1
|
Villa-Roel N, Park C, Andueza A, Baek KI, Su A, Blaser MC, Leshnower BG, Yoganathan A, Aikawa E, Jo H. Side- and Disease-Dependent Changes in Human Aortic Valve Cell Population and Transcriptomic Heterogeneity Determined by Single-Cell RNA Sequencing. Genes (Basel) 2024; 15:1623. [PMID: 39766890 PMCID: PMC11675841 DOI: 10.3390/genes15121623] [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: 11/21/2024] [Revised: 12/13/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Calcific aortic valve disease (CAVD) is a highly prevalent disease, especially in the elderly population, but there are no effective drug therapies other than aortic valve repair or replacement. CAVD develops preferentially on the fibrosa side, while the ventricularis side remains relatively spared through unknown mechanisms. We hypothesized that the fibrosa is prone to the disease due to side-dependent differences in transcriptomic patterns and cell phenotypes. METHODS To test this hypothesis, we performed single-cell RNA sequencing using a new method to collect endothelial-enriched samples independently from the fibrosa and ventricularis sides of freshly obtained human aortic valve leaflets from five donors, ranging from non-diseased to fibrocalcific stages. RESULTS From the 82,356 aortic valve cells analyzed, we found 27 cell clusters, including seven valvular endothelial cell (VEC), nine valvular interstitial cell (VIC), and seven immune, three transitional, and one stromal cell population. We identified several side-dependent VEC subtypes with unique gene expression patterns. Homeostatic VIC clusters were abundant in non-diseased tissues, while VICs enriched with fibrocalcific genes and pathways were more prevalent in diseased leaflets. Furthermore, homeostatic macrophage (MΦ) clusters decreased while inflammatory MΦ and T-cell clusters increased with disease progression. A foamy MΦ cluster was increased in the fibrosa of mildly diseased tissues. Some side-dependent VEC clusters represented non-diseased, protective phenotypes, while others were CAVD-associated and were characterized by genes enriched in pathways of inflammation, endothelial-mesenchymal transition, apoptosis, proliferation, and fibrosis. Interestingly, we found several activator protein-1 (AP-1)-related transcription factors (FOSB, FOS, JUN, JUNB) and EGR1 to be upregulated in the fibrosa and diseased aortic valve leaflets. CONCLUSIONS Our results showed that VECs are highly heterogeneous in a side- and CAVD-dependent manner. Unique VEC clusters and their differentially regulated genes and pathways found in the fibrosa of diseased tissues may represent novel pathogenic mechanisms and potential therapeutic targets.
Collapse
Affiliation(s)
- Nicolas Villa-Roel
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, 1760 Haygood Drive, Health Sciences Research Bldg E170, Atlanta, GA 30322, USA (C.P.); (A.A.); (K.I.B.); (A.S.); (A.Y.)
| | - Christian Park
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, 1760 Haygood Drive, Health Sciences Research Bldg E170, Atlanta, GA 30322, USA (C.P.); (A.A.); (K.I.B.); (A.S.); (A.Y.)
| | - Aitor Andueza
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, 1760 Haygood Drive, Health Sciences Research Bldg E170, Atlanta, GA 30322, USA (C.P.); (A.A.); (K.I.B.); (A.S.); (A.Y.)
| | - Kyung In Baek
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, 1760 Haygood Drive, Health Sciences Research Bldg E170, Atlanta, GA 30322, USA (C.P.); (A.A.); (K.I.B.); (A.S.); (A.Y.)
| | - Ally Su
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, 1760 Haygood Drive, Health Sciences Research Bldg E170, Atlanta, GA 30322, USA (C.P.); (A.A.); (K.I.B.); (A.S.); (A.Y.)
| | - Mark C. Blaser
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (M.C.B.); (E.A.)
| | | | - Ajit Yoganathan
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, 1760 Haygood Drive, Health Sciences Research Bldg E170, Atlanta, GA 30322, USA (C.P.); (A.A.); (K.I.B.); (A.S.); (A.Y.)
| | - Elena Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (M.C.B.); (E.A.)
| | - Hanjoong Jo
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, 1760 Haygood Drive, Health Sciences Research Bldg E170, Atlanta, GA 30322, USA (C.P.); (A.A.); (K.I.B.); (A.S.); (A.Y.)
- Department of Medicine, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
2
|
Nasim S, Abujamra BA, Chaparro D, Nogueira PDS, Riva A, Hutcheson JD, Kos L. Multiple cell types including melanocytes contribute to elastogenesis in the developing murine aortic valve. Sci Rep 2024; 14:25481. [PMID: 39461968 PMCID: PMC11513076 DOI: 10.1038/s41598-024-73673-5] [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: 01/04/2024] [Accepted: 09/19/2024] [Indexed: 10/28/2024] Open
Abstract
Elastic fibers are crucial for aortic valve (AoV) function and are generated and maintained by valvular interstitial cells (VICs). VICs exhibit diverse phenotypes, yet the specific subpopulation responsible for producing and regulating elastic fibers remains unclear. This gap in knowledge is significant, given that elastin (Eln) abnormalities lead to congenital AoV defects and initiate AoV diseases. This study characterizes the timing of Eln expression in murine AoV, revealing it peaks during late embryogenesis and early postnatal stages, decreasing in adulthood. Spatial transcriptomics and RT-qPCR indicate that Eln expression correlates with genes associated to elastogenesis, including Acta2, a smooth muscle cell marker. While Eln expression is not exclusive to a single VIC subpopulation, RNAscope and immunofluorescence demonstrate a population of Eln-expressing VICs that co-express alpha smooth muscle actin and melanocytic markers. As previously reported in adult mice, we show a relationship between AoV pigment and elastic fiber patterning during early postnatal stages and further show that melanocytes may play a critical role in elastogenesis. In summary, Eln is expressed in the AoV during early postnatal stages by cells co-expressing markers of various types, highlighting the complexity of VICs phenotypes and their role in elastic fiber regulation.
Collapse
Affiliation(s)
- Sana Nasim
- Department of Biomedical Engineering, Florida International University, Miami, FL, 33174, USA
| | - Beatriz Abdo Abujamra
- Department of Biological Sciences, Florida International University, Miami, FL, 33199, USA
| | - Daniel Chaparro
- Department of Biomedical Engineering, Florida International University, Miami, FL, 33174, USA
| | - Perony Da Silva Nogueira
- Biomolecular Sciences Institute, Florida International University, Miami, FL, 33199, USA
- Department of Biological Sciences, Florida International University, Miami, FL, 33199, USA
| | | | - Joshua D Hutcheson
- Department of Biomedical Engineering, Florida International University, Miami, FL, 33174, USA.
- Biomolecular Sciences Institute, Florida International University, Miami, FL, 33199, USA.
| | - Lidia Kos
- Biomolecular Sciences Institute, Florida International University, Miami, FL, 33199, USA.
- Department of Biological Sciences, Florida International University, Miami, FL, 33199, USA.
| |
Collapse
|
3
|
Tan C, Ge ZD, Kurup S, Dyakiv Y, Liu T, Muller WA, Kume T. FOXC1 and FOXC2 Ablation Causes Abnormal Valvular Endothelial Cell Junctions and Lymphatic Vessel Formation in Myxomatous Mitral Valve Degeneration. Arterioscler Thromb Vasc Biol 2024; 44:1944-1959. [PMID: 38989578 PMCID: PMC11335087 DOI: 10.1161/atvbaha.124.320316] [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: 09/12/2023] [Accepted: 06/26/2024] [Indexed: 07/12/2024]
Abstract
BACKGROUND Mitral valve (MV) disease including myxomatous degeneration is the most common form of valvular heart disease with an age-dependent frequency. Genetic evidence indicates that mutations of the human transcription factor FOXC1 are associated with MV defects, including MV regurgitation. In this study, we sought to determine whether murine Foxc1 and its closely related factor, Foxc2, are required in valvular endothelial cells (VECs) for the maintenance of MV leaflets, including VEC junctions and the stratified trilaminar ECM (extracellular matrix). METHODS Adult mice carrying tamoxifen-inducible, vascular endothelial cell (EC), and lymphatic EC-specific, compound Foxc1;Foxc2 mutations (ie, EC-Foxc-DKO and lymphatic EC-Foxc-DKO mice, respectively) were used to study the function of Foxc1 and Foxc2 in the maintenance of MVs. The EC and lymphatic EC mutations of Foxc1/c2 were induced at 7 to 8 weeks of age by tamoxifen treatment, and abnormalities in the MVs of these mutant mice were assessed via whole-mount immunostaining, immunohistochemistry/RNAscope, Movat pentachrome/Masson Trichrome staining, and Evans blue injection. RESULTS EC deletions of Foxc1 and Foxc2 in mice resulted in abnormally extended and thicker MVs by causing defects in the regulation of ECM organization with increased proteoglycan and decreased collagen. Notably, reticular adherens junctions were found in VECs of control MV leaflets, and these reticular structures were severely disrupted in EC-Foxc-DKO mice. PROX1 (prospero homeobox protein 1), a key regulator in a subset of VECs on the fibrosa side of MVs, was downregulated in EC-Foxc1/c2 mutant VECs. Furthermore, we determined the precise location of lymphatic vessels in murine MVs, and these lymphatic vessels were aberrantly expanded and dysfunctional in EC-Foxc1/c2 mutant MVs. Lymphatic EC deletion of Foxc1/c2 also resulted in similar structural/ECM abnormalities as seen in EC-Foxc1/c2 mutant MVs. CONCLUSIONS Our results indicate that Foxc1 and Foxc2 are required for maintaining the integrity of the MV, including VEC junctions, ECM organization, and lymphatic vessel formation/function to prevent myxomatous MV degeneration.
Collapse
Affiliation(s)
- Can Tan
- Department of Medicine, Feinberg Cardiovascular and Renal Research Institute (C.T., S.K., Y.D., T.L., T.K.), Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Zhi-Dong Ge
- Departments of Pediatrics, Surgery, and Pathology, Cardiovascular-Thoracic Surgery and the Heart Center, Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital of Chicago (Z.-D.G.), Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Shreya Kurup
- Department of Medicine, Feinberg Cardiovascular and Renal Research Institute (C.T., S.K., Y.D., T.L., T.K.), Feinberg School of Medicine, Northwestern University, Chicago, IL
- Honors College, University of Illinois at Chicago (S.K.)
| | - Yaryna Dyakiv
- Department of Medicine, Feinberg Cardiovascular and Renal Research Institute (C.T., S.K., Y.D., T.L., T.K.), Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Ting Liu
- Department of Medicine, Feinberg Cardiovascular and Renal Research Institute (C.T., S.K., Y.D., T.L., T.K.), Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - William A. Muller
- Department of Pathology (W.A.M.), Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Tsutomu Kume
- Department of Medicine, Feinberg Cardiovascular and Renal Research Institute (C.T., S.K., Y.D., T.L., T.K.), Feinberg School of Medicine, Northwestern University, Chicago, IL
| |
Collapse
|
4
|
Small AM, Yutzey KE, Binstadt BA, Voigts Key K, Bouatia-Naji N, Milan D, Aikawa E, Otto CM, St Hilaire C. Unraveling the Mechanisms of Valvular Heart Disease to Identify Medical Therapy Targets: A Scientific Statement From the American Heart Association. Circulation 2024; 150:e109-e128. [PMID: 38881493 PMCID: PMC11542557 DOI: 10.1161/cir.0000000000001254] [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] [Indexed: 06/18/2024]
Abstract
Valvular heart disease is a common cause of morbidity and mortality worldwide and has no effective medical therapy. Severe disease is managed with valve replacement procedures, which entail high health care-related costs and postprocedural morbidity and mortality. Robust ongoing research programs have elucidated many important molecular pathways contributing to primary valvular heart disease. However, there remain several key challenges inherent in translating research on valvular heart disease to viable molecular targets that can progress through the clinical trials pathway and effectively prevent or modify the course of these common conditions. In this scientific statement, we review the basic cellular structures of the human heart valves and discuss how these structures change in primary valvular heart disease. We focus on the most common primary valvular heart diseases, including calcific aortic stenosis, bicuspid aortic valves, mitral valve prolapse, and rheumatic heart disease, and outline the fundamental molecular discoveries contributing to each. We further outline potential therapeutic molecular targets for primary valvular heart disease and discuss key knowledge gaps that might serve as future research priorities.
Collapse
|
5
|
Majumdar U, Choudhury TZ, Manivannan S, Ueyama Y, Basu M, Garg V. Single-cell RNA-sequencing analysis of aortic valve interstitial cells demonstrates the regulation of integrin signaling by nitric oxide. Front Cardiovasc Med 2022; 9:742850. [PMID: 36386365 PMCID: PMC9640371 DOI: 10.3389/fcvm.2022.742850] [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] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 09/30/2022] [Indexed: 11/22/2022] Open
Abstract
Calcific aortic valve disease (CAVD) is an increasingly prevalent condition among the elderly population that is associated with significant morbidity and mortality. Insufficient understanding of the underlying disease mechanisms has hindered the development of pharmacologic therapies for CAVD. Recently, we described nitric oxide (NO) mediated S-nitrosylation as a novel mechanism for preventing the calcific process. We demonstrated that NO donor or an S-nitrosylating agent, S-nitrosoglutathione (GSNO), inhibits spontaneous calcification in porcine aortic valve interstitial cells (pAVICs) and this was supported by single-cell RNA sequencing (scRNAseq) that demonstrated NO donor and GSNO inhibited myofibroblast activation of pAVICs. Here, we investigated novel signaling pathways that are critical for the calcification of pAVICs that are altered by NO and GSNO by performing an in-depth analysis of the scRNA-seq dataset. Transcriptomic analysis revealed 1,247 differentially expressed genes in pAVICs after NO donor or GSNO treatment compared to untreated cells. Pathway-based analysis of the differentially expressed genes revealed an overrepresentation of the integrin signaling pathway, along with the Rho GTPase, Wnt, TGF-β, and p53 signaling pathways. We demonstrate that ITGA8 and VCL, two of the identified genes from the integrin signaling pathway, which are known to regulate cell-extracellular matrix (ECM) communication and focal adhesion, were upregulated in both in vitro and in vivo calcific conditions. Reduced expression of these genes after treatment with NO donor suggests that NO inhibits calcification by targeting myofibroblast adhesion and ECM remodeling. In addition, withdrawal of NO donor after 3 days of exposure revealed that NO-mediated transcriptional and translational regulation is a transient event and requires continuous NO exposure to inhibit calcification. Overall, our data suggest that NO and S-nitrosylation regulate the integrin signaling pathway to maintain healthy cell-ECM interaction and prevent CAVD.
Collapse
Affiliation(s)
- Uddalak Majumdar
- Center for Cardiovascular Research, Nationwide Children’s Hospital, Columbus, OH, United States
- The Heart Center, Nationwide Children’s Hospital, Columbus, OH, United States
| | - Talita Z. Choudhury
- Center for Cardiovascular Research, Nationwide Children’s Hospital, Columbus, OH, United States
- The Heart Center, Nationwide Children’s Hospital, Columbus, OH, United States
| | - Sathiyanarayanan Manivannan
- Center for Cardiovascular Research, Nationwide Children’s Hospital, Columbus, OH, United States
- The Heart Center, Nationwide Children’s Hospital, Columbus, OH, United States
| | - Yukie Ueyama
- Center for Cardiovascular Research, Nationwide Children’s Hospital, Columbus, OH, United States
- The Heart Center, Nationwide Children’s Hospital, Columbus, OH, United States
| | - Madhumita Basu
- Center for Cardiovascular Research, Nationwide Children’s Hospital, Columbus, OH, United States
- The Heart Center, Nationwide Children’s Hospital, Columbus, OH, United States
- Department of Pediatrics, The Ohio State University, Columbus, OH, United States
| | - Vidu Garg
- Center for Cardiovascular Research, Nationwide Children’s Hospital, Columbus, OH, United States
- The Heart Center, Nationwide Children’s Hospital, Columbus, OH, United States
- Department of Pediatrics, The Ohio State University, Columbus, OH, United States
- Department of Molecular Genetics, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
6
|
Nasim S, Pandey P, Kanashiro-Takeuchi RM, He J, Hutcheson JD, Kos L. Pigmentation Affects Elastic Fiber Patterning and Biomechanical Behavior of the Murine Aortic Valve. Front Cardiovasc Med 2021; 8:754560. [PMID: 34957247 PMCID: PMC8702816 DOI: 10.3389/fcvm.2021.754560] [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: 08/06/2021] [Accepted: 11/04/2021] [Indexed: 11/29/2022] Open
Abstract
The aortic valve (AoV) maintains unidirectional blood distribution from the left ventricle of the heart to the aorta for systemic circulation. The AoV leaflets rely on a precise extracellular matrix microarchitecture of collagen, elastin, and proteoglycans for appropriate biomechanical performance. We have previously demonstrated a relationship between the presence of pigment in the mouse AoV with elastic fiber patterning using multiphoton imaging. Here, we extended those findings using wholemount confocal microscopy revealing that elastic fibers were diminished in the AoV of hypopigmented mice (KitWv and albino) and were disorganized in the AoV of K5-Edn3 transgenic hyperpigmented mice when compared to wild type C57BL/6J mice. We further used atomic force microscopy to measure stiffness differences in the wholemount AoV leaflets of mice with different levels of pigmentation. We show that AoV leaflets of K5-Edn3 had overall higher stiffness (4.42 ± 0.35 kPa) when compared to those from KitWv (2.22 ± 0.21 kPa), albino (2.45 ± 0.16 kPa), and C57BL/6J (3.0 ± 0.16 kPa) mice. Despite the striking elastic fiber phenotype and noted stiffness differences, adult mutant mice were found to have no overt cardiac differences as measured by echocardiography. Our results indicate that pigmentation, but not melanocytes, is required for proper elastic fiber organization in the mouse AoV and dictates its biomechanical properties.
Collapse
Affiliation(s)
- Sana Nasim
- Department of Biomedical Engineering, Florida International University, Miami, FL, United States
| | - Popular Pandey
- Biomolecular Sciences Institute, Florida International University, Miami, FL, United States.,Department of Physics, Florida International University, Miami, FL, United States
| | - Rosemeire M Kanashiro-Takeuchi
- Department of Molecular and Cellular Pharmacology, Leonard M Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Jin He
- Biomolecular Sciences Institute, Florida International University, Miami, FL, United States.,Department of Physics, Florida International University, Miami, FL, United States
| | - Joshua D Hutcheson
- Department of Biomedical Engineering, Florida International University, Miami, FL, United States.,Biomolecular Sciences Institute, Florida International University, Miami, FL, United States
| | - Lidia Kos
- Biomolecular Sciences Institute, Florida International University, Miami, FL, United States.,Department of Biological Sciences, Florida International University, Miami, FL, United States
| |
Collapse
|