Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Proc Natl Acad Sci U S A ; 118(27)2021 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-34183396

RESUMO

The onset of venous thromboembolism, including pulmonary embolism, represents a significant health burden affecting more than 1 million people annually worldwide. Current treatment options are based on anticoagulation, which is suboptimal for preventing further embolic events. In order to develop better treatments for thromboembolism, we sought to understand the structural and mechanical properties of blood clots and how this influences embolism in vivo. We developed a murine model in which fibrin γ-chain cross-linking by activated Factor XIII is eliminated (FGG3X) and applied methods to study thromboembolism at whole-body and organ levels. We show that FGG3X mice have a normal phenotype, with overall coagulation parameters and platelet aggregation and function largely unaffected, except for total inhibition of fibrin γ-chain cross-linking. Elimination of fibrin γ-chain cross-linking resulted in thrombi with reduced strength that were prone to fragmentation. Analysis of embolism in vivo using Xtreme optical imaging and light sheet microscopy demonstrated that the elimination of fibrin γ-chain cross-linking resulted in increased embolization without affecting clot size or lysis. Our findings point to a central previously unrecognized role for fibrin γ-chain cross-linking in clot stability. They also indirectly indicate mechanistic targets for the prevention of thrombosis through selective modulation of fibrin α-chain but not γ-chain cross-linking by activated Factor XIII to reduce thrombus size and burden, while maintaining clot stability and preventing embolism.


Assuntos
Reagentes de Ligações Cruzadas/química , Fator XIIIa/metabolismo , Fibrinogênio/metabolismo , Embolia Pulmonar/etiologia , Embolia Pulmonar/patologia , Veia Cava Inferior/patologia , Trombose Venosa/complicações , Animais , Coagulação Sanguínea , Plaquetas/metabolismo , Modelos Animais de Doenças , Camundongos Endogâmicos C57BL , Imagem Óptica , Embolia Pulmonar/sangue , Trombose Venosa/sangue
2.
Am J Respir Crit Care Med ; 205(8): 903-916, 2022 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-35044899

RESUMO

Rationale: Patients with chronic obstructive pulmonary disease (COPD) experience excess cardiovascular morbidity and mortality, and exacerbations further increase the risk of such events. COPD is associated with persistent blood and airway neutrophilia and systemic and tissue hypoxia. Hypoxia augments neutrophil elastase release, enhancing capacity for tissue injury. Objective: To determine whether hypoxia-driven neutrophil protein secretion contributes to endothelial damage in COPD. Methods: The healthy human neutrophil secretome generated under normoxic or hypoxic conditions was characterized by quantitative mass spectrometry, and the capacity for neutrophil-mediated endothelial damage was assessed. Histotoxic protein concentrations were measured in normoxic versus hypoxic neutrophil supernatants and plasma from patients experiencing COPD exacerbation and healthy control subjects. Measurements and Main Results: Hypoxia promoted PI3Kγ-dependent neutrophil elastase secretion, with greater release seen in neutrophils from patients with COPD. Supernatants from neutrophils incubated under hypoxia caused pulmonary endothelial cell damage, and identical supernatants from COPD neutrophils increased neutrophil adherence to endothelial cells. Proteomics revealed differential neutrophil protein secretion under hypoxia and normoxia, and hypoxia augmented secretion of a subset of histotoxic granule and cytosolic proteins, with significantly greater release seen in COPD neutrophils. The plasma of patients with COPD had higher content of hypoxia-upregulated neutrophil-derived proteins and protease activity, and vascular injury markers. Conclusions: Hypoxia drives a destructive "hypersecretory" neutrophil phenotype conferring enhanced capacity for endothelial injury, with a corresponding signature of neutrophil degranulation and vascular injury identified in plasma of patients with COPD. Thus, hypoxic enhancement of neutrophil degranulation may contribute to increased cardiovascular risk in COPD. These insights may identify new therapeutic opportunities for endothelial damage in COPD.


Assuntos
Doença Pulmonar Obstrutiva Crônica , Lesões do Sistema Vascular , Células Endoteliais/metabolismo , Humanos , Hipóxia/metabolismo , Elastase de Leucócito/metabolismo , Neutrófilos/metabolismo , Doença Pulmonar Obstrutiva Crônica/metabolismo , Lesões do Sistema Vascular/metabolismo
3.
Int J Mol Sci ; 23(11)2022 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-35682592

RESUMO

(1) Background: Systemic infection is associated with increased neuroinflammation and accelerated cognitive decline in AD patients. Activated neutrophils produce neutrophil-derived microvesicles (NMV), which are internalised by human brain microvascular endothelial cells and increase their permeability in vitro, suggesting that NMV play a role in blood-brain barrier (BBB) integrity during infection. The current study investigated whether microRNA content of NMV from AD patients is significantly different compared to healthy controls and could impact cerebrovascular integrity. (2) Methods: Neutrophils isolated from peripheral blood samples of five AD and five healthy control donors without systemic infection were stimulated to produce NMV. MicroRNAs isolated from NMV were analysed by RNA-Seq, and online bioinformatic tools were used to identify significantly differentially expressed microRNAs in the NMV. Target and pathway analyses were performed to predict the impact of the candidate microRNAs on vascular integrity. (3) Results: There was no significant difference in either the number of neutrophils (p = 0.309) or the number of NMV (p = 0.3434) isolated from AD donors compared to control. However, 158 microRNAs were significantly dysregulated in AD NMV compared to controls, some of which were associated with BBB dysfunction, including miR-210, miR-20b-5p and miR-126-5p. Pathway analysis revealed numerous significantly affected pathways involved in regulating vascular integrity, including the TGFß and PDGFB pathways, as well as Hippo, IL-2 and DNA damage signalling. (4) Conclusions: NMV from AD patients contain miRNAs that may alter the integrity of the BBB and represent a novel neutrophil-mediated mechanism for BBB dysfunction in AD and the accelerated cognitive decline seen as a result of a systemic infection.


Assuntos
Doença de Alzheimer , MicroRNAs , Doença de Alzheimer/metabolismo , Barreira Hematoencefálica/metabolismo , Células Endoteliais/metabolismo , Humanos , MicroRNAs/metabolismo , Neutrófilos/metabolismo , RNA-Seq
4.
J Cell Sci ; 132(11)2019 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-31076511

RESUMO

Endothelial cell (EC) sensing of fluid shear stress direction is a critical determinant of vascular health and disease. Unidirectional flow induces EC alignment and vascular homeostasis, whereas bidirectional flow has pathophysiological effects. ECs express several mechanoreceptors that respond to flow, but the mechanism for sensing shear stress direction is poorly understood. We determined, by using in vitro flow systems and magnetic tweezers, that ß1 integrin is a key sensor of force direction because it is activated by unidirectional, but not bidirectional, shearing forces. ß1 integrin activation by unidirectional force was amplified in ECs that were pre-sheared in the same direction, indicating that alignment and ß1 integrin activity has a feedforward interaction, which is a hallmark of system stability. En face staining and EC-specific genetic deletion studies in the murine aorta revealed that ß1 integrin is activated and is essential for EC alignment at sites of unidirectional flow but is not activated at sites of bidirectional flow. In summary, ß1 integrin sensing of unidirectional force is a key mechanism for decoding blood flow mechanics to promote vascular homeostasis.This article has an associated First Person interview with the first author of the paper.


Assuntos
Aorta/fisiologia , Integrina beta1/metabolismo , Fluxo Sanguíneo Regional/fisiologia , Animais , Linhagem Celular , Feminino , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Integrina beta1/genética , Mecanorreceptores/fisiologia , Camundongos , Camundongos Knockout , Estresse Fisiológico/fisiologia
5.
Int J Mol Sci ; 20(20)2019 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-31652502

RESUMO

The blood-brain barrier (BBB), composed of brain microvascular endothelial cells (BMEC) that are tightly linked by tight junction (TJ) proteins, restricts the movement of molecules between the periphery and the central nervous system. Elevated systemic levels of neutrophils have been detected in patients with altered BBB function, but the role of neutrophils in BMEC dysfunction is unknown. Neutrophils are key players of the immune response and, when activated, produce neutrophil-derived microvesicles (NMV). NMV have been shown to impact the integrity of endothelial cells throughout the body and we hypothesize that NMV released from circulating neutrophils interact with BMEC and induce endothelial cell dysfunction. Therefore, the current study investigated the interaction of NMV with human BMEC and determined whether they altered gene expression and function in vitro. Using flow cytometry and confocal imaging, NMV were shown to be internalized by the human cerebral microvascular endothelial cell line hCMEC/D3 via a variety of energy-dependent mechanisms, including endocytosis and macropinocytosis. The internalization of NMV significantly altered the transcriptomic profile of hCMEC/D3, specifically inducing the dysregulation of genes associated with TJ, ubiquitin-mediated proteolysis and vesicular transport. Functional studies confirmed NMV significantly increased permeability and decreased the transendothelial electrical resistance (TEER) of a confluent monolayer of hCMEC/D3. These findings indicate that NMV interact with and affect gene expression of BMEC as well as impacting their integrity. We conclude that NMV may play an important role in modulating the permeability of BBB during an infection.


Assuntos
Barreira Hematoencefálica/metabolismo , Células Endoteliais/metabolismo , Vesículas Extracelulares/metabolismo , Neutrófilos/metabolismo , Barreira Hematoencefálica/citologia , Permeabilidade Capilar , Células Cultivadas , Endocitose , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Humanos
6.
Blood ; 127(4): 487-95, 2016 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-26608329

RESUMO

Previous studies have shown effects of thrombin and fibrinogen γ' on clot structure. However, structural information was obtained using electron microscopy, which requires sample dehydration. Our aim was to investigate the role of thrombin and fibrinogen γ' in modulating fibrin structure under fully hydrated conditions. Fibrin fibers were studied using turbidimetry, atomic force microscopy, electron microscopy, and magnetic tweezers in purified and plasma solutions. Increased thrombin induced a pronounced decrease in average protofibril content per fiber, with a relatively minor decrease in fiber size, leading to the formation of less compact fiber structures. Atomic force microscopy under fully hydrated conditions confirmed that fiber diameter was only marginally decreased. Decreased protofibril content of the fibers produced by high thrombin resulted in weakened clot architecture as analyzed by magnetic tweezers in purified systems and by thromboelastometry in plasma and whole blood. Fibers produced with fibrinogen γ' showed reduced protofibril packing over a range of thrombin concentrations. High-magnification electron microscopy demonstrated reduced protofibril packing in γ' fibers and unraveling of fibers into separate protofibrils. Decreased protofibril packing was confirmed in plasma for high thrombin concentrations and fibrinogen-deficient plasma reconstituted with γ' fibrinogen. These findings demonstrate that, in fully hydrated conditions, thrombin and fibrinogen γ' have dramatic effects on protofibril content and that protein density within fibers correlates with strength of the fibrin network. We conclude that regulation of protofibril content of fibers is an important mechanism by which thrombin and fibrinogen γ' modulate fibrin clot structure and strength.


Assuntos
Coagulação Sanguínea , Fibrinogênios Anormais/metabolismo , Fibrinogênios Anormais/ultraestrutura , Trombina/metabolismo , Trombina/ultraestrutura , Viscosidade Sanguínea , Humanos , Microscopia de Força Atômica , Nefelometria e Turbidimetria , Trombose/metabolismo
7.
Circ Res ; 119(3): 450-62, 2016 07 22.
Artigo em Inglês | MEDLINE | ID: mdl-27245171

RESUMO

RATIONALE: Blood flow-induced shear stress controls endothelial cell (EC) physiology during atherosclerosis via transcriptional mechanisms that are incompletely understood. The mechanosensitive transcription factor TWIST is expressed during embryogenesis, but its role in EC responses to shear stress and focal atherosclerosis is unknown. OBJECTIVE: To investigate whether TWIST regulates endothelial responses to shear stress during vascular dysfunction and atherosclerosis and compare TWIST function in vascular development and disease. METHODS AND RESULTS: The expression and function of TWIST1 was studied in EC in both developing vasculature and during the initiation of atherosclerosis. In zebrafish, twist was expressed in early embryonic vasculature where it promoted angiogenesis by inducing EC proliferation and migration. In adult porcine and murine arteries, TWIST1 was expressed preferentially at low shear stress regions as evidenced by quantitative polymerase chain reaction and en face staining. Moreover, studies of experimental murine carotid arteries and cultured EC revealed that TWIST1 was induced by low shear stress via a GATA4-dependent transcriptional mechanism. Gene silencing in cultured EC and EC-specific genetic deletion in mice demonstrated that TWIST1 promoted atherosclerosis by inducing inflammation and enhancing EC proliferation associated with vascular leakiness. CONCLUSIONS: TWIST expression promotes developmental angiogenesis by inducing EC proliferation and migration. In addition to its role in development, TWIST is expressed preferentially at low shear stress regions of adult arteries where it promotes atherosclerosis by inducing EC proliferation and inflammation. Thus, pleiotropic functions of TWIST control vascular disease and development.


Assuntos
Aterosclerose/metabolismo , Velocidade do Fluxo Sanguíneo/fisiologia , Endotélio Vascular/metabolismo , Proteínas Nucleares/biossíntese , Proteína 1 Relacionada a Twist/biossíntese , Animais , Aterosclerose/patologia , Movimento Celular/fisiologia , Proliferação de Células/fisiologia , Células Cultivadas , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Endotélio Vascular/patologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Células Endoteliais da Veia Umbilical Humana/patologia , Humanos , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Suínos , Peixe-Zebra
8.
Arterioscler Thromb Vasc Biol ; 37(11): 2087-2101, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28882872

RESUMO

OBJECTIVE: Atherosclerosis develops near branches and bends of arteries that are exposed to low shear stress (mechanical drag). These sites are characterized by excessive endothelial cell (EC) proliferation and inflammation that promote lesion initiation. The transcription factor HIF1α (hypoxia-inducible factor 1α) is canonically activated by hypoxia and has a role in plaque neovascularization. We studied the influence of shear stress on HIF1α activation and the contribution of this noncanonical pathway to lesion initiation. APPROACH AND RESULTS: Quantitative polymerase chain reaction and en face staining revealed that HIF1α was expressed preferentially at low shear stress regions of porcine and murine arteries. Low shear stress induced HIF1α in cultured EC in the presence of atmospheric oxygen. The mechanism involves the transcription factor nuclear factor-κB that induced HIF1α transcripts and induction of the deubiquitinating enzyme Cezanne that stabilized HIF1α protein. Gene silencing revealed that HIF1α enhanced proliferation and inflammatory activation in EC exposed to low shear stress via induction of glycolysis enzymes. We validated this observation by imposing low shear stress in murine carotid arteries (partial ligation) that upregulated the expression of HIF1α, glycolysis enzymes, and inflammatory genes and enhanced EC proliferation. EC-specific genetic deletion of HIF1α in hypercholesterolemic apolipoprotein E-defecient mice reduced inflammation and endothelial proliferation in partially ligated arteries, indicating that HIF1α drives inflammation and vascular dysfunction at low shear stress regions. CONCLUSIONS: Mechanical low shear stress activates HIF1α at atheroprone regions of arteries via nuclear factor-κB and Cezanne. HIF1α promotes atherosclerosis initiation at these sites by inducing excessive EC proliferation and inflammation via the induction of glycolysis enzymes.


Assuntos
Aterosclerose/metabolismo , Células Endoteliais/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Inflamação/metabolismo , Mecanotransdução Celular , Placa Aterosclerótica , Animais , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Aterosclerose/genética , Aterosclerose/patologia , Proliferação de Células , Células Cultivadas , Modelos Animais de Doenças , Endopeptidases/metabolismo , Células Endoteliais/patologia , Indução Enzimática , Feminino , Predisposição Genética para Doença , Glicólise , Células Endoteliais da Veia Umbilical Humana/metabolismo , Células Endoteliais da Veia Umbilical Humana/patologia , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Inflamação/genética , Inflamação/patologia , Mediadores da Inflamação/metabolismo , Camundongos Knockout , NF-kappa B/metabolismo , Oxigênio/metabolismo , Fenótipo , Estabilidade Proteica , Proteólise , Interferência de RNA , Fluxo Sanguíneo Regional , Estresse Mecânico , Sus scrofa , Fatores de Tempo , Transfecção , Ubiquitinação , Regulação para Cima
9.
Arterioscler Thromb Vasc Biol ; 37(1): 130-143, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27834691

RESUMO

OBJECTIVE: Atherosclerosis is initiated at branches and bends of arteries exposed to disturbed blood flow that generates low shear stress. This mechanical environment promotes lesions by inducing endothelial cell (EC) apoptosis and dysfunction via mechanisms that are incompletely understood. Although transcriptome-based studies have identified multiple shear-responsive genes, most of them have an unknown function. To address this, we investigated whether zebrafish embryos can be used for functional screening of mechanosensitive genes that regulate EC apoptosis in mammalian arteries. APPROACH AND RESULTS: First, we demonstrated that flow regulates EC apoptosis in developing zebrafish vasculature. Specifically, suppression of blood flow in zebrafish embryos (by targeting cardiac troponin) enhanced that rate of EC apoptosis (≈10%) compared with controls exposed to flow (≈1%). A panel of candidate regulators of apoptosis were identified by transcriptome profiling of ECs from high and low shear stress regions of the porcine aorta. Genes that displayed the greatest differential expression and possessed 1 to 2 zebrafish orthologues were screened for the regulation of apoptosis in zebrafish vasculature exposed to flow or no-flow conditions using a knockdown approach. A phenotypic change was observed in 4 genes; p53-related protein (PERP) and programmed cell death 2-like protein functioned as positive regulators of apoptosis, whereas angiopoietin-like 4 and cadherin 13 were negative regulators. The regulation of perp, cdh13, angptl4, and pdcd2l by shear stress and the effects of perp and cdh13 on EC apoptosis were confirmed by studies of cultured EC exposed to flow. CONCLUSIONS: We conclude that a zebrafish model of flow manipulation coupled to gene knockdown can be used for functional screening of mechanosensitive genes in vascular ECs, thus providing potential therapeutic targets to prevent or treat endothelial injury at atheroprone sites.


Assuntos
Apoptose , Aterosclerose/genética , Células Endoteliais/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Mecanotransdução Celular/genética , Proteínas de Peixe-Zebra/genética , Peixe-Zebra/genética , Animais , Animais Geneticamente Modificados , Aterosclerose/metabolismo , Aterosclerose/patologia , Aterosclerose/fisiopatologia , Células Cultivadas , Embrião não Mamífero/irrigação sanguínea , Células Endoteliais/patologia , Feminino , Perfilação da Expressão Gênica/métodos , Técnicas de Silenciamento de Genes , Redes Reguladoras de Genes , Estudos de Associação Genética , Predisposição Genética para Doença , Humanos , Camundongos , Fenótipo , Interferência de RNA , Fluxo Sanguíneo Regional , Estresse Mecânico , Suínos , Transcriptoma , Transfecção , Peixe-Zebra/embriologia , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/metabolismo
10.
Arterioscler Thromb Vasc Biol ; 36(2): 308-16, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26743168

RESUMO

OBJECTIVE: Factor XIII (FXIII) cross-links fibrin upon activation by thrombin. Activation involves cleavage at residue 37 by thrombin, releasing an activation peptide. A common polymorphism (valine to leucine variant at residue 34, V34L), located in the activation peptide, has been associated with increased activation rates and paradoxically a protective effect in cardiovascular disease. There is, currently, no data available on the effects of V34L from in vivo models of thrombosis. We examined the effect of FXIII V34L on clot formation and cross-linking in vivo. APPROACH AND RESULTS: We generated a panel of full-length recombinant human FXIII-A2 variants with amino acid substitutions in the activation peptide to investigate the effect of these variants on activation rate, and we used wild-type, V34L, and alanine to glycine variant at residue 33 variants to study the effects of varying FXIII activation rate on thrombus formation in a murine model of FeCl3 injury. FXIII activation assay showed that residues 29, 30, 33, and 34 play a critical role in thrombin interaction. Full-length recombinant human FXIII-A2 V34L has significant effects on clot formation, structure, and lysis in vitro, using turbidity assay. This variant influenced fibrin cross-linking but not size of the thrombus in vivo. CONCLUSIONS: Mutations in the activation peptide of full-length recombinant FXIII regulate activation rates by thrombin, and V34L influences in vivo thrombus formation by increased cross-linking of the clot.


Assuntos
Coagulação Sanguínea , Fator XIIIa/metabolismo , Fibrina/metabolismo , Trombina/metabolismo , Trombose Venosa/metabolismo , Substituição de Aminoácidos , Animais , Testes de Coagulação Sanguínea , Modelos Animais de Doenças , Deficiência do Fator XIII/sangue , Deficiência do Fator XIII/genética , Deficiência do Fator XIII/metabolismo , Fator XIIIa/administração & dosagem , Fator XIIIa/genética , Genótipo , Humanos , Masculino , Camundongos da Linhagem 129 , Camundongos Endogâmicos CBA , Camundongos Knockout , Mutação , Fenótipo , Proteínas Recombinantes/metabolismo , Fatores de Tempo , Trombose Venosa/sangue , Trombose Venosa/genética
11.
J Biol Chem ; 290(40): 24067-78, 2015 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-26269588

RESUMO

The endothelium is critically involved in the pathogenesis of atherosclerosis by producing pro-inflammatory mediators, including IL-1ß. Coronary arteries from patients with ischemic heart disease express large amounts of IL-1ß in the endothelium. However, the mechanism by which endothelial cells (ECs) release IL-1ß remains to be elucidated. We investigated neutrophil elastase (NE), a potent serine protease detected in vulnerable areas of human carotid plaques, as a potential "trigger" for IL-1ß processing and release. This study tested the hypothesis that NE potentiates the processing and release of IL-1ß from human coronary endothelium. We found that NE cleaves the pro-isoform of IL-1ß in ECs and causes significant secretion of bioactive IL-1ß via extracellular vesicles. This release was attenuated significantly by inhibition of neutrophil elastase but not caspase-1. Transient increases in intracellular Ca(2+) levels were observed prior to secretion. Inside ECs, and after NE treatment only, IL-1ß was detected within LAMP-1-positive multivesicular bodies. The released vesicles contained bioactive IL-1ß. In vivo, in experimental atherosclerosis, NE was detected in mature atherosclerotic plaques, predominantly in the endothelium, alongside IL-1ß. This study reveals a novel mechanistic link between NE expression in atherosclerotic plaques and concomitant pro-inflammatory bioactive IL-1ß secretion from ECs. This could reveal additional potential anti-IL-1ß therapeutic targets and provide further insights into the inflammatory process by which vascular disease develops.


Assuntos
Vasos Coronários/metabolismo , Endotélio Vascular/metabolismo , Regulação Enzimológica da Expressão Gênica , Interleucina-1beta/metabolismo , Elastase de Leucócito/metabolismo , Animais , Apolipoproteínas E/genética , Apoptose , Cálcio/metabolismo , Sobrevivência Celular , Células Cultivadas , Células Endoteliais/enzimologia , Humanos , Imuno-Histoquímica , Inflamação/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Microcirculação , Fosforilação , Placa Aterosclerótica/metabolismo
12.
Blood ; 118(15): 4265-73, 2011 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-21803846

RESUMO

The activation of endothelial cells is critical to initiating an inflammatory response. Activation induces the fusion of Weibel-Palade Bodies (WPB) with the plasma membrane, thus transferring P-selectin and VWF to the cell surface, where they act in the recruitment of leukocytes and platelets, respectively. CD63 has long been an established component of WPB, but the functional significance of its presence within an organelle that acts in inflammation and hemostasis was unknown. We find that ablating CD63 expression leads to a loss of P-selectin-dependent function: CD63-deficient HUVECs fail to recruit leukocytes, CD63-deficient mice exhibit a significant reduction in both leukocyte rolling and recruitment and we show a failure of leukocyte extravasation in a peritonitis model. Loss of CD63 has a similar phenotype to loss of P-selectin itself, thus CD63 is an essential cofactor to P-selectin.


Assuntos
Células Endoteliais/metabolismo , Migração e Rolagem de Leucócitos , Leucócitos/metabolismo , Selectina-P/metabolismo , Tetraspanina 30/metabolismo , Animais , Linhagem Celular , Membrana Celular/genética , Membrana Celular/metabolismo , Membrana Celular/patologia , Células Endoteliais/patologia , Técnicas de Silenciamento de Genes , Leucócitos/patologia , Camundongos , Camundongos Knockout , Selectina-P/genética , Peritonite/genética , Peritonite/metabolismo , Peritonite/patologia , Tetraspanina 30/genética , Corpos de Weibel-Palade/genética , Corpos de Weibel-Palade/metabolismo
14.
Sci Adv ; 8(35): eabo7958, 2022 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-36044575

RESUMO

Endothelial cell (EC) sensing of disturbed blood flow triggers atherosclerosis, a disease of arteries that causes heart attack and stroke, through poorly defined mechanisms. The Notch pathway plays a central role in blood vessel growth and homeostasis, but its potential role in sensing of disturbed flow has not been previously studied. Here, we show using porcine and murine arteries and cultured human coronary artery EC that disturbed flow activates the JAG1-NOTCH4 signaling pathway. Light-sheet imaging revealed enrichment of JAG1 and NOTCH4 in EC of atherosclerotic plaques, and EC-specific genetic deletion of Jag1 (Jag1ECKO) demonstrated that Jag1 promotes atherosclerosis at sites of disturbed flow. Mechanistically, single-cell RNA sequencing in Jag1ECKO mice demonstrated that Jag1 suppresses subsets of ECs that proliferate and migrate. We conclude that JAG1-NOTCH4 sensing of disturbed flow enhances atherosclerosis susceptibility by regulating EC heterogeneity and that therapeutic targeting of this pathway may treat atherosclerosis.


Assuntos
Aterosclerose , Proteína Jagged-1 , Placa Aterosclerótica , Receptor Notch4 , Animais , Aterosclerose/genética , Aterosclerose/metabolismo , Vasos Coronários/metabolismo , Células Endoteliais/metabolismo , Humanos , Proteína Jagged-1/genética , Proteína Jagged-1/metabolismo , Camundongos , Placa Aterosclerótica/metabolismo , Receptor Notch4/genética , Receptor Notch4/metabolismo , Transdução de Sinais , Suínos
15.
Int J Biochem Cell Biol ; 141: 106097, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34655813

RESUMO

Microvesicles are formed through shedding from the plasma membrane, a process shared by almost all human cells. Microvesicles are highly abundant and have been detected in blood, urine, cerebrospinal fluid, and saliva. They contain a library of cargo derived from their parental cell during formation, including proteases, micro-RNAs and lipids and delivery of this parental cell-derived cargo to other cells can alter target cell function and drive disease. Cell specific molecules on the surface of microvesicles, obtained during microvesicle formation, allows their parental cell to be identified and populations of microvesicles to be investigated for roles in the pathogenesis of various diseases. For instance, recent work by our group has identified a role for neutrophil microvesicles in atherosclerosis. Microvesicle profiles could in future be associated with certain diseases and act as a biomarker to allow for earlier diagnosis. This short review will discuss some of the processes central to all microvesicles before focusing on neutrophil microvesicles, their potential role in cardiovascular disease and the mechanisms that may underpin this.


Assuntos
Membrana Celular , Neutrófilos , Aterosclerose , Comunicação Celular , Micropartículas Derivadas de Células
16.
Elife ; 102021 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-34633287

RESUMO

Fibrinogen is essential for blood coagulation. The C-terminus of the fibrinogen α-chain (αC-region) is composed of an αC-domain and αC-connector. Two recombinant fibrinogen variants (α390 and α220) were produced to investigate the role of subregions in modulating clot stability and resistance to lysis. The α390 variant, truncated before the αC-domain, produced clots with a denser structure and thinner fibres. In contrast, the α220 variant, truncated at the start of the αC-connector, produced clots that were porous with short, stunted fibres and visible fibre ends. These clots were mechanically weak and susceptible to lysis. Our data demonstrate differential effects for the αC-subregions in fibrin polymerisation, clot mechanical strength, and fibrinolytic susceptibility. Furthermore, we demonstrate that the αC-subregions are key for promoting longitudinal fibre growth. Together, these findings highlight critical functions of the αC-subregions in relation to clot structure and stability, with future implications for development of novel therapeutics for thrombosis.


Assuntos
Coagulação Sanguínea/fisiologia , Fibrinogênio/química , Fibrinogênio/metabolismo , Fibrinólise , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Animais , Células CHO , Cricetulus , Fibrina/química , Humanos , Camundongos Knockout , Proteínas Recombinantes/química
17.
J Leukoc Biol ; 85(1): 55-63, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18927400

RESUMO

Ly-6G is a member of the Ly-6 family of GPI-linked proteins, which is expressed on murine neutrophils. Antibodies against Ly-6G cause neutropenia, and fatal reactions also develop if mice are primed with TNF-alpha prior to antibody treatment. We have investigated the mechanisms behind these responses to Ly-6G ligation in the belief that similar mechanisms may be involved in neutropenia and respiratory disorders associated with alloantibody ligation of the related Ly-6 family member, NB1, in humans. Neutrophil adhesion, microvascular obstruction, breathing difficulties, and death initiated by anti-Ly-6G antibodies in TNF-alpha-primed mice were shown to be highly complement-dependent, partly mediated by CD11b, CD18, and FcgammaR and associated with clustering of Ly-6G. Neutrophil depletion, on the other hand, was only partly complement-dependent and was not altered by blockade of CD11b, CD18, or FcgammaR. Unlike other neutrophil-activating agents, Ly-6G ligation did not induce neutropenia via sequestration in the lungs. Cross-linking Ly-6G mimicked the responses seen with whole antibody in vivo and also activated murine neutrophils in vitro. Although this suggests that the responses are, in part, mediated by nonspecific properties of antibody ligation, neutrophil depletion requires an additional mechanism possibly specific to the natural function of Ly-6G.


Assuntos
Antígenos Ly/imunologia , Proteínas do Sistema Complemento/imunologia , Neutropenia/imunologia , Neutrófilos/imunologia , Insuficiência Respiratória/imunologia , Animais , Anticorpos Monoclonais/farmacologia , Antígeno CD11b/imunologia , Antígenos CD18/imunologia , Reagentes de Ligações Cruzadas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microcirculação/imunologia , Neutropenia/mortalidade , Neutropenia/fisiopatologia , Receptores de IgG/genética , Receptores de IgG/imunologia , Respiração , Insuficiência Respiratória/mortalidade , Insuficiência Respiratória/fisiopatologia , Fator de Necrose Tumoral alfa/imunologia
18.
Nat Rev Cardiol ; 17(1): 52-63, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31366922

RESUMO

Flowing blood generates a frictional force called shear stress that has major effects on vascular function. Branches and bends of arteries are exposed to complex blood flow patterns that exert low or low oscillatory shear stress, a mechanical environment that promotes vascular dysfunction and atherosclerosis. Conversely, physiologically high shear stress is protective. Endothelial cells are critical sensors of shear stress but the mechanisms by which they decode complex shear stress environments to regulate physiological and pathophysiological responses remain incompletely understood. Several laboratories have advanced this field by integrating specialized shear-stress models with systems biology approaches, including transcriptome, methylome and proteome profiling and functional screening platforms, for unbiased identification of novel mechanosensitive signalling pathways in arteries. In this Review, we describe these studies, which reveal that shear stress regulates diverse processes and demonstrate that multiple pathways classically known to be involved in embryonic development, such as BMP-TGFß, WNT, Notch, HIF1α, TWIST1 and HOX family genes, are regulated by shear stress in arteries in adults. We propose that mechanical activation of these pathways evolved to orchestrate vascular development but also drives atherosclerosis in low shear stress regions of adult arteries.


Assuntos
Aterosclerose/genética , Endotélio Vascular/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Mecanotransdução Celular/genética , Animais , Aterosclerose/metabolismo , Aterosclerose/fisiopatologia , Endotélio Vascular/fisiopatologia , Predisposição Genética para Doença , Humanos , Neovascularização Fisiológica/genética , Fenótipo , Fluxo Sanguíneo Regional , Fatores de Risco , Estresse Mecânico , Remodelação Vascular/genética
19.
Cardiovasc Res ; 116(7): 1300-1310, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-31504243

RESUMO

AIMS: Atherosclerosis develops near branches and bends of arteries that are exposed to disturbed blood flow which exerts low wall shear stress (WSS). These mechanical conditions alter endothelial cells (EC) by priming them for inflammation and by inducing turnover. Homeobox (Hox) genes are developmental genes involved in the patterning of embryos along their anterior-posterior and proximal-distal axes. Here we identified Hox genes that are regulated by WSS and investigated their functions in adult arteries. METHODS AND RESULTS: EC were isolated from inner (low WSS) and outer (high WSS) regions of the porcine aorta and the expression of Hox genes was analysed by quantitative real-time PCR. Several Hox genes (HoxA10, HoxB4, HoxB7, HoxB9, HoxD8, HoxD9) were significantly enriched at the low WSS compared to the high WSS region. Similarly, studies of cultured human umbilical vein EC (HUVEC) or porcine aortic EC revealed that the expression of multiple Hox genes (HoxA10, HoxB9, HoxD8, HoxD9) was enhanced under low (4 dyn/cm2) compared to high (13 dyn/cm2) WSS conditions. Gene silencing studies identified Hox genes (HoxB9, HoxD8, HoxD9) that are positive regulators of inflammatory molecule expression in EC exposed to low WSS, and others (HoxB9, HoxB7, HoxB4) that regulated EC turnover. We subsequently focused on HoxB9 because it was strongly up-regulated by low WSS and, uniquely, was a driver of both inflammation and proliferation. At a mechanistic level, we demonstrate using cultured EC and murine models that bone morphogenic protein 4 (BMP4) is an upstream regulator of HoxB9 which elicits inflammation via induction of numerous inflammatory mediators including TNF and downstream NF-κB activation. Moreover, the BMP4-HoxB9-TNF pathway was potentiated by hypercholesterolaemic conditions. CONCLUSIONS: Low WSS induces multiple Hox genes that control the activation state and turnover of EC. Notably, low WSS activates a BMP4-HoxB9-TNF signalling pathway to initiate focal arterial inflammation, thereby demonstrating integration of the BMP and Hox systems in vascular pathophysiology.


Assuntos
Aorta Torácica/metabolismo , Doenças da Aorta/metabolismo , Aterosclerose/metabolismo , Proteína Morfogenética Óssea 4/metabolismo , Proteínas de Homeodomínio/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Inflamação/metabolismo , Placa Aterosclerótica , Animais , Aorta Torácica/patologia , Aorta Torácica/fisiopatologia , Doenças da Aorta/genética , Doenças da Aorta/patologia , Doenças da Aorta/fisiopatologia , Aterosclerose/genética , Aterosclerose/patologia , Aterosclerose/fisiopatologia , Proteína Morfogenética Óssea 4/genética , Células Cultivadas , Modelos Animais de Doenças , Proteínas de Homeodomínio/genética , Células Endoteliais da Veia Umbilical Humana/patologia , Humanos , Inflamação/genética , Inflamação/patologia , Inflamação/fisiopatologia , Camundongos Endogâmicos C57BL , Camundongos Knockout para ApoE , Fluxo Sanguíneo Regional , Transdução de Sinais , Estresse Mecânico , Sus scrofa , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
20.
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa