Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Front Oncol ; 5: 164, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26258071

RESUMO

BACKGROUND: Lung cancer is a devastating disease with limited treatment options. Many lung cancers have changes in their microenvironment including upregulation of the extracellular matrix glycosaminoglycan, hyaluronan (HA), which we have previously demonstrated can regulate the activity of the extracellular serine protease, hyaluronan binding protein 2 (HABP2). This study examined the functional role of HABP2 on HA-mediated human lung cancer dynamics. METHODS: Immunohistochemical analysis was performed on lung cancer patient samples using anti-HABP2 antibody. Stable control, shRNA, and HABP2 overexpressing human lung adenocarcinoma cells were evaluated using immunoblot analysis, migration, extravasation, and urokinase plasminogen activator (uPA) activation assays with or without high-molecular weight HA or low-molecular weight HA (LMW-HA). In human lung cancer xenograft models, primary tumor growth rates and lung metastasis were analyzed using consecutive tumor volume measurements and nestin immunoreactivity in nude mouse lungs. RESULTS: We provide evidence that HABP2 is an important regulator of lung cancer progression. HABP2 expression was increased in several subtypes of patient non-small cell lung cancer samples. Further, HABP2 overexpression increased LMW-HA-induced uPA activation, migration, and extravasation in human lung adenocarcinoma cells. In vivo, overexpression of HABP2 in human lung adenocarcinoma cells increased primary tumor growth rates in nude mice by ~2-fold and lung metastasis by ~10-fold compared to vector control cells (n = 5/condition). CONCLUSION: Our data suggest a possible direct effect of HABP2 on uPA activation and lung cancer progression. Our observations suggest that exploration of HABP2 in non-small cell lung carcinoma merits further study both as a diagnostic and therapeutic option.

2.
Cancer ; 121(16): 2681-8, 2015 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-26043235

RESUMO

Mu opioids are among the most widely used drugs for patients with cancer with both acute and chronic pain as well as in the perioperative period. Several retrospective studies have suggested that opioid use might promote tumor progression and as a result negatively impact survival in patients with advanced cancer; however, in the absence of appropriate prospective validation, any changes in recommendations for opioid use are not warranted. In this review, the authors present preclinical and clinical data that support their hypothesis that the mu opioid receptor is a potential target for cancer therapy because of its plausible role in tumor progression. The authors also propose the hypothesis that peripheral opioid antagonists such as methylnaltrexone, which reverses the peripheral effects of mu opioids but maintains centrally mediated analgesia and is approved by the US Food and Drug Administration for the treatment of opioid-induced constipation, can be used to target the mu opioid receptor.


Assuntos
Antagonistas de Entorpecentes/uso terapêutico , Neoplasias/tratamento farmacológico , Receptores Opioides mu/fisiologia , Animais , Humanos , Camundongos , Terapia de Alvo Molecular , Naltrexona/análogos & derivados , Naltrexona/uso terapêutico , Compostos de Amônio Quaternário/uso terapêutico , Receptores Opioides mu/genética
3.
Adv Cancer Res ; 123: 191-209, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25081530

RESUMO

Vascular integrity or the maintenance of blood vessel continuity is a fundamental process regulated by endothelial cell-cell junctions. Defects in endothelial barrier function are an initiating factor in several disease processes including tumor angiogenesis and metastasis. The glycosaminoglycan, hyaluronan (HA), maintains vascular integrity through specific mechanisms including HA-binding protein signaling in caveolin-enriched microdomains, a subset of lipid rafts. Certain disease states, including cancer, increase enzymatic hyaluronidase activity and reactive oxygen species generation, which break down high molecular weight HA (HMW-HA) to low molecular weight fragments (LMW-HA). LMW-HA can activate specific HA-binding proteins during tumor progression to promote disruption of endothelial cell-cell contacts. In contrast, exogenous administration of HMW-HA promotes enhancement of vascular integrity. This review focuses on the roles of HA in regulating angiogenic and metastatic processes based on its size and the HA-binding proteins present. Further, potential therapeutic applications of HMW-HA in treating cancer are discussed.


Assuntos
Endotélio/metabolismo , Regulação Neoplásica da Expressão Gênica , Ácido Hialurônico/química , Neoplasias/metabolismo , Animais , Antineoplásicos/química , Caveolinas/química , Progressão da Doença , Glicosaminoglicanos/química , Humanos , Receptores de Hialuronatos , Microdomínios da Membrana/química , Peso Molecular , Metástase Neoplásica , Neovascularização Patológica , Estrutura Terciária de Proteína , Espécies Reativas de Oxigênio , Transdução de Sinais/fisiologia
4.
J Biol Chem ; 289(35): 24043-58, 2014 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-25023279

RESUMO

Angiogenesis or the formation of new blood vessels is important in the growth and metastatic potential of various cancers. Therefore, understanding the mechanism(s) by which angiogenesis occurs can have important therapeutic implications in numerous malignancies. We and others have demonstrated that low molecular weight hyaluronan (LMW-HA, ∼2500 Da) promotes endothelial cell (EC) barrier disruption and angiogenesis. However, the mechanism(s) by which this occurs is poorly defined. Our data indicate that treatment of human EC with LMW-HA induced CD44v10 association with the receptor-tyrosine kinase, EphA2, transactivation (tyrosine phosphorylation) of EphA2, and recruitment of the PDZ domain scaffolding protein, PATJ, to the cell periphery. Silencing (siRNA) CD44, EphA2, PATJ, or Dbs (RhoGEF) expression blocked LMW-HA-mediated angiogenesis (EC proliferation, migration, and tubule formation). In addition, silencing EphA2, PATJ, Src, or Dbs expression blocked LMW-HA-mediated RhoA activation. To translate our in vitro findings, we utilized a novel anginex/liposomal targeting of murine angiogenic endothelium with either CD44 or EphA2 siRNA and observed inhibition of LMW-HA-induced angiogenesis in implanted Matrigel plugs. Taken together, these results indicate LMW-HA-mediated transactivation of EphA2 is required for PATJ and Dbs membrane recruitment and subsequent RhoA activation required for angiogenesis. These results suggest that targeting downstream effectors of LMW-HA could be a useful therapeutic intervention for angiogenesis-associated diseases including tumor progression.


Assuntos
Efrina-A2/genética , Ácido Hialurônico/fisiologia , Neoplasias/patologia , Neovascularização Patológica/fisiopatologia , Receptores Proteína Tirosina Quinases/genética , Ativação Transcricional , Animais , Progressão da Doença , Efrina-A2/fisiologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Ácido Hialurônico/química , Camundongos , Camundongos Endogâmicos C57BL , Peso Molecular , Receptores Proteína Tirosina Quinases/fisiologia
5.
Mol Biol Cell ; 25(13): 2006-16, 2014 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-24829380

RESUMO

Vascular integrity and the maintenance of blood vessel continuity are fundamental features of the circulatory system maintained through endothelial cell-cell junctions. Defects in the endothelial barrier become an initiating factor in several pathologies, including ischemia/reperfusion, tumor angiogenesis, pulmonary edema, sepsis, and acute lung injury. Better understanding of mechanisms stimulating endothelial barrier enhancement may provide novel therapeutic strategies. We previously reported that oxidized phospholipids (oxidized 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphocholine [OxPAPC]) promote endothelial cell (EC) barrier enhancement both in vitro and in vivo. This study examines the initiating mechanistic events triggered by OxPAPC to increase vascular integrity. Our data demonstrate that OxPAPC directly binds the cell membrane-localized chaperone protein, GRP78, associated with its cofactor, HTJ-1. OxPAPC binding to plasma membrane-localized GRP78 leads to GRP78 trafficking to caveolin-enriched microdomains (CEMs) on the cell surface and consequent activation of sphingosine 1-phosphate receptor 1, Src and Fyn tyrosine kinases, and Rac1 GTPase, processes essential for cytoskeletal reorganization and EC barrier enhancement. Using animal models of acute lung injury with vascular hyperpermeability, we observed that HTJ-1 knockdown blocked OxPAPC protection from interleukin-6 and ventilator-induced lung injury. Our data indicate for the first time an essential role of GRP78 and HTJ-1 in OxPAPC-mediated CEM dynamics and enhancement of vascular integrity.


Assuntos
Células Endoteliais/metabolismo , Proteínas de Choque Térmico/fisiologia , Fosfatidilcolinas/fisiologia , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Animais , Caveolinas/metabolismo , Células Cultivadas , Impedância Elétrica , Chaperona BiP do Retículo Endoplasmático , Endotélio Vascular/citologia , Endotélio Vascular/fisiologia , Proteínas de Choque Térmico HSP40/metabolismo , Humanos , Masculino , Microdomínios da Membrana/metabolismo , Proteínas de Membrana/metabolismo , Camundongos Endogâmicos C57BL , Oxirredução , Transporte Proteico , Artéria Pulmonar/citologia , Receptores de Lisoesfingolipídeo/metabolismo
6.
PLoS One ; 9(3): e91577, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24662916

RESUMO

Recent epidemiologic studies implying differences in cancer recurrence based on anesthetic regimens raise the possibility that the mu opioid receptor (MOR) can influence cancer progression. Based on our previous observations that overexpression of MOR in human non-small cell lung cancer (NSCLC) cells increased tumor growth and metastasis, this study examined whether MOR regulates growth factor receptor signaling and epithelial mesenchymal transition (EMT) in human NSCLC cells. We utilized specific siRNA, shRNA, chemical inhibitors and overexpression vectors in human H358 NSCLC cells that were either untreated or treated with various concentrations of DAMGO, morphine, fentanyl, EGF or IGF. Cell function assays, immunoblot and immunoprecipitation assays were then performed. Our results indicate MOR regulates opioid and growth factor-induced EGF receptor signaling (Src, Gab-1, PI3K, Akt and STAT3 activation) which is crucial for consequent human NSCLC cell proliferation and migration. In addition, human NSCLC cells treated with opioids, growth factors or MOR overexpression exhibited an increase in snail, slug and vimentin and decrease ZO-1 and claudin-1 protein levels, results consistent with an EMT phenotype. Further, these effects were reversed with silencing (shRNA) or chemical inhibition of MOR, Src, Gab-1, PI3K, Akt and STAT3 (p<0.05). Our data suggest a possible direct effect of MOR on opioid and growth factor-signaling and consequent proliferation, migration and EMT transition during lung cancer progression. Such an effect provides a plausible explanation for the epidemiologic findings.


Assuntos
Analgésicos Opioides/farmacologia , Carcinoma Pulmonar de Células não Pequenas/patologia , Movimento Celular/efeitos dos fármacos , Fator de Crescimento Epidérmico/farmacologia , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Neoplasias Pulmonares/patologia , Receptores Opioides mu/metabolismo , Anestésicos/efeitos adversos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Progressão da Doença , Receptores ErbB/metabolismo , Inativação Gênica , Humanos , Receptores Opioides mu/deficiência , Receptores Opioides mu/genética , Transdução de Sinais/efeitos dos fármacos
7.
Cancer Biol Ther ; 14(7): 679-91, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23792636

RESUMO

Cytoskeletal and focal adhesion abnormalities are observed in several types of cancer, including lung cancer. We have previously reported that paxillin (PXN) was mutated, amplified, and overexpressed in a significant number of lung cancer patient samples, that PXN protein was upregulated in more advanced stages of lung cancer compared with lower stages, and that the PXN gene was also amplified in some pre-neoplastic lung lesions. Among the mutations investigated, we previously found that PXN variant A127T in lung cancer cells enhanced cell proliferation and focal adhesion formation and colocalized with the anti-apoptotic protein B Cell Lymphoma 2 (BCL-2), which is known to localize to the mitochondria, among other sites. To further explore the effects of activating mutations of PXN on mitochondrial function, we cloned and expressed wild-type PXN and variants containing the most commonly occurring PXN mutations (P46S, P52L, G105D, A127T, P233L, T255I, D399N, E423K, P487L, and K506R) in a GFP-tagged vector using HEK-293 human embryonic kidney cells. Utilizing live-cell imaging to systematically study the effects of wild-type PXN vs. mutants, we created a model that recapitulates the salient features of the measured dynamics and conclude that compared with wild-type, some mutant clones confer enhanced focal adhesion and lamellipodia formation (A127T, P233L, and P487L) and some confer increased association with BCL-2, Dynamin-related Protein-1 (DRP-1), and Mitofusion-2 (MFN-2) proteins (P233L and D399N). Further, PXN mutants, through their interactions with BCL-2 and DRP-1, could regulate cisplatin drug resistance in human lung cancer cells. The data reported herein suggest that mutant PXN variants play a prominent role in mitochondrial dynamics with direct implications on lung cancer progression and hence, deserve further exploration as therapeutic targets.


Assuntos
Adesões Focais/genética , Neoplasias Pulmonares/genética , Dinâmica Mitocondrial/genética , Paxilina/genética , Adesões Focais/metabolismo , Células HEK293 , Humanos , Neoplasias Pulmonares/metabolismo , Mutação , Paxilina/metabolismo
8.
J Biol Chem ; 288(4): 2191-200, 2013 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-23212923

RESUMO

Vascular endothelial cell (EC) barrier integrity is critical to vessel homeostasis whereas barrier dysfunction is a key feature of inflammatory disorders and tumor angiogenesis. We previously reported that hepatocyte growth factor (HGF)-mediated increases in EC barrier integrity are signaled through a dynamic complex present in lipid rafts involving its receptor, c-Met. We extended these observations to confirm that S1PR1 (sphingosine 1-phosphate receptor 1) and integrin ß4 (ITGB4) are essential participants in HGF-induced EC barrier enhancement. Immunoprecipitation experiments demonstrated HGF-mediated recruitment of c-Met, ITGB4 and S1PR1 to caveolin-enriched lipid rafts in human lung EC with direct interactions of c-Met with both S1PR1 and ITGB4 accompanied by c-Met-dependent S1PR1 and ITGB4 transactivation. Reduced S1PR1 expression (siRNA) attenuated both ITGB4 and Rac1 activation as well as c-Met/ITGB4 interaction and resulted in decreased transendothelial electrical resistance. Furthermore, reduced ITGB4 expression attenuated HGF-induced c-Met activation, c-Met/S1PR1 interaction, and effected decreases in S1P- and HGF-induced EC barrier enhancement. Finally, the c-Met inhibitor, XL880, suppressed HGF-induced c-Met activation as well as S1PR1 and ITGB4 transactivation. These results support a critical role for S1PR1 and ITGB4 transactivation as rate-limiting events in the transduction of HGF signals via a dynamic c-Met complex resulting in enhanced EC barrier integrity.


Assuntos
Células Endoteliais/citologia , Fator de Crescimento de Hepatócito/metabolismo , Integrina beta4/metabolismo , Proteínas Proto-Oncogênicas c-met/metabolismo , Receptores de Lisoesfingolipídeo/metabolismo , Membrana Celular/metabolismo , Eletrofisiologia , Humanos , Pulmão/metabolismo , Microdomínios da Membrana/química , Microdomínios da Membrana/metabolismo , Microcirculação , Modelos Biológicos , RNA Interferente Pequeno/metabolismo , Receptores de Esfingosina-1-Fosfato , Treonina/química , Ativação Transcricional , Tirosina/química , Proteínas rac1 de Ligação ao GTP/metabolismo
10.
Anesthesiology ; 116(4): 857-67, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22343475

RESUMO

BACKGROUND: Recent epidemiologic studies suggesting that there were differences in cancer recurrence contingent on anesthetic regimens have raised the possibility that µ-opioid agonists can influence cancer progression. Based on our previous studies indicating the µ-opioid receptor (MOR) is up-regulated in several types of non-small cell lung cancer, this study examined the functional significance of MOR overexpression to elucidate a possible mechanism for the epidemiologic findings. METHODS: Stable vector control and MOR1 overexpressing human bronchioloalveolar carcinoma cells were evaluated using immunoblot analysis, proliferation and transendothelial extravasation assays with or without Akt inhibitor, mTOR inhibitor (temsirolimus), or the peripheral MOR antagonist, methylnaltrexone. In human lung cancer xenograft models, primary tumor growth rates and lung metastasis were analyzed using consecutive tumor volume measurements and nestin immunoreactivity in lungs of the nude mouse model. RESULTS: The authors provide evidence that MOR is an important regulator of lung cancer progression. MOR overexpression increased Akt and mTOR activation, proliferation, and extravasation in human bronchioloalveolar carcinoma cells. In vivo, overexpression of MOR in human bronchoalveolar carcinoma cells increased primary tumor growth rates in nude mice by approximately 2.5-fold and lung metastasis by approximately 20-fold compared with vector control cells (n = 4 per condition). CONCLUSIONS: The overexpression data suggest a possible direct effect of MOR on Akt and mTOR activation and lung cancer progression. Such an effect provides a plausible explanation for the epidemiologic findings. The authors' observations further suggest that exploration of MOR in non-small cell lung carcinoma merits further study both as a diagnostic and therapeutic option.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/secundário , Neoplasias Pulmonares/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores Opioides mu/biossíntese , Serina-Treonina Quinases TOR/metabolismo , Animais , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Linhagem Celular Tumoral , Progressão da Doença , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Masculino , Camundongos , Camundongos Nus , Proteínas Proto-Oncogênicas c-akt/fisiologia , Receptores Opioides mu/genética , Receptores Opioides mu/fisiologia , Serina-Treonina Quinases TOR/fisiologia , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
11.
Am J Physiol Lung Cell Mol Physiol ; 301(2): L137-47, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21571904

RESUMO

Hyaluronan (HA) has diverse functions in normal lung homeostasis and pulmonary disease. HA constitutes the major glycosaminoglycan in lung tissue, with HA degradation products, produced by hyaluronidase enzymes and reactive oxygen species, being implicated in several lung diseases, including acute lung injury, asthma, chronic obstructive pulmonary disease, and pulmonary hypertension. The differential activities of HA and its degradation products are due, in part, to regulation of multiple HA-binding proteins, including cluster of differentiation 44 (CD44), Toll-like receptor 4 (TLR4), HA-binding protein 2 (HABP2), and receptor for HA-mediated motility (RHAMM). Recent research indicates that exogenous administration of high-molecular-weight HA can serve as a novel therapeutic intervention for lung diseases, including lipopolysaccharide (LPS)-induced acute lung injury, sepsis/ventilator-induced lung injury, and airway hyperreactivity. This review focuses on the regulatory role of HA and HA-binding proteins in lung pathology and discusses the capacity of HA to augment and inhibit various lung diseases.


Assuntos
Receptores de Hialuronatos/metabolismo , Ácido Hialurônico/metabolismo , Pneumopatias/metabolismo , Pulmão/metabolismo , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/prevenção & controle , Animais , Hiper-Reatividade Brônquica/tratamento farmacológico , Hiper-Reatividade Brônquica/prevenção & controle , Bronquite/prevenção & controle , Doença Crônica , Proteínas da Matriz Extracelular/metabolismo , Glucuronosiltransferase/metabolismo , Humanos , Hialuronan Sintases , Ácido Hialurônico/química , Ácido Hialurônico/uso terapêutico , Lipopolissacarídeos , Pneumopatias/induzido quimicamente , Pneumopatias/tratamento farmacológico , Pneumopatias/prevenção & controle , Peso Molecular , Isoformas de Proteínas/metabolismo , Enfisema Pulmonar/prevenção & controle , Espécies Reativas de Oxigênio/metabolismo , Receptor 4 Toll-Like/metabolismo , Lesão Pulmonar Induzida por Ventilação Mecânica/prevenção & controle
12.
Anesth Analg ; 112(3): 558-67, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21156980

RESUMO

BACKGROUND: The possibility that µ opioid agonists can influence cancer recurrence is a subject of recent interest. Epidemiologic studies suggested that there were differences in cancer recurrence in breast and prostate cancer contingent on anesthetic regimens. In this study, we identify a possible mechanism for these epidemiologic findings on the basis of µ opioid receptor (MOR) regulation of Lewis lung carcinoma (LLC) tumorigenicity in cell and animal models. METHODS: We used human lung tissue and human non-small cell lung cancer (NSCLC) cell lines and evaluated MOR expression using immunoblot and immunohistochemical analysis. LLC cells were treated with the peripheral opioid antagonist methylnaltrexone (MNTX) or MOR shRNA and evaluated for proliferation, invasion, and soft agar colony formation in vitro and primary tumor growth and lung metastasis in C57BL/6 and MOR knockout mice using VisEn fluorescence mediated tomography imaging and immunohistochemical analysis. RESULTS: We provide several lines of evidence that the MOR may be a potential target for lung cancer, a disease with high mortality and few treatment options. We first observed that there is ∼5- to 10-fold increase in MOR expression in lung samples from patients with NSCLC and in several human NSCLC cell lines. The MOR agonists morphine and [D-Ala(2), N-MePhe(4), Gly-ol]-enkephalin (DAMGO) increased in vitro LLC cell growth. Treatment with MNTX or silencing MOR expression inhibited LLC invasion and anchorage-independent growth by 50%-80%. Injection of MOR silenced LLC lead to a ∼65% reduction in mouse lung metastasis. In addition, MOR knockout mice do not develop significant tumors when injected with LLC in comparison with wild-type controls. Finally, continuous infusion of the peripheral opioid antagonist MNTX attenuates primary LLC tumor growth and reduces lung metastasis. CONCLUSIONS: Taken together, our data suggest a possible direct effect of opiates on lung cancer progression, and provide a plausible explanation for the epidemiologic findings. Our observations further suggest a possible therapeutic role for opioid antagonists.


Assuntos
Carcinoma Pulmonar de Lewis/metabolismo , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Progressão da Doença , Neoplasias Pulmonares/metabolismo , Receptores Opioides mu/fisiologia , Animais , Carcinoma Pulmonar de Lewis/patologia , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular , Linhagem Celular Tumoral , Feminino , Humanos , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Invasividade Neoplásica/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Am J Cardiovasc Dis ; 1(3): 200-13, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22254199

RESUMO

Vascular integrity or the maintenance of blood vessel continuity is a fundamental process regulated, in part, by the endothelial glycocalyx and cell-cell junctions. Defects in endothelial barrier function are an initiating factor in several disease processes including atherosclerosis, ischemia/reperfusion, tumor angiogenesis, cancer metastasis, diabetes, sepsis and acute lung injury. The glycosaminoglycan, hyaluronan (HA), maintains vascular integrity through endothelial glycocalyx modulation, caveolin-enriched microdomain regulation and interaction with endothelial HA binding proteins. Certain disease states increase hyaluronidase activity and reactive oxygen species (ROS) generation which break down high molecular weight HA to low molecular weight fragments causing damage to the endothelial glycocalyx. Further, these HA fragments can activate specific HA binding proteins upregulated in vascular disease to promote actin cytoskeletal reorganization and inhibition of endothelial cell-cell contacts. This review focuses on the crucial role of HA in vascular integrity and how HA degradation promotes vascular barrier disruption.

14.
Mol Biol Cell ; 21(22): 4042-56, 2010 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-20861316

RESUMO

Nonmuscle myosin light chain kinase (nmMLCK), a multi-functional cytoskeletal protein critical to vascular homeostasis, is highly regulated by tyrosine phosphorylation. We identified multiple novel c-Abl-mediated nmMLCK phosphorylation sites by mass spectroscopy analysis (including Y231, Y464, Y556, Y846) and examined their influence on nmMLCK function and human lung endothelial cell (EC) barrier regulation. Tyrosine phosphorylation of nmMLCK increased kinase activity, reversed nmMLCK-mediated inhibition of Arp2/3-mediated actin polymerization, and enhanced binding to the critical actin-binding phosphotyrosine protein, cortactin. EC challenge with sphingosine 1-phosphate (S1P), a potent barrier-enhancing agonist, resulted in c-Abl and phosphorylated nmMLCK recruitment into caveolin-enriched microdomains, rapid increases in Abl kinase activity, and spatial targeting of c-Abl to barrier-promoting cortical actin structures. Conversely, reduced c-Abl expression in EC (siRNA) markedly attenuated S1P-mediated cortical actin formation, reduced the EC modulus of elasticity (assessed by atomic force microscopy), reduced nmMLCK and cortactin tyrosine phosphorylation, and attenuated S1P-mediated barrier enhancement. These studies indicate an essential role for Abl kinase in vascular barrier regulation via posttranslational modification of nmMLCK and strongly support c-Abl-cortactin-nmMLCK interaction as a novel determinant of cortical actin-based cytoskeletal rearrangement critical to S1P-mediated EC barrier enhancement.


Assuntos
Células Endoteliais/metabolismo , Quinase de Cadeia Leve de Miosina/metabolismo , Proteínas Proto-Oncogênicas c-abl/metabolismo , Tirosina/metabolismo , Actinas/metabolismo , Sequência de Aminoácidos , Sítios de Ligação/genética , Western Blotting , Permeabilidade Capilar/efeitos dos fármacos , Caveolinas/metabolismo , Linhagem Celular , Citoesqueleto/efeitos dos fármacos , Citoesqueleto/metabolismo , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Ativação Enzimática , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Lisofosfolipídeos/farmacologia , Espectrometria de Massas , Microdomínios da Membrana/efeitos dos fármacos , Microdomínios da Membrana/metabolismo , Microscopia de Força Atômica , Microscopia Confocal , Dados de Sequência Molecular , Quinase de Cadeia Leve de Miosina/genética , Fosforilação , Ligação Proteica , Proteínas Proto-Oncogênicas c-abl/genética , Interferência de RNA , Esfingosina/análogos & derivados , Esfingosina/farmacologia , Tirosina/genética
15.
Future Oncol ; 6(8): 1237-42, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20799870

RESUMO

The possibility that anesthetic drugs can influence cancer recurrence rate is a subject of recent interest. Based on early in vitro data demonstrating opiates on breast cancer xenografts and two recent epidemiologic studies suggesting differences in recurrence rates in both breast and prostate cancer contingents dependent on whether patients received a combined regional-general anesthetic or a general anesthetic with opioid analgesia, there has been recent interest in the role of the micro-opioid receptor (MOR) in angiogenesis and oncogenic signaling. We recently demonstrated that morphine causes reciprocal transactivation of the MOR and VEGF receptors and that MOR-knockout mice do not develop significant tumors when injected with lung cancer cells as do their wild-type controls. Furthermore, infusion of the peripheral MOR antagonist methylnaltrexone markedly attenuates tumor growth in experimental mouse models. These experimental data support the hypothesis that opioids affect tumor progression and suggest the MOR as a potential target for chemotherapeutic drugs.


Assuntos
Analgésicos Opioides/farmacologia , Anestesia Geral/efeitos adversos , Neoplasias da Mama/prevenção & controle , Recidiva Local de Neoplasia/prevenção & controle , Neoplasias da Próstata/prevenção & controle , Animais , Neoplasias da Mama/patologia , Neoplasias da Mama/cirurgia , Feminino , Humanos , Masculino , Camundongos , Camundongos Knockout , Recidiva Local de Neoplasia/patologia , Recidiva Local de Neoplasia/cirurgia , Neoplasias da Próstata/patologia , Neoplasias da Próstata/cirurgia , Receptores Opioides/metabolismo , Resultado do Tratamento
16.
Am J Physiol Lung Cell Mol Physiol ; 299(5): L639-51, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20709728

RESUMO

Endothelial cell (EC) barrier dysfunction results in increased vascular permeability, a perturbation observed in inflammatory states, tumor angiogenesis, atherosclerosis, and both sepsis and acute lung injury. Therefore, agents that enhance EC barrier integrity have important therapeutic implications. We observed that binding of high-molecular-weight hyaluronan (HMW-HA) to its cognate receptor CD44 within caveolin-enriched microdomains (CEM) enhances human pulmonary EC barrier function. Immunocytochemical analysis indicated that HMW-HA promotes redistribution of a significant population of CEM to areas of cell-cell contact. Quantitative proteomic analysis of CEM isolated from human EC demonstrated HMW-HA-mediated recruitment of cytoskeletal regulatory proteins (annexin A2, protein S100-A10, and filamin A/B). Inhibition of CEM formation [caveolin-1 small interfering RNA (siRNA) and cholesterol depletion] or silencing (siRNA) of CD44, annexin A2, protein S100-A10, or filamin A/B expression abolished HMW-HA-induced actin cytoskeletal reorganization and EC barrier enhancement. To confirm our in vitro results in an in vivo model of inflammatory lung injury with vascular hyperpermeability, we observed that the protective effects of HMW-HA on LPS-induced pulmonary vascular leakiness were blocked in caveolin-1 knockout mice. Furthermore, targeted inhibition of CD44 expression in the mouse pulmonary vasculature significantly reduced HMW-HA-mediated protection from LPS-induced hyperpermeability. These data suggest that HMW-HA, via CD44-mediated CEM signaling events, represents a potentially useful therapeutic agent for syndromes of increased vascular permeability.


Assuntos
Vasos Sanguíneos/metabolismo , Permeabilidade Capilar , Ácido Hialurônico/química , Ácido Hialurônico/metabolismo , Pulmão/irrigação sanguínea , Lesão Pulmonar Aguda , Animais , Caveolina 1/metabolismo , Linhagem Celular , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Humanos , Receptores de Hialuronatos/genética , Receptores de Hialuronatos/metabolismo , Inflamação/metabolismo , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Peso Molecular , Proteoma/análise , RNA Interferente Pequeno/metabolismo
17.
J Biol Chem ; 285(24): 18575-85, 2010 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-20360610

RESUMO

Non-small cell lung cancer (NSCLC) has a poor prognosis and improved therapies are needed. Expression of EphA2 is increased in NSCLC metastases. In this study, we investigated EphA2 mutations in NSCLC and examined molecular pathways involved in NSCLC. Tumor and cell line DNA was sequenced. One EphA2 mutation was modeled by expression in BEAS2B cells, and functional and biochemical studies were conducted. A G391R mutation was detected in H2170 and 2/28 squamous cell carcinoma patient samples. EphA2 G391R caused constitutive activation of EphA2 with increased phosphorylation of Src, cortactin, and p130(Cas). Wild-type (WT) and G391R cells had 20 and 40% increased invasiveness; this was attenuated with knockdown of Src, cortactin, or p130(Cas). WT and G391R cells demonstrated a 70% increase in focal adhesion area. Mammalian target of rapamycin (mTOR) phosphorylation was increased in G391R cells with increased survival (55%) compared with WT (30%) and had increased sensitivity to rapamycin. A recurrent EphA2 mutation is present in lung squamous cell carcinoma and increases tumor invasion and survival through activation of focal adhesions and actin cytoskeletal regulatory proteins as well as mTOR. Further study of EphA2 as a therapeutic target is warranted.


Assuntos
Carcinoma de Células Escamosas/metabolismo , Adesões Focais/metabolismo , Regulação Neoplásica da Expressão Gênica , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Neoplasias Pulmonares/metabolismo , Mutação , Proteínas Serina-Treonina Quinases/metabolismo , Receptor EphA2/biossíntese , Linhagem Celular Tumoral , Sobrevivência Celular , Análise Mutacional de DNA , Humanos , Imuno-Histoquímica/métodos , Invasividade Neoplásica , Análise de Sequência com Séries de Oligonucleotídeos , Serina-Treonina Quinases TOR , Transfecção
18.
J Angiogenes Res ; 2(1): 5, 2010 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-20298531

RESUMO

BACKGROUND: Recent cancer therapies include drugs that target both tumor growth and angiogenesis including mammalian target of rapamycin (mTOR) inhibitors. Since mTOR inhibitor therapy is associated with significant side effects, we examined potential agents that can reduce the therapeutic dose. METHODS: Methylnaltrexone (MNTX), a peripheral mu opioid receptor (MOR) antagonist, in combination with the mTOR inhibitors temsirolimus and/or rapamycin, was evaluated for inhibition of VEGF-induced human pulmonary microvascular endothelial cell (EC) proliferation and migration as well as in vivo angiogenesis (mouse Matrigel plug assay). RESULTS: MNTX inhibited VEGF-induced EC proliferation and migration with an IC50 of approximately 100 nM. Adding 10 nM MNTX to EC shifted the IC50 of temsirolimus inhibition of VEGF-induced proliferation and migration from approximately 10 nM to approximately 1 nM and from approximately 50 to approximately 10 nM respectively. We observed similar effects with rapamycin. On a mechanistic level, we observed that MNTX increased EC plasma membrane-associated tyrosine phosphate activity. Inhibition of tyrosine phosphatase activity (3,4-dephostatin) blocked the synergy between MNTX and temsirolimus and increased VEGF-induced tyrosine phosphorylation of Src with enhanced PI3 kinase and mTOR Complex 2-dependent phosphorylation of Akt and subsequent activation of mTOR Complex 1 (rapamycin and temsirolimus target), while silencing Src, Akt or mTOR complex 2 components blocked VEGF-induced angiogenic events. CONCLUSIONS: Our data indicate that MNTX exerts a synergistic effect with rapamycin and temsirolimus on inhibition of VEGF-induced human EC proliferation and migration and in vivo angiogenesis. Therefore, addition of MNTX could potentially lower the dose of mTOR inhibitors which could improve therapeutic index.

19.
J Biol Chem ; 284(50): 34964-75, 2009 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-19833721

RESUMO

Reactive oxygen species (ROS) generation, particularly by the endothelial NADPH oxidase family of proteins, plays a major role in the pathophysiology associated with lung inflammation, ischemia/reperfusion injury, sepsis, hyperoxia, and ventilator-associated lung injury. We examined potential regulators of ROS production and discovered that hyperoxia treatment of human pulmonary artery endothelial cells induced recruitment of the vesicular regulator, dynamin 2, the non-receptor tyrosine kinase, c-Abl, and the NADPH oxidase subunit, p47(phox), to caveolin-enriched microdomains (CEMs). Silencing caveolin-1 (which blocks CEM formation) and/or c-Abl expression with small interference RNA inhibited hyperoxia-mediated tyrosine phosphorylation and association of dynamin 2 with p47(phox) and ROS production. In addition, treatment of human pulmonary artery endothelial cells with dynamin 2 small interfering RNA or the dynamin GTPase inhibitor, Dynasore, attenuated hyperoxia-mediated ROS production and p47(phox) recruitment to CEMs. Using purified recombinant proteins, we observed that c-Abl tyrosine-phosphorylated dynamin 2, and this phosphorylation increased p47(phox)/dynamin 2 association (change in the dissociation constant (K(d)) from 85.8 to 6.9 nm). Furthermore, exposure of mice to hyperoxia increased ROS production, c-Abl activation, dynamin 2 association with p47(phox), and pulmonary leak, events that were attenuated in the caveolin-1 knock-out mouse confirming a role for CEMs in ROS generation. These results suggest that hyperoxia induces c-Abl-mediated dynamin 2 phosphorylation required for recruitment of p47(phox) to CEMs and subsequent ROS production in lung endothelium.


Assuntos
Caveolina 1/metabolismo , Dinamina II/metabolismo , Células Endoteliais , Microdomínios da Membrana , NADPH Oxidases/metabolismo , Proteínas Proto-Oncogênicas c-abl/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Animais , Líquido da Lavagem Broncoalveolar/citologia , Caveolina 1/genética , Células Cultivadas , Dinamina II/genética , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Ativação Enzimática , Humanos , Hiperóxia/metabolismo , Pulmão/citologia , Pulmão/metabolismo , Masculino , Microdomínios da Membrana/química , Microdomínios da Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NADPH Oxidases/genética , Proteínas Proto-Oncogênicas c-abl/genética , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo
20.
Clin Cancer Res ; 15(18): 5714-23, 2009 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-19723643

RESUMO

PURPOSE: African Americans have higher incidence and poorer response to lung cancer treatment compared with Caucasians. However, the underlying molecular mechanisms for the significant ethnic difference are not known. The present study examines the ethnic differences in the type and frequency of MET proto-oncogene (MET) mutation in lung cancer and correlated them with other frequently mutated genes such as epidermal growth factor receptor (EGFR), KRAS2, and TP53. EXPERIMENTAL DESIGN: Using tumor tissue genomic DNA from 141 Asian, 76 Caucasian, and 66 African American lung cancer patients, exons coding for MET and EGFR were PCR amplified, and mutations were detected by sequencing. Mutation carriers were further screened for KRAS2 and TP53 mutations. Functional implications of important MET mutations were explored by molecular modeling and hepatocyte growth factor binding studies. RESULTS: Unlike the frequently encountered somatic mutations in EGFR, MET mutations in lung tumors were germline. MET-N375S, the most frequent mutation of MET, occurred in 13% of East Asians compared with none in African Americans. The frequency of MET mutations was highest among male smokers and squamous cell carcinoma. The MET-N375S mutation seems to confer resistance to MET inhibition based on hepatocyte growth factor ligand binding, molecular modeling, and apoptotic susceptibility to MET inhibitor studies. CONCLUSIONS: MET in lung cancer tissues contained nonsynonymous mutations in the semaphorin and juxtamembrane domains but not in the tyrosine kinase domain. All the MET mutations were germline. East Asians, African-Americans, and Caucasians had different MET genotypes and haplotypes. MET mutations in the semaphorin domain affected ligand binding.


Assuntos
Neoplasias Pulmonares/enzimologia , Neoplasias Pulmonares/genética , Proteínas Proto-Oncogênicas c-met/genética , Proteínas Proto-Oncogênicas c-met/metabolismo , Grupos Raciais/genética , Idoso , Idoso de 80 Anos ou mais , Povo Asiático/genética , População Negra/genética , Análise Mutacional de DNA , Inibidores Enzimáticos/farmacologia , Receptores ErbB/genética , Receptores ErbB/metabolismo , Etnicidade/genética , Éxons , Feminino , Genótipo , Humanos , Indóis/farmacologia , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/metabolismo , Masculino , Pessoa de Meia-Idade , Modelos Moleculares , Mutação , Piperazinas/farmacologia , Reação em Cadeia da Polimerase , Prognóstico , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas c-met/antagonistas & inibidores , Semaforinas/química , Semaforinas/genética , Sulfonamidas/farmacologia , População Branca/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA