Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Cell Rep ; 43(6): 114297, 2024 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-38824643

RESUMO

The mechanical environment generated through the adhesive interaction of endothelial cells (ECs) with the matrix controls nuclear tension, preventing aberrant gene synthesis and the transition from restrictive to leaky endothelium, a hallmark of acute lung injury (ALI). However, the mechanisms controlling tension transmission to the nucleus and EC-restrictive fate remain elusive. Here, we demonstrate that, in a kinase-independent manner, focal adhesion kinase (FAK) safeguards tension transmission to the nucleus to maintain EC-restrictive fate. In FAK-depleted ECs, robust activation of the RhoA-Rho-kinase pathway increased EC tension and phosphorylation of the nuclear envelope protein, emerin, activating DNMT3a. Activated DNMT3a methylates the KLF2 promoter, impairing the synthesis of KLF2 and its target S1PR1 to induce the leaky EC transcriptome. Repleting FAK (wild type or kinase dead) or inhibiting RhoA-emerin-DNMT3a activities in damaged lung ECs restored KLF2 transcription of the restrictive EC transcriptome. Thus, FAK sensing and control of tension transmission to the nucleus govern restrictive endothelium to maintain lung homeostasis.


Assuntos
Núcleo Celular , Células Endoteliais , Fatores de Transcrição Kruppel-Like , Transcriptoma , Proteína rhoA de Ligação ao GTP , Animais , Humanos , Camundongos , Núcleo Celular/metabolismo , DNA Metiltransferase 3A , Células Endoteliais/metabolismo , Quinase 1 de Adesão Focal/metabolismo , Quinase 1 de Adesão Focal/genética , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Proteína-Tirosina Quinases de Adesão Focal/genética , Células Endoteliais da Veia Umbilical Humana/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , Fatores de Transcrição Kruppel-Like/genética , Proteínas de Membrana/metabolismo , Proteínas de Membrana/genética , Proteínas Nucleares/metabolismo , Proteínas Nucleares/genética , Fosforilação , Regiões Promotoras Genéticas/genética , Quinases Associadas a rho/metabolismo , Quinases Associadas a rho/genética , Proteína rhoA de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP/genética , Transcriptoma/genética , Masculino , Feminino
2.
Arterioscler Thromb Vasc Biol ; 42(7): 886-902, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35477279

RESUMO

BACKGROUND: The vascular endothelium maintains tissue-fluid homeostasis by controlling the passage of large molecules and fluid between the blood and interstitial space. The interaction of catenins and the actin cytoskeleton with VE-cadherin (vascular endothelial cadherin) is the primary mechanism for stabilizing AJs (adherens junctions), thereby preventing lung vascular barrier disruption. Members of the Rho (Ras homology) family of GTPases and conventional GEFs (guanine exchange factors) of these GTPases have been demonstrated to play important roles in regulating endothelial permeability. Here, we evaluated the role of DOCK4 (dedicator of cytokinesis 4)-an unconventional Rho family GTPase GEF in vascular function. METHODS: We generated mice deficient in DOCK4' used DOCK4 silencing and reconstitution approaches in human pulmonary artery endothelial cells' used assays to evaluate protein localization, endothelial cell permeability, and small GTPase activation. RESULTS: Our data show that DOCK4-deficient mice are viable. However, these mice have hemorrhage selectively in the lung, incomplete smooth muscle cell coverage in pulmonary vessels, increased basal microvascular permeability, and impaired response to S1P (sphingosine-1-phosphate)-induced reversal of thrombin-induced permeability. Consistent with this, DOCK4 rapidly translocates to the cell periphery and associates with the detergent-insoluble fraction following S1P treatment, and its absence prevents S1P-induced Rac-1 activation and enhancement of barrier function. Moreover, DOCK4-silenced pulmonary artery endothelial cells exhibit enhanced basal permeability in vitro that is associated with enhanced Rho GTPase activation. CONCLUSIONS: Our findings indicate that DOCK4 maintains AJs necessary for lung vascular barrier function by establishing the normal balance between RhoA (Ras homolog family member A) and Rac-1-mediated actin cytoskeleton remodeling, a previously unappreciated function for the atypical GEF family of molecules. Our studies also identify S1P as a potential upstream regulator of DOCK4 activity.


Assuntos
Células Endoteliais , Proteínas rho de Ligação ao GTP , Junções Aderentes/metabolismo , Animais , Permeabilidade Capilar/fisiologia , Células Cultivadas , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Proteínas Ativadoras de GTPase/genética , Proteínas Ativadoras de GTPase/metabolismo , Fatores de Troca do Nucleotídeo Guanina/genética , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Pulmão/metabolismo , Camundongos , Proteínas rho de Ligação ao GTP/metabolismo
3.
J Exp Med ; 219(5)2022 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-35404389

RESUMO

Monocytes undergo phenotypic and functional changes in response to inflammatory cues, but the molecular signals that drive different monocyte states remain largely undefined. We show that monocytes acquire macrophage markers upon glomerulonephritis and may be derived from CCR2+CX3CR1+ double-positive monocytes, which are preferentially recruited, dwell within glomerular capillaries, and acquire proinflammatory characteristics in the nephritic kidney. Mechanistically, the transition to immature macrophages begins within the vasculature and relies on CCR2 in circulating cells and TNFR2 in parenchymal cells, findings that are recapitulated in vitro with monocytes cocultured with TNF-TNFR2-activated endothelial cells generating CCR2 ligands. Single-cell RNA sequencing of cocultures defines a CCR2-dependent monocyte differentiation path associated with the acquisition of immune effector functions and generation of CCR2 ligands. Immature macrophages are detected in the urine of lupus nephritis patients, and their frequency correlates with clinical disease. In conclusion, CCR2-dependent functional specialization of monocytes into macrophages begins within the TNF-TNFR2-activated vasculature and may establish a CCR2-based autocrine, feed-forward loop that amplifies renal inflammation.


Assuntos
Células Endoteliais , Monócitos , Receptores CCR2 , Receptores Tipo II do Fator de Necrose Tumoral , Humanos , Ligantes , Macrófagos , Receptores CCR2/genética , Receptores Tipo II do Fator de Necrose Tumoral/genética
4.
Sci Rep ; 7: 42758, 2017 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-28218251

RESUMO

Store-operated Ca2+ entry (SOCE) mediates the increase in intracellular calcium (Ca2+) in endothelial cells (ECs) that regulates several EC functions including tissue-fluid homeostasis. Stromal-interaction molecule 1 (STIM1), upon sensing the depletion of (Ca2+) from the endoplasmic reticulum (ER) store, organizes as puncta that trigger store-operated Ca2+ entry (SOCE) via plasmalemmal Ca2+-selective Orai1 channels. While the STIM1 and Orai1 binding interfaces have been mapped, signaling mechanisms activating STIM1 recruitment of Orai1 and STIM1-Orai1 interaction remains enigmatic. Here, we show that ER Ca2+-store depletion rapidly induces STIM1 phosphorylation at Y361 via proline-rich kinase 2 (Pyk2) in ECs. Surprisingly, the phospho-defective STIM1-Y361F mutant formed puncta but failed to recruit Orai1, thereby preventing. SOCE Furthermore, studies in mouse lungs, expression of phosphodefective STIM1-Y361F mutant in ECs prevented the increase in vascular permeability induced by the thrombin receptor, protease activated receptor 1 (PAR1). Hence, Pyk2-dependent phosphorylation of STIM1 at Y361 is a critical phospho-switch enabling recruitment of Orai1 into STIM1 puncta leading to SOCE. Therefore, Y361 in STIM1 represents a novel target for limiting SOCE-associated vascular leak.


Assuntos
Cálcio/metabolismo , Quinase 2 de Adesão Focal/metabolismo , Proteínas de Neoplasias/química , Proteínas de Neoplasias/metabolismo , Proteína ORAI1/metabolismo , Molécula 1 de Interação Estromal/química , Molécula 1 de Interação Estromal/metabolismo , Tirosina/metabolismo , Animais , Sítios de Ligação , Células Cultivadas , Retículo Endoplasmático/metabolismo , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Humanos , Camundongos , Mutação , Proteínas de Neoplasias/genética , Fosforilação , Ligação Proteica , Molécula 1 de Interação Estromal/genética
5.
Arterioscler Thromb Vasc Biol ; 36(2): 380-8, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26743170

RESUMO

OBJECTIVE: Increased vascular permeability is a hallmark of sepsis and acute respiratory distress syndrome. Angiopoietin (Ang2) induces vascular leak, and excess Ang2 generation is associated with patient mortality from these diseases. However, mechanisms dampening Ang2 generation during injury remain unclear. Interestingly, microRNA (miR)-150 levels were decreased in septic patients. miR regulate signaling networks by silencing mRNAs containing complementary sequences. Thus, we hypothesized that miR-150 suppresses Ang2 generation and thereby resolves vascular injury. APPROACH AND RESULTS: Wild-type or miR-150(-/-) mice or endothelial cells were exposed to lipopolysaccharide or sepsis, and Ang2 levels, adherens junction reannealing, endothelial barrier function, and mortality were determined. Although Ang2 transiently increased during lipopolysaccharide-induced injury in wild-type endothelial cells and lungs, miR-150 expression was elevated only during recovery from injury. Deletion of miR-150 caused a persistent increase in Ang2 levels and impaired adherens junctions reannealing after injury, resulting thereby in an irreversible increase in vascular permeability. Also, miR-150(-/-) mice died rapidly after sepsis. Rescuing miR-150 expression in endothelial cells prevented Ang2 generation, thereby restoring vascular barrier function in miR-150(-/-) mice. miR-150 terminated Ang2 generation by targeting the transcription factor, early growth response 2. Thus, early growth response 2 or Ang2 depletion in miR-150(-/-) endothelial cells restored junctional reannealing and reinstated barrier function. Importantly, upregulating miR-150 expression by injecting a chemically synthesized miR-150 mimic into wild-type mice vasculature decreased early growth response 2 and Ang2 levels and hence mortality from sepsis. CONCLUSIONS: miR-150 is a novel suppressor of Ang2 generation with a key role in resolving vascular injury and reducing mortality resulting from sepsis.


Assuntos
Angiopoietina-2/metabolismo , Células Endoteliais/metabolismo , MicroRNAs/metabolismo , Artéria Pulmonar/metabolismo , Doenças Vasculares/metabolismo , Remodelação Vascular , Junções Aderentes/metabolismo , Junções Aderentes/patologia , Angiopoietina-2/genética , Animais , Permeabilidade Capilar , Células Cultivadas , Modelos Animais de Doenças , Proteína 2 de Resposta de Crescimento Precoce/metabolismo , Células Endoteliais/patologia , Regulação da Expressão Gênica , Genótipo , Lipopolissacarídeos , Camundongos Endogâmicos C57BL , Camundongos Knockout , MicroRNAs/genética , Fenótipo , Artéria Pulmonar/patologia , Sepse/complicações , Transdução de Sinais , Fatores de Tempo , Transfecção , Doenças Vasculares/etiologia , Doenças Vasculares/genética , Doenças Vasculares/patologia , Doenças Vasculares/terapia
6.
J Cell Sci ; 128(5): 878-87, 2015 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-25588843

RESUMO

Activation of sphingosine-1-phosphate receptor 1 (S1PR1) plays a key role in repairing endothelial barrier function. We addressed the role of phosphorylation of the three intracellular tyrosine residues of S1PR1 in endothelial cells in regulating the receptor responsiveness and endothelial barrier function regulated by sphingosine 1-phosphate (S1P)-mediated activation of S1PR1. We demonstrated that phosphorylation of only Y143 site was required for S1PR1 internalization in response to S1P. Maximal S1PR1 internalization was seen in 20 min but S1PR1 returned to the cell surface within 1 h accompanied by Y143-dephosphorylation. Cell surface S1PR1 loss paralleled defective endothelial barrier enhancement induced by S1P. Expression of phospho-defective (Y143F) or phospho-mimicking (Y143D) mutants, respectively, failed to internalize or showed unusually high receptor internalization, consistent with the requirement of Y143 in regulating cell surface S1PR1 expression. Phosphorylation of the five S1PR1 C-terminal serine residues did not affect the role of Y143 phosphorylation in signaling S1PR1 internalization. Thus, rapid reduction of endothelial cell surface expression of S1PR1 subsequent to Y143 phosphorylation is a crucial mechanism of modulating S1PR1 signaling, and hence the endothelial barrier repair function of S1P.


Assuntos
Regulação para Baixo/fisiologia , Células Endoteliais/metabolismo , Lisofosfolipídeos/metabolismo , Receptores de Lisoesfingolipídeo/biossíntese , Transdução de Sinais/fisiologia , Esfingosina/análogos & derivados , Substituição de Aminoácidos , Células Cultivadas , Células Endoteliais/citologia , Humanos , Lisofosfolipídeos/genética , Mutação de Sentido Incorreto , Fosforilação , Receptores de Lisoesfingolipídeo/genética , Esfingosina/genética , Esfingosina/metabolismo , Receptores de Esfingosina-1-Fosfato , Tirosina/genética , Tirosina/metabolismo
7.
Pulm Circ ; 4(4): 535-51, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25610592

RESUMO

The endothelial monolayer partitioning underlying tissue from blood components in the vessel wall maintains tissue fluid balance and host defense through dynamically opening intercellular junctions. Edemagenic agonists disrupt endothelial barrier function by signaling the opening of the intercellular junctions leading to the formation of protein-rich edema in the interstitial tissue, a hallmark of tissue inflammation that, if left untreated, causes fatal diseases, such as acute respiratory distress syndrome. In this review, we discuss how intercellular junctions are maintained under normal conditions and after stimulation of endothelium with edemagenic agonists. We have focused on reviewing the new concepts dealing with the alteration of adherens junctions after inflammatory stimulus.

8.
J Biol Chem ; 288(6): 4241-50, 2013 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-23212915

RESUMO

Stable adherens junctions (AJs) are required for formation of restrictive endothelial barrier. Vascular endothelial cadherin from contiguous endothelial cells forms AJs, which are stabilized intracellularly by binding of p120-catenin and cortical actin. Mechanisms inducing cortical actin formation and enabling its linkage with p120-catenin remain enigmatic. We altered the function of neural Wiskott-Aldrich syndrome protein (N-WASP), which induces actin polymerization through actin-related protein 2/3 complex (Arp2/3), to address the role of N-WASP in regulating AJ stability and thereby endothelial permeability. We show that depletion of N-WASP in endothelial cells impaired AJ adhesion and favored the organization of actin from cortical actin to stress fibers, resulting thereby in formation of leaky endothelial barrier. Exposure of the N-WASP-depleted endothelial cell monolayer to the permeability-increasing mediator, thrombin, exaggerated AJ disruption and stress fiber formation, leading to an irreversible increase in endothelial permeability. We show that N-WASP binds p120-catenin through its verprolin cofilin acid (VCA) domain, induces cortical actin formation through Arp2, and links p120-catenin with cortical actin. The interaction of N-WASP with p120-catenin, actin, and Arp2 requires phosphorylation of N-WASP at the Tyr-256 residue by focal adhesion kinase. Expression of the VCA domain of N-WASP or phosphomimicking (Y256D)-N-WASP mutant in endothelial cells stabilizes AJs and facilitates barrier recovery after thrombin stimulation. Our study demonstrates that N-WASP, by mediating p120-catenin interaction with actin-polymerizing machinery, maintains AJs and mitigates disruption of endothelial barrier function by edemagenic agents, therefore representing a novel target for preventing leaky endothelial barrier syndrome.


Assuntos
Proteína 2 Relacionada a Actina/metabolismo , Junções Aderentes/metabolismo , Cateninas/metabolismo , Células Endoteliais/metabolismo , Fibras de Estresse/metabolismo , Proteína Neuronal da Síndrome de Wiskott-Aldrich/metabolismo , Proteína 2 Relacionada a Actina/genética , Complexo 2-3 de Proteínas Relacionadas à Actina/genética , Complexo 2-3 de Proteínas Relacionadas à Actina/metabolismo , Junções Aderentes/genética , Animais , Células COS , Cateninas/genética , Chlorocebus aethiops , Humanos , Fosforilação/genética , Ligação Proteica/genética , Estrutura Terciária de Proteína , Fibras de Estresse/genética , Proteína Neuronal da Síndrome de Wiskott-Aldrich/genética , delta Catenina
9.
Microbiol Res ; 165(3): 250-8, 2010 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-19616419

RESUMO

Various factors are thought to be responsible for Candida albicans virulence, such as lipases, proteases and adhesins. Many of these factors are GPI-anchored cell surface proteins responsible for pathogenicity. Hwp2 is a putative GPI-anchored protein. The purpose of this study is to characterize the role of Hwp2 regarding filamentation on various filamentation-inducing and non-inducing solid and liquid media, virulence in a mouse model of disseminated candidiasis, and drug resistance to six widely used antifungal agents, by creating a homozygous null hwp2 strain and comparing it with the parental and a revertant HWP2(+)strain. It was observed that an hwp2Delta strain was highly filamentation-deficient on solid agar media as opposed to most liquid media tested. Furthermore, the mutant strain was slightly reduced in virulence compared to the wild strain since all mice infected with the control strain died after 6 days of injection compared with 11 days for the mutant. These results indicate a possible role for Hwp2 in adhesion and invasiveness. Finally a previously unidentified 37-amino-acid-long, stretch of Hwp2, possibly involved in protein aggregation, was found to align with high sequence identity and exclusively to C. albicans cell wall proteins.


Assuntos
Candida albicans/patogenicidade , Parede Celular/química , Proteínas Fúngicas/fisiologia , Glicoproteínas de Membrana/fisiologia , Fatores de Virulência/fisiologia , Sequência de Aminoácidos , Animais , Antifúngicos/farmacologia , Candida albicans/citologia , Candida albicans/efeitos dos fármacos , Candida albicans/crescimento & desenvolvimento , Candidíase/microbiologia , Candidíase/patologia , Adesão Celular , Meios de Cultura/química , Proteínas Fúngicas/genética , Deleção de Genes , Glicosilfosfatidilinositóis/química , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Alinhamento de Sequência , Análise de Sobrevida , Virulência , Fatores de Virulência/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA