1
|
Ogawa Y, Yamamoto A, Yamazawa S, Ikemura M, Hirata Y, Inuzuka R. Decreased smooth muscle cells and fibrous thickening of the tunica media in peripheral pulmonary artery stenosis in Alagille syndrome. Cardiovasc Pathol 2025; 74:107677. [PMID: 39069193 DOI: 10.1016/j.carpath.2024.107677] [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] [Received: 03/20/2024] [Revised: 07/14/2024] [Accepted: 07/21/2024] [Indexed: 07/30/2024] Open
Abstract
Alagille syndrome is caused by mutations in genes involved in NOTCH signaling, specifically JAG1 and NOTCH2, and is associated with a high rate of peripheral pulmonary artery stenosis. In this study, we report the case of an infant with Alagille syndrome caused by a JAG1 mutation, who succumbed to acute exacerbation of right heart failure due to severe peripheral pulmonary artery stenosis. The autopsy revealed that the peripheral pulmonary arteries were significantly stenosed, exhibiting hypoplasia and thickened vessel walls. Histological examination of the pulmonary artery walls showed a decrease in smooth muscle cells in the tunica media and an increase in collagen and elastic fibers, although the intrapulmonary arteries were intact. These findings are important for understanding the pathogenesis of Alagille syndrome and developing treatment strategies for peripheral pulmonary artery stenosis.
Collapse
Affiliation(s)
- Yosuke Ogawa
- Department of Pediatrics, The University of Tokyo Hospital, Tokyo, Japan
| | - Amane Yamamoto
- Department of Pathology, The University of Tokyo Hospital, Tokyo, Japan
| | - Sho Yamazawa
- Department of Pathology, The University of Tokyo Hospital, Tokyo, Japan
| | - Masako Ikemura
- Department of Pathology, The University of Tokyo Hospital, Tokyo, Japan
| | - Yasutaka Hirata
- Department of Cardiac Surgery, The University of Tokyo Hospital, Tokyo, Japan
| | - Ryo Inuzuka
- Department of Pediatrics, The University of Tokyo Hospital, Tokyo, Japan.
| |
Collapse
|
2
|
Yan W, Cheng J, Wu H, Gao Z, Li Z, Cao C, Meng Q, Wu Y, Ren S, Zhao F, Wang H, Liu P, Wang J, Hu X, Ao Y. Vascular Smooth Muscle Cells Transdifferentiate into Chondrocyte-Like Cells and Facilitate Meniscal Fibrocartilage Regeneration. RESEARCH (WASHINGTON, D.C.) 2024; 7:0555. [PMID: 39717465 PMCID: PMC11665451 DOI: 10.34133/research.0555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 11/25/2024] [Accepted: 12/02/2024] [Indexed: 12/25/2024]
Abstract
The effective and translational strategy to regenerate knee meniscal fibrocartilage remained challenging. Herein, we first identified vascular smooth muscle cells (VSMCs) transdifferentiated into fibrochondrocytes and participated in spontaneous meniscal regeneration using smooth muscle cell lineage tracing transgenic mice meniscal defect model. Then, we identified low-intensity pulsed ultrasound (LIPUS) acoustic stimulus enhanced fibrochondrogenic transdifferentiation of VSMCs in vitro and in vivo. Mechanistically, LIPUS stimulus could up-regulate mechanosensitive ion channel Piezo1 expression and then activate the transforming growth factor β1 (TGFβ1) signal, following repression of the Notch signal, consequently enhancing fibrochondrogenic transdifferentiation of VSMCs. Finally, we demonstrated that the regular LIPUS stimulus enhanced anisotropic native-like meniscal fibrocartilage tissue regeneration in a beagle canine subtotal meniscectomy model at 6 months postoperatively. The single-cell RNA sequencing analysis confirmed the role of VSMC fibrochondrogenic transdifferentiation in meniscal regeneration.
Collapse
Affiliation(s)
- Wenqiang Yan
- Department of Sports Medicine, Peking University Third Hospital,
Institute of Sports Medicine of Peking University, Beijing, China
- Beijing Key Laboratory of Sports Injuries, Beijing, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, China
| | - Jin Cheng
- Department of Sports Medicine, Peking University Third Hospital,
Institute of Sports Medicine of Peking University, Beijing, China
- Beijing Key Laboratory of Sports Injuries, Beijing, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, China
| | - Haoda Wu
- Department of Sports Medicine, Peking University Third Hospital,
Institute of Sports Medicine of Peking University, Beijing, China
- Beijing Key Laboratory of Sports Injuries, Beijing, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, China
| | - Zeyuan Gao
- Department of Sports Medicine, Peking University Third Hospital,
Institute of Sports Medicine of Peking University, Beijing, China
- Beijing Key Laboratory of Sports Injuries, Beijing, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, China
| | - Zong Li
- Department of Sports Medicine, Peking University Third Hospital,
Institute of Sports Medicine of Peking University, Beijing, China
- Beijing Key Laboratory of Sports Injuries, Beijing, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, China
| | - Chenxi Cao
- Department of Sports Medicine, Peking University Third Hospital,
Institute of Sports Medicine of Peking University, Beijing, China
- Beijing Key Laboratory of Sports Injuries, Beijing, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, China
| | - Qingyang Meng
- Department of Sports Medicine, Peking University Third Hospital,
Institute of Sports Medicine of Peking University, Beijing, China
- Beijing Key Laboratory of Sports Injuries, Beijing, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, China
| | - Yue Wu
- Department of Sports Medicine, Peking University Third Hospital,
Institute of Sports Medicine of Peking University, Beijing, China
- Beijing Key Laboratory of Sports Injuries, Beijing, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, China
| | - Shuang Ren
- Department of Sports Medicine, Peking University Third Hospital,
Institute of Sports Medicine of Peking University, Beijing, China
- Beijing Key Laboratory of Sports Injuries, Beijing, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, China
| | - Fengyuan Zhao
- Department of Sports Medicine, Peking University Third Hospital,
Institute of Sports Medicine of Peking University, Beijing, China
- Beijing Key Laboratory of Sports Injuries, Beijing, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, China
| | - Hongde Wang
- Department of Sports Medicine, Peking University Third Hospital,
Institute of Sports Medicine of Peking University, Beijing, China
- Beijing Key Laboratory of Sports Injuries, Beijing, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, China
| | - Ping Liu
- Department of Sports Medicine, Peking University Third Hospital,
Institute of Sports Medicine of Peking University, Beijing, China
- Beijing Key Laboratory of Sports Injuries, Beijing, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, China
| | - Jianquan Wang
- Department of Sports Medicine, Peking University Third Hospital,
Institute of Sports Medicine of Peking University, Beijing, China
- Beijing Key Laboratory of Sports Injuries, Beijing, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, China
| | - Xiaoqing Hu
- Department of Sports Medicine, Peking University Third Hospital,
Institute of Sports Medicine of Peking University, Beijing, China
- Beijing Key Laboratory of Sports Injuries, Beijing, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, China
| | - Yingfang Ao
- Department of Sports Medicine, Peking University Third Hospital,
Institute of Sports Medicine of Peking University, Beijing, China
- Beijing Key Laboratory of Sports Injuries, Beijing, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, China
| |
Collapse
|
3
|
Yu Y, Cai Y, Yang F, Yang Y, Cui Z, Shi D, Bai R. Vascular smooth muscle cell phenotypic switching in atherosclerosis. Heliyon 2024; 10:e37727. [PMID: 39309965 PMCID: PMC11416558 DOI: 10.1016/j.heliyon.2024.e37727] [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: 05/30/2024] [Revised: 08/25/2024] [Accepted: 09/09/2024] [Indexed: 09/25/2024] Open
Abstract
Atherosclerosis (AS) is a complex pathology process involving intricate interactions among various cells and biological processes. Vascular smooth muscle cells (VSMCs) are the predominant cell type in normal arteries, and under atherosclerotic stimuli, VSMCs respond to altered blood flow and microenvironment changes by downregulating contractile markers and switching their phenotype. This review overviews the diverse phenotypes of VSMCs, including the canonical contractile VSMCs, synthetic VSMCs, and phenotypes resembling macrophages, foam cells, myofibroblasts, osteoblasts/chondrocytes, and mesenchymal stem cells. We summarize their presumed protective and pro-atherosclerotic roles in AS development. Additionally, we underscore the molecular mechanisms and regulatory pathways governing VSMC phenotypic switching, encompassing transcriptional regulation, biochemical factors, plaque microenvironment, epigenetics, miRNAs, and the cytoskeleton, emphasizing their significance in AS development. Finally, we outline probable future research directions targeting VSMCs, offering insights into potential therapeutic strategies for AS management.
Collapse
Affiliation(s)
- Yanqiao Yu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, 100091, China
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yajie Cai
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, 100091, China
| | - Furong Yang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, 100091, China
| | - Yankai Yang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, 100091, China
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Zhuorui Cui
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, 100091, China
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Dazhuo Shi
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, 100091, China
| | - Ruina Bai
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, 100091, China
| |
Collapse
|
4
|
Zhao J, Sormani L, Jacquelin S, Li H, Styke C, Zhou C, Beesley J, Oon L, Kaur S, Sim SL, Wong HY, Dight J, Hashemi G, Shafiee A, Roy E, Patel J, Khosrotehrani K. Distinct roles of SOX9 in self-renewal of progenitors and mesenchymal transition of the endothelium. Angiogenesis 2024; 27:545-560. [PMID: 38733496 PMCID: PMC11303482 DOI: 10.1007/s10456-024-09927-7] [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: 12/04/2023] [Accepted: 04/24/2024] [Indexed: 05/13/2024]
Abstract
Regenerative capabilities of the endothelium rely on vessel-resident progenitors termed endothelial colony forming cells (ECFCs). This study aimed to investigate if these progenitors are impacted by conditions (i.e., obesity or atherosclerosis) characterized by increased serum levels of oxidized low-density lipoprotein (oxLDL), a known inducer of Endothelial-to-Mesenchymal Transition (EndMT). Our investigation focused on understanding the effects of EndMT on the self-renewal capabilities of progenitors and the associated molecular alterations. In the presence of oxLDL, ECFCs displayed classical features of EndMT, through reduced endothelial gene and protein expression, function as well as increased mesenchymal genes, contractility, and motility. Additionally, ECFCs displayed a dramatic loss in self-renewal capacity in the presence of oxLDL. RNA-sequencing analysis of ECFCs exposed to oxLDL validated gene expression changes suggesting EndMT and identified SOX9 as one of the highly differentially expressed genes. ATAC sequencing analysis identified SOX9 binding sites associated with regions of dynamic chromosome accessibility resulting from oxLDL exposure, further pointing to its importance. EndMT phenotype and gene expression changes induced by oxLDL in vitro or high fat diet (HFD) in vivo were reversed by the silencing of SOX9 in ECFCs or the endothelial-specific conditional knockout of Sox9 in murine models. Overall, our findings support that EndMT affects vessel-resident endothelial progenitor's self-renewal. SOX9 activation is an early transcriptional event that drives the mesenchymal transition of endothelial progenitor cells. The identification of the molecular network driving EndMT in vessel-resident endothelial progenitors presents a new avenue in understanding and preventing a range of condition where this process is involved.
Collapse
Affiliation(s)
- Jilai Zhao
- Frazer Institute, The University of Queensland, Dermatology Research Centre, Experimental Dermatology Group, Brisbane, QLD, 4102, Australia
| | - Laura Sormani
- Frazer Institute, The University of Queensland, Dermatology Research Centre, Experimental Dermatology Group, Brisbane, QLD, 4102, Australia
| | - Sebastien Jacquelin
- Mater Research, Translational Research Institute, Macrophage Biology Laboratory, Brisbane, QLD, 4102, Australia
| | - Haiming Li
- Frazer Institute, The University of Queensland, Dermatology Research Centre, Experimental Dermatology Group, Brisbane, QLD, 4102, Australia
| | - Cassandra Styke
- Frazer Institute, The University of Queensland, Dermatology Research Centre, Experimental Dermatology Group, Brisbane, QLD, 4102, Australia
| | - Chenhao Zhou
- Frazer Institute, The University of Queensland, Dermatology Research Centre, Experimental Dermatology Group, Brisbane, QLD, 4102, Australia
| | - Jonathan Beesley
- Cancer Research Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
| | - Linus Oon
- Frazer Institute, The University of Queensland, Dermatology Research Centre, Experimental Dermatology Group, Brisbane, QLD, 4102, Australia
| | - Simranpreet Kaur
- Frazer Institute, The University of Queensland, Dermatology Research Centre, Experimental Dermatology Group, Brisbane, QLD, 4102, Australia
- Mater Research, Translational Research Institute, Macrophage Biology Laboratory, Brisbane, QLD, 4102, Australia
| | - Seen-Ling Sim
- Frazer Institute, The University of Queensland, Dermatology Research Centre, Experimental Dermatology Group, Brisbane, QLD, 4102, Australia
| | - Ho Yi Wong
- Frazer Institute, The University of Queensland, Dermatology Research Centre, Experimental Dermatology Group, Brisbane, QLD, 4102, Australia
| | - James Dight
- Frazer Institute, The University of Queensland, Dermatology Research Centre, Experimental Dermatology Group, Brisbane, QLD, 4102, Australia
| | - Ghazaleh Hashemi
- Frazer Institute, The University of Queensland, Dermatology Research Centre, Experimental Dermatology Group, Brisbane, QLD, 4102, Australia
| | - Abbas Shafiee
- Frazer Institute, The University of Queensland, Dermatology Research Centre, Experimental Dermatology Group, Brisbane, QLD, 4102, Australia
| | - Edwige Roy
- Frazer Institute, The University of Queensland, Dermatology Research Centre, Experimental Dermatology Group, Brisbane, QLD, 4102, Australia
| | - Jatin Patel
- Centre for Ageing Research Program, Queensland University of Technology, Brisbane, QLD, 4102, Australia
| | - Kiarash Khosrotehrani
- Frazer Institute, The University of Queensland, Dermatology Research Centre, Experimental Dermatology Group, Brisbane, QLD, 4102, Australia.
| |
Collapse
|
5
|
Ritsvall O, Albinsson S. Emerging role of YAP/TAZ in vascular mechanotransduction and disease. Microcirculation 2024; 31:e12838. [PMID: 38011540 DOI: 10.1111/micc.12838] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 11/08/2023] [Accepted: 11/13/2023] [Indexed: 11/29/2023]
Abstract
Cells have an incredible ability to physically interact with neighboring cells and their environment. They can detect and respond to mechanical forces by converting mechanical stimuli into biochemical signals in a process known as mechanotransduction. This is a key process for the adaption of vascular smooth muscle and endothelial cells to altered flow and pressure conditions. Mechanical stimuli, referring to a physical force exerted on cells, are primarily sensed by transmembrane proteins and the actin cytoskeleton, which initiate a cascade of intracellular events, including the activation of signaling pathways, ion channels, and transcriptional regulators. Recent work has highlighted an important role of the transcriptional coactivators YAP/TAZ for mechanotransduction in vascular cells. Interestingly, the activity of YAP/TAZ decreases with age, providing a potential mechanism for the detrimental effects of aging in the vascular wall. In this review, we summarize the current knowledge on the functional role of YAP and TAZ in vascular endothelial and smooth muscle cells for mechanotransduction in homeostasis and disease. In particular, the review is focused on in vivo observations from conditional knockout (KO) models of YAP/TAZ and the potential implications these studies may have for our understanding of vascular disease development.
Collapse
Affiliation(s)
- Olivia Ritsvall
- Department of Experimental Medical Science, Molecular Vascular Physiology, Lund University, Lund, Sweden
| | - Sebastian Albinsson
- Department of Experimental Medical Science, Molecular Vascular Physiology, Lund University, Lund, Sweden
| |
Collapse
|
6
|
Nasr M, Fay A, Lupieri A, Malet N, Darmon A, Zahreddine R, Swiader A, Wahart A, Viaud J, Nègre-Salvayre A, Hirsch E, Monteyne D, Perez-Morgà D, Dupont N, Codogno P, Ramel D, Morel E, Laffargue M, Gayral S. PI3KCIIα-Dependent Autophagy Program Protects From Endothelial Dysfunction and Atherosclerosis in Response to Low Shear Stress in Mice. Arterioscler Thromb Vasc Biol 2024; 44:620-634. [PMID: 38152888 DOI: 10.1161/atvbaha.123.319978] [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: 08/08/2023] [Accepted: 12/12/2023] [Indexed: 12/29/2023]
Abstract
BACKGROUND The ability to respond to mechanical forces is a basic requirement for maintaining endothelial cell (ECs) homeostasis, which is continuously subjected to low shear stress (LSS) and high shear stress (HSS). In arteries, LSS and HSS have a differential impact on EC autophagy processes. However, it is still unclear whether LSS and HSS differently tune unique autophagic machinery or trigger specific autophagic responses in ECs. METHODS Using fluid flow system to generate forces on EC and multiscale imaging analyses on ApoE-/- mice whole arteries, we studied the cellular and molecular mechanism involved in autophagic response to LSS or HSS on the endothelium. RESULTS We found that LSS and HSS trigger autophagy activation by mobilizing specific autophagic signaling modules. Indeed, LSS-induced autophagy in endothelium was independent of the class III PI3K (phosphoinositide 3-kinase) VPS34 (vacuolar sorting protein 34) but controlled by the α isoform of class II PI3K (phosphoinositide 3-kinase class II α [PI3KCIIα]). Accordingly, reduced PI3KCIIα expression in ApoE-/- mice (ApoE-/-PI3KCIIα+/-) led to EC dysfunctions associated with increased plaque deposition in the LSS regions. Mechanistically, we revealed that PI3KCIIα inhibits mTORC1 (mammalian target of rapamycin complex 1) activation and that rapamycin treatment in ApoE-/-PI3KCIIα+/- mice specifically rescue autophagy in arterial LSS regions. Finally, we demonstrated that absence of PI3KCIIα led to decreased endothelial primary cilium biogenesis in response to LSS and that ablation of primary cilium mimics PI3KCIIα-decreased expression in EC dysfunction, suggesting that this organelle could be the mechanosensor linking PI3KCIIα and EC homeostasis. CONCLUSIONS Our data reveal that mechanical forces variability within the arterial system determines EC autophagic response and supports a central role of PI3KCIIα/mTORC1 axis to prevent EC dysfunction in LSS regions.
Collapse
Affiliation(s)
- Mouin Nasr
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Institut national de la Santé et de la Recherche (INSERM) 1297, University of Toulouse 3, France (M.N., A.F., A.L., N.M., A.D., R.Z., A.S., A.W., J.V., A.N.-S., D.R., M.L., S.G.)
| | - Alexis Fay
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Institut national de la Santé et de la Recherche (INSERM) 1297, University of Toulouse 3, France (M.N., A.F., A.L., N.M., A.D., R.Z., A.S., A.W., J.V., A.N.-S., D.R., M.L., S.G.)
| | - Adrien Lupieri
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Institut national de la Santé et de la Recherche (INSERM) 1297, University of Toulouse 3, France (M.N., A.F., A.L., N.M., A.D., R.Z., A.S., A.W., J.V., A.N.-S., D.R., M.L., S.G.)
| | - Nicole Malet
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Institut national de la Santé et de la Recherche (INSERM) 1297, University of Toulouse 3, France (M.N., A.F., A.L., N.M., A.D., R.Z., A.S., A.W., J.V., A.N.-S., D.R., M.L., S.G.)
| | - Anne Darmon
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Institut national de la Santé et de la Recherche (INSERM) 1297, University of Toulouse 3, France (M.N., A.F., A.L., N.M., A.D., R.Z., A.S., A.W., J.V., A.N.-S., D.R., M.L., S.G.)
| | - Rana Zahreddine
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Institut national de la Santé et de la Recherche (INSERM) 1297, University of Toulouse 3, France (M.N., A.F., A.L., N.M., A.D., R.Z., A.S., A.W., J.V., A.N.-S., D.R., M.L., S.G.)
| | - Audrey Swiader
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Institut national de la Santé et de la Recherche (INSERM) 1297, University of Toulouse 3, France (M.N., A.F., A.L., N.M., A.D., R.Z., A.S., A.W., J.V., A.N.-S., D.R., M.L., S.G.)
| | - Amandine Wahart
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Institut national de la Santé et de la Recherche (INSERM) 1297, University of Toulouse 3, France (M.N., A.F., A.L., N.M., A.D., R.Z., A.S., A.W., J.V., A.N.-S., D.R., M.L., S.G.)
| | - Julien Viaud
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Institut national de la Santé et de la Recherche (INSERM) 1297, University of Toulouse 3, France (M.N., A.F., A.L., N.M., A.D., R.Z., A.S., A.W., J.V., A.N.-S., D.R., M.L., S.G.)
| | - Anne Nègre-Salvayre
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Institut national de la Santé et de la Recherche (INSERM) 1297, University of Toulouse 3, France (M.N., A.F., A.L., N.M., A.D., R.Z., A.S., A.W., J.V., A.N.-S., D.R., M.L., S.G.)
| | - Emilio Hirsch
- Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Torino, Turin, Italy (E.H.)
| | - Daniel Monteyne
- IBMM-DBM, Department of Molecular Parasitology, University of Brussels, Gosselies, Belgium (D.M., D.P.-M.)
| | - David Perez-Morgà
- IBMM-DBM, Department of Molecular Parasitology, University of Brussels, Gosselies, Belgium (D.M., D.P.-M.)
| | - Nicolas Dupont
- Institut Necker-Enfants Malades (INEM), INSERM U1151-CNRS UMR 8253, Université Paris Descartes-Sorbonne Paris Cité, France (N.D., P.C., E.M.)
| | - Patrice Codogno
- Institut Necker-Enfants Malades (INEM), INSERM U1151-CNRS UMR 8253, Université Paris Descartes-Sorbonne Paris Cité, France (N.D., P.C., E.M.)
| | - Damien Ramel
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Institut national de la Santé et de la Recherche (INSERM) 1297, University of Toulouse 3, France (M.N., A.F., A.L., N.M., A.D., R.Z., A.S., A.W., J.V., A.N.-S., D.R., M.L., S.G.)
| | - Etienne Morel
- Institut Necker-Enfants Malades (INEM), INSERM U1151-CNRS UMR 8253, Université Paris Descartes-Sorbonne Paris Cité, France (N.D., P.C., E.M.)
| | - Muriel Laffargue
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Institut national de la Santé et de la Recherche (INSERM) 1297, University of Toulouse 3, France (M.N., A.F., A.L., N.M., A.D., R.Z., A.S., A.W., J.V., A.N.-S., D.R., M.L., S.G.)
| | - Stephanie Gayral
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Institut national de la Santé et de la Recherche (INSERM) 1297, University of Toulouse 3, France (M.N., A.F., A.L., N.M., A.D., R.Z., A.S., A.W., J.V., A.N.-S., D.R., M.L., S.G.)
| |
Collapse
|
7
|
Gonzalez Galofre ZN, Kilpatrick AM, Marques M, Sá da Bandeira D, Ventura T, Gomez Salazar M, Bouilleau L, Marc Y, Barbosa AB, Rossi F, Beltran M, van de Werken HJG, van IJcken WFJ, Henderson NC, Forbes SJ, Crisan M. Runx1+ vascular smooth muscle cells are essential for hematopoietic stem and progenitor cell development in vivo. Nat Commun 2024; 15:1653. [PMID: 38395882 PMCID: PMC10891074 DOI: 10.1038/s41467-024-44913-z] [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: 02/25/2020] [Accepted: 01/09/2024] [Indexed: 02/25/2024] Open
Abstract
Hematopoietic stem cells (HSCs) produce all essential cellular components of the blood. Stromal cell lines supporting HSCs follow a vascular smooth muscle cell (vSMC) differentiation pathway, suggesting that some hematopoiesis-supporting cells originate from vSMC precursors. These pericyte-like precursors were recently identified in the aorta-gonad-mesonephros (AGM) region; however, their role in the hematopoietic development in vivo remains unknown. Here, we identify a subpopulation of NG2+Runx1+ perivascular cells that display a sclerotome-derived vSMC transcriptomic profile. We show that deleting Runx1 in NG2+ cells impairs the hematopoietic development in vivo and causes transcriptional changes in pericytes/vSMCs, endothelial cells and hematopoietic cells in the murine AGM. Importantly, this deletion leads also to a significant reduction of HSC reconstitution potential in the bone marrow in vivo. This defect is developmental, as NG2+Runx1+ cells were not detected in the adult bone marrow, demonstrating the existence of a specialised pericyte population in the HSC-generating niche, unique to the embryo.
Collapse
Affiliation(s)
- Zaniah N Gonzalez Galofre
- Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, UK
- Centre for Regenerative Medicine/Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, UK
| | - Alastair M Kilpatrick
- Centre for Regenerative Medicine/Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, UK
| | - Madalena Marques
- Centre for Regenerative Medicine/Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, UK
| | - Diana Sá da Bandeira
- Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, UK
- Centre for Regenerative Medicine/Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, UK
| | - Telma Ventura
- Centre for Regenerative Medicine/Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, UK
| | - Mario Gomez Salazar
- Centre for Regenerative Medicine/Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, UK
| | - Léa Bouilleau
- Centre for Regenerative Medicine/Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, UK
| | - Yvan Marc
- Centre for Regenerative Medicine/Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, UK
| | - Ana B Barbosa
- Centre for Regenerative Medicine/Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, UK
| | - Fiona Rossi
- Centre for Regenerative Medicine/Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, UK
| | - Mariana Beltran
- Centre for Inflammation Research/Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, UK
| | - Harmen J G van de Werken
- Cancer Computational Biology Center, Erasmus MC Cancer Institute, University Medical Center, 3000 CA, Rotterdam, The Netherlands
- Department of Urology, Erasmus MC Cancer Institute, University Medical Center, 3000 CA, Rotterdam, The Netherlands
- Department of Immunology, Erasmus MC Cancer Institute, University Medical Center, 3000 CA, Rotterdam, The Netherlands
| | - Wilfred F J van IJcken
- Center for Biomics, Department of Cell Biology, Erasmus MC University Medical Centre, 3015 GE, Rotterdam, The Netherlands
| | - Neil C Henderson
- Centre for Inflammation Research/Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, UK
- MRC Human Genetics Unit, Institute of Genetics and Cancer, The University of Edinburgh, Edinburgh, UK
| | - Stuart J Forbes
- Centre for Regenerative Medicine/Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, UK
| | - Mihaela Crisan
- Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, UK.
- Centre for Regenerative Medicine/Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
8
|
Faleeva M, Ahmad S, Theofilatos K, Lynham S, Watson G, Whitehead M, Marhuenda E, Iskratsch T, Cox S, Shanahan CM. Sox9 Accelerates Vascular Aging by Regulating Extracellular Matrix Composition and Stiffness. Circ Res 2024; 134:307-324. [PMID: 38179698 PMCID: PMC10826924 DOI: 10.1161/circresaha.123.323365] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 12/21/2023] [Accepted: 12/30/2023] [Indexed: 01/06/2024]
Abstract
BACKGROUND Vascular calcification and increased extracellular matrix (ECM) stiffness are hallmarks of vascular aging. Sox9 (SRY-box transcription factor 9) has been implicated in vascular smooth muscle cell (VSMC) osteo/chondrogenic conversion; however, its relationship with aging and calcification has not been studied. METHODS Immunohistochemistry was performed on human aortic samples from young and aged patients. Young and senescent primary human VSMCs were induced to produce ECM, and Sox9 expression was manipulated using adenoviral overexpression and depletion. ECM properties were characterized using atomic force microscopy and proteomics, and VSMC phenotype on hydrogels and the ECM were examined using confocal microscopy. RESULTS In vivo, Sox9 was not spatially associated with vascular calcification but correlated with the senescence marker p16 (cyclin-dependent kinase inhibitor 2A). In vitro Sox9 showed mechanosensitive responses with increased expression and nuclear translocation in senescent cells and on stiff matrices. Sox9 was found to regulate ECM stiffness and organization by orchestrating changes in collagen (Col) expression and reducing VSMC contractility, leading to the formation of an ECM that mirrored that of senescent cells. These ECM changes promoted phenotypic modulation of VSMCs, whereby senescent cells plated on ECM synthesized from cells depleted of Sox9 returned to a proliferative state, while proliferating cells on a matrix produced by Sox9 expressing cells showed reduced proliferation and increased DNA damage, reiterating features of senescent cells. LH3 (procollagen-lysine, 2-oxoglutarate 5-dioxygenase 3) was identified as an Sox9 target and key regulator of ECM stiffness. LH3 is packaged into extracellular vesicles and Sox9 promotes extracellular vesicle secretion, leading to increased LH3 deposition within the ECM. CONCLUSIONS These findings highlight the crucial role of ECM structure and composition in regulating VSMC phenotype. We identify a positive feedback cycle, whereby cellular senescence and increased ECM stiffening promote Sox9 expression, which, in turn, drives further ECM modifications to further accelerate stiffening and senescence.
Collapse
Affiliation(s)
- Maria Faleeva
- British Heart Foundation (BHF) Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences (M.F., S.A., K.T., G.W., M.W., C.M.S.) King’s College London, United Kingdom
| | - Sadia Ahmad
- British Heart Foundation (BHF) Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences (M.F., S.A., K.T., G.W., M.W., C.M.S.) King’s College London, United Kingdom
| | - Konstantinos Theofilatos
- British Heart Foundation (BHF) Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences (M.F., S.A., K.T., G.W., M.W., C.M.S.) King’s College London, United Kingdom
| | - Steven Lynham
- Proteomics Facility, Centre of Excellence for Mass Spectrometry (S.L.) King’s College London, United Kingdom
| | - Gabriel Watson
- British Heart Foundation (BHF) Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences (M.F., S.A., K.T., G.W., M.W., C.M.S.) King’s College London, United Kingdom
| | - Meredith Whitehead
- British Heart Foundation (BHF) Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences (M.F., S.A., K.T., G.W., M.W., C.M.S.) King’s College London, United Kingdom
| | - Emilie Marhuenda
- School of Engineering and Material Science, Queen Mary University of London, United Kingdom (E.M., T.I.)
| | - Thomas Iskratsch
- School of Engineering and Material Science, Queen Mary University of London, United Kingdom (E.M., T.I.)
| | - Susan Cox
- Randall Centre for Cell & Molecular Biophysics, Faculty of Life Sciences & Medicine (S.C.) King’s College London, United Kingdom
| | - Catherine M. Shanahan
- British Heart Foundation (BHF) Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences (M.F., S.A., K.T., G.W., M.W., C.M.S.) King’s College London, United Kingdom
| |
Collapse
|
9
|
Mašek J, Andersson ER. Jagged-mediated development and disease: Mechanistic insights and therapeutic implications for Alagille syndrome. Curr Opin Cell Biol 2024; 86:102302. [PMID: 38194749 DOI: 10.1016/j.ceb.2023.102302] [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: 07/14/2023] [Revised: 11/23/2023] [Accepted: 11/24/2023] [Indexed: 01/11/2024]
Abstract
Notch signaling controls multiple aspects of embryonic development and adult homeostasis. Alagille syndrome is usually caused by a single mutation in the jagged canonical Notch ligand 1 (JAG1), and manifests with liver disease and cardiovascular symptoms that are a direct consequence of JAG1 haploinsufficiency. Recent insights into Jag1/Notch-controlled developmental and homeostatic processes explain how pathology develops in the hepatic and cardiovascular systems and, together with recent elucidation of mechanisms modulating liver regeneration, provide a basis for therapeutic efforts. Importantly, disease presentation can be regulated by genetic modifiers, that may also be therapeutically leverageable. Here, we summarize recent insights into how Jag1 controls processes of relevance to Alagille syndrome, focused on Jag1/Notch functions in hepatic and cardiovascular development and homeostasis.
Collapse
Affiliation(s)
- Jan Mašek
- Department of Cell Biology, Faculty of Science, Charles University, Prague, Czech Republic. https://twitter.com/JanMasekLab
| | - Emma R Andersson
- Dept of Cell and Molecular Biology, Karolinska Institutet, Sweden.
| |
Collapse
|
10
|
Sunshine HL, Cicchetto AC, Kaczor-Urbanowicz KE, Ma F, Pi D, Symons C, Turner M, Shukla V, Christofk HR, Vallim TA, Iruela-Arispe ML. Endothelial Jagged1 levels and distribution are post-transcriptionally controlled by ZFP36 decay proteins. Cell Rep 2024; 43:113627. [PMID: 38157296 PMCID: PMC10884959 DOI: 10.1016/j.celrep.2023.113627] [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: 06/12/2023] [Revised: 11/02/2023] [Accepted: 12/12/2023] [Indexed: 01/03/2024] Open
Abstract
Vascular morphogenesis requires a delicate gradient of Notch signaling controlled, in part, by the distribution of ligands (Dll4 and Jagged1). How Jagged1 (JAG1) expression is compartmentalized in the vascular plexus remains unclear. Here, we show that Jag1 mRNA is a direct target of zinc-finger protein 36 (ZFP36), an RNA-binding protein involved in mRNA decay that we find robustly induced by vascular endothelial growth factor (VEGF). Endothelial cells lacking ZFP36 display high levels of JAG1 and increase angiogenic sprouting in vitro. Furthermore, mice lacking Zfp36 in endothelial cells display mispatterned and increased levels of JAG1 in the developing retinal vascular plexus. Abnormal levels of JAG1 at the sprouting front alters NOTCH1 signaling, increasing the number of tip cells, a phenotype that is rescued by imposing haploinsufficiency of Jag1. Our findings reveal an important feedforward loop whereby VEGF stimulates ZFP36, consequently suppressing Jag1 to enable adequate levels of Notch signaling during sprouting angiogenesis.
Collapse
Affiliation(s)
- Hannah L Sunshine
- Molecular, Cellular, and Integrative Physiology Graduate Program, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Cell and Development Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Andrew C Cicchetto
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Karolina Elżbieta Kaczor-Urbanowicz
- Center for Oral and Head/Neck Oncology Research, UCLA Biosystems & Function, UCLA School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095-1668, USA; UCLA Section of Orthodontics, UCLA School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA; UCLA Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Feiyang Ma
- Department of Cell and Development Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Danielle Pi
- Department of Cell and Development Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Chloe Symons
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Martin Turner
- Immunology Programme, The Babraham Institute, CB22 3AT Cambridge, UK
| | - Vipul Shukla
- Department of Cell and Development Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Center for Human Immunobiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Heather R Christofk
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA 90095-1606, USA
| | - Thomas A Vallim
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA 90095-1606, USA
| | - M Luisa Iruela-Arispe
- Department of Cell and Development Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
11
|
Romay MC, Knutsen RH, Ma F, Mompeón A, Hernandez GE, Salvador J, Mirkov S, Batra A, Sullivan DP, Procissi D, Buchanan S, Kronquist E, Ferrante EA, Muller WA, Walshon J, Steffens A, McCortney K, Horbinski C, Tournier‑Lasserve E, Sonabend AM, Sorond FA, Wang MM, Boehm M, Kozel BA, Iruela-Arispe ML. Age-related loss of Notch3 underlies brain vascular contractility deficiencies, glymphatic dysfunction, and neurodegeneration in mice. J Clin Invest 2024; 134:e166134. [PMID: 38015629 PMCID: PMC10786701 DOI: 10.1172/jci166134] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 11/14/2023] [Indexed: 11/30/2023] Open
Abstract
Vascular aging affects multiple organ systems, including the brain, where it can lead to vascular dementia. However, a concrete understanding of how aging specifically affects the brain vasculature, along with molecular readouts, remains vastly incomplete. Here, we demonstrate that aging is associated with a marked decline in Notch3 signaling in both murine and human brain vessels. To clarify the consequences of Notch3 loss in the brain vasculature, we used single-cell transcriptomics and found that Notch3 inactivation alters regulation of calcium and contractile function and promotes a notable increase in extracellular matrix. These alterations adversely impact vascular reactivity, manifesting as dilation, tortuosity, microaneurysms, and decreased cerebral blood flow, as observed by MRI. Combined, these vascular impairments hinder glymphatic flow and result in buildup of glycosaminoglycans within the brain parenchyma. Remarkably, this phenomenon mirrors a key pathological feature found in brains of patients with CADASIL, a hereditary vascular dementia associated with NOTCH3 missense mutations. Additionally, single-cell RNA sequencing of the neuronal compartment in aging Notch3-null mice unveiled patterns reminiscent of those observed in neurodegenerative diseases. These findings offer direct evidence that age-related NOTCH3 deficiencies trigger a progressive decline in vascular function, subsequently affecting glymphatic flow and culminating in neurodegeneration.
Collapse
Affiliation(s)
- Milagros C. Romay
- Department of Cell and Development Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | | | - Feiyang Ma
- Department of Cell and Development Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Ana Mompeón
- Department of Cell and Development Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Gloria E. Hernandez
- Department of Cell and Development Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, California, USA
| | - Jocelynda Salvador
- Department of Cell and Development Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Snezana Mirkov
- Department of Cell and Development Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Ayush Batra
- Department of Pathology
- Department of Neurology, and
| | | | - Daniele Procissi
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois, USA
| | - Samuel Buchanan
- Department of Cell and Development Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Elise Kronquist
- National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, USA
| | - Elisa A. Ferrante
- National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, USA
- Laboratory of Cardiovascular Regenerative Medicine, NIH, Bethesda, Maryland, USA
| | | | - Jordain Walshon
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Alicia Steffens
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Kathleen McCortney
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Craig Horbinski
- Department of Pathology
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Elisabeth Tournier‑Lasserve
- Inserm NeuroDiderot, Université Paris Cité, Paris, France
- Service de Génétique Neurovasculaire, Assistance Publique–Hôpitaux de Paris, Hôpital Saint-Louis, Paris, France
| | - Adam M. Sonabend
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Chicago, Illinois, USA
| | | | - Michael M. Wang
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
- VA Ann Arbor Healthcare System, Ann Arbor, Michigan, USA
| | - Manfred Boehm
- National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, USA
- Laboratory of Cardiovascular Regenerative Medicine, NIH, Bethesda, Maryland, USA
| | - Beth A. Kozel
- National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, USA
| | - M. Luisa Iruela-Arispe
- Department of Cell and Development Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
12
|
Eley L, Richardson RV, Alqahtani A, Chaudhry B, Henderson DJ. eNOS plays essential roles in the developing heart and aorta linked to disruption of Notch signalling. Dis Model Mech 2024; 17:dmm050265. [PMID: 38111957 PMCID: PMC10846539 DOI: 10.1242/dmm.050265] [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: 04/24/2023] [Accepted: 12/12/2023] [Indexed: 12/20/2023] Open
Abstract
eNOS (NOS3) is the enzyme that generates nitric oxide, a signalling molecule and regulator of vascular tone. Loss of eNOS function is associated with increased susceptibility to atherosclerosis, hypertension, thrombosis and stroke. Aortopathy and cardiac hypertrophy have also been found in eNOS null mice, but their aetiology is unclear. We evaluated eNOS nulls before and around birth for cardiac defects, revealing severe abnormalities in the ventricular myocardium and pharyngeal arch arteries. Moreover, in the aortic arch, there were fewer baroreceptors, which sense changes in blood pressure. Adult eNOS null survivors showed evidence of cardiac hypertrophy, aortopathy and cartilaginous metaplasia in the periductal region of the aortic arch. Notch1 and neuregulin were dysregulated in the forming pharyngeal arch arteries and ventricles, suggesting that these pathways may be relevant to the defects observed. Dysregulation of eNOS leads to embryonic and perinatal death, suggesting mutations in eNOS are candidates for causing congenital heart defects in humans. Surviving eNOS mutants have a deficiency of baroreceptors that likely contributes to high blood pressure and may have relevance to human patients who suffer from hypertension associated with aortic arch abnormalities.
Collapse
Affiliation(s)
- Lorraine Eley
- Bioscience Institute, Newcastle University, Centre for Life, Central Parkway, Newcastle upon Tyne, NE1 3BZ, UK
| | - Rachel V. Richardson
- Bioscience Institute, Newcastle University, Centre for Life, Central Parkway, Newcastle upon Tyne, NE1 3BZ, UK
| | - Ahlam Alqahtani
- Bioscience Institute, Newcastle University, Centre for Life, Central Parkway, Newcastle upon Tyne, NE1 3BZ, UK
| | - Bill Chaudhry
- Bioscience Institute, Newcastle University, Centre for Life, Central Parkway, Newcastle upon Tyne, NE1 3BZ, UK
| | - Deborah J. Henderson
- Bioscience Institute, Newcastle University, Centre for Life, Central Parkway, Newcastle upon Tyne, NE1 3BZ, UK
| |
Collapse
|
13
|
Alonso-Herranz L, Albarrán-Juárez J, Bentzon JF. Mechanisms of fibrous cap formation in atherosclerosis. Front Cardiovasc Med 2023; 10:1254114. [PMID: 37671141 PMCID: PMC10475556 DOI: 10.3389/fcvm.2023.1254114] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 08/09/2023] [Indexed: 09/07/2023] Open
Abstract
The fibrous cap is formed by smooth muscle cells that accumulate beneath the plaque endothelium. Cap rupture is the main cause of coronary thrombosis, leading to infarction and sudden cardiac death. Therefore, the qualities of the cap are primary determinants of the clinical outcome of coronary and carotid atherosclerosis. In this mini-review, we discuss current knowledge about the formation of the fibrous cap, including cell recruitment, clonal expansion, and central molecular signaling pathways. We also examine the differences between mouse and human fibrous caps and explore the impact of anti-atherosclerotic therapies on the state of the fibrous cap. We propose that the cap should be understood as a neo-media to substitute for the original media that becomes separated from the surface endothelium during atherogenesis and that embryonic pathways involved in the development of the arteria media contribute to cap formation.
Collapse
Affiliation(s)
- Laura Alonso-Herranz
- Atherosclerosis Research Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Julián Albarrán-Juárez
- Atherosclerosis Research Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Jacob Fog Bentzon
- Atherosclerosis Research Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Steno Diabetes Center Aarhus and Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| |
Collapse
|
14
|
Murtada SI, Kawamura Y, Cavinato C, Wang M, Ramachandra AB, Spronck B, Li DS, Tellides G, Humphrey JD. Biomechanical and transcriptional evidence that smooth muscle cell death drives an osteochondrogenic phenotype and severe proximal vascular disease in progeria. Biomech Model Mechanobiol 2023; 22:1333-1347. [PMID: 37149823 PMCID: PMC10544720 DOI: 10.1007/s10237-023-01722-5] [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: 09/09/2022] [Accepted: 04/11/2023] [Indexed: 05/08/2023]
Abstract
Hutchinson-Gilford Progeria Syndrome results in rapid aging and severe cardiovascular sequelae that accelerate near end-of-life. We found a progressive disease process in proximal elastic arteries that was less evident in distal muscular arteries. Changes in aortic structure and function were then associated with changes in transcriptomics assessed via both bulk and single cell RNA sequencing, which suggested a novel sequence of progressive aortic disease: adverse extracellular matrix remodeling followed by mechanical stress-induced smooth muscle cell death, leading a subset of remnant smooth muscle cells to an osteochondrogenic phenotype that results in an accumulation of proteoglycans that thickens the aortic wall and increases pulse wave velocity, with late calcification exacerbating these effects. Increased central artery pulse wave velocity is known to drive left ventricular diastolic dysfunction, the primary diagnosis in progeria children. It appears that mechanical stresses above ~ 80 kPa initiate this progressive aortic disease process, explaining why elastic lamellar structures that are organized early in development under low wall stresses appear to be nearly normal whereas other medial constituents worsen progressively in adulthood. Mitigating early mechanical stress-driven smooth muscle cell loss/phenotypic modulation promises to have important cardiovascular implications in progeria patients.
Collapse
Affiliation(s)
- Sae-Il Murtada
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Yuki Kawamura
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT, USA
| | - Cristina Cavinato
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Molly Wang
- Department of Surgery, Yale School of Medicine, New Haven, CT, USA
| | | | - Bart Spronck
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
- Department of Biomedical Engineering, Maastricht University, Maastricht, Netherlands
| | - David S Li
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - George Tellides
- Department of Surgery, Yale School of Medicine, New Haven, CT, USA
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA
| | - Jay D Humphrey
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA.
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
15
|
Wen T, Duan Y, Gao D, Zhang X, Zhang X, Liang L, Yang Z, Zhang P, Zhang J, Sun J, Feng Y, Zheng Q, Han H, Yan X. miR-342-5p promotes vascular smooth muscle cell phenotypic transition through a negative-feedback regulation of Notch signaling via targeting FOXO3. Life Sci 2023:121828. [PMID: 37270171 DOI: 10.1016/j.lfs.2023.121828] [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: 04/12/2023] [Revised: 05/28/2023] [Accepted: 05/30/2023] [Indexed: 06/05/2023]
Abstract
AIM Under various pathological conditions such as cancer, vascular smooth muscle cells (vSMCs) transit their contractile phenotype into phenotype(s) characterized by proliferation and secretion, a process called vSMC phenotypic transition (vSMC-PT). Notch signaling regulates vSMC development and vSMC-PT. This study aims to elucidate how the Notch signal is regulated. MAIN METHODS Gene-modified mice with a SM22α-CreERT2 transgene were generated to activate/block Notch signaling in vSMCs. Primary vSMCs and MOVAS cells were cultured in vitro. RNA-seq, qRT-PCR and Western blotting were used to evaluated gene expression level. EdU incorporation, Transwell and collagen gel contraction assays were conducted to determine the proliferation, migration and contraction, respectively. KEY FINDINGS Notch activation upregulated, while Notch blockade downregulated, miR-342-5p and its host gene Evl in vSMCs. However, miR-342-5p overexpression promoted vSMC-PT as shown by altered gene expression profile, increased migration and proliferation, and decreased contraction, while miR-342-5p blockade exhibited the opposite effects. Moreover, miR-342-5p overexpression significantly suppressed Notch signaling, and Notch activation partially abolished miR-342-5p-induced vSMC-PT. Mechanically, miR-342-5p directly targeted FOXO3, and FOXO3 overexpression rescued miR-342-5p-induced Notch repression and vSMC-PT. In a simulated tumor microenvironment, miR-342-5p was upregulated by tumor cell-derived conditional medium (TCM), and miR-342-5p blockade abrogated TCM-induced vSMC-PT. Meanwhile, conditional medium from miR-342-5p-overexpressing vSMCs significantly enhanced tumor cell proliferation, while miR-342-5p blockade had the opposite effects. Consistently, in a co-inoculation tumor model, miR-342-5p blockade in vSMCs significantly delayed tumor growth. SIGNIFICANCE miR-342-5p promotes vSMC-PT through a negative-feedback regulation of Notch signaling via downregulating FOXO3, which could be a potential target for cancer therapy.
Collapse
Affiliation(s)
- Ting Wen
- Faculty of Life Sciences, Northwest University, Xi'an 710069, China; State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an 710032, China
| | - Yanyan Duan
- Faculty of Life Sciences, Northwest University, Xi'an 710069, China; State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an 710032, China
| | - Dan Gao
- Faculty of Life Sciences, Northwest University, Xi'an 710069, China; State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an 710032, China
| | - Xinxin Zhang
- College of Pulmonary and Critical Care Medicine, The 8th Medical Centre of Chinese PLA General Hospital, Beijing 100091, China
| | - Xiaoyan Zhang
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an 710032, China
| | - Liang Liang
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an 710032, China
| | - Ziyan Yang
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an 710032, China
| | - Peiran Zhang
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an 710032, China
| | - Jiayulin Zhang
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an 710032, China
| | - Jiaxing Sun
- Department of Ophthalmology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Yixuan Feng
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an 710032, China
| | - Qijun Zheng
- Department of Cardiovascular Surgery, Shenzhen People's Hospital, Shenzhen 518020, China.
| | - Hua Han
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an 710032, China; Department of Gastroenterology, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038, China.
| | - Xianchun Yan
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
16
|
Medzikovic L, Aryan L, Ruffenach G, Li M, Savalli N, Sun W, Sarji S, Hong J, Sharma S, Olcese R, Fishbein G, Eghbali M. Myocardial fibrosis and calcification are attenuated by microRNA-129-5p targeting Asporin and Sox9 in cardiac fibroblasts. JCI Insight 2023; 8:e168655. [PMID: 37154157 PMCID: PMC10243800 DOI: 10.1172/jci.insight.168655] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 03/15/2023] [Indexed: 05/10/2023] Open
Abstract
Myocardial fibrosis and calcification associate with adverse outcomes in nonischemic heart failure. Cardiac fibroblasts (CF) transition into myofibroblasts (MF) and osteogenic fibroblasts (OF) to promote myocardial fibrosis and calcification. However, common upstream mechanisms regulating both CF-to-MF transition and CF-to-OF transition remain unknown. microRNAs are promising targets to modulate CF plasticity. Our bioinformatics revealed downregulation of miR-129-5p and upregulation of its targets small leucine-rich proteoglycan Asporin (ASPN) and transcription factor SOX9 as common in mouse and human heart failure (HF). We experimentally confirmed decreased miR-129-5p and enhanced SOX9 and ASPN expression in CF in human hearts with myocardial fibrosis and calcification. miR-129-5p repressed both CF-to-MF and CF-to-OF transition in primary CF, as did knockdown of SOX9 and ASPN. Sox9 and Aspn are direct targets of miR-129-5p that inhibit downstream β-catenin expression. Chronic Angiotensin II infusion downregulated miR-129-5p in CF in WT and TCF21-lineage CF reporter mice, and it was restored by miR-129-5p mimic. Importantly, miR-129-5p mimic not only attenuated progression of myocardial fibrosis, calcification marker expression, and SOX9 and ASPN expression in CF but also restored diastolic and systolic function. Together, we demonstrate miR-129-5p/ASPN and miR-129-5p/SOX9 as potentially novel dysregulated axes in CF-to-MF and CF-to-OF transition in myocardial fibrosis and calcification and the therapeutic relevance of miR-129-5p.
Collapse
Affiliation(s)
| | - Laila Aryan
- Department of Anesthesiology & Perioperative Medicine
| | | | - Min Li
- Department of Anesthesiology & Perioperative Medicine
| | | | - Wasila Sun
- Department of Anesthesiology & Perioperative Medicine
| | - Shervin Sarji
- Department of Anesthesiology & Perioperative Medicine
| | - Jason Hong
- Department of Anesthesiology & Perioperative Medicine
- Division of Pulmonary & Critical Care Medicine
| | - Salil Sharma
- Department of Anesthesiology & Perioperative Medicine
| | - Riccardo Olcese
- Department of Anesthesiology & Perioperative Medicine
- Department of Physiology, and
| | - Gregory Fishbein
- Department of Physiology, and
- Department of Pathology & Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | | |
Collapse
|
17
|
Tani S, Okada H, Onodera S, Chijimatsu R, Seki M, Suzuki Y, Xin X, Rowe DW, Saito T, Tanaka S, Chung UI, Ohba S, Hojo H. Stem cell-based modeling and single-cell multiomics reveal gene-regulatory mechanisms underlying human skeletal development. Cell Rep 2023; 42:112276. [PMID: 36965484 DOI: 10.1016/j.celrep.2023.112276] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 01/19/2023] [Accepted: 03/02/2023] [Indexed: 03/27/2023] Open
Abstract
Although the skeleton is essential for locomotion, endocrine functions, and hematopoiesis, the molecular mechanisms of human skeletal development remain to be elucidated. Here, we introduce an integrative method to model human skeletal development by combining in vitro sclerotome induction from human pluripotent stem cells and in vivo endochondral bone formation by implanting the sclerotome beneath the renal capsules of immunodeficient mice. Histological and scRNA-seq analyses reveal that the induced bones recapitulate endochondral ossification and are composed of human skeletal cells and mouse circulatory cells. The skeletal cell types and their trajectories are similar to those of human embryos. Single-cell multiome analysis reveals dynamic changes in chromatin accessibility associated with multiple transcription factors constituting cell-type-specific gene-regulatory networks (GRNs). We further identify ZEB2, which may regulate the GRNs in human osteogenesis. Collectively, these results identify components of GRNs in human skeletal development and provide a valuable model for its investigation.
Collapse
Affiliation(s)
- Shoichiro Tani
- Laboratory of Clinical Biotechnology, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan; Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan.
| | - Hiroyuki Okada
- Laboratory of Clinical Biotechnology, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan; Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Shoko Onodera
- Department of Biochemistry, Tokyo Dental College, Tokyo 101-0061, Japan
| | - Ryota Chijimatsu
- Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan; Center for Comprehensive Genomic Medicine, Okayama University Hospital, Okayama 700-8558, Japan
| | - Masahide Seki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba 277-8562, Japan
| | - Yutaka Suzuki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba 277-8562, Japan
| | - Xiaonan Xin
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - David W Rowe
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Taku Saito
- Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Sakae Tanaka
- Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Ung-Il Chung
- Laboratory of Clinical Biotechnology, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan; Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Tokyo 113-8655, Japan
| | - Shinsuke Ohba
- Laboratory of Clinical Biotechnology, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan; Department of Cell Biology, Institute of Biomedical Sciences, Nagasaki University, Nagasaki 852-8588, Japan; Department of Oral Anatomy and Developmental Biology, Graduate School of Dentistry, Osaka University, Osaka 565-0871, Japan.
| | - Hironori Hojo
- Laboratory of Clinical Biotechnology, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan; Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Tokyo 113-8655, Japan.
| |
Collapse
|
18
|
Chatzeli L, Bordeu I, Han S, Bisetto S, Waheed Z, Koo BK, Alcolea MP, Simons BD. A cellular hierarchy of Notch and Kras signaling controls cell fate specification in the developing mouse salivary gland. Dev Cell 2023; 58:94-109.e6. [PMID: 36693323 PMCID: PMC7614884 DOI: 10.1016/j.devcel.2022.12.009] [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: 04/25/2022] [Revised: 10/14/2022] [Accepted: 12/20/2022] [Indexed: 01/24/2023]
Abstract
The development of the mouse salivary gland involves a tip-driven process of branching morphogenesis that takes place in concert with differentiation into acinar, myoepithelial, and ductal (basal and luminal) sub-lineages. By combining clonal lineage tracing with a three-dimensional (3D) reconstruction of the branched epithelial network and single-cell RNA-seq analysis, we show that in tips, a heterogeneous population of renewing progenitors transition from a Krt14+ multipotent state to unipotent states via two transcriptionally distinct bipotent states, one restricted to the Krt14+ basal and myoepithelial lineage and the other to the Krt8+ acinar and luminal lineage. Using genetic perturbations, we show how the differential expression of Notch signaling correlates with spatial segregation, exits from multipotency, and promotes the Krt8+ lineage, whereas Kras activation promotes proacinar fate. These findings provide a mechanistic basis for how positional cues within growing tips regulate the process of lineage segregation and ductal patterning.
Collapse
Affiliation(s)
- Lemonia Chatzeli
- Wellcome Trust, Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, UK; Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB2 0AW, UK.
| | - Ignacio Bordeu
- Wellcome Trust, Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, UK; Department of Applied Mathematics and Theoretical Physics, Centre for Mathematical Sciences, University of Cambridge, Cambridge CB3 0WA, UK; Departamento de Física, Facultad de Ciencias Físicas y Matemáticas, Universidad de Chile, 837.0415 Santiago, Chile
| | - Seungmin Han
- Wellcome Trust, Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, UK; Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB2 0AW, UK
| | - Sara Bisetto
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB2 0AW, UK
| | - Zahra Waheed
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB2 0AW, UK
| | - Bon-Kyoung Koo
- Center for Genome Engineering, Institute for Basic Science, Expo-ro 55, Yuseong-gu, Daejeon 34126, Republic of Korea
| | - Maria P Alcolea
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB2 0AW, UK; Department of Oncology, The Hutchison Building, Box 197 Cambridge Biomedical Campus, University of Cambridge, Cambridge, CB2 0XZ, UK
| | - Benjamin D Simons
- Wellcome Trust, Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, UK; Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB2 0AW, UK; Department of Applied Mathematics and Theoretical Physics, Centre for Mathematical Sciences, University of Cambridge, Cambridge CB3 0WA, UK.
| |
Collapse
|
19
|
Murtada SI, Kawamura Y, Cavinato C, Wang M, Ramachandra AB, Spronck B, Tellides G, Humphrey JD. Smooth Muscle Cell Death Drives an Osteochondrogenic Phenotype and Severe Proximal Vascular Disease in Progeria. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.10.523266. [PMID: 36711514 PMCID: PMC9882088 DOI: 10.1101/2023.01.10.523266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Hutchinson-Gilford Progeria Syndrome results in rapid aging and severe cardiovascular sequelae that accelerate near end of life. We associate progressive deterioration of arterial structure and function with single cell transcriptional changes, which reveals a rapid disease process in proximal elastic arteries that largely spares distal muscular arteries. These data suggest a novel sequence of progressive vascular disease in progeria: initial extracellular matrix remodeling followed by mechanical stress-induced smooth muscle cell death in proximal arteries, leading a subset of remnant smooth muscle cells to an osteochondrogenic phenotypic modulation that results in an accumulation of proteoglycans that thickens the wall and increases pulse wave velocity, with late calcification exacerbating these effects. Increased pulse wave velocity drives left ventricular diastolic dysfunction, the primary diagnosis in progeria children. Mitigating smooth muscle cell loss / phenotypic modulation promises to have important cardiovascular implications in progeria patients.
Collapse
Affiliation(s)
- Sae-Il Murtada
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Yuki Kawamura
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT, USA
| | - Cristina Cavinato
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Mo Wang
- Department of Surgery, Yale School of Medicine, New Haven, CT, USA
| | | | - Bart Spronck
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
- Department of Biomedical Engineering, Maastricht University, Maastricht, Netherlands
| | - George Tellides
- Department of Surgery, Yale School of Medicine, New Haven, CT, USA
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA
| | - Jay D. Humphrey
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
20
|
Hashimoto D, Fujimoto K, Kim SW, Lee YS, Nakata M, Suzuki K, Wada Y, Asamura S, Yamada G. Emerging structural and pathological analyses on the erectile organ, corpus cavernous containing sinusoids. Reprod Med Biol 2023; 22:e12539. [PMID: 37663955 PMCID: PMC10472535 DOI: 10.1002/rmb2.12539] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/08/2023] [Accepted: 08/18/2023] [Indexed: 09/05/2023] Open
Abstract
Background The corpus cavernosum (CC) containing sinusoids plays fundamental roles for erection. Analysis of pathological changes in the erectile system is studied by recent experimental systems. Various in vitro models utilizing genital mesenchymal-derived cells and explant culture systems are summarized. Methods 3D reconstruction of section images of murine CC was created. Ectopic chondrogenesis in aged mouse CC was shown by a gene expression study revealing the prominent expression of Sox9. Various experimental strategies utilizing mesenchyme-derived primary cells and tissue explants are introduced. Main Findings Possible roles of Sox9 in chondrogenesis and its regulation by several signals are suggested. The unique character of genital mesenchyme is shown by various analyses of external genitalia (ExG) derived cells and explant cultures. Such strategies are also applied to the analysis of erectile contraction/relaxation responses to many signals and aging process. Conclusion Erectile dysfunction (ED) is one of the essential topics for the modern aged society. More comprehensive studies are necessary to reveal the nature of the erectile system by combining multiple cell culture strategies.
Collapse
Affiliation(s)
- Daiki Hashimoto
- Department of Developmental Genetics, Institute of Advanced MedicineWakayama Medical UniversityWakayamaJapan
- Department of Physiology and Regenerative Medicine, Faculty of MedicineKindai UniversityOsakaJapan
| | - Kota Fujimoto
- Department of Developmental Genetics, Institute of Advanced MedicineWakayama Medical UniversityWakayamaJapan
- Department of Plastic and Reconstructive Surgery, Graduate School of MedicineWakayama Medical UniversityWakayamaJapan
| | - Sang Woon Kim
- Department of Urology, Urological Science InstituteYonsei University College of MedicineSeoulSouth Korea
| | - Yong Seung Lee
- Department of Urology, Urological Science InstituteYonsei University College of MedicineSeoulSouth Korea
| | - Masanori Nakata
- Department of Physiology, Faculty of MedicineWakayama Medical UniversityWakayamaJapan
| | - Kentaro Suzuki
- Faculty of Life and Environmental SciencesUniversity of YamanashiYamanashiJapan
| | - Yoshitaka Wada
- Department of Plastic and Reconstructive Surgery, Graduate School of MedicineWakayama Medical UniversityWakayamaJapan
| | - Shinichi Asamura
- Department of Plastic and Reconstructive Surgery, Graduate School of MedicineWakayama Medical UniversityWakayamaJapan
| | - Gen Yamada
- Department of Developmental Genetics, Institute of Advanced MedicineWakayama Medical UniversityWakayamaJapan
- Department of Plastic and Reconstructive Surgery, Graduate School of MedicineWakayama Medical UniversityWakayamaJapan
| |
Collapse
|
21
|
Yu Q, Liu JX, Zheng X, Yan X, Zhao P, Yin C, Li W, Song Z. Sox9 mediates autophagy-dependent vascular smooth muscle cell phenotypic modulation and transplant arteriosclerosis. iScience 2022; 25:105161. [PMID: 36204267 PMCID: PMC9531173 DOI: 10.1016/j.isci.2022.105161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 08/04/2022] [Accepted: 09/15/2022] [Indexed: 11/16/2022] Open
Affiliation(s)
- Qihong Yu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan 430030, China
| | - Jin-Xin Liu
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xichuan Zheng
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xueke Yan
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Peng Zhao
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Chuanzheng Yin
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Wei Li
- Departments of Gerontology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Corresponding author
| | - Zifang Song
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Corresponding author
| |
Collapse
|
22
|
Breikaa RM, Denman K, Ueyama Y, McCallinhart PE, Khan AQ, Agarwal G, Trask AJ, Garg V, Lilly B. Loss of Jagged1 in mature endothelial cells causes vascular dysfunction with alterations in smooth muscle phenotypes. Vascul Pharmacol 2022; 145:107087. [PMID: 35792302 DOI: 10.1016/j.vph.2022.107087] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/27/2022] [Accepted: 06/28/2022] [Indexed: 11/24/2022]
Abstract
BACKGROUND Notch signaling is an evolutionarily conserved pathway that functions via direct cell-cell contact. The Notch ligand Jagged1 (Jag1) has been extensively studied in vascular development, particularly for its role in smooth muscle cell maturation. Endothelial cell-expressed Jag1 is essential for blood vessel formation by signaling to nascent vascular smooth muscle cells and promoting their differentiation. Given the established importance of Jag1 in endothelial cell/smooth muscle crosstalk during development, we sought to determine the extent of this communication in the adult vasculature for blood vessel function and homeostasis. METHODS We conditionally deleted Jag1 in endothelial cells of adult mice and examined the phenotypic consequences on smooth muscle cells of the vasculature. RESULTS Our results show that genetic loss of Jag1 in endothelial cells has a significant impact on Notch signaling and vascular smooth muscle function in mature blood vessels. Endothelial cell-specific deletion of Jag1 causes a concomitant loss of JAG1 and NOTCH3 expression in vascular smooth muscle cells, resulting in a transition to a less differentiated state. Aortic vascular smooth muscle cells isolated from the endothelial cell-specific Jag1 deficient mice retain an altered phenotype in culture with fixed changes in gene expression and reduced Notch signaling. Utilizing comparative RNA-sequence analysis, we found that Jag1 deficiency preferentially affects extracellular matrix and adhesion protein gene expression. Vasoreactivity studies revealed a reduced contractile response and impaired agonist-induced relaxation in endothelial cell Jag1-deficient aortas compared to controls. CONCLUSIONS These data are the first to demonstrate that Jag1 in adult endothelial cells is required for the regulation and homeostasis of smooth muscle cell function in arterial vessels partially through the autoregulation of Notch signaling and cell matrix/adhesion components in smooth muscle cells.
Collapse
Affiliation(s)
- Randa M Breikaa
- Center for Cardiovascular Research and The Heart Center, Nationwide Children's Hospital, The Ohio State University, Columbus, OH, USA; Molecular, Cellular and Developmental Biology Program, The Ohio State University, Columbus, OH, USA
| | - Kimberly Denman
- Department of Mechanical and Aerospace Engineering, The Ohio State University, Columbus, OH, USA
| | - Yukie Ueyama
- Center for Cardiovascular Research and The Heart Center, Nationwide Children's Hospital, The Ohio State University, Columbus, OH, USA
| | - Patricia E McCallinhart
- Center for Cardiovascular Research and The Heart Center, Nationwide Children's Hospital, The Ohio State University, Columbus, OH, USA
| | - Aiman Q Khan
- Center for Cardiovascular Research and The Heart Center, Nationwide Children's Hospital, The Ohio State University, Columbus, OH, USA
| | - Gunjan Agarwal
- Department of Mechanical and Aerospace Engineering, The Ohio State University, Columbus, OH, USA
| | - Aaron J Trask
- Center for Cardiovascular Research and The Heart Center, Nationwide Children's Hospital, The Ohio State University, Columbus, OH, USA; Department of Pediatrics, The Ohio State University, Columbus, OH, USA
| | - Vidu Garg
- Center for Cardiovascular Research and The Heart Center, Nationwide Children's Hospital, The Ohio State University, Columbus, OH, USA; Department of Pediatrics, The Ohio State University, Columbus, OH, USA
| | - Brenda Lilly
- Center for Cardiovascular Research and The Heart Center, Nationwide Children's Hospital, The Ohio State University, Columbus, OH, USA; Department of Pediatrics, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
23
|
Notch signaling pathway: architecture, disease, and therapeutics. Signal Transduct Target Ther 2022; 7:95. [PMID: 35332121 PMCID: PMC8948217 DOI: 10.1038/s41392-022-00934-y] [Citation(s) in RCA: 518] [Impact Index Per Article: 172.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 02/16/2022] [Accepted: 02/16/2022] [Indexed: 02/07/2023] Open
Abstract
The NOTCH gene was identified approximately 110 years ago. Classical studies have revealed that NOTCH signaling is an evolutionarily conserved pathway. NOTCH receptors undergo three cleavages and translocate into the nucleus to regulate the transcription of target genes. NOTCH signaling deeply participates in the development and homeostasis of multiple tissues and organs, the aberration of which results in cancerous and noncancerous diseases. However, recent studies indicate that the outcomes of NOTCH signaling are changeable and highly dependent on context. In terms of cancers, NOTCH signaling can both promote and inhibit tumor development in various types of cancer. The overall performance of NOTCH-targeted therapies in clinical trials has failed to meet expectations. Additionally, NOTCH mutation has been proposed as a predictive biomarker for immune checkpoint blockade therapy in many cancers. Collectively, the NOTCH pathway needs to be integrally assessed with new perspectives to inspire discoveries and applications. In this review, we focus on both classical and the latest findings related to NOTCH signaling to illustrate the history, architecture, regulatory mechanisms, contributions to physiological development, related diseases, and therapeutic applications of the NOTCH pathway. The contributions of NOTCH signaling to the tumor immune microenvironment and cancer immunotherapy are also highlighted. We hope this review will help not only beginners but also experts to systematically and thoroughly understand the NOTCH signaling pathway.
Collapse
|
24
|
He X, Li X, Han Y, Chen G, Xu T, Cai D, Sun Y, Wang S, Lai Y, Teng Z, Huang S, Liao W, Liao Y, Bin J, Xiu J. CircRNA Chordc1 protects mice from abdominal aortic aneurysm by contributing to the phenotype and growth of vascular smooth muscle cells. MOLECULAR THERAPY. NUCLEIC ACIDS 2022; 27:81-98. [PMID: 34938608 PMCID: PMC8649900 DOI: 10.1016/j.omtn.2021.11.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 09/23/2021] [Accepted: 11/04/2021] [Indexed: 12/15/2022]
Abstract
Circular RNAs (circRNAs) have important potential in modulating vascular smooth muscle cell (VSMC) activity, but their roles in abdominal aortic aneurysm (AAA) are unknown. We performed in situ hybridization and immunohistochemistry and determined that circChordc1 (cysteine and histidine-rich domain containing 1) was markedly downregulated in aneurysm tissue compared with normal arteries. A gene gain and loss strategy was used to confirm that circChordc1 transformed VSMCs into a contracted phenotype and improved their growth, which significantly suppressed aneurysm formation and reduced the risk of rupture in mouse models of angiotensin (Ang) II- and CaCl2-induced AAA. RNA pull-down, immunoprecipitation, and immunoblotting indicated that circChordc1 facilitated the VSMC phenotype and growth determination by binding to vimentin and ANXA2 (annexin A2), which not only increased vimentin phosphorylation to promote its degradation but also promoted the interaction between ANXA2 and glycogen synthase kinase 3 beta (GSK3β) to induce the nuclear entry of β-catenin. Thus, our present study revealed that circChordc1 optimized the VSMC phenotype and improved their growth by inducing vimentin degradation and increasing the activity of the GSK3β/β-catenin pathway, thereby extenuating vascular wall remodeling and reversing pathological aneurysm progression.
Collapse
Affiliation(s)
- Xiang He
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, 510005 Guangzhou, China.,Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou, China
| | - Xinzhong Li
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, 510005 Guangzhou, China.,Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou, China
| | - Yuan Han
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, 510005 Guangzhou, China.,Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou, China
| | - Guojun Chen
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, 510005 Guangzhou, China
| | - Tong Xu
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, 510005 Guangzhou, China
| | - Donghua Cai
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, 510005 Guangzhou, China
| | - Yili Sun
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, 510005 Guangzhou, China
| | - Shifei Wang
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, 510005 Guangzhou, China
| | - Yanxian Lai
- Department of Cardiology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, 1 Panfu Road, Guangzhou 510180, China
| | - Zhonghua Teng
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, 510005 Guangzhou, China
| | - Senlin Huang
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, 510005 Guangzhou, China
| | - Wangjun Liao
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yulin Liao
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, 510005 Guangzhou, China
| | - Jianping Bin
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, 510005 Guangzhou, China.,Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou, China
| | - Jiancheng Xiu
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, 510005 Guangzhou, China.,Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou, China
| |
Collapse
|
25
|
Favre J, Vessieres E, Guihot AL, Proux C, Grimaud L, Rivron J, Garcia MC, Réthoré L, Zahreddine R, Davezac M, Fébrissy C, Adlanmerini M, Loufrani L, Procaccio V, Foidart JM, Flouriot G, Lenfant F, Fontaine C, Arnal JF, Henrion D. Membrane estrogen receptor alpha (ERα) participates in flow-mediated dilation in a ligand-independent manner. eLife 2021; 10:68695. [PMID: 34842136 PMCID: PMC8676342 DOI: 10.7554/elife.68695] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 11/26/2021] [Indexed: 12/13/2022] Open
Abstract
Estrogen receptor alpha (ERα) activation by estrogens prevents atheroma through its nuclear action, whereas plasma membrane-located ERα accelerates endothelial healing. The genetic deficiency of ERα was associated with a reduction in flow-mediated dilation (FMD) in one man. Here, we evaluated ex vivo the role of ERα on FMD of resistance arteries. FMD, but not agonist (acetylcholine, insulin)-mediated dilation, was reduced in male and female mice lacking ERα (Esr1-/- mice) compared to wild-type mice and was not dependent on the presence of estrogens. In C451A-ERα mice lacking membrane ERα, not in mice lacking AF2-dependent nuclear ERα actions, FMD was reduced, and restored by antioxidant treatments. Compared to wild-type mice, isolated perfused kidneys of C451A-ERα mice revealed a decreased flow-mediated nitrate production and an increased H2O2 production. Thus, endothelial membrane ERα promotes NO bioavailability through inhibition of oxidative stress and thereby participates in FMD in a ligand-independent manner.
Collapse
Affiliation(s)
- Julie Favre
- Angers University, MITOVASC, CNRS UMR 6015, INSERM U1083, Angers, France
| | - Emilie Vessieres
- Angers University, MITOVASC, CNRS UMR 6015, INSERM U1083, Angers, France.,CARFI facility, Angers University, Angers, France
| | - Anne-Laure Guihot
- Angers University, MITOVASC, CNRS UMR 6015, INSERM U1083, Angers, France.,CARFI facility, Angers University, Angers, France
| | - Coralyne Proux
- Angers University, MITOVASC, CNRS UMR 6015, INSERM U1083, Angers, France.,CARFI facility, Angers University, Angers, France
| | - Linda Grimaud
- Angers University, MITOVASC, CNRS UMR 6015, INSERM U1083, Angers, France
| | - Jordan Rivron
- Angers University, MITOVASC, CNRS UMR 6015, INSERM U1083, Angers, France.,CARFI facility, Angers University, Angers, France
| | - Manuela Cl Garcia
- Angers University, MITOVASC, CNRS UMR 6015, INSERM U1083, Angers, France.,CARFI facility, Angers University, Angers, France
| | - Léa Réthoré
- Angers University, MITOVASC, CNRS UMR 6015, INSERM U1083, Angers, France
| | - Rana Zahreddine
- INSERM U1297, Paul Sabatier University (Toulouse III) , University Hospital (UHC) of Toulouse, Toulouse, France
| | - Morgane Davezac
- INSERM U1297, Paul Sabatier University (Toulouse III) , University Hospital (UHC) of Toulouse, Toulouse, France
| | - Chanaelle Fébrissy
- INSERM U1297, Paul Sabatier University (Toulouse III) , University Hospital (UHC) of Toulouse, Toulouse, France
| | - Marine Adlanmerini
- INSERM U1297, Paul Sabatier University (Toulouse III) , University Hospital (UHC) of Toulouse, Toulouse, France
| | - Laurent Loufrani
- Angers University, MITOVASC, CNRS UMR 6015, INSERM U1083, Angers, France.,University Hospital (CHU) of Angers, Angers, France
| | - Vincent Procaccio
- Angers University, MITOVASC, CNRS UMR 6015, INSERM U1083, Angers, France.,University Hospital (CHU) of Angers, Angers, France
| | - Jean-Michel Foidart
- Groupe Interdisciplinaire de Génoprotéomique Appliquée, Université de Liège, Liège, Belgium
| | - Gilles Flouriot
- INSERM U1085, IRSET (Institut de Recherche en Santé, Environnement et Travail), University of Rennes, Rennes, France
| | - Françoise Lenfant
- INSERM U1297, Paul Sabatier University (Toulouse III) , University Hospital (UHC) of Toulouse, Toulouse, France
| | - Coralie Fontaine
- INSERM U1297, Paul Sabatier University (Toulouse III) , University Hospital (UHC) of Toulouse, Toulouse, France
| | - Jean-François Arnal
- INSERM U1297, Paul Sabatier University (Toulouse III) , University Hospital (UHC) of Toulouse, Toulouse, France
| | - Daniel Henrion
- Angers University, MITOVASC, CNRS UMR 6015, INSERM U1083, Angers, France.,CARFI facility, Angers University, Angers, France.,University Hospital (CHU) of Angers, Angers, France
| |
Collapse
|
26
|
Herrero-Cervera A, Espinós-Estévez C, Martín-Vañó S, Taberner-Cortés A, Aguilar-Ballester M, Vinué Á, Piqueras L, Martínez-Hervás S, González-Navarro H. Dissecting Abdominal Aortic Aneurysm Is Aggravated by Genetic Inactivation of LIGHT (TNFSF14). Biomedicines 2021; 9:biomedicines9111518. [PMID: 34829747 PMCID: PMC8615201 DOI: 10.3390/biomedicines9111518] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/18/2021] [Accepted: 10/19/2021] [Indexed: 11/16/2022] Open
Abstract
Abdominal aortic aneurysm (AAA), is a complex disorder characterized by vascular vessel wall remodeling. LIGHT (TNFSF14) is a proinflammatory cytokine associated with vascular disease. In the present study, the impact of genetic inactivation of Light was investigated in dissecting AAA induced by angiotensin II (AngII) in the Apolipoprotein E-deficient (Apoe−/−) mice. Studies in aortic human (ah) vascular smooth muscle cells (VSMC) to study potential translation to human pathology were also performed. AngII-treated Apoe−/−Light−/− mice displayed increased abdominal aorta maximum diameter and AAA severity compared with Apoe−/− mice. Notably, reduced smooth muscle α-actin+ area and Acta2 and Col1a1 gene expression were observed in AAA from Apoe−/−Light−/− mice, suggesting a loss of VSMC contractile phenotype compared with controls. Decreased Opn and augmented Sox9 expression, which are associated with detrimental and non-contractile osteochondrogenic VSMC phenotypes, were also seen in AngII-treated Apoe−/−Light−/− mouse AAA. Consistent with a role of LIGHT preserving VSMC contractile characteristics, LIGHT-treatment of ahVSMCs diminished the expression of SOX9 and of the pluripotency marker CKIT. These effects were partly mediated through lymphotoxin β receptor (LTβR) as the silencing of its gene ablated LIGHT effects on ahVSMCs. These studies suggest a protective role of LIGHT through mechanisms that prevent VSMC trans-differentiation in an LTβR-dependent manner.
Collapse
Affiliation(s)
- Andrea Herrero-Cervera
- INCLIVA, Institute of Health Research, 46010 Valencia, Spain; (A.H.-C.); (S.M.-V.); (A.T.-C.); (M.A.-B.); (Á.V.); (L.P.); (S.M.-H.)
| | | | - Susana Martín-Vañó
- INCLIVA, Institute of Health Research, 46010 Valencia, Spain; (A.H.-C.); (S.M.-V.); (A.T.-C.); (M.A.-B.); (Á.V.); (L.P.); (S.M.-H.)
| | - Alida Taberner-Cortés
- INCLIVA, Institute of Health Research, 46010 Valencia, Spain; (A.H.-C.); (S.M.-V.); (A.T.-C.); (M.A.-B.); (Á.V.); (L.P.); (S.M.-H.)
| | - María Aguilar-Ballester
- INCLIVA, Institute of Health Research, 46010 Valencia, Spain; (A.H.-C.); (S.M.-V.); (A.T.-C.); (M.A.-B.); (Á.V.); (L.P.); (S.M.-H.)
| | - Ángela Vinué
- INCLIVA, Institute of Health Research, 46010 Valencia, Spain; (A.H.-C.); (S.M.-V.); (A.T.-C.); (M.A.-B.); (Á.V.); (L.P.); (S.M.-H.)
| | - Laura Piqueras
- INCLIVA, Institute of Health Research, 46010 Valencia, Spain; (A.H.-C.); (S.M.-V.); (A.T.-C.); (M.A.-B.); (Á.V.); (L.P.); (S.M.-H.)
- Department of Pharmacology, University of Valencia, 46010 Valencia, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
| | - Sergio Martínez-Hervás
- INCLIVA, Institute of Health Research, 46010 Valencia, Spain; (A.H.-C.); (S.M.-V.); (A.T.-C.); (M.A.-B.); (Á.V.); (L.P.); (S.M.-H.)
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
- Endocrinology and Nutrition Service, Clinic Hospital of Valencia, 46010 Valencia, Spain
- Department of Medicine, University of Valencia, 46010 Valencia, Spain
| | - Herminia González-Navarro
- INCLIVA, Institute of Health Research, 46010 Valencia, Spain; (A.H.-C.); (S.M.-V.); (A.T.-C.); (M.A.-B.); (Á.V.); (L.P.); (S.M.-H.)
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
- Biochemistry and Molecular Biology Department, University of Valencia, 46010 Valencia, Spain
- Correspondence: ; Tel.: +34-96-386-44-03; Fax: +34-96-398-78-60
| |
Collapse
|
27
|
Martos-Rodríguez CJ, Albarrán-Juárez J, Morales-Cano D, Caballero A, MacGrogan D, de la Pompa JL, Carramolino L, Bentzon JF. Fibrous Caps in Atherosclerosis Form by Notch-Dependent Mechanisms Common to Arterial Media Development. Arterioscler Thromb Vasc Biol 2021; 41:e427-e439. [PMID: 34261328 DOI: 10.1161/atvbaha.120.315627] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Objective Atheromatous fibrous caps are produced by smooth muscle cells (SMCs) that are recruited to the subendothelial space. We tested whether the recruitment mechanisms are the same as in embryonic artery development, which relies prominently on Notch signaling to form the subendothelial medial SMC layers. Approach and Results Notch elements were expressed in regions of fibrous cap in human and mouse plaques. To assess the causal role of Notch signaling in cap formation, we studied atherosclerosis in mice where the Notch pathway was inactivated in SMCs by conditional knockout of the essential effector transcription factor RBPJ (recombination signal-binding protein for immunoglobulin kappa J region). The recruitment of cap SMCs was significantly reduced without major effects on plaque size. Lineage tracing revealed the accumulation of SMC-derived plaque cells in the cap region was unaltered but that Notch-defective cells failed to re-acquire the SMC phenotype in the cap. Conversely, to analyze whether the loss of Notch signaling is required for SMC-derived cells to accumulate in atherogenesis, we studied atherosclerosis in mice with constitutive activation of Notch signaling in SMCs achieved by conditional expression of the Notch intracellular domain. Forced Notch signaling inhibited the ability of medial SMCs to contribute to plaque cells, including both cap SMCs and osteochondrogenic cells, and significantly reduced atherosclerosis development. Conclusions Sequential loss and gain of Notch signaling is needed to build the cap SMC population. The shared mechanisms with embryonic arterial media assembly suggest that the cap forms as a neo-media that restores the connection between endothelium and subendothelial SMCs, transiently disrupted in early atherogenesis.
Collapse
MESH Headings
- Actins/genetics
- Actins/metabolism
- Animals
- Arteries/metabolism
- Arteries/pathology
- Atherosclerosis/genetics
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Cell Lineage
- Cells, Cultured
- Disease Progression
- Fibrosis
- Humans
- Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics
- Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism
- Jagged-1 Protein/genetics
- Jagged-1 Protein/metabolism
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Phenotype
- Plaque, Atherosclerotic
- Rats
- Receptors, Notch/genetics
- Receptors, Notch/metabolism
- Signal Transduction
- Tunica Media/metabolism
- Tunica Media/pathology
- Mice
Collapse
Affiliation(s)
- Carlos J Martos-Rodríguez
- Experimental Pathology of Atherosclerosis Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain (C.J.M.-R., D.M.-C., A.C., L.C., J.F.B.)
| | - Julián Albarrán-Juárez
- Heart Diseases, Department of Clinical Medicine (J.A.-J., A.C., J.F.B.), Aarhus University, Denmark
| | - Daniel Morales-Cano
- Experimental Pathology of Atherosclerosis Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain (C.J.M.-R., D.M.-C., A.C., L.C., J.F.B.)
| | - Ainoa Caballero
- Experimental Pathology of Atherosclerosis Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain (C.J.M.-R., D.M.-C., A.C., L.C., J.F.B.)
- Heart Diseases, Department of Clinical Medicine (J.A.-J., A.C., J.F.B.), Aarhus University, Denmark
| | - Donal MacGrogan
- Intercellular Signalling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (D.M., J.L.d.l.P.)
- Ciber de Enfermedades Cardiovasculares, Madrid, Spain (D.M., J.L.d.l.P.)
| | - José Luis de la Pompa
- Intercellular Signalling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (D.M., J.L.d.l.P.)
- Ciber de Enfermedades Cardiovasculares, Madrid, Spain (D.M., J.L.d.l.P.)
| | - Laura Carramolino
- Experimental Pathology of Atherosclerosis Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain (C.J.M.-R., D.M.-C., A.C., L.C., J.F.B.)
| | - Jacob F Bentzon
- Experimental Pathology of Atherosclerosis Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain (C.J.M.-R., D.M.-C., A.C., L.C., J.F.B.)
- Heart Diseases, Department of Clinical Medicine (J.A.-J., A.C., J.F.B.), Aarhus University, Denmark
- Steno Diabetes Center Aarhus, Department of Clinical Medicine (J.F.B.), Aarhus University, Denmark
| |
Collapse
|
28
|
Hashimoto D, Kajimoto M, Ueda Y, Hyuga T, Fujimoto K, Inoue S, Suzuki K, Kataoka T, Kimura K, Yamada G. 3D reconstruction and histopathological analyses on murine corporal body. Reprod Med Biol 2021; 20:199-207. [PMID: 33850453 PMCID: PMC8022099 DOI: 10.1002/rmb2.12369] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/15/2021] [Accepted: 01/17/2021] [Indexed: 12/15/2022] Open
Abstract
PURPOSE Erectile dysfunction (ED) is one of the increasing diseases with aging society. The basis of ED derived from local penile abnormality is poorly understood because of the complex three-dimensional (3D) distribution of sinusoids in corpus cavernosum (CC). Understanding the 3D histological structure of penis is thus necessary. Analyses on the status of regulatory signals for such abnormality are also performed. METHODS To analyze the 3D structure of sinusoid, 3D reconstruction from serial sections of murine CC were performed. Histological analyses between young (2 months old) and aged (14 months old) CC were performed. As for chondrogenic signaling status of aged CC, SOX9 and RBPJK staining was examined. RESULTS Sinusoids prominently developed in the outer regions of CC adjacent to tunica albuginea. Aged CC samples contained ectopic chondrocytes in such regions. Associating with the appearance of chondrocytes, the expression of SOX9, chondrogenic regulator, was upregulated. The expression of RBPJK, one of the Notch signal regulators, was downregulated in the aged CC. CONCLUSIONS Prominent sinusoids distribute in the outer region of CC which may possess important roles for erection. A possibility of ectopic chondrogenesis induced by alteration of SOX9/Notch signaling with aging is indicated.
Collapse
Affiliation(s)
- Daiki Hashimoto
- Department of Developmental GeneticsInstitute of Advanced MedicineWakayama Medical UniversityWakayamaJapan
| | - Mizuki Kajimoto
- Department of Developmental GeneticsInstitute of Advanced MedicineWakayama Medical UniversityWakayamaJapan
| | - Yuko Ueda
- Department of UrologyWakayama Medical UniversityWakayamaJapan
| | - Taiju Hyuga
- Department of Developmental GeneticsInstitute of Advanced MedicineWakayama Medical UniversityWakayamaJapan
| | - Kota Fujimoto
- Department of Developmental GeneticsInstitute of Advanced MedicineWakayama Medical UniversityWakayamaJapan
| | - Saaya Inoue
- Department of Developmental GeneticsInstitute of Advanced MedicineWakayama Medical UniversityWakayamaJapan
| | - Kentaro Suzuki
- Department of Developmental GeneticsInstitute of Advanced MedicineWakayama Medical UniversityWakayamaJapan
| | - Tomoya Kataoka
- Department of Clinical PharmaceuticsGraduate School of Medical SciencesNagoya City UniversityNagoyaJapan
| | - Kazunori Kimura
- Department of Clinical PharmaceuticsGraduate School of Medical SciencesNagoya City UniversityNagoyaJapan
- Department of Hospital PharmacyGraduate School of Pharmaceutical SciencesNagoya City UniversityNagoyaJapan
| | - Gen Yamada
- Department of Developmental GeneticsInstitute of Advanced MedicineWakayama Medical UniversityWakayamaJapan
| |
Collapse
|
29
|
MicroRNA-34a: the bad guy in age-related vascular diseases. Cell Mol Life Sci 2021; 78:7355-7378. [PMID: 34698884 PMCID: PMC8629897 DOI: 10.1007/s00018-021-03979-4] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/08/2021] [Accepted: 10/12/2021] [Indexed: 12/12/2022]
Abstract
The age-related vasculature alteration is the prominent risk factor for vascular diseases (VD), namely, atherosclerosis, abdominal aortic aneurysm, vascular calcification (VC) and pulmonary arterial hypertension (PAH). The chronic sterile low-grade inflammation state, alias inflammaging, characterizes elderly people and participates in VD development. MicroRNA34-a (miR-34a) is emerging as an important mediator of inflammaging and VD. miR-34a increases with aging in vessels and induces senescence and the acquisition of the senescence-associated secretory phenotype (SASP) in vascular smooth muscle (VSMCs) and endothelial (ECs) cells. Similarly, other VD risk factors, including dyslipidemia, hyperglycemia and hypertension, modify miR-34a expression to promote vascular senescence and inflammation. miR-34a upregulation causes endothelial dysfunction by affecting ECs nitric oxide bioavailability, adhesion molecules expression and inflammatory cells recruitment. miR-34a-induced senescence facilitates VSMCs osteoblastic switch and VC development in hyperphosphatemia conditions. Conversely, atherogenic and hypoxic stimuli downregulate miR-34a levels and promote VSMCs proliferation and migration during atherosclerosis and PAH. MiR34a genetic ablation or miR-34a inhibition by anti-miR-34a molecules in different experimental models of VD reduce vascular inflammation, senescence and apoptosis through sirtuin 1 Notch1, and B-cell lymphoma 2 modulation. Notably, pleiotropic drugs, like statins, liraglutide and metformin, affect miR-34a expression. Finally, human studies report that miR-34a levels associate to atherosclerosis and diabetes and correlate with inflammatory factors during aging. Herein, we comprehensively review the current knowledge about miR-34a-dependent molecular and cellular mechanisms activated by VD risk factors and highlight the diagnostic and therapeutic potential of modulating its expression in order to reduce inflammaging and VD burn and extend healthy lifespan.
Collapse
|
30
|
Wang L, Chennupati R, Jin YJ, Li R, Wang S, Günther S, Offermanns S. YAP/TAZ Are Required to Suppress Osteogenic Differentiation of Vascular Smooth Muscle Cells. iScience 2020; 23:101860. [PMID: 33319178 PMCID: PMC7726335 DOI: 10.1016/j.isci.2020.101860] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 09/10/2020] [Accepted: 11/20/2020] [Indexed: 12/22/2022] Open
Abstract
Vascular smooth muscle cells (VSMCs) represent the prevailing cell type of arterial vessels and are essential for blood vessel structure and homeostasis. They have substantial potential for phenotypic plasticity when exposed to various stimuli in their local microenvironment. How VSMCs maintain their differentiated contractile phenotype is still poorly understood. Here we demonstrate that the Hippo pathway effectors YAP and TAZ play a critical role in maintaining the differentiated contractile phenotype of VSMCs. In the absence of YAP/TAZ, VSMCs lose their differentiated phenotype and undergo osteogenic differentiation, which results in vascular calcification. Osteogenic transdifferentiation was accompanied by the upregulation of Wnt target genes. The absence of YAP/TAZ in VSMCs led to Disheveled 3 (DVL3) nuclear translocation and upregulation of osteogenesis-associated genes independent of canonical Wnt/β-catenin signaling activation. Our data indicate that cytoplasmic YAP/TAZ interact with DVL3 to avoid its nuclear translocation and osteogenic differentiation, thereby maintaining the differentiated phenotype of VSMCs. YAP/TAZ play an important role in maintaining vascular SMCs contractile phenotype Loss of YAP/TAZ in vSMCs leads to reduced expression of smooth muscle marker genes Loss of YAP/TAZ in vSMCs results in reduced artery contractility Deficiency of YAP/TAZ in vSMCs leads to osteogenic transdifferentiation
Collapse
Affiliation(s)
- Lei Wang
- Max Planck Institute for Heart and Lung Research, Department of Pharmacology, Bad Nauheim 61231, Germany
| | - Ramesh Chennupati
- Max Planck Institute for Heart and Lung Research, Department of Pharmacology, Bad Nauheim 61231, Germany
| | - Young-June Jin
- Max Planck Institute for Heart and Lung Research, Department of Pharmacology, Bad Nauheim 61231, Germany
| | - Rui Li
- Max Planck Institute for Heart and Lung Research, Department of Pharmacology, Bad Nauheim 61231, Germany
| | - ShengPeng Wang
- Max Planck Institute for Heart and Lung Research, Department of Pharmacology, Bad Nauheim 61231, Germany.,Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Yanta District, Xi'an 710061, China
| | - Stefan Günther
- Bioinformatics and Deep Sequencing Platform, Max Planck Institute for Heart and Lung Research, Bad Nauheim 61231, Germany
| | - Stefan Offermanns
- Max Planck Institute for Heart and Lung Research, Department of Pharmacology, Bad Nauheim 61231, Germany.,Center for Molecular Medicine, Medical Faculty, Goethe University, Frankfurt am Main 60590, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Frankfurt Rhine-Main, 13347 Berlin, Germany
| |
Collapse
|
31
|
Zhang M, Yuan SZ, Sun H, Sun L, Zhou D, Yan J. miR-199b-5p promoted chondrogenic differentiation of C3H10T1/2 cells by regulating JAG1. J Tissue Eng Regen Med 2020; 14:1618-1629. [PMID: 32870569 DOI: 10.1002/term.3122] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 07/17/2020] [Accepted: 08/26/2020] [Indexed: 12/13/2022]
Abstract
Mesenchymal stem cells (MSCs) are considered a promising candidate for use in cell-based therapy for cartilage repair. To promote understanding of the molecular control of chondrogenesis differentiation in MSCs, we compared the changes in microRNAs during in vitro chondrogenesis process of human bone-marrow mesenchymal stem cells (hBMSCs). MiR-199b-5p was up-regulated significantly during this process. The aim of the study was to investigate the effects of miR-199b-5p on chondrogenic differentiation of C3H10T1/2 MSC cells and explore the underlying mechanisms. MiR-199b-5p mimics or inhibitor were transfected into C3H10T1/2 cells, respectively, and then, the effects of miR-199b-5p on chondrogenic differentiation of C3H10T1/2 cells were detected. The results indicated that miR-199b-5p overexpression inhibited the growth of C3H10T1/2 cells but promoted transforming growth factor-β3 (TGF-β3)-induced C3H10T1/2 cells of chondrogenic differentiation, as supported by enhancing the gene and protein expression of chondrocyte specific markers of SOX9, aggrecan, and collagen type II (Col2a1). In contrast, inhibiting miR-199b-5p notably promoted the proliferation of C3H10T1/2 cells but decreased chondrogenic differentiation. Furthermore, mechanism studies revealed that JAG1 was a direct target of miR-199b-5p by dual luciferase reporter assays. While silencing of JAG1 by isRNA resulted an increase of chondrogenic differentiation. Further, JAG1 knockdown was demonstrated to block the effect of miR-199b-5p inhibition. In conclusion, the present study revealed for the first time that miR-199b-5p was the positive regulators to modulate chondrogenic differentiation of C3H10T1/2 cells by targeting JAG1. These findings may provide a novel insight on miRNA-mediated MSC therapy for cartilage related disorders.
Collapse
Affiliation(s)
- Miao Zhang
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Shu Zheng Yuan
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Haimei Sun
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Lei Sun
- Musculosketetal Tissue Bank, Beijing Jishuitan Hospital, Beijing, China
| | - Deshan Zhou
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Jihong Yan
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| |
Collapse
|
32
|
Extracellular vesicle signalling in atherosclerosis. Cell Signal 2020; 75:109751. [PMID: 32860954 PMCID: PMC7534042 DOI: 10.1016/j.cellsig.2020.109751] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 08/20/2020] [Accepted: 08/21/2020] [Indexed: 12/12/2022]
Abstract
Atherosclerosis is a major cardiovascular disease and in 2016, the World Health Organisation (WHO) estimated 17.5 million global deaths, corresponding to 31% of all global deaths, were driven by inflammation and deposition of lipids into the arterial wall. This leads to the development of plaques which narrow the vessel lumen, particularly in the coronary and carotid arteries. Atherosclerotic plaques can become unstable and rupture, leading to myocardial infarction or stroke. Extracellular vesicles (EVs) are a heterogeneous population of vesicles secreted from cells with a wide range of biological functions. EVs participate in cell-cell communication and signalling via transport of cargo including enzymes, DNA, RNA and microRNA in both physiological and patholophysiological settings. EVs are present in atherosclerotic plaques and have been implicated in cellular signalling processes in atherosclerosis development, including immune responses, inflammation, cell proliferation and migration, cell death and vascular remodeling during progression of the disease. In this review, we summarise the current knowledge regarding EV signalling in atherosclerosis progression and the potential of utilising EV signatures as biomarkers of disease.
Collapse
|
33
|
SM22α + vascular mural cells are essential for vessel stability in tumors and undergo phenotype transition regulated by Notch signaling. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:124. [PMID: 32616053 PMCID: PMC7331127 DOI: 10.1186/s13046-020-01630-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 06/25/2020] [Indexed: 11/17/2022]
Abstract
Background Malformation of blood vessels represents a hallmark of cancers, but the role and regulation of vascular mural cells (vMCs), including vascular smooth muscle cells (vSMCs) and pericytes, in tumors has not been fully understood. SM22α has been identified as a marker of vSMCs. This study aims at elucidating the function and regulation of SM22α+ mural cells (SM22-MCs) in tumor stroma. Methods Gene-modified mice with a SM22α-CreERT2 transgene were employed to deplete SM22-MCs or activate/block Notch signaling in these cells. vSMCs from mouse dorsal aorta (vSMCs-DA) were cultured in vitro. RNA-seq was used to compare gene expression profiles. qRT-PCR and western blotting were used to determine gene expression level. Immunofluorescence was used to observe morphological alterations in tumors. Results SM22-MCs are essential for stabilizing tumor vasculature. Notch signaling was downregulated in tumor-derived SM22-MCs and vSMCs-DA treated with cancer cell-derived conditioned medium. Notch activation in SM22-MCs normalized tumor vasculature and repressed tumor growth. On the other hand, Notch disruption aggravated abnormal tumor vasculature and promoted growth and metastasis. Gene expression profiling of vSMCs-DA showed that Notch activation enhances their contractile phenotype and suppresses their secretory phenotype, further attenuating the invasion and proliferation of tumor cells. In contrast, Notch blockade in vSMCs-DA mitigated their contractile phenotype while strengthened the secretory phenotype. Conclusion SM22-MCs facilitate vessel stability in tumors, and they gain a secretory phenotype and promote tumor malignancy in the absence of Notch signaling.
Collapse
|
34
|
Yianni V, Sharpe PT. Epigenetic mechanisms driving lineage commitment in mesenchymal stem cells. Bone 2020; 134:115309. [PMID: 32145460 DOI: 10.1016/j.bone.2020.115309] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 03/02/2020] [Accepted: 03/02/2020] [Indexed: 12/15/2022]
Abstract
The increasing application of approaches that allow tracing of individual cells over time, together with transcriptomic and epigenomic analyses is changing the way resident stromal stem cells (mesenchymal stem cells) are viewed. Rather than being a defined, homogeneous cell population as described following in vitro expansion, in vivo, these cells are highly programmed according to their resident tissue location. This programming is evidenced by different epigenetic landscapes and gene transcription signatures in cells before any in vitro expansion. This has potentially profound implications for the heterotypic use of these cells in therapeutic tissue engineering applications.
Collapse
Affiliation(s)
- Val Yianni
- Centre for Craniofacial & Regenerative Biology, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, United Kingdom of Great Britain and Northern Ireland
| | - Paul T Sharpe
- Centre for Craniofacial & Regenerative Biology, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, United Kingdom of Great Britain and Northern Ireland.
| |
Collapse
|
35
|
Induced osteogenic differentiation of human smooth muscle cells as a model of vascular calcification. Sci Rep 2020; 10:5951. [PMID: 32249802 PMCID: PMC7136202 DOI: 10.1038/s41598-020-62568-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 03/14/2020] [Indexed: 01/09/2023] Open
Abstract
Vascular calcification is a severe pathological event in the manifestation of atherosclerosis. Pathogenic triggers mediating osteogenic differentiation of arterial smooth muscle cells (SMC) in humans remain insufficiently understood and are to a large extent investigated in animal models or cells derived thereof. Here, we describe an in vitro model based on SMC derived from healthy and diseased humans that allows to comprehensively investigate vascular calcification mechanisms. Comparing the impact of the commonly used SMC culture media VascuLife, DMEM, and M199, cells were characterised by immunofluorescence, flow cytometry, qPCR, and regarding their contractility and proliferative capacity. Irrespective of the arterial origin, the clinical background and the expansion medium used, all cells expressed typical molecular SMC marker while contractility varied between donors. Interestingly, the ability to induce an osteogenic differentiation strongly depended on the culture medium, with only SMC cultured in DMEM depositing calcified matrix upon osteogenic stimulation, which correlated with increased alkaline phosphatase activity, increased inorganic phosphate level and upregulation of osteogenic gene markers. Our optimized model is suitable for donor-oriented as well as broader screening of potential pathogenic mediators triggering vascular calcification. Translational studies aiming to identify and to evaluate therapeutic targets in a personalized fashion would be feasible.
Collapse
|
36
|
van Engeland NCA, Suarez Rodriguez F, Rivero-Müller A, Ristori T, Duran CL, Stassen OMJA, Antfolk D, Driessen RCH, Ruohonen S, Ruohonen ST, Nuutinen S, Savontaus E, Loerakker S, Bayless KJ, Sjöqvist M, Bouten CVC, Eriksson JE, Sahlgren CM. Vimentin regulates Notch signaling strength and arterial remodeling in response to hemodynamic stress. Sci Rep 2019; 9:12415. [PMID: 31455807 PMCID: PMC6712036 DOI: 10.1038/s41598-019-48218-w] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 07/30/2019] [Indexed: 01/12/2023] Open
Abstract
The intermediate filament (IF) cytoskeleton has been proposed to regulate morphogenic processes by integrating the cell fate signaling machinery with mechanical cues. Signaling between endothelial cells (ECs) and vascular smooth muscle cells (VSMCs) through the Notch pathway regulates arterial remodeling in response to changes in blood flow. Here we show that the IF-protein vimentin regulates Notch signaling strength and arterial remodeling in response to hemodynamic forces. Vimentin is important for Notch transactivation by ECs and vimentin knockout mice (VimKO) display disrupted VSMC differentiation and adverse remodeling in aortic explants and in vivo. Shear stress increases Jagged1 levels and Notch activation in a vimentin-dependent manner. Shear stress induces phosphorylation of vimentin at serine 38 and phosphorylated vimentin interacts with Jagged1 and increases Notch activation potential. Reduced Jagged1-Notch transactivation strength disrupts lateral signal induction through the arterial wall leading to adverse remodeling. Taken together we demonstrate that vimentin forms a central part of a mechanochemical transduction pathway that regulates multilayer communication and structural homeostasis of the arterial wall.
Collapse
Affiliation(s)
- Nicole C A van Engeland
- Åbo Akademi University, Faculty of Science and Engineering, Biosciences, Turku, Finland.,Eindhoven University of Technology, Department of Biomedical Engineering, 5600, MB, Eindhoven, The Netherlands
| | - Freddy Suarez Rodriguez
- Åbo Akademi University, Faculty of Science and Engineering, Biosciences, Turku, Finland.,Turku Bioscience, Åbo Akademi University and University of Turku, Turku, Finland
| | - Adolfo Rivero-Müller
- Åbo Akademi University, Faculty of Science and Engineering, Biosciences, Turku, Finland.,Department of Biochemistry and Molecular Biology, Medical University of Lublin, Lublin, Poland
| | - Tommaso Ristori
- Åbo Akademi University, Faculty of Science and Engineering, Biosciences, Turku, Finland.,Eindhoven University of Technology, Department of Biomedical Engineering, 5600, MB, Eindhoven, The Netherlands.,Institute of Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Camille L Duran
- Department of Molecular & Cellular Medicine, Texas A&M University Health Science Center, College Station, TX, 77843, Texas, USA
| | - Oscar M J A Stassen
- Åbo Akademi University, Faculty of Science and Engineering, Biosciences, Turku, Finland.,Turku Bioscience, Åbo Akademi University and University of Turku, Turku, Finland
| | - Daniel Antfolk
- Åbo Akademi University, Faculty of Science and Engineering, Biosciences, Turku, Finland.,Turku Bioscience, Åbo Akademi University and University of Turku, Turku, Finland
| | - Rob C H Driessen
- Eindhoven University of Technology, Department of Biomedical Engineering, 5600, MB, Eindhoven, The Netherlands
| | - Saku Ruohonen
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Turku, Finland
| | - Suvi T Ruohonen
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Turku, Finland.,Turku Center for Disease Modelling, University of Turku, Turku, Finland
| | - Salla Nuutinen
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Turku, Finland
| | - Eriika Savontaus
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Turku, Finland.,Turku Center for Disease Modelling, University of Turku, Turku, Finland
| | - Sandra Loerakker
- Eindhoven University of Technology, Department of Biomedical Engineering, 5600, MB, Eindhoven, The Netherlands.,Institute of Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Kayla J Bayless
- Department of Molecular & Cellular Medicine, Texas A&M University Health Science Center, College Station, TX, 77843, Texas, USA
| | - Marika Sjöqvist
- Åbo Akademi University, Faculty of Science and Engineering, Biosciences, Turku, Finland.,Turku Bioscience, Åbo Akademi University and University of Turku, Turku, Finland
| | - Carlijn V C Bouten
- Eindhoven University of Technology, Department of Biomedical Engineering, 5600, MB, Eindhoven, The Netherlands.,Institute of Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - John E Eriksson
- Turku Bioscience, Åbo Akademi University and University of Turku, Turku, Finland
| | - Cecilia M Sahlgren
- Åbo Akademi University, Faculty of Science and Engineering, Biosciences, Turku, Finland. .,Eindhoven University of Technology, Department of Biomedical Engineering, 5600, MB, Eindhoven, The Netherlands. .,Turku Bioscience, Åbo Akademi University and University of Turku, Turku, Finland. .,Institute of Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands.
| |
Collapse
|
37
|
Ziyad S, Riordan JD, Cavanaugh AM, Su T, Hernandez GE, Hilfenhaus G, Morselli M, Huynh K, Wang K, Chen JN, Dupuy AJ, Iruela-Arispe ML. A Forward Genetic Screen Targeting the Endothelium Reveals a Regulatory Role for the Lipid Kinase Pi4ka in Myelo- and Erythropoiesis. Cell Rep 2019; 22:1211-1224. [PMID: 29386109 PMCID: PMC5828030 DOI: 10.1016/j.celrep.2018.01.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 11/05/2017] [Accepted: 01/05/2018] [Indexed: 11/19/2022] Open
Abstract
Given its role as the source of definitive hematopoietic cells, we sought to determine whether mutations initiated in the hemogenic endothelium would yield hematopoietic abnormalities or malignancies. Here, we find that endothelium-specific transposon mutagenesis in mice promotes hematopoietic pathologies that are both myeloid and lymphoid in nature. Frequently mutated genes included previously recognized cancer drivers and additional candidates, such as Pi4ka, a lipid kinase whose mutation was found to promote myeloid and erythroid dysfunction. Subsequent validation experiments showed that targeted inactivation of the Pi4ka catalytic domain or reduction in mRNA expression inhibited myeloid and erythroid cell differentiation in vitro and promoted anemia in vivo through a mechanism involving deregulation of AKT, MAPK, SRC, and JAK-STAT signaling. Finally, we provide evidence linking PI4KAP2, previously considered a pseudogene, to human myeloid and erythroid leukemia.
Collapse
Affiliation(s)
- Safiyyah Ziyad
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jesse D Riordan
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA 52242, USA
| | - Ann M Cavanaugh
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Trent Su
- Institute for Quantitative and Computational Biology and Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Gloria E Hernandez
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Georg Hilfenhaus
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Marco Morselli
- Institute for Quantitative and Computational Biology and Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA 90095, USA; Institute of Genomics and Proteomics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Kristine Huynh
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Kevin Wang
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jau-Nian Chen
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Adam J Dupuy
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA 52242, USA
| | - M Luisa Iruela-Arispe
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
38
|
Janssen JN, Batschkus S, Schimmel S, Bode C, Schminke B, Miosge N. The Influence of TGF-β3, EGF, and BGN on SOX9 and RUNX2 Expression in Human Chondrogenic Progenitor Cells. J Histochem Cytochem 2018; 67:117-127. [PMID: 30431382 DOI: 10.1369/0022155418811645] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Osteoarthritis (OA) is the most common chronic joint disease and leads to the degradation of the extracellular matrix by an imbalance between anabolic and catabolic processes. TGF-β3 (transforming growth factor beta-3) and epidermal growth factor (EGF) influence the osteochondrogenic potential of chondrocytes. In this study, we compared the expression of mediators and receptors in the TGF-β3 and EGF pathways, as well as biglycan (BGN), in healthy and diseased chondrocytes. Furthermore, we used chondrogenic progenitor cells (CPCs) for in vitro stimulation and knockdown experiments to elucidate the effects of TGF-β3 and EGF on the chondrogenic potential. Our results demonstrate that the expression of TGF-beta receptor type-1 (TGFBRI) and epidermal growth factor receptor (EGFR) is altered in diseased chondrocytes as well as in CPCs. Moreover, TGF-β3 and EGF stimulation influenced the expression levels of BGN, SRY (sex determining region Y)-box 9 (SOX9), and Runt-related transcription factor 2 (RUNX2) in CPCs. Therefore, changes in TGFBRI and EGFR expression likely contribute to the degenerative and regenerative effects seen in late stages of OA.
Collapse
Affiliation(s)
| | | | - Stefan Schimmel
- Tissue Regeneration Work Group, Department of Prosthodontics
| | - Christa Bode
- Tissue Regeneration Work Group, Department of Prosthodontics
| | - Boris Schminke
- Department of Oral and Maxillofacial Surgery, University Medical Center, Göttingen, Germany
| | - Nicolai Miosge
- Tissue Regeneration Work Group, Department of Prosthodontics
| |
Collapse
|
39
|
Yianni V, Sharpe PT. Molecular Programming of Perivascular Stem Cell Precursors. Stem Cells 2018; 36:1890-1904. [PMID: 30068019 DOI: 10.1002/stem.2895] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 07/09/2018] [Accepted: 07/15/2018] [Indexed: 12/15/2022]
Abstract
Pericytes have been shown to act as precursors of resident adult stem cells in stromal tissues in vivo. When expanded in vitro these cells are capable of giving rise to multiple mesenchymal cell types, irrespective of their tissue of origin. This phenomenon of multi-lineage differentiation is only observed in culture, whereas in vivo, stromal stem cell differentiation is restricted to tissue-specific cell types. An important unanswered question is how a single, widely distributed cell type (a pericyte) gives rise to stem cells with tissue-specific functions and attributes. Using a combination of transcriptomics and epigenomics we have compared the molecular status of two populations of stromal stem cell precursors. Using a LacZ transgene insertion that is expressed in pericytes but not in stem cells, we were able to compare pericyte populations from two different tissues, mouse incisors and bone marrow. Pericytes, freshly isolated from mouse incisors and bone marrow, exhibited transcriptomes and epigenetic landscapes that were extensively different, reflecting their tissue of origin and future in vivo differentiation potential. Dspp, an odontoblast differentiation gene, as well as additional odontogenic genes, are shown to be expressed in dental pulp-derived pericytes. These genetic loci are also decorated with histone modifications indicative of a transcriptionally active chromatin state. In bone marrow pericytes, a major osteogenic differentiation gene, Runx2, is not expressed but is marked by both active and repressive histones and therefore primed to be expressed. Polycomb repressor complex 1 analysis showed that key genes involved in the induction of adipogenesis, chondrogenesis, and myogenesis are targeted by Ring1b and therefore stably repressed. This indicates that pericyte populations are molecularly obstructed from differentiating down certain lineages in vivo. Stem Cells 2018;36:1890-15.
Collapse
Affiliation(s)
- Val Yianni
- Centre for Craniofacial and Regenerative Biology (CCRB), Dental Institute, Kings College London, London, SE1 9RT, United Kingdom
| | - Paul T Sharpe
- Centre for Craniofacial and Regenerative Biology (CCRB), Dental Institute, Kings College London, London, SE1 9RT, United Kingdom
| |
Collapse
|
40
|
Sun J, Luo Z, Wang G, Wang Y, Wang Y, Olmedo M, Morandi MM, Barton S, Kevil CG, Shu B, Shang X, Dong Y. Notch ligand Jagged1 promotes mesenchymal stromal cell-based cartilage repair. Exp Mol Med 2018; 50:1-10. [PMID: 30242147 PMCID: PMC6155067 DOI: 10.1038/s12276-018-0151-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 05/17/2018] [Accepted: 06/07/2018] [Indexed: 01/09/2023] Open
Abstract
Placenta-derived mesenchymal stromal cells (PMSCs) provide a promising cell source for tissue regeneration. However, rapid induction of PMSC chondrogenic differentiation during therapeutic transplantation remains extremely challenging. Here we undertook a study to determine if Notch inhibition by soluble Jagged1 (JAG1) peptides could be utilized to accelerate PMSC-induced cartilage regeneration in a mouse post-traumatic osteoarthritis (PTOA) model. Our results showed that treatment of PMSCs with soluble JAG1 significantly enhanced chondrogenesis in culture as shown by increased alcian blue staining and decreased Notch target Hes1 expression when compared to those in lgG-treated control cells. Importantly, significantly enhanced cartilage formation and decreased joint inflammation were observed when JAG1-treated PMSCs were injected into mouse PTOA knee joints. Finally, in vivo cell tracing showed that more JAG1-treated PMSCs remained in knee joint tissues and that JAG1-treated PMSCs exhibited greater PMSC chondrogenic differentiation than lgG-treated control PMSCs at 4 weeks after injection. These data indicate that transient Notch inhibition by soluble JAG1 could be used to enhance PMSC survival and chondrogenic differentiation, thereby increasing the therapeutic potential of PMSCs for cartilage regeneration. Stem cells derived from placental tissue may help in treating damaged joints, thanks to an improved method for encouraging them to form cartilage. Placenta-derived stem cells are readily available, as the placenta is usually discarded after birth. These stem cells can easily be grown into cartilage tissue in a laboratory, but not when transplanted. Researchers led by Yufeng Dong (Louisiana State University Health Sciences Center, Shreveport, USA) and Xifu Shang (Anhui Provincial Hospital, Hefei, China) treated stem cells with a naturally occurring protein, JAG1, which blocks the signal that prevents them from forming cartilage. They found that JAG1 increased the stem cells’ capacity for cartilage formation. When JAG1-stimulated stem cells were injected into a mouse model of joint disease, cartilage formation was improved, and joint inflammation was reduced.
Collapse
Affiliation(s)
- Junkui Sun
- Department of Orthopaedics, the First Affiliated Hospital, Zhengzhou University, Henan, 450001, China.,Department of Orthopaedics, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Zhengliang Luo
- Department of Orthopedic Surgery, Anhui Provincial Hospital, 17 Lujiang Rd, Hefei, China
| | - Guangxi Wang
- Department of Orthopaedics, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Yuping Wang
- Department of Obstetrics and Gynecology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Yisheng Wang
- Department of Orthopaedics, the First Affiliated Hospital, Zhengzhou University, Henan, 450001, China
| | - Margaret Olmedo
- Department of Orthopaedics, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Massimo Max Morandi
- Department of Orthopaedics, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Shane Barton
- Department of Orthopaedics, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Christopher G Kevil
- Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Bing Shu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xifu Shang
- Department of Orthopedic Surgery, Anhui Provincial Hospital, 17 Lujiang Rd, Hefei, China.
| | - Yufeng Dong
- Department of Orthopaedics, Louisiana State University Health Sciences Center, Shreveport, LA, USA.
| |
Collapse
|
41
|
Affiliation(s)
- Mark W Majesky
- From the Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, and Departments of Pediatrics and Pathology, University of Washington.
| |
Collapse
|
42
|
Kenyon JD, Sergeeva O, Somoza RA, Li M, Caplan AI, Khalil AM, Lee Z. Analysis of -5p and -3p Strands of miR-145 and miR-140 During Mesenchymal Stem Cell Chondrogenic Differentiation. Tissue Eng Part A 2018; 25:80-90. [PMID: 29676203 DOI: 10.1089/ten.tea.2017.0440] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The chondrogenic differentiation of mesenchymal stem cells (MSCs) is mediated by transcription factors and small noncoding RNAs such as microRNAs (miRNAs). Each miRNA is initially transcribed as a long transcript, which matures to produce -5p and -3p strands. It is widely believed that the mature and functional miRNA from any given pre-miRNA, usually the -5p strand, is functional, while the opposing -3p strand is degraded. However, recent cartilage literature started to show functional -3p strands for a few miRNAs. This study aimed at examining both -5p and -3p strands of two key miRNAs miR-140 and miR-145, known to be involved in the chondrogenic differentiation of MSCs. The level (copy number) of both -5p and -3p strands of miR-145 and miR-140 along the time line of MSC chondrogenic differentiation was determined by polymerase chain reaction. The gene expression profiles of several genes related to MSC chondrogenesis were compared with these miRNA profiles along the same timeline. While miR-145-3p is declining in step with miR-145-5p in pellet cultures during the process, the -3p strand is only 1-2% of the total miR-145 products. In contrast, the mature -3p and -5p products of miR-140 are found to increase with near-equal molar expression throughout chondrogenic differentiation. Numerous genes are expressed by cartilage progenitor cells during development. One such target gene, Sox9, is a regulatory target of the dominant miR-145-5p, consistent with the data. Further experimental validations are warranted to confirm that ACAN, FOXO1, and RUNX3 as direct targets of miR-145-5p in the context of MSC chondrogenesis. Similarly, TRSP1 and ACAN are worth further validation as direct targets of miR-145-3p. For miR-140, SOX4 shall be further validated as a direct target of miR-140-5p, while KLF4, PTHLH, and WNT5A can be validated as direct targets of miR-140-3p.
Collapse
Affiliation(s)
- Jonathan D Kenyon
- 1 Department of Biology, Skeletal Research Center, Case Western Reserve University, Cleveland, Ohio
| | - Olga Sergeeva
- 2 Department of Radiology, Case Western Reserve University, Cleveland, Ohio
| | - Rodrigo A Somoza
- 1 Department of Biology, Skeletal Research Center, Case Western Reserve University, Cleveland, Ohio
| | - Ming Li
- 3 Department of Epidemiology and Biostatistics, Case Western Reserve University, Cleveland, Ohio
| | - Arnold I Caplan
- 1 Department of Biology, Skeletal Research Center, Case Western Reserve University, Cleveland, Ohio
| | - Ahmad M Khalil
- 4 Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio
| | - Zhenghong Lee
- 2 Department of Radiology, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
43
|
Steiger D, Yokota T, Li J, Ren S, Minamisawa S, Wang Y. The serine/threonine-protein kinase/endoribonuclease IRE1α protects the heart against pressure overload-induced heart failure. J Biol Chem 2018; 293:9652-9661. [PMID: 29769316 DOI: 10.1074/jbc.ra118.003448] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2018] [Revised: 05/04/2018] [Indexed: 12/26/2022] Open
Abstract
Heart failure is associated with induction of endoplasmic reticulum (ER) stress and the unfolded protein response (UPR). The serine/threonine protein kinase/endoribonuclease IRE1α is a key protein in ER stress signal transduction. IRE1α activity can induce both protective UPR and apoptotic downstream signaling events, but the specific role for IRE1α activity in the heart is unknown. A major aim of this study was to characterize the specific contribution of IRE1α in cardiac physiology and pathogenesis. We used both cultured myocytes and a transgenic mouse line with inducible and cardiomyocyte-specific IRE1α overexpression as experimental models to achieve targeted IRE1α activation. IRE1α expression induced a potent but transient ER stress response in cardiomyocytes and did not cause significant effects in the intact heart under normal physiological conditions. Furthermore, the IRE1α-activated transgenic heart responding to pressure overload exhibited preserved function and reduced fibrotic area, associated with increased adaptive UPR signaling and with blunted inflammatory and pathological gene expression. Therefore, we conclude that IRE1α induces transient ER stress signaling and confers a protective effect against pressure overload-induced pathological remodeling in the heart. To our knowledge, this report provides first direct evidence of a specific and protective role for IRE1α in the heart and reveals an interaction between ER stress signaling and inflammatory regulation in the pathologically stressed heart.
Collapse
Affiliation(s)
- DeAnna Steiger
- From the Departments of Anesthesiology, Physiology, and Medicine, David Geffen School of Medicine, UCLA, Los Angeles, California 90095 and
| | - Tomohiro Yokota
- From the Departments of Anesthesiology, Physiology, and Medicine, David Geffen School of Medicine, UCLA, Los Angeles, California 90095 and
| | - Jin Li
- From the Departments of Anesthesiology, Physiology, and Medicine, David Geffen School of Medicine, UCLA, Los Angeles, California 90095 and
| | - Shuxun Ren
- From the Departments of Anesthesiology, Physiology, and Medicine, David Geffen School of Medicine, UCLA, Los Angeles, California 90095 and
| | - Susumu Minamisawa
- the Department of Cell Physiology, Jikei University School of Medicine, Tokyo 105-8461, Japan
| | - Yibin Wang
- From the Departments of Anesthesiology, Physiology, and Medicine, David Geffen School of Medicine, UCLA, Los Angeles, California 90095 and
| |
Collapse
|
44
|
Li IMH, Liu K, Neal A, Clegg PD, De Val S, Bou-Gharios G. Differential tissue specific, temporal and spatial expression patterns of the Aggrecan gene is modulated by independent enhancer elements. Sci Rep 2018; 8:950. [PMID: 29343853 PMCID: PMC5772622 DOI: 10.1038/s41598-018-19186-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 12/22/2017] [Indexed: 02/06/2023] Open
Abstract
The transcriptional mechanism through which chondrocytes control the spatial and temporal composition of the cartilage tissue has remained largely elusive. The central aim of this study was to identify whether transcriptional enhancers played a role in the organisation of the chondrocytes in cartilaginous tissue. We focused on the Aggrecan gene (Acan) as it is essential for the normal structure and function of cartilage and it is expressed developmentally in different stages of chondrocyte maturation. Using transgenic reporter studies in mice we identified four elements, two of which showed individual chondrocyte developmental stage specificity. In particular, one enhancer (-80) distinguishes itself from the others by being predominantly active in adult cartilage. Furthermore, the -62 element uniquely drove reporter activity in early chondrocytes. The remaining chondrocyte specific enhancers, +28 and -30, showed no preference to chondrocyte type. The transcription factor SOX9 interacted with all the enhancers in vitro and mutation of SOX9 binding sites in one of the enhancers (-30) resulted in a loss of its chondrocyte specificity and ectopic enhancer reporter activity. Thus, the Acan enhancers orchestrate the precise spatiotemporal expression of this gene in cartilage types at different stages of development and adulthood.
Collapse
Affiliation(s)
- Ian M H Li
- Institute of Ageing and Chronic Disease, University of Liverpool, William Henry Duncan Building, Liverpool, L7 8TX, UK
| | - Ke Liu
- Institute of Ageing and Chronic Disease, University of Liverpool, William Henry Duncan Building, Liverpool, L7 8TX, UK
| | - Alice Neal
- Ludwig Cancer Research Ltd, University of Oxford, Oxford, UK
| | - Peter D Clegg
- Institute of Ageing and Chronic Disease, University of Liverpool, William Henry Duncan Building, Liverpool, L7 8TX, UK
| | - Sarah De Val
- Ludwig Cancer Research Ltd, University of Oxford, Oxford, UK
| | - George Bou-Gharios
- Institute of Ageing and Chronic Disease, University of Liverpool, William Henry Duncan Building, Liverpool, L7 8TX, UK.
| |
Collapse
|
45
|
Panda DK, Bai X, Sabbagh Y, Zhang Y, Zaun HC, Karellis A, Koromilas AE, Lipman ML, Karaplis AC. Defective interplay between mTORC1 activity and endoplasmic reticulum stress-unfolded protein response in uremic vascular calcification. Am J Physiol Renal Physiol 2018; 314:F1046-F1061. [PMID: 29357413 DOI: 10.1152/ajprenal.00350.2017] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Vascular calcification increases the risk of cardiovascular disease and death in patients with chronic kidney disease (CKD). Increased activity of mammalian target of rapamycin complex 1 (mTORC1) and endoplasmic reticulum (ER) stress-unfolded protein response (UPR) are independently reported to partake in the pathogenesis of vascular calcification in CKD. However, the association between mTORC1 activity and ER stress-UPR remains unknown. We report here that components of the uremic state [activation of the receptor for advanced glycation end products (RAGE) and hyperphosphatemia] potentiate vascular smooth muscle cell (VSMC) calcification by inducing persistent and exaggerated activity of mTORC1. This gives rise to prolonged and excessive ER stress-UPR as well as attenuated levels of sestrin 1 ( Sesn1) and Sesn3 feeding back to inhibit mTORC1 activity. Activating transcription factor 4 arising from the UPR mediates cell death via expression of CCAAT/enhancer-binding protein (c/EBP) homologous protein (CHOP), impairs the generation of pyrophosphate, a potent inhibitor of mineralization, and potentiates VSMC transdifferentiation to the osteochondrocytic phenotype. Short-term treatment of CKD mice with rapamycin, an inhibitor of mTORC1, or tauroursodeoxycholic acid, a bile acid that restores ER homeostasis, normalized mTORC1 activity, molecular markers of UPR, and calcium content of aortas. Collectively, these data highlight that increased and/or protracted mTORC1 activity arising from the uremic state leads to dysregulated ER stress-UPR and VSMC calcification. Manipulation of the mTORC1-ER stress-UPR pathway opens up new therapeutic strategies for the prevention and treatment of vascular calcification in CKD.
Collapse
Affiliation(s)
- Dibyendu K Panda
- Division of Nephrology, Department of Medicine and Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University , Montreal, Quebec , Canada
| | - Xiuying Bai
- Division of Endocrinology and Metabolism, Department of Medicine and Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University , Montreal, Quebec , Canada
| | - Yves Sabbagh
- Rare Disease, Sanofi Genzyme, Framingham, Massachusetts
| | - Yan Zhang
- Division of Nephrology, Department of Medicine and Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University , Montreal, Quebec , Canada
| | - Hans-Christian Zaun
- Division of Nephrology, Department of Medicine and Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University , Montreal, Quebec , Canada
| | - Angeliki Karellis
- Division of Endocrinology and Metabolism, Department of Medicine and Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University , Montreal, Quebec , Canada
| | - Antonis E Koromilas
- Department of Oncology and Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University , Montreal, Quebec , Canada
| | - Mark L Lipman
- Division of Nephrology, Department of Medicine and Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University , Montreal, Quebec , Canada
| | - Andrew C Karaplis
- Division of Endocrinology and Metabolism, Department of Medicine and Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University , Montreal, Quebec , Canada
| |
Collapse
|
46
|
Mack JJ, Mosqueiro TS, Archer BJ, Jones WM, Sunshine H, Faas GC, Briot A, Aragón RL, Su T, Romay MC, McDonald AI, Kuo CH, Lizama CO, Lane TF, Zovein AC, Fang Y, Tarling EJ, de Aguiar Vallim TQ, Navab M, Fogelman AM, Bouchard LS, Iruela-Arispe ML. NOTCH1 is a mechanosensor in adult arteries. Nat Commun 2017; 8:1620. [PMID: 29158473 PMCID: PMC5696341 DOI: 10.1038/s41467-017-01741-8] [Citation(s) in RCA: 220] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 10/13/2017] [Indexed: 02/06/2023] Open
Abstract
Endothelial cells transduce mechanical forces from blood flow into intracellular signals required for vascular homeostasis. Here we show that endothelial NOTCH1 is responsive to shear stress, and is necessary for the maintenance of junctional integrity, cell elongation, and suppression of proliferation, phenotypes induced by laminar shear stress. NOTCH1 receptor localizes downstream of flow and canonical NOTCH signaling scales with the magnitude of fluid shear stress. Reduction of NOTCH1 destabilizes cellular junctions and triggers endothelial proliferation. NOTCH1 suppression results in changes in expression of genes involved in the regulation of intracellular calcium and proliferation, and preventing the increase of calcium signaling rescues the cell-cell junctional defects. Furthermore, loss of Notch1 in adult endothelium increases hypercholesterolemia-induced atherosclerosis in the descending aorta. We propose that NOTCH1 is atheroprotective and acts as a mechanosensor in adult arteries, where it integrates responses to laminar shear stress and regulates junctional integrity through modulation of calcium signaling.
Collapse
Affiliation(s)
- Julia J Mack
- Department of Molecular, Cell & Developmental Biology, University of California, Los Angeles, CA, 90095, USA
| | - Thiago S Mosqueiro
- Institute for Quantitative and Computational Biology, University of California, Los Angeles, CA, 90095, USA
| | - Brian J Archer
- Department of Bioengineering, University of California, Los Angeles, CA, 90095, USA
| | - William M Jones
- Department of Molecular, Cell & Developmental Biology, University of California, Los Angeles, CA, 90095, USA
| | - Hannah Sunshine
- Interdepartmental Graduate Program in Molecular, Cellular and Integrative Physiology, University of California, Los Angeles, CA, 90095, USA
| | - Guido C Faas
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Anais Briot
- Department of Molecular, Cell & Developmental Biology, University of California, Los Angeles, CA, 90095, USA
| | - Raquel L Aragón
- Molecular Biology Interdisciplinary Graduate Program, Molecular Biology Institute, University of California, Los Angeles, CA, 90095, USA
| | - Trent Su
- Department of Biological Chemistry, University of California, Los Angeles, CA, 90095, USA
| | - Milagros C Romay
- Department of Molecular, Cell & Developmental Biology, University of California, Los Angeles, CA, 90095, USA
| | - Austin I McDonald
- Molecular Biology Interdisciplinary Graduate Program, Molecular Biology Institute, University of California, Los Angeles, CA, 90095, USA
| | - Cheng-Hsiang Kuo
- Department of Medicine, University of Chicago, Chicago, IL, 60637, USA
| | - Carlos O Lizama
- Cardiovascular Research Institute, University of California, San Francisco, CA, 94158, USA
| | - Timothy F Lane
- Department of Biological Chemistry, University of California, Los Angeles, CA, 90095, USA
- Department of Ob-Gyn, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, CA, 90095, USA
| | - Ann C Zovein
- Cardiovascular Research Institute, University of California, San Francisco, CA, 94158, USA
| | - Yun Fang
- Department of Medicine, University of Chicago, Chicago, IL, 60637, USA
| | - Elizabeth J Tarling
- Department of Biological Chemistry, University of California, Los Angeles, CA, 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, CA, 90095, USA
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Thomas Q de Aguiar Vallim
- Department of Biological Chemistry, University of California, Los Angeles, CA, 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, CA, 90095, USA
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Mohamad Navab
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Alan M Fogelman
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Louis S Bouchard
- Department of Bioengineering, University of California, Los Angeles, CA, 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, CA, 90095, USA
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA, 90095, USA
| | - M Luisa Iruela-Arispe
- Department of Molecular, Cell & Developmental Biology, University of California, Los Angeles, CA, 90095, USA.
- Molecular Biology Institute, University of California, Los Angeles, CA, 90095, USA.
| |
Collapse
|
47
|
Siebel C, Lendahl U. Notch Signaling in Development, Tissue Homeostasis, and Disease. Physiol Rev 2017; 97:1235-1294. [PMID: 28794168 DOI: 10.1152/physrev.00005.2017] [Citation(s) in RCA: 674] [Impact Index Per Article: 84.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 05/19/2017] [Accepted: 05/26/2017] [Indexed: 02/07/2023] Open
Abstract
Notch signaling is an evolutionarily highly conserved signaling mechanism, but in contrast to signaling pathways such as Wnt, Sonic Hedgehog, and BMP/TGF-β, Notch signaling occurs via cell-cell communication, where transmembrane ligands on one cell activate transmembrane receptors on a juxtaposed cell. Originally discovered through mutations in Drosophila more than 100 yr ago, and with the first Notch gene cloned more than 30 yr ago, we are still gaining new insights into the broad effects of Notch signaling in organisms across the metazoan spectrum and its requirement for normal development of most organs in the body. In this review, we provide an overview of the Notch signaling mechanism at the molecular level and discuss how the pathway, which is architecturally quite simple, is able to engage in the control of cell fates in a broad variety of cell types. We discuss the current understanding of how Notch signaling can become derailed, either by direct mutations or by aberrant regulation, and the expanding spectrum of diseases and cancers that is a consequence of Notch dysregulation. Finally, we explore the emerging field of Notch in the control of tissue homeostasis, with examples from skin, liver, lung, intestine, and the vasculature.
Collapse
Affiliation(s)
- Chris Siebel
- Department of Discovery Oncology, Genentech Inc., DNA Way, South San Francisco, California; and Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - Urban Lendahl
- Department of Discovery Oncology, Genentech Inc., DNA Way, South San Francisco, California; and Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
48
|
Lan L, Jiang Y, Zhang W, Li T, Ying B, Zhu S. Expression of Notch signaling pathway during osteoarthritis in the temporomandibular joint. J Craniomaxillofac Surg 2017; 45:1338-1348. [PMID: 28684076 DOI: 10.1016/j.jcms.2017.05.029] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 04/28/2017] [Accepted: 05/29/2017] [Indexed: 02/05/2023] Open
Abstract
PURPOSE Our study aim was to characterize the expression of Notch molecules during temporomandibular joint osteoarthritis (TMJOA), thus exploring the mechanism and roles that Notch signaling possibly plays in the initiation and progression of TMJOA. MATERIALS AND METHODS A total of 126 mice were divided randomly into experimental groups, a sham-surgery group and a normal control group. In the experimental group, total discectomy was performed in the right temporomandibular joint (TMJ) to induce TMJOA; the sham-operation group underwent the same procedure without disc removal, and the normal control group was left undisturbed. Fourteen mice in each group were sacrificed in batches respectively at 1, 2, and 4 weeks postoperatively. Histology was performed to examine TMJOA in eight condyles each group, and a modified Mankin scoring system was used for evaluation. Immunohistochemistry was carried out to characterize the expression of the Notch markers Notch1, Jagged1, Hes1, and Hes5. Real-time polymerase chain reaction (RT-PCR) was performed for further detection and analysis of Notch markers in six condyles in each group. RESULTS Notch1, Jagged1, and Hes5 were activated in the experimental group, with expression levels that increased dramatically over time, whereas the control group showed no fluctuation. Hes1 expression was suppressed at the beginning but was up-regulated afterward. CONCLUSIONS Our data suggest that Notch signaling is activated in TMJOA with a much more abundant expression in osteoarthritis cartilage.
Collapse
Affiliation(s)
- Lingli Lan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yangmei Jiang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Weina Zhang
- Department of Stomatology, Ningbo First Hospital, Ningbo 315000, China
| | - Ting Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Binbin Ying
- Department of Stomatology, Ningbo First Hospital, Ningbo 315000, China
| | - Songsong Zhu
- Centre of Orthognathic and TMJ Surgery, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
49
|
Rippe C, Zhu B, Krawczyk KK, Bavel EV, Albinsson S, Sjölund J, Bakker ENTP, Swärd K. Hypertension reduces soluble guanylyl cyclase expression in the mouse aorta via the Notch signaling pathway. Sci Rep 2017; 7:1334. [PMID: 28465505 PMCID: PMC5430981 DOI: 10.1038/s41598-017-01392-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 03/28/2017] [Indexed: 02/06/2023] Open
Abstract
Hypertension is a dominating risk factor for cardiovascular disease. To characterize the genomic response to hypertension, we administered vehicle or angiotensin II to mice and performed gene expression analyses. AngII treatment resulted in a robust increase in blood pressure and altered expression of 235 genes in the aorta, including Gucy1a3 and Gucy1b3 which encode subunits of soluble guanylyl cyclase (sGC). Western blotting and immunohistochemistry confirmed repression of sGC associated with curtailed relaxation via sGC activation. Analysis of transcription factor binding motifs in promoters of differentially expressed genes identified enrichment of motifs for RBPJ, a component of the Notch signaling pathway, and the Notch coactivators FRYL and MAML2 were reduced. Gain and loss of function experiments demonstrated that JAG/NOTCH signaling controls sGC expression together with MAML2 and FRYL. Reduced expression of sGC, correlating with differential expression of MAML2, in stroke prone and spontaneously hypertensive rats was also seen, and RNA-Seq data demonstrated correlations between JAG1, NOTCH3, MAML2 and FRYL and the sGC subunits GUCY1A3 and GUCY1B3 in human coronary artery. Notch signaling thus provides a constitutive drive on expression of the major nitric oxide receptor (GUCY1A3/GUCY1B3) in arteries from mice, rats, and humans, and this control mechanism is disturbed in hypertension.
Collapse
Affiliation(s)
- Catarina Rippe
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Baoyi Zhu
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | | | - Ed Van Bavel
- Department of Biomedical Engineering and Physics, Academic Medical Center, Amsterdam, The Netherlands
| | | | - Jonas Sjölund
- Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Erik N T P Bakker
- Department of Biomedical Engineering and Physics, Academic Medical Center, Amsterdam, The Netherlands
| | - Karl Swärd
- Department of Experimental Medical Science, Lund University, Lund, Sweden.
| |
Collapse
|
50
|
Miller SR, Perera SN, Baker CVH. Constitutively active Notch1 converts cranial neural crest-derived frontonasal mesenchyme to perivascular cells in vivo. Biol Open 2017; 6:317-325. [PMID: 28183698 PMCID: PMC5374403 DOI: 10.1242/bio.023887] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Perivascular/mural cells originate from either the mesoderm or the cranial neural crest. Regardless of their origin, Notch signalling is necessary for their formation. Furthermore, in both chicken and mouse, constitutive Notch1 activation (via expression of the Notch1 intracellular domain) is sufficient in vivo to convert trunk mesoderm-derived somite cells to perivascular cells, at the expense of skeletal muscle. In experiments originally designed to investigate the effect of premature Notch1 activation on the development of neural crest-derived olfactory ensheathing glial cells (OECs), we used in ovo electroporation to insert a tetracycline-inducible NotchΔE construct (encoding a constitutively active mutant of mouse Notch1) into the genome of chicken cranial neural crest cell precursors, and activated NotchΔE expression by doxycycline injection at embryonic day 4. NotchΔE-targeted cells formed perivascular cells within the frontonasal mesenchyme, and expressed a perivascular marker on the olfactory nerve. Hence, constitutively activating Notch1 is sufficient in vivo to drive not only somite cells, but also neural crest-derived frontonasal mesenchyme and perhaps developing OECs, to a perivascular cell fate. These results also highlight the plasticity of neural crest-derived mesenchyme and glia. Summary: Sustained Notch1 activation is sufficient to drive cranial neural crest-derived frontonasal mesenchyme to adopt a perivascular (mural) cell fate in developing chick embryos.
Collapse
Affiliation(s)
- Sophie R Miller
- Department of Physiology, Development and Neuroscience, University of Cambridge, Anatomy Building, Downing Street, Cambridge CB2 3DY, UK
| | - Surangi N Perera
- Department of Physiology, Development and Neuroscience, University of Cambridge, Anatomy Building, Downing Street, Cambridge CB2 3DY, UK
| | - Clare V H Baker
- Department of Physiology, Development and Neuroscience, University of Cambridge, Anatomy Building, Downing Street, Cambridge CB2 3DY, UK
| |
Collapse
|