Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
J Biol Chem ; 290(24): 14884-92, 2015 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-25931117

RESUMEN

Autophagy is the targeted degradation of proteins and organelles critical for homeostasis and cell survival. Transforming growth factor ß (TGF-ß) differentially regulates autophagy in a context-specific manner, although the precise intracellular mechanisms remain less clear. Importantly, how TGF-ß controls autophagic responses in endothelial cells (EC) during angiogenesis is unknown. Here we identified endoglin, an EC-specific TGF-ß co-receptor essential for angiogenesis, as a key determinant of autophagy. Among the two opposing TGF-ß Smad pathways in the EC system (Smad1/5/8 and Smad2/3), we found Smad2 as the major transcriptional regulator of autophagy that targets beclin1 (BECN1) gene expression. Smad2, but not Smad3, acts as a repressor upstream of the BECN1 promoter region. Overall, endoglin promotes autophagy by impeding Smad2 transcriptional repressor activity. Notably, increased beclin1 levels upon Smad2 knockdown directly correlated with enhanced autophagy during angiogenesis. Taken together, these results establish endoglin as a critical mediator of autophagy and demonstrate a new transcriptional mechanism by which Smad2 inhibits angiogenesis.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Autofagia/fisiología , Endotelio/metabolismo , Péptidos y Proteínas de Señalización Intracelular/fisiología , Proteína Smad2/fisiología , Animales , Secuencia de Bases , Beclina-1 , Células Cultivadas , Inmunoprecipitación de Cromatina , Cartilla de ADN , Endoglina , Endotelio/citología , Ratones , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína Smad2/genética
2.
J Biol Chem ; 289(37): 25486-96, 2014 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-25070888

RESUMEN

Endoglin is a transforming growth factor ß (TGF-ß) co-receptor essential for angiogenesis and tumor vascularization. Endoglin modulates the crucial balance between pro- and anti-angiogenic signaling by activin receptor-like kinase (ALK) 1, 5, and TGF-ß type II (TßRII) receptors. Despite its established role in physiology and disease, the mechanism of endoglin down-regulation remains unknown. Here we report that the conserved juxtamembrane cytoplasmic tyrosine motif ((612)YIY(614)) is a critical determinant of angiogenesis. Src directly phosphorylates this motif to induce endoglin internalization and degradation via the lysosome. We identified epidermal growth factor (EGF) and vascular endothelial growth factor (VEGF) as Src-activators that induce endoglin turnover following (612)YIY(614) phosphorylation. Interestingly, Src phosphorylation of endoglin-(612)YIY(614) was also an important process for receptor down-regulation by TRACON105 (TRC105), an endoglin-targeting antibody currently in clinical trials. The regulation of (612)YIY(614) phosphorylation was critical for angiogenesis, as both the phosphomimetic and unphosphorylatable mutants impaired endothelial functions including proliferation, migration, and capillary tube formation. Collectively, these findings establish Src and pro-angiogenic mitogens as critical mediators of endoglin stability and function.


Asunto(s)
Secuencias de Aminoácidos/genética , Antígenos CD/genética , Neovascularización Fisiológica/genética , Receptores de Superficie Celular/genética , Familia-src Quinasas/metabolismo , Receptores de Activinas Tipo II/genética , Receptores de Activinas Tipo II/metabolismo , Proliferación Celular/genética , Endoglina , Células Endoteliales/citología , Células Endoteliales/metabolismo , Humanos , Fosforilación , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Estabilidad Proteica , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo
3.
Biochem Biophys Res Commun ; 452(3): 468-72, 2014 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-25159840

RESUMEN

Peptidylprolyl cis/trans isomerase, NIMA-interacting 1 (PIN1) modulates phospho-signaling by catalyzing rotation of the bond between a phosphorylated serine or threonine before proline in proteins. As depletion of PIN1 increased inflammatory protein expression in cultured endothelial cells treated with bacterial endotoxin (lipopolysaccharide, LPS) and interferon-γ, we hypothesized that PIN1 knockout would increase sensitivity to LPS-induced lung inflammation in mice. Mortality due to a high dose of LPS (30mg/kg) was greater in knockout than wildtype mice. Lung myeloperoxidase activity, reflecting neutrophils, was increased to a 35% higher level in PIN1 knockout mouse lung, as compared with wildtype, after treatment with a sublethal dose of 3mgLPS/kg, ip. Unexpectedly, plasma tumor necrosis factor-α (TNF) was approximately 50% less than in wildtype mice. Knockout mice, however, were more sensitive than wildtype to TNF-induced neutrophil accumulation. The neutrophil adhesion molecule, E-selectin, was also elevated in lungs of knockout mice treated with TNF, suggesting that PIN1 depletion increases endothelial sensitivity to TNF. Indeed, TNF induced more reactive oxygen species in cultured endothelial cells depleted of PIN1 with short hairpin RNA than in control cells. Collectively, the results indicate that while PIN1 normally facilitates TNF production in LPS-treated mice, it suppresses pulmonary and endothelial reactions to the cytokine. Tissue or cell-specific effects of PIN1 may affect the overall inflammatory response to LPS and other stimuli.


Asunto(s)
Pulmón/enzimología , Isomerasa de Peptidilprolil/genética , Neumonía/enzimología , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Células Cultivadas , Selectina E/genética , Selectina E/metabolismo , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Expresión Génica , Lipopolisacáridos/farmacología , Pulmón/efectos de los fármacos , Pulmón/patología , Ratones , Ratones Noqueados , Peptidilprolil Isomerasa de Interacción con NIMA , Infiltración Neutrófila/efectos de los fármacos , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , Neutrófilos/patología , Isomerasa de Peptidilprolil/deficiencia , Neumonía/inducido químicamente , Neumonía/genética , Neumonía/patología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Factor de Necrosis Tumoral alfa/farmacología
4.
Biochem Biophys Res Commun ; 414(3): 581-6, 2011 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-21982763

RESUMEN

The peptidyl-proline isomerase, protein never in mitosis gene A interacting-1 (PIN1) binds and isomerizes proteins phosphorylated on serine/threonine before a proline. It was previously found that depletion of PIN1 greatly increased induction of cyclooxygenase-2 and inducible nitric oxide synthase by lowering calpain activity in murine aortic endothelial cells (MAEC). Here we investigated the effect of PIN1 on the endogenous inhibitor of heterodimeric µ- and m-calpains, calpastatin. MAEC were transduced with small hairpin (sh) RNA to knock down PIN1 (KD) or an inactive Control shRNA. Cells were also treated with non-targeted double stranded small inhibitory RNA (siRNA) or siRNA designed to deplete calpastatin. Despite reducing calpain activity, PIN1 KD did not significantly affect the expression of µ- and m-calpains, or calpastatin, compared to Control shRNA. Instead, depletion of PIN1 increased the inhibitory activity of calpastatin. Calpastatin co-immunoprecipitated with endogenous PIN1 and was pulled down with glutathione-S-transferase (GST)-PIN1 fusion protein. Adding GST-PIN1 to KD cell extracts lacking PIN1 reduced calpastatin inhibitory activity. Substrate binding and catalytic domain mutants of PIN1 failed to do so. These results suggest that protein interaction and the proline isomerase functions of PIN1 are required for it to inhibit calpastatin. Furthermore, depletion of calpastatin raised calpain activity and reduced calpain inhibitory activity to similar levels in KD and Control MAEC, indicating that calpastatin is required for PIN1 depletion to lower calpain activity. Thus, PIN1 apparently restrains the ability of calpastatin to inhibit calpain, maintaining calpain activity in endothelial cells. PIN1 may act directly via phosphorylated serine/threonine-proline motifs in calpastatin, or indirectly via other PIN1 substrates that control calpastatin.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Calpaína/antagonistas & inhibidores , Endotelio Vascular/metabolismo , Isomerasa de Peptidilprolil/metabolismo , Animales , Endotelio Vascular/enzimología , Técnicas de Silenciamiento del Gen , Ratones , Mitosis , Peptidilprolil Isomerasa de Interacción con NIMA , Isomerasa de Peptidilprolil/genética , Fosforilación , ARN Interferente Pequeño/genética
5.
Mol Cancer ; 9: 217, 2010 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-20712901

RESUMEN

BACKGROUND: Targeting Signal Transducer and Activator of Transcription 3 (STAT3) signaling is an attractive therapeutic approach for most types of human cancers with constitutively activated STAT3. A novel small molecular STAT3 inhibitor, FLLL32 was specifically designed from dietary agent, curcumin to inhibit constitutive STAT3 signaling in multiple myeloma, glioblastoma, liver cancer, and colorectal cancer cells. RESULTS: FLLL32 was found to be a potent inhibitor of STAT3 phosphorylation, STAT3 DNA binding activity, and the expression of STAT3 downstream target genes in vitro, leading to the inhibition of cell proliferation as well as the induction of Caspase-3 and PARP cleavages in human multiple myeloma, glioblastoma, liver cancer, and colorectal cancer cell lines. However, FLLL32 exhibited little inhibition on some tyrosine kinases containing SH2 or both SH2 and SH3 domains, and other protein and lipid kinases using a kinase profile assay. FLLL32 was also more potent than four previously reported JAK2 and STAT3 inhibitors as well as curcumin to inhibit cell viability in these cancer cells. Furthermore, FLLL32 selectively inhibited the induction of STAT3 phosphorylation by Interleukin-6 but not STAT1 phosphorylation by IFN-γ. CONCLUSION: Our findings indicate that FLLL32 exhibits potent inhibitory activity to STAT3 and has potential for targeting multiple myeloma, glioblastoma, liver cancer, and colorectal cancer cells expressing constitutive STAT3 signaling.


Asunto(s)
División Celular/efectos de los fármacos , Curcumina/análogos & derivados , ADN/efectos de los fármacos , Factor de Transcripción STAT3/antagonistas & inhibidores , Línea Celular Tumoral , Curcumina/farmacología , ADN/metabolismo , Humanos , Fosforilación , Factor de Transcripción STAT3/metabolismo
6.
Mol Cancer ; 9: 165, 2010 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-20576164

RESUMEN

BACKGROUND: We characterized the biologic effects of a novel small molecule STAT3 pathway inhibitor that is derived from the natural product curcumin. We hypothesized this lead compound would specifically inhibit the STAT3 signaling pathway to induce apoptosis in melanoma cells. RESULTS: FLLL32 specifically reduced STAT3 phosphorylation at Tyr705 (pSTAT3) and induced apoptosis at micromolar amounts in human melanoma cell lines and primary melanoma cultures as determined by annexin V/propidium iodide staining and immunoblot analysis. FLLL32 treatment reduced expression of STAT3-target genes, induced caspase-dependent apoptosis, and reduced mitochondrial membrane potential. FLLL32 displayed specificity for STAT3 over other homologous STAT proteins. In contrast to other STAT3 pathway inhibitors (WP1066, JSI-124, Stattic), FLLL32 did not abrogate IFN-gamma-induced pSTAT1 or downstream STAT1-mediated gene expression as determined by Real Time PCR. In addition, FLLL32 did not adversely affect the function or viability of immune cells from normal donors. In peripheral blood mononuclear cells (PBMCs), FLLL32 inhibited IL-6-induced pSTAT3 but did not reduce signaling in response to immunostimulatory cytokines (IFN-gamma, IL 2). Treatment of PBMCs or natural killer (NK) cells with FLLL32 also did not decrease viability or granzyme b and IFN-gamma production when cultured with K562 targets as compared to vehicle (DMSO). CONCLUSIONS: These data suggest that FLLL32 represents a lead compound that could serve as a platform for further optimization to develop improved STAT3 specific inhibitors for melanoma therapy.


Asunto(s)
Apoptosis/efectos de los fármacos , Curcumina/análogos & derivados , Citocinas/fisiología , Melanoma/patología , Factor de Transcripción STAT3/antagonistas & inhibidores , Western Blotting , Línea Celular Tumoral , Curcumina/farmacología , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Expresión Génica/fisiología , Humanos , Melanoma/inmunología , Melanoma/metabolismo , Reacción en Cadena de la Polimerasa , Transducción de Señal/fisiología
7.
Oncotarget ; 8(8): 12675-12685, 2017 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-27177332

RESUMEN

Angiogenesis is the formation of new blood vessels from existing vasculature critical for embryonic development and vascular remodeling. Its dysregulation underlies numerous pathologic states ranging from ischemia to tumor growth and as such identifying new targeted- therapies is of significant interest for angiogenesis-based medicine. Here we evaluated the potential angiostatic properties of capsicodendrin (CPCD), a natural compound isolated from Cinnamosma macrocarpa, a plant belonging to the Malagasy Cinnamosma. CPCD potently inhibits endothelial proliferation, migration and capillary tube formation at nanomolar to low micromolar concentrations without inducing cytotoxic effects. We show that CPCD directly inactivates VEGFR2 and downstream AKT signaling, thereby strongly inducing autophagy as determined by increased expression of beclin1, autophagy-related gene (Atg) 3, Atg5 and LC3 cleavage. Ectopic AKT overexpression counteracts the inhibitory effects of CPCD on proliferation and capillary tubule formation. Importantly, CPCD treatment in vivo inhibits sprouting angiogenesis as evidenced by strongly reduced intersegmental vessel (ISV) sprouting and subintestinal vessel (SIV) formation during zebrafish embryonic development, and correlates with increased presence of LC3II along the ISVs despite overall reduced vasculature. These findings demonstrate CPCD as a potent inhibitor of the VEGFR2/AKT pathway at nanomolar concentrations and inducer of autophagy-related angiostatic effects.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Neovascularización Fisiológica/efectos de los fármacos , Extractos Vegetales/farmacología , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Animales , Autofagia/efectos de los fármacos , Línea Celular , Células Endoteliales/efectos de los fármacos , Técnica del Anticuerpo Fluorescente , Humanos , Magnoliaceae , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/efectos de los fármacos , Pez Cebra
8.
Clin Cancer Res ; 11(8): 2998-3008, 2005 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-15837754

RESUMEN

PURPOSE: Down-regulation of Bcl-2 by the antisense G3139, currently under clinical evaluations, could restore chemosensitivity in otherwise resistant malignant cells. To date, the mechanism of intracellular accumulation of G3139 following in vivo administration remains to be elucidated. This study aimed to assess whether detectable intracellular concentrations of G3139 are achievable in vivo and how these relate to Bcl-2 down-regulation. EXPERIMENTAL DESIGN: Cellular uptake of G3139 was studied in leukemia myeloid cell lines and blasts collected from treated patients using a newly developed, novel, and highly sensitive ELISA-based assay. Real-time reverse transcription-PCR was used to quantify Bcl-2 mRNA changes in treated cells. RESULTS: The assay was fully validated and showed a limit of quantification of 50 pmol/L. When exposed to 0.33 to 10 mumol/L G3139, K562 cells exhibited intracellular concentrations in the range of 2.1 to 11.4 pmol/mg protein. When G3139 was delivered with cationic lipids, a 10- to 25-fold increase of the intracellular concentrations was observed. There was an accumulation of G3139 in the nuclei, and the ratio of nucleus to cytoplasm was increased 7-fold by cationic lipids. Intracellular concentrations of G3139 were correlated with Bcl-2 mRNA down-regulation. Robust intracellular concentrations of G3139 were achieved in vivo in bone marrow (range, 3.4-40.6 pmol/mg protein) and peripheral blood mononuclear cells (range, 0.47-19.4 pmol/mg protein) from acute myeloid leukemia patients treated with G3139. CONCLUSIONS: This is the first evidence that measurable intracellular levels of G3139 are achievable in vivo in acute myeloid leukemia patients and that Bcl-2 down-regulation is likely to depend on the achievable intracellular concentrations rather than on plasma concentrations.


Asunto(s)
Leucemia Mieloide/metabolismo , Oligonucleótidos Antisentido/farmacocinética , Proteínas Proto-Oncogénicas c-bcl-2/genética , Tionucleótidos/farmacocinética , Enfermedad Aguda , Secuencia de Bases , Línea Celular Tumoral , Cromatografía Líquida de Alta Presión/métodos , Ensayo de Inmunoadsorción Enzimática/métodos , Humanos , Células K562 , Leucemia Mieloide/genética , Leucemia Mieloide/patología , Oligonucleótidos Antisentido/sangre , Oligonucleótidos Antisentido/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reproducibilidad de los Resultados , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tionucleótidos/sangre , Tionucleótidos/metabolismo , Factores de Tiempo
9.
Biochem Pharmacol ; 65(7): 1189-97, 2003 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-12663054

RESUMEN

Angiotensin II (ANG II) participates in many cardiovascular disease states, but the mechanisms involved are not completely defined. Doses of ANG II that do not affect blood pressure significantly can still cause early changes in vascular endothelial performance and cell-specific protein 3-nitrotyrosine formation (protein-3NT, marker of peroxynitrite formation) in vivo. Here, we have tested the hypothesis that ANG II induces endothelial cell peroxynitrite (ONOO-) formation in vitro, and investigated the mechanisms involved. Endothelial cells were incubated with ANG II (1nM-250 microM), and protein nitration was assessed by immunoblotting. ANG II caused concentration-dependent increases in protein-3NT above detectable basal control levels, at concentrations greater than 100nM. This response was inhibited significantly by co-incubation with losartan or diphenyleneiodonium chloride. Endothelial cell lysates incubated with nitrated protein standards demonstrated significant protein-3NT modification activity only in the presence of serum. However, endothelial cell lysates did not modify the free amino acid form of 3NT (free-3NT) in identical experimental conditions, assessed by capillary electrophoresis. Finally, free-3NT was cytotoxic to cultured endothelial cells (fitted LC(50)=98 microM). These data demonstrate that stimulation of angiotensin receptor subtype 1 by ANG II can cause increased endothelial cell protein nitration in vitro in the absence of other cell types or stimuli, at concentrations that are pathophysiologically relevant. Furthermore, endothelial cells selectively modified nitrated protein tyrosine residues only in the presence of a cofactor(s), and did not modify the free modified amino acid. Protein nitration may be a regulated endothelial signaling process, while free-3NT may be toxic to endothelial cells.


Asunto(s)
Angiotensina II/farmacología , Endotelio Vascular/efectos de los fármacos , Especies de Nitrógeno Reactivo/metabolismo , Tirosina/análogos & derivados , Animales , Endotelio Vascular/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Tirosina/metabolismo
10.
Cardiovasc Toxicol ; 4(2): 199-206, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15371635

RESUMEN

HIV-protease inhibitor (HIV-PI) drugs are critical for highly active antiretroviral therapy (HAART) efficacy, but several recent reports have suggested that metabolic and/or cardiovascular toxicities are associated with these drugs. Given the importance of the HIV-PI drug class and the widespread and chronic use of these agents in an expanding patient population, further understanding of this potential drug toxicity is imperative. Here, we investigated a role for direct endothelial toxicity induced by saquinavir (SAQ), the first HIV-PI drug marketed in the United States and still an important component of HAART therapies. In initial studies using isolated vascular tissues, we observed selective impairment of endothelium-dependent vasodilation with no effect on contractile responses. Subsequent studies using human endothelial cells in culture at clinically relevant concentrations (5 and 10 microM, 2-48 h) demonstrated concentration-dependent increases in cell death, mainly via apoptosis rather than necrosis (determined via Annexin-V positive membrane labeling). Live cell imaging also demonstrated increased intracellular oxidant production (as measured by DCF fluorescence), which could be abrogated by incubation with the antioxidant N-acetylcysteine (NAC). NAC also prevented SAQ- induced apoptotic cell death. These data demonstrate that SAQ has direct toxicological effects on endothelial cells, and that the toxicity apparently involves apoptotic pathway activation via reactive oxygen and/or nitrogen species.


Asunto(s)
Apoptosis , Células Endoteliales/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Inhibidores de la Proteasa del VIH/toxicidad , Saquinavir/toxicidad , Animales , Aorta Torácica/efectos de los fármacos , Aorta Torácica/fisiología , Línea Celular , Supervivencia Celular/efectos de los fármacos , Células Endoteliales/citología , Endotelio Vascular/citología , Endotelio Vascular/fisiología , Humanos , Técnicas In Vitro , Masculino , Necrosis , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Venas Umbilicales , Vasodilatación/efectos de los fármacos
11.
Int J Mol Med ; 12(4): 533-40, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12964031

RESUMEN

Endotoxin (LPS) is a cause of adult respiratory distress syndrome (ARDS), a disease which is preceded by acute lung injury involving the pulmonary endothelium. Experimentally, LPS causes acute DNA strand breakage in mouse lung endothelial cells (MLEC). Engagement of integrin cell adhesion receptors inhibits acute DNA breakage, which could be of use in reducing lung injury. Because integrins presumably inhibit DNA damage or activate repair, we hypothesized that the DNA-damage response protein, poly(ADP-ribose) polymerase-1 (PARP-1), regulates the protective action of integrins, as well as sensitivity to LPS. Therefore, the effect of LPS on MLEC cultured from wild-type and PARP-1 knockout mice was determined. Fluorescence microscopic measures were used to assess plasma membrane integrity, PARP activity, DNA strand breakage and DNA repair in attached cells. LPS caused a concentration-dependent increase in the permeability of wild-type MLEC. Engagement of beta1 integrins with an antibody protected wild-type MLEC from this LPS-induced injury. Wild-type cells treated with the PARP-inhibitor, 3-aminobenzamide, and PARP-1 knockout MLEC were also resistant. LPS caused acute DNA breakage in both wild-type and knockout MLEC, but PARP was activated only in wild-type cells. LPS-induced DNA breakage was inhibited by 3-aminobenzamide, but not by knockout. Anti-beta1 integrin antibody also inhibited the DNA breakage and PARP activation caused by LPS in wild-type MLEC. However, integrin engagement did not prevent DNA breakage in PARP-1 knockout cells, despite a similar level of beta1 integrin in wild-type and knockout cells. Thus, integrin engagement, 3-aminobenzamide, and PARP-1 deletion protected MLEC from increases in membrane permeability caused by LPS. PARP-1 deletion also impaired the ability of integrin engagement to inhibit LPS-induced DNA breakage, suggesting that knockout may affect nuclear factors necessary for integrin-mediated suppression of LPS-induced DNA breakage.


Asunto(s)
Daño del ADN , Endotelio/metabolismo , Endotoxinas/metabolismo , Eliminación de Gen , Integrinas/metabolismo , Pulmón/citología , Poli(ADP-Ribosa) Polimerasas/genética , Animales , Benzamidas/farmacología , Western Blotting , Caspasas/metabolismo , Membrana Celular/metabolismo , Células Cultivadas , Reparación del ADN , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Cinética , Lipopolisacáridos/química , Pulmón/patología , Ratones , Ratones Noqueados , Microscopía Fluorescente , Factores de Tiempo
12.
In Vitro Cell Dev Biol Anim ; 39(1-2): 43-50, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-12892526

RESUMEN

Given the utility of murine models and the physiological and pathological significance of the aortic endothelium, we developed a simplified, nonenzymatic method for isolation and culture of murine aortic endothelial cells (MAECs). Aortic explants were initially cultured on fibronectin-coated plastic. Murine aortic endothelial cells migrated from the explants and proliferated. This expansion allowed for cultures to be established from the aortas of one or three mice. Murine aortic endothelial cells were then purified from expanded cultures by fluorescence-activated cell sorting for the uptake of 1,1'-dioctadecyl-3,3,3',3'-tetramethyl-indocarbocyanine perchlorate-labeled acetylated low-density lipoprotein. The majority of the cells in expanded cultures were as positive as human umbilical vein endothelial cells labeled in the same way. The most positive half of the labeled MAEC population was placed back in culture, and the cells formed "cobblestone" monolayers at confluence. Smooth muscle alpha-actin, which was present in aortic tissue and to a lesser extent in explant cultures before sorting, was not detected in selected MAECs. Western blotting and immunostaining also demonstrated the presence of the endothelial markers, platelet endothelial cell adhesion molecule-1, factor VIII-related antigen, and Bandeiraea simplicifolia lectin 1 binding. Murine aortic endothelial cells retained expected inflammatory functions: vascular cell adhesion molecule-1 protein was induced by bacterial endotoxin, and NO production was synergistically induced by the combination of endotoxin and interferon-gamma. Our simple, efficient method will facilitate investigations of aortic endothelial cell function in vitro using murine models.


Asunto(s)
Aorta/anatomía & histología , Técnicas de Cultivo de Célula/métodos , Células Endoteliales/citología , Células Endoteliales/metabolismo , Endotelio Vascular/citología , Inflamación/metabolismo , Animales , Separación Celular , Células Cultivadas , Medios de Cultivo/química , Técnicas de Cultivo , Células Endoteliales/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Citometría de Flujo , Humanos , Interferón gamma/farmacología , Lipopolisacáridos/farmacología , Ratones , Ratones Endogámicos C57BL , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico Sintasa de Tipo II , Nitritos/metabolismo , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Molécula 1 de Adhesión Celular Vascular/metabolismo
13.
J Inflamm (Lond) ; 6: 20, 2009 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-19545424

RESUMEN

BACKGROUND: The peptidyl-proline isomerase, Protein Never in Mitosis Gene A Interacting-1 (PIN1), regulates turnover of inducible nitric oxide synthase (iNOS) in murine aortic endothelial cells (MAEC) stimulated with E. coli endotoxin (LPS) and interferon-gamma (IFN). Degradation of iNOS was reduced by a calpain inhibitor, suggesting that PIN1 may affect induction of other calpain-sensitive inflammatory proteins, such as cyclooxygenase (COX)-2, in MAEC. METHODS: MAEC, transduced with lentivirus encoding an inactive control short hairpin (sh) RNA or one targeting PIN1 that reduced PIN1 by 85%, were used. Cells were treated with LPS/IFN, calpain inhibitors (carbobenzoxy-valinyl-phenylalaninal (zVF), PD150606), cycloheximide and COX inhibitors to determine the effect of PIN1 depletion on COX-2 and calpain. RESULTS: LPS or IFN alone did not induce COX-2. However, treatment with 10 mug LPS plus 20 ng IFN per ml induced COX-2 protein 10-fold in Control shRNA MAEC. Induction was significantly greater (47-fold) in PIN1 shRNA cells. COX-2-dependent prostaglandin E2 production increased 3-fold in KD MAEC, but did not increase in Control cells. The additional increase in COX-2 protein due to PIN1 depletion was post-transcriptional, as induction of COX-2 mRNA by LPS/IFN was the same in cells containing or lacking PIN1. Instead, the loss of COX-2 protein, after treatment with cycloheximide to block protein synthesis, was reduced in cells lacking PIN1 in comparison with Control cells, indicating that degradation of the enzyme was reduced. zVF and PD150606 each enhanced the induction of COX-2 by LPS/IFN. zVF also slowed the loss of COX-2 after treatment with cycloheximide, and COX-2 was degraded by exogenous mu-calpain in vitro. In contrast to iNOS, physical interaction between COX-2 and PIN1 was not detected, suggesting that effects of PIN1 on calpain, rather than COX-2 itself, affect COX-2 degradation. While cathepsin activity was unaltered, depletion of PIN1 reduced calpain activity by 55% in comparison with Control shRNA cells. CONCLUSION: PIN1 reduced calpain activity and slowed the degradation of COX-2 in MAEC, an effect recapitulated by an inhibitor of calpain. Given the sensitivity of COX-2 and iNOS to calpain, PIN1 may normally limit induction of these and other calpain substrates by maintaining calpain activity in endothelial cells.

14.
Am J Physiol Cell Physiol ; 295(3): C819-27, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18650263

RESUMEN

The peptidyl-proline isomerase Protein Never in Mitosis Gene A Interacting-1 (PIN1) increases the level or activity of several transcription factors that can induce the inducible nitric oxide (NO) synthase (iNOS). PIN1 can also regulate mRNA and protein turnover. Here, the effect of depletion of PIN1 on induction of iNOS by Escherichia coli endotoxin (LPS) and interferon-gamma (IFNgamma) in murine aortic endothelial cells (MAEC) was determined. Suppression of PIN1 by 85% with small hairpin RNA enhanced the induction of NO and iNOS protein by LPS-IFNgamma. There was no effect on induction of iNOS mRNA, suggesting a posttranscriptional effect. The enhanced levels of iNOS protein were functionally significant since LPS-IFNgamma was cytotoxic to MAEC lacking PIN1 but not MAEC harboring an inactive control construct, and because cytotoxicity was blocked by the NO synthase inhibitor N(omega)-nitro-L-arginine methyl ester. Consistent with posttranscriptional action, knockdown of PIN1 increased the stability of iNOS protein in cycloheximide-treated cells. Furthermore, loss of iNOS was blocked by the calpain inhibitor carbobenzoxy-valinyl-phenylalaninal but not by the selective proteasome inhibitor epoxomicin. Immunoprecipitation indicated that PIN1 can interact with iNOS. Pull down of iNOS with a wild-type glutathione-S-transferase-PIN1 fusion protein, but not with a mutant of the amino terminal phospho-(serine/threonine)-proline binding WW domain of PIN1, indicated that this domain mediates interaction. The results suggest that PIN1 associates with iNOS and can limit its induction by facilitating calpain-mediated degradation in MAEC.


Asunto(s)
Células Endoteliales/enzimología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Isomerasa de Peptidilprolil/metabolismo , Procesamiento Proteico-Postraduccional , Animales , Calpaína/antagonistas & inhibidores , Calpaína/metabolismo , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Dipéptidos/farmacología , Células Endoteliales/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Proteínas de Escherichia coli , Interferón gamma/metabolismo , Lipopolisacáridos/farmacología , Ratones , NG-Nitroarginina Metil Éster/farmacología , Peptidilprolil Isomerasa de Interacción con NIMA , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Oligopéptidos/farmacología , Isomerasa de Peptidilprolil/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma , Unión Proteica , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Interferencia de ARN , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Proteínas Recombinantes/metabolismo , Factores de Tiempo
15.
J Pharmacol Exp Ther ; 321(1): 318-26, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17202402

RESUMEN

Engagement of integrin cell adhesion receptors suppresses bleomycin (BLM)-induced DNA strand breakage in endothelial cells. Previous investigation of cells from poly(ADP-ribose) polymerase (PARP)-1 knockout mice and with an inhibitor of the enzyme indicated that this facilitator of base excision repair (BER) is required for integrin-mediated suppression of DNA strand breakage. Here, small inhibitory RNA (siRNA) was used to assess the requirement for the BER proteins, DNA ligase III (Lig3) alpha, PARP-1, and X-ray repair complementing defective repair in Chinese hamster cells 1 (XRCC1), and for the long-patch BER ligase, DNA ligase I (Lig1), in integrin-mediated protection from BLM-induced DNA breakage. Murine lung endothelial cells (MLECs) were transfected with siRNA, treated with anti-beta1 integrin antibody, and then BLM. 3'-OH in DNA and accumulation of phosphorylated histone H2AX (gammaH2AX), which reflects double-strand breakage, were measured. Integrin antibody inhibited the increases in 3'-OH caused by BLM in MLECs transfected with either control or Lig1 siRNA. However, after knockdown of Lig3alpha, PARP-1, or XRCC1, suppression of DNA breakage by integrin antibody was limited. BLM increased gammaH2AX levels, and integrin treatment inhibited this by 57 to 73% in MLECs transfected with control siRNA. Integrin engagement also inhibited increases in gammaH2AX in BLM-treated cells transfected with Lig1 siRNA. In contrast, Lig3alpha, PARP-1, and XRCC1 siRNAs prevented integrin-mediated inhibition of BLM-induced gammaH2AX levels. The results suggest that the BER proteins, Lig3alpha, PARP-1, and XRCC1, are required for integrin-mediated suppression of BLM-induced DNA breakage.


Asunto(s)
Antimetabolitos Antineoplásicos/toxicidad , Bleomicina/antagonistas & inhibidores , Bleomicina/toxicidad , Daño del ADN , Reparación del ADN/efectos de los fármacos , Células Endoteliales/metabolismo , Integrinas/fisiología , Pulmón/metabolismo , Animales , Western Blotting , Supervivencia Celular/efectos de los fármacos , ADN Ligasa (ATP) , ADN Ligasas/genética , Proteínas de Unión al ADN/genética , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Técnica del Anticuerpo Fluorescente , Proteínas Fluorescentes Verdes/biosíntesis , Proteínas Fluorescentes Verdes/genética , Histonas/genética , Etiquetado Corte-Fin in Situ , Pulmón/citología , Pulmón/efectos de los fármacos , Ratones , Poli(ADP-Ribosa) Polimerasa-1 , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Poli(ADP-Ribosa) Polimerasas/genética , ARN Interferente Pequeño/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína 1 de Reparación por Escisión del Grupo de Complementación Cruzada de las Lesiones por Rayos X
16.
J Inflamm (Lond) ; 4: 22, 2007 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-17997851

RESUMEN

BACKGROUND: It was hypothesized that a pro-atherogenic, high saturated fat and cholesterol diet (HCD) would increase the inflammatory response to E. coli endotoxin (LPS) and increase its concentration in plasma after administration to mice. METHODS: C57Bl/6 mice were fed a HCD or a control diet (CD) for 4 weeks, and then treated with saline, 0.5, 1 or 2 mg LPS/kg, ip. Liver injury (alanine:2-oxoglutarate aminotransferase and aspartate aminotransferase, collagen staining), circulating cytokines (tumor necrosis factor-alpha, interleukin-6 and interferon-gamma), factors that can bind LPS (serum amyloid A, apolipoprotein A1, LPS binding protein, and CD14), and plasma levels of LPS were measured. The hepatic response was assessed by measuring vascular cell adhesion molecule (VCAM)-1, inducible nitric oxide synthase (iNOS) and signal transducer and activator of transcription-1 proteins, and VCAM-1 and iNOS mRNAs. Hepatic mRNA encoding the LPS receptor, Toll like receptor 4, was also determined. RESULTS: Two mg LPS/kg killed 100% of mice fed HCD within 5 d, while no mice fed CD died. All mice treated with 0 to 1 mg LPS/kg survived 24 h. HCD increased plasma alanine:2-oxoglutarate aminotransferase and aspartate aminotransferase, and the enzymes were increased more by LPS in HCD than CD mice. Induction of plasma tumor necrosis factor-alpha, interleukin-6, and interferon-gamma by LPS was greater with HCD than CD. Hepatic VCAM-1 and iNOS protein and mRNA were induced by LPS more in mice fed HCD than CD. Tyrosine phosphorylation of signal transducer and activator of transcription-1 caused by LPS was prolonged in HCD compared with CD mice. Despite the hepatic effects of HCD, diet had no effect on the LPS plasma concentration-time profile. HCD alone did not affect circulating levels of plasma apolipoprotein A1 or LPS binding protein. However, plasma concentrations of serum amyloid A and CD14, and hepatic toll-like receptor-4 mRNA were increased in mice fed HCD. CONCLUSION: HCD increased the sensitivity of mice to LPS without affecting its plasma level. Although increased serum amyloid A and CD14 in the circulation may inhibit LPS actions, their overexpression, along with hepatic toll-like receptor-4 or other factors, may contribute to the heightened sensitivity to LPS.

17.
Pharm Res ; 23(6): 1251-64, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16718617

RESUMEN

PURPOSE: To develop and validate an ultrasensitive and specific hybridization-based enzyme-linked immunosorbent assay method for quantification of two phosphorothioate oligonucleotides (PS ODNs) (G3139 and GTI-2040) in biological fluids. METHODS: This assay was based on hybridization of analytes to the biotin-labeled capture ODNs followed by ligation with digoxigenin-labeled detection ODN. The bound duplex was then detected by anti-digoxigenin-alkaline phosphatase using Attophos (Promega, Madison, WI, USA) as substrate. S1 nuclease and major factors such as the hybridization temperature, concentration of capture probe, and the use of detergent were evaluated toward assay sensitivity, selectivity, and accuracy. RESULTS: The method is selective to the parent drugs with minimal cross-reactivity (<6%) with 3'-end deletion oligomers for both G3139 and GTI-2040. A linear range of 0.05 to 10 nM (r2 > 0.99) was observed for GTI-2040 in a variety of biological matrices. For both G3139 and GTI-2040, the within-day precision and accuracy values were found to be <20% and 90-110%, respectively; the between-day precision and accuracy were determined to be <20% and 90-120%. Addition of S1 nuclease combined with washing step greatly improved the assay linearity and selectivity. The utility of this assay was demonstrated by simultaneous determination of GTI-2040 in plasma and its intracellular levels in treated acute myeloid leukemia patients. CONCLUSIONS: The validated hybridization enzyme-linked immunosorbent assay method is specific for quantitation of PS ODNs in biological samples to picomolar level. This method provides a powerful technique to evaluate plasma pharmacokinetics and intracellular uptake of PS ODNs in patients and shows its utility in clinical evaluations.


Asunto(s)
Ensayo de Inmunoadsorción Enzimática/métodos , Oligonucleótidos Antisentido/análisis , Tionucleótidos/análisis , Enfermedad Aguda , Animales , Sondas de ADN , Eritrocitos/metabolismo , Humanos , Células K562 , Leucemia Mieloide/sangre , Leucemia Mieloide/metabolismo , Leucocitos Mononucleares/metabolismo , Ratones , Hibridación de Ácido Nucleico , Oligodesoxirribonucleótidos , Oligonucleótidos Antisentido/sangre , Oligonucleótidos Antisentido/farmacocinética , Ratas , Reproducibilidad de los Resultados , Endonucleasas Específicas del ADN y ARN con un Solo Filamento , Temperatura , Tionucleótidos/sangre , Tionucleótidos/orina
18.
Mol Pharmacol ; 68(2): 439-46, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15901851

RESUMEN

Engagement of integrin cell adhesion receptors in mouse lung endothelial cells induces global sensitivity of DNA to nuclease digestion, reflecting alterations in chromatin structure. These structural changes may contribute to the antigenotoxic effects of integrin engagement in lung endothelium. Because histone acetylation and poly(ADP-ribosyl)ation modulate chromatin structure, we investigated the effects of beta1 integrin engagement with antibody on these post-translational modifications and the presence of histones at discrete DNA sequences in the mouse lung endothelial cell genome using chromatin immunoprecipitation. Integrin engagement increased acetylation of core histone H3. The presence of acetylated histone H3 at intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1) promoters, and a nonpromoter sequence was also increased. As with integrin engagement, the histone deacetylase inhibitor trichostatin A caused global hypersensitivity of DNA to nuclease digestion and induced acetylation of histone H3 and its coimmunoprecipitation with VCAM-1 and ICAM-1 promoters and nonpromoter DNA. In contrast to acetyl-histone H3, the association of linker histone H1 with specific DNA sequences was either reduced or unaffected by integrin engagement and trichostatin A. Although integrin engagement and trichostatin A treatment did not affect histone H1 poly(ADP-ribosyl)ation, deletion of poly(ADP-ribose) polymerase-1 increased core histone H3 acetylation and increased its level at the iNOS promoter while decreasing the amount of histone H1. The results suggest that integrin engagement, as well as trichostatin A and PARP-1 deletion, regulate chromatin structure via core histone H3 acetylation and reduced linker histone H1-DNA association.


Asunto(s)
Células Endoteliales/metabolismo , Histonas/metabolismo , Integrinas/metabolismo , Pulmón/metabolismo , Acetilación , Animales , Células Cultivadas , Pulmón/citología , Ratones , Ratones Noqueados
19.
Mol Pharmacol ; 66(2): 302-11, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15266021

RESUMEN

Nitric oxide (NO) can be produced in large amounts by up-regulation of inducible NO synthase (iNOS). iNOS is induced in many cell types by pro-inflammatory agents, such as bacterial lipopolysaccharide (LPS) and cytokines. Overproduction by endothelial cells (EC) may contribute to vascular diseases. In contrast to macrophages, murine aortic endothelial cells (MAEC) produced no NO in response to either LPS or interferon gamma (IFNgamma), whereas combined treatment was highly synergistic. In this study, we investigated the mechanisms of synergy in MAEC. LPS activated p38 mitogen-activated protein kinase (MAPK), whereas IFNgamma activated Janus kinase and signal transducer and activator of transcription-1 (STAT1). Both pathways were required for iNOS induction because herbimycin A, a tyrosine kinase inhibitor, and 4-(4-fluorophenyl)-2-(4-hydroxyphenyl)-5-(4-pyridyl)1H-imidazole. HCl (SB202190), a p38 MAPKalpha/beta inhibitor, each blocked induction. LPS increased the phosphorylation of STAT1alpha at serine 727 in IFNgamma-treated MAEC. SB202190, but not 2'-amino-3'-methoxyflavone (PD98059), an inhibitor of p44/p42 MAPK activation, abolished the phosphorylation and induction of iNOS. SB202190 did not affect tyrosine 701 phosphorylation or nuclear translocation of STAT1. However, STAT1-DNA binding activity was reduced by SB202190. Although LPS stimulated the DNA binding activity of nuclear factor kappaB and activating protein-1, combined treatment with IFNgamma did not enhance activation, and SB202190 did not inhibit it. The results indicate that p38 MAPKalpha and/or beta are required for the synergistic induction of iNOS by LPS and IFNgamma in MAEC. Furthermore, the synergistic induction is associated with phosphorylation of STAT1alpha serine 727 in MAEC. This observation may explain potentially beneficial effects of p38 MAPK inhibitors in vascular inflammatory diseases.


Asunto(s)
Endotelio Vascular/efectos de los fármacos , Interferón gamma/farmacología , Lipopolisacáridos/farmacología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Óxido Nítrico Sintasa/metabolismo , Factor de Transcripción AP-1/metabolismo , Animales , Aorta/citología , Proteínas de Unión al ADN/metabolismo , Sinergismo Farmacológico , Endotelio Vascular/enzimología , Activación Enzimática , Imidazoles/farmacología , Ratones , FN-kappa B/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II , Fosforilación/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Piridinas/farmacología , Factor de Transcripción STAT1 , Serina/genética , Transactivadores/metabolismo , Transcripción Genética , Proteínas Quinasas p38 Activadas por Mitógenos
20.
Crit Care Med ; 30(5): 1071-82, 2002 May.
Artículo en Inglés | MEDLINE | ID: mdl-12006805

RESUMEN

OBJECTIVE: To synthesize novel inhibitors of the nuclear enzyme poly(adenosine 5'-diphosphate [ADP]-ribose) synthetase (PARS), also known as poly(ADP-ribose) polymerase (PARP), and to test them in in vitro models of oxidant-induced cytotoxicity and in endotoxin and splanchnic occlusion-reperfusion-induced shock. DESIGN: Randomized, prospective laboratory study. SETTING: Research laboratory. SUBJECTS: Murine macrophages, thymocytes, and endothelial cells; Balb/c mice and Wistar rats. INTERVENTIONS: Macrophages and endothelial cells were treated with peroxynitrite and bleomycin to induce PARS activation, and thymocytes were treated with peroxynitrite to induce cell necrosis. Novel PARS inhibitors were synthesized and used to reduce PARS activation and to reverse cytotoxicity. Balb/c mice were subjected to splanchnic occlusion and reperfusion and were pretreated with various doses (1-10 mg/kg intraperitoneally) of PJ34, a selected, potent, water-soluble PARS inhibitor. The passage of fluorescein isothiocyanate-conjugated dextran (4 kDa) was analyzed in everted gut ileal sacs incubated ex vivo as an index of gut permeability. Wistar rats were subjected to Escherichia coli bacterial lipopolysaccharide (40 mg/kg intraperitoneally). PJ34 was also used at 10 mg/kg intraperitoneally, 1 hr before lipopolysaccharide or at 25 mg/kg intraperitoneally 1 hr after lipopolysaccharide treatment. Serum concentrations of indicators or multiple organ injury, concentrations of various proinflammatory mediators, and tissue concentrations of myeloperoxidase and malondialdehyde were measured. In addition, survival rates and vascular contractile and relaxant responses were recorded. MEASUREMENTS AND MAIN RESULTS: Appropriate modifications of the phenanthridinone core structure yielded significant increases in the potency of the compounds, both as PARS inhibitors and as cytoprotective agents. The compound N-(6-oxo-5,6-dihydro-phenanthridin-2-yl) -N,N-dimethylacetamide (designated as PJ34) was one of the potent PARS inhibitors of the series, and it dose-dependently protected against thymocyte necrosis, with a half-maximal restoration of cell viability of 35 nM and complete protection at 200 nM. PARS activation also was visualized by immunohistochemistry and was dose-dependently suppressed by PJ34. The effect of PJ34 was dose-dependently reversed by excess nicotinamide adenine dinucleotide (oxidized). The PARS inhibitors dose-dependently suppressed proinflammatory cytokine and chemokine production and restored viability in immunostimulated macrophages. PJ34 was selected for the subsequent in vivo studies. PJ34 significantly protected against splanchnic reperfusion-induced intestinal hyperpermeability in the mouse. PJ34 reduced peak plasma concentrations of tumor necrosis factor-alpha, interleukin-1beta, and nitrite/nitrate in the plasma of lipopolysaccharide-treated rats. PJ34 ameliorated the lipopolysaccharide-induced increases in indexes of liver and kidney failure and concentrations of myeloperoxidase and malondialdehyde in the lung and gut. Lipopolysaccharide elicited vascular dysfunction, which was normalized by PJ34. Lipopolysaccharide-induced mortality was reduced by PJ34 (both pre- and posttreatment). CONCLUSIONS: The novel series of phenanthridinone PARS inhibitors have potent cytoprotective effects in vitro and significant protective effects in shock and reperfusion injury in rodent models in vivo.


Asunto(s)
Citoprotección , Inhibidores Enzimáticos/farmacología , Fenantrenos/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Choque/prevención & control , Animales , Bleomicina/farmacología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Activación Enzimática , Interleucina-1/sangre , Riñón/química , Lipopolisacáridos/farmacología , Pulmón/química , Malondialdehído/análisis , Ratones , Ratones Endogámicos BALB C , Nitratos/sangre , Nitritos/sangre , Peroxidasa/análisis , Ácido Peroxinitroso/farmacología , Ratas , Ratas Wistar , Factor de Necrosis Tumoral alfa/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA