Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 296
Filtrar
1.
J Cell Biol ; 120(6): 1491-9, 1993 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-8383691

RESUMO

Thrombin, a potent activator of cellular responses, proteolytically cleaves, and thereby activates its receptor. In the present study, we compared the effects of the thrombin receptor 14-amino acid peptide (TRP-14; SFLLRNPNDKYEPF), which comprises the NH2 terminus after cleavage of the thrombin receptor, and of the native alpha-thrombin on endothelial monolayer permeability. Addition of TRP-14 (1-200 microM) to bovine pulmonary artery endothelial cells increased [Ca2+]i in a dose-dependent manner. The peak increase in [Ca2+]i in response to 100 microM TRP-14 or 0.1 microM alpha-thrombin was similar (i.e., 931 +/- 74 nM and 1032 +/- 80 nM, respectively), which was followed by a slow decrease with t1/2 values of 0.73 and 0.61 min, respectively. Extracellular Ca2+ chelation with 5 mM EGTA abolished the sustained increases in [Ca2+]i induced by either TRP-14 or alpha-thrombin. alpha-thrombin (0.1 microM) increased transendothelial [125I]albumin permeability, whereas TRP-14 (1-100 microM) had no effect. Coincubation of 100 microM TRP-14 with 1 microM DIP-alpha-thrombin also did not increase permeability over control values. Stimulation of BPAEC with 0.1 microM alpha-thrombin induced translocation of protein kinase C (PKC) from the cytosol to the plasma membrane indicative of PKC activation, whereas TRP-14 had no effect at any concentration. TRP-14 at 100 microM desensitized BPAEC to thrombin-induced increases in [Ca2+]i and transendothelial permeability. The Ca2+ desensitization was reversed after approximately 60 min, and this recovery paralleled the recovery of the permeability response. These findings indicate that the TRP-14-induced Ca2+ mobilization in the absence of PKC activation is insufficient to increase endothelial permeability. In contrast, the increase in endothelial permeability after alpha-thrombin occurred in conjunction with Ca2+ mobilization as well as PKC activation. TRP-14 pretreatment prevented the alpha-thrombin-induced increase in endothelial permeability secondary to desensitization of the Ca2+ signal. The results suggest that combined cytosolic Ca2+ mobilization mediated by TRP-14 and PKC activation mediated by a TRP-14-independent pathway are dual signals responsible for the thrombin-induced increase in vascular endothelial permeability.


Assuntos
Endotélio Vascular/fisiologia , Fragmentos de Peptídeos/farmacologia , Receptores de Superfície Celular/fisiologia , Trombina/farmacologia , Sequência de Aminoácidos , Animais , Cálcio/metabolismo , Bovinos , Permeabilidade da Membrana Celular/efeitos dos fármacos , Células Cultivadas , Citosol/metabolismo , Endotélio Vascular/efeitos dos fármacos , Radioisótopos do Iodo , Cinética , Dados de Sequência Molecular , Fragmentos de Peptídeos/síntese química , Artéria Pulmonar , Receptores de Superfície Celular/metabolismo , Receptores de Trombina , Soroalbumina Bovina/metabolismo
2.
J Cell Biol ; 119(4): 935-44, 1992 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-1385447

RESUMO

Thrombin-induced expression of endothelial adhesivity toward neutrophils (PMN) was studied using human umbilical vein endothelial cells (HUVEC). HUVEC were challenged with human alpha-thrombin for varying durations up to 120 min, after which the cells were fixed with 1% paraformaldehyde and 51Cr-labeled human PMN were added to determine PMN adhesion. Endothelial adhesivity increased within 15 min after alpha-thrombin exposure, and the response persisted up to 120 min. Expression of endothelial adhesion proteins, P-selectin (GMP-140, PADGEM, CD62), and intercellular adhesion molecule-1 (ICAM-1; CD54) on the endothelial surface was quantitated by increase in the specific binding of anti-P-selectin mAb G1 and anti-ICAM-1 mAb RR1/1 labeled with 125I. P-selectin expression was maximal at 5-15 min alpha-thrombin exposure and decayed to basal levels within 90 min. In contrast, ICAM-1 activity increased at 30 min and remained elevated for 120 min after alpha-thrombin challenge. The initial endothelial adhesivity was dependent on P-selectin expression since PMN adhesion occurring within the first 30 min after alpha-thrombin challenge was inhibited by mAb G1. The later prolonged PMN adhesion was ICAM-1 dependent since this response was inhibited by mAb RR1/1 and to the same degree by the anti-CD18 mAb IB4. Anti-ELAM-1 mAb BB11 had no effect on adhesion of PMN to the alpha-thrombin-challenged cells. The initial P-selectin expression and PMN adhesion responses were reproduced by the 14-amino peptide (SFLLRNPNDKYEPF) (thrombin-receptor activity peptide; TRP-14) which comprised the NH2 terminus created by thrombin's proteolytic action on its receptors. However, TRP-14-induced PMN adhesion was transient, and TRP-14 did not cause ICAM-1 expression. The ICAM-1-dependent PMN adhesion mediated by alpha-thrombin was protein synthesis independent since ICAM-1 expression and PMN adhesion were not inhibited by cycloheximide pretreatment of HUVEC. Moreover, Northern blot analysis indicated absence of ICAM-1 mRNA signal up to 180 min after alpha-thrombin challenge. In conclusion, thrombin-induced endothelial adhesivity involves early- and late-phase responses. The initial reversible PMN adhesion is mediated by rapid P-selectin expression via TRP-14 generation. Thrombin-induced PMN adhesion is stabilized by a protein synthesis-independent upregulation of the constitutive ICAM-1 activity which enables the interaction of ICAM-1 with the CD18 beta 2 integrin on PMN.


Assuntos
Moléculas de Adesão Celular/biossíntese , Adesão Celular , Endotélio Vascular/metabolismo , Neutrófilos/fisiologia , Glicoproteínas da Membrana de Plaquetas/biossíntese , Trombina/farmacologia , Sequência de Aminoácidos , Adesão Celular/efeitos dos fármacos , Células Cultivadas , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Humanos , Molécula 1 de Adesão Intercelular , Dados de Sequência Molecular , Selectina-P , Fragmentos de Peptídeos/farmacologia , Biossíntese de Proteínas , Receptores de Superfície Celular , Receptores de Trombina
3.
J Cell Biol ; 150(5): 1057-70, 2000 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-10973995

RESUMO

We tested the hypothesis that the albumin-docking protein gp60, which is localized in caveolae, couples to the heterotrimeric GTP binding protein G(i), and thereby activates plasmalemmal vesicle formation and the directed migration of vesicles in endothelial cells (ECs). We used the water-soluble styryl pyridinium dye N-(3-triethylaminopropyl)-4-(p-dibutylaminostyryl) pyridinium dibromide (FM 1-43) to quantify vesicle trafficking by confocal and digital fluorescence microscopy. FM 1-43 and fluorescently labeled anti-gp60 antibody (Ab) were colocalized in endocytic vesicles within 5 min of gp60 activation. Vesicles migrated to the basolateral surface where they released FM 1-43, the fluid phase styryl probe. FM 1-43 fluorescence disappeared from the basolateral EC surface without the loss of anti-gp60 Ab fluorescence. Activation of cell-surface gp60 by cross-linking (using anti-gp60 Ab and secondary Ab) in EC grown on microporous filters increased transendothelial (125)I-albumin permeability without altering liquid permeability (hydraulic conductivity), thus, indicating the dissociation of hydraulic conductivity from the albumin permeability pathway. The findings that the sterol-binding agent, filipin, prevented gp60-activated vesicle formation and that caveolin-1 and gp60 were colocalized in vesicles suggest the caveolar origin of endocytic vesicles. Pertussis toxin pretreatment and expression of the dominant negative construct encoding an 11-amino acid G(alphai) carboxyl-terminal peptide inhibited endothelial (125)I-albumin endocytosis and vesicle formation induced by gp60 activation. Expression of dominant negative Src (dn-Src) and overexpression of wild-type caveolin-1 also prevented gp60-activated endocytosis. Caveolin-1 overexpression resulted in the sequestration of G(alphai) with the caveolin-1, whereas dn-Src inhibited G(alphai) binding to caveolin-1. Thus, vesicle formation induced by gp60 and migration of vesicles to the basolateral membrane requires the interaction of gp60 with caveolin-1, followed by the activation of the downstream G(i)-coupled Src kinase signaling pathway.


Assuntos
Caveolinas , Endocitose/fisiologia , Endotélio Vascular/fisiologia , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Proteínas de Membrana/fisiologia , Sialoglicoproteínas/metabolismo , Quinases da Família src/metabolismo , Animais , Bovinos , Caveolina 1 , Membrana Celular/fisiologia , Membrana Celular/ultraestrutura , Permeabilidade da Membrana Celular , Células Cultivadas , Grânulos Citoplasmáticos/fisiologia , Grânulos Citoplasmáticos/ultraestrutura , Endotélio Vascular/ultraestrutura , Filipina/farmacologia , Corantes Fluorescentes , Humanos , Proteínas de Membrana/genética , Microcirculação , Microscopia Confocal , Microscopia de Fluorescência , Toxina Pertussis , Circulação Pulmonar , Compostos de Piridínio/farmacocinética , Compostos de Amônio Quaternário/farmacocinética , Proteínas Recombinantes/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Transfecção , Fatores de Virulência de Bordetella/farmacologia
4.
J Clin Invest ; 90(6): 2508-16, 1992 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-1281836

RESUMO

We studied the effects of fibrinogen degradation product (FDP) fragment D on endothelial monolayer integrity and the mechanisms of fragment D-induced endothelial cell detachment from the substratum. Incubation of bovine pulmonary artery endothelial cells (BPAEC) with fragment D caused concentration- and time-dependent cell detachment from the substratum. The optimal response occurred at fragment D concentrations of 2 microM and required an incubation time of 24 h. BPAEC challenged with fragment D increased the concentration and activity of urokinase-type plasminogen activator (uPA) in the conditioned medium within 2 to 4 h of incubation. Fragment D also induced the release of tissue-type plasminogen activator, but to a lesser extent than uPA. Fragment D concurrently increased plasminogen activator (PA) activity in a concentration-dependent manner. Increased PA activity was followed by augmentation of cell-associated plasmin activity and subsequent increase in the degradation of 125I-fibrinogen and 125I-vitronectin precoated in the subendothelial matrix. Pretreatment of BPAEC with anti-uPA antibody, and inhibitors of uPA (dansyl-GGACK) and plasmin (aprotinin) prevented approximately 60% of the fragment D-induced endothelial cell detachment. We conclude that FDP fragment D increases secretion of endothelial PAs and enhances the generation of plasmin, thereby contributing to proteolysis of extracellular matrix and endothelial cell detachment. Fragment D may be a critical mediator linking activation of fibrinolysis to vascular endothelial injury in inflammatory disorders.


Assuntos
Adesão Celular , Endotélio Vascular/citologia , Produtos de Degradação da Fibrina e do Fibrinogênio/metabolismo , Fibrinólise , Sequência de Aminoácidos , Animais , Bovinos , Células Cultivadas , Fibrinogênio/metabolismo , Fibrinolisina/antagonistas & inibidores , Fibrinolisina/metabolismo , Glicoproteínas/metabolismo , Técnicas In Vitro , Dados de Sequência Molecular , Ativador de Plasminogênio Tecidual/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Vitronectina
5.
J Clin Invest ; 89(3): 981-8, 1992 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-1347298

RESUMO

(TNF alpha)-induced sequestration of neutrophils (PMN) in lungs and of the resultant PMN-dependent pulmonary edema. Guinea pig lungs perfused with Ringers-albumin were challenged with TNF alpha (1,000 U/ml) for 90 min, followed by addition of fresh perfusate containing 2 x 10(7) human PMN. TNF alpha challenge caused sequestration of PMN in the pulmonary vascular bed as indicated by a threefold increase in lung tissue myeloperoxidase activity (MPO). The activation of the sequestered PMN with phorbol 12-myristate 13-acetate (PMA; 5 x 10(-9) M) produced threefold increases in pulmonary artery (Ppa) and pulmonary capillary hydrostatic (Pcap) pressures, and twofold increases in lung wet-to-dry weight (W/D) ratio and capillary filtration coefficient (Kf,c) over baseline. TNF alpha prestimulation was required for these responses since activation of PMN with PMA in control lungs produced smaller increases in Ppa and Pcap (P less than 0.01) and did not change the W/D and Kf,c. TNF alpha prestimulation also induced the expression of intercellular adhesion molecule (ICAM-1) on pulmonary vascular endothelial cells. Monoclonal antibodies (mAbs) to the neutrophil CD18 integrin (beta-chain of CD11/CD18 complex) (mAb IB4) and to its endothelial cell ligand ICAM-1 (mAb RR1/1) were used to examine the role of PMN adhesion in the TNF alpha-induced responses. Pretreatment of PMN with mAb IB4 prevented PMN uptake and increases in Ppa, Pcap, Kf,c, and W/D ratio. Addition of mAb RR1/1 to the perfusate reduced PMN uptake by 58%, and prevented the increases in Ppa, Pcap, Kf,c, and W/D ratio, as with mAb IB4. The findings indicate that TNF alpha prestimulation of lungs mediates PMN uptake and that this requires the expression of ICAM-1 and its interaction with CD18 integrin on PMN. The activation of PMN sequestered by ICAM-1-dependent mechanism contributes to the development of pulmonary vascular injury and edema.


Assuntos
Antígenos CD/fisiologia , Moléculas de Adesão Celular/fisiologia , Edema Pulmonar/etiologia , Fator de Necrose Tumoral alfa/farmacologia , Animais , Anticorpos Monoclonais/imunologia , Antígenos CD18 , Permeabilidade Capilar/efeitos dos fármacos , Moléculas de Adesão Celular/análise , Cicloeximida/farmacologia , Cobaias , Humanos , Molécula 1 de Adesão Intercelular , Neutrófilos/fisiologia , Circulação Pulmonar , Acetato de Tetradecanoilforbol/farmacologia , Vasoconstrição
6.
J Clin Invest ; 84(3): 793-801, 1989 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-2569479

RESUMO

Plasmin resulted in increased neutrophil adherence to cultured ovine pulmonary artery endothelial cell monolayers in a concentration-dependent manner (10(-12)-10(-7) M). The adherence response increased fivefold above baseline within 60 min after addition of plasmin (10(-8) M) and the response persisted up to 30 min after removal of plasmin. The neutrophil adherence was mediated by the action of plasmin on neutrophils rather than endothelial cells. The response was the result of an increase in functional activity of CD18 neutrophil cell surface adhesive glycoprotein. Neutrophil adherence was inhibited by pretreatment of neutrophils with MAbs IB4 and 60.3 targeted against the beta chain of the CD18, whereas control isotypic MAb 60.5 against HLA class I antigen had no effect. The plasmin catalytic site was not involved in the response. Lys-plasminogen had reduced adherence-promoting activity relative to plasmin, whereas glu-plasminogen had no effect. Elastase-derived plasminogen fragments corresponding to kringle 1+2+3 and kringle 4 (both of which contained the lysine-binding sites) possessed neutrophil adherence-promoting activities similar to plasmin, whereas miniplasminogen (which contains the catalytic site but no lysine-binding sites) had minimal effect, indicating the involvement of lysine-binding sites in the response. Blocking lysine-binding sites of plasmin and elastase-derived plasminogen fragments with tranexamic acid (IC50 of 5 mM) inhibited neutrophil adherence. A monospecific polyclonal antibody against the lysine-binding sites also reduced the neutrophil adherence-promoting activity of plasmin. The results indicate that plasmin induces neutrophil adherence to the endothelium and that the effect is mediated by lysine-binding sites on plasmin.


Assuntos
Adesão Celular , Endotélio Vascular/fisiologia , Fibrinolisina/farmacologia , Lisina/metabolismo , Neutrófilos/fisiologia , Animais , Sítios de Ligação/efeitos dos fármacos , Ligação Competitiva , Antígenos CD18 , Catálise , Adesão Celular/efeitos dos fármacos , Fibrinolisina/metabolismo , Humanos , Cinética , Lisina/fisiologia , Glicoproteínas de Membrana/imunologia , Fragmentos de Peptídeos/farmacologia , Plasminogênio/farmacologia , Artéria Pulmonar , Ovinos
7.
J Clin Invest ; 89(3): 794-802, 1992 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-1541673

RESUMO

We examined the effects of tumor necrosis factor-alpha (TNF alpha) stimulation of endothelial cells on the increase in endothelial permeability induced by H2O2. Bovine pulmonary microvascular endothelial cells (BPMVEC) were grown to confluence on a microporous filter and the 125I-albumin clearance rate across the monolayer was determined. Pretreatment with TNF alpha (100 U/ml) for 6 h had no direct effect on transendothelial 125I-albumin permeability. However, TNF alpha pretreatment enhanced the susceptibility of BPMVEC to H2O2; that is, H2O2 (10 microM) alone had no direct effect, whereas H2O2 increased 125I-albumin permeability more than threefold when added to monolayers pretreated for 6 h with TNF alpha. Determination of lactate dehydrogenase release indicated that increased permeability was not due to cytolysis. We measured the intracellular contents of GSH and catalase to determine their possible role in mediating the increased susceptibility to H2O2. TNF alpha treatment (100 U/ml for 6 h) decreased total GSH content and concomitantly increased the oxidized GSH content, but did not alter the cellular catalase activity. The role of GSH was examined by pretreating endothelial cells with 2 mM GSH for 3 h, which produced an 80% increase in intracellular GSH content. GSH repletion inhibited the increased sensitivity of the TNF alpha-treated endothelial cells to H2O2. We tested the effects of xanthine oxidase (XO) inhibition since XO activation may be a source of oxidants responsible for the decrease in cellular GSH content. Pretreatment with 0.5 mM oxypurinol attenuated the synergistic effect of TNF alpha and H2O2 on endothelial permeability. The results indicate that decreased oxidant buffering capacity secondary to TNF alpha-induced reduction in intracellular GSH content mediates the increased susceptibility of endothelial cells to H2O2. This mechanism may contribute to oxidant-dependent vascular endothelial injury in septicemia associated with TNF alpha release.


Assuntos
Permeabilidade Capilar/efeitos dos fármacos , Endotélio Vascular/efeitos dos fármacos , Glutationa/análise , Peróxido de Hidrogênio/toxicidade , Fator de Necrose Tumoral alfa/farmacologia , Animais , Catalase/análise , Bovinos , Células Cultivadas , Glutationa/farmacologia , Pulmão/irrigação sanguínea , Pulmão/efeitos dos fármacos , Oxipurinol/farmacologia
8.
J Clin Invest ; 101(11): 2427-37, 1998 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-9616214

RESUMO

The binding of beta2 (CD18) integrins on PMN cell membrane to intercellular adhesion molecule (ICAM) counter-receptors on the surface of vascular endothelial cells mediates PMN adhesion to endothelial cells. Neutrophil inhibitory factor (NIF), a 41-kD glycoprotein isolated from the canine hookworm (Ancylostoma caninum), is a beta2 integrin antagonist that inhibits PMN adhesion to endothelial cells. We transferred the NIF gene into CD1 mouse lungs by intravenous injection of cationic liposomes to study the effects of in vivo NIF expression on LPS-induced lung PMN sequestration and the development of lung injury. RT-PCR and Northern blot analysis indicated the lung-selective expression of the NIF transgene, and immunocytochemistry showed prominent NIF expression in pulmonary microvessel endothelial cells. NIF staining was also observed in intraluminal leukocytes present in pulmonary microvessels. This may be the result of NIF binding to leukocytes after its secretion from the transduced lung cells, since there was no evidence of NIF gene expression in circulating leukocytes. Pulmonary vascular NIF expression abrogated the lung tissue PMN uptake and airspace migration of PMN and prevented lung vascular injury (as measured by the lung tissue uptake of [125I]labeled albumin) after the intraperitoneal LPS challenge (200 microg/mouse). Expression of a control protein, chloramphenicol acetyltransferase (CAT), by the same strategy, had no effect on these responses. In vitro studies showed that NIF prevented mouse PMN adhesion consistent with the inhibition of lung uptake after LPS challenge in NIF transgene-expressing mice. We conclude that pulmonary vascular expression of NIF, a specific beta2 integrin- binding protein, is a potentially useful gene transfer strategy in modulating the infiltration of PMN across the alveolar-capillary epithelial barrier and in preventing lung vascular endothelial injury.


Assuntos
Antígenos CD18/fisiologia , Glicoproteínas/fisiologia , Proteínas de Helminto/fisiologia , Lipopolissacarídeos/toxicidade , Pulmão/patologia , Proteínas de Membrana , Neutrófilos/efeitos dos fármacos , Animais , Adesão Celular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Técnicas de Transferência de Genes , Glicoproteínas/genética , Proteínas de Helminto/genética , Pulmão/irrigação sanguínea , Masculino , Camundongos , Camundongos Transgênicos , Neutrófilos/fisiologia
9.
J Clin Invest ; 85(6): 1991-8, 1990 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-2347922

RESUMO

We examined the effects of activation of endothelial protein kinase C (PKC) of the endothelial barrier function. Exposure of confluent bovine pulmonary artery endothelial cell monolayers to phorbol 12-myristate 13-acetate (PMA) resulted in concentration-dependent (10(-8)-10(-6) M) increases in PKC activity and in the transendothelial flux of 125I-albumin. Exposure of the endothelium to 1-oleoyl 2-acetyl glycerol (OAG) also increased the transendothelial flux of 125I-albumin in a concentration-dependent manner. Neither 4 alpha-phorbol didecanoate nor 1-mono-oleoyl glycerol, which do not activate PKC, altered permeability. The increase in 125I-albumin permeability induced by PMA was inhibited by 25 microM H7 (a PKC inhibitor), but not by the control compound HA1004 (25 microM). After 16 h of exposure to PMA, 125I-albumin permeability returned to baseline and a significant reduction in cytosolic PKC activity was noted. Further challenge with PMA at this time resulted in no significant increase in PKC activity indicating downregulation of the enzyme; moreover, this PMA challenge did not increase endothelial permeability. Exposure of endothelial monolayers to phospholipase C (PLC), which increases membrane phosphatidylinositide turnover, or to alpha-thrombin also induced concentration-dependent activation of PKC and increases in 125I-albumin endothelial permeability. The thrombin- and PLC-induced permeability increases were inhibited by H7, but not by HA1004. The activation of endothelial PKC directly by PMA or OAG and by PLC and alpha-thrombin increases the transendothelial albumin permeability, indicating that PKC activation is an important signal transduction pathway by which extracellular mediators increase endothelial macromolecular transport.


Assuntos
Endotélio Vascular/fisiologia , Proteína Quinase C/fisiologia , Albumina Sérica/metabolismo , Animais , Bovinos , Membrana Celular/metabolismo , Células Cultivadas , Citosol/enzimologia , Ativação Enzimática , Técnicas In Vitro , Permeabilidade , Artéria Pulmonar/citologia , Acetato de Tetradecanoilforbol/farmacologia , Trombina/farmacologia , Fatores de Tempo , Fosfolipases Tipo C/fisiologia
10.
Mol Cell Biol ; 21(16): 5554-65, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11463837

RESUMO

The procoagulant thrombin promotes the adhesion of polymorphonuclear leukocytes to endothelial cells by a mechanism involving expression of intercellular adhesion molecule 1 (ICAM-1) via an NF-kappaB-dependent pathway. We now provide evidence that protein kinase C-delta (PKC-delta) and the p38 mitogen-activated protein (MAP) kinase pathway play a critical role in the mechanism of thrombin-induced ICAM-1 gene expression in endothelial cells. We observed the phosphorylation of PKC-delta and p38 MAP kinase within 1 min after thrombin challenge of human umbilical vein endothelial cells. Pretreatment of these cells with the PKC-delta inhibitor rottlerin prevented the thrombin-induced phosphorylation of p38 MAP kinase, suggesting that p38 MAP kinase signals downstream of PKC-delta. Inhibition of PKC-delta or p38 MAP kinase by pharmacological and genetic approaches markedly decreased the thrombin-induced NF-kappaB activity and resultant ICAM-1 expression. The effects of PKC-delta inhibition were secondary to inhibition of IKKbeta activation and of subsequent NF-kappaB binding to the ICAM-1 promoter. The effects of p38 MAP kinase inhibition occurred downstream of IkappaBalpha degradation without affecting the DNA binding function of nuclear NF-kappaB. Thus, PKC-delta signals thrombin-induced ICAM-1 gene transcription by a dual mechanism involving activation of IKKbeta, which mediates NF-kappaB binding to the ICAM-1 promoter, and p38 MAP kinase, which enhances transactivation potential of the bound NF-kappaB p65 (RelA).


Assuntos
Endotélio Vascular/fisiologia , Molécula 1 de Adesão Intercelular/genética , Isoenzimas/fisiologia , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Proteína Quinase C/fisiologia , Trombina/fisiologia , Células Cultivadas , Ativação Enzimática , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , NF-kappa B/fisiologia , Proteína Quinase C-delta , Transdução de Sinais/efeitos dos fármacos , Trombina/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno
11.
Handb Exp Pharmacol ; (176 Pt 1): 107-44, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16999218

RESUMO

An important function of the endothelium is to regulate the transport of liquid and solutes across the semi-permeable vascular endothelial barrier. Two cellular pathways controlling endothelial barrier function have been identified. The transcellular pathway transports plasma proteins of the size of albumin or greater via the process of transcytosis in vesicle carriers originating from cell surface caveolae. Specific signalling cues are able to induce the internalisation of caveolae and their movement to the basal side of the endothelium. Caveolin-1, the primary structural protein required for the formation of caveolae, is also important in regulating vesicle trafficking through the cell by controlling the activity and localisation of signalling molecules that mediate vesicle fission, endocytosis, fusion and finally exocytosis. An important function of the transcytotic pathways is to regulate the delivery of albumin and immunoglobulins, thereby controlling tissue oncotic pressure and host-defence. The paracellular pathway induced during inflammation is formed by gaps between endothelial cells at the level of adherens and tight junctional complexes. Paracellular permeability is increased by second messenger signalling pathways involving Ca2+ influx via activation of store-operated channels, protein kinase Calpha (PKCalpha), and Rho kinase that together participate in the stimulation of myosin light chain phosphorylation, actin-myosin contraction, and disruption of the junctions. In this review of the field, we discuss the current understanding of the signalling pathways regulating paracellular and transcellular endothelial permeability.


Assuntos
Junções Aderentes/metabolismo , Permeabilidade Capilar , Endotélio Vascular/metabolismo , Vesículas Transportadoras/metabolismo , Angiopoietina-1/farmacologia , Animais , Transporte Biológico , Sinalização do Cálcio , Cavéolas/metabolismo , Caveolina 1/metabolismo , AMP Cíclico/farmacologia , Edema/metabolismo , Endotélio Vascular/efeitos dos fármacos , Humanos , Inflamação/metabolismo , Lisofosfolipídeos/farmacologia , Esfingosina/análogos & derivados , Esfingosina/farmacologia , Proteína rhoA de Ligação ao GTP/metabolismo
12.
Cancer Res ; 55(23 Suppl): 5736s-5742s, 1995 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-7493338

RESUMO

An IgM lambda human tumor cell-reactive monoclonal antibody was developed that reacts with cells of ovarian cancer, colorectal cancer, breast cancer, and certain other malignancies. The monoclonal antibody AC6C3-2B12, which was obtained from a recent recloning, was purified from tissue culture supernatants and analyzed by high-performance liquid chromatography and sodium dodecyl sulfate-PAGE. An animal model was developed in which human tumors grew either as solid peritoneal metastases or as s.c. nodules utilizing the human colorectal carcinoma cell line SW620. The biodistribution of 111In-labeled IgM conjugate was studied after i.v. or i.p. administration in nude mice bearing an s.c. xenograft or peritoneal tumor lumps of a human colorectal carcinoma (SW620). IgM administered i.v. cleared rapidly from blood and was deposited mainly in the liver [50% injected dose/g (ID)/g)], pancreas (20% ID/g), and kidney (10% ID/g) at 24 h. Tumor deposition was low (< or = 1.0% ID/g) in the s.c. tumor xenograft. In contrast, high tumor targeting (29% ID/g) was found in peritoneal tumor lumps after i.p. administration of 111In-labeled IgM. The biological half-life of IgM in the tumor was 100 h. Long peritoneal residence time (t 1/2 = 67 h) and low liver uptake (7% ID/g) were observed after i.p. administration. Blood activity was < 1% of the injected activity. Tumor:normal organ ratios were high (range, 2-290) from 2 to 144 h after i.p. administration. Whole body autoradiograms at 24 h after i.p. 111In-labeled IgM administration confirmed the biodistribution results. In normal beagle dogs, 75% of the i.p.-administered 111In-IgM decayed in the peritoneal cavity. The majority of the remaining radioactivity was taken up by mediastinal lymph nodes. Biological half-life in both locations was approximately 137 h. The i.p. administration of intact, specific radiolabeled IgM provides prolonged retention of radioactivity in tumor, low normal tissue uptake, a long peritoneal residence time, and very limited spillover of IgM into the circulation. This approach offers a promising new method for the diagnosis and treatment of certain patients with peritoneal carcinomatosis.


Assuntos
Anticorpos Antineoplásicos/uso terapêutico , Neoplasias Colorretais/radioterapia , Imunoglobulina M/uso terapêutico , Radioisótopos de Índio/uso terapêutico , Radioimunoterapia , Animais , Autorradiografia , Cães , Humanos , Camundongos , Camundongos Nus , Ácido Pentético
13.
Circulation ; 100(12): 1330-7, 1999 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-10491379

RESUMO

Background-The inability to inhibit multiple mediators of septic shock represents a major hurdle in the treatment of septic shock. In vivo inhibition of nuclear factor (NF)-kappaB activation, a transcription factor regulating expression of many proinflammatory genes, could provide a useful strategy for the treatment of septic shock. Methods and Results-In rats challenged with lipopolysaccharide (LPS) 8 mg/kg IV, we determined the time course of NF-kappaB activation and expression of multiple inflammatory signals: tumor necrosis factor-alpha (TNF-alpha), cyclooxygenase-2 (COX-2), cytokine-inducible neutrophil chemoattractant (CINC), and intercellular adhesion molecule-1 (ICAM)-1. We studied the effects of in vivo inhibition of NF-kappaB activation using pyrrolidine dithiocarbamate (PDTC) on the expression of these mediators. NF-kappaB activation preceded the induction of TNF-alpha, COX-2, CINC, and ICAM-1 mRNAs. PDTC prevented the LPS-induced NF-kappaB activation but did not inhibit activation of the transcription factors AP-1, Sp-1, and CREB. PDTC inhibited the LPS-induced expression of TNF-alpha, COX-2, CINC, and ICAM-1 mRNA and proteins and reduced the LPS-induced increases in plasma TNF-alpha, 6-keto-prostaglandin F(1alpha), and CINC concentrations. Inhibition of expression of these mediators prevented the increases in myeloperoxidase activity (a measure of neutrophil sequestration) in the heart, lungs, and liver. Conclusions-NF-kappaB activation correlates with LPS-induced expression of TNF-alpha, COX-2, CINC, and ICAM-1 genes in vivo. PDTC inhibits NF-kappaB activation and expression of these proinflammatory genes and their products. Thus, blocking NF-kappaB activation may be an effective strategy in the treatment of septic shock.


Assuntos
Quimiocinas CXC , Peptídeos e Proteínas de Sinalização Intercelular , NF-kappa B/antagonistas & inibidores , Pirrolidinas/farmacologia , Choque Séptico/metabolismo , Tiocarbamatos/farmacologia , 6-Cetoprostaglandina F1 alfa/sangue , Animais , Fatores Quimiotáticos/metabolismo , Ciclo-Oxigenase 2 , Expressão Gênica/efeitos dos fármacos , Substâncias de Crescimento/metabolismo , Molécula 1 de Adesão Intercelular/metabolismo , Isoenzimas/metabolismo , Masculino , Prostaglandina-Endoperóxido Sintases/metabolismo , Ratos , Ratos Sprague-Dawley , Choque Séptico/tratamento farmacológico , Fator de Necrose Tumoral alfa/metabolismo
14.
J Leukoc Biol ; 60(2): 199-206, 1996 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-8773581

RESUMO

Efficient polymorphonuclear neutrophil (PMN) migration depends on specific interactions between PMNs, endothelial cells, and extracellular matrix (ECM) proteins. We investigated the relationship between PMN migration and the ECM molecule fibronectin (FN). We used an in vitro migration assay system to show that human PMNs migrated across an FN-coated filter barrier toward a formyl-Met-Leu-Phe (fMLP) chemoattractant gradient in greater numbers than across (uncoated) bare fitters. In 1 h of fMLP stimulation, 69 +/- 6% of the PMNs had migrated across the FN-coated filters, whereas 46 +/- 5% of PMNs migrated across bare filters. This effect was specific to FN; coating the filters with the ECM protein vitronectin did not enhance migration. Monoclonal antibodies against FN or against the alpha5 or beta1 integrin subunits of the FN receptor inhibited the enhanced PMN migration response across FN-coated filters. These findings indicate that the extracellular matrix protein FN enhances PMN migration and that this response is mediated by the alpha5beta1 FN receptor.


Assuntos
Fibronectinas/farmacologia , Neutrófilos/citologia , Neutrófilos/efeitos dos fármacos , Receptores de Vitronectina , Anticorpos/farmacologia , Especificidade de Anticorpos , Movimento Celular/efeitos dos fármacos , Fibronectinas/imunologia , Fibronectinas/metabolismo , Filtração/instrumentação , Humanos , Integrina beta1/farmacologia , Integrinas/imunologia , Integrinas/metabolismo , N-Formilmetionina Leucil-Fenilalanina/farmacologia , Estimulação Química
15.
J Leukoc Biol ; 55(1): 58-63, 1994 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-7904295

RESUMO

We examined the hypothesis that neutrophil (PMN)-mediated injury of the vascular endothelium is dependent on adhesion of PMNs to endothelial cells via the leukocyte adhesion glycoprotein CD11/CD18. We compared the PMN activation responses [i.e. adhesion to cultured endothelial cells, superoxide (O2-) production, degranulation, and cytosolic [Ca2+] ([Ca2+]i)] and endothelial injury elicited by opsonized zymosan (OZ, which is phagocytosed by PMNs) or phorbol 12-myristate 13-acetate (PMA, a protein kinase C activator). The basal adherence of nonstimulated PMNs to bovine pulmonary artery endothelial cells (BPAEC) was 9.0 +/- 1.1 PMN/field. PMA and OZ increased PMN adherence to BPAEC (to 31.1 +/- 1.4 and 39.8 +/- 3.8 PMN/field, respectively), which in both cases was inhibited by anti-CD18 monoclonal antibody (mAb) IB4. Stimulation of PMNs with PMA or OZ produced injury to 73% and 53% of BPAEC examined, respectively, which corresponded to 6.8-fold and 3.5-fold increases in transendothelial 125I-albumin permeability from baseline. Pretreatment of PMNs with mAb IB4 prevented endothelial injury in both cases. Both PMA and OZ increased the production of O2- (by 7.6-fold and 3.1-fold over control, respectively) and promoted the release of myeloperoxidase (5.2-fold and 9.1-fold over control, respectively) (P < .01). IB4 did not inhibit the PMA- or OZ-induced increases in O2-. IB4 did not inhibit the PMA-induced myeloperoxidase release but reduced by approximately 29% the OZ-induced myeloperoxidase release. Stimulation of PMNs layered on BPAEC with OZ (0.5 mg/ml) caused an approximately 7-fold increase in PMN [Ca2+]i over baseline, which decayed to a steady-state level above baseline at 10 min. IB4 (10 micrograms/ml) alone did not alter baseline [Ca2+]i and did not inhibit the OZ-induced increase in [Ca2+]i. In contrast to OZ, stimulation of PMNs with PMA did not increase [Ca2+]i. The results indicate that the protective effects of the anti-CD18 mAb IB4 were associated predominantly with its antiadherence property. Therefore, CD18 integrin-mediated PMN adhesion to the endothelium is a critical determinant of endothelial injury irrespective of the PMN-activating stimulus.


Assuntos
Antígenos CD/fisiologia , Endotélio Vascular/patologia , Neutrófilos/fisiologia , Acetato de Tetradecanoilforbol/farmacologia , Zimosan/farmacologia , Animais , Anticorpos Monoclonais/imunologia , Antígenos CD11 , Antígenos CD18 , Cálcio/metabolismo , Bovinos , Adesão Celular , Endotélio Vascular/fisiologia , Humanos , Neutrófilos/efeitos dos fármacos , Permeabilidade , Fagocitose , Superóxidos/metabolismo
16.
J Leukoc Biol ; 39(4): 385-97, 1986 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-3456403

RESUMO

Antisheep antileukocyte serum (ALS) was produced in rabbits, purified, and adsorbed against erythrocytes and platelets. The ALS was infused intra-arterially over a 3-hour period into anesthetized sheep (n = 6) prepared with lung lymph fistulas. Pulmonary vascular resistance (PVR) and pulmonary lymph flow (Qlym) increased two- to threefold while the lymph-to-plasma protein concentration ratio (L/P) did not change from baseline, suggesting an increase in pulmonary vascular permeability to proteins. The arterial leukocyte count decreased from 4,935 +/- 840 to 1,385 +/- 325 cells/microliters, the neutrophil count decreased from 1,045 +/- 265 to 340 +/- 130 cells/microliters, and the platelet count decreased from 2.8 X 10(5) +/- 0.2 X 10(5) to 0.65 X 10(5) +/- 0.12 X 10(5) cells/microliters. ALS induced neutrophil aggregation in vitro, but not platelet aggregation. In the present study, we also examined the effects of ALS-induced leukopenia on the increase in pulmonary vascular permeability after intravenous alpha-thrombin challenge to assess the role of leukocytes in mediating the increased permeability response. Sheep (n = 5) were made leukopenic by an intramuscular injection of ALS; Qlym was in the normal range 4 to 5 hours after the ALS administration. In the leukopenic group, thrombin challenge caused an increase in Qlym from 7.8 +/- 0.7 to 12.9 +/- 1.9 ml/hr (a 64% increase) while L/P decreased from 0.86 +/- 0.04 to 0.70 +/- 0.05 (a 19% decrease). In contrast, thrombin-induced intravascular coagulation in control sheep (n = 5) produced a 250% increase in Qlym with an increase in L/P. The results indicate that leukocytes are required for the increase in lung vascular permeability after thrombin-induced intravascular coagulation.


Assuntos
Anticorpos , Hemodinâmica , Leucopenia/fisiopatologia , Pulmão/fisiopatologia , Circulação Pulmonar , Animais , Pressão Sanguínea , Contagem de Leucócitos , Linfa/fisiologia , Neutrófilos/imunologia , Contagem de Plaquetas , Coelhos/imunologia , Ovinos , Trombina/fisiologia , Resistência Vascular
17.
Cardiovasc Res ; 11(4): 310-6, 1977 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-142580

RESUMO

Myocardial function, flow and O2 consumption were compared in cardiac hypertrophy induced by pressure-loading (P) and by volume overloading (V). Increases in LV-to-body weight ratios in P and V hypertrophied hearts were comparable. Indices of LV function such as cardiac output, stroke volume, stroke work, minute work, peak LV dP/dt, ratio of peak LV dP/dt-to-isovolumic pressure and -to-LVEDP, and Vmax were significantly reduced from normal only in hypertrophy induced by V. Left ventricular coronary flow was reduced from 167.1 +/- 27.2 in normal dogs to 146.2 +/- 17.1 cm3/min-100 g-1 in P hypertrophy, and was reduced further to 82.5 +/- 8.2 cm3/min-100 g-1 in V hypertrophy. Flows decreased similarly in epicardium and endocardium in both hypertrophied hearts. Cardiac O2 extraction in P and V hearts was greater than in control hearts. Myocardial O2 consumption was maintained at control values in P hypertrophy, and decreased by 54 +/- 3% in V hypertrophy. These findings indicate that LV function is impaired at rest in hypertrophy induced by V and is normal in hypertrophy induced by P.


Assuntos
Cardiomegalia/fisiopatologia , Circulação Coronária , Miocárdio/metabolismo , Animais , Velocidade do Fluxo Sanguíneo , Débito Cardíaco , Cardiomegalia/etiologia , Cardiomegalia/metabolismo , Cães , Feminino , Hemodinâmica , Masculino , Tamanho do Órgão , Consumo de Oxigênio
18.
Physiol Genomics ; 4(2): 137-145, 2000 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-11120874

RESUMO

We investigated the function of proteinase-activated receptor-1 (PAR-1) in the regulation of pulmonary microvascular permeability in response to thrombin challenge using PAR-1 knockout mice (-/-). Lungs were isolated and perfused with albumin (5 g/100 ml)-Krebs solution at constant flow (2 ml/min). Lung wet weight and pulmonary artery pressure (P(pa)) were continuously monitored. We determined the capillary filtration coefficient (K(fc)) and (125)I-labeled albumin (BSA) permeability-surface area product (PS) to assess changes in pulmonary microvessel permeability to liquid and albumin, respectively. Normal and PAR-1-null lung preparations received in the perfusate: 1) thrombin or 2) selective PAR-1 agonist peptide (TFLLRNPNDK-NH(2)). In control PAR-1 (+/+) mouse lungs, (125)I-albumin PS and K(fc) were significantly increased over baseline (by approximately 7- and 1.5-fold, respectively) within 20 min of alpha-thrombin (100 nM) challenge. PAR-1 agonist peptide (5 microM) gave similar results, whereas control peptide (5 microM; FTLLRNPNDK-NH(2)) was ineffective. At relatively high concentrations, thrombin (500 nM) or PAR-1 agonist peptide (10 microM) also induced increases in P(pa) and lung wet weight. All effects of thrombin (100 or 500 nM) or PAR-1 agonist peptide (5 or 10 microM) were prevented in PAR-1-null lung preparations. Baseline measures of microvessel permeability and P(pa) in the PAR-1-null preparations were indistinguishable from those in normal lungs. Moreover, PAR-1-null preparations gave normal vasoconstrictor response to thromboxane analog, U-46619 (100 nM). The results indicate that the PAR-1 receptor is critical in mediating the permeability-increasing and vasoconstrictor effects of thrombin in pulmonary microvessels.


Assuntos
Permeabilidade Capilar/efeitos dos fármacos , Circulação Pulmonar/efeitos dos fármacos , Receptores de Trombina/genética , Trombina/farmacologia , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacologia , Animais , Relação Dose-Resposta a Droga , Genótipo , Técnicas In Vitro , Pulmão/irrigação sanguínea , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Masculino , Camundongos , Camundongos Knockout , Cadeias Leves de Miosina/metabolismo , Oligopeptídeos/farmacologia , Fosforilação/efeitos dos fármacos , RNA/genética , RNA/metabolismo , Receptor PAR-1 , Receptores de Trombina/fisiologia , Vasoconstritores/farmacologia
19.
J Invest Dermatol ; 93(2 Suppl): 62S-67S, 1989 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-2546995

RESUMO

The endothelial barrier in all organ beds allows the free exchange of water, but is restrictive to varying degrees to the transport of solutes such as albumin. For example, in the brain microvessels, the endothelial barrier restricts the transport of protein, whereas in fenestrated and continuous endothelial cells of the renal and lung endothelial cells, the endothelial barrier is semipermeable. The endothelial monolayer demonstrates selectivity, i.e., the permeation of molecules is inversely related to the molecular weight. Although the "pore" theory has been used to describe the transport across the endothelial barrier, the transport of solutes is also dependent on the charge of solutes and the endothelial cell, and the ability of the solute to bind to or be taken up by endothelial cells. Receptor-mediated trancytosis of albumin may contribute to albumin transport in addition to transport by paracellular pathways (i.e., through a so-called pore). Water permeability across the endothelium is determined by the interaction of albumin with glycocalyx and interstitial components of the endothelium (the "fiber matrix"). Ambient concentration of albumin serves to lower endothelial hydraulic conductivity. Increased endothelial permeability to solutes and water in inflammatory states is dependent on the shape and configuration of endothelial cells as determined by alterations in cytoskeletal elements, such as f-actin, and as regulated by intracellular second messengers such as free cytosolic calcium.


Assuntos
Permeabilidade Capilar , Endotélio Vascular/metabolismo , Animais , Transporte Biológico Ativo , Células Cultivadas , Técnicas Citológicas , Eletroquímica , Endotélio Vascular/citologia , Humanos , Receptores de Albumina , Receptores de Superfície Celular/fisiologia , Albumina Sérica/metabolismo , Água/metabolismo
20.
FEBS Lett ; 470(1): 70-6, 2000 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-10722848

RESUMO

C-terminal kinesin motor proteins, such as the Drosophila NCD and yeast KAR3, are involved in chromosomal segregation. Previously we have described two orthologs of NCD in Caenorhabditis elegans, KLP-3 and KLP-17, which also participate in chromosome movement. Here we report cDNA cloning of klp-15 and klp-16, and the expression pattern of the genes encoding C-terminal motor kinesins including klp-15 and klp-16. Interestingly KLP-15 and KLP-16 form a unique class of C-terminal kinesins, distinct from the previously known C-terminal motors in other organisms. Using in situ hybridization and RNA interference assay, we show that although all of these motors mediate chromosome segregation, they do so in a combination of unique and overlapping manners, suggesting a complex hierarchy of kinesin motor function in metazoans.


Assuntos
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans/genética , Proteínas de Helminto/fisiologia , Cinesinas/fisiologia , Proteínas Associadas aos Microtúbulos , Sequência de Aminoácidos , Animais , Sequência de Bases , Northern Blotting , Caenorhabditis elegans/embriologia , Segregação de Cromossomos , Clonagem Molecular , DNA de Helmintos , Expressão Gênica , Proteínas de Helminto/genética , Hibridização In Situ , Cinesinas/genética , Dados de Sequência Molecular , RNA de Helmintos , Homologia de Sequência de Aminoácidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA