Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
1.
Semin Cell Dev Biol ; 155(Pt C): 50-61, 2024 03 01.
Article in English | MEDLINE | ID: mdl-37714806

ABSTRACT

The coronary vasculature consists of a complex hierarchal network of arteries, veins, and capillaries which collectively function to perfuse the myocardium. However, the pathways controlling the temporally and spatially restricted mechanisms underlying the formation of this vascular network remain poorly understood. In recent years, the increasing use and refinement of transgenic mouse models has played an instrumental role in offering new insights into the cellular origins of the coronary vasculature, as well as identifying a continuum of transitioning cell states preceding the full maturation of the coronary vasculature. Coupled with the emergence of single cell RNA sequencing platforms, these technologies have begun to uncover the key regulatory factors mediating the convergence of distinct cellular origins to ensure the formation of a collectively functional, yet phenotypically diverse, vascular network. Furthermore, improved understanding of the key regulatory factors governing coronary vessel formation in the embryo may provide crucial clues into future therapeutic strategies to reactivate these developmentally functional mechanisms to drive the revascularisation of the ischaemic adult heart.


Subject(s)
Coronary Vessels , Neovascularization, Physiologic , Animals , Mice , Coronary Vessels/metabolism , Neovascularization, Physiologic/genetics , Heart , Myocardium/metabolism , Mice, Transgenic
2.
Circulation ; 149(2): 135-154, 2024 01 09.
Article in English | MEDLINE | ID: mdl-38084582

ABSTRACT

BACKGROUND: Endothelial cell (EC) generation and turnover by self-proliferation contributes to vascular repair and regeneration. The ability to accurately measure the dynamics of EC generation would advance our understanding of cellular mechanisms of vascular homeostasis and diseases. However, it is currently challenging to evaluate the dynamics of EC generation in large vessels such as arteries because of their infrequent proliferation. METHODS: By using dual recombination systems based on Cre-loxP and Dre-rox, we developed a genetic system for temporally seamless recording of EC proliferation in vivo. We combined genetic recording of EC proliferation with single-cell RNA sequencing and gene knockout to uncover cellular and molecular mechanisms underlying EC generation in arteries during homeostasis and disease. RESULTS: Genetic proliferation tracing reveals that ≈3% of aortic ECs undergo proliferation per month in adult mice during homeostasis. The orientation of aortic EC division is generally parallel to blood flow in the aorta, which is regulated by the mechanosensing protein Piezo1. Single-cell RNA sequencing analysis reveals 4 heterogeneous aortic EC subpopulations with distinct proliferative activity. EC cluster 1 exhibits transit-amplifying cell features with preferential proliferative capacity and enriched expression of stem cell markers such as Sca1 and Sox18. EC proliferation increases in hypertension but decreases in type 2 diabetes, coinciding with changes in the extent of EC cluster 1 proliferation. Combined gene knockout and proliferation tracing reveals that Hippo/vascular endothelial growth factor receptor 2 signaling pathways regulate EC proliferation in large vessels. CONCLUSIONS: Genetic proliferation tracing quantitatively delineates the dynamics of EC generation and turnover, as well as EC division orientation, in large vessels during homeostasis and disease. An EC subpopulation in the aorta exhibits more robust cell proliferation during homeostasis and type 2 diabetes, identifying it as a potential therapeutic target for vascular repair and regeneration.


Subject(s)
Diabetes Mellitus, Type 2 , Vascular Endothelial Growth Factor A , Animals , Mice , Vascular Endothelial Growth Factor A/metabolism , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Aorta/metabolism , Endothelial Cells/metabolism , Homeostasis , Ion Channels/metabolism
3.
Cell Mol Life Sci ; 81(1): 350, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39141086

ABSTRACT

Heparan sulfate (HS) proteoglycans are important regulators of cellular responses to soluble mediators such as chemokines, cytokines and growth factors. We profiled changes in expression of genes encoding HS core proteins, biosynthesis enzymes and modifiers during macrophage polarisation, and found that the most highly regulated gene was Sulf2, an extracellular HS 6-O-sulfatase that was markedly downregulated in response to pro-inflammatory stimuli. We then generated Sulf2+/- bone marrow chimeric mice and examined inflammatory responses in antigen-induced arthritis, as a model of rheumatoid arthritis. Resolution of inflammation was impaired in myeloid Sulf2+/- chimeras, with elevated joint swelling and increased abundance of pro-arthritic Th17 cells in synovial tissue. Transcriptomic and in vitro analyses indicated that Sulf2 deficiency increased type I interferon signaling in bone marrow-derived macrophages, leading to elevated expression of the Th17-inducing cytokine IL6. This establishes that dynamic remodeling of HS by Sulf2 limits type I interferon signaling in macrophages, and so protects against Th17-driven pathology.


Subject(s)
Macrophages , Mice, Inbred C57BL , Signal Transduction , Th17 Cells , Animals , Th17 Cells/immunology , Th17 Cells/metabolism , Mice , Macrophages/metabolism , Macrophages/immunology , Sulfatases/metabolism , Sulfatases/genetics , Sulfotransferases/metabolism , Sulfotransferases/genetics , Myeloid Cells/metabolism , Myeloid Cells/immunology , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Arthritis, Experimental/metabolism , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Inflammation/metabolism , Inflammation/pathology , Mice, Knockout , Interleukin-6/metabolism , Interleukin-6/genetics , Heparitin Sulfate/metabolism
4.
Circ Res ; 130(3): 352-365, 2022 02 04.
Article in English | MEDLINE | ID: mdl-34995101

ABSTRACT

BACKGROUND: Unraveling how new coronary arteries develop may provide critical information for establishing novel therapeutic approaches to treating ischemic cardiac diseases. There are 2 distinct coronary vascular populations derived from different origins in the developing heart. Understanding the formation of coronary arteries may provide insights into new ways of promoting coronary artery formation after myocardial infarction. METHODS: To understand how intramyocardial coronary arteries are generated to connect these 2 coronary vascular populations, we combined genetic lineage tracing, light sheet microscopy, fluorescence micro-optical sectioning tomography, and tissue-specific gene knockout approaches to understand their cellular and molecular mechanisms. RESULTS: We show that a subset of intramyocardial coronary arteries form by angiogenic extension of endocardium-derived vascular tunnels in the neonatal heart. Three-dimensional whole-mount fluorescence imaging showed that these endocardium-derived vascular tunnels or tubes adopt an arterial fate in neonates. Mechanistically, we implicate Mettl3 (methyltransferase-like protein 3) and Notch signaling in regulating endocardium-derived intramyocardial coronary artery formation. Functionally, these intramyocardial arteries persist into adulthood and play a protective role after myocardial infarction. CONCLUSIONS: A subset of intramyocardial coronary arteries form by extension of endocardium-derived vascular tunnels in the neonatal heart.


Subject(s)
Coronary Vessels/embryology , Endocardium/embryology , Animals , Coronary Vessels/growth & development , Coronary Vessels/metabolism , Endocardium/growth & development , Endocardium/metabolism , Methyltransferases/genetics , Methyltransferases/metabolism , Mice , Mice, Inbred C57BL , Organogenesis
5.
Pharmacol Res ; 209: 107447, 2024 Oct 05.
Article in English | MEDLINE | ID: mdl-39374886

ABSTRACT

BACKGROUND: C-type natriuretic peptide (CNP) is a significant player in the maintenance of cardiac and vascular homeostasis regulating local blood flow, platelet and leukocyte activation, heart structure and function, angiogenesis and metabolic balance. Since such processes are perturbed in myocardial infarction (MI), we explored the role of cardiomyocyte-derived CNP, and pharmacological administration of the peptide, in offsetting the pathological consequences of MI. METHODS: Wild type (WT) and cardiomyocyte-restricted CNP null (cmCNP-/-) mice were subjected to left anterior descending coronary artery (LADCA) ligation and acute effects on infarct size and longer-term outcomes of cardiac repair explored. Heart structure and function were assessed by combined echocardiographic and molecular analyses. Pharmacological administration of CNP (0.2 mg/kg/day; s.c.) was utilized to assess therapeutic potential. RESULTS: Compared to WT littermates, cmCNP-/- mice had a modestly increased infarct size following LADCA ligation but without significant deterioration of cardiac structural and functional indices. However, cmCNP-/- animals exhibited overtly worse heart morphology and contractility 6 weeks following MI, with particularly deleterious reductions in left ventricular ejection fraction, dilatation, fibrosis and revascularization. This phenotype was largely recapitulated in animals with global deletion of natriuretic peptide receptor (NPR)-C (NPR-C-/-). Pharmacological administration of CNP rescued the deleterious pathology in WT and cmCNP-/-, but not NPR-C-/-, animals. CONCLUSIONS AND IMPLICATIONS: Cardiomyocytes synthesize and release CNP as an intrinsic protective mechanism in response to MI that reduces cardiac structural and functional deficits; these salutary actions are primarily NPR-C-dependent. Pharmacological targeting of CNP may represent a new therapeutic option for MI.

6.
Basic Res Cardiol ; 117(1): 17, 2022 03 31.
Article in English | MEDLINE | ID: mdl-35357563

ABSTRACT

Cardiac contractile strength is recognised as being highly pH-sensitive, but less is known about the influence of pH on cardiac gene expression, which may become relevant in response to changes in myocardial metabolism or vascularization during development or disease. We sought evidence for pH-responsive cardiac genes, and a physiological context for this form of transcriptional regulation. pHLIP, a peptide-based reporter of acidity, revealed a non-uniform pH landscape in early-postnatal myocardium, dissipating in later life. pH-responsive differentially expressed genes (pH-DEGs) were identified by transcriptomics of neonatal cardiomyocytes cultured over a range of pH. Enrichment analysis indicated "striated muscle contraction" as a pH-responsive biological process. Label-free proteomics verified fifty-four pH-responsive gene-products, including contractile elements and the adaptor protein CRIP2. Using transcriptional assays, acidity was found to reduce p300/CBP acetylase activity and, its a functional readout, inhibit myocardin, a co-activator of cardiac gene expression. In cultured myocytes, acid-inhibition of p300/CBP reduced H3K27 acetylation, as demonstrated by chromatin immunoprecipitation. H3K27ac levels were more strongly reduced at promoters of acid-downregulated DEGs, implicating an epigenetic mechanism of pH-sensitive gene expression. By tandem cytoplasmic/nuclear pH imaging, the cardiac nucleus was found to exercise a degree of control over its pH through Na+/H+ exchangers at the nuclear envelope. Thus, we describe how extracellular pH signals gain access to the nucleus and regulate the expression of a subset of cardiac genes, notably those coding for contractile proteins and CRIP2. Acting as a proxy of a well-perfused myocardium, alkaline conditions are permissive for expressing genes related to the contractile apparatus.


Subject(s)
Cell Nucleus , Myocardium , Animals , Gene Expression , Mammals , Myocardial Contraction , Myocardium/metabolism , Myocytes, Cardiac/metabolism
7.
Int J Mol Sci ; 23(7)2022 Mar 29.
Article in English | MEDLINE | ID: mdl-35409127

ABSTRACT

Signalling activities are tightly regulated to control cellular responses. Heparan sulfate proteoglycans (HSPGs) at the cell membrane and extracellular matrix regulate ligand availability and interaction with a range of key receptors. SULF1 and SULF2 enzymes modify HSPG sulfation by removing 6-O sulfates to regulate cell signalling but are considered functionally identical. Our in vitro mRNA and protein analyses of two diverse human endothelial cell lines, however, highlight their markedly distinct regulatory roles of maintaining specific HSPG sulfation patterns through feedback regulation of HS 6-O transferase (HS6ST) activities and highly divergent roles in vascular endothelial growth factor (VEGF) and Transforming growth factor ß (TGFß) cell signalling activities. Unlike Sulf2, Sulf1 over-expression in dermal microvascular HMec1 cells promotes TGFß and VEGF cell signalling by simultaneously upregulating HS6ST1 activity. In contrast, Sulf1 over-expression in venous ea926 cells has the opposite effect as it attenuates both TGFß and VEGF signalling while Sulf2 over-expression maintains the control phenotype. Exposure of these cells to VEGF-A, TGFß1, and their inhibitors further highlights their endothelial cell type-specific responses and integral growth factor interactions to regulate cell signalling and selective feedback regulation of HSPG sulfation that additionally exploits alternative Sulf2 RNA-splicing to regulate net VEGF-A and TGFß cell signalling activities.


Subject(s)
Sulfotransferases , Vascular Endothelial Growth Factor A , Endothelial Cells/metabolism , Heparan Sulfate Proteoglycans , Sulfotransferases/genetics , Sulfotransferases/metabolism , Transforming Growth Factor beta , Vascular Endothelial Growth Factor A/genetics
8.
Circ Res ; 123(1): 86-99, 2018 06 22.
Article in English | MEDLINE | ID: mdl-29764841

ABSTRACT

RATIONALE: Organs of the body require vascular networks to supply oxygen and nutrients and maintain physiological function. The blood vessels of different organs are structurally and functionally heterogeneous in nature. To more precisely dissect their distinct in vivo function in individual organs, without potential interference from off-site targets, it is necessary to genetically target them in an organ-specific manner. OBJECTIVE: The objective of this study was to generate a genetic system that targets vascular endothelial cells in an organ- or tissue-specific manner and to exemplify the potential application of intersectional genetics for precise, target-specific gene manipulation in vivo. METHODS AND RESULTS: We took advantage of 2 orthogonal recombination systems, Dre-rox and Cre-loxP, to create a genetic targeting system based on intersectional genetics. Using this approach, Cre activity was only detectable in cells that had expressed both Dre and Cre. Applying this new system, we generated a coronary endothelial cell-specific Cre (CoEC-Cre) and a brain endothelial cell-specific Cre (BEC-Cre). Through lineage tracing, gene knockout and overexpression experiments, we demonstrated that CoEC-Cre and BEC-Cre efficiently and specifically target blood vessels in the heart and brain, respectively. By deletion of vascular endothelial growth factor receptor 2 using BEC-Cre, we showed that vascular endothelial growth factor signaling regulates angiogenesis in the central nervous system and also controls the integrity of the blood-brain barrier. CONCLUSIONS: We provide 2 examples to illustrate the use of intersectional genetics for more precise gene targeting in vivo, namely manipulation of genes in blood vessels of the heart and brain. More broadly, this system provides a valuable strategy for tissue-specific gene manipulation that can be widely applied to other fields of biomedical research.


Subject(s)
Blood Vessels , Brain/blood supply , Coronary Vessels , Gene Targeting/methods , Animals , Blood-Brain Barrier , Cell Hypoxia , Endothelial Cells , Gene Knockout Techniques , In Situ Hybridization/methods , Mice , Neovascularization, Physiologic , Organ Specificity , Receptors, Vascular Endothelial Growth Factor/physiology , Vascular Endothelial Growth Factor A/physiology
9.
J Mol Cell Cardiol ; 102: 94-107, 2017 01.
Article in English | MEDLINE | ID: mdl-27914791

ABSTRACT

Sarcomere assembly is a highly orchestrated and dynamic process which adapts, during perinatal development, to accommodate growth of the heart. Sarcomeric components, including titin, undergo an isoform transition to adjust ventricular filling. Many sarcomeric genes have been implicated in congenital cardiomyopathies, such that understanding developmental sarcomere transitions will inform the aetiology and treatment. We sought to determine whether Thymosin ß4 (Tß4), a peptide that regulates the availability of actin monomers for polymerization in non-muscle cells, plays a role in sarcomere assembly during cardiac morphogenesis and influences adult cardiac function. In Tß4 null mice, immunofluorescence-based sarcomere analyses revealed shortened thin filament, sarcomere and titin spring length in cardiomyocytes, associated with precocious up-regulation of the short titin isoforms during the postnatal splicing transition. By magnetic resonance imaging, this manifested as diminished stroke volume and limited contractile reserve in adult mice. Extrapolating to an in vitro cardiomyocyte model, the altered postnatal splicing was corrected with addition of synthetic Tß4, whereby normal sarcomere length was restored. Our data suggest that Tß4 is required for setting correct sarcomere length and for appropriate splicing of titin, not only in the heart but also in skeletal muscle. Distinguishing between thin filament extension and titin splicing as the primary defect is challenging, as these events are intimately linked. The regulation of titin splicing is a previously unrecognised role of Tß4 and gives preliminary insight into a mechanism by which titin isoforms may be manipulated to correct cardiac dysfunction.


Subject(s)
Connectin/genetics , RNA Splicing , Sarcomeres/metabolism , Thymosin/deficiency , Animals , Echocardiography , Heart/diagnostic imaging , Heart/physiopathology , Hemodynamics , Male , Mice , Mice, Knockout , Myocardial Contraction/genetics , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/metabolism , Sarcomeres/ultrastructure
10.
Microcirculation ; 24(1)2017 01.
Article in English | MEDLINE | ID: mdl-27859990

ABSTRACT

Neovascularization of the ischemic myocardium postinfarction is necessary to restore blood flow to vulnerable cardiomyocytes and will be indispensable for prospective regenerative strategies, to perfuse newly formed myocardium. Therapeutic attempts to enhance new vessel formation have, to date, yielded modest clinical benefits, and innovative approaches are now needed. Intrinsic mechanisms are initiated by the heart in an attempt to rebuild injured vessels, but these are poorly understood. Insight into the underlying mechanisms may reveal targets for therapeutically augmenting this low-level neovascular response. Starting from a limited number of descriptive studies, this review summarizes what is known of coronary neovascularization and explores putative mechanisms and cellular sources which may endogenously contribute, or that may be pharmacologically triggered, to support vasculo- or angiogenesis. As injury responses in the adult frequently recapitulate embryological processes, a particular focus is placed on the developmental mechanisms of coronary vessel formation. An understanding of the cellular sources and the regulatory pathways used by the embryo may reveal novel targets for reactivating coronary vessel and myocardial regeneration.


Subject(s)
Myocardial Ischemia/therapy , Neovascularization, Physiologic , Animals , Coronary Vessels/embryology , Coronary Vessels/growth & development , Humans , Regeneration
11.
Nature ; 474(7353): 640-4, 2011 Jun 08.
Article in English | MEDLINE | ID: mdl-21654746

ABSTRACT

A significant bottleneck in cardiovascular regenerative medicine is the identification of a viable source of stem/progenitor cells that could contribute new muscle after ischaemic heart disease and acute myocardial infarction. A therapeutic ideal--relative to cell transplantation--would be to stimulate a resident source, thus avoiding the caveats of limited graft survival, restricted homing to the site of injury and host immune rejection. Here we demonstrate in mice that the adult heart contains a resident stem or progenitor cell population, which has the potential to contribute bona fide terminally differentiated cardiomyocytes after myocardial infarction. We reveal a novel genetic label of the activated adult progenitors via re-expression of a key embryonic epicardial gene, Wilm's tumour 1 (Wt1), through priming by thymosin ß4, a peptide previously shown to restore vascular potential to adult epicardium-derived progenitor cells with injury. Cumulative evidence indicates an epicardial origin of the progenitor population, and embryonic reprogramming results in the mobilization of this population and concomitant differentiation to give rise to de novo cardiomyocytes. Cell transplantation confirmed a progenitor source and chromosome painting of labelled donor cells revealed transdifferentiation to a myocyte fate in the absence of cell fusion. Derived cardiomyocytes are shown here to structurally and functionally integrate with resident muscle; as such, stimulation of this adult progenitor pool represents a significant step towards resident-cell-based therapy in human ischaemic heart disease.


Subject(s)
Adult Stem Cells/cytology , Cell Differentiation , Heart Injuries , Myocytes, Cardiac/cytology , Animals , Cellular Reprogramming , Gene Expression Regulation , Mice , Myocardial Infarction/pathology , Myocytes, Cardiac/metabolism , Thymosin/metabolism , WT1 Proteins/genetics , WT1 Proteins/metabolism
12.
Blood ; 124(15): e33-44, 2014 Oct 09.
Article in English | MEDLINE | ID: mdl-25030063

ABSTRACT

The recruitment of monocytes and their differentiation into macrophages at sites of inflammation are key events in determining the outcome of the inflammatory response and initiating the return to tissue homeostasis. To study monocyte trafficking and macrophage differentiation in vivo, we have generated a novel transgenic reporter mouse expressing a green fluorescent protein (GFP) under the control of the human CD68 promoter. CD68-GFP mice express high levels of GFP in both monocyte and embryo-derived tissue resident macrophages in adult animals. The human CD68 promoter drives GFP expression in all CD115(+) monocytes of adult blood, spleen, and bone marrow; we took advantage of this to directly compare the trafficking of bone marrow-derived CD68-GFP monocytes to that of CX3CR1(GFP) monocytes in vivo using a sterile zymosan peritonitis model. Unlike CX3CR1(GFP) monocytes, which downregulate GFP expression on differentiation into macrophages in this model, CD68-GFP monocytes retain high-level GFP expression for 72 hours after differentiation into macrophages, allowing continued cell tracking during resolution of inflammation. In summary, this novel CD68-GFP transgenic reporter mouse line represents a powerful resource for analyzing monocyte mobilization and monocyte trafficking as well as studying the fate of recruited monocytes in models of acute and chronic inflammation.


Subject(s)
Antigens, CD/genetics , Antigens, Differentiation, Myelomonocytic/genetics , Cell Differentiation , Green Fluorescent Proteins/metabolism , Macrophages, Peritoneal/cytology , Monocytes/cytology , Promoter Regions, Genetic/genetics , Adoptive Transfer , Animals , Bone Marrow/metabolism , CX3C Chemokine Receptor 1 , Chronic Disease , Embryonic Development , Flow Cytometry , Fluorescent Antibody Technique , Genes, Reporter , Humans , Inflammation/pathology , Leukocytes/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Mycobacterium Infections/pathology , Mycobacterium bovis/physiology , Phenotype , Receptors, Chemokine/metabolism , Spleen/metabolism
14.
Nat Cell Biol ; 9(10): 1131-41, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17891141

ABSTRACT

The bHLH transcription factor Hand1 is essential for placentation and cardiac morphogenesis in the developing embryo. Here we implicate Hand1 as a molecular switch that determines whether a trophoblast stem cell continues to proliferate or commits to differentiation. We identify a novel interaction of Hand1 with a protein that contains an I-mfa (inhibitor of myogenic factor) domain that anchors Hand1 in the nucleolus where it negatively regulates Hand1 activity. In the trophoblast stem-cell line Rcho-1, nucleolar sequestration of Hand1 accompanies sustained cell proliferation and renewal, whereas release of Hand1 into the nucleus leads to its activation, thus committing cells to a differentiated giant-cell fate. Site-specific phosphorylation is required for nucleolar release of Hand1, for its dimerization and biological function, and this is mediated by the non-canonical polo-like kinase Plk4 (Sak). Sak is co-expressed in Rcho-1 cells, localizes to the nucleolus during G2 and phosphorylates Hand1 as a requirement for trophoblast stem-cell commitment to a giant-cell fate. This study defines a novel cellular mechanism for regulating Hand1 that is a crucial step in the stem-cell differentiation pathway.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation , Cell Nucleolus/metabolism , Stem Cells/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/physiology , Blotting, Northern , Blotting, Western , Cell Proliferation , Gene Expression Regulation, Developmental , Giant Cells/cytology , Giant Cells/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Immunoprecipitation , Mice , Myogenic Regulatory Factors/metabolism , NIH 3T3 Cells , Phosphorylation , Protein Binding , Protein Phosphatase 2 , Protein Serine-Threonine Kinases/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/cytology , Trophoblasts/cytology , Trophoblasts/metabolism , Two-Hybrid System Techniques
15.
Circ Res ; 111(4): e89-102, 2012 Aug 03.
Article in English | MEDLINE | ID: mdl-22723298

ABSTRACT

RATIONALE: Compromised development of blood vessel walls leads to vascular instability that may predispose to aneurysm with risk of rupture and lethal hemorrhage. There is currently a lack of insight into developmental insults that may define the molecular and cellular characteristics of initiating and perpetrating factors in adult aneurismal disease. OBJECTIVE: To investigate a role for the actin-binding protein thymosin ß4 (Tß4), previously shown to be proangiogenic, in mural cell development and vascular wall stability. METHODS AND RESULTS: Phenotypic analyses of both global and endothelial-specific loss-of-function Tß4 mouse models revealed a proportion of Tß4-null embryos with vascular hemorrhage coincident with a reduction in smooth muscle cell coverage of their developing vessels. Mechanistic studies revealed that extracellular Tß4 can stimulate differentiation of mesodermal progenitor cells to a mature mural cell phenotype through activation of the transforming growth factor-beta (TGFß) pathway and that reduced TGFß signaling correlates with the severity of hemorrhagic phenotype in Tß4-null vasculature. CONCLUSIONS: Tß4 is a novel endothelial secreted trophic factor that functions synergistically with TGFß to regulate mural cell development and vascular wall stability. These findings have important implications for understanding congenital anomalies that may be causative for adult-onset vascular instability.


Subject(s)
Endothelial Cells/metabolism , Hemorrhage/etiology , Mesenchymal Stem Cells/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Paracrine Communication , Thymosin/metabolism , Animals , Aorta/metabolism , Aorta/pathology , Cell Differentiation , Cells, Cultured , Coculture Techniques , Endothelial Cells/pathology , Genes, Reporter , Genotype , Gestational Age , Hemorrhage/metabolism , Hemorrhage/pathology , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Humans , Mesenchymal Stem Cells/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/embryology , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Phenotype , Signal Transduction , Smad Proteins/metabolism , Thymosin/deficiency , Thymosin/genetics , Transfection , Transforming Growth Factor beta/metabolism
16.
Circ Res ; 111(2): e19-31, 2012 Jul 06.
Article in English | MEDLINE | ID: mdl-22647876

ABSTRACT

RATIONALE: Nkx2.5 is one of the most widely studied cardiac-specific transcription factors, conserved from flies to man, with multiple essential roles in both the developing and adult heart. Specific dominant mutations in NKX2.5 have been identified in adult congenital heart disease patients presenting with conduction system anomalies and recent genome-wide association studies implicate the NKX2.5 locus, as causative for lethal arrhythmias ("sudden cardiac death") that occur at a frequency in the population of 1 in 1000 per annum worldwide. Haploinsufficiency for Nkx2.5 in the mouse phenocopies human conduction disease pathology yet the phenotypes, described in both mouse and man, are highly pleiotropic, implicit of unknown modifiers and/or factors acting in epistasis with Nkx2.5/NKX2.5. OBJECTIVE: To identify bone fide upstream genetic modifier(s) of Nkx2.5/NKX2.5 function and to determine epistatic effects relevant to the manifestation of NKX2.5-dependent adult congenital heart disease. METHODS AND RESULTS: A study of cardiac function in prospero-related homeobox protein 1 (Prox1) heterozygous mice, using pressure-volume loop and micromannometry, revealed rescue of hemodynamic parameters in Nkx2.5(Cre/+); Prox1(loxP/+) animals versus Nkx2.5(Cre/+) controls. Anatomic studies, on a Cx40(EGFP) background, revealed Cre-mediated knock-down of Prox1 restored the anatomy of the atrioventricular node and His-Purkinje network both of which were severely hypoplastic in Nkx2.5(Cre/+) littermates. Steady state surface electrocardiography recordings and high-speed multiphoton imaging, to assess Ca(2+) handling, revealed atrioventricular conduction and excitation-contraction were also normalized by Prox1 haploinsufficiency, as was expression of conduction genes thought to act downstream of Nkx2.5. Chromatin immunoprecipitation on adult hearts, in combination with both gain and loss-of-function reporter assays in vitro, revealed that Prox1 recruits the corepressor HDAC3 to directly repress Nkx2.5 via a proximal upstream enhancer as a mechanism for regulating Nkx2.5 function in adult cardiac conduction. CONCLUSIONS: Here we identify Prox1 as a direct upstream modifier of Nkx2.5 in the maintenance of the adult conduction system and rescue of Nkx2.5 conduction disease phenotypes. This study is the first example of rescue of Nkx2.5 function and establishes a model for ensuring electrophysiological function within the adult heart alongside insight into a novel Prox1-HDAC3-Nkx2.5 signaling pathway for therapeutic targeting in conduction disease.


Subject(s)
Epistasis, Genetic/genetics , Heart Conduction System/physiopathology , Heart Diseases/genetics , Heart Diseases/metabolism , Histone Deacetylases/genetics , Homeodomain Proteins/genetics , Phenotype , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , Animals , Heart Diseases/physiopathology , Histone Deacetylases/physiology , Homeobox Protein Nkx-2.5 , Homeodomain Proteins/physiology , Mice , Mice, Transgenic , NIH 3T3 Cells , Transcription Factors/physiology , Tumor Suppressor Proteins/physiology
17.
Nature ; 445(7124): 177-82, 2007 Jan 11.
Article in English | MEDLINE | ID: mdl-17108969

ABSTRACT

Cardiac failure has a principal underlying aetiology of ischaemic damage arising from vascular insufficiency. Molecules that regulate collateral growth in the ischaemic heart also regulate coronary vasculature formation during embryogenesis. Here we identify thymosin beta4 (Tbeta4) as essential for all aspects of coronary vessel development in mice, and demonstrate that Tbeta4 stimulates significant outgrowth from quiescent adult epicardial explants, restoring pluripotency and triggering differentiation of fibroblasts, smooth muscle cells and endothelial cells. Tbeta4 knockdown in the heart is accompanied by significant reduction in the pro-angiogenic cleavage product N-acetyl-seryl-aspartyl-lysyl-proline (AcSDKP). Although injection of AcSDKP was unable to rescue Tbeta4 mutant hearts, it significantly enhanced endothelial cell differentiation from adult epicardially derived precursor cells. This study identifies Tbeta4 and AcSDKP as potent stimulators of coronary vasculogenesis and angiogenesis, and reveals Tbeta4-induced adult epicardial cells as a viable source of vascular progenitors for continued renewal of regressed vessels at low basal level or sustained neovascularization following cardiac injury.


Subject(s)
Cell Movement , Neovascularization, Physiologic , Pericardium/cytology , Pericardium/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Thymosin/metabolism , Animals , Cell Differentiation , Cell Lineage , Coronary Vessels/cytology , Coronary Vessels/embryology , Coronary Vessels/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Female , Mice , Myoblasts, Cardiac/cytology , Myoblasts, Cardiac/metabolism , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Organ Specificity , Pericardium/embryology , Pregnancy , Thymosin/deficiency , Thymosin/genetics
18.
Nat Genet ; 34(2): 203-8, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12766769

ABSTRACT

Fraser syndrome (OMIM 219000) is a multisystem malformation usually comprising cryptophthalmos, syndactyly and renal defects. Here we report autozygosity mapping and show that the locus FS1 at chromosome 4q21 is associated with Fraser syndrome, although the condition is genetically heterogeneous. Mutation analysis identified five frameshift mutations in FRAS1, which encodes one member of a family of novel proteins related to an extracellular matrix (ECM) blastocoelar protein found in sea urchin. The FRAS1 protein contains a series of N-terminal cysteine-rich repeat motifs previously implicated in BMP metabolism, suggesting that it has a role in both structure and signal propagation in the ECM. It has been speculated that Fraser syndrome is a human equivalent of the blebbed phenotype in the mouse, which has been associated with mutations in at least five loci including bl. As mapping data were consistent with homology of FRAS1 and bl, we screened DNA from bl/bl mice and identified a premature termination of mouse Fras1. Thus, the bl mouse is a model for Fraser syndrome in humans, a disorder caused by disrupted epithelial integrity in utero.


Subject(s)
Blister/genetics , Denys-Drash Syndrome/genetics , Extracellular Matrix Proteins/genetics , Animals , Base Sequence , Blister/pathology , Chromosomes, Human, Pair 4/genetics , DNA/genetics , DNA Mutational Analysis , Denys-Drash Syndrome/pathology , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Inbred Strains , Mice, Mutant Strains , Molecular Sequence Data , Pedigree , Phenotype
19.
Int Immunopharmacol ; 115: 109702, 2023 Feb.
Article in English | MEDLINE | ID: mdl-37724952

ABSTRACT

Atherosclerosis is a progressive, degenerative vascular disease and a leading cause of morbidity and mortality. In response to endothelial damage, platelet derived growth factor (PDGF)-BB induced phenotypic modulation of medial smooth muscle cells (VSMCs) promotes atherosclerotic lesion formation and destabilisation of the vessel wall. VSMC sensitivity to PDGF-BB is determined by endocytosis of Low density lipoprotein receptor related protein 1 (LRP1)-PDGFR ß complexes to balance receptor recycling with lysosomal degradation. Consequently, LRP1 is implicated in various arterial diseases. Having identified Tß4 as a regulator of LRP1-mediated endocytosis to protect against aortic aneurysm, we sought to determine whether Tß4 may additionally function to protect against atherosclerosis, by regulating LRP1-mediated growth factor signalling. By single cell transcriptomic analysis, Tmsb4x, encoding Tß4, strongly correlated with contractile gene expression and was significantly down-regulated in cells that adopted a modulated phenotype in atherosclerosis. We assessed susceptibility to atherosclerosis of global Tß4 knockout mice using the ApoE-/- hypercholesterolaemia model. Inflammation, elastin integrity, VSMC phenotype and signalling were analysed in the aortic root and descending aorta. Tß4KO; ApoE-/- mice develop larger atherosclerotic plaques than control mice, with medial layer degeneration characterised by accelerated VSMC phenotypic modulation. Defects in Tß4KO; ApoE-/- mice phenocopied those in VSMC-specific LRP1 nulls and, moreover, were underpinned by hyperactivated LRP1-PDGFRß signalling. We identify an atheroprotective role for endogenous Tß4 in maintaining differentiated VSMC phenotype via LRP1-mediated PDGFRß signalling.


Subject(s)
Atherosclerosis , Muscle, Smooth, Vascular , Animals , Mice , Apolipoproteins E/genetics , Becaplermin , LDL-Receptor Related Proteins , Lipoproteins, LDL
20.
Curr Opin Physiol ; 35: None, 2023 Oct.
Article in English | MEDLINE | ID: mdl-38328689

ABSTRACT

The complex and hierarchical vascular network of arteries, veins, and capillaries features considerable endothelial heterogeneity, yet the regulatory pathways directing arteriovenous specification, differentiation, and identity are still not fully understood. Recent advances in analysis of endothelial-specific gene-regulatory elements, single-cell RNA sequencing, and cell lineage tracing have both emphasized the importance of transcriptional regulation in this process and shed considerable light on the mechanism and regulation of specification within the endothelium. In this review, we discuss recent advances in our understanding of how endothelial cells acquire arterial and venous identity and the role different transcription factors play in this process.

SELECTION OF CITATIONS
SEARCH DETAIL