheng Li
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Xiaoming Shi
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Ka Zhang
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Hao Kan
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Li Geng
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Dongxu He
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| |
Collapse
|
13
|
Abedini A, Levinsohn J, Klötzer KA, Dumoulin B, Ma Z, Frederick J, Dhillon P, Balzer MS, Shrestha R, Liu H, Vitale S, Bergeson AM, Devalaraja-Narashimha K, Grandi P, Bhattacharyya T, Hu E, Pullen SS, Boustany-Kari CM, Guarnieri P, Karihaloo A, Traum D, Yan H, Coleman K, Palmer M, Sarov-Blat L, Morton L, Hunter CA, Kaestner KH, Li M, Susztak K. Single-cell multi-omic and spatial profiling of human kidneys implicates the fibrotic microenvironment in kidney disease progression. Nat Genet 2024; 56:1712-1724. [PMID: 39048792 PMCID: PMC11592391 DOI: 10.1038/s41588-024-01802-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 05/15/2024] [Indexed: 07/27/2024]
Abstract
Kidneys are intricate three-dimensional structures in the body, yet the spatial and molecular principles of kidney health and disease remain inadequately understood. We generated high-quality datasets for 81 samples, including single-cell, single-nuclear, spot-level (Visium) and single-cell resolution (CosMx) spatial-RNA expression and single-nuclear open chromatin, capturing cells from healthy, diabetic and hypertensive diseased human kidneys. Combining these data, we identify cell types and map them to their locations within the tissue. Unbiased deconvolution of the spatial data identifies the following four distinct microenvironments: glomerular, immune, tubule and fibrotic. We describe the complex organization of microenvironments in health and disease and find that the fibrotic microenvironment is able to molecularly classify human kidneys and offers an improved prognosis compared to traditional histopathology. We provide a comprehensive spatially resolved molecular roadmap of the human kidney and the fibrotic process, demonstrating the clinical utility of spatial transcriptomics.
Collapse
Affiliation(s)
- Amin Abedini
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Penn/CHOP Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Jonathan Levinsohn
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Penn/CHOP Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Konstantin A Klötzer
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Penn/CHOP Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Bernhard Dumoulin
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Penn/CHOP Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Ziyuan Ma
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Penn/CHOP Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Julia Frederick
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Penn/CHOP Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Poonam Dhillon
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Penn/CHOP Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Michael S Balzer
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Penn/CHOP Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Nephrology, Charité - Universitätsmedizin, Berlin, Germany
| | - Rojesh Shrestha
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Penn/CHOP Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Hongbo Liu
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Penn/CHOP Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Steven Vitale
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Penn/CHOP Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Andi M Bergeson
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Penn/CHOP Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | | | - Paola Grandi
- Genomic Sciences, GSK-Cellzome, Heidelberg, Germany
| | | | - Erding Hu
- Research and Development, GSK, Crescent Drive, Philadelphia, PA, USA
| | - Steven S Pullen
- Department of Cardiometabolic Diseases Research, Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT, USA
| | - Carine M Boustany-Kari
- Department of Cardiometabolic Diseases Research, Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT, USA
| | - Paolo Guarnieri
- Department of Cardiometabolic Diseases Research, Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT, USA
| | | | - Daniel Traum
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Hanying Yan
- Department of Epidemiology, Biostatistics and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Kyle Coleman
- Department of Epidemiology, Biostatistics and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Matthew Palmer
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Lea Sarov-Blat
- Research and Development, GSK, Crescent Drive, Philadelphia, PA, USA
| | - Lori Morton
- Cardiovascular and Renal Research, Regeneron Pharmaceuticals Inc., Tarrytown, NY, USA
| | - Christopher A Hunter
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, USA
| | - Klaus H Kaestner
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Mingyao Li
- Department of Epidemiology, Biostatistics and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Katalin Susztak
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA.
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA.
- Penn/CHOP Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA.
- Department of Genetics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
14
|
Udugampolage NS, Frolova S, Taurino J, Pini A, Martelli F, Voellenkle C. Coding and Non-Coding Transcriptomic Landscape of Aortic Complications in Marfan Syndrome. Int J Mol Sci 2024; 25:7367. [PMID: 39000474 PMCID: PMC11242319 DOI: 10.3390/ijms25137367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 06/21/2024] [Accepted: 06/27/2024] [Indexed: 07/16/2024] Open
Abstract
Marfan syndrome (MFS) is a rare congenital disorder of the connective tissue, leading to thoracic aortic aneurysms (TAA) and dissection, among other complications. Currently, the most efficient strategy to prevent life-threatening dissection is preventive surgery. Periodic imaging applying complex techniques is required to monitor TAA progression and to guide the timing of surgical intervention. Thus, there is an acute demand for non-invasive biomarkers for diagnosis and prognosis, as well as for innovative therapeutic targets of MFS. Unraveling the intricate pathomolecular mechanisms underlying the syndrome is vital to address these needs. High-throughput platforms are particularly well-suited for this purpose, as they enable the integration of different datasets, such as transcriptomic and epigenetic profiles. In this narrative review, we summarize relevant studies investigating changes in both the coding and non-coding transcriptome and epigenome in MFS-induced TAA. The collective findings highlight the implicated pathways, such as TGF-β signaling, extracellular matrix structure, inflammation, and mitochondrial dysfunction. Potential candidates as biomarkers, such as miR-200c, as well as therapeutic targets emerged, like Tfam, associated with mitochondrial respiration, or miR-632, stimulating endothelial-to-mesenchymal transition. While these discoveries are promising, rigorous and extensive validation in large patient cohorts is indispensable to confirm their clinical relevance and therapeutic potential.
Collapse
Affiliation(s)
| | - Svetlana Frolova
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, 20097 Milan, Italy; (S.F.); (C.V.)
- Department of Biosciences, University of Milan, 20122 Milan, Italy
| | - Jacopo Taurino
- Cardiovascular-Genetic Center, IRCCS Policlinico San Donato, 20097 Milan, Italy; (N.S.U.); (J.T.); (A.P.)
| | - Alessandro Pini
- Cardiovascular-Genetic Center, IRCCS Policlinico San Donato, 20097 Milan, Italy; (N.S.U.); (J.T.); (A.P.)
| | - Fabio Martelli
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, 20097 Milan, Italy; (S.F.); (C.V.)
| | - Christine Voellenkle
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, 20097 Milan, Italy; (S.F.); (C.V.)
| |
Collapse
|
15
|
Martinez AN, Tortelote GG, Pascale CL, Ekanem UOI, Leite APDO, McCormack IG, Dumont AS. Dimethyl Fumarate Mediates Sustained Vascular Smooth Muscle Cell Remodeling in a Mouse Model of Cerebral Aneurysm. Antioxidants (Basel) 2024; 13:773. [PMID: 39061841 PMCID: PMC11274241 DOI: 10.3390/antiox13070773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 06/20/2024] [Accepted: 06/23/2024] [Indexed: 07/28/2024] Open
Abstract
Cerebral aneurysms (CA) are a type of vascular disease that causes significant morbidity and mortality with rupture. Dysfunction of the vascular smooth muscle cells (VSMCs) from circle of Willis (CoW) vessels mediates CA formation, as they are the major cell type of the arterial wall and play a role in maintaining vessel integrity. Dimethyl fumarate (DMF), a first-line oral treatment for relapsing-remitting multiple sclerosis, has been shown to inhibit VSMC proliferation and reduce CA formation in a mouse model. Potential unwanted side effects of DMF on VSMC function have not been investigated yet. The present study characterizes the impact of DMF on VSMC using single-cell RNA-sequencing (scRNA-seq) in CoW vessels following CA induction and further explores its role in mitochondrial function using in vitro VSMC cultures. Two weeks of DMF treatment following CA induction impaired the transcription of the glutathione redox system and downregulated mitochondrial respiration genes in VSMCs. In vitro, DMF treatment increased lactate formation and enhanced the mitochondrial production of reactive oxygen species (ROS). These effects rendered VSMCs vulnerable to oxidative stress and led to mitochondrial dysfunction and enhancement of apoptosis. Taken together, our data support the concept that the DMF-mediated antiproliferative effect on VSMCs is linked to disturbed antioxidative functions resulting in altered mitochondrial metabolism. This negative impact of DMF treatment on VSMCs may be linked to preexisting alterations of cerebrovascular function due to renal hypertension. Therefore, before severe adverse effects emerge, it would be clinically relevant to develop indices or biomarkers linked to this disturbed antioxidative function to monitor patients undergoing DMF treatment.
Collapse
Affiliation(s)
- Alejandra N. Martinez
- Department of Neurosurgery, The Tulane Center for Clinical Neurosciences, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA 70012, USA (A.S.D.)
| | - Giovane G. Tortelote
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, LA 70112, USA;
| | - Crissey L. Pascale
- Department of Neurosurgery, The Tulane Center for Clinical Neurosciences, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA 70012, USA (A.S.D.)
| | - Uduak-Obong I. Ekanem
- Department of Neurosurgery, The Tulane Center for Clinical Neurosciences, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA 70012, USA (A.S.D.)
| | - Ana Paula de O. Leite
- Department of Pharmacology, The Tulane Center for Sex-Based Biology and Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Isabella G. McCormack
- Department of Neurosurgery, The Tulane Center for Clinical Neurosciences, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA 70012, USA (A.S.D.)
| | - Aaron S. Dumont
- Department of Neurosurgery, The Tulane Center for Clinical Neurosciences, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA 70012, USA (A.S.D.)
| |
Collapse
|
16
|
Bramel EE, Camejo WAE, Creamer TJ, Restrepo L, Saqib M, Bagirzadeh R, Zeng A, Mitchell JT, Stein-O’Brien GL, Pedroza AJ, Fischbein MP, Dietz HC, MacFarlane EG. Intrinsic Gata4 expression sensitizes the aortic root to dilation in a Loeys-Dietz syndrome mouse model. RESEARCH SQUARE 2024:rs.3.rs-4420617. [PMID: 38883722 PMCID: PMC11177966 DOI: 10.21203/rs.3.rs-4420617/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
Loeys-Dietz syndrome (LDS) is an aneurysm disorder caused by mutations that decrease transforming growth factor-β (TGF-β) signaling. Although aneurysms develop throughout the arterial tree, the aortic root is a site of heightened risk. To identify molecular determinants of this vulnerability, we investigated the heterogeneity of vascular smooth muscle cells (VSMCs) in the aorta of Tgfbr1 M318R/+ LDS mice by single cell and spatial transcriptomics. Reduced expression of components of the extracellular matrix-receptor apparatus and upregulation of stress and inflammatory pathways were observed in all LDS VSMCs. However, regardless of genotype, a subset of Gata4-expressing VSMCs predominantly located in the aortic root intrinsically displayed a less differentiated, proinflammatory profile. A similar population was also identified among aortic VSMCs in a human scRNAseq dataset. Postnatal VSMC-specific Gata4 deletion reduced aortic root dilation in LDS mice, suggesting that this factor sensitizes the aortic root to the effects of impaired TGF-β signaling.
Collapse
Affiliation(s)
- Emily E. Bramel
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Predoctoral Training in Human Genetics and Genomics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Wendy A. Espinoza Camejo
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Predoctoral Training in Human Genetics and Genomics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Tyler J. Creamer
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Leda Restrepo
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Muzna Saqib
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Rustam Bagirzadeh
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Anthony Zeng
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jacob T. Mitchell
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Predoctoral Training in Human Genetics and Genomics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Genevieve L. Stein-O’Brien
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Albert J. Pedroza
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Michael P. Fischbein
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Harry C. Dietz
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Elena Gallo MacFarlane
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
17
|
Liu X, Zeng Q, Yang H, Li W, Chen Q, Yin K, Pan Z, Wang K, Luo M, Shu C, Zhou Z. Single-Nucleus Multiomic Analyses Identifies Gene Regulatory Dynamics of Phenotypic Modulation in Human Aneurysmal Aortic Root. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400444. [PMID: 38552156 PMCID: PMC11165511 DOI: 10.1002/advs.202400444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Indexed: 06/12/2024]
Abstract
Aortic root aneurysm is a potentially life-threatening condition that may lead to aortic rupture and is often associated with genetic syndromes, such as Marfan syndrome (MFS). Although studies with MFS animal models have provided valuable insights into the pathogenesis of aortic root aneurysms, this understanding of the transcriptomic and epigenomic landscape in human aortic root tissue remains incomplete. This knowledge gap has impeded the development of effective targeted therapies. Here, this study performs the first integrative analysis of single-nucleus multiomic (gene expression and chromatin accessibility) and spatial transcriptomic sequencing data of human aortic root tissue under healthy and MFS conditions. Cell-type-specific transcriptomic and cis-regulatory profiles in the human aortic root are identified. Regulatory and spatial dynamics during phenotypic modulation of vascular smooth muscle cells (VSMCs), the cardinal cell type, are delineated. Moreover, candidate key regulators driving the phenotypic modulation of VSMC, such as FOXN3, TEAD1, BACH2, and BACH1, are identified. In vitro experiments demonstrate that FOXN3 functions as a novel key regulator for maintaining the contractile phenotype of human aortic VSMCs through targeting ACTA2. These findings provide novel insights into the regulatory and spatial dynamics during phenotypic modulation in the aneurysmal aortic root of humans.
Collapse
Affiliation(s)
- Xuanyu Liu
- State Key Laboratory of Cardiovascular DiseaseNational Center for Cardiovascular DiseasesBeijing Key Laboratory for Molecular Diagnostics of Cardiovascular DiseasesCenter of Laboratory MedicineFuwai HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100037China
| | - Qingyi Zeng
- State Key Laboratory of Cardiovascular DiseaseNational Center for Cardiovascular DiseasesBeijing Key Laboratory for Molecular Diagnostics of Cardiovascular DiseasesCenter of Laboratory MedicineFuwai HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100037China
| | - Hang Yang
- State Key Laboratory of Cardiovascular DiseaseNational Center for Cardiovascular DiseasesBeijing Key Laboratory for Molecular Diagnostics of Cardiovascular DiseasesCenter of Laboratory MedicineFuwai HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100037China
| | - Wenke Li
- State Key Laboratory of Cardiovascular DiseaseNational Center for Cardiovascular DiseasesBeijing Key Laboratory for Molecular Diagnostics of Cardiovascular DiseasesCenter of Laboratory MedicineFuwai HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100037China
| | - Qianlong Chen
- State Key Laboratory of Cardiovascular DiseaseNational Center for Cardiovascular DiseasesBeijing Key Laboratory for Molecular Diagnostics of Cardiovascular DiseasesCenter of Laboratory MedicineFuwai HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100037China
| | - Kunlun Yin
- State Key Laboratory of Cardiovascular DiseaseNational Center for Cardiovascular DiseasesBeijing Key Laboratory for Molecular Diagnostics of Cardiovascular DiseasesCenter of Laboratory MedicineFuwai HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100037China
| | - Zihang Pan
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesState Key Laboratory of Vascular Homeostasis and RemodelingPeking UniversityBeijing100191China
| | - Kai Wang
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesState Key Laboratory of Vascular Homeostasis and RemodelingPeking UniversityBeijing100191China
| | - Mingyao Luo
- State Key Laboratory of Cardiovascular DiseaseNational Center for Cardiovascular DiseasesBeijing Key Laboratory for Molecular Diagnostics of Cardiovascular DiseasesCenter of Laboratory MedicineFuwai HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100037China
- Center of Vascular SurgeryFuwai HospitalChinese Academy of Medical SciencesBeijing100037China
- Department of Vascular SurgeryFuwai Yunnan Cardiovascular HospitalAffiliated Cardiovascular Hospital of Kunming Medical UniversityKunmingYunnan650102China
- Department of Vascular SurgeryCentral‐China Subcenter of National Center for Cardiovascular DiseasesHenan Cardiovascular Disease CenterFuwai Central‐China Cardiovascular HospitalCentral China Fuwai Hospital of Zhengzhou UniversityZhengzhou450046China
| | - Chang Shu
- State Key Laboratory of Cardiovascular DiseaseNational Center for Cardiovascular DiseasesBeijing Key Laboratory for Molecular Diagnostics of Cardiovascular DiseasesCenter of Laboratory MedicineFuwai HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100037China
- Center of Vascular SurgeryFuwai HospitalChinese Academy of Medical SciencesBeijing100037China
| | - Zhou Zhou
- State Key Laboratory of Cardiovascular DiseaseNational Center for Cardiovascular DiseasesBeijing Key Laboratory for Molecular Diagnostics of Cardiovascular DiseasesCenter of Laboratory MedicineFuwai HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100037China
| |
Collapse
|
18
|
Ye Q, Chen M, Ma L. Genetic liability to elevated circulating IP-10, IFNγ and SCGFβ levels in relation to thoracic aortic aneurysm: A mendelian randomization study. Cytokine 2024; 178:156569. [PMID: 38484620 DOI: 10.1016/j.cyto.2024.156569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 02/26/2024] [Accepted: 03/04/2024] [Indexed: 04/12/2024]
Abstract
Inflammation is associated with thoracic aortic aneurysm (TAA) but the effects of each circulating inflammatory factor on TAA remain unclear. In this study, we explored the relationship between circulating inflammatory factors and TAA risk using Mendelian randomization (MR) approach based on summary statistics from the latest genome-wide association study (GWAS) of 41 circulating inflammatory factors in 8293 Finns and a GWAS involving 1351 TAA cases and 18,295 controls of European ancestry. In univariable MR, higher interferon gamma-induced protein 10 (IP-10) levels, higher interferon gamma (IFNγ) levels and higher stem cell growth factor beta (SCGFβ) levels were associated with an increased risk of TAA (OR = 1.37, 95 % CI = 1.17-1.59, p = 7.42 × 10-5; OR = 1.43, 95 % CI = 1.19-1.74, p = 2.04 × 10-4; OR = 1.27, 95 % CI = 1.09-1.48, p = 2.40 × 10-3, respectively). In multivariable MR, the patterns of associations for the three cytokines remained adjusting for each other or smoking, but were attenuated differently with adjustment for other cardiovascular risk factors, especially for lipids and body mass index. Bidirectional MR approach did not identify any significant associations between cytokines and risk factors. Our results indicated that circulating cytokines may play mediation roles in the pathogenesis of TAA. Further studies are needed to determine whether these biomarkers can be used to prevent and treat TAA.
Collapse
Affiliation(s)
- Qianxi Ye
- Department of Cardiovascular Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, China
| | - Miao Chen
- Department of Cardiovascular Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, China
| | - Liang Ma
- Department of Cardiovascular Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, China.
| |
Collapse
|
19
|
Jiang D, Zheng S, Xu X, Yue H, Liang W, Wu Z. Uncovering Druggable Targets in Aortic Dissection: An Association Study Integrating Mendelian Randomization, pQTL, and Protein-Protein Interaction Network. Biomedicines 2024; 12:1204. [PMID: 38927411 PMCID: PMC11200553 DOI: 10.3390/biomedicines12061204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 05/16/2024] [Accepted: 05/27/2024] [Indexed: 06/28/2024] Open
Abstract
Aortic dissection (AD) is a life-threatening acute aortic syndrome. There are limitations and challenges in the discovery and application of biomarkers and drug targets for AD. Mendelian randomization (MR) analysis is a reliable analytical method to identify effective therapeutic targets. We aimed to identify novel therapeutic targets for AD and investigate their potential side-effects based on MR analysis. Data from protein quantitative trait loci (pQTLs) were used for MR analyses to identify potential therapeutic targets. We probed druggable proteins involved in the pathogenesis of aortic dissection from deCODE. In this study, a two-sample MR analysis was conducted, with druggable proteins as the exposure factor and data on genome-wide association studies (GWAS) of AD as the outcome. After conducting a two-sample MR, summary data-based Mendelian randomization (SMR) analysis and colocalization analysis were performed. A protein-protein interaction (PPI) network was also constructed to delve into the interactions between identified proteins. After MR analysis and the Steiger test, we identified five proteins as potential therapeutic targets for AD. SMR analysis and colocalization analysis also confirmed our findings. Finally, we identified ASPN (OR = 1.36, 95% CI: 1.20, 1.54, p = 4.22 × 10-5) and SPOCK2 (OR = 0.57, 95% CI: 0.41, 0.78, p = 4.52 × 10-4) as the core therapeutic targets. Through PPI network analysis, we identified six druggable targets, enabling the subsequent identification of six promising drugs from DrugBank for treating AD. This discovery of specific proteins as novel therapeutic targets represents a significant advancement in AD treatment. These findings provide more effective treatment options for AD.
Collapse
Affiliation(s)
| | | | | | | | | | - Zhong Wu
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, No. 37 Guoxue Road, Wuhou District, Chengdu 610041, China; (D.J.)
| |
Collapse
|
20
|
Rojas MG, Pereira-Simon S, Zigmond ZM, Varona Santos J, Perla M, Santos Falcon N, Stoyell-Conti FF, Salama A, Yang X, Long X, Duque JC, Salman LH, Tabbara M, Martinez L, Vazquez-Padron RI. Single-Cell Analyses Offer Insights into the Different Remodeling Programs of Arteries and Veins. Cells 2024; 13:793. [PMID: 38786017 PMCID: PMC11119253 DOI: 10.3390/cells13100793] [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: 03/06/2024] [Revised: 04/18/2024] [Accepted: 04/29/2024] [Indexed: 05/25/2024] Open
Abstract
Arteries and veins develop different types of occlusive diseases and respond differently to injury. The biological reasons for this discrepancy are not well understood, which is a limiting factor for the development of vein-targeted therapies. This study contrasts human peripheral arteries and veins at the single-cell level, with a focus on cell populations with remodeling potential. Upper arm arteries (brachial) and veins (basilic/cephalic) from 30 organ donors were compared using a combination of bulk and single-cell RNA sequencing, proteomics, flow cytometry, and histology. The cellular atlases of six arteries and veins demonstrated a 7.8× higher proportion of contractile smooth muscle cells (SMCs) in arteries and a trend toward more modulated SMCs. In contrast, veins showed a higher abundance of endothelial cells, pericytes, and macrophages, as well as an increasing trend in fibroblasts. Activated fibroblasts had similar proportions in both types of vessels but with significant differences in gene expression. Modulated SMCs and activated fibroblasts were characterized by the upregulation of MYH10, FN1, COL8A1, and ITGA10. Activated fibroblasts also expressed F2R, POSTN, and COMP and were confirmed by F2R/CD90 flow cytometry. Activated fibroblasts from veins were the top producers of collagens among all fibroblast populations from both types of vessels. Venous fibroblasts were also highly angiogenic, proinflammatory, and hyper-responders to reactive oxygen species. Differences in wall structure further explain the significant contribution of fibroblast populations to remodeling in veins. Fibroblasts are almost exclusively located outside the external elastic lamina in arteries, while widely distributed throughout the venous wall. In line with the above, ECM-targeted proteomics confirmed a higher abundance of fibrillar collagens in veins vs. more basement ECM components in arteries. The distinct cellular compositions and transcriptional programs of reparative populations in arteries and veins may explain differences in acute and chronic wall remodeling between vessels. This information may be relevant for the development of antistenotic therapies.
Collapse
Affiliation(s)
- Miguel G. Rojas
- Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (M.G.R.); (S.P.-S.); (J.V.S.); (A.S.)
| | - Simone Pereira-Simon
- Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (M.G.R.); (S.P.-S.); (J.V.S.); (A.S.)
| | | | - Javier Varona Santos
- Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (M.G.R.); (S.P.-S.); (J.V.S.); (A.S.)
| | - Mikael Perla
- Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (M.G.R.); (S.P.-S.); (J.V.S.); (A.S.)
| | - Nieves Santos Falcon
- Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (M.G.R.); (S.P.-S.); (J.V.S.); (A.S.)
| | - Filipe F. Stoyell-Conti
- Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (M.G.R.); (S.P.-S.); (J.V.S.); (A.S.)
| | - Alghidak Salama
- Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (M.G.R.); (S.P.-S.); (J.V.S.); (A.S.)
| | - Xiaofeng Yang
- Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Xiaochun Long
- Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Juan C. Duque
- Department of Medicine, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Loay H. Salman
- Division of Nephrology and Hypertension, Albany Medical College, Albany, NY 12208, USA
| | - Marwan Tabbara
- Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (M.G.R.); (S.P.-S.); (J.V.S.); (A.S.)
| | - Laisel Martinez
- Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (M.G.R.); (S.P.-S.); (J.V.S.); (A.S.)
| | - Roberto I. Vazquez-Padron
- Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (M.G.R.); (S.P.-S.); (J.V.S.); (A.S.)
- Bruce W. Carter Veterans Affairs Medical Center, Miami, FL 33125, USA;
| |
Collapse
|
21
|
Gibson Hughes TA, Dona MSI, Sobey CG, Pinto AR, Drummond GR, Vinh A, Jelinic M. Aortic Cellular Heterogeneity in Health and Disease: Novel Insights Into Aortic Diseases From Single-Cell RNA Transcriptomic Data Sets. Hypertension 2024; 81:738-751. [PMID: 38318714 DOI: 10.1161/hypertensionaha.123.20597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2024]
Abstract
Aortic diseases such as atherosclerosis, aortic aneurysms, and aortic stiffening are significant complications that can have significant impact on end-stage cardiovascular disease. With limited pharmacological therapeutic strategies that target the structural changes in the aorta, surgical intervention remains the only option for some patients with these diseases. Although there have been significant contributions to our understanding of the cellular architecture of the diseased aorta, particularly in the context of atherosclerosis, furthering our insight into the cellular drivers of disease is required. The major cell types of the aorta are well defined; however, the advent of single-cell RNA sequencing provides unrivaled insights into the cellular heterogeneity of each aortic cell type and the inferred biological processes associated with each cell in health and disease. This review discusses previous concepts that have now been enhanced with recent advances made by single-cell RNA sequencing with a focus on aortic cellular heterogeneity.
Collapse
Affiliation(s)
- Tayla A Gibson Hughes
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia (T.A.G.H., C.G.S., A.R.P., G.R.D., A.V., M.J.)
| | - Malathi S I Dona
- Baker Heart and Diabetes Research Institute, Melbourne, Victoria, Australia (M.S.I.D., A.R.P.)
| | - Christopher G Sobey
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia (T.A.G.H., C.G.S., A.R.P., G.R.D., A.V., M.J.)
| | - Alexander R Pinto
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia (T.A.G.H., C.G.S., A.R.P., G.R.D., A.V., M.J.)
- Baker Heart and Diabetes Research Institute, Melbourne, Victoria, Australia (M.S.I.D., A.R.P.)
| | - Grant R Drummond
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia (T.A.G.H., C.G.S., A.R.P., G.R.D., A.V., M.J.)
| | - Antony Vinh
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia (T.A.G.H., C.G.S., A.R.P., G.R.D., A.V., M.J.)
| | - Maria Jelinic
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia (T.A.G.H., C.G.S., A.R.P., G.R.D., A.V., M.J.)
| |
Collapse
|
22
|
Fu Y, Zhou Y, Wang K, Li Z, Kong W. Extracellular Matrix Interactome in Modulating Vascular Homeostasis and Remodeling. Circ Res 2024; 134:931-949. [PMID: 38547250 DOI: 10.1161/circresaha.123.324055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/02/2024]
Abstract
The ECM (extracellular matrix) is a major component of the vascular microenvironment that modulates vascular homeostasis. ECM proteins include collagens, elastin, noncollagen glycoproteins, and proteoglycans/glycosaminoglycans. ECM proteins form complex matrix structures, such as the basal lamina and collagen and elastin fibers, through direct interactions or lysyl oxidase-mediated cross-linking. Moreover, ECM proteins directly interact with cell surface receptors or extracellular secreted molecules, exerting matricellular and matricrine modulation, respectively. In addition, extracellular proteases degrade or cleave matrix proteins, thereby contributing to ECM turnover. These interactions constitute the ECM interactome network, which is essential for maintaining vascular homeostasis and preventing pathological vascular remodeling. The current review mainly focuses on endogenous matrix proteins in blood vessels and discusses the interaction of these matrix proteins with other ECM proteins, cell surface receptors, cytokines, complement and coagulation factors, and their potential roles in maintaining vascular homeostasis and preventing pathological remodeling.
Collapse
Affiliation(s)
- Yi Fu
- Department of Physiology and Pathophysiology (Y.F., K.W., Z.L., W.K.), School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Yuan Zhou
- Department of Biomedical Informatics (Y.Z.), School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Kai Wang
- Department of Physiology and Pathophysiology (Y.F., K.W., Z.L., W.K.), School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Zhuofan Li
- Department of Physiology and Pathophysiology (Y.F., K.W., Z.L., W.K.), School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Wei Kong
- Department of Physiology and Pathophysiology (Y.F., K.W., Z.L., W.K.), School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| |
Collapse
|
23
|
Sharma D, Worssam MD, Pedroza AJ, Dalal AR, Alemany H, Kim HJ, Kundu R, Fischbein M, Cheng P, Wirka R, Quertermous T. Comprehensive Integration of Multiple Single-Cell Transcriptomic Data Sets Defines Distinct Cell Populations and Their Phenotypic Changes in Murine Atherosclerosis. Arterioscler Thromb Vasc Biol 2024; 44:391-408. [PMID: 38152886 PMCID: PMC11285358 DOI: 10.1161/atvbaha.123.320030] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 12/12/2023] [Indexed: 12/29/2023]
Abstract
BACKGROUND The application of single-cell transcriptomic (single-cell RNA sequencing) analysis to the study of atherosclerosis has provided unique insights into the molecular and genetic mechanisms that mediate disease risk and pathophysiology. However, nonstandardized methodologies and relatively high costs associated with the technique have limited the size and replication of existing data sets and created disparate or contradictory findings that have fostered misunderstanding and controversy. METHODS To address these uncertainties, we have performed a conservative integration of multiple published single-cell RNA sequencing data sets into a single meta-analysis, performed extended analysis of native resident vascular cells, and used in situ hybridization to map the disease anatomic location of the identified cluster cells. To investigate the transdifferentiation of smooth muscle cells to macrophage phenotype, we have developed a classifying algorithm based on the quantification of reporter transgene expression. RESULTS The reporter gene expression tool indicates that within the experimental limits of the examined studies, transdifferentiation of smooth muscle cell to the macrophage lineage is extremely rare. Validated transition smooth muscle cell phenotypes were defined by clustering, and the location of these cells was mapped to lesion anatomy with in situ hybridization. We have also characterized 5 endothelial cell phenotypes and linked these cellular species to different vascular structures and functions. Finally, we have identified a transcriptomically unique cellular phenotype that constitutes the aortic valve. CONCLUSIONS Taken together, these analyses resolve a number of outstanding issues related to differing results reported with vascular disease single-cell RNA sequencing studies, and significantly extend our understanding of the role of resident vascular cells in anatomy and disease.
Collapse
Affiliation(s)
- Disha Sharma
- Division of Cardiovascular Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94305
| | - Matthew DeForest Worssam
- Division of Cardiovascular Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94305
| | - Albert J. Pedroza
- Division of Cardiothoracic surgery, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94305
| | - Alex R. Dalal
- Division of Cardiothoracic surgery, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94305
| | - Haizea Alemany
- Division of Cardiovascular Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94305
| | - Hyun-Jung Kim
- Division of Cardiovascular Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94305
| | | | - Michael Fischbein
- Division of Cardiothoracic surgery, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94305
| | - Paul Cheng
- Division of Cardiovascular Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94305
| | - Robert Wirka
- Division of Cardiology, McAllister Heart Institute, UNC School of Medicine, 111 Mason Farm Road, MBRB 3312B, Chapel Hill, NC 27599-7126
| | - Thomas Quertermous
- Division of Cardiovascular Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94305
| |
Collapse
|
24
|
Gonzalez BA, Harmeyer SW, Song T, Sadayappan S, Yutzey KE. Dynamic changes in mitral valve extracellular matrix, tissue mechanics and function in a mouse model of Marfan syndrome. Matrix Biol 2024; 126:1-13. [PMID: 38185344 DOI: 10.1016/j.matbio.2024.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/21/2023] [Accepted: 01/04/2024] [Indexed: 01/09/2024]
Abstract
OBJECTIVE Mouse models of Marfan syndrome (MFS) with Fibrillin 1 (Fbn1) variant C1041G exhibit cardiovascular abnormalities, including myxomatous valve disease (MVD) and aortic aneurism, with structural extracellular matrix (ECM) dysregulation. In this study, we examine the structure-function-mechanics relations of the mitral valve related to specific transitions in ECM composition and organization in progressive MVD in MFS mice from Postnatal day (P)7 to 1 year-of-age. APPROACH AND RESULTS Mechanistic links between mechanical forces and biological changes in MVD progression were examined in Fbn1C1041G/+ MFS mice. By echocardiography, mitral valve dysfunction is prevalent at 2 months with a decrease in cardiac function at 6 months, followed by a preserved cardiac function at 12 months. Mitral valve (MV) regurgitation occurs in a subset of mice at 2-6 months, while progressive dilatation of the aorta occurs from 2 to 12 months. Mitral valve tissue mechanical assessments using a uniaxial Permeabilizable Fiber System demonstrate decreased stiffness of MFS MVs at all stages. Histological and microscopic analysis of ECM content, structure, and fiber orientation demonstrate that alterations in ECM mechanics, composition, and organization precede functional abnormalities in Fbn1C1041G/+MFS MVs. At 2 months, ECM abnormalities are detected with an increase in proteoglycans and decreased stiffness of the mitral valve. By 6-12 months, collagen fiber remodeling is increased with abnormal fiber organization in MFS mitral valve leaflets. At the same time, matrifibrocyte gene expression characteristic of collagen-rich connective tissue is increased, as detected by RNA in situ hybridization and qPCR. Together, these studies demonstrate early prevalence of proteoglycans at 2 months followed by upregulation of collagen structure and organization with age in MVs of MFS mice. CONCLUSIONS Altogether, our data indicate dynamic regulation of mitral valve structure, tissue mechanics, and function that reflect changes in ECM composition, organization, and gene expression in progressive MVD. Notably, increased collagen fiber organization and orientation, potentially dependent on increased matrifibrocyte cell activity, is apparent with altered mitral valve mechanics and function in aging MFS mice.
Collapse
Affiliation(s)
- Brittany A Gonzalez
- The Heart Institute, Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Samuel W Harmeyer
- The Heart Institute, Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Taejeong Song
- Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Sakthivel Sadayappan
- Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Katherine E Yutzey
- The Heart Institute, Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati OH, USA.
| |
Collapse
|
25
|
Shi Y, Chen J, Cai L, Zhang X, Chen Z, Yang J, Jiang Y, Lu Y. Uncovering the Hidden World of Aqueous Humor Proteins for Discovery of Biomarkers for Marfan Syndrome. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2303161. [PMID: 38088571 PMCID: PMC10853735 DOI: 10.1002/advs.202303161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 10/23/2023] [Indexed: 12/19/2023]
Abstract
Ectopia lentis is a hallmark of Marfan syndrome (MFS), a genetic connective tissue disorder affecting 1/5000 to 1/10 000 individuals worldwide. Early detection in ophthalmology clinics and timely intervention of cardiovascular complications can be lifesaving. In this study, a modified proteomics workflow with liquid chromatography-tandem mass spectrometry (LC-MS/MS)-based data-independent acquisition (DIA) and field asymmetric ion mobility spectrometry (FAIMS) to profile the proteomes of aqueous humor (AH) and lens tissue from MFS children with ectopia lentis is utilized. Over 2300 and 2938 comparable proteins are identified in AH and the lens capsule, respectively. Functional enrichment analyses uncovered dysregulation of complement and coagulation-related pathways, collagen binding, and cell adhesion in MFS. Through weighted correlation network analysis (WGCNA) and machine learning, distinct modules associated with clinical traits are constructed and a unique biomarker panel (Q14376, Q99972, P02760, Q07507; gene names: GALE, MYOC, AMBP, DPT) is defined. These biomarkers are further validated using advanced parallel reaction monitoring (PRM) in an independent patient cohort. The results provide novel insights into the proteome characterization of ectopia lentis and offer a promising approach for developing a valuable biomarker panel to aid in the early diagnosis of Marfan syndrome via AH proteome.
Collapse
Affiliation(s)
- Yumeng Shi
- Eye Institute and Department of Ophthalmology, Eye and ENT HospitalFudan UniversityShanghai200031China
- NHC Key Laboratory of MyopiaFudan UniversityShanghai200031China
- Key Laboratory of MyopiaChinese Academy of Medical SciencesShanghai200031China
- Shanghai Key Laboratory of Visual Impairment and RestorationShanghai200031China
| | - Jiahui Chen
- Eye Institute and Department of Ophthalmology, Eye and ENT HospitalFudan UniversityShanghai200031China
- NHC Key Laboratory of MyopiaFudan UniversityShanghai200031China
- Key Laboratory of MyopiaChinese Academy of Medical SciencesShanghai200031China
- Shanghai Key Laboratory of Visual Impairment and RestorationShanghai200031China
| | - Lei Cai
- Eye Institute and Department of Ophthalmology, Eye and ENT HospitalFudan UniversityShanghai200031China
- NHC Key Laboratory of MyopiaFudan UniversityShanghai200031China
- Key Laboratory of MyopiaChinese Academy of Medical SciencesShanghai200031China
- Shanghai Key Laboratory of Visual Impairment and RestorationShanghai200031China
| | - Xueling Zhang
- Eye Institute and Department of Ophthalmology, Eye and ENT HospitalFudan UniversityShanghai200031China
- NHC Key Laboratory of MyopiaFudan UniversityShanghai200031China
- Key Laboratory of MyopiaChinese Academy of Medical SciencesShanghai200031China
- Shanghai Key Laboratory of Visual Impairment and RestorationShanghai200031China
| | - Zexu Chen
- Eye Institute and Department of Ophthalmology, Eye and ENT HospitalFudan UniversityShanghai200031China
- NHC Key Laboratory of MyopiaFudan UniversityShanghai200031China
- Key Laboratory of MyopiaChinese Academy of Medical SciencesShanghai200031China
- Shanghai Key Laboratory of Visual Impairment and RestorationShanghai200031China
| | - Jin Yang
- Eye Institute and Department of Ophthalmology, Eye and ENT HospitalFudan UniversityShanghai200031China
- NHC Key Laboratory of MyopiaFudan UniversityShanghai200031China
- Key Laboratory of MyopiaChinese Academy of Medical SciencesShanghai200031China
- Shanghai Key Laboratory of Visual Impairment and RestorationShanghai200031China
| | - Yongxiang Jiang
- Eye Institute and Department of Ophthalmology, Eye and ENT HospitalFudan UniversityShanghai200031China
- NHC Key Laboratory of MyopiaFudan UniversityShanghai200031China
- Key Laboratory of MyopiaChinese Academy of Medical SciencesShanghai200031China
- Shanghai Key Laboratory of Visual Impairment and RestorationShanghai200031China
| | - Yi Lu
- Eye Institute and Department of Ophthalmology, Eye and ENT HospitalFudan UniversityShanghai200031China
- NHC Key Laboratory of MyopiaFudan UniversityShanghai200031China
- Key Laboratory of MyopiaChinese Academy of Medical SciencesShanghai200031China
- Shanghai Key Laboratory of Visual Impairment and RestorationShanghai200031China
| |
Collapse
|
26
|
Raghavan A, Pirruccello JP, Ellinor PT, Lindsay ME. Using Genomics to Identify Novel Therapeutic Targets for Aortic Disease. Arterioscler Thromb Vasc Biol 2024; 44:334-351. [PMID: 38095107 PMCID: PMC10843699 DOI: 10.1161/atvbaha.123.318771] [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: 06/26/2023] [Accepted: 11/21/2023] [Indexed: 01/04/2024]
Abstract
Aortic disease, including dissection, aneurysm, and rupture, carries significant morbidity and mortality and is a notable cause of sudden cardiac death. Much of our knowledge regarding the genetic basis of aortic disease has relied on the study of individuals with Mendelian aortopathies and, until recently, the genetic determinants of population-level variance in aortic phenotypes remained unclear. However, the application of machine learning methodologies to large imaging datasets has enabled researchers to rapidly define aortic traits and mine dozens of novel genetic associations for phenotypes such as aortic diameter and distensibility. In this review, we highlight the emerging potential of genomics for identifying causal genes and candidate drug targets for aortic disease. We describe how deep learning technologies have accelerated the pace of genetic discovery in this field. We then provide a blueprint for translating genetic associations to biological insights, reviewing techniques for locus and cell type prioritization, high-throughput functional screening, and disease modeling using cellular and animal models of aortic disease.
Collapse
Affiliation(s)
- Avanthi Raghavan
- Cardiology Division, Massachusetts General Hospital, Boston, Massachusetts, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, Massachusetts, USA
- Cardiovascular Disease Initiative, Broad Institute, Cambridge, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - James P. Pirruccello
- Division of Cardiology, University of California San Francisco, San Francisco, CA, USA
| | - Patrick T. Ellinor
- Cardiology Division, Massachusetts General Hospital, Boston, Massachusetts, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, Massachusetts, USA
- Cardiovascular Disease Initiative, Broad Institute, Cambridge, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Mark E. Lindsay
- Cardiology Division, Massachusetts General Hospital, Boston, Massachusetts, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, Massachusetts, USA
- Cardiovascular Disease Initiative, Broad Institute, Cambridge, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
27
|
Rojas MG, Zigmond ZM, Pereira-Simon S, Santos Falcon N, Suresh Kumar M, Stoyell-Conti FF, Kosanovic C, Griswold AJ, Salama A, Yang X, Tabbara M, Vazquez-Padron RI, Martinez L. The intricate cellular ecosystem of human peripheral veins as revealed by single-cell transcriptomic analysis. PLoS One 2024; 19:e0296264. [PMID: 38206912 PMCID: PMC10783777 DOI: 10.1371/journal.pone.0296264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 12/09/2023] [Indexed: 01/13/2024] Open
Abstract
The venous system has been historically understudied despite its critical roles in blood distribution, heart function, and systemic immunity. This study dissects the microanatomy of upper arm veins at the single cell level, and how it relates to wall structure, remodeling processes, and inflammatory responses to injury. We applied single-cell RNA sequencing to 4 non-diseased human veins (3 basilic, 1 cephalic) obtained from organ donors, followed by bioinformatic and histological analyses. Unsupervised clustering of 20,006 cells revealed a complex ecosystem of endothelial cell (EC) types, smooth muscle cell (SMCs) and pericytes, various types of fibroblasts, and immune cell populations. The venous endothelium showed significant upregulation of cell adhesion genes, with arteriovenous zonation EC phenotypes highlighting the heterogeneity of vasa vasorum (VV) microvessels. Venous SMCs had atypical contractile phenotypes and showed widespread localization in the intima and media. MYH11+DESlo SMCs were transcriptionally associated with negative regulation of contraction and pro-inflammatory gene expression. MYH11+DEShi SMCs showed significant upregulation of extracellular matrix genes and pro-migratory mediators. Venous fibroblasts ranging from secretory to myofibroblastic phenotypes were 4X more abundant than SMCs and widely distributed throughout the wall. Fibroblast-derived angiopoietin-like factors were identified as versatile signaling hubs to regulate angiogenesis and SMC proliferation. An abundant monocyte/macrophage population was detected and confirmed by histology, including pro-inflammatory and homeostatic phenotypes, with cell counts positively correlated with age. Ligand-receptor interactome networks identified the venous endothelium in the main lumen and the VV as a niche for monocyte recruitment and infiltration. This study underscores the transcriptional uniqueness of venous cells and their relevance for vascular inflammation and remodeling processes. Findings from this study may be relevant for molecular investigations of upper arm veins used for vascular access creation, where single-cell analyses of cell composition and phenotypes are currently lacking.
Collapse
Affiliation(s)
- Miguel G. Rojas
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida, United States of America
| | - Zachary M. Zigmond
- Bruce W. Carter Veterans Affairs Medical Center, Miami, Florida, United States of America
| | - Simone Pereira-Simon
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida, United States of America
| | - Nieves Santos Falcon
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida, United States of America
| | - Maya Suresh Kumar
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida, United States of America
| | - Filipe F. Stoyell-Conti
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida, United States of America
| | - Christina Kosanovic
- John P. Hussman Institute for Human Genomics, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida, United States of America
| | - Anthony J. Griswold
- John P. Hussman Institute for Human Genomics, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida, United States of America
| | - Alghidak Salama
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida, United States of America
| | - Xiaofeng Yang
- Department of Cardiovascular Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, United States of America
| | - Marwan Tabbara
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida, United States of America
| | - Roberto I. Vazquez-Padron
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida, United States of America
- Bruce W. Carter Veterans Affairs Medical Center, Miami, Florida, United States of America
| | - Laisel Martinez
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida, United States of America
| |
Collapse
|
28
|
Kalyanaraman H, Casteel DE, Cabriales JA, Tat J, Zhuang S, Chan A, Dretchen KL, Boss GR, Pilz RB. The Antioxidant/Nitric Oxide-Quenching Agent Cobinamide Prevents Aortic Disease in a Mouse Model of Marfan Syndrome. JACC Basic Transl Sci 2024; 9:46-62. [PMID: 38362350 PMCID: PMC10864892 DOI: 10.1016/j.jacbts.2023.07.014] [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: 02/13/2023] [Revised: 07/18/2023] [Accepted: 07/24/2023] [Indexed: 02/17/2024]
Abstract
Major pathologic changes in the proximal aorta underlie the life-threatening aortic aneurysms and dissections in Marfan Syndrome; current treatments delay aneurysm development without addressing the primary pathology. Because excess oxidative stress and nitric oxide/protein kinase G signaling likely contribute to the aortopathy, we hypothesized that cobinamide, a strong antioxidant that can attenuate nitric oxide signaling, could be uniquely suited to prevent aortic disease. In a well-characterized mouse model of Marfan Syndrome, cobinamide dramatically reduced elastin breaks, prevented excess collagen deposition and smooth muscle cell apoptosis, and blocked DNA, lipid, and protein oxidation and excess nitric oxide/protein kinase G signaling in the ascending aorta. Consistent with preventing pathologic changes, cobinamide diminished aortic root dilation without affecting blood pressure. Cobinamide exhibited excellent safety and pharmacokinetic profiles indicating it could be a practical treatment. We conclude that cobinamide deserves further study as a disease-modifying treatment of Marfan Syndrome.
Collapse
Affiliation(s)
- Hema Kalyanaraman
- Department of Medicine, University of California-San Diego, La Jolla, California, USA
| | - Darren E. Casteel
- Department of Medicine, University of California-San Diego, La Jolla, California, USA
| | - Justin A. Cabriales
- Department of Medicine, University of California-San Diego, La Jolla, California, USA
| | - John Tat
- Department of Medicine, University of California-San Diego, La Jolla, California, USA
| | - Shunhui Zhuang
- Department of Medicine, University of California-San Diego, La Jolla, California, USA
| | - Adriano Chan
- Department of Medicine, University of California-San Diego, La Jolla, California, USA
| | | | - Gerry R. Boss
- Department of Medicine, University of California-San Diego, La Jolla, California, USA
| | - Renate B. Pilz
- Department of Medicine, University of California-San Diego, La Jolla, California, USA
| |
Collapse
|
29
|
Lin W, Xiong J, Jiang Y, Liu H, Bian J, Wang J, Shao Y, Ni B. Fibrillin-1 mutation contributes to Marfan syndrome by inhibiting Cav1.2-mediated cell proliferation in vascular smooth muscle cells. Channels (Austin) 2023; 17:2192377. [PMID: 36972239 PMCID: PMC10054150 DOI: 10.1080/19336950.2023.2192377] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 03/14/2023] [Indexed: 03/29/2023] Open
Abstract
Marfan syndrome (MFS) is an autosomal dominant connective tissue disorder caused by mutation in fibrillin-1 (FBN1). However, the molecular mechanism underlying MFS remains poorly understood. The study aimed to explore how the L-type calcium channel (CaV1.2) modulates disease progression of MFS and to identify a potential effective target for attenuating MFS. KEGG enrichment analysis showed that the calcium signaling pathway gene set was significantly enriched. We demonstrated that FBN1 deficiency exhibited inhibition on both the expression of Cav1.2 and proliferation of vascular smooth muscle cells (VSMCs). Then, we examined whether FBN1 mediates Cav1.2 via regulating TGF-β1. Higher levels of TGF-β1 were observed in the serum and aortic tissues from patients with MFS. TGF-β1 modulated Cav1.2 expression in a concentration-dependent manner. We evaluated the role of Cav1.2 in MFS by small interfering RNA and Cav1.2 agonist Bay K8644. The effect of Cav1.2 on cell proliferation was dependent on c-Fos activity. These results demonstrated FBN1 deficiency decreased the expression levels of Cav1.2 via regulation of TGF-β1, and downregulation of Cav1.2 inhibited cell proliferation of human aortic smooth muscle cells (HASMCs) in MFS patients. These findings suggest that Cav1.2 may be an appealing therapeutic target for MFS.
Collapse
Affiliation(s)
- Wenfeng Lin
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jiaqi Xiong
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yefan Jiang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hao Liu
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jinhui Bian
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Juejin Wang
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yongfeng Shao
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Buqing Ni
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
30
|
Seeburun S, Wu S, Hemani D, Pham L, Ju D, Xie Y, Kata P, Li L. Insights into elastic fiber fragmentation: Mechanisms and treatment of aortic aneurysm in Marfan syndrome. Vascul Pharmacol 2023; 153:107215. [PMID: 37640090 PMCID: PMC10872825 DOI: 10.1016/j.vph.2023.107215] [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: 08/06/2023] [Accepted: 08/25/2023] [Indexed: 08/31/2023]
Abstract
Marfan syndrome (MFS) is an autosomal dominant connective tissue disorder caused by mutations in fibrillin 1 (FBN1) gene. These mutations result in defects in the skeletal, ocular, and cardiovascular systems. Aortic aneurysm is the leading cause of premature mortality in untreated MFS patients. Elastic fiber fragmentation in the aortic vessel wall is a hallmark of MFS-associated aortic aneurysms. FBN1 mutations result in FBN1 fragments that also contribute to elastic fiber fragmentation. Although recent research has advanced our understanding of MFS, the contribution of elastic fiber fragmentation to the pathogenesis of aneurysm formation remains poorly understood. This review provides a comprehensive overview of the molecular mechanisms of elastic fiber fragmentation and its role in the pathogenesis of aortic aneurysm progression. Increased comprehension of elastic fragmentation has significant clinical implications for developing targeted interventions to block aneurysm progression, which would benefit not only individuals with Marfan syndrome but also other patients with aneurysms. Moreover, this review highlights an overlooked connection between inhibiting aneurysm and the restoration of elastic fibers in the vessel wall with various aneurysm inhibitors, including drugs and chemicals. Investigating the underlying molecular mechanisms could uncover innovative therapeutic strategies to inhibit elastin fragmentation and prevent the progression of aneurysms.
Collapse
Affiliation(s)
- Sheilabi Seeburun
- Department of Internal Medicine, Wayne State University, Detroit MI, USA
| | - Shichao Wu
- Department of Internal Medicine, Wayne State University, Detroit MI, USA
| | - Darshi Hemani
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit MI, USA
| | - Lucynda Pham
- Department of Internal Medicine, Wayne State University, Detroit MI, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit MI, USA
| | - Donghong Ju
- Department of Internal Medicine, Wayne State University, Detroit MI, USA
- Department of Oncology, Wayne State University, Detroit MI, USA
| | - Youming Xie
- Department of Oncology, Wayne State University, Detroit MI, USA
| | - Priyaranjan Kata
- Department of Internal Medicine, Wayne State University, Detroit MI, USA
| | - Li Li
- Department of Internal Medicine, Wayne State University, Detroit MI, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit MI, USA
| |
Collapse
|
31
|
Yurdagul A, Aikawa E. Discovering a Rare Smooth Muscle Cell Population Specific to Men in Ascending Aortic Aneurysm Using Spatial Transcriptomics. Arterioscler Thromb Vasc Biol 2023; 43:2298-2300. [PMID: 37916413 PMCID: PMC10842258 DOI: 10.1161/atvbaha.123.320235] [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: 11/03/2023]
Affiliation(s)
- Arif Yurdagul
- Department of Molecular and Cellular Physiology, LSU Health Sciences Center in Shreveport, Shreveport, LA, 71130
| | - Elena Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
32
|
Mizrak D, Zhao Y, Feng H, Macaulay J, Tang Y, Sultan Z, Zhao G, Guo Y, Zhang J, Yang B, Eugene Chen Y. Single-Molecule Spatial Transcriptomics of Human Thoracic Aortic Aneurysms Uncovers Calcification-Related CARTPT-Expressing Smooth Muscle Cells. Arterioscler Thromb Vasc Biol 2023; 43:2285-2297. [PMID: 37823268 PMCID: PMC10842613 DOI: 10.1161/atvbaha.123.319329] [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: 03/21/2023] [Accepted: 09/28/2023] [Indexed: 10/13/2023]
Abstract
BACKGROUND Although single-cell RNA-sequencing is commonly applied to dissect the heterogeneity in human tissues, it involves the preparation of single-cell suspensions via cell dissociation, causing loss of spatial information. In this study, we employed high-resolution single-cell transcriptome imaging to reveal rare smooth muscle cell (SMC) types in human thoracic aortic aneurysm (TAA) tissue samples. METHODS Single-molecule spatial distribution of transcripts from 140 genes was analyzed in fresh-frozen human TAA samples with region and sex-matched controls. In vitro studies and tissue staining were performed to examine human CART prepropeptide (CARTPT) regulation and function. RESULTS We captured thousands of cells per sample including a spatially distinct CARTPT-expressing SMC subtype enriched in male TAA samples. Immunoassays confirmed human CART (cocaine- and amphetamine-regulated transcript) protein enrichment in male TAA tissue and truncated CARTPT secretion into cell culture medium. Oxidized low-density lipoprotein, a cardiovascular risk factor, induced CARTPT expression, whereas CARTPT overexpression in human aortic SMCs increased the expression of key osteochondrogenic transcription factors and reduced contractile gene expression. Recombinant human CART treatment of human SMCs further confirmed this phenotype. Alizarin red staining revealed calcium deposition in male TAA samples showing similar localization with human CART staining. CONCLUSIONS Here, we demonstrate the feasibility of single-molecule imaging in uncovering rare SMC subtypes in the diseased human aorta, a difficult tissue to dissociate. We identified a spatially distinct CARTPT-expressing SMC subtype enriched in male human TAA samples. Our functional studies suggest that human CART promotes osteochondrogenic switch of aortic SMCs, potentially leading to medial calcification of the thoracic aorta.
Collapse
Affiliation(s)
- Dogukan Mizrak
- Department of Cardiac Surgery, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Yang Zhao
- Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Hao Feng
- Department of Cardiac Surgery, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Jane Macaulay
- Department of Cardiac Surgery, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Ying Tang
- Department of Cardiac Surgery, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Zain Sultan
- Department of Cardiac Surgery, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Guizhen Zhao
- Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Yanhong Guo
- Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Jifeng Zhang
- Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Bo Yang
- Department of Cardiac Surgery, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Y. Eugene Chen
- Department of Cardiac Surgery, University of Michigan Medical Center, Ann Arbor, MI, USA
- Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA
| |
Collapse
|
33
|
Scipione CA, Hyduk SJ, Polenz CK, Cybulsky MI. Unveiling the Hidden Landscape of Arterial Diseases at Single-Cell Resolution. Can J Cardiol 2023; 39:1781-1794. [PMID: 37716639 DOI: 10.1016/j.cjca.2023.09.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 08/25/2023] [Accepted: 09/11/2023] [Indexed: 09/18/2023] Open
Abstract
High-resolution single-cell technologies have shed light on the pathogenesis of cardiovascular diseases by enabling the discovery of novel cellular and transcriptomic signatures associated with various conditions, and uncovering new contributions of inflammatory processes, immunity, metabolic stress, and risk factors. We review the information obtained from studies using single-cell technologies in tissues with atherosclerosis and aortic aneurysms. Insights are provided on the biology of endothelial, smooth muscle, and immune cells in the arterial intima and media. In addition to cellular diversity, numerous examples of plasticity and phenotype switching are highlighted and presented in the context of normal cell functions.
Collapse
Affiliation(s)
- Corey A Scipione
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada; Departments of Laboratory Medicine and Pathobiology and Immunology, University of Toronto, Toronto, Ontario, Canada.
| | - Sharon J Hyduk
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Chanele K Polenz
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada; Departments of Laboratory Medicine and Pathobiology and Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Myron I Cybulsky
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada; Departments of Laboratory Medicine and Pathobiology and Immunology, University of Toronto, Toronto, Ontario, Canada; Peter Munk Cardiac Centre, University Health Network, Toronto, Ontario, Canada.
| |
Collapse
|
34
|
Pedroza AJ, Cheng P, Dalal AR, Baeumler K, Kino A, Tognozzi E, Shad R, Yokoyama N, Nakamura K, Mitchel O, Hiesinger W, MacFarlane EG, Fleischmann D, Woo YJ, Quertermous T, Fischbein MP. Early clinical outcomes and molecular smooth muscle cell phenotyping using a prophylactic aortic arch replacement strategy in Loeys-Dietz syndrome. J Thorac Cardiovasc Surg 2023; 166:e332-e376. [PMID: 37500053 DOI: 10.1016/j.jtcvs.2023.07.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 06/12/2023] [Accepted: 07/11/2023] [Indexed: 07/29/2023]
Abstract
OBJECTIVES Patients with Loeys-Dietz syndrome demonstrate a heightened risk of distal thoracic aortic events after valve-sparing aortic root replacement. This study assesses the clinical risks and hemodynamic consequences of a prophylactic aortic arch replacement strategy in Loeys-Dietz syndrome and characterizes smooth muscle cell phenotype in Loeys-Dietz syndrome aneurysmal and normal-sized downstream aorta. METHODS Patients with genetically confirmed Loeys-Dietz syndrome (n = 8) underwent prophylactic aortic arch replacement during valve-sparing aortic root replacement. Four-dimensional flow magnetic resonance imaging studies were performed in 4 patients with Loeys-Dietz syndrome (valve-sparing aortic root replacement + arch) and compared with patients with contemporary Marfan syndrome (valve-sparing aortic root replacement only, n = 5) and control patients (without aortopathy, n = 5). Aortic tissues from 4 patients with Loeys-Dietz syndrome and 2 organ donors were processed for anatomically segmented single-cell RNA sequencing and histologic assessment. RESULTS Patients with Loeys-Dietz syndrome valve-sparing aortic root replacement + arch had no deaths, major morbidity, or aortic events in a median of 2 years follow-up. Four-dimensional magnetic resonance imaging demonstrated altered flow parameters in patients with postoperative aortopathy relative to controls, but no clear deleterious changes due to arch replacement. Integrated analysis of aortic single-cell RNA sequencing data (>49,000 cells) identified a continuum of abnormal smooth muscle cell phenotypic modulation in Loeys-Dietz syndrome defined by reduced contractility and enriched extracellular matrix synthesis, adhesion receptors, and transforming growth factor-beta signaling. These modulated smooth muscle cells populated the Loeys-Dietz syndrome tunica media with gradually reduced density from the overtly aneurysmal root to the nondilated arch. CONCLUSIONS Patients with Loeys-Dietz syndrome demonstrated excellent surgical outcomes without overt downstream flow or shear stress disturbances after concomitant valve-sparing aortic root replacement + arch operations. Abnormal smooth muscle cell-mediated aortic remodeling occurs within the normal diameter, clinically at-risk Loeys-Dietz syndrome arch segment. These initial clinical and pathophysiologic findings support concomitant arch replacement in Loeys-Dietz syndrome.
Collapse
Affiliation(s)
- Albert J Pedroza
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, Calif
| | - Paul Cheng
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, Calif
| | - Alex R Dalal
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, Calif
| | - Kathrin Baeumler
- Department of Radiology, Stanford University School of Medicine, Stanford, Calif
| | - Aya Kino
- Department of Radiology, Stanford University School of Medicine, Stanford, Calif
| | - Emily Tognozzi
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, Calif
| | - Rohan Shad
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, Calif
| | - Nobu Yokoyama
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, Calif
| | - Ken Nakamura
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, Calif
| | - Olivia Mitchel
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, Calif
| | - William Hiesinger
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, Calif
| | - Elena Gallo MacFarlane
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Md
| | - Dominik Fleischmann
- Department of Radiology, Stanford University School of Medicine, Stanford, Calif
| | - Y Joseph Woo
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, Calif
| | - Thomas Quertermous
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, Calif
| | - Michael P Fischbein
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, Calif.
| |
Collapse
|
35
|
Shin J, Tkachenko S, Gomez D, Tripathi R, Owens GK, Cherepanova OA. Smooth muscle cells-specific loss of OCT4 accelerates neointima formation after acute vascular injury. Front Cardiovasc Med 2023; 10:1276945. [PMID: 37942066 PMCID: PMC10627795 DOI: 10.3389/fcvm.2023.1276945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Accepted: 10/09/2023] [Indexed: 11/10/2023] Open
Abstract
Introduction There is growing evidence that smooth muscle cell (SMC) phenotypic transitions play critical roles during normal developmental and tissue recovery processes and in pathological conditions such as atherosclerosis. However, the molecular mechanisms responsible for these transitions are not well understood. Recently, we found that the embryonic stem cell/induced pluripotent stem cell (iPSC) factor OCT4, which was believed to be silenced in somatic cells, plays an atheroprotective role in SMC, and regulates angiogenesis after corneal alkali burn and hindlimb ischemia by mediating microvascular SMC and pericyte migration. However, the kinetics of OCT4 activation in arterial SMC and its role in acute pathological conditions are still unknown. Methods and Results Here, using an Oct4-IRES-GFP reporter mouse model, we found that OCT4 is reactivated in the carotid artery 18 hours post-acute ligation-induced injury, a common in vivo model of the SMC phenotypic transitions. Next, using a tamoxifen-inducible Myh11-CreERT2 Oct4 knockout mouse model, we found that the loss of OCT4, specifically in SMC, led to accelerated neointima formation and increased tunica media following carotid artery ligation, at least in part by increasing SMC proliferation within the media. Bulk RNA sequencing analysis on the cultured SMC revealed significant down-regulation of the SMC contractile markers and dysregulation of the genes belonging to the regulation of cell proliferation and, positive and negative regulation for cell migration ontological groups following genetic inactivation of Oct4. We also found that loss of Oct4 resulted in suppression of contractile SMC markers after the injury and in cultured aortic SMC. Further mechanistic studies revealed that OCT4 regulates SMC contractile genes, ACTA2 and TAGLN, at least in part by direct binding to the promoters of these genes. Conclusion These results demonstrate that the pluripotency factor OCT4 is quickly activated in SMC after the acute vascular injury and inhibits SMC hyperproliferation, which may be protective in preventing excessive neointima formation.
Collapse
Affiliation(s)
- Junchul Shin
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Svyatoslav Tkachenko
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, United States
| | - Delphine Gomez
- Department of Medicine, Division of Cardiology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Rupande Tripathi
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States
| | - Gary K. Owens
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States
| | - Olga A. Cherepanova
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| |
Collapse
|
36
|
Kwartler CS, Pedroza AJ, Kaw A, Guan P, Ma S, Duan XY, Kernell C, Wang C, Pinelo JEE, Bowen MSB, Chen J, Zhong Y, Sinha S, Shen X, Fischbein MP, Milewicz DM. Nuclear Smooth Muscle α-actin Participates in Vascular Smooth Muscle Cell Differentiation. NATURE CARDIOVASCULAR RESEARCH 2023; 2:937-955. [PMID: 38919852 PMCID: PMC11198982 DOI: 10.1038/s44161-023-00337-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 08/23/2023] [Indexed: 06/27/2024]
Abstract
Missense variants throughout ACTA2, encoding smooth muscle α-actin (αSMA), predispose to adult-onset thoracic aortic disease, but variants disrupting arginine 179 (R179) lead to Smooth Muscle Dysfunction Syndrome (SMDS) characterized by diverse childhood-onset vascular diseases. Here we show that αSMA localizes to the nucleus in wildtype (WT) smooth muscle cells (SMCs), enriches in the nucleus with SMC differentiation, and associates with chromatin remodeling complexes and SMC contractile gene promotors. The ACTA2 p.R179 αSMA variant shows decreased nuclear localization. Primary SMCs from Acta2 SMC-R179C/+ mice are less differentiated than WT SMCs in vitro and in vivo and have global changes in chromatin accessibility. Induced pluripotent stem cells from patients with ACTA2 p.R179 variants fail to fully differentiate from neuroectodermal progenitor cells to SMCs, and single-cell transcriptomic analyses of an ACTA2 p.R179H patient's aortic tissue show increased SMC plasticity. Thus, nuclear αSMA participates in SMC differentiation, and loss of this nuclear activity occurs with ACTA2 p.R179 pathogenic variants.
Collapse
Affiliation(s)
- Callie S. Kwartler
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Albert J. Pedroza
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA 94305
| | - Anita Kaw
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Pujun Guan
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Shuangtao Ma
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030
- Current address: Department Medicine, Michigan State University, East Lansing, MI 48824
| | - Xue-yan Duan
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Caroline Kernell
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Charis Wang
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Jose Emiliano Esparza Pinelo
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Mikayla S. Borthwick Bowen
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Jiyuan Chen
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Yuan Zhong
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Smithville, TX 78957
| | - Sanjay Sinha
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Xuetong Shen
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, China
| | | | - Dianna M. Milewicz
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030
| |
Collapse
|
37
|
Feng X, Zheng X, Lin A, Yang S, Zhang S, Wu D, Wu W, Han X. FBN1 knockout promotes cervical artery dissection by inducing N-glycosylation alternation of extracellular matrix proteins in rat VSMCs. Cell Signal 2023; 110:110834. [PMID: 37532137 DOI: 10.1016/j.cellsig.2023.110834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 07/20/2023] [Accepted: 07/30/2023] [Indexed: 08/04/2023]
Abstract
FBN1 mutation promotes the degeneration of microfibril structures and extracellular matrix (ECM) integrity in the tunica media of the aorta in Marfan syndrome. However, whether FBN1 modulates cervical artery dissection (CAD) development and the potential molecular mechanisms of abnormal FBN1 in CAD remains elusive. In this study, FBN1 deficiency participated in the development of CAD and influenced the proliferation, apoptosis, and migration of vascular smooth muscle cells. FBN1 knockout induced alternations in mRNA levels of the transcriptome, protein expression of the proteome, and abundance of N-glycosylation of the N-glycoproteome. Comprehensive analysis of multiple omics showed up-regulation in mRNA levels of ECM proteins; yet, both the ECM protein levels and relative abundance of N-glycosylation were decreased. Moreover, we performed in vivo experiments to confirm the altered glycosylation of proteins in vascular smooth muscle cells. In conclusion, FBN1 deletion in vascular smooth muscle cells can result in altered N-glycosylation of ECM protein, which were critical for the stability of ECM and the process of CAD. This may open the way for a novel therapeutic strategy to treat people with CAD.
Collapse
Affiliation(s)
- Xiaochao Feng
- Department of Neurology, Shanghai Fifth People(')s Hospital of Fudan University, Shanghai, China
| | - Xixi Zheng
- Human Phenome Institute of Fudan University, Shanghai, China
| | - Aiqi Lin
- Department of Neurology, Huashan Hospital of Fudan University, Shanghai, China
| | - Shilin Yang
- Department of Neurology, Huashan Hospital of Fudan University, Shanghai, China
| | - Shufan Zhang
- Department of Neurology, Huashan Hospital of Fudan University, Shanghai, China
| | - Danhong Wu
- Department of Neurology, Shanghai Fifth People(')s Hospital of Fudan University, Shanghai, China
| | - Weicheng Wu
- Human Phenome Institute of Fudan University, Shanghai, China; Fudan University-Rugao People's Hospital Joint Research Institute of Longevity and Aging, Jiangsu, China.
| | - Xiang Han
- Department of Neurology, Shanghai Fifth People(')s Hospital of Fudan University, Shanghai, China; Department of Neurology, Huashan Hospital of Fudan University, Shanghai, China.
| |
Collapse
|
38
|
Chakraborty A, Li Y, Zhang C, Li Y, Rebello KR, Li S, Xu S, Vasquez HG, Zhang L, Luo W, Wang G, Chen K, Coselli JS, LeMaire SA, Shen YH. Epigenetic Induction of Smooth Muscle Cell Phenotypic Alterations in Aortic Aneurysms and Dissections. Circulation 2023; 148:959-977. [PMID: 37555319 PMCID: PMC10529114 DOI: 10.1161/circulationaha.123.063332] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 07/17/2023] [Indexed: 08/10/2023]
Abstract
BACKGROUND Smooth muscle cell (SMC) phenotypic switching has been increasingly detected in aortic aneurysm and dissection (AAD) tissues. However, the diverse SMC phenotypes in AAD tissues and the mechanisms driving SMC phenotypic alterations remain to be identified. METHODS We examined the transcriptomic and epigenomic dynamics of aortic SMC phenotypic changes in mice with angiotensin II-induced AAD by using single-cell RNA sequencing and single-cell sequencing assay for transposase-accessible chromatin. SMC phenotypic alteration in aortas from patients with ascending thoracic AAD was examined by using single-cell RNA sequencing analysis. RESULTS Single-cell RNA sequencing analysis revealed that aortic stress induced the transition of SMCs from a primary contractile phenotype to proliferative, extracellular matrix-producing, and inflammatory phenotypes. Lineage tracing showed the complete transformation of SMCs to fibroblasts and macrophages. Single-cell sequencing assay for transposase-accessible chromatin analysis indicated that these phenotypic alterations were controlled by chromatin remodeling marked by the reduced chromatin accessibility of contractile genes and the induced chromatin accessibility of genes involved in proliferation, extracellular matrix, and inflammation. IRF3 (interferon regulatory factor 3), a proinflammatory transcription factor activated by cytosolic DNA, was identified as a key driver of the transition of aortic SMCs from a contractile phenotype to an inflammatory phenotype. In cultured SMCs, cytosolic DNA signaled through its sensor STING (stimulator of interferon genes)-TBK1 (tank-binding kinase 1) to activate IRF3, which bound and recruited EZH2 (enhancer of zeste homolog 2) to contractile genes to induce repressive H3K27me3 modification and gene suppression. In contrast, double-stranded DNA-STING-IRF3 signaling induced inflammatory gene expression in SMCs. In Sting-/- mice, the aortic stress-induced transition of SMCs into an inflammatory phenotype was prevented, and SMC populations were preserved. Finally, profound SMC phenotypic alterations toward diverse directions were detected in human ascending thoracic AAD tissues. CONCLUSIONS Our study reveals the dynamic epigenetic induction of SMC phenotypic alterations in AAD. DNA damage and cytosolic leakage drive SMCs from a contractile phenotype to an inflammatory phenotype.
Collapse
Affiliation(s)
- Abhijit Chakraborty
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery (A.C., Y.L., C.Z., K.R.R., Y.L., S.X., W.L., H.G.V., L.Z., J.S.C., S.A.L., Y.H.S.), Baylor College of Medicine, Houston, TX
- Department of Cardiovascular Surgery, The Texas Heart Institute, Houston (A.C., Y.L., C.Z., K.R.R., Y.L., W.L., H.G.V., L.Z., J.S.C., S.A.L.)
| | - Yanming Li
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery (A.C., Y.L., C.Z., K.R.R., Y.L., S.X., W.L., H.G.V., L.Z., J.S.C., S.A.L., Y.H.S.), Baylor College of Medicine, Houston, TX
- Department of Cardiovascular Surgery, The Texas Heart Institute, Houston (A.C., Y.L., C.Z., K.R.R., Y.L., W.L., H.G.V., L.Z., J.S.C., S.A.L.)
| | - Chen Zhang
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery (A.C., Y.L., C.Z., K.R.R., Y.L., S.X., W.L., H.G.V., L.Z., J.S.C., S.A.L., Y.H.S.), Baylor College of Medicine, Houston, TX
- Department of Cardiovascular Surgery, The Texas Heart Institute, Houston (A.C., Y.L., C.Z., K.R.R., Y.L., W.L., H.G.V., L.Z., J.S.C., S.A.L.)
| | - Yang Li
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery (A.C., Y.L., C.Z., K.R.R., Y.L., S.X., W.L., H.G.V., L.Z., J.S.C., S.A.L., Y.H.S.), Baylor College of Medicine, Houston, TX
- Department of Cardiovascular Surgery, The Texas Heart Institute, Houston (A.C., Y.L., C.Z., K.R.R., Y.L., W.L., H.G.V., L.Z., J.S.C., S.A.L.)
| | - Kimberly R Rebello
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery (A.C., Y.L., C.Z., K.R.R., Y.L., S.X., W.L., H.G.V., L.Z., J.S.C., S.A.L., Y.H.S.), Baylor College of Medicine, Houston, TX
- Department of Cardiovascular Surgery, The Texas Heart Institute, Houston (A.C., Y.L., C.Z., K.R.R., Y.L., W.L., H.G.V., L.Z., J.S.C., S.A.L.)
| | - Shengyu Li
- Center for Bioinformatics and Computational Biology, Houston Methodist Research Institute, TX (S.L., G.W.)
| | - Samantha Xu
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery (A.C., Y.L., C.Z., K.R.R., Y.L., S.X., W.L., H.G.V., L.Z., J.S.C., S.A.L., Y.H.S.), Baylor College of Medicine, Houston, TX
| | - Hernan G Vasquez
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery (A.C., Y.L., C.Z., K.R.R., Y.L., S.X., W.L., H.G.V., L.Z., J.S.C., S.A.L., Y.H.S.), Baylor College of Medicine, Houston, TX
- Department of Cardiovascular Surgery, The Texas Heart Institute, Houston (A.C., Y.L., C.Z., K.R.R., Y.L., W.L., H.G.V., L.Z., J.S.C., S.A.L.)
| | - Lin Zhang
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery (A.C., Y.L., C.Z., K.R.R., Y.L., S.X., W.L., H.G.V., L.Z., J.S.C., S.A.L., Y.H.S.), Baylor College of Medicine, Houston, TX
- Department of Cardiovascular Surgery, The Texas Heart Institute, Houston (A.C., Y.L., C.Z., K.R.R., Y.L., W.L., H.G.V., L.Z., J.S.C., S.A.L.)
| | - Wei Luo
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery (A.C., Y.L., C.Z., K.R.R., Y.L., S.X., W.L., H.G.V., L.Z., J.S.C., S.A.L., Y.H.S.), Baylor College of Medicine, Houston, TX
- Department of Cardiovascular Surgery, The Texas Heart Institute, Houston (A.C., Y.L., C.Z., K.R.R., Y.L., W.L., H.G.V., L.Z., J.S.C., S.A.L.)
| | - Guangyu Wang
- Center for Bioinformatics and Computational Biology, Houston Methodist Research Institute, TX (S.L., G.W.)
| | - Kaifu Chen
- Department of Pediatrics, Harvard Medical School, Boston, MA (K.C.)
| | - Joseph S Coselli
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery (A.C., Y.L., C.Z., K.R.R., Y.L., S.X., W.L., H.G.V., L.Z., J.S.C., S.A.L., Y.H.S.), Baylor College of Medicine, Houston, TX
- Cardiovascular Research Institute (J.S.C., S.A.L., Y.H.S.), Baylor College of Medicine, Houston, TX
- Department of Cardiovascular Surgery, The Texas Heart Institute, Houston (A.C., Y.L., C.Z., K.R.R., Y.L., W.L., H.G.V., L.Z., J.S.C., S.A.L.)
| | - Scott A LeMaire
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery (A.C., Y.L., C.Z., K.R.R., Y.L., S.X., W.L., H.G.V., L.Z., J.S.C., S.A.L., Y.H.S.), Baylor College of Medicine, Houston, TX
- Cardiovascular Research Institute (J.S.C., S.A.L., Y.H.S.), Baylor College of Medicine, Houston, TX
- Department of Cardiovascular Surgery, The Texas Heart Institute, Houston (A.C., Y.L., C.Z., K.R.R., Y.L., W.L., H.G.V., L.Z., J.S.C., S.A.L.)
| | - Ying H Shen
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery (A.C., Y.L., C.Z., K.R.R., Y.L., S.X., W.L., H.G.V., L.Z., J.S.C., S.A.L., Y.H.S.), Baylor College of Medicine, Houston, TX
- Cardiovascular Research Institute (J.S.C., S.A.L., Y.H.S.), Baylor College of Medicine, Houston, TX
| |
Collapse
|
39
|
Lei C, Kan H, Xian X, Chen W, Xiang W, Song X, Wu J, Yang D, Zheng Y. FAM3A reshapes VSMC fate specification in abdominal aortic aneurysm by regulating KLF4 ubiquitination. Nat Commun 2023; 14:5360. [PMID: 37660071 PMCID: PMC10475135 DOI: 10.1038/s41467-023-41177-x] [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: 11/05/2022] [Accepted: 08/24/2023] [Indexed: 09/04/2023] Open
Abstract
Reprogramming of vascular smooth muscle cell (VSMC) differentiation plays an essential role in abdominal aortic aneurysm (AAA). However, the underlying mechanisms are still unclear. We explore the expression of FAM3A, a newly identified metabolic cytokine, and whether and how FAM3A regulates VSMC differentiation in AAA. We discover that FAM3A is decreased in the aortas and plasma in AAA patients and murine models. Overexpression or supplementation of FAM3A significantly attenuate the AAA formation, manifested by maintenance of the well-differentiated VSMC status and inhibition of VSMC transformation toward macrophage-, chondrocyte-, osteogenic-, mesenchymal-, and fibroblast-like cell subpopulations. Importantly, FAM3A induces KLF4 ubiquitination and reduces its phosphorylation and nuclear localization. Here, we report FAM3A as a VSMC fate-shaping regulator in AAA and reveal the underlying mechanism associated with KLF4 ubiquitination and stability, which may lead to the development of strategies based on FAM3A to restore VSMC homeostasis in AAA.
Collapse
Affiliation(s)
- Chuxiang Lei
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Dongcheng District, Beijing, 100730, China
| | - Haoxuan Kan
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Dongcheng District, Beijing, 100730, China
| | - Xiangyu Xian
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Dongcheng District, Beijing, 100730, China
| | - Wenlin Chen
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Dongcheng District, Beijing, 100730, China
| | - Wenxuan Xiang
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Dongcheng District, Beijing, 100730, China
| | - Xiaohong Song
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Dongcheng District, Beijing, 100730, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Dongcheng District, Beijing, 100730, China
| | - Jianqiang Wu
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Dongcheng District, Beijing, 100730, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Dongcheng District, Beijing, 100730, China
| | - Dan Yang
- Department of Computational Biology and Bioinformatics, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Haidian District, Beijing, 100193, China.
| | - Yuehong Zheng
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Dongcheng District, Beijing, 100730, China.
| |
Collapse
|
40
|
Weiss D, Rego BV, Cavinato C, Li DS, Kawamura Y, Emuna N, Humphrey JD. Effects of Age, Sex, and Extracellular Matrix Integrity on Aortic Dilatation and Rupture in a Mouse Model of Marfan Syndrome. Arterioscler Thromb Vasc Biol 2023; 43:e358-e372. [PMID: 37470181 PMCID: PMC10528515 DOI: 10.1161/atvbaha.123.319122] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 07/11/2023] [Indexed: 07/21/2023]
Abstract
BACKGROUND Transmural failure of the aorta is responsible for substantial morbidity and mortality; it occurs when mechanical stress exceeds strength. The aortic root and ascending aorta are susceptible to dissection and rupture in Marfan syndrome, a connective tissue disorder characterized by a progressive reduction in elastic fiber integrity. Whereas competent elastic fibers endow the aorta with compliance and resilience, cross-linked collagen fibers confer stiffness and strength. We hypothesized that postnatal reductions in matrix cross-linking increase aortopathy when turnover rates are high. METHODS We combined ex vivo biaxial mechanical testing with multimodality histological examinations to quantify expected age- and sex-dependent structural vulnerability of the ascending aorta in Fbn1C1041G/+ Marfan versus wild-type mice without and with 4-week exposures to β-aminopropionitrile, an inhibitor of lysyl oxidase-mediated cross-linking of newly synthesized elastic and collagen fibers. RESULTS We found a strong β-aminopropionitrile-associated sexual dimorphism in aortic dilatation in Marfan mice and aortic rupture in wild-type mice, with dilatation correlating with compromised elastic fiber integrity and rupture correlating with compromised collagen fibril organization. A lower incidence of rupture of β-aminopropionitrile-exposed Marfan aortas associated with increased lysyl oxidase, suggesting a compensatory remodeling of collagen that slows disease progression in the otherwise compromised Fbn1C1041G/+ aorta. CONCLUSIONS Collagen fiber structure and function in the Marfan aorta are augmented, in part, by increased lysyl oxidase in female and especially male mice, which improves structural integrity, particularly via fibrils in the adventitia. Preserving or promoting collagen cross-linking may represent a therapeutic target for an otherwise vulnerable aorta.
Collapse
Affiliation(s)
- Dar Weiss
- Department of Biomedical Engineering, Yale University, New Haven, CT (D.W., B.V.R., C.C., D.S.L., Y.K., N.E., J.D.H.)
| | - Bruno V Rego
- Department of Biomedical Engineering, Yale University, New Haven, CT (D.W., B.V.R., C.C., D.S.L., Y.K., N.E., J.D.H.)
| | - Cristina Cavinato
- Department of Biomedical Engineering, Yale University, New Haven, CT (D.W., B.V.R., C.C., D.S.L., Y.K., N.E., J.D.H.)
| | - David S Li
- Department of Biomedical Engineering, Yale University, New Haven, CT (D.W., B.V.R., C.C., D.S.L., Y.K., N.E., J.D.H.)
| | - Yuki Kawamura
- Department of Biomedical Engineering, Yale University, New Haven, CT (D.W., B.V.R., C.C., D.S.L., Y.K., N.E., J.D.H.)
| | - Nir Emuna
- Department of Biomedical Engineering, Yale University, New Haven, CT (D.W., B.V.R., C.C., D.S.L., Y.K., N.E., J.D.H.)
| | - Jay D Humphrey
- Department of Biomedical Engineering, Yale University, New Haven, CT (D.W., B.V.R., C.C., D.S.L., Y.K., N.E., J.D.H.)
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT (J.D.H.)
| |
Collapse
|
41
|
Zhang X, Che Y, Mao L, Li D, Deng J, Guo Y, Zhao Q, Zhang X, Wang L, Gao X, Chen Y, Zhang T. H3.3B controls aortic dissection progression by regulating vascular smooth muscle cells phenotypic transition and vascular inflammation. Genomics 2023; 115:110685. [PMID: 37454936 DOI: 10.1016/j.ygeno.2023.110685] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 06/13/2023] [Accepted: 07/13/2023] [Indexed: 07/18/2023]
Abstract
Aortic dissection is a devastating cardiovascular disease with a high lethality. Histone variants maintain the genomic integrity and play important roles in development and diseases. However, the role of histone variants in aortic dissection has not been well identified. In the present study, H3f3b knockdown reduced the synthetic genes expression of VSMCs, while overexpressing H3f3b exacerbated the cellular immune response of VSMCs induced by inflammatory cytokines. Combined RNA-seq and ChIP-seq analyses revealed that histone variant H3.3B directly bound to the genes related to extracellular matrix, VSMC synthetic phenotype, cytokine responses and TGFβ signaling pathway, and regulated their expressions. In addition, VSMC-specific H3f3b knockin aggravated aortic dissection development in mice, while H3f3b knockout significantly reduced the incidence of aortic dissection. In term of mechanisms, H3.3B regulated Spp1 and Ccl2 genes, inducing the apoptosis of VSMCs and recruiting macrophages. This study demonstrated the vital roles of H3.3B in phenotypic transition of VSMCs, loss of media VSMCs, and vascular inflammation in aortic dissection.
Collapse
Affiliation(s)
- Xuelin Zhang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Yang Che
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Lin Mao
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Dandan Li
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Jianqing Deng
- Vascular Surgery Department, The First Medical Center of PLA General Hospital, Beijing 100853, China
| | - Yilong Guo
- Vascular Surgery Department, The First Medical Center of PLA General Hospital, Beijing 100853, China
| | - Quanyi Zhao
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China; Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen 518057, China
| | - Xingzhong Zhang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Li Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China; Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen 518057, China; Key Laboratory of Application of Pluripotent Stem Cells in Heart Regeneration,Chinese Academy of Medical Sciences, Beijing 100037, China.
| | - Xiang Gao
- Department of Vascular Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, China.
| | - Yinan Chen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China; Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen 518057, China.
| | - Tao Zhang
- Vascular Surgery Department, Peking University People's Hospital, Beijing 100044, China.
| |
Collapse
|
42
|
Rager C, Klöpper T, Pfeil U, Tasch S, Whittaker MR, Exintaris B, Mietens A, Middendorff R. Reference Gene U2 Enables Direct Comparison between Relative Gene Expression Levels of Vascular Smooth Muscle Cells in Tissue and Culture Using Real-Time Quantitative PCR. Cells 2023; 12:2135. [PMID: 37681867 PMCID: PMC10487071 DOI: 10.3390/cells12172135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 08/17/2023] [Indexed: 09/09/2023] Open
Abstract
In nearly every lab, real-time quantitative polymerase chain reaction (qPCR) is used to quantify gene expression. However, a comparison of different samples requires the careful selection of suitable reference genes (RGs), sometimes referred to as housekeeping genes. In the case of vascular smooth muscle cells (vSMCs), it is important to know under which conditions gene expression in isolated and cultured vSMCs can be compared with vSMCs in a healthy blood vessel. We isolated the vSMC-containing layer of the rat aorta (tunica media) and used one (longitudinal) half for direct RNA extraction, while the other half served to isolate and culture vSMCs prior to RNA extraction. First, the expression of the routinely used RGs beta-actin (Actb) and Glyceraldehyde-3-phosphate dehydrogenase (Gapdh) is investigated in intact media and corresponding cultured vSMCs. Significant differences in their Ct values show that these RGs could not be used for such direct comparisons; therefore, we select 15 different RGs. Only the gene expression of the small ribonuclear protein (snRNP) U2 shows no significant differences between the absolute Ct values of cultured vSMCs and the intact media; moreover, no differences were found between male and female rats in our experimental setup. In conclusion, U2 was shown to be an appropriate (sex-independent) RG to compare relative expression levels of vSMCs in culture to those vSMCs within their physiological tissue environment.
Collapse
Affiliation(s)
- Christine Rager
- Signalling Transduction Group, Institute of Anatomy and Cell Biology, Faculty of Medicine, Justus-Liebig-University, 35392 Giessen, Germany; (C.R.)
- Drug Discovery Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Parkville, Melbourne, VIC 3052, Australia
| | - Tobias Klöpper
- Signalling Transduction Group, Institute of Anatomy and Cell Biology, Faculty of Medicine, Justus-Liebig-University, 35392 Giessen, Germany; (C.R.)
| | - Uwe Pfeil
- Cardiopulmonary Neurobiology Group, Institute of Anatomy and Cell Biology, Faculty of Medicine, Justus-Liebig-University, 35392 Giessen, Germany
| | - Sabine Tasch
- Signalling Transduction Group, Institute of Anatomy and Cell Biology, Faculty of Medicine, Justus-Liebig-University, 35392 Giessen, Germany; (C.R.)
| | - Michael Raymond Whittaker
- Drug Discovery Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Parkville, Melbourne, VIC 3052, Australia
| | - Betty Exintaris
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Parkville, Melbourne, VIC 3052, Australia
| | - Andrea Mietens
- Signalling Transduction Group, Institute of Anatomy and Cell Biology, Faculty of Medicine, Justus-Liebig-University, 35392 Giessen, Germany; (C.R.)
| | - Ralf Middendorff
- Signalling Transduction Group, Institute of Anatomy and Cell Biology, Faculty of Medicine, Justus-Liebig-University, 35392 Giessen, Germany; (C.R.)
| |
Collapse
|
43
|
Enkhjargal B, De Leon SSP, Tsukahara Y, Liu H, Huangfu Y, Wang Y, Seabra PM, Yang X, Goodman J, Wan X, Chitalia V, Han J, Seta F. Redox Dysregulation of Vascular Smooth Muscle Sirtuin-1 in Thoracic Aortic Aneurysm in Marfan Syndrome. Arterioscler Thromb Vasc Biol 2023; 43:e339-e357. [PMID: 37288573 PMCID: PMC10524979 DOI: 10.1161/atvbaha.123.319145] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 05/26/2023] [Indexed: 06/09/2023]
Abstract
BACKGROUND Thoracic aortic aneurysms (TAAs) are abnormal aortic dilatations and a major cardiovascular complication of Marfan syndrome. We previously demonstrated a critical role for vascular smooth muscle (VSM) SirT1 (sirtuin-1), a lysine deacetylase, against maladaptive aortic remodeling associated with chronic oxidative stress and aberrant activation of MMPs (matrix metalloproteinases). METHODS In this study, we investigated whether redox dysregulation of SirT1 contributed to the pathogenesis of TAA using fibrillin-1 hypomorphic mice (Fbn1mgR/mgR), an established model of Marfan syndrome prone to aortic dissection/rupture. RESULTS Oxidative stress markers 3-nitrotyrosine and 4-hydroxynonenal were significantly elevated in aortas of patients with Marfan syndrome. Moreover, reversible oxidative post-translational modifications (rOPTM) of protein cysteines, particularly S-glutathionylation, were dramatically increased in aortas of Fbn1mgR/mgR mice, before induction of severe oxidative stress markers. Fbn1mgR/mgR aortas and VSM cells exhibited an increase in rOPTM of SirT1, coinciding with the upregulation of acetylated proteins, an index of decreased SirT1 activity, and increased MMP2/9 activity. Mechanistically, we demonstrated that TGFβ (transforming growth factor beta), which was increased in Fbn1mgR/mgR aortas, stimulated rOPTM of SirT1, decreasing its deacetylase activity in VSM cells. VSM cell-specific deletion of SirT1 in Fbn1mgR/mgR mice (SMKO-Fbn1mgR/mgR) caused a dramatic increase in aortic MMP2 expression and worsened TAA progression, leading to aortic rupture in 50% of SMKO-Fbn1mgR/mgR mice, compared with 25% of Fbn1mgR/mgR mice. rOPTM of SirT1, rOPTM-mediated inhibition of SirT1 activity, and increased MMP2/9 activity were all exacerbated by the deletion of Glrx (glutaredoxin-1), a specific deglutathionylation enzyme, while being corrected by overexpression of Glrx or of an oxidation-resistant SirT1 mutant in VSM cells. CONCLUSIONS Our novel findings strongly suggest a causal role of S-glutathionylation of SirT1 in the pathogenesis of TAA. Prevention or reversal of SirT1 rOPTM may be a novel therapeutic strategy to prevent TAA and TAA dissection/ruptures in individuals with Marfan syndrome, for which, thus far, no targeted therapy has been developed.
Collapse
Affiliation(s)
- Budbazar Enkhjargal
- Vascular Biology Section, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | | | - Yuko Tsukahara
- Vascular Biology Section, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Hanxiao Liu
- Vascular Biology Section, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Yuhao Huangfu
- Vascular Biology Section, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Yu Wang
- Vascular Biology Section, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Pedro Maria Seabra
- Vascular Biology Section, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Xiaoqiu Yang
- Vascular Biology Section, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Jena Goodman
- Vascular Biology Section, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Xueping Wan
- Vascular Biology Section, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Vipul Chitalia
- Renal Section, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Jingyan Han
- Vascular Biology Section, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Francesca Seta
- Vascular Biology Section, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
44
|
Hu Y, Cai Z, He B. Smooth Muscle Heterogeneity and Plasticity in Health and Aortic Aneurysmal Disease. Int J Mol Sci 2023; 24:11701. [PMID: 37511460 PMCID: PMC10380637 DOI: 10.3390/ijms241411701] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 07/16/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
Vascular smooth muscle cells (VSMCs) are the predominant cell type in the medial layer of the aorta, which plays a critical role in the maintenance of aortic wall integrity. VSMCs have been suggested to have contractile and synthetic phenotypes and undergo phenotypic switching to contribute to the deteriorating aortic wall structure. Recently, the unprecedented heterogeneity and diversity of VSMCs and their complex relationship to aortic aneurysms (AAs) have been revealed by high-resolution research methods, such as lineage tracing and single-cell RNA sequencing. The aortic wall consists of VSMCs from different embryonic origins that respond unevenly to genetic defects that directly or indirectly regulate VSMC contractile phenotype. This difference predisposes to hereditary AAs in the aortic root and ascending aorta. Several VSMC phenotypes with different functions, for example, secreting VSMCs, proliferative VSMCs, mesenchymal stem cell-like VSMCs, immune-related VSMCs, proinflammatory VSMCs, senescent VSMCs, and stressed VSMCs are identified in non-hereditary AAs. The transformation of VSMCs into different phenotypes is an adaptive response to deleterious stimuli but can also trigger pathological remodeling that exacerbates the pathogenesis and development of AAs. This review is intended to contribute to the understanding of VSMC diversity in health and aneurysmal diseases. Papers that give an update on VSMC phenotype diversity in health and aneurysmal disease are summarized and recent insights on the role of VSMCs in AAs are discussed.
Collapse
Affiliation(s)
- Yunwen Hu
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Zhaohua Cai
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Ben He
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| |
Collapse
|
45
|
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: 7] [Impact Index Per Article: 3.5] [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
|
46
|
Rodríguez-Rovira I, López-Sainz A, Palomo-Buitrago ME, Pérez B, Jiménez-Altayó F, Campuzano V, Egea G. Hyperuricaemia Does Not Interfere with Aortopathy in a Murine Model of Marfan Syndrome. Int J Mol Sci 2023; 24:11293. [PMID: 37511051 PMCID: PMC10379183 DOI: 10.3390/ijms241411293] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 06/30/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
Redox stress is involved in the aortic aneurysm pathogenesis in Marfan syndrome (MFS). We recently reported that allopurinol, a xanthine oxidoreductase inhibitor, blocked aortopathy in a MFS mouse model acting as an antioxidant without altering uric acid (UA) plasma levels. Hyperuricaemia is ambiguously associated with cardiovascular injuries as UA, having antioxidant or pro-oxidant properties depending on the concentration and accumulation site. We aimed to evaluate whether hyperuricaemia causes harm or relief in MFS aortopathy pathogenesis. Two-month-old male wild-type (WT) and MFS mice (Fbn1C1041G/+) were injected intraperitoneally for several weeks with potassium oxonate (PO), an inhibitor of uricase (an enzyme that catabolises UA to allantoin). Plasma UA and allantoin levels were measured via several techniques, aortic root diameter and cardiac parameters by ultrasonography, aortic wall structure by histopathology, and pNRF2 and 3-NT levels by immunofluorescence. PO induced a significant increase in UA in blood plasma both in WT and MFS mice, reaching a peak at three and four months of age but decaying at six months. Hyperuricaemic MFS mice showed no change in the characteristic aortic aneurysm progression or aortic wall disarray evidenced by large elastic laminae ruptures. There were no changes in cardiac parameters or the redox stress-induced nuclear translocation of pNRF2 in the aortic tunica media. Altogether, the results suggest that hyperuricaemia interferes neither with aortopathy nor cardiopathy in MFS mice.
Collapse
Affiliation(s)
- Isaac Rodríguez-Rovira
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain
| | - Angela López-Sainz
- Department of Cardiology, Hospital Clínic de Barcelona, IDIBAPS, 08036 Barcelona, Spain
| | | | - Belen Pérez
- Department of Pharmacology, School of Medicine, Autonomous University of Barcelona, Bellaterra, 08192 Barcelona, Spain
| | - Francesc Jiménez-Altayó
- Department of Pharmacology, School of Medicine, Autonomous University of Barcelona, Bellaterra, 08192 Barcelona, Spain
| | - Victoria Campuzano
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, 28029 Madrid, Spain
| | - Gustavo Egea
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain
- UZA/UA Center of Medical Genetics, University of Antwerp, 2650 Edegem, Belgium
| |
Collapse
|
47
|
Nakamura K, Dalal AR, Yokoyama N, Pedroza AJ, Kusadokoro S, Mitchel O, Gilles C, Masoudian B, Leipzig M, Casey KM, Hiesinger W, Uchida T, Fischbein MP. Lineage-Specific Induced Pluripotent Stem Cell-Derived Smooth Muscle Cell Modeling Predicts Integrin Alpha-V Antagonism Reduces Aortic Root Aneurysm Formation in Marfan Syndrome Mice. Arterioscler Thromb Vasc Biol 2023; 43:1134-1153. [PMID: 37078287 PMCID: PMC10330156 DOI: 10.1161/atvbaha.122.318448] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Accepted: 04/05/2023] [Indexed: 04/21/2023]
Abstract
BACKGROUND The role of increased smooth muscle cell (SMC) integrin αv signaling in Marfan syndrome (MFS) aortic aneurysm remains unclear. Herein, we examine the mechanism and potential efficacy of integrin αv blockade as a therapeutic strategy to reduce aneurysm progression in MFS. METHODS Induced pluripotent stem cells (iPSCs) were differentiated into aortic SMCs of the second heart field (SHF) and neural crest (NC) lineages, enabling in vitro modeling of MFS thoracic aortic aneurysms. The pathological role of integrin αv during aneurysm formation was confirmed by blockade of integrin αv with GLPG0187 in Fbn1C1039G/+ MFS mice. RESULTS iPSC-derived MFS SHF SMCs overexpress integrin αv relative to MFS NC and healthy control SHF cells. Furthermore, integrin αv downstream targets (FAK [focal adhesion kinase]/AktThr308/mTORC1 [mechanistic target of rapamycin complex 1]) were activated, especially in MFS SHF. Treatment of MFS SHF SMCs with GLPG0187 reduced p-FAK/p-AktThr308/mTORC1 activity back to control SHF levels. Functionally, MFS SHF SMCs had increased proliferation and migration compared to MFS NC SMCs and control SMCs, which normalized with GLPG0187 treatment. In the Fbn1C1039G/+ MFS mouse model, integrin αv, p-AktThr308, and downstream targets of mTORC1 proteins were elevated in the aortic root/ascending segment compared to littermate wild-type control. Mice treated with GLPG0187 (age 6-14 weeks) had reduced aneurysm growth, elastin fragmentation, and reduction of the FAK/AktThr308/mTORC1 pathway. GLPG0187 treatment reduced the amount and severity of SMC modulation assessed by single-cell RNA sequencing. CONCLUSIONS The integrin αv-FAK-AktThr308 signaling pathway is activated in iPSC SMCs from MFS patients, specifically from the SHF lineage. Mechanistically, this signaling pathway promotes SMC proliferation and migration in vitro. As biological proof of concept, GLPG0187 treatment slowed aneurysm growth and p-AktThr308 signaling in Fbn1C1039G/+ mice. Integrin αv blockade via GLPG0187 may be a promising therapeutic approach to inhibit MFS aneurysmal growth.
Collapse
Affiliation(s)
- Ken Nakamura
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| | - Alex R. Dalal
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| | - Nobu Yokoyama
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| | - Albert J. Pedroza
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| | - Sho Kusadokoro
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| | - Olivia Mitchel
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| | - Casey Gilles
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| | - Bahar Masoudian
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| | - Matthew Leipzig
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| | - Kerriann M. Casey
- Department of Comparative Medicine, Stanford University School of Medicine. Stanford CA, USA
| | - William Hiesinger
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| | - Tetsuro Uchida
- Second Department of Surgery, Yamagata University Faculty of Medicine. Yamagata, Japan
| | - Michael P. Fischbein
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| |
Collapse
|
48
|
Pedroza AJ, Dalal AR, Shad R, Yokoyama N, Nakamura K, Mitchel O, Gilles C, Hiesinger W, Fischbein MP. Smooth Muscle Cell Klf4 Expression Is Not Required for Phenotype Modulation or Aneurysm Formation in Marfan Syndrome Mice-Brief Report. Arterioscler Thromb Vasc Biol 2023; 43:971-978. [PMID: 37128911 PMCID: PMC10434826 DOI: 10.1161/atvbaha.122.318509] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 04/13/2023] [Indexed: 05/03/2023]
Abstract
BACKGROUND Smooth muscle cell (SMC) phenotypic reprogramming toward a mixed synthetic-proteolytic state is a central feature of aortic root aneurysm in Marfan syndrome (MFS). Previous work identified Klf4 as a potential mediator of SMC plasticity in MFS. METHODS MFS (Fbn1C1041G/+) mouse strains with an inducible vascular SMC fluorescent reporter (MFSSMC) with or without SMC-specific deletion of Klf4 exons 2 to 3 (MFSSMC-Klf4Δ) were generated. Simultaneous SMC tracing and Klf4 loss-of-function (Klf4Δ mice) was induced at 6 weeks of age. Aneurysm growth was assessed via serial echocardiography (4-24 weeks). Twenty-four-week-old mice were assessed via histology, RNA in situ hybridization, and aortic single-cell RNA sequencing. RESULTS MFS mice demonstrated progressive aortic root dilatation compared with control (WTSMC) mice regardless of Klf4 genotype (P<0.001), but there was no difference in aneurysm growth in MFSSMC-Klf4Δ versus MFSSMC (P=0.884). Efficient SMC Klf4 deletion was confirmed via lineage-stratified genotyping, RNA in situ hybridization, and immunohistochemistry. Single-cell RNA sequencing of traced SMCs revealed a highly similar pattern of phenotype modulation marked by loss of contractile markers (eg, Myh11, Cnn1) and heightened expression of matrix genes (eg, Col1a1, Fn1) between Klf4 genotypes. Pseudotemporal quantitation of SMC dedifferentiation confirmed that Klf4 deletion did not alter the global extent of phenotype modulation, but reduced expression of 23 genes during this phenotype transition in MFSSMC-Klf4Δmice, including multiple chondrogenic genes expressed by only the most severely dedifferentiated SMCs (eg, Cytl1, Tnfrsf11b). CONCLUSIONS Klf4 is not required to initiate SMC phenotype modulation in MFS aneurysm but may exert regulatory control over chondrogenic genes expressed in highly dedifferentiated SMCs.
Collapse
Affiliation(s)
- Albert J. Pedroza
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| | - Alex R. Dalal
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| | - Rohan Shad
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| | - Nobu Yokoyama
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| | - Ken Nakamura
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| | - Olivia Mitchel
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| | - Casey Gilles
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| | - William Hiesinger
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| | - Michael P. Fischbein
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| |
Collapse
|
49
|
Worssam MD, Lambert J, Oc S, Taylor JCK, Taylor AL, Dobnikar L, Chappell J, Harman JL, Figg NL, Finigan A, Foote K, Uryga AK, Bennett MR, Spivakov M, Jørgensen HF. Cellular mechanisms of oligoclonal vascular smooth muscle cell expansion in cardiovascular disease. Cardiovasc Res 2023; 119:1279-1294. [PMID: 35994249 PMCID: PMC10202649 DOI: 10.1093/cvr/cvac138] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 07/08/2022] [Accepted: 08/05/2022] [Indexed: 11/14/2022] Open
Abstract
AIMS Quiescent, differentiated adult vascular smooth muscle cells (VSMCs) can be induced to proliferate and switch phenotype. Such plasticity underlies blood vessel homeostasis and contributes to vascular disease development. Oligoclonal VSMC contribution is a hallmark of end-stage vascular disease. Here, we aim to understand cellular mechanisms underpinning generation of this VSMC oligoclonality. METHODS AND RESULTS We investigate the dynamics of VSMC clone formation using confocal microscopy and single-cell transcriptomics in VSMC-lineage-traced animal models. We find that activation of medial VSMC proliferation occurs at low frequency after vascular injury and that only a subset of expanding clones migrate, which together drives formation of oligoclonal neointimal lesions. VSMC contribution in small atherosclerotic lesions is typically from one or two clones, similar to observations in mature lesions. Low frequency (<0.1%) of clonal VSMC proliferation is also observed in vitro. Single-cell RNA-sequencing revealed progressive cell state changes across a contiguous VSMC population at onset of injury-induced proliferation. Proliferating VSMCs mapped selectively to one of two distinct trajectories and were associated with cells showing extensive phenotypic switching. A proliferation-associated transitory state shared pronounced similarities with atypical SCA1+ VSMCs from uninjured mouse arteries and VSMCs in healthy human aorta. We show functionally that clonal expansion of SCA1+ VSMCs from healthy arteries occurs at higher rate and frequency compared with SCA1- cells. CONCLUSION Our data suggest that activation of proliferation at low frequency is a general, cell-intrinsic feature of VSMCs. We show that rare VSMCs in healthy arteries display VSMC phenotypic switching akin to that observed in pathological vessel remodelling and that this is a conserved feature of mouse and human healthy arteries. The increased proliferation of modulated VSMCs from healthy arteries suggests that these cells respond more readily to disease-inducing cues and could drive oligoclonal VSMC expansion.
Collapse
Affiliation(s)
- Matt D Worssam
- Section of Cardiorespiratory Medicine, Department of Medicine, University of Cambridge, Papworth Road, Cambridge Biomedical Campus, Cambridge CB2 0BB, UK
| | - Jordi Lambert
- Section of Cardiorespiratory Medicine, Department of Medicine, University of Cambridge, Papworth Road, Cambridge Biomedical Campus, Cambridge CB2 0BB, UK
| | - Sebnem Oc
- Section of Cardiorespiratory Medicine, Department of Medicine, University of Cambridge, Papworth Road, Cambridge Biomedical Campus, Cambridge CB2 0BB, UK
| | - James C K Taylor
- Section of Cardiorespiratory Medicine, Department of Medicine, University of Cambridge, Papworth Road, Cambridge Biomedical Campus, Cambridge CB2 0BB, UK
| | - Annabel L Taylor
- Section of Cardiorespiratory Medicine, Department of Medicine, University of Cambridge, Papworth Road, Cambridge Biomedical Campus, Cambridge CB2 0BB, UK
| | - Lina Dobnikar
- Section of Cardiorespiratory Medicine, Department of Medicine, University of Cambridge, Papworth Road, Cambridge Biomedical Campus, Cambridge CB2 0BB, UK
- Babraham Institute, Cambridge, UK
| | - Joel Chappell
- Section of Cardiorespiratory Medicine, Department of Medicine, University of Cambridge, Papworth Road, Cambridge Biomedical Campus, Cambridge CB2 0BB, UK
| | - Jennifer L Harman
- Section of Cardiorespiratory Medicine, Department of Medicine, University of Cambridge, Papworth Road, Cambridge Biomedical Campus, Cambridge CB2 0BB, UK
| | - Nichola L Figg
- Section of Cardiorespiratory Medicine, Department of Medicine, University of Cambridge, Papworth Road, Cambridge Biomedical Campus, Cambridge CB2 0BB, UK
| | - Alison Finigan
- Section of Cardiorespiratory Medicine, Department of Medicine, University of Cambridge, Papworth Road, Cambridge Biomedical Campus, Cambridge CB2 0BB, UK
| | - Kirsty Foote
- Section of Cardiorespiratory Medicine, Department of Medicine, University of Cambridge, Papworth Road, Cambridge Biomedical Campus, Cambridge CB2 0BB, UK
| | - Anna K Uryga
- Section of Cardiorespiratory Medicine, Department of Medicine, University of Cambridge, Papworth Road, Cambridge Biomedical Campus, Cambridge CB2 0BB, UK
| | - Martin R Bennett
- Section of Cardiorespiratory Medicine, Department of Medicine, University of Cambridge, Papworth Road, Cambridge Biomedical Campus, Cambridge CB2 0BB, UK
| | - Mikhail Spivakov
- Functional Gene Control Group, MRC London Institute of Medical Sciences, London, UK
- Institute of Clinical Sciences, Imperial College London, London, UK
| | - Helle F Jørgensen
- Section of Cardiorespiratory Medicine, Department of Medicine, University of Cambridge, Papworth Road, Cambridge Biomedical Campus, Cambridge CB2 0BB, UK
| |
Collapse
|
50
|
Yang Y, Xie E, Liu Y, Peng Z, Yu C, Hua K, Yang X. Calcium promotes vascular smooth muscle cell phenotypic switching in Marfan syndrome. Biochem Biophys Res Commun 2023; 665:124-132. [PMID: 37156050 DOI: 10.1016/j.bbrc.2023.05.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 04/19/2023] [Accepted: 05/03/2023] [Indexed: 05/10/2023]
Abstract
Fibrillin 1 (Fbn1) mutations cause Marfan syndrome (MFS), with aortic root dilatation, dissection, and rupture. Few studies reported the blood calcium and lipid profile of MFS, and the effect of vascular smooth muscle cell (VSMC) phenotypic switching on MFS aortic aneurysm is unclear. Here, we aimed to investigate the role of calcium-related VSMC phenotypic switching in MFS. We retrospectively collected MFS patients' clinical data, performed bioinformatics analysis to screen the enriched biological process in MFS patients and mice, and detected markers of VSMC phenotypic switching on Fbn1C1039G/+ mice and primary aortic vascular smooth muscle cells. We found that patients with MFS have elevated blood calcium levels and dyslipidemia. Furthermore, the calcium concentration levels were increased with age in MFS mice, accompanied by the promoted VSMC phenotypic switching, and SERCA2 contributed to maintaining the contractile phenotype of VSMCs. This study provides the first evidence that the increased calcium is associated with the promoted VSMC phenotype switching in MFS. SERCA may become a novel therapeutic target for suppressing aneurysm progression in MFS.
Collapse
Affiliation(s)
- Yunxiao Yang
- Department of Cardiovascular Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China
| | - Enzehua Xie
- Department of Cardiovascular Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College/National Center for Cardiovascular Diseases, Beijing, 100047, China
| | - Yuhua Liu
- Department of Cardiovascular Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China
| | - Zhan Peng
- Department of Cardiovascular Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China
| | - Cuntao Yu
- Department of Cardiovascular Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College/National Center for Cardiovascular Diseases, Beijing, 100047, China.
| | - Kun Hua
- Department of Cardiovascular Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China.
| | - Xiubin Yang
- Department of Cardiovascular Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China.
| |
Collapse
|