Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
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.
Pharmaceuticals (Basel) ; 14(8)2021 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-34451807

RESUMO

Protein kinase inhibitors (PKIs) are important therapeutic agents. As of 31 May 2021, the United States Food and Drug Administration (USFDA) has approved 70 PKIs. Most of the PKIs are employed to treat cancer and inflammatory diseases. Imatinib was the first PKI approved by USFDA in 2001. This review summarizes the compound patents and the essential polymorph patents of the PKIs approved by the USFDA from 2001 to 31 May 2021. The dates on the generic drug availability of the PKIs in the USA market have also been forecasted. It is expected that 19 and 48 PKIs will be genericized by 2025 and 2030, respectively, due to their compound patent expiry. This may reduce the financial toxicity associated with the existing PKIs. There are nearly 535 reported PKs. However, the USFDA approved PKIs target only about 10-15% of the total said PKs. As a result, there are still a large number of unexplored PKs. As the field advances during the next 20 years, one can anticipate that PKIs with many scaffolds, chemotypes, and pharmacophores will be developed.

3.
Expert Opin Ther Pat ; 31(11): 1059-1074, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34082658

RESUMO

INTRODUCTION: Mucormycosis is an uncommon but life-threatening infection with nonspecific clinical manifestations that make its diagnosis/treatment difficult. The current literature indicates that mucormycosis case incidences are on the rise in developing and developed countries, and, unfortunately, there are only a few treatments available. Accordingly, it is essential to provide more treatment options for mucormycosis. AREA COVERED: This patent review focuses on the granted patents and patent applications related to medication for mucormycosis treatment from the publication year of the amphotericin-B patent application (1958) till 30 January 2021. EXPERT OPINION: Mucormycosis has few available treatments, including amphotericin-B, isavuconazonium sulfate, posaconazole, or their combination. A few anti-mucormycosis medicines are under clinical development. The exact burden of mucormycosis is unknown, but it is expected to be higher than the reported cases because of mucormycosis epidemiological changes. This patent review has shown that scientists are progressing toward developing a new treatment for mucormycosis in the form of new chemical compounds, new drug combinations, and dosage forms, vaccines, plant products, drug repurposing, and derivatives of the biomolecules. This progress is encouraging to fight this devastating illness.


Assuntos
Antifúngicos/farmacologia , Desenvolvimento de Medicamentos , Mucormicose/tratamento farmacológico , Animais , Reposicionamento de Medicamentos , Humanos , Mucormicose/epidemiologia , Mucormicose/microbiologia , Patentes como Assunto
4.
FASEB J ; 34(9): 12805-12819, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32772419

RESUMO

Increased endothelial permeability leads to excessive exudation of plasma proteins and leukocytes in the interstitium, which characterizes several vascular diseases including acute lung injury. The myosin light chain kinase long (MYLK-L) isoform is canonically known to regulate the endothelial permeability by phosphorylating myosin light chain (MLC-P). Compared to the short MYLK isoform, MYLK-L contains an additional stretch of ~919 amino acid at the N-terminus of unknown function. We show that thapsigargin and thrombin-induced SOCE was markedly reduced in Mylk-L-/- endothelial cells (EC) or MYLK-L-depleted human EC. These agonists also failed to increase endothelial permeability in MYLK-L-depleted EC and Mylk-L-/- lungs, thus demonstrating the novel role of MYLK-L-induced SOCE in increasing vascular permeability. MYLK-L augmented SOCE by increasing endoplasmic reticulum (ER)-plasma membrane (PM) junctions and STIM1 translocation to these junctions. Transduction of N-MYLK domain (amino acids 1-919 devoid of catalytic activity) into Mylk-L-/- EC rescued SOCE to the level seen in control EC in a STIM1-dependent manner. N-MYLK-induced SOCE augmented endothelial permeability without MLC-P via an actin-binding motif, DVRGLL. Liposomal-mediated delivery of N-MYLK mutant but not ∆DVRGLL-N-MYLK mutant in Mylk-L-/- mice rescued vascular permeability increase in response to endotoxin, indicating that targeting of DVRGLL motif within MYLK-L may limit SOCE-induced vascular hyperpermeability.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Permeabilidade Capilar , Membrana Celular/enzimologia , Retículo Endoplasmático/enzimologia , Quinase de Cadeia Leve de Miosina/metabolismo , Lesão Pulmonar Aguda/metabolismo , Animais , Células Endoteliais da Veia Umbilical Humana , Humanos , Isoenzimas/metabolismo , Pulmão/metabolismo , Pulmão/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas de Neoplasias/metabolismo , Molécula 1 de Interação Estromal/metabolismo
5.
Cell Rep ; 30(12): 4096-4109.e5, 2020 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-32209471

RESUMO

Acute lung injury (ALI) is a lethal inflammatory lung disorder whose incidence is on the rise. Alveolar macrophages normally act to resolve inflammation, but when dysregulated they can provoke ALI. We demonstrate that monocyte-derived macrophages (CD11b+ macrophages) recruited into the airspace upregulate the anti-inflammatory function of alveolar macrophages by suppressing their stimulator of type 1 interferon gene (STING) signaling. Depletion of CD11b+ macrophages in mice (macrophagedep mice) after endotoxin or after Pseudomonas aeruginosa causes expansion of the inflammatory alveolar macrophage population, leading to neutrophil accumulation, irreversible loss of lung vascular barrier function, and lethality. We show that CD11b+ macrophages suppress alveolar macrophage-STING signaling via sphingosine kinase-2 (SPHK2) generation of sphingosine-1-phosphate (S1P). Thus, adoptive transfer of wild-type (WT) or STING-/-, but not SPHK2-/-, CD11b monocytes from murine bone marrow into injured macrophagedep mice rescue anti-inflammatory alveolar macrophages and reverse lung vascular injury. SPHK2-induced S1P generation in CD11b+ macrophages has the potential to educate alveolar macrophages to resolve ALI.


Assuntos
Antígeno CD11b/metabolismo , Inflamação/patologia , Lisofosfolipídeos/metabolismo , Macrófagos Alveolares/metabolismo , Proteínas de Membrana/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Esfingosina/análogos & derivados , Transferência Adotiva , Animais , Citocinas/metabolismo , Humanos , Mediadores da Inflamação/metabolismo , Pulmão/irrigação sanguínea , Pulmão/patologia , Macrófagos Alveolares/microbiologia , Camundongos Endogâmicos C57BL , Nucleotídeos Cíclicos/metabolismo , Pseudomonas aeruginosa/fisiologia , Transdução de Sinais , Esfingosina/metabolismo , Células U937
6.
Cell Rep ; 27(3): 793-805.e4, 2019 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-30995477

RESUMO

Alveolar macrophages (AMs), upon sensing pathogens, trigger host defense by activating toll-like receptor 4 (TLR4), but the counterbalancing mechanisms that deactivate AM inflammatory signaling and prevent lethal edema, the hallmark of acute lung injury (ALI), remain unknown. Here, we demonstrate the essential role of AM protease-activating receptor 2 (PAR2) in rapidly suppressing inflammation to prevent long-lasting injury. We show that thrombin, released during TLR4-induced lung injury, directly activates PAR2 to generate cAMP, which abolishes Ca2+ entry through the TRPV4 channel. Deletion of PAR2 and thus the accompanying cAMP generation augments Ca2+ entry via TRPV4, causing sustained activation of the transcription factor NFAT to produce long-lasting TLR4-mediated inflammatory lung injury. Rescuing thrombin-sensitive PAR2 expression or blocking TRPV4 activity in PAR2-null AMs restores their capacity to resolve inflammation and reverse lung injury. Thus, activation of the thrombin-induced PAR2-cAMP cascade in AMs suppresses TLR4 inflammatory signaling to reinstate tissue integrity.


Assuntos
Sinalização do Cálcio , AMP Cíclico/metabolismo , Inflamação/prevenção & controle , Macrófagos Alveolares/metabolismo , Receptor PAR-2/metabolismo , Canais de Cátion TRPV/metabolismo , Receptor 4 Toll-Like/metabolismo , Lesão Pulmonar Aguda/metabolismo , Lesão Pulmonar Aguda/patologia , Animais , Cálcio/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Inflamação/metabolismo , Lipopolissacarídeos/toxicidade , Macrófagos Alveolares/citologia , Macrófagos Alveolares/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fatores de Transcrição NFATC/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Receptor PAR-2/deficiência , Receptor PAR-2/genética , Canais de Cátion TRPV/antagonistas & inibidores , Canais de Cátion TRPV/genética , Trombina/metabolismo
7.
Am J Respir Cell Mol Biol ; 61(2): 257-265, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30811958

RESUMO

Dysfunctional endothelial cell (EC) barrier and increased lung vascular permeability is a cardinal feature of acute lung injury and sepsis that may result in a pathophysiological condition characterized by alveolar flooding, pulmonary edema, and subsequent hypoxemia. In lung ECs, activation of Rho-associated kinase-1 (ROCK1) phosphorylates myosin light chain (MLC)-associated phosphatase at its inhibitory site, which favors phosphorylation of MLC, stress fiber formation, and hyperpermeability during acute lung injury. The role of microRNA-144 (miR-144) has been well investigated in many human diseases, including cardiac ischemia/reperfusion-induced injury, lung cancer, and lung viral infection; however, its role in pulmonary EC barrier regulation remains obscure. Here, we investigated the miR-144-mediated mechanism in the protection of endothelial barrier function in an LPS-induced lung injury model. By using transendothelial electrical resistance and transwell permeability assay to examine in vitro permeability and immunofluorescence microscopy to determine barrier integrity, we showed that ectopic expression of miR-144 effectively blocked lung EC barrier disruption and hyperpermeability in response to proinflammatory agents. Furthermore, using a gain-and-loss-of-function strategy, overexpression of miR-144 significantly decreased ROCK1 expression. Concomitantly, miR-144 inhibits ROCK1-mediated phosphorylation of MLC phosphataseThr853 and thus phosphorylation of MLCThr18/Ser19 to counteract stress fiber formation in LPS-activated EC. Finally, in LPS-challenged mice, intranasal delivery of miR-144 mimic via liposomes attenuated endotoxemia-induced increases in lung wet/dry ratio, vascular permeability, and inflammation. In conclusion, these data suggest that miR-144-attenuated activation of inflammatory ROCK1/MLC pathway in vascular ECs is a promising therapeutic strategy to counter inflammatory lung injury.


Assuntos
Células Endoteliais/metabolismo , Pulmão/metabolismo , MicroRNAs/metabolismo , Quinases Associadas a rho/metabolismo , Animais , Impedância Elétrica , Células Endoteliais/efeitos dos fármacos , Humanos , Inflamação , Lipopolissacarídeos , Lipossomos/metabolismo , Pulmão/irrigação sanguínea , Pulmão/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microcirculação , Fosfatase de Miosina-de-Cadeia-Leve/metabolismo , Permeabilidade , Traumatismo por Reperfusão , Transdução de Sinais
9.
Int J Biol Macromol ; 119: 23-31, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30053390

RESUMO

Venom peptides anoplin, cupiennin 1a, latarcin 1, latarcin 3a, latarcin 5, melittin, and pandinin 2 are known to have antibacterial properties. In the current study, we examined whether the antimicrobial properties of these venom peptides have any connection to the binding and inhibition of bacterial ATP synthase. Venom peptides inhibited Escherichia coli wild type and mutant membrane-bound F1Fo ATP synthase to varying degrees. Although significant loss of oxidative phosphorylation was observed for wild type, very little loss occurred for null and mutant E. coli strains in the presence of venom peptides. This study also reaffirms that ßDELSEED-motif residues of ATP synthase are required for peptide binding. Modified venom peptides with C-terminal amide (NH2) groups caused augmented inhibition of ATP synthase and E. coli cell death. Growth patterns of wild type, null, and mutant strains in the presence of melittin, anoplin, cupiennin 1a, latarcin 1, latarcin 3a, latarcin 5, pandinin 2, and their modified variants suggested the possibility of additional molecular targets. Our results demonstrate that the antimicrobial properties of venom peptides are connected to the binding and inhibition of bacterial ATP synthase. Moreover, selective inhibition of ATP synthase by venom peptides suggests a viable alternative to combat antibiotic-resistant microbial infections.


Assuntos
Adenosina Trifosfatases/antagonistas & inibidores , Adenosina Trifosfatases/química , Peptídeos Catiônicos Antimicrobianos/química , Peptídeos Catiônicos Antimicrobianos/farmacologia , Peçonhas/química , Peçonhas/farmacologia , Adenosina Trifosfatases/genética , Relação Dose-Resposta a Droga , Ativação Enzimática/efeitos dos fármacos , Escherichia coli/enzimologia , Modelos Moleculares , Conformação Molecular , Mutação , Ligação Proteica , Relação Estrutura-Atividade
10.
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
11.
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
12.
FASEB J ; 30(1): 102-10, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26316271

RESUMO

Stability of endothelial cell (EC) adherens junctions (AJs) is central for prevention of tissue edema, the hallmark of chronic inflammatory diseases including acute respiratory distress syndrome. Here, we demonstrate a previously unsuspected role of sphingosine kinase 1 (SPHK1) in the mechanism by which transient receptor potential channel 1 (Trpc1)-mediated Ca(2+) entry destabilizes AJs. Trpc1(-/-) monolayers showed a 2.2-fold increase in vascular endothelial (VE)-cadherin cell-surface expression above wild-type (WT) monolayers. Thrombin increased endothelial permeability (evident by a 5-fold increase in interendothelial gap area and 60% decrease in transendothelial electrical resistance) in WT but not Trpc1(-/-) ECs. Trpc1(-/-) mice resisted the hyperpermeability effects of the edemagenic agonists used and exhibited 60% less endotoxin-induced mortality. Because sphingosine-1-phosphate (S1P) strengthens AJs, we determined if TRPC1 functioned by inhibiting SPHK1 activity, which generates S1P. Intriguingly, Trpc1(-/-) ECs or ECs transducing a TRPC1-inactive mutant showed a 1.5-fold increase in basal SPHK1 expression compared with WT ECs, resulting in a 2-fold higher S1P level. SPHK1 inhibitor SK1-I decreased basal transendothelial electrical resistance more in WT ECs (48 and 72% reduction at 20 and 50 µM, respectively) than in Trpc1(-/-) ECs. However, SK1-I pretreatment rescued thrombin-induced EC permeability in Trpc1(-/-) ECs. Thus, TRPC1 suppression of basal SPHK1 activity enables EC-barrier destabilization by edemagenic agonists.


Assuntos
Junções Aderentes/metabolismo , Células Endoteliais/metabolismo , Endotélio/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Canais de Cátion TRPC/metabolismo , Animais , Caderinas/metabolismo , Cálcio/metabolismo , Permeabilidade da Membrana Celular , Camundongos Knockout , Transdução de Sinais/fisiologia , Canais de Cátion TRPC/genética
13.
J Exp Med ; 212(11): 1883-99, 2015 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-26392222

RESUMO

Leukocyte transendothelial migration (TEM) is a tightly regulated, multistep process that is critical to the inflammatory response. A transient increase in endothelial cytosolic free calcium ion concentration (↑[Ca(2+)]i) is required for TEM. However, the mechanism by which endothelial ↑[Ca(2+)]i regulates TEM and the channels mediating this ↑[Ca(2+)]i are unknown. Buffering ↑[Ca(2+)]i in endothelial cells does not affect leukocyte adhesion or locomotion but selectively blocks TEM, suggesting a role for ↑[Ca(2+)]i specifically for this step. Transient receptor potential canonical 6 (TRPC6), a Ca(2+) channel expressed in endothelial cells, colocalizes with platelet/endothelial cell adhesion molecule-1 (PECAM) to surround leukocytes during TEM and clusters when endothelial PECAM is engaged. Expression of dominant-negative TRPC6 or shRNA knockdown in endothelial cells arrests neutrophils apically over the junction, similar to when PECAM is blocked. Selectively activating endothelial TRPC6 rescues TEM during an ongoing PECAM blockade, indicating that TRPC6 functions downstream of PECAM. Furthermore, endothelial TRPC6 is required for trafficking of lateral border recycling compartment membrane, which facilitates TEM. Finally, mice lacking TRPC6 in the nonmyeloid compartment (i.e., endothelium) exhibit a profound defect in neutrophil TEM with no effect on leukocyte trafficking. Our findings identify endothelial TRPC6 as the calcium channel mediating the ↑[Ca(2+)]i required for TEM at a step downstream of PECAM homophilic interactions.


Assuntos
Movimento Celular , Células Endoteliais/fisiologia , Leucócitos/fisiologia , Canais de Cátion TRPC/fisiologia , Animais , Cálcio/metabolismo , Citosol/metabolismo , Ácido Egtázico/análogos & derivados , Ácido Egtázico/farmacologia , Humanos , Inflamação/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Molécula-1 de Adesão Celular Endotelial a Plaquetas/fisiologia , Canal de Cátion TRPC6
14.
Vascul Pharmacol ; 70: 45-54, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25869521

RESUMO

Rho kinase mediates the effects of inflammatory permeability factors by increasing actomyosin-generated traction forces on endothelial adherens junctions, resulting in disassembly of intercellular junctions and increased vascular leakage. In vitro, this is accompanied by the Rho kinase-driven formation of prominent radial F-actin fibers, but the in vivo relevance of those F-actin fibers has been debated, suggesting other Rho kinase-mediated events to occur in vascular leak. Here, we delineated the contributions of the highly homologous isoforms of Rho kinase (ROCK1 and ROCK2) to vascular hyperpermeability responses. We show that ROCK2, rather than ROCK1 is the critical Rho kinase for regulation of thrombin receptor-mediated vascular permeability. Novel traction force mapping in endothelial monolayers, however, shows that ROCK2 is not required for the thrombin-induced force enhancements. Rather, ROCK2 is pivotal to baseline junctional tension as a novel mechanism by which Rho kinase primes the endothelium for hyperpermeability responses, independent from subsequent ROCK1-mediated contractile stress-fiber formation during the late phase of the permeability response.


Assuntos
Permeabilidade Capilar , Células Endoteliais/enzimologia , Junções Intercelulares/enzimologia , Quinases Associadas a rho/metabolismo , Animais , Permeabilidade Capilar/efeitos dos fármacos , Células Cultivadas , Impedância Elétrica , Células Endoteliais/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/enzimologia , Humanos , Junções Intercelulares/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Inibidores de Proteínas Quinases/farmacologia , Interferência de RNA , Transdução de Sinais , Fibras de Estresse/enzimologia , Trombina/farmacologia , Fatores de Tempo , Transfecção , Quinases Associadas a rho/antagonistas & inibidores , Quinases Associadas a rho/genética
15.
Am J Physiol Cell Physiol ; 308(8): C581-93, 2015 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-25673771

RESUMO

Pulmonary arterial hypertension (PAH) is a progressive disease that, if left untreated, eventually leads to right heart failure and death. Elevated pulmonary arterial pressure (PAP) in patients with PAH is mainly caused by an increase in pulmonary vascular resistance (PVR). Sustained vasoconstriction and excessive pulmonary vascular remodeling are two major causes for elevated PVR in patients with PAH. Excessive pulmonary vascular remodeling is mediated by increased proliferation of pulmonary arterial smooth muscle cells (PASMC) due to PASMC dedifferentiation from a contractile or quiescent phenotype to a proliferative or synthetic phenotype. Increased cytosolic Ca(2+) concentration ([Ca(2+)]cyt) in PASMC is a key stimulus for cell proliferation and this phenotypic transition. Voltage-dependent Ca(2+) entry (VDCE) and store-operated Ca(2+) entry (SOCE) are important mechanisms for controlling [Ca(2+)]cyt. Stromal interacting molecule proteins (e.g., STIM2) and Orai2 both contribute to SOCE and we have previously shown that STIM2 and Orai2, specifically, are upregulated in PASMC from patients with idiopathic PAH and from animals with experimental pulmonary hypertension in comparison to normal controls. In this study, we show that STIM2 and Orai2 are upregulated in proliferating PASMC compared with contractile phenotype of PASMC. Additionally, a switch in Ca(2+) regulation is observed in correlation with a phenotypic transition from contractile PASMC to proliferative PASMC. PASMC in a contractile phenotype or state have increased VDCE, while in the proliferative phenotype or state PASMC have increased SOCE. The data from this study indicate that upregulation of STIM2 and Orai2 is involved in the phenotypic transition of PASMC from a contractile state to a proliferative state; the enhanced SOCE due to upregulation of STIM2 and Orai2 plays an important role in PASMC proliferation.


Assuntos
Canais de Cálcio/biossíntese , Hipertensão Pulmonar/metabolismo , Glicoproteínas de Membrana/biossíntese , Miócitos de Músculo Liso/citologia , Canais de Cátion TRPC/biossíntese , Remodelação Vascular/fisiologia , Animais , Cálcio/metabolismo , Cálcio/farmacologia , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio/genética , Canais de Cálcio Tipo L/metabolismo , Sinalização do Cálcio/fisiologia , Desdiferenciação Celular , Proliferação de Células , Células Cultivadas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Contração Muscular/fisiologia , Músculo Liso Vascular/citologia , Nifedipino/farmacologia , Proteína ORAI2 , Artéria Pulmonar/citologia , Interferência de RNA , RNA Interferente Pequeno , Ratos , Ratos Sprague-Dawley , Molécula 2 de Interação Estromal , Canais de Cátion TRPC/genética , Canal de Cátion TRPC6 , Fator de Crescimento Transformador beta/farmacologia , Resistência Vascular , Vasoconstrição
16.
Am J Respir Cell Mol Biol ; 53(3): 355-67, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25569851

RESUMO

Hypoxic pulmonary vasoconstriction (HPV) is an important physiological response that optimizes the ventilation/perfusion ratio. Chronic hypoxia causes vascular remodeling, which is central to the pathogenesis of hypoxia-induced pulmonary hypertension (HPH). We have previously shown that Notch3 is up-regulated in HPH and that activation of Notch signaling enhances store-operated Ca(2+) entry (SOCE), an important mechanism that contributes to pulmonary arterial smooth muscle cell (PASMC) proliferation and contraction. Here, we investigate the role of Notch signaling in HPV and hypoxia-induced enhancement of SOCE. We examined SOCE in human PASMCs exposed to hypoxia and pulmonary arterial pressure in mice using the isolated perfused/ventilated lung method. Wild-type and canonical transient receptor potential (TRPC) 6(-/-) mice were exposed to chronic hypoxia to induce HPH. Inhibition of Notch signaling with a γ-secretase inhibitor attenuates hypoxia-enhanced SOCE in PASMCs and hypoxia-induced increase in pulmonary arterial pressure. Our results demonstrate that hypoxia activates Notch signaling and up-regulates TRPC6 channels. Additionally, treatment with a Notch ligand can mimic hypoxic responses. Finally, inhibition of TRPC6, either pharmacologically or genetically, attenuates HPV, hypoxia-enhanced SOCE, and the development of HPH. These results demonstrate that hypoxia-induced activation of Notch signaling mediates HPV and the development of HPH via functional activation and up-regulation of TRPC6 channels. Understanding the molecular mechanisms that regulate cytosolic free Ca(2+) concentration and PASMC proliferation is critical to elucidation of the pathogenesis of HPH. Targeting Notch regulation of TRPC6 will be beneficial in the development of novel therapies for pulmonary hypertension associated with hypoxia.


Assuntos
Sinalização do Cálcio , Hipertensão Pulmonar/metabolismo , Receptor Notch1/metabolismo , Vasoconstrição , Animais , Proteínas de Ligação ao Cálcio/metabolismo , Hipóxia Celular , Células Cultivadas , Humanos , Hipertensão Pulmonar/fisiopatologia , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Masculino , Proteínas de Membrana/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/fisiopatologia , Miócitos de Músculo Liso/metabolismo , Artéria Pulmonar/metabolismo , Artéria Pulmonar/fisiopatologia , Proteínas Serrate-Jagged , Canais de Cátion TRPC/antagonistas & inibidores , Canais de Cátion TRPC/genética , Canais de Cátion TRPC/metabolismo , Canal de Cátion TRPC6
17.
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.

18.
Am J Physiol Lung Cell Mol Physiol ; 305(4): L291-300, 2013 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-23771883

RESUMO

Loss of lung-fluid homeostasis is the hallmark of acute lung injury (ALI). Association of catenins and actin cytoskeleton with vascular endothelial (VE)-cadherin is generally considered the main mechanism for stabilizing adherens junctions (AJs), thereby preventing disruption of lung vascular barrier function. The present study identifies endothelial focal adhesion kinase (FAK), a nonreceptor tyrosine kinase that canonically regulates focal adhesion turnover, as a novel AJ-stabilizing mechanism. In wild-type mice, induction of ALI by intraperitoneal administration of lipopolysaccharide or cecal ligation and puncture markedly decreased FAK expression in lungs. Using a mouse model in which FAK was conditionally deleted only in endothelial cells (ECs), we show that loss of EC-FAK mimicked key features of ALI (diffuse lung hemorrhage, increased transvascular albumin influx, edema, and neutrophil accumulation in the lung). EC-FAK deletion disrupted AJs due to impairment of the fine balance between the activities of RhoA and Rac1 GTPases. Deletion of EC-FAK facilitated RhoA's interaction with p115-RhoA guanine exchange factor, leading to activation of RhoA. Activated RhoA antagonized Rac1 activity, destabilizing AJs. Inhibition of Rho kinase, a downstream effector of RhoA, reinstated normal endothelial barrier function in FAK-/- ECs and lung vascular integrity in EC-FAK-/- mice. Our findings demonstrate that EC-FAK plays an essential role in maintaining AJs and thereby lung vascular barrier function by establishing the normal balance between RhoA and Rac1 activities.


Assuntos
Endotélio/enzimologia , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Deleção de Genes , Pulmão/irrigação sanguínea , Pulmão/fisiopatologia , Neuropeptídeos/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Animais , Líquidos Corporais/metabolismo , Células Endoteliais/enzimologia , Células Endoteliais/patologia , Endotélio/patologia , Endotélio/fisiopatologia , Estabilidade Enzimática , Proteína-Tirosina Quinases de Adesão Focal/deficiência , Humanos , Integrases/metabolismo , Pulmão/enzimologia , Pulmão/patologia , Lesão Pulmonar/enzimologia , Lesão Pulmonar/patologia , Lesão Pulmonar/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Especificidade de Órgãos
19.
Hepatol Int ; 7(2): 622-35, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26201795

RESUMO

AIMS: To evaluate the participation of nitric oxide (NO) and prostaglandin (PGI2) on hyperdynamic state in endotoxemia-induced portal hypertension (EIP) induced by chronic endotoxemia. METHODS: The portal pressure (PP) and mean arterial pressure (MAP) were recorded, in vivo before and after administration of L-NAME (NOS inhibitor) and indomethacin (specific blocker of COX). The vasoactive responses to acetylcholine of thoracic rat aortic rings were studied in vitro before and after nitric oxide and cyclooxygenase blockade using multichannel organ bath. The mRNA expression for isoforms of (cyclooxygenase) COX and nitric oxide synthase (NOS) were analyzed using RT-PCR. RESULTS: Administration of both L-NAME and indomethacin in EIP rabbits significantly reduced (p < 0.05) the PP and reversed the MAP to normal as compared to sham-operated (SO) rabbits. There was impaired vasodilatory response to acetylcholine in EIP rabbits. L-NAME caused a significant reduction in acetylcholine-induced vasorelaxation in SO rabbits than EIP due to preexisting hyperemia in EIP. Indomethacin partially restored vasoresponsiveness to acetylcholine in EIP group. The mRNA expression of eNOS (endothelial NOS) and COX-1 (constitutive COX) were significantly higher in SO than EIP rabbits. iNOS (inducible NOS) and COX-2 (inducible COX) mRNA expression was seen only in EIP rabbits. CONCLUSIONS: A significant component of acetylcholine-mediated vasorelaxation in EIP model is modulated by eNOS. There was increased production of contractile prostaglandin in EIP rabbits. iNOS and COX-2 play an important role in the hemodynamic abnormalities of PHT. This novel model of PHT produced by chronic splanchnic endotoxemia in rabbit, mimics impaired vasodilation and vasoreactivity akin to other models of PHT.

20.
J Exp Med ; 209(11): 1953-68, 2012 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-23045603

RESUMO

Lung vascular endothelial barrier disruption and the accompanying inflammation are primary pathogenic features of acute lung injury (ALI); however, the basis for the development of both remains unclear. Studies have shown that activation of transient receptor potential canonical (TRPC) channels induces Ca(2+) entry, which is essential for increased endothelial permeability. Here, we addressed the role of Toll-like receptor 4 (TLR4) intersection with TRPC6-dependent Ca(2+) signaling in endothelial cells (ECs) in mediating lung vascular leakage and inflammation. We find that the endotoxin (lipopolysaccharide; LPS) induces Ca(2+) entry in ECs in a TLR4-dependent manner. Moreover, deletion of TRPC6 renders mice resistant to endotoxin-induced barrier dysfunction and inflammation, and protects against sepsis-induced lethality. TRPC6 induces Ca(2+) entry in ECs, which is secondary to the generation of diacylglycerol (DAG) induced by LPS. Ca(2+) entry mediated by TRPC6, in turn, activates the nonmuscle myosin light chain kinase (MYLK), which not only increases lung vascular permeability but also serves as a scaffold to promote the interaction of myeloid differentiation factor 88 and IL-1R-associated kinase 4, which are required for NF-κB activation and lung inflammation. Our findings suggest that TRPC6-dependent Ca(2+) entry into ECs, secondary to TLR4-induced DAG generation, participates in mediating both lung vascular barrier disruption and inflammation induced by endotoxin.


Assuntos
Cálcio/metabolismo , Pulmão/metabolismo , Canais de Cátion TRPC/metabolismo , Receptor 4 Toll-Like/metabolismo , Animais , Transplante de Medula Óssea , Sinalização do Cálcio , Permeabilidade Capilar/efeitos dos fármacos , Células Cultivadas , Citocinas/metabolismo , Diglicerídeos/metabolismo , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Immunoblotting , Imuno-Histoquímica , Inflamação/induzido quimicamente , Inflamação/metabolismo , Quinases Associadas a Receptores de Interleucina-1/metabolismo , Lipopolissacarídeos/toxicidade , Pulmão/irrigação sanguínea , Pulmão/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/metabolismo , Quinase de Cadeia Leve de Miosina/genética , Quinase de Cadeia Leve de Miosina/metabolismo , NF-kappa B/metabolismo , Ligação Proteica , Canais de Cátion TRPC/genética , Canal de Cátion TRPC6
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA