1
|
Bonet F, Campuzano O, Córdoba-Caballero J, Alcalde M, Sarquella-Brugada G, Braza-Boïls A, Brugada R, Hernández-Torres F, Quezada-Feijoo M, Ramos M, Mangas A, Ranea JAG, Toro R. Role of miRNA-mRNA Interactome in Pathophysiology of Arrhythmogenic Cardiomyopathy. Biomedicines 2024; 12:1807. [PMID: 39200271 PMCID: PMC11351583 DOI: 10.3390/biomedicines12081807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 07/25/2024] [Accepted: 07/29/2024] [Indexed: 09/02/2024] Open
Abstract
Arrhythmogenic cardiomyopathy is an inherited entity characterized by irregular cell-cell adhesion, cardiomyocyte death and fibro-fatty replacement of ventricular myocytes, leading to malignant ventricular arrythmias, contractile dysfunction and sudden cardiac death. Pathogenic variants in genes that encode desmosome are the predominant cause of arrhythmogenic cardiomyopathy. Moreover, signalling pathways such as Wnt/ß-catenin and transforming growth factor-β have been involved in the disease progression. However, still little is known about the molecular pathophysiological mechanisms that underlie arrhythmogenic cardiomyopathy pathogenesis. We used mRNA and small RNA sequencing to analyse the transcriptome of health and arrhythmogenic cardiomyopathy of autopsied human hearts. Our results showed 697 differentially expressed genes and eight differentially expressed miRNAs. Functional enrichment revealed mitochondrial respiratory-related pathways, impaired response to oxidative stress, apoptotic signalling pathways and inflammatory response-related and extracellular matrix response pathways. Furthermore, analysis of the miRNA-mRNA interactome identified eleven negatively correlated miRNA-target pairs for arrhythmogenic cardiomyopathy. Our finding revealed novel arrhythmogenic cardiomyopathy-related miRNAs with important regulatory function in disease pathogenesis, highlighting their value as potential key targets for therapeutic approaches.
Collapse
Affiliation(s)
- Fernando Bonet
- Research Unit, Biomedical Research and Innovation Institute of Cadiz (INiBICA), Puerta del Mar University Hospital, 11009 Cádiz, Spain; (F.B.); (J.C.-C.); (A.M.)
| | - Oscar Campuzano
- Medical Science Department, School of Medicine, University of Girona, 17003 Girona, Spain; (G.S.-B.); (R.B.)
- Institut d’Investigació Biomèdica de Girona (IDIBGI-CERCA), 17190 Salt, Spain;
- Centro Investigación Biomédica en Red, Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain;
| | - José Córdoba-Caballero
- Research Unit, Biomedical Research and Innovation Institute of Cadiz (INiBICA), Puerta del Mar University Hospital, 11009 Cádiz, Spain; (F.B.); (J.C.-C.); (A.M.)
- Department of Molecular Biology and Biochemistry, University of Málaga, 29071 Málaga, Spain;
| | - Mireia Alcalde
- Institut d’Investigació Biomèdica de Girona (IDIBGI-CERCA), 17190 Salt, Spain;
- Centro Investigación Biomédica en Red, Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain;
| | - Georgia Sarquella-Brugada
- Medical Science Department, School of Medicine, University of Girona, 17003 Girona, Spain; (G.S.-B.); (R.B.)
- Pediatric Arrhythmias, Inherited Cardiac Diseases and Sudden Death Unit, Cardiology Department, Sant Joan de Déu Hospital, 08950 Barcelona, Spain
- Arrítmies Pediàtriques, Cardiologia Genètica i Mort Sobtada, Malalties Cardiovasculars en el Desenvolupament, Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain
| | - Aitana Braza-Boïls
- Centro Investigación Biomédica en Red, Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain;
- Cardiopatías Familiares, Muerte Súbita y Mecanismos de Enfermedad (CAFAMUSME) Research Group, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
| | - Ramon Brugada
- Medical Science Department, School of Medicine, University of Girona, 17003 Girona, Spain; (G.S.-B.); (R.B.)
- Institut d’Investigació Biomèdica de Girona (IDIBGI-CERCA), 17190 Salt, Spain;
- Centro Investigación Biomédica en Red, Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain;
- Cardiology Service, Hospital Josep Trueta de Girona, 17007 Girona, Spain
| | - Francisco Hernández-Torres
- Medina Foundation, Technology Park of Health Sciences, 18016 Granada, Spain;
- Department of Biochemistry and Molecular Biology III and Immunology, Faculty of Medicine, University of Granada, 18016 Granada, Spain
| | - Maribel Quezada-Feijoo
- Cardiology Department, Hospital Central de la Cruz Roja, 28003 Madrid, Spain; (M.Q.-F.)
- Medicine School, Alfonso X el Sabio University, 28007 Madrid, Spain
| | - Monica Ramos
- Cardiology Department, Hospital Central de la Cruz Roja, 28003 Madrid, Spain; (M.Q.-F.)
- Medicine School, Alfonso X el Sabio University, 28007 Madrid, Spain
| | - Alipio Mangas
- Research Unit, Biomedical Research and Innovation Institute of Cadiz (INiBICA), Puerta del Mar University Hospital, 11009 Cádiz, Spain; (F.B.); (J.C.-C.); (A.M.)
- Medicine Department, School of Medicine, University of Cadiz, 11003 Cádiz, Spain
- Lipid and Atherosclerotic Unit, Puerta del Mar University Hospital, 11009 Cadiz, Spain
| | - Juan A. G. Ranea
- Department of Molecular Biology and Biochemistry, University of Málaga, 29071 Málaga, Spain;
- Institute of Biomedical Research in Málaga and platform of nanomedicine (IBIMA Plataforma BIONAND), 29071 Málaga, Spain
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
- Spanish National Bioinformatics Institute (INB/ELIXIR-ES), Instituto de Salud Carlos III (ISCIII), 28020 Madrid, Spain
| | - Rocío Toro
- Research Unit, Biomedical Research and Innovation Institute of Cadiz (INiBICA), Puerta del Mar University Hospital, 11009 Cádiz, Spain; (F.B.); (J.C.-C.); (A.M.)
- Medicine Department, School of Medicine, University of Cadiz, 11003 Cádiz, Spain
| |
Collapse
|
2
|
Li C, Ren S, Xiong H, Chen J, Jiang T, Guo J, Yan C, Chen Z, Yang X, Xu X. MiR-145-5p overexpression rejuvenates aged adipose stem cells and accelerates wound healing. Biol Open 2024; 13:bio060117. [PMID: 38315073 PMCID: PMC10903265 DOI: 10.1242/bio.060117] [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: 09/04/2023] [Accepted: 01/08/2024] [Indexed: 02/07/2024] Open
Abstract
Adipose-derived stem cells (ADSCs) have been widely applied in translational and regenerative medicine. During aging, there is a recognized functional decline in ADSCs, which compromises their therapeutic effectiveness. Currently, the mechanisms of aging-induced stem cell dysfunction remain unclear, hence there is a need to elucidate these mechanisms and propose strategies for reversing this functional impairment. In this study, we found that ADSCs isolated from old donors (O-ADSCs) presented inferior phenotypes and decreased miR-145-5p levels compared to those from young donors (Y-ADSCs). To interrogate the role of miR-145-5p in ADSCs, gain- and loss-of-function assays were performed. The results indicated that miR-145-5p overexpression in O-ADSCs promoted cellular proliferation and migration, while reducing cell senescence. Further study demonstrated that miR-145-5p could regulate ADSCs function by targeting bone morphogenetic protein binding endothelial cell precursor-derived regulator (BMPER), which is a crucial modulator in angiogenesis. Moreover, in vivo experiments showed that miR-145-5p-overexpressing O-ADSCs accelerated wound healing by promoting wound re-epithelialization and angiogenesis. Collectively, this study indicates that miR-145-5p works as a positive regulator for optimizing O-ADSCs function, and may be a novel therapeutic target for restoring aging-associated impairments in stem cell function.
Collapse
Affiliation(s)
- Chengcheng Li
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, NO.1277 Jiefang Avenue, Wuhan 430022, China
| | - Sen Ren
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Hewei Xiong
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jing Chen
- Department of Dermatology, Wuhan No.1 Hospital, Wuhan 430000, Hubei, China
| | - Tao Jiang
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, NO.1277 Jiefang Avenue, Wuhan 430022, China
| | - Jiahe Guo
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, NO.1277 Jiefang Avenue, Wuhan 430022, China
| | - Chengqi Yan
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, NO.1277 Jiefang Avenue, Wuhan 430022, China
| | - Zhenbing Chen
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, NO.1277 Jiefang Avenue, Wuhan 430022, China
| | - Xiaofan Yang
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, NO.1277 Jiefang Avenue, Wuhan 430022, China
| | - Xiang Xu
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, NO.1277 Jiefang Avenue, Wuhan 430022, China
| |
Collapse
|
3
|
BMPER Improves Vascular Remodeling and the Contractile Vascular SMC Phenotype. Int J Mol Sci 2023; 24:ijms24054950. [PMID: 36902380 PMCID: PMC10002482 DOI: 10.3390/ijms24054950] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 02/19/2023] [Accepted: 03/01/2023] [Indexed: 03/08/2023] Open
Abstract
Dedifferentiated vascular smooth muscle cells (vSMCs) play an essential role in neointima formation, and we now aim to investigate the role of the bone morphogenetic protein (BMP) modulator BMPER (BMP endothelial cell precursor-derived regulator) in neointima formation. To assess BMPER expression in arterial restenosis, we used a mouse carotid ligation model with perivascular cuff placement. Overall BMPER expression after vessel injury was increased; however, expression in the tunica media was decreased compared to untreated control. Consistently, BMPER expression was decreased in proliferative, dedifferentiated vSMC in vitro. C57BL/6_Bmper+/- mice displayed increased neointima formation 21 days after carotid ligation and enhanced expression of Col3A1, MMP2, and MMP9. Silencing of BMPER increased the proliferation and migration capacity of primary vSMCs, as well as reduced contractibility and expression of contractile markers, whereas stimulation with recombinant BMPER protein had the opposite effect. Mechanistically, we showed that BMPER binds insulin-like growth factor-binding protein 4 (IGFBP4), resulting in the modulation of IGF signaling. Furthermore, perivascular application of recombinant BMPER protein prevented neointima formation and ECM deposition in C57BL/6N mice after carotid ligation. Our data demonstrate that BMPER stimulation causes a contractile vSMC phenotype and suggest that BMPER has the potential for a future therapeutic agent in occlusive cardiovascular diseases.
Collapse
|
4
|
Elmasry K, Habib S, Moustafa M, Al-Shabrawey M. Bone Morphogenetic Proteins and Diabetic Retinopathy. Biomolecules 2021; 11:biom11040593. [PMID: 33919531 PMCID: PMC8073699 DOI: 10.3390/biom11040593] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/16/2021] [Accepted: 04/16/2021] [Indexed: 12/12/2022] Open
Abstract
Bone morphogenetic proteins (BMPs) play an important role in bone formation and repair. Recent studies underscored their essential role in the normal development of several organs and vascular homeostasis in health and diseases. Elevated levels of BMPs have been linked to the development of cardiovascular complications of diabetes mellitus. However, their particular role in the pathogenesis of microvascular dysfunction associated with diabetic retinopathy (DR) is still under-investigated. Accumulated evidence from our and others’ studies suggests the involvement of BMP signaling in retinal inflammation, hyperpermeability and pathological neovascularization in DR and age-related macular degeneration (AMD). Therefore, targeting BMP signaling in diabetes is proposed as a potential therapeutic strategy to halt the development of microvascular dysfunction in retinal diseases, particularly in DR. The goal of this review article is to discuss the biological functions of BMPs, their underlying mechanisms and their potential role in the pathogenesis of DR in particular.
Collapse
Affiliation(s)
- Khaled Elmasry
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA;
- Culver Vision discovery Institute, Augusta University, Augusta, GA 30912, USA;
- Department of Anatomy, Mansoura Faculty of Medicine, Mansoura University, Dakahlia Governorate 35516, Egypt
| | - Samar Habib
- Department of Medical Parasitology, Mansoura Faculty of Medicine, Mansoura University, Dakahlia Governorate 35516, Egypt;
- Department of Obstetrics and Gynecology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Mohamed Moustafa
- Culver Vision discovery Institute, Augusta University, Augusta, GA 30912, USA;
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Mohamed Al-Shabrawey
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA;
- Culver Vision discovery Institute, Augusta University, Augusta, GA 30912, USA;
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA 30912, USA
- Correspondence: ; Tel.: +1-(706)721-4278 or +1-(706)721-4279
| |
Collapse
|
5
|
An X, Ma H, Liu Y, Li F, Song Y, Li G, Bai Y, Cao B. Effects of miR-101-3p on goat granulosa cells in vitro and ovarian development in vivo via STC1. J Anim Sci Biotechnol 2020; 11:102. [PMID: 33072314 PMCID: PMC7557009 DOI: 10.1186/s40104-020-00506-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 08/18/2020] [Indexed: 12/23/2022] Open
Abstract
Background MiRNAs act as pivotal post-transcriptional gene mediators in the regulation of diverse biological processes, including proliferation, development and apoptosis. Our previous study has showed that miR-101-3p is differentially expressed in dairy goat ovaries compared single with multiple litters. The objective of this research was to explore the potential function and molecular mechanism of miR-101-3p via its target STC1 in goat ovarian growth and development. Results cDNA libraries were constructed using goat granulosa cells transfected with miR-101-3p mimics and negative control by RNA-sequencing. In total, 142 differentially expressed unigenes (DEGs) were detected between two libraries, including 78 down-regulated and 64 up-regulated genes. GO and KEGG enrichment analysis showed the potential impacts of DEGs on ovarian development. STC1 was singled out from DEGs for further research owing to it regulates reproductive-related processes. In vitro, bioinformatics analysis and 3′-UTR assays confirmed that STC1 was a target of miR-101-3p. ELISA was performed to detect the estrogen (E2) and progesterone (P4) levels. CCK8, EdU and flow cytometry assays were performed to detect the proliferation and apoptosis of granulosa cells. Results showed that miR-101-3p regulated STAR, CYP19A1, CYP11A1 and 3β-HSD steroid hormone synthesis-associated genes by STC1 depletion, thus promoted E2 and P4 secretions. MiR-101-3p also affected the key protein PI3K, PTEN, AKT and mTOR in PI3K-AKT pathway by STC1, thereby suppressing proliferation and promoting apoptosis of granulosa cells. In vivo, the distribution and expression levels of miR-101-3p in mouse ovaries were determined through fluorescence in situ hybridisation (FISH). Immunohistochemistry results showed that STC1 expression was suppressed in mouse ovaries in miR-101-3p-agonist and siRNA-STC1 groups. Small and stunted ovarian fragments, decreased numbers of follicles at diverse stages were observed using Hematoxylin-eosin (HE) staining, thereby showing unusual ovarian development after miR-101-3p overexpression or STC1 depletion. Inhibition of miR-101-3p manifested opposite results. Conclusions Taken together, our results demonstrated a regulatory mechanism of miR-101-3p via STC1 in goat granulosa cells, and offered the first in vivo example of miR-101-3p and STC1 functions required for ovarian development.
Collapse
Affiliation(s)
- Xiaopeng An
- College of Animal Science and Technology, Northwest A&F University, No. 22 Xinong Road, Yangling, Shaanxi 712100 P.R. China
| | - Haidong Ma
- College of Animal Science and Technology, Northwest A&F University, No. 22 Xinong Road, Yangling, Shaanxi 712100 P.R. China.,College of Biological Science and Engineering, Shaanxi University and Technology, Hanzhong, Shaanxi, 723001 P.R. China
| | - Yuhan Liu
- College of Animal Science and Technology, Northwest A&F University, No. 22 Xinong Road, Yangling, Shaanxi 712100 P.R. China
| | - Fu Li
- College of Animal Science and Technology, Northwest A&F University, No. 22 Xinong Road, Yangling, Shaanxi 712100 P.R. China
| | - Yuxuan Song
- College of Animal Science and Technology, Northwest A&F University, No. 22 Xinong Road, Yangling, Shaanxi 712100 P.R. China
| | - Guang Li
- College of Animal Science and Technology, Northwest A&F University, No. 22 Xinong Road, Yangling, Shaanxi 712100 P.R. China
| | - Yueyu Bai
- Henan Animal Health Supervision Institution, No. 91 Jingsan Road, Zhengzhou, Henan 450008 P.R. China
| | - Binyun Cao
- College of Animal Science and Technology, Northwest A&F University, No. 22 Xinong Road, Yangling, Shaanxi 712100 P.R. China
| |
Collapse
|
6
|
Borasch K, Richardson K, Plendl J. Cardiogenesis with a focus on vasculogenesis and angiogenesis. Anat Histol Embryol 2020; 49:643-655. [PMID: 32319704 DOI: 10.1111/ahe.12549] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 02/04/2020] [Accepted: 02/20/2020] [Indexed: 12/21/2022]
Abstract
The initial intraembryonic vasculogenesis occurs in the cardiogenic mesoderm. Here, a cell population of proendocardial cells detaches from the mesoderm that subsequently generates the single endocardial tube by forming vascular plexuses. In the course of embryogenesis, the endocardium retains vasculogenic, angiogenic and haematopoietic potential. The coronary blood vessels that sustain the rapidly expanding myocardium develop in the course of the formation of the cardiac loop by vasculogenesis and angiogenesis from progenitor cells of the proepicardial serosa at the venous pole of the heart as well as from the endocardium and endothelial cells of the sinus venosus. Prospective coronary endothelial cells and progenitor cells of the coronary blood vessel walls (smooth muscle cells, perivascular cells) originate from different cell populations that are in close spatial as well as regulatory connection with each other. Vasculo- and angiogenesis of the coronary blood vessels are for a large part regulated by the epicardium and epicardium-derived cells. Vasculogenic and angiogenic signalling pathways include the vascular endothelial growth factors, the angiopoietins and the fibroblast growth factors and their receptors.
Collapse
Affiliation(s)
- Katrin Borasch
- Department of Veterinary Medicine, Institute of Veterinary Anatomy, Freie University Berlin, Berlin, Germany
| | - Kenneth Richardson
- College of Veterinary Medicine, School of Veterinary and Life Sciences, Murdoch University, Murdoch, WA, Australia
| | - Johanna Plendl
- Department of Veterinary Medicine, Institute of Veterinary Anatomy, Freie University Berlin, Berlin, Germany
| |
Collapse
|
7
|
Aluganti Narasimhulu C, Singla DK. The Role of Bone Morphogenetic Protein 7 (BMP-7) in Inflammation in Heart Diseases. Cells 2020; 9:cells9020280. [PMID: 31979268 PMCID: PMC7073173 DOI: 10.3390/cells9020280] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 01/17/2020] [Accepted: 01/21/2020] [Indexed: 12/31/2022] Open
Abstract
Bone morphogenetic protein-7 is (BMP-7) is a potent anti-inflammatory growth factor belonging to the Transforming Growth Factor Beta (TGF-β) superfamily. It plays an important role in various biological processes, including embryogenesis, hematopoiesis, neurogenesis and skeletal morphogenesis. BMP-7 stimulates the target cells by binding to specific membrane-bound receptor BMPR 2 and transduces signals through mothers against decapentaplegic (Smads) and mitogen activated protein kinase (MAPK) pathways. To date, rhBMP-7 has been used clinically to induce the differentiation of mesenchymal stem cells bordering the bone fracture site into chondrocytes, osteoclasts, the formation of new bone via calcium deposition and to stimulate the repair of bone fracture. However, its use in cardiovascular diseases, such as atherosclerosis, myocardial infarction, and diabetic cardiomyopathy is currently being explored. More importantly, these cardiovascular diseases are associated with inflammation and infiltrated monocytes where BMP-7 has been demonstrated to be a key player in the differentiation of pro-inflammatory monocytes, or M1 macrophages, into anti-inflammatory M2 macrophages, which reduces developed cardiac dysfunction. Therefore, this review focuses on the molecular mechanisms of BMP-7 treatment in cardiovascular disease and its role as an anti-fibrotic, anti-apoptotic and anti-inflammatory growth factor, which emphasizes its potential therapeutic significance in heart diseases.
Collapse
|
8
|
Urness LD, Wang X, Doan H, Shumway N, Noyes CA, Gutierrez-Magana E, Lu R, Mansour SL. Spatial and temporal inhibition of FGFR2b ligands reveals continuous requirements and novel targets in mouse inner ear morphogenesis. Development 2018; 145:dev.170142. [PMID: 30504125 DOI: 10.1242/dev.170142] [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] [Received: 07/17/2018] [Accepted: 11/19/2018] [Indexed: 12/25/2022]
Abstract
Morphogenesis of the inner ear epithelium requires coordinated deployment of several signaling pathways, and disruptions cause abnormalities of hearing and/or balance. The FGFR2b ligands FGF3 and FGF10 are expressed throughout otic development and are required individually for normal morphogenesis, but their prior and redundant roles in otic placode induction complicates investigation of subsequent combinatorial functions in morphogenesis. To interrogate these roles and identify new effectors of FGF3 and FGF10 signaling at the earliest stages of otic morphogenesis, we used conditional gene ablation after otic placode induction, and temporal inhibition of signaling with a secreted, dominant-negative FGFR2b ectodomain. We show that both ligands are required continuously after otocyst formation for maintenance of otic neuroblasts and for patterning and proliferation of the epithelium, leading to normal morphogenesis of both the cochlear and vestibular domains. Furthermore, the first genome-wide identification of proximal targets of FGFR2b signaling in the early otocyst reveals novel candidate genes for inner ear development and function.
Collapse
Affiliation(s)
- Lisa D Urness
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112-5330, USA
| | - Xiaofen Wang
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112-5330, USA
| | - Huy Doan
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112-5330, USA
| | - Nathan Shumway
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112-5330, USA
| | - C Albert Noyes
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112-5330, USA
| | | | - Ree Lu
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112-5330, USA
| | - Suzanne L Mansour
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112-5330, USA .,Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT 84112-5330, USA
| |
Collapse
|
9
|
Ramai D, Lai J, Monzidelis C, Reddy S. Coronary Artery Development: Origin, Malformations, and Translational Vascular Reparative Therapy. J Cardiovasc Pharmacol Ther 2018; 23:292-300. [PMID: 29642708 DOI: 10.1177/1074248418769633] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
After thickening of the cardiac chamber walls during embryogenesis, oxygen and nutrients can no longer be adequately supplied to cardiac cells via passive diffusion; therefore, a primitive vascular network develops to supply these vital structures. This plexus further matures into coronary arteries and veins, which ensures continued development of the heart. Various models have been proposed to account for the growth of the coronary arteries. However, lineage-tracing studies in the last decade have identified 3 major sources, namely, the proepicardium, the sinus venosus, and endocardium. Although the exact contribution of each source remains unknown, the emerging model depicts alternative pathways and progenitor cells, which ensure successful coronary angiogenesis. We aim to explore the current trends in coronary artery development, the cellular and molecular signals regulating heart vascularization, and its implications for heart disease and vascular regeneration.
Collapse
Affiliation(s)
- Daryl Ramai
- Department of Medicine, The Brooklyn Hospital Center, Academic Affiliate of The Icahn School of Medicine at Mount Sinai, Clinical Affiliate of The Mount Sinai Hospital, Brooklyn, NY, USA
- Department of Anatomical Sciences, School of Medicine, St George’s University, Grenada, West Indies
| | - Jonathan Lai
- Department of Anatomical Sciences, School of Medicine, St George’s University, Grenada, West Indies
| | - Constantine Monzidelis
- Department of Medicine, The Brooklyn Hospital Center, Academic Affiliate of The Icahn School of Medicine at Mount Sinai, Clinical Affiliate of The Mount Sinai Hospital, Brooklyn, NY, USA
| | - Sarath Reddy
- Division of Cardiology, The Brooklyn Hospital Center, Academic Affiliate of The Icahn School of Medicine at Mount Sinai, Clinical Affiliate of The Mount Sinai Hospital, Brooklyn, NY, USA
| |
Collapse
|
10
|
Esser JS, Steiner RE, Deckler M, Schmitt H, Engert B, Link S, Charlet A, Patterson C, Bode C, Zhou Q, Moser M. Extracellular bone morphogenetic protein modulator BMPER and twisted gastrulation homolog 1 preserve arterial-venous specification in zebrafish blood vessel development and regulate Notch signaling in endothelial cells. FEBS J 2018; 285:1419-1436. [PMID: 29473997 DOI: 10.1111/febs.14414] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 01/26/2018] [Accepted: 02/19/2018] [Indexed: 01/16/2023]
Abstract
The bone morphogenetic protein (BMP) signaling pathway plays a central role during vasculature development. Mutations or dysregulation of the BMP pathway members have been linked to arteriovenous malformations. In the present study, we investigated the effect of the BMP modulators bone morphogenetic protein endothelial precursor-derived regulator (BMPER) and twisted gastrulation protein homolog 1 (TWSG1) on arteriovenous specification during zebrafish development and analyzed downstream Notch signaling pathway in human endothelial cells. Silencing of bmper and twsg1b in zebrafish embryos by morpholinos resulted in a pronounced enhancement of venous ephrinB4a marker expression and concomitant dysregulated arterial ephrinb2a marker expression detected by in situ hybridization. As arteriovenous specification was disturbed, we assessed the impact of BMPER and TWSG1 protein stimulation on the Notch signaling pathway on endothelial cells from different origin. Quantitative real-time PCR (qRT-PCR) and western blot analysis showed increased expression of Notch target gene hairy and enhancer of split, HEY1/2 and EPHRINB2. Consistently, silencing of BMPER in endothelial cells by siRNAs decreased Notch signaling and downstream effectors. BMP receptor antagonist DMH1 abolished BMPER and BMP4 induced Notch signaling pathway activation. In conclusion, we found that in endothelial cells, BMPER and TWSG1 are necessary for regular Notch signaling activity and in zebrafish embryos BMPER and TWSG1 preserve arteriovenous specification to prevent malformations.
Collapse
Affiliation(s)
- Jennifer Susanne Esser
- Department of Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University Freiburg, Germany
| | - Rahel Elisabeth Steiner
- Department of Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University Freiburg, Germany
| | - Meike Deckler
- Department of Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University Freiburg, Germany
| | - Hannah Schmitt
- Department of Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University Freiburg, Germany
| | - Bianca Engert
- Department of Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University Freiburg, Germany
| | - Sandra Link
- Department of Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University Freiburg, Germany
| | - Anne Charlet
- Department of Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University Freiburg, Germany
| | - Cam Patterson
- Weill Cornell Medical Center, New York Presbyterian Hospital, NY, USA
| | - Christoph Bode
- Department of Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University Freiburg, Germany
| | - Qian Zhou
- Department of Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University Freiburg, Germany
| | - Martin Moser
- Department of Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University Freiburg, Germany
| |
Collapse
|
11
|
McGarvey AC, Rybtsov S, Souilhol C, Tamagno S, Rice R, Hills D, Godwin D, Rice D, Tomlinson SR, Medvinsky A. A molecular roadmap of the AGM region reveals BMPER as a novel regulator of HSC maturation. J Exp Med 2017; 214:3731-3751. [PMID: 29093060 PMCID: PMC5716029 DOI: 10.1084/jem.20162012] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 06/16/2017] [Accepted: 09/01/2017] [Indexed: 12/16/2022] Open
Abstract
Through transcriptional profiling of the mouse AGM region, McGarvey et al. identify potential niche regulators of HSC development. They show a new function of BMPER in regulating HSC maturation, likely via its modulation of BMP signalling. In the developing embryo, hematopoietic stem cells (HSCs) emerge from the aorta-gonad-mesonephros (AGM) region, but the molecular regulation of this process is poorly understood. Recently, the progression from E9.5 to E10.5 and polarity along the dorso-ventral axis have been identified as clear demarcations of the supportive HSC niche. To identify novel secreted regulators of HSC maturation, we performed RNA sequencing over these spatiotemporal transitions in the AGM region and supportive OP9 cell line. Screening several proteins through an ex vivo reaggregate culture system, we identify BMPER as a novel positive regulator of HSC development. We demonstrate that BMPER is associated with BMP signaling inhibition, but is transcriptionally induced by BMP4, suggesting that BMPER contributes to the precise control of BMP activity within the AGM region, enabling the maturation of HSCs within a BMP-negative environment. These findings and the availability of our transcriptional data through an accessible interface should provide insight into the maintenance and potential derivation of HSCs in culture.
Collapse
Affiliation(s)
- Alison C McGarvey
- Stem Cell Bioinformatics Group, Institute for Stem Cell Research, Medical Research Council Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, Scotland, UK
| | - Stanislav Rybtsov
- Ontogeny of Haematopoietic Stem Cells Group, Institute for Stem Cell Research, Medical Research Council Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, Scotland, UK
| | - Céline Souilhol
- Ontogeny of Haematopoietic Stem Cells Group, Institute for Stem Cell Research, Medical Research Council Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, Scotland, UK
| | - Sara Tamagno
- Ontogeny of Haematopoietic Stem Cells Group, Institute for Stem Cell Research, Medical Research Council Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, Scotland, UK
| | - Ritva Rice
- University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - David Hills
- Ontogeny of Haematopoietic Stem Cells Group, Institute for Stem Cell Research, Medical Research Council Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, Scotland, UK
| | - Duncan Godwin
- Stem Cell Bioinformatics Group, Institute for Stem Cell Research, Medical Research Council Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, Scotland, UK
| | - David Rice
- University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Simon R Tomlinson
- Stem Cell Bioinformatics Group, Institute for Stem Cell Research, Medical Research Council Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, Scotland, UK
| | - Alexander Medvinsky
- Ontogeny of Haematopoietic Stem Cells Group, Institute for Stem Cell Research, Medical Research Council Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, Scotland, UK
| |
Collapse
|
12
|
Lockyer P, Mao H, Fan Q, Li L, Yu-Lee LY, Eissa NT, Patterson C, Xie L, Pi X. LRP1-Dependent BMPER Signaling Regulates Lipopolysaccharide-Induced Vascular Inflammation. Arterioscler Thromb Vasc Biol 2017; 37:1524-1535. [PMID: 28596374 DOI: 10.1161/atvbaha.117.309521] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2016] [Accepted: 05/30/2017] [Indexed: 01/10/2023]
Abstract
OBJECTIVE Bacterial endotoxin (lipopolysaccharide)-mediated sepsis involves dysregulated systemic inflammation, which injures the lung and other organs, often fatally. Vascular endothelial cells act as both targets and mediators of lipopolysaccharide-induced inflammatory responses. Dysfunction of endothelium results in increases of proinflammatory cytokine production and permeability leakage. BMPER (bone morphogenetic protein-binding endothelial regulator), an extracellular modulator of bone morphogenetic protein signaling, has been identified as a vital component in chronic endothelial inflammatory responses and atherosclerosis. However, it is unclear whether BMPER also regulates inflammatory response in an acute setting such as sepsis. To address this question, we investigated the role of BMPER during lipopolysaccharide-induced acute lung injury. APPROACH AND RESULTS Mice missing 1 allele of BMPER (BMPER+/- mice used in the place of BMPER-/- mice that die at birth) were used for lipopolysaccharide challenge. Lipopolysaccharide-induced pulmonary inflammation and injury was reduced in BMPER+/- mice as shown by several measures, including survival rate, infiltration of inflammatory cells, edema, and production of proinflammatory cytokines. Mechanistically, we have demonstrated that BMPER is required and sufficient for the activation of nuclear factor of activated T cells c1. This BMPER-induced nuclear factor of activated T cells activation is coordinated by multiple signaling pathways, including bone morphogenetic protein-independent low-density lipoprotein receptor-related protein 1-extracellular signal-regulated kinase activation, calcineurin signaling, and low-density lipoprotein receptor-related protein 1β-mediated nuclear factor 45 nuclear export in response to BMPER treatment. CONCLUSIONS We conclude that BMPER plays a pivotal role in pulmonary inflammatory response, which provides new therapeutic options against sepsis shock. The new signaling pathway initiated by BMPER/low-density lipoprotein receptor-related protein 1 axis broadens our understanding about BMPER's role in vascular homeostasis.
Collapse
Affiliation(s)
- Pamela Lockyer
- From the Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill (P.L.); Department of Medicine, Section of Athero & Lipo, Cardiovascular Research Institute (H.M., Q.F., L.L., L.X., X.P.), Departments of Molecular and Cellular Biology and Medicine, Section of Immunology Allergy and Rheumatology, Integrative Molecular and Biomedical Sciences (L.Y.Y.L.), and Departments of Medicine and Pathology and Immunology (N.T.E.), Baylor College of Medicine, Houston, TX; and New York-Presbyterian Hospital, New York (C.P.)
| | - Hua Mao
- From the Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill (P.L.); Department of Medicine, Section of Athero & Lipo, Cardiovascular Research Institute (H.M., Q.F., L.L., L.X., X.P.), Departments of Molecular and Cellular Biology and Medicine, Section of Immunology Allergy and Rheumatology, Integrative Molecular and Biomedical Sciences (L.Y.Y.L.), and Departments of Medicine and Pathology and Immunology (N.T.E.), Baylor College of Medicine, Houston, TX; and New York-Presbyterian Hospital, New York (C.P.)
| | - Qiying Fan
- From the Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill (P.L.); Department of Medicine, Section of Athero & Lipo, Cardiovascular Research Institute (H.M., Q.F., L.L., L.X., X.P.), Departments of Molecular and Cellular Biology and Medicine, Section of Immunology Allergy and Rheumatology, Integrative Molecular and Biomedical Sciences (L.Y.Y.L.), and Departments of Medicine and Pathology and Immunology (N.T.E.), Baylor College of Medicine, Houston, TX; and New York-Presbyterian Hospital, New York (C.P.)
| | - Luge Li
- From the Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill (P.L.); Department of Medicine, Section of Athero & Lipo, Cardiovascular Research Institute (H.M., Q.F., L.L., L.X., X.P.), Departments of Molecular and Cellular Biology and Medicine, Section of Immunology Allergy and Rheumatology, Integrative Molecular and Biomedical Sciences (L.Y.Y.L.), and Departments of Medicine and Pathology and Immunology (N.T.E.), Baylor College of Medicine, Houston, TX; and New York-Presbyterian Hospital, New York (C.P.)
| | - Li-Yuan Yu-Lee
- From the Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill (P.L.); Department of Medicine, Section of Athero & Lipo, Cardiovascular Research Institute (H.M., Q.F., L.L., L.X., X.P.), Departments of Molecular and Cellular Biology and Medicine, Section of Immunology Allergy and Rheumatology, Integrative Molecular and Biomedical Sciences (L.Y.Y.L.), and Departments of Medicine and Pathology and Immunology (N.T.E.), Baylor College of Medicine, Houston, TX; and New York-Presbyterian Hospital, New York (C.P.)
| | - N Tony Eissa
- From the Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill (P.L.); Department of Medicine, Section of Athero & Lipo, Cardiovascular Research Institute (H.M., Q.F., L.L., L.X., X.P.), Departments of Molecular and Cellular Biology and Medicine, Section of Immunology Allergy and Rheumatology, Integrative Molecular and Biomedical Sciences (L.Y.Y.L.), and Departments of Medicine and Pathology and Immunology (N.T.E.), Baylor College of Medicine, Houston, TX; and New York-Presbyterian Hospital, New York (C.P.)
| | - Cam Patterson
- From the Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill (P.L.); Department of Medicine, Section of Athero & Lipo, Cardiovascular Research Institute (H.M., Q.F., L.L., L.X., X.P.), Departments of Molecular and Cellular Biology and Medicine, Section of Immunology Allergy and Rheumatology, Integrative Molecular and Biomedical Sciences (L.Y.Y.L.), and Departments of Medicine and Pathology and Immunology (N.T.E.), Baylor College of Medicine, Houston, TX; and New York-Presbyterian Hospital, New York (C.P.)
| | - Liang Xie
- From the Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill (P.L.); Department of Medicine, Section of Athero & Lipo, Cardiovascular Research Institute (H.M., Q.F., L.L., L.X., X.P.), Departments of Molecular and Cellular Biology and Medicine, Section of Immunology Allergy and Rheumatology, Integrative Molecular and Biomedical Sciences (L.Y.Y.L.), and Departments of Medicine and Pathology and Immunology (N.T.E.), Baylor College of Medicine, Houston, TX; and New York-Presbyterian Hospital, New York (C.P.)
| | - Xinchun Pi
- From the Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill (P.L.); Department of Medicine, Section of Athero & Lipo, Cardiovascular Research Institute (H.M., Q.F., L.L., L.X., X.P.), Departments of Molecular and Cellular Biology and Medicine, Section of Immunology Allergy and Rheumatology, Integrative Molecular and Biomedical Sciences (L.Y.Y.L.), and Departments of Medicine and Pathology and Immunology (N.T.E.), Baylor College of Medicine, Houston, TX; and New York-Presbyterian Hospital, New York (C.P.).
| |
Collapse
|
13
|
Sharma B, Chang A, Red-Horse K. Coronary Artery Development: Progenitor Cells and Differentiation Pathways. Annu Rev Physiol 2016; 79:1-19. [PMID: 27959616 DOI: 10.1146/annurev-physiol-022516-033953] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Coronary artery disease (CAD) is the number one cause of death worldwide and involves the accumulation of plaques within the artery wall that can occlude blood flow to the heart and cause myocardial infarction. The high mortality associated with CAD makes the development of medical interventions that repair and replace diseased arteries a high priority for the cardiovascular research community. Advancements in arterial regenerative medicine could benefit from a detailed understanding of coronary artery development during embryogenesis and of how these pathways might be reignited during disease. Recent research has advanced our knowledge on how the coronary vasculature is built and revealed unexpected features of progenitor cell deployment that may have implications for organogenesis in general. Here, we highlight these recent findings and discuss how they set the stage to interrogate developmental pathways during injury and disease.
Collapse
Affiliation(s)
- Bikram Sharma
- Department of Biology, Stanford University, Stanford, California 94305;
| | - Andrew Chang
- Department of Biology, Stanford University, Stanford, California 94305; .,Department of Developmental Biology, Stanford University, Stanford, California 94305
| | - Kristy Red-Horse
- Department of Biology, Stanford University, Stanford, California 94305;
| |
Collapse
|
14
|
Abstract
The discovery of the transforming growth factor β (TGF-β) family ligands and the realization that their bioactivities need to be tightly controlled temporally and spatially led to intensive research that has identified a multitude of extracellular modulators of TGF-β family ligands, uncovered their functions in developmental and pathophysiological processes, defined the mechanisms of their activities, and explored potential modulator-based therapeutic applications in treating human diseases. These studies revealed a diverse repertoire of extracellular and membrane-associated molecules that are capable of modulating TGF-β family signals via control of ligand availability, processing, ligand-receptor interaction, and receptor activation. These molecules include not only soluble ligand-binding proteins that were conventionally considered as agonists and antagonists of TGF-β family of growth factors, but also extracellular matrix (ECM) proteins and proteoglycans that can serve as "sink" and control storage and release of both the TGF-β family ligands and their regulators. This extensive network of soluble and ECM modulators helps to ensure dynamic and cell-specific control of TGF-β family signals. This article reviews our knowledge of extracellular modulation of TGF-β growth factors by diverse proteins and their molecular mechanisms to regulate TGF-β family signaling.
Collapse
Affiliation(s)
- Chenbei Chang
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama 35294
| |
Collapse
|
15
|
Associations between allelic polymorphism of the BMP Binding Endothelial Regulator and phenotypic variation of cattle. Mol Cell Probes 2015; 29:358-364. [DOI: 10.1016/j.mcp.2015.09.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 09/23/2015] [Accepted: 09/23/2015] [Indexed: 11/22/2022]
|
16
|
Ye L, Jiang WG. Bone morphogenetic proteins in tumour associated angiogenesis and implication in cancer therapies. Cancer Lett 2015; 380:586-597. [PMID: 26639195 DOI: 10.1016/j.canlet.2015.10.036] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 09/17/2015] [Accepted: 10/12/2015] [Indexed: 02/09/2023]
Abstract
Bone morphogenetic protein (BMP) belongs to transforming growth factor-β superfamily. To date, more than 20 BMPs have been identified in humans. BMPs play a critical role in embryonic and postnatal development, and also in maintaining homeostasis in different organs and tissues by regulating cell differentiation, proliferation, survival and motility. They play important roles in the development and progression of certain malignancies, including prostate cancer, breast cancer, lung cancer, etc. Recently, more evidence shows that BMPs are also involved in tumour associated angiogenesis. For example BMP can either directly regulate the functions of vascular endothelial cells or indirectly influence the angiogenesis via regulation of angiogenic factors, such as vascular endothelial growth factor (VEGF). Such crosstalk can also be reflected in the interaction with other angiogenic factors, like hepatocyte growth factor (HGF) and basic fibroblast growth factor (bFGF). All these factors are involved in the orchestration of the angiogenic process during tumour development and progression. Review of the relevant studies will provide a comprehensive prospective on current understanding and shed light on the corresponding therapeutic opportunity.
Collapse
Affiliation(s)
- Lin Ye
- Metastasis & Angiogenesis Research Group, Cardiff University-Peking University Cancer Institute, Institute of Cancer and Genetics, Cardiff University School of Medicine, Cardiff CF14 4XN, UK.
| | - Wen G Jiang
- Metastasis & Angiogenesis Research Group, Cardiff University-Peking University Cancer Institute, Institute of Cancer and Genetics, Cardiff University School of Medicine, Cardiff CF14 4XN, UK
| |
Collapse
|
17
|
Huan C, Yang T, Liang J, Xie T, Cheng L, Liu N, Kurkciyan A, Monterrosa Mena J, Wang C, Dai H, Noble PW, Jiang D. Methylation-mediated BMPER expression in fibroblast activation in vitro and lung fibrosis in mice in vivo. Sci Rep 2015; 5:14910. [PMID: 26442443 PMCID: PMC4595647 DOI: 10.1038/srep14910] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 09/11/2015] [Indexed: 12/20/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive lung disease. Although the pathogenesis is poorly understood, evidence suggests that genetic and epigenetic alterations, such as DNA methylation, may play a key role. Bone morphogenetic proteins (BMPs) are members of the transforming growth factor-β (TGF-β) superfamily and are important regulators in IPF. Here we identified BMP endothelial cell precursor-derived regulator (BMPER) as a key regulator of fibroblast activation. BMPER is a secreted glycoprotein that binds directly to BMPs and may regulate TGF-β/BMP signaling, but its role in lung fibrosis is not clear. BMPER is highly expressed in human IPF lung fibroblasts compared to normal lung fibroblasts. Demethylation agent 5′-azacytidine decreased BMPER expression in fibroblasts, and attenuated the invasion and migration of IPF lung fibroblasts. Furthermore, siRNA-mediated reduction of BMPER in the human lung fibroblasts impaired cell migration and invasion. 5′-azacytidine treatment additionally regulated BMPER expression and reduced lung fibrosis in mice in vivo. These findings demonstrate that methylation of specific genes in fibroblasts may offer a new therapeutic strategy for IPF by modulating fibroblast activation.
Collapse
Affiliation(s)
- Caijuan Huan
- Department of Respiratory and Critical Care Medicine, Beijing Key Laboratory of Respiratory and Pulmonary Circulation Disorders, Beijing Chao-Yang Hospital-Beijing Institute of Respiratory Medicine, Capital Medical University, Beijing 100020, China.,Cedars-Sinai Medical Center, Department of Medicine, Los Angeles, CA 90048, USA
| | - Ting Yang
- Department of Respiratory and Critical Care Medicine, Beijing Key Laboratory of Respiratory and Pulmonary Circulation Disorders, Beijing Chao-Yang Hospital-Beijing Institute of Respiratory Medicine, Capital Medical University, Beijing 100020, China
| | - Jiurong Liang
- Cedars-Sinai Medical Center, Department of Medicine, Los Angeles, CA 90048, USA
| | - Ting Xie
- Cedars-Sinai Medical Center, Department of Medicine, Los Angeles, CA 90048, USA
| | - Luis Cheng
- Cedars-Sinai Medical Center, Department of Medicine, Los Angeles, CA 90048, USA
| | - Ningshan Liu
- Cedars-Sinai Medical Center, Department of Medicine, Los Angeles, CA 90048, USA
| | - Adrianne Kurkciyan
- Cedars-Sinai Medical Center, Department of Medicine, Los Angeles, CA 90048, USA
| | | | - Chen Wang
- China-Japan Friendship Hospital, Beijing, China
| | - Huaping Dai
- Department of Respiratory and Critical Care Medicine, Beijing Key Laboratory of Respiratory and Pulmonary Circulation Disorders, Beijing Chao-Yang Hospital-Beijing Institute of Respiratory Medicine, Capital Medical University, Beijing 100020, China
| | - Paul W Noble
- Cedars-Sinai Medical Center, Department of Medicine, Los Angeles, CA 90048, USA
| | - Dianhua Jiang
- Cedars-Sinai Medical Center, Department of Medicine, Los Angeles, CA 90048, USA
| |
Collapse
|
18
|
BMPER Promotes Epithelial-Mesenchymal Transition in the Developing Cardiac Cushions. PLoS One 2015; 10:e0139209. [PMID: 26418455 PMCID: PMC4587915 DOI: 10.1371/journal.pone.0139209] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 09/10/2015] [Indexed: 01/25/2023] Open
Abstract
Formation of the cardiac valves is an essential component of cardiovascular development. Consistent with the role of the bone morphogenetic protein (BMP) signaling pathway in cardiac valve formation, embryos that are deficient for the BMP regulator BMPER (BMP-binding endothelial regulator) display the cardiac valve anomaly mitral valve prolapse. However, how BMPER deficiency leads to this defect is unknown. Based on its expression pattern in the developing cardiac cushions, we hypothesized that BMPER regulates BMP2-mediated signaling, leading to fine-tuned epithelial-mesenchymal transition (EMT) and extracellular matrix deposition. In the BMPER-/- embryo, EMT is dysregulated in the atrioventricular and outflow tract cushions compared with their wild-type counterparts, as indicated by a significant increase of Sox9-positive cells during cushion formation. However, proliferation is not impaired in the developing BMPER-/- valves. In vitro data show that BMPER directly binds BMP2. In cultured endothelial cells, BMPER blocks BMP2-induced Smad activation in a dose-dependent manner. In addition, BMP2 increases the Sox9 protein level, and this increase is inhibited by co-treatment with BMPER. Consistently, in the BMPER-/- embryos, semi-quantitative analysis of Smad activation shows that the canonical BMP pathway is significantly more active in the atrioventricular cushions during EMT. These results indicate that BMPER negatively regulates BMP-induced Smad and Sox9 activity during valve development. Together, these results identify BMPER as a regulator of BMP2-induced cardiac valve development and will contribute to our understanding of valvular defects.
Collapse
|
19
|
Esser JS, Rahner S, Deckler M, Bode C, Patterson C, Moser M. Fibroblast Growth Factor Signaling Pathway in Endothelial Cells Is Activated by BMPER to Promote Angiogenesis. Arterioscler Thromb Vasc Biol 2015; 35:358-67. [DOI: 10.1161/atvbaha.114.304345] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Jennifer S. Esser
- From the Department for Cardiology and Angiology, Heart Center University of Freiburg, Albert-Ludwigs University Freiburg, Freiburg, Germany (J.S.E., S.R., M.D., C.B., M.M.); UNC McAllister Heart Institute, Department of Medicine, University of North Carolina, Chapel Hill (C.P.); and New York-Presbyterian Hospital/Weill-Cornell Medical Center, New York, NY (C.P.)
| | - Susanne Rahner
- From the Department for Cardiology and Angiology, Heart Center University of Freiburg, Albert-Ludwigs University Freiburg, Freiburg, Germany (J.S.E., S.R., M.D., C.B., M.M.); UNC McAllister Heart Institute, Department of Medicine, University of North Carolina, Chapel Hill (C.P.); and New York-Presbyterian Hospital/Weill-Cornell Medical Center, New York, NY (C.P.)
| | - Meike Deckler
- From the Department for Cardiology and Angiology, Heart Center University of Freiburg, Albert-Ludwigs University Freiburg, Freiburg, Germany (J.S.E., S.R., M.D., C.B., M.M.); UNC McAllister Heart Institute, Department of Medicine, University of North Carolina, Chapel Hill (C.P.); and New York-Presbyterian Hospital/Weill-Cornell Medical Center, New York, NY (C.P.)
| | - Christoph Bode
- From the Department for Cardiology and Angiology, Heart Center University of Freiburg, Albert-Ludwigs University Freiburg, Freiburg, Germany (J.S.E., S.R., M.D., C.B., M.M.); UNC McAllister Heart Institute, Department of Medicine, University of North Carolina, Chapel Hill (C.P.); and New York-Presbyterian Hospital/Weill-Cornell Medical Center, New York, NY (C.P.)
| | - Cam Patterson
- From the Department for Cardiology and Angiology, Heart Center University of Freiburg, Albert-Ludwigs University Freiburg, Freiburg, Germany (J.S.E., S.R., M.D., C.B., M.M.); UNC McAllister Heart Institute, Department of Medicine, University of North Carolina, Chapel Hill (C.P.); and New York-Presbyterian Hospital/Weill-Cornell Medical Center, New York, NY (C.P.)
| | - Martin Moser
- From the Department for Cardiology and Angiology, Heart Center University of Freiburg, Albert-Ludwigs University Freiburg, Freiburg, Germany (J.S.E., S.R., M.D., C.B., M.M.); UNC McAllister Heart Institute, Department of Medicine, University of North Carolina, Chapel Hill (C.P.); and New York-Presbyterian Hospital/Weill-Cornell Medical Center, New York, NY (C.P.)
| |
Collapse
|
20
|
Chen HI, Poduri A, Numi H, Kivela R, Saharinen P, McKay AS, Raftrey B, Churko J, Tian X, Zhou B, Wu JC, Alitalo K, Red-Horse K. VEGF-C and aortic cardiomyocytes guide coronary artery stem development. J Clin Invest 2014; 124:4899-914. [PMID: 25271623 DOI: 10.1172/jci77483] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Accepted: 08/28/2014] [Indexed: 11/17/2022] Open
Abstract
Coronary arteries (CAs) stem from the aorta at 2 highly stereotyped locations, deviations from which can cause myocardial ischemia and death. CA stems form during embryogenesis when peritruncal blood vessels encircle the cardiac outflow tract and invade the aorta, but the underlying patterning mechanisms are poorly understood. Here, using murine models, we demonstrated that VEGF-C-deficient hearts have severely hypoplastic peritruncal vessels, resulting in delayed and abnormally positioned CA stems. We observed that VEGF-C is widely expressed in the outflow tract, while cardiomyocytes develop specifically within the aorta at stem sites where they surround maturing CAs in both mouse and human hearts. Mice heterozygous for islet 1 (Isl1) exhibited decreased aortic cardiomyocytes and abnormally low CA stems. In hearts with outflow tract rotation defects, misplaced stems were associated with shifted aortic cardiomyocytes, and myocardium induced ectopic connections with the pulmonary artery in culture. These data support a model in which CA stem development first requires VEGF-C to stimulate vessel growth around the outflow tract. Then, aortic cardiomyocytes facilitate interactions between peritruncal vessels and the aorta. Derangement of either step can lead to mispatterned CA stems. Studying this niche for cardiomyocyte development, and its relationship with CAs, has the potential to identify methods for stimulating vascular regrowth as a treatment for cardiovascular disease.
Collapse
|
21
|
Dyer LA, Wu Y, Patterson C. Protein isolation from the developing embryonic mouse heart valve region. J Vis Exp 2014:51911. [PMID: 25285454 DOI: 10.3791/51911] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Western blot analysis is a commonly employed technique for detecting and quantifying protein levels. However, for small tissue samples, this analysis method may not be sufficiently sensitive to detect a protein of interest. To overcome these difficulties, we examined protocols for obtaining protein from adult human cardiac valves and modified these protocols for the developing early embryonic mouse counterparts. In brief, the mouse embryonic aortic valve regions, including the aortic valve and surrounding aortic wall, are collected in the minimal possible volume of a Tris-based lysis buffer with protease inhibitors. If required based on the breeding strategy, embryos are genotyped prior to pooling four embryonic aortic valve regions for homogenization. After homogenization, an SDS-based sample buffer is used to denature the sample for running on an SDS-PAGE gel and subsequent western blot analysis. Although the protein concentration remains too low to quantify using spectrophotometric protein quantification assays and have sample remaining for subsequent analyses, this technique can be used to successfully detect and semi-quantify phosphorylated proteins via western blot from pooled samples of four embryonic day 13.5 mouse aortic valve regions, each of which yields approximately 1 μg of protein. This technique will be of benefit for studying cell signaling pathway activation and protein expression levels during early embryonic mouse valve development.
Collapse
Affiliation(s)
- Laura A Dyer
- McAllister Heart Institute, University of North Carolina at Chapel Hill;
| | - Yaxu Wu
- McAllister Heart Institute, University of North Carolina at Chapel Hill
| | - Cam Patterson
- New York-Presbyterian Hospital/Weill-Cornell Medical Center
| |
Collapse
|
22
|
Kim JD, Lee HW, Jin SW. Diversity is in my veins: role of bone morphogenetic protein signaling during venous morphogenesis in zebrafish illustrates the heterogeneity within endothelial cells. Arterioscler Thromb Vasc Biol 2014; 34:1838-45. [PMID: 25060789 DOI: 10.1161/atvbaha.114.303219] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Endothelial cells are a highly diverse group of cells which display distinct cellular responses to exogenous stimuli. Although the aptly named vascular endothelial growth factor-A signaling pathway is hailed as the most important signaling input for endothelial cells, additional factors also participate in regulating diverse aspects of endothelial behaviors and functions. Given this heterogeneity, these additional factors seem to play a critical role in creating a custom-tailored environment to regulate behaviors and functions of distinct subgroups of endothelial cells. For instance, molecular cues that modulate morphogenesis of arterial vascular beds can be distinct from those that govern morphogenesis of venous vascular beds. Recently, we have found that bone morphogenetic protein signaling selectively promotes angiogenesis from venous vascular beds without eliciting similar responses from arterial vascular beds in zebrafish, indicating that bone morphogenetic protein signaling functions as a context-dependent regulator during vascular morphogenesis. In this review, we will provide an overview of the molecular mechanisms that underlie proangiogenic effects of bone morphogenetic protein signaling on venous vascular beds in the context of endothelial heterogeneity and suggest a more comprehensive picture of the molecular mechanisms of vascular morphogenesis during development.
Collapse
Affiliation(s)
- Jun-Dae Kim
- From the Yale Cardiovascular Research Center, Section of Cardiovascular Medicine (J.-D.K., H.W.L., S.-W.J.) and Department of Internal Medicine (J.-D.K., H.W.L., S.-W.J.), Yale University School of Medicine, New Haven, CT; and School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Korea (S.-W.J.)
| | - Heon-Woo Lee
- From the Yale Cardiovascular Research Center, Section of Cardiovascular Medicine (J.-D.K., H.W.L., S.-W.J.) and Department of Internal Medicine (J.-D.K., H.W.L., S.-W.J.), Yale University School of Medicine, New Haven, CT; and School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Korea (S.-W.J.)
| | - Suk-Won Jin
- From the Yale Cardiovascular Research Center, Section of Cardiovascular Medicine (J.-D.K., H.W.L., S.-W.J.) and Department of Internal Medicine (J.-D.K., H.W.L., S.-W.J.), Yale University School of Medicine, New Haven, CT; and School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Korea (S.-W.J.).
| |
Collapse
|