Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
2.
Am J Respir Cell Mol Biol ; 48(6): 725-32, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23392573

RESUMEN

Chronic second-hand smoke (SHS) exposure comprises the main risk factor for nonsmokers to develop chronic obstructive pulmonary disease (COPD). However, the mechanisms behind the chronic inflammation and lung destruction remain incompletely understood. In this study, we show that chronic exposure of Sprague-Dawley rats to SHS results in a significant increase of proinflammatory cytokine IL-18 and chemokine (C-C motif) ligand 5 in the bronchoalveolar lavage fluid (BALF) and a significant decrease of vascular endothelial growth factor (VEGF) in the lung tissue. SHS exposure resulted in progressive alveolar airspace enlargement, cell death, pulmonary vessel loss, vessel muscularization, collagen deposition, and right ventricular hypertrophy. Alveolar macrophages displayed a foamy phenotype and a decreased expression of the natural inhibitor of IL-18, namely, IL-18 binding protein (IL-18BP). Moreover, IL-18 down-regulated the expression of VEGF receptor-1 and VEGFR receptor-2, and induced apoptosis in pulmonary microvascular endothelial cells in vitro. We also observed a trend toward increased concentrations of IL-18 in the BALF of patients with COPD. Our findings suggest that IL-18-mediated endothelial cell death may contribute to vascular destruction and disappearance in SHS-induced COPD. Moreover, IL-18 and IL-18BP are potential new targets for therapeutics.


Asunto(s)
Células Endoteliales/patología , Interleucina-18/inmunología , Enfisema Pulmonar/patología , Contaminación por Humo de Tabaco/efectos adversos , Animales , Líquido del Lavado Bronquioalveolar/inmunología , Permeabilidad Capilar , Cardiomegalia/inducido químicamente , Cardiomegalia/patología , Muerte Celular , Línea Celular , Quimiocina CCL5/inmunología , Células Endoteliales/inmunología , Células Endoteliales/metabolismo , Humanos , Inmunohistoquímica , Exposición por Inhalación/efectos adversos , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/inmunología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Interleucina-18/genética , Interleucina-18/metabolismo , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Enfermedad Pulmonar Obstructiva Crónica/inmunología , Enfermedad Pulmonar Obstructiva Crónica/patología , Enfisema Pulmonar/inmunología , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/patología , Ratas , Ratas Sprague-Dawley , Productos de Tabaco/efectos adversos , Factor A de Crecimiento Endotelial Vascular , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Pérdida de Peso
3.
Am J Physiol Lung Cell Mol Physiol ; 302(10): L1014-22, 2012 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-22387295

RESUMEN

In severe pulmonary arterial hypertension (PAH), vascular lesions are composed of phenotypically altered vascular and inflammatory cells that form clusters or tumorlets. Because macrophages are found in increased numbers in intravascular and perivascular space in human PAH, here we address the question whether macrophages play a role in pulmonary vascular remodeling and whether accumulation of macrophages in the lung vasculature could be compromised by the immune system. We used the mouse macrophage cell line RAW 264.7 because these cells are resistant to apoptosis, have high proliferative capacity, and resemble cells in the plexiform lesions that tend to pile up instead of maintaining a monolayer. Cells were characterized by immunocytochemistry with cell surface markers (Lycopersicon Esculentum Lectin, CD117, CD133, FVIII, CD31, VEGFR-2, and S100). Activated, but not quiescent, T cells were able to suppress RAW 264.7 cell proliferative and migration activity in vitro. The carboxyfluorescein diacetate-labeled RAW 264.7 cells were injected into the naïve Sprague Dawley (SD) rat and athymic nude rat. Twelve days later, cells were found in the lung vasculature of athymic nude rats that lack functional T cells, contributing to vascular remodeling. No labeled RAW 264.7 cells were detected in the lungs of immune-competent SD rats. Our data demonstrate that T cells can inhibit in vitro migration and in vivo accumulation of macrophage-like cells.


Asunto(s)
Comunicación Celular/inmunología , Pulmón/irrigación sanguínea , Macrófagos/citología , Arteria Pulmonar/citología , Linfocitos T/citología , Animales , Biomarcadores/análisis , Línea Celular , Movimiento Celular/inmunología , Proliferación Celular , Hipertensión Pulmonar Primaria Familiar , Fluoresceínas , Colorantes Fluorescentes , Hipertensión Pulmonar/inmunología , Hipertensión Pulmonar/patología , Hipertensión Pulmonar/fisiopatología , Inmunohistoquímica , Pulmón/citología , Pulmón/inmunología , Macrófagos/inmunología , Macrófagos/trasplante , Masculino , Ratones , Modelos Biológicos , Arteria Pulmonar/inmunología , Ratas , Ratas Desnudas , Ratas Sprague-Dawley , Linfocitos T/inmunología
4.
Am J Physiol Lung Cell Mol Physiol ; 302(10): L977-91, 2012 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-22307907

RESUMEN

Many chronic pulmonary diseases are associated with pulmonary hypertension (PH) and pulmonary vascular remodeling, which is a term that continues to be used to describe a wide spectrum of vascular abnormalities. Pulmonary vascular structural changes frequently increase pulmonary vascular resistance, causing PH and right heart failure. Although rat models had been standard models of PH research, in more recent years the availability of genetically engineered mice has made this species attractive for many investigators. Here we review a large amount of data derived from experimental PH reports published since 1996. These studies using wild-type and genetically designed mice illustrate the challenges and opportunities provided by these models. Hemodynamic measurements are difficult to obtain in mice, and right heart failure has not been investigated in mice. Anatomical, cellular, and genetic differences distinguish mice and rats, and pharmacogenomics may explain the degree of PH and the particular mode of pulmonary vascular adaptation and also the response of the right ventricle.


Asunto(s)
Modelos Animales de Enfermedad , Ventrículos Cardíacos/fisiopatología , Hipertensión Pulmonar/fisiopatología , Hipertrofia Ventricular Derecha/fisiopatología , Arteria Pulmonar/fisiopatología , Animales , Presión Sanguínea , Hipertensión Pulmonar Primaria Familiar , Ventrículos Cardíacos/patología , Hipertensión Pulmonar/patología , Hipertrofia Ventricular Derecha/patología , Ratones , Ratones Transgénicos , Arteria Pulmonar/patología , Ratas , Resistencia Vascular , Función Ventricular Derecha , Remodelación Ventricular
5.
Am J Physiol Lung Cell Mol Physiol ; 302(7): L711-8, 2012 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-22307908

RESUMEN

Efficient clearance of apoptotic cells from the lung by alveolar macrophages is important for the maintenance of tissue structure and function. Lung tissue from humans with emphysema contains increased numbers of apoptotic cells and decreased levels of vascular endothelial growth factor (VEGF). Mice treated with VEGF receptor inhibitors have increased numbers of apoptotic cells and develop emphysema. We hypothesized that VEGF regulates apoptotic cell clearance by alveolar macrophages (AM) via its interaction with VEGF receptor 1 (VEGF R1). Our data show that the uptake of apoptotic cells by murine AMs and human monocyte-derived macrophages is inhibited by depletion of VEGF and that VEGF activates Rac1. Antibody blockade or pharmacological inhibition of VEGF R1 activity also decreased apoptotic cell uptake ex vivo. Conversely, overexpression of VEGF significantly enhanced apoptotic cell uptake by AMs in vivo. These results indicate that VEGF serves a positive regulatory role via its interaction with VEGF R1 to activate Rac1 and enhance AM apoptotic cell clearance.


Asunto(s)
Apoptosis , Pulmón/metabolismo , Macrófagos Alveolares/fisiología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Animales , Anticuerpos Monoclonales , Línea Celular , Doxiciclina/administración & dosificación , Doxiciclina/farmacología , Enfisema/inmunología , Enfisema/metabolismo , Humanos , Indoles/administración & dosificación , Indoles/farmacología , Células Jurkat , Pulmón/citología , Macrófagos Alveolares/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Pirroles/administración & dosificación , Pirroles/farmacología , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptor 1 de Factores de Crecimiento Endotelial Vascular/inmunología
6.
J Clin Invest ; 118(2): 394-402, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18246188

RESUMEN

Emphysema is one manifestation of a group of chronic, obstructive, and frequently progressive destructive lung diseases. Cigarette smoking and air pollution are the main causes of emphysema in humans, and cigarette smoking causes emphysema in rodents. This review examines the concept of a homeostatically active lung structure maintenance program that, when attacked by proteases and oxidants, leads to the loss of alveolar septal cells and airspace enlargement. Inflammatory and noninflammatory mechanisms of disease pathogenesis, as well as the role of the innate and adaptive immune systems, are being explored in genetically altered animals and in exposure models of this disease. These recent scientific advances support a model whereby alveolar destruction resulting from a coalescence of mechanical forces, such as hyperinflation, and more recently recognized cellular and molecular events, including apoptosis, cellular senescence, and failed lung tissue repair, produces the clinically recognized syndrome of emphysema.


Asunto(s)
Enfisema Pulmonar/etiología , Enfisema Pulmonar/metabolismo , Animales , Modelos Animales de Enfermedad , Cobayas , Humanos , Inmunidad Innata , Ratones , Estrés Oxidativo , Péptido Hidrolasas/metabolismo , Enfisema Pulmonar/genética , Ratas , alfa 1-Antitripsina/metabolismo
7.
Respir Res ; 11: 179, 2010 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-21162738

RESUMEN

BACKGROUND: The presence of anti-endothelial cell antibodies and pathogenic T cells may reflect an autoimmune component in the pathogenesis of emphysema. Whether immune modulatory strategies can protect against the development of emphysema is not known. METHODS: Sprague Dawley rats were immunized with human umbilical vein endothelial cells (HUVEC) to induce autoimmune emphysema and treated with intrathymic HUVEC-injection and pristane. Measurements of alveolar airspace enlargement, cytokine levels, immuno histochemical, western blot analysis, and T cell repertoire of the lung tissue were performed. RESULTS: The immunomodulatory strategies protected lungs against cell death as demonstrated by reduced numbers of TUNEL and active caspase-3 positive cells and reduced levels of active caspase-3, when compared with lungs from HUVEC-immunized rats. Immunomodulatory strategies also suppressed anti-endothelial antibody production and preserved CNTF, IL-1alpha and VEGF levels. The immune deviation effects of the intrathymic HUVEC-injection were associated with an expansion of CD4+CD25+Foxp3+ regulatory T cells. Pristane treatment decreased the proportion of T cells expressing receptor beta-chain, Vß16.1 in the lung tissue. CONCLUSIONS: Our data demonstrate that interventions classically employed to induce central T cell tolerance (thymic inoculation of antigen) or to activate innate immune responses (pristane treatment) can prevent the development of autoimmune emphysema.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/prevención & control , Autoinmunidad/inmunología , Citocinas/inmunología , Enfisema/inmunología , Enfisema/prevención & control , Inmunomodulación/inmunología , Animales , Masculino , Ratas , Ratas Sprague-Dawley
8.
Am J Physiol Lung Cell Mol Physiol ; 297(5): L954-64, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19684203

RESUMEN

We recently reported that vasa vasorum expansion occurs in the pulmonary artery (PA) adventitia of chronically hypoxic animals and that extracellular ATP is a pro-angiogenic factor for isolated vasa vasorum endothelial cells (VVEC). However, the sources of extracellular ATP in the PA vascular wall, as well as the molecular mechanisms underlying its release, remain elusive. Studies were undertaken to explore whether VVEC release ATP in response to hypoxia and to determine signaling pathways involved in this process. We found that hypoxia (1-3% O2) resulted in time- and O2-dependent ATP release from VVEC. Preincubation with the inhibitors of vesicular transport (monensin, brefeldin A, and N-ethylmaleimide) significantly decreased ATP accumulation in the VVEC conditioned media, suggesting that hypoxia-induced ATP release occurs through vesicular exocytosis. Additionally, both hypoxia and exogenously added ATP resulted in the activation of PI3K and accumulation of GTP-bound RhoA in a time-dependent manner. Pharmacological inhibition of PI3K and ROCK or knockout of RhoA by small interfering RNA significantly abolished hypoxia-induced ATP release from VVEC. Moreover, RhoA and ROCK play a critical role in ATP-induced increases in VVEC DNA synthesis, migration, and tube formation, indicating a functional contribution of PI3K, Rho, and ROCK to both the autocrine mechanism of ATP release and ATP-mediated angiogenic activation of VVEC. Taken together, our findings provide novel evidence for the signaling mechanisms that link hypoxia-induced increases in extracellular ATP and vasa vasorum expansion.


Asunto(s)
Adenosina Trifosfato/farmacología , Células Endoteliales/enzimología , Neovascularización Fisiológica/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Vasa Vasorum/citología , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Animales , Bovinos , Hipoxia de la Célula/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Colágeno/metabolismo , ADN/biosíntesis , Combinación de Medicamentos , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Espacio Extracelular/efectos de los fármacos , Espacio Extracelular/metabolismo , Laminina/metabolismo , Masculino , Proteoglicanos/metabolismo , Arteria Pulmonar/citología , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Vesículas Transportadoras/efectos de los fármacos , Vesículas Transportadoras/metabolismo
9.
Circ Res ; 100(6): 923-9, 2007 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-17332430

RESUMEN

Vascular remodeling, rather than vasoconstriction, is believed to account for high vascular resistance in severe pulmonary arterial hypertension (PAH). We have found previously that acute Rho kinase inhibition nearly normalizes PAH in chronically hypoxic rats that have no occlusive neointimal lesions. Here we examined whether Rho kinase-mediated vasoconstriction was also important in a rat model of severe occlusive PAH. Adult rats were exposed to chronic hypoxia ( approximately 10% O(2)) after subcutaneous injection of the vascular endothelial growth factor receptor inhibitor SUGEN 5416. Hemodynamic measurements were made in anesthetized rats after 2 weeks of hypoxia (early group) and 3 weeks of hypoxia plus 2 weeks of normoxia (late group). Both groups developed PAH, with greater severity in the late group. In the early group, intravenous fasudil was more effective than intravenous bradykinin, inhaled NO, or intravenous iloprost in reducing right ventricular systolic pressure. Despite more occlusive vascular lesions, fasudil also markedly reduced right ventricular systolic pressure in late-stage rats. Blood-perfused lungs from late-stage rats showed spontaneous vasoconstriction, which was reversed partially by the endothelin A receptor blocker BQ123 and completely by fasudil or Y-27632. Phosphorylation of MYPT1, a downstream target of Rho kinase, was increased in lungs from both groups of rats, and fasudil (intravenous) reversed the increased phosphorylation in the late group. Thus, in addition to structural occlusion, Rho kinase-mediated vasoconstriction is an important component of severe PAH in SUGEN 5416/hypoxia-exposed rats, and PAH can be significantly reduced in the setting of a severely remodeled lung circulation if an unconventional vasodilator is used.


Asunto(s)
Hipertensión Pulmonar/fisiopatología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Arteria Pulmonar/fisiopatología , Vasoconstricción , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/análogos & derivados , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/farmacología , Animales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Antagonistas de los Receptores de la Endotelina A , Hipertensión Pulmonar/inducido químicamente , Hipertensión Pulmonar/enzimología , Hipoxia , Indoles , Masculino , Fosfatasa de Miosina de Cadena Ligera/metabolismo , Técnicas de Cultivo de Órganos , Péptidos Cíclicos/farmacología , Fosforilación/efectos de los fármacos , Arteria Pulmonar/enzimología , Pirroles , Ratas , Ratas Sprague-Dawley , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Vasoconstricción/efectos de los fármacos , Vasodilatadores/farmacología , Quinasas Asociadas a rho
10.
FASEB J ; 21(13): 3640-52, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17567571

RESUMEN

Severe pulmonary hypertension (PH) is characterized by complex precapillary arteriolar lesions, which contain phenotypically altered smooth muscle (SM) and endothelial cells (EC). We have demonstrated that VEGF receptor blockade by SU5416 {3-[(2,4-dimethylpyrrol-5-yl)methylidenyl]-indolin 2-one} in combination with chronic hypoxia causes severe angioproliferative PH associated with arterial occlusion in rats. We postulate that endothelial-mesenchymal transdifferentiation can take place in the occlusive lesions and that endothelium-derived mesenchymal cells can further differentiate toward a SM phenotype. To examine this hypothesis, we incubated human pulmonary microvascular endothelial cells (HPMVEC) with SU5416 and analyzed these cells utilizing quantitative-PCR, immunofluorescent staining and flow cytometry analysis. In vitro studies in HPMVEC demonstrated that SU5416 suppressed PGI2S gene expression while potently inducing COX-2, VEGF, and TGF-beta1 expression; and caused transdifferentiation of mature vascular endothelial cells (defined by Dil-ac-LDL, Lectin and Factor VIII) to SM-like (as defined by expression of alpha-SM actin) "transitional" cells, coexpressing both endothelial and SM markers. SU5416 expanded the number of CD34 and/or c-kit positive cells and caused transdifferentiation of CD34 positive cells but not negative cells. In conclusion, our data show that SU5416 generated a selection pressure that killed some EC and expanded progenitor-like cells to transdifferentiate to SM-like and neuronal-like cells.


Asunto(s)
Antígenos CD34/inmunología , Apoptosis , Diferenciación Celular , Endotelio Vascular/efectos de los fármacos , Músculo Liso/citología , Receptores de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Transdiferenciación Celular , Células Cultivadas , Técnica del Anticuerpo Fluorescente , Humanos
13.
FASEB J ; 19(9): 1178-80, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15897232

RESUMEN

We have demonstrated that VEGF receptor blockade in combination with chronic hypoxia causes in rats severe angioproliferative pulmonary hypertension (SAPH) associated with arterial occlusion by proliferating endothelial cells, and we postulate that the established, lumen-occluding lesions are the result of the emergence of apoptosis-resistant proliferating cells. To study the dependence of exuberant endothelial cell proliferation on initial apoptosis, we adapted the CELLMAX artificial capillary system to analyze the effects of a VEGF receptor antagonist (SU5416) on human pulmonary microvascular endothelial cells under pulsatile shear stress. Immunohistochemical staining for caspase-3 and PCNA and flow cytometry for Annexin-V and BrdU supported our concept, since SU5416 caused initial apoptosis (35.8% at 24 h after the SU5416 addition and 4.8% in control cells) whereas the surviving cells became hyperproliferative (PCNA positive). Flow cytometry showed that apoptosis inhibition prevented the proliferation following the initial apoptosis. These lumen-filling endothelial cells were apoptosis resistant, grew without serum, and were phenotypically altered in that they express the tumor marker survivin. Hyperproliferative apoptosis-resistant cells were also generated by adding apoptosed cells instead of the VEGF receptor blocker to the CELLMAX system. In conclusion, endothelial cell death resulted in the selection of an apoptosis-resistant, proliferating phenotypically altered endothelial cell phenotype.


Asunto(s)
Apoptosis , Células Endoteliales/patología , Hipertensión Pulmonar/patología , Apoptosis/efectos de los fármacos , Caspasa 3 , Inhibidores de Caspasas , Caspasas/análisis , Caveolina 1/análisis , Línea Celular , Proliferación Celular/efectos de los fármacos , Cicloheximida/farmacología , Humanos , Indoles/farmacología , Pirroles/farmacología , Receptores de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Resistencia al Corte , Factor de Necrosis Tumoral alfa/farmacología , Proteína bcl-X/análisis
14.
Peptides ; 26(8): 1292-300, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15878794

RESUMEN

Bradykinin is an important modulator of endothelial cell function and has also a powerful cardioprotective effect. Here we report that treatment of severely pulmonary hypertensive rats (that recapitulate several of the physiological and pathological characteristics of the human pulmonary vascular disease, including dramatic right ventricular hypertrophy, pericardial effusion and death) with a newly synthesized long-acting bradykinin B2 receptor agonist B9972 caused reduction of the pulmonary artery pressure (PAP=51+/-2.0 versus 68+/-2.8 of untreated animals) and of right ventricular hypertrophy (Rv/Lv+S=0.55+/-0.02 versus 0.73+/-0.03 of untreated rats) and activation of Akt. Long-term stimulation with B9972 in our animal model of SPH resulted in decreased expression of the B2 receptor in lung vasculature. Treatment with B9972 decreased the number of plexiform lesions in the lungs by inducing cell apoptosis in the obliterated vessels and by restoring caveolin-1 expression. B9972 also promoted eNOS activation. In vitro B9972 caused activation of caspase-3 as well as Erk and induction of prostacyclin production in rat pulmonary microvascular EC. Taken together our data suggest that a stable bradykinin B2 agonist B9972 demonstrates the capacity to reduce severe pulmonary hypertension, right ventricular hypertrophy and induce apoptosis of hyperproliferative cells in pre-capillary pulmonary arterioles.


Asunto(s)
Hipertensión Pulmonar/tratamiento farmacológico , Hipertrofia Ventricular Derecha/tratamiento farmacológico , Oligopéptidos/farmacología , Arteria Pulmonar/efectos de los fármacos , Receptor de Bradiquinina B2/agonistas , Animales , Apoptosis/efectos de los fármacos , Determinación de la Presión Sanguínea/métodos , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Células Endoteliales/enzimología , Hipertensión Pulmonar/fisiopatología , Hipertrofia Ventricular Derecha/fisiopatología , Masculino , Oligopéptidos/administración & dosificación , Oligopéptidos/síntesis química , Arteria Pulmonar/fisiopatología , Ratas , Ratas Sprague-Dawley , Receptor de Bradiquinina B2/biosíntesis , Receptor de Bradiquinina B2/metabolismo , Índice de Severidad de la Enfermedad , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
15.
Peptides ; 26(8): 1288-91, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15878795

RESUMEN

Lung and prostate cancers are major health problems worldwide. Treatments with standard chemotherapy agents are relatively ineffective. Combination chemotherapy gives better treatment than a single agent because the drugs can inhibit the cancer in different pathways, but new therapeutic agents are needed for the treatment of both tumor types. Bradykinin (BK) antagonists offer advantages of combination therapy in one compound. These promising multitargeted anti-cancer compounds selectively stimulate apoptosis in cancers and also inhibit both angiogenesis and matrix metalloprotease (MMP) action in treated lung and prostate tumors in nude mice. The highly potent, metabolism-resistant bradykinin antagonist peptide dimer, B-9870 [SUIM-(DArg-Arg-Pro-Hyp-Gly-Igl-Ser-DIgl-Oic-Arg)2] (SUIM=suberimidyl; Hyp=4-hydroxyproline; Igl=alpha-(2-indanyl)glycine; Oic=octahydroindole-2-carboxylic acid) and its non-peptide mimetic, BKM-570 [2,3,4,5,6-pentafluorocinnamoyl-(o-2,6-dichlorobenzyl)-L-tyrosine-N-(4-amino-2,2,6,6-tetramethylpiperidyl)amide] are superior to the widely used but toxic chemotherapeutic drugs cisplatin and taxotere. In certain combinations, they act synergistically with standard anti-cancer drugs. Due to its structure and biological activity, BKM-570 is an attractive lead compound for derivatization and evaluation for lung and prostate cancer drugs.


Asunto(s)
Antineoplásicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Bradiquinina/antagonistas & inhibidores , Oligopéptidos/farmacología , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Cobayas , Humanos , Íleon/efectos de los fármacos , Inhibidores de la Metaloproteinasa de la Matriz , Ratones , Ratones Desnudos , Oligopéptidos/administración & dosificación , Oligopéptidos/química , Fenilalanina/análogos & derivados , Fenilalanina/farmacología , Conejos , Ratas , Relación Estructura-Actividad , Tirosina/análogos & derivados , Tirosina/farmacología
16.
Int Immunopharmacol ; 2(13-14): 1781-6, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12489792

RESUMEN

Bradykinin (BK) is an autocrine growth factor for lung and prostate cancers. BK also facilitates tumor extension by increasing tissue permeability and stimulating angiogenesis. Peptide BK antagonists are in development as potential new drugs for lung cancer. Newer nonpeptide BK antagonists have even higher potency against lung cancer, in vitro and in vivo. These compounds have now been applied to the study of prostate cancers, and have been found to be effective. Prostate cancer cell line PC3 is derived from a late-stage, hormone-independent, metastatic tumor; its growth is difficult to inhibit. Our established BK antagonists, while less effective against this line of prostate cancer in xenografts in nude mice than against lung cancer, are active and have led the way to development of new peptide and nonpeptide agents for prostate cancer. In addition to inhibiting cancer cell growth directly, they inhibit angiogenesis mediated by vascular endothelial growth factor, and inhibit increased tissue permeability mediated by membrane metalloproteases in these tumors. This class of compounds offers hope for development of new drugs for refractory prostate cancer.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Antineoplásicos/uso terapéutico , Antagonistas de los Receptores de Bradiquinina , Neoplasias de la Próstata/tratamiento farmacológico , Inhibidores de Proteasas/uso terapéutico , Inhibidores de la Angiogénesis/química , Animales , Antineoplásicos/química , Membrana Celular/enzimología , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Humanos , Concentración 50 Inhibidora , Masculino , Metaloendopeptidasas/antagonistas & inhibidores , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Inhibidores de Proteasas/química , Receptores de Bradiquinina/biosíntesis , Relación Estructura-Actividad , Células Tumorales Cultivadas , Cordón Umbilical/citología , Cordón Umbilical/efectos de los fármacos , Cordón Umbilical/metabolismo
17.
Front Physiol ; 4: 91, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23720629

RESUMEN

Chronic Obstructive Pulmonary Disease (COPD) is one of the foremost causes of death worldwide. It is primarily caused by tobacco smoke, making it an easily preventable disease, but facilitated by genetic α-1 antitrypsin deficiency. In addition to active smokers, health problems also occur in people involuntarily exposed to second hand smoke (SHS). Currently, the relationship between SHS and COPD is not well established. Knowledge of pathogenic mechanisms is limited, thereby halting the advancement of new treatments for this socially and economically detrimental disease. Here, we attempt to summarize tobacco smoke studies undertaken in animal models, applying both mainstream (direct, nose only) and side stream (indirect, whole body) smoke exposures. This overview of 155 studies compares cellular and molecular mechanisms as well as proteolytic, inflammatory, and vasoreactive responses underlying COPD development. This is a difficult task, as listing of exposure parameters is limited for most experiments. We show that both mainstream and SHS studies largely present similar inflammatory cell populations dominated by macrophages as well as elevated chemokine/cytokine levels, such as TNF-α. Additionally, SHS, like mainstream smoke, has been shown to cause vascular remodeling and neutrophil elastase-mediated proteolytic matrix breakdown with failure to repair. Disease mechanisms and therapeutic interventions appear to coincide in both exposure scenarios. One of the more widely applied interventions, the anti-oxidant therapy, is successful for both mainstream and SHS. The comparison of direct with indirect smoke exposure studies in this review emphasizes that, even though there are many overlapping pathways, it is not conclusive that SHS is using exactly the same mechanisms as direct smoke in COPD pathogenesis, but should be considered a preventable health risk. Some characteristics and therapeutic alternatives uniquely exist in SHS-related COPD.

18.
PLoS One ; 7(5): e38038, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22675432

RESUMEN

BACKGROUND: Given the relative abundance and toxic potential of acrolein in inhaled cigarette smoke, it is surprising how little is known about the pulmonary and systemic effects of acrolein. Here we test the hypothesis whether systemic administration of acrolein could cause endoplasmic reticulum (ER) stress, and lung cell apoptosis, leading to the enlargement of the alveolar air spaces in rats. METHODS: Acute and chronic effects of intraperitoneally administered acrolein were tested. Mean alveolar airspace area was measured by using light microscopy and imaging system software. TUNEL staining and immunohistochemistry (IHC) for active caspase 3 and Western blot analysis for active caspase 3, and caspase 12 were performed to detect apoptosis. The ER-stress related gene expression in the lungs was determined by Quantitative real-time PCR analysis. Acrolein-protein adducts in the lung tissue were detected by IHC. RESULTS: Acute administration of acrolein caused a significant elevation of activated caspase 3, upregulation of VEGF expression and induced ER stress proteins in the lung tissue. The chronic administration of acrolein in rats led to emphysematous lung tissue remodeling. TUNEL staining and IHC for cleaved caspase 3 showed a large number of apoptotic septal cells in the acrolein-treated rat lungs. Chronic acrolein administration cause the endoplasmic reticulum stress response manifested by significant upregulation of ATF4, CHOP and GADd34 expression. In smokers with COPD there was a considerable accumulation of acrolein-protein adducts in the inflammatory, airway and vascular cells. CONCLUSIONS: Systemic administration of acrolein causes endoplasmic reticulum stress response, lung cell apoptosis, and chronic administration leads to the enlargement of the alveolar air spaces and emphysema in rats. The substantial accumulation of acrolein-protein adducts in the lungs of COPD patients suggest a role of acrolein in the pathogenesis of emphysema.


Asunto(s)
Acroleína/toxicidad , Estrés del Retículo Endoplásmico/efectos de los fármacos , Enfisema Pulmonar/inducido químicamente , Acroleína/administración & dosificación , Acroleína/metabolismo , Animales , Caspasa 3/metabolismo , Expresión Génica/efectos de los fármacos , Pulmón/efectos de los fármacos , Pulmón/patología , Masculino , Unión Proteica , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/patología , Enfisema Pulmonar/metabolismo , Enfisema Pulmonar/patología , Ratas , Ratas Sprague-Dawley , Respuesta de Proteína Desplegada/efectos de los fármacos , Respuesta de Proteína Desplegada/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA