Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters











Publication year range
1.
Nat Cardiovasc Res ; 3(9): 1035-1048, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39232138

ABSTRACT

Atherosclerotic cardiovascular disease (ASCVD) is the leading cause of mortality worldwide. Laminar shear stress from blood flow, sensed by vascular endothelial cells, protects from ASCVD by upregulating the transcription factors KLF2 and KLF4, which induces an anti-inflammatory program that promotes vascular resilience. Here we identify clustered γ-protocadherins as therapeutically targetable, potent KLF2 and KLF4 suppressors whose upregulation contributes to ASCVD. Mechanistic studies show that γ-protocadherin cleavage results in translocation of the conserved intracellular domain to the nucleus where it physically associates with and suppresses signaling by the Notch intracellular domain. γ-Protocadherins are elevated in human ASCVD endothelium; their genetic deletion or antibody blockade protects from ASCVD in mice without detectably compromising host defense against bacterial or viral infection. These results elucidate a fundamental mechanism of vascular inflammation and reveal a method to target the endothelium rather than the immune system as a protective strategy in ASCVD.


Subject(s)
Atherosclerosis , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors , Atherosclerosis/metabolism , Atherosclerosis/genetics , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Animals , Humans , Disease Models, Animal , Signal Transduction , Cadherins/metabolism , Cadherins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Male , Receptors, Notch/metabolism , Receptors, Notch/genetics , Cadherin Related Proteins , Plaque, Atherosclerotic/metabolism , Plaque, Atherosclerotic/genetics , Plaque, Atherosclerotic/pathology
2.
Nat Cardiovasc Res ; 3(7): 785-798, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39196179

ABSTRACT

Vascular remodeling to match arterial diameter to tissue requirements commonly fails in ischemic disease. Endothelial cells sense fluid shear stress (FSS) from blood flow to maintain FSS within a narrow range in healthy vessels. Thus, high FSS induces vessel outward remodeling, but mechanisms are poorly understood. We previously reported that Smad1/5 is maximally activated at physiological FSS. Smad1/5 limits Akt activation, suggesting that inhibiting Smad1/5 may facilitate outward remodeling. Here we report that high FSS suppresses Smad1/5 by elevating KLF2, which induces the bone morphogenetic protein (BMP) pathway inhibitor, BMP-binding endothelial regulator (BMPER), thereby de-inhibiting Akt. In mice, surgically induced high FSS elevated BMPER expression, inactivated Smad1/5 and induced vessel outward remodeling. Endothelial BMPER deletion impaired blood flow recovery and vascular remodeling. Blocking endothelial cell Smad1/5 activation with BMP9/10 blocking antibodies improved vascular remodeling in mouse models of type 1 and type 2 diabetes. Suppression of Smad1/5 is thus a potential therapeutic approach for ischemic disease.


Subject(s)
Kruppel-Like Transcription Factors , Smad1 Protein , Smad5 Protein , Vascular Remodeling , Animals , Smad5 Protein/metabolism , Smad5 Protein/genetics , Smad1 Protein/metabolism , Smad1 Protein/genetics , Kruppel-Like Transcription Factors/metabolism , Kruppel-Like Transcription Factors/genetics , Vascular Remodeling/physiology , Humans , Stress, Mechanical , Disease Models, Animal , Mice , Mice, Inbred C57BL , Male , Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells , Mice, Knockout , Proto-Oncogene Proteins c-akt/metabolism , Mechanotransduction, Cellular , Cells, Cultured , Signal Transduction
3.
bioRxiv ; 2024 Jan 21.
Article in English | MEDLINE | ID: mdl-38293157

ABSTRACT

Atherosclerotic cardiovascular disease (ASCVD) is the leading cause of mortality worldwide1. Laminar shear stress (LSS) from blood flow in straight regions of arteries protects against ASCVD by upregulating the Klf2/4 anti-inflammatory program in endothelial cells (ECs)2-8. Conversely, disturbed shear stress (DSS) at curves or branches predisposes these regions to plaque formation9,10. We previously reported a whole genome CRISPR knockout screen11 that identified novel inducers of Klf2/4. Here we report suppressors of Klf2/4 and characterize one candidate, protocadherin gamma A9 (Pcdhga9), a member of the clustered protocadherin gene family12. Pcdhg deletion increases Klf2/4 levels in vitro and in vivo and suppresses inflammatory activation of ECs. Pcdhg suppresses Klf2/4 by inhibiting the Notch pathway via physical interaction of cleaved Notch1 intracellular domain (NICD Val1744) with nuclear Pcdhg C-terminal constant domain (CCD). Pcdhg inhibition by EC knockout (KO) or blocking antibody protects from atherosclerosis. Pcdhg is elevated in the arteries of human atherosclerosis. This study identifies a novel fundamental mechanism of EC resilience and therapeutic target for treating inflammatory vascular disease.

4.
bioRxiv ; 2023 Oct 25.
Article in English | MEDLINE | ID: mdl-37961694

ABSTRACT

Fluid shear stress (FSS) from blood flow is sensed by vascular endothelial cells (ECs) to determine vessel stability, remodeling and susceptibility to atherosclerosis and other inflammatory diseases but the regulatory networks that govern these behaviors are only partially understood. We used cSTAR, a powerful new computational method, to define EC transcriptomic states under low shear stress (LSS) that triggers vessel inward remodeling, physiological shear stress (PSS) that stabilizes vessels, high shear stress (HSS) that triggers outward remodeling, and oscillatory shear stress (OSS) that confers disease susceptibility, all in comparison to cells under static conditions (STAT). We combined these results with the LINCS database where EC transcriptomic responses to drug treatments to define a preliminary regulatory network in which the cyclin-dependent kinases CDK1/2 play a central role in promoting vessel stability. Experimental analysis showed that PSS induced a strong late G1 cell cycle arrest in which CDK2 was activated. EC deletion of CDK2 in mice resulted in inward artery remodeling and both pulmonary and systemic hypertension. These results validate use of cSTAR to determine EC state and in vivo vessel behavior, reveal unexpected features of EC phenotype under different FSS conditions, and identify CDK2 as a key element within the EC regulatory network that governs artery remodeling.

5.
Sci Total Environ ; 889: 164173, 2023 Sep 01.
Article in English | MEDLINE | ID: mdl-37201824

ABSTRACT

Microplastic (MP) pollution is a serious global environmental problem, particularly in marine ecosystems. However, the pollution patterns of MPs in the ocean and atmosphere, particularly the sea-air interrelationship, remain unclear. Therefore, the abundance, distribution patterns, and sources of MPs in the seawater and atmosphere of the South China Sea (SCS) were comparatively investigated. The results showed that MPs were prevalent in the SCS with an average abundance of 103.4 ± 98.3 items/m3 in the seawater and 4.62 ± 3.60 items/100 m3 in the atmosphere. The spatial analysis indicated that the pollution patterns of seawater MPs were mainly determined by land-based discharge and sea surface currents, whereas atmospheric MPs were predominantly determined by air parcel trajectory and wind conditions. The highest MP abundance of 490 items/m3 in seawater was found at a station near Vietnam with current vortices. However, the highest MP abundance of 14.6 items/100 m3 in the atmosphere was found in air parcels with low-speed southerly winds from Malaysia. Similar MP compositions (e.g., polyethylene terephthalate, polystyrene, and polyethylene) were observed in the two environmental compartments. Furthermore, similar MP characteristics (e.g., shape, color, and size) in the seawater and atmosphere of the same region suggested a close relationship between the MPs in the two compartments. For this purpose, cluster analysis and calculation of the MP diversity integrated index were performed. The results showed an obvious dispersion between the two compartment clusters and a higher diversity integrated index of MPs in seawater than in the atmosphere, thus implying higher compositional diversity and more complex sources of MPs in seawater relative to the atmosphere. These findings deepen our understanding of MP fate and patterns in the semi-enclosed marginal sea environment and highlight the potential interrelationship of MPs in the air-sea system.


Subject(s)
Microplastics , Water Pollutants, Chemical , Plastics , Ecosystem , Water Pollutants, Chemical/analysis , Environmental Monitoring , Seawater , China , Atmosphere
6.
Biochem Pharmacol ; 206: 115290, 2022 12.
Article in English | MEDLINE | ID: mdl-36241094

ABSTRACT

The endothelium is a mechanosensitive organ whose pleiotropic actions regulate vessel structure to adjust tissue perfusion. To do so, it possesses ion channels, receptor complexes, and signaling pathways responding to blood flow, whose activation will either maintain vascular integrity and quiescence or, on the contrary, remodel the vessel's structure in both health and disease. Recent studies have demonstrated the crucial role of endothelial inflammation, endothelial to mesenchymal transition (EndMT), and perturbed hemodynamics in the progression of pulmonary arterial hypertension and essential hypertension. These two distinct diseases share some common mechanistic cues, pointing towards new potential therapeutic approaches to treat them. In this review, we summarize these common mechanisms to map future drug development strategies targeting flow sensing mechanisms and vascular remodeling.


Subject(s)
Hypertension , Vascular Remodeling , Humans , Vascular Remodeling/physiology , Epithelial-Mesenchymal Transition , Endothelium , Signal Transduction/physiology , Hypertension/drug therapy
7.
J Am Heart Assoc ; 11(14): e025337, 2022 07 19.
Article in English | MEDLINE | ID: mdl-35861829

ABSTRACT

Background Atherosclerosis occurs preferentially in regions of low and disturbed fluid shear stress (FSS) but is limited in regions of high laminar FSS as a result of inhibition of endothelial inflammatory pathways. Recent work has identified endothelial to mesenchymal transition (EndMT) driven by TGFß2 (transforming growth factor beta 2)-Smad2/3 (mothers against decapentaplegic) signaling as a critical component of atherogenesis. However, interactions between FSS and EndMT in this context have not been investigated. Methods and Results Endothelial cells were treated with TGFß2 and inflammatory cytokines (interleukin 1ß and tumor necrosis factor alpha) with or without high FSS in a parallel plate flow chamber. Smad2/3 nuclear translocation and target gene expression, assayed by immunofluorescence and quantitative polymerase chain reaction, revealed that high FSS blocked the Smad2/3-EndMT pathway. In vivo, mice were injected with TGFß2 and inflammatory cytokines, then regions of the aorta under low versus high FSS were examined. TGFß2 and inflammatory cytokine treatment stimulated Smad2/3 nuclear translocation and target gene expression predominantly in regions of low FSS with little effect in regions of high FSS. Conclusions High FSS inhibits endothelial Smad2/3 activation and EndMT in response to inflammatory mediators, resulting in selective EndMT at athero-susceptible, low FSS regions of arteries.


Subject(s)
Cytokines , Endothelial Cells , Animals , Cells, Cultured , Cytokines/metabolism , Endothelial Cells/metabolism , Epithelial-Mesenchymal Transition , Mice , Signal Transduction , Stress, Mechanical
8.
Proc Natl Acad Sci U S A ; 118(51)2021 12 21.
Article in English | MEDLINE | ID: mdl-34911761

ABSTRACT

Arterial remodeling is an important adaptive mechanism that maintains normal fluid shear stress in a variety of physiologic and pathologic conditions. Inward remodeling, a process that leads to reduction in arterial diameter, plays a critical role in progression of such common diseases as hypertension and atherosclerosis. Yet, despite its pathogenic importance, molecular mechanisms controlling inward remodeling remain undefined. Mitogen-activated protein kinases (MAPKs) perform a number of functions ranging from control of proliferation to migration and cell-fate transitions. While the MAPK ERK1/2 signaling pathway has been extensively examined in the endothelium, less is known about the role of the MEKK3/ERK5 pathway in vascular remodeling. To better define the role played by this signaling cascade, we studied the effect of endothelial-specific deletion of its key upstream MAP3K, MEKK3, in adult mice. The gene's deletion resulted in a gradual inward remodeling of both pulmonary and systematic arteries, leading to spontaneous hypertension in both vascular circuits and accelerated progression of atherosclerosis in hyperlipidemic mice. Molecular analysis revealed activation of TGFß-signaling both in vitro and in vivo. Endothelial-specific TGFßR1 knockout prevented inward arterial remodeling in MEKK3 endothelial knockout mice. These data point to the unexpected participation of endothelial MEKK3 in regulation of TGFßR1-Smad2/3 signaling and inward arterial remodeling in artery diseases.


Subject(s)
Hypertension, Pulmonary/pathology , MAP Kinase Kinase Kinase 1/metabolism , MAP Kinase Kinase Kinase 3/metabolism , Transforming Growth Factor beta/metabolism , Vascular Remodeling/physiology , Animals , Gene Deletion , Gene Expression Regulation/drug effects , Genotype , Hindlimb/blood supply , Human Umbilical Vein Endothelial Cells , Humans , Hypertension, Pulmonary/metabolism , Ischemia , MAP Kinase Kinase Kinase 1/genetics , MAP Kinase Kinase Kinase 3/genetics , Mice , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Selective Estrogen Receptor Modulators/toxicity , Signal Transduction , Tamoxifen/toxicity , Transforming Growth Factor beta/genetics
9.
Proc Natl Acad Sci U S A ; 118(37)2021 09 14.
Article in English | MEDLINE | ID: mdl-34504019

ABSTRACT

Endothelial cell (EC) sensing of wall fluid shear stress (FSS) from blood flow governs vessel remodeling to maintain FSS at a specific magnitude or set point in healthy vessels. Low FSS triggers inward remodeling to restore normal FSS but the regulatory mechanisms are unknown. In this paper, we describe the signaling network that governs inward artery remodeling. FSS induces Smad2/3 phosphorylation through the type I transforming growth factor (TGF)-ß family receptor Alk5 and the transmembrane protein Neuropilin-1, which together increase sensitivity to circulating bone morphogenetic protein (BMP)-9. Smad2/3 nuclear translocation and target gene expression but not phosphorylation are maximal at low FSS and suppressed at physiological high shear. Reducing flow by carotid ligation in rodents increases Smad2/3 nuclear localization, while the resultant inward remodeling is blocked by the EC-specific deletion of Alk5. The flow-activated MEKK3/Klf2 pathway mediates the suppression of Smad2/3 nuclear translocation at high FSS, mainly through the cyclin-dependent kinase (CDK)-2-dependent phosphosphorylation of the Smad linker region. Thus, low FSS activates Smad2/3, while higher FSS blocks nuclear translocation to induce inward artery remodeling, specifically at low FSS. These results are likely relevant to inward remodeling in atherosclerotic vessels, in which Smad2/3 is activated through TGF-ß signaling.


Subject(s)
Carotid Arteries/physiology , Carotid Artery Diseases/prevention & control , Endothelial Cells/physiology , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Stress, Mechanical , Vascular Remodeling , Animals , Carotid Arteries/cytology , Carotid Artery Diseases/metabolism , Carotid Artery Diseases/pathology , Endothelial Cells/cytology , Humans , Male , Mice , Mice, Inbred C57BL , Phosphorylation , Signal Transduction , Smad2 Protein/genetics , Smad3 Protein/genetics , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
10.
Nat Commun ; 6: 7937, 2015 Aug 03.
Article in English | MEDLINE | ID: mdl-26235885

ABSTRACT

Cerebral cavernous malformations 2 (CCM2) loss is associated with the familial form of CCM disease. The protein kinase MEKK3 (MAP3K3) is essential for embryonic angiogenesis in mice and interacts physically with CCM2, but how this interaction is mediated and its relevance to cerebral vasculature are unknown. Here we report that Mekk3 plays an intrinsic role in embryonic vascular development. Inducible endothelial Mekk3 knockout in neonatal mice is lethal due to multiple intracranial haemorrhages and brain blood vessels leakage. We discover direct interaction between CCM2 harmonin homology domain (HHD) and the N terminus of MEKK3, and determine a 2.35 Å cocrystal structure. We find Mekk3 deficiency impairs neurovascular integrity, which is partially dependent on Rho-ROCK signalling, and that disruption of MEKK3:CCM2 interaction leads to similar neurovascular leakage. We conclude that CCM2:MEKK3-mediated regulation of Rho signalling is required for maintenance of neurovascular integrity, unravelling a mechanism by which CCM2 loss leads to disease.


Subject(s)
Blood Vessels/embryology , Cerebrovascular Circulation/genetics , Intracranial Hemorrhages/genetics , MAP Kinase Kinase Kinase 3/genetics , Microfilament Proteins/genetics , Neovascularization, Physiologic/genetics , Animals , Animals, Newborn , Blood Vessels/metabolism , Capillary Permeability/genetics , Crystallization , Hemangioma, Cavernous, Central Nervous System/genetics , MAP Kinase Kinase Kinase 3/metabolism , Mice , Mice, Knockout , Microfilament Proteins/metabolism , Signal Transduction , rho GTP-Binding Proteins/metabolism , rho-Associated Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL