Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Mol Pharmacol ; 84(6): 899-910, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24077967

RESUMO

AP301 [Cyclo(CGQRETPEGAEAKPWYC)], a cyclic peptide comprising the human tumor necrosis factor lectin-like domain (TIP domain) sequence, is currently being developed as a treatment for lung edema and has been shown to reduce extravascular lung water and improve lung function in mouse, rat, and pig models. The current paradigm for liquid homeostasis in the adult mammalian lung is that passive apical uptake of sodium via the amiloride-sensitive epithelial Na⁺ channel (ENaC) and nonselective cyclic-nucleotide-gated cation channels creates the major driving force for reabsorption of water through the alveolar epithelium in addition to other ion channels such as potassium and chloride channels. AP301 can increase amiloride-sensitive current in A549 cells as well as in freshly isolated type II alveolar epithelial cells from different species. ENaC is expressed endogenously in all of these cell types. Consequently, this study was undertaken to determine whether ENaC is the specific target of AP301. The effect of AP301 in A549 cells as well as in human embryonic kidney cells and Chinese hamster ovary cells heterologously expressing human ENaC subunits (α, ß, γ, and δ) was measured in patch clamp experiments. The congener TIP peptide AP318 [Cyclo(4-aminobutanoic acid-GQRETPEGAEAKPWYD)] activated ENaC by increasing single-channel open probability. AP301 increased current in proteolytically activated (cleaved) but not near-silent (uncleaved) ENaC in a reversible manner. αßγ- or δßγ-ENaC coexpression was required for maximal activity. No increase in current was observed after deglycosylation of extracellular domains of ENaC. Thus, our data suggest that the specific interaction of AP301 with both endogenously and heterologously expressed ENaC requires precedent binding to glycosylated extracellular loop(s).


Assuntos
Canais Epiteliais de Sódio/metabolismo , Peptídeos Cíclicos/farmacologia , Edema Pulmonar/tratamento farmacológico , Animais , Células CHO , Linhagem Celular Tumoral , Cricetinae , Cricetulus , Glicosilação , Células HEK293 , Humanos , Técnicas de Patch-Clamp , Subunidades Proteicas/metabolismo
2.
Pulm Pharmacol Ther ; 26(3): 356-63, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23313096

RESUMO

Pulmonary permeability oedema is a frequent complication in a number of life-threatening lung conditions, such as ALI and ARDS. Apart from ventilation strategies, no specific therapy yet exists for treatment of these potentially fatal illnesses. The oedema-reducing capacity of the lectin-like domain of TNF (TIP) and of synthetic peptides, mTIP and hTIP, which mimic the TIP domain of mouse and human TNF, have been demonstrated in various studies in rodents. Cell-based electrophysiological studies have revealed that the alveolar fluid clearing capacity of TNF and the TIP peptides is due to activation of the amiloride-sensitive Na(+) current in alveolar epithelial cells and that the primary site of action is on the apical side of these cells. AP301, a synthetic cyclic peptide mimicking the TIP domain of human TNF is currently undergoing clinical trials as a therapy for pulmonary permeability oedema. AP301 has been shown to improve alveolar liquid clearance and lung function in a porcine model of ALI. For non-clinical regulatory assessment, dog, pig and rat are standard animal models; accordingly, pre-clinical toxicological and pharmacological safety studies have been conducted with AP301 in dogs and rats. Hitherto, no studies have assessed the pharmacodynamic effect of AP301 on primary canine or porcine type II AEC. The current study describes the effect of AP301 on the amiloride-sensitive Na(+) current in type II AEC isolated from dog, pig and rat lungs. In whole cell patch clamp experiments with dog type II AEC, an increase in the amiloride-sensitive Na(+) current from 3.7 pA to 49.4 pA was observed in the presence of AP301; in pig type II AEC, an increase from 10.0 pA to 159.6 pA was observed, and in rat AEC, from 6.9 pA to 62.4 pA. In whole cell patch clamp experiments in A549 cells, AP301-induced enhancement of the amiloride-sensitive current was eliminated when Na(+) in the bath solution was replaced with N-methyl-d-glucamine (NMDG), and when the cells were pre-incubated with 5-aminoimidazole-4-carboxamide-1-ß-d-ribofuranoside (AICAR), an inhibitor of ENaC, but enhancement was unaffected by addition of cyclic nucleotide-gated (CNG) channel inhibitors Zn(2+) or l-cis-diltiazem prior to AP301. These results provide strong evidence that AP301 activates the amiloride-sensitive Na(+) current through ENaC in type II AEC from dog, pig and rat. To our knowledge, this is the first cell-based analysis of the oedema-clearing effect of AP301 observed in the porcine model of pulmonary oedema. Furthermore, the results validate the dog and pig models in non-clinical assessment of AP301.


Assuntos
Canais Epiteliais de Sódio/metabolismo , Peptídeos Cíclicos/farmacologia , Alvéolos Pulmonares/metabolismo , Amilorida/farmacologia , Animais , Linhagem Celular Tumoral , Diltiazem/farmacologia , Cães , Relação Dose-Resposta a Droga , Lectinas/farmacologia , Peptídeos Cíclicos/metabolismo , Ratos , Suínos , Fator de Necrose Tumoral alfa/farmacologia , Zinco/farmacologia
3.
J Immunol ; 186(3): 1608-17, 2011 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-21178007

RESUMO

The pore-forming toxin Panton-Valentine leukocidin (PVL) is carried by community-acquired methicillin-resistant Staphylococcus aureus and associated with necrotizing pneumonia together with poor prognosis of infected patients. Although the cell-death-inducing properties of PVL have previously been examined, the pulmonary immune response to PVL is largely unknown. Using an unbiased transcriptional profiling approach, we show that PVL induces only 29 genes in mouse alveolar macrophages, which are associated with TLR signaling. Further studies indicate that PVL directly binds to TLR2 and induces immune responses via NF-κB in a TLR2, CD14, MyD88, IL-1R-associated kinase 1, and TNFR-associated factor 6-dependent manner. PVL-mediated inflammation is independent of pore formation but strongly depends on the LukS subunit and is suppressed in CD14/TLR2(-/-) cells. In vivo PVL or LukS induced a robust inflammatory response in lungs, which was diminished in CD14/TLR2(-/-) mice. These results highlight the proinflammatory properties of PVL and identify CD14/TLR2 as an essential receptor complex for PVL-induced lung inflammation.


Assuntos
Toxinas Bacterianas/toxicidade , Exotoxinas/toxicidade , Imunidade Inata , Mediadores da Inflamação/fisiologia , Leucocidinas/toxicidade , Receptores de Lipopolissacarídeos/fisiologia , Staphylococcus aureus Resistente à Meticilina/imunologia , Pneumonia/imunologia , Pneumonia/patologia , Receptor 2 Toll-Like/fisiologia , Animais , Linhagem Celular , Humanos , Imunidade Inata/genética , Mediadores da Inflamação/metabolismo , Interleucina-8/biossíntese , Interleucina-8/metabolismo , Receptores de Lipopolissacarídeos/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/patologia , Receptor 2 Toll-Like/deficiência , Receptor 2 Toll-Like/genética , Regulação para Cima/genética , Regulação para Cima/imunologia
4.
Biochem Pharmacol ; 98(4): 740-53, 2015 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-26254591

RESUMO

Dysfunction of the epithelial sodium channel (ENaC), which regulates salt and water homeostasis in epithelia, causes several human pathological conditions, including pulmonary oedema. This is a potentially lethal complication of acute lung injury at least partially caused by dysfunctional alveolar liquid clearance, which in turn impairs alveolar gas exchange. Solnatide (named TIP-peptide, AP301), a 17 residue peptide mimicking the lectin-like domain of TNF has been shown to activate ENaC in several experimental animal models of acute lung injury and is being evaluated as a potential therapy for pulmonary oedema. The peptide has recently completed phase 1 and 2a clinical trials. In this study, we identify a glycosylation-dependent mechanism that preserves ENaC function and expression. Since our previous data suggested that the pore-forming subunits of ENaC are essential for maximal current activation by solnatide, we performed single- and multi-N-glycosylation site mutations in αN232,293,312,397,511Q- and δN166,211,384Q-subunits, in order to identify crucial residues for interaction with solnatide within the extracellular loop of the channel. Additionally, we generated αL576X and αN232,293,312,397,511Q,L576X deletion mutants of ENaC-α, since we have previously demonstrated that the carboxy terminal domain of this subunit is also involved in its interaction with solnatide. In cells expressing αN232,293,312,397,511Q,L576Xßγ-hENaC or δN166,311,384Q,D552Xßγ-hENaC activation by solnatide, as measured in whole cell patch clamp mode, was completely abolished, whereas it was attenuated in αL576Xßγ-hENaC- and δD552Xßγ-hENaC-expressing cells. Taken together, our findings delineate an N-glycan dependent interaction between the TIP-peptide and ENaC leading to normalization of both sodium and fluid absorption in oedematous alveoli to non-oedematous levels.


Assuntos
Canais Epiteliais de Sódio/metabolismo , Peptídeos Cíclicos/química , Peptídeos Cíclicos/metabolismo , Glicosilação , Células HEK293 , Humanos
5.
J Med Chem ; 53(22): 8021-9, 2010 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-20979368

RESUMO

The amiloride-sensitive epithelial sodium channel (ENaC) plays a prominent role in sodium uptake from alveolar fluid and is the major component in alveolar fluid clearance in normal and diseased lungs. The lectin-like domain of TNF-α has been shown to activate amiloride-sensitive sodium uptake in type II alveolar epithelial cells. Therefore, several synthetic peptides that mimic the lectin-like domain of TNF-α (TIP) were synthesized and their ability to enhance sodium current through ENaC was studied in A549 cells with the patch clamp technique. Our data suggest that a free positively charged N-terminal amino group on residue 1 and/or a free negatively charged carboxyl group on residue 17 of the TIP peptide is essential for the ENaC-activating effect. Ventilation strategies apart, no standard treatment exists for pulmonary permeability edema. Therefore, novel therapies activating sodium uptake from the alveolar fluid via ENaC could improve clinical outcome.


Assuntos
Agonistas do Canal de Sódio Epitelial , Lectinas/química , Peptídeos Cíclicos/síntese química , Peptidomiméticos/síntese química , Fator de Necrose Tumoral alfa/química , Amilorida/farmacologia , Cátions Monovalentes , Linhagem Celular Tumoral , Canais Epiteliais de Sódio/fisiologia , Humanos , Modelos Moleculares , Técnicas de Patch-Clamp , Peptídeos Cíclicos/química , Peptídeos Cíclicos/farmacologia , Peptidomiméticos/química , Peptidomiméticos/farmacologia , Potássio/metabolismo , Estrutura Terciária de Proteína , Sódio/metabolismo , Relação Estrutura-Atividade
6.
J Immunol ; 180(6): 4116-23, 2008 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-18322222

RESUMO

Type I IFN (IFN-I) signaling is detrimental to cells and mice infected with Listeria monocytogenes. In this study, we investigate the impact of IFN-I on the activity of listeriolysin O (LLO), a pore-forming toxin and virulence protein released by L. monocytogenes. Treatment of macrophages with IFN-beta increased the ability of sublytic LLO concentrations to cause transient permeability of the plasma membrane. At higher LLO concentrations, IFN-beta enhanced the complete breakdown of membrane integrity and cell death. This activity of IFN-beta required Stat1. Perturbation of the plasma membrane by LLO resulted in activation of the p38MAPK pathway. IFN-beta pretreatment enhanced LLO-mediated signaling through this pathway, consistent with its ability to increase membrane damage. p38MAPK activation in response to LLO was independent of TLR4, a putative LLO receptor, and inhibition of p38MAPK neither enhanced nor prevented LLO-induced death. IFN-beta caused cells to express increased amounts of caspase 1 and to produce a detectable caspase 1 cleavage product after LLO treatment. Contrasting recent reports with another pore-forming toxin, this pathway did not aid cell survival as caspase 1-deficient cells were equally sensitive to lysis by LLO. Key lipogenesis enzymes were suppressed in IFN-beta-treated cells, which may exacerbate the membrane damage caused by LLO.


Assuntos
Permeabilidade da Membrana Celular/imunologia , Proteínas de Choque Térmico/fisiologia , Proteínas Hemolisinas/fisiologia , Interferon beta/fisiologia , Macrófagos/citologia , Macrófagos/imunologia , Regulação para Cima/imunologia , Adjuvantes Imunológicos/fisiologia , Adjuvantes Imunológicos/toxicidade , Animais , Toxinas Bacterianas/toxicidade , Morte Celular/imunologia , Células Cultivadas , Citotoxinas/fisiologia , Proteínas de Choque Térmico/toxicidade , Proteínas Hemolisinas/toxicidade , Interferon beta/toxicidade , Células L , Listeria monocytogenes/crescimento & desenvolvimento , Listeria monocytogenes/imunologia , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais/imunologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa