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












Base de datos
Intervalo de año de publicación
1.
FASEB J ; 35(3): e21427, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33629776

RESUMEN

Porphyrins are used for cancer diagnostic and therapeutic applications, but the mechanism of how porphyrins accumulate in cancer cells remains elusive. Knowledge of how porphyrins enter cancer cells can aid the development of more accurate cancer diagnostics and therapeutics. To gain insight into porphyrin uptake mechanisms in cancer cells, we developed a flow cytometry assay to quantify cellular uptake of meso-tetra (4-carboxyphenyl) porphyrin (TCPP), a porphyrin that is currently being developed for cancer diagnostics. We found that TCPP enters cancer cells through clathrin-mediated endocytosis. The LDL receptor, previously implicated in the cellular uptake of other porphyrins, only contributes modestly to uptake. We report that TCPP instead binds strongly ( KD=42nM ) to CD320, the cellular receptor for cobalamin/transcobalamin II (Cbl/TCN2). Additionally, TCPP competes with Cbl/TCN2 for CD320 binding, suggesting that CD320 is a novel receptor for TCPP. Knockdown of CD320 inhibits TCPP uptake by up to 40% in multiple cancer cell lines, including lung, breast, and prostate cell lines, which supports our hypothesis that CD320 both binds to and transports TCPP into cancer cells. Our findings provide some novel insights into why porphyrins concentrate in cancer cells. Additionally, our study describes a novel function for the CD320 receptor which has been reported to transport only Cbl/TCN2 complexes.


Asunto(s)
Neoplasias/metabolismo , Porfirinas/farmacología , Vitamina B 12/farmacología , Transporte Biológico/efectos de los fármacos , Endocitosis/efectos de los fármacos , Endocitosis/fisiología , Humanos , Neoplasias/tratamiento farmacológico , Porfirinas/metabolismo , Receptores de LDL/efectos de los fármacos , Receptores de LDL/metabolismo , Vitamina B 12/metabolismo
2.
Cell Rep ; 33(5): 108332, 2020 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-33147457

RESUMEN

We report here that the autocrine signaling mediated by growth and differentiation factor 6 (GDF6), a member of the bone morphogenetic protein (BMP) family of cytokines, maintains Ewing sarcoma growth by preventing Src hyperactivation. Surprisingly, Ewing sarcoma depends on the prodomain, not the BMP domain, of GDF6. We demonstrate that the GDF6 prodomain is a ligand for CD99, a transmembrane protein that has been widely used as a marker of Ewing sarcoma. The binding of the GDF6 prodomain to the CD99 extracellular domain results in recruitment of CSK (C-terminal Src kinase) to the YQKKK motif in the intracellular domain of CD99, inhibiting Src activity. GDF6 silencing causes hyperactivation of Src and p21-dependent growth arrest. We demonstrate that two GDF6 prodomain mutants linked to Klippel-Feil syndrome are hyperactive in CD99-Src signaling. These results reveal a cytokine signaling pathway that regulates the CSK-Src axis and cancer cell proliferation and suggest the gain-of-function activity for disease-causing GDF6 mutants.


Asunto(s)
Antígeno 12E7/metabolismo , Factor 6 de Diferenciación de Crecimiento/metabolismo , Sarcoma de Ewing/metabolismo , Sarcoma de Ewing/patología , Transducción de Señal , Familia-src Quinasas/metabolismo , Animales , Proteína Tirosina Quinasa CSK/metabolismo , Proliferación Celular , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica , Factor 6 de Diferenciación de Crecimiento/química , Humanos , Síndrome de Klippel-Feil/genética , Ratones SCID , Mutación/genética , Proteínas de Fusión Oncogénica/metabolismo , Dominios Proteicos , Proteoma/metabolismo , Proteómica , Proteína Proto-Oncogénica c-fli-1/metabolismo , Proteína EWS de Unión a ARN/metabolismo , Transcripción Genética
3.
Transfusion ; 57(11): 2690-2700, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28880373

RESUMEN

BACKGROUND: Although transfusion is a lifesaving intervention, it may be associated with significant morbidity in injured patients. We hypothesize that stored red blood cells (RBCs) induce proinflammatory activation of human pulmonary microvascular endothelial cells (HMVECs) resulting in neutrophil (PMN) adhesion and predisposition to acute lung injury (ALI). STUDY DESIGN AND METHODS: Ten units of RBCs were collected; 50% (by weight) were leukoreduced (LR-RBCs) and the remainder was unmodified and stored in additive solution-5 (AS-5). An additional 10 units of RBCs were collected, leukoreduced, and stored in AS-3. HMVECs were incubated with [10%-40%]FINAL of the supernatants on Day (D)1 to D42 of storage, lipid extracts, and purified lipids. Endothelial surface expression of intercellular adhesion molecule-1 (ICAM-1), interleukin (IL)-8 release, and PMN adhesion to HMVECs were measured. HMVEC signaling via the BLT2 receptor was evaluated. Supernatants and lipids were also employed as the first event in a two-event model of ALI. RESULTS: The supernatants [10%-40%]FINAL from D21 LR-RBCs and D42 RBCs and LR-RBCs and the lipids from D42 stored in AS-5 induced increased ICAM-1 surface expression on endothelium, IL-8 release, and PMN adhesion. In addition, the supernatants [20%-40%]FINAL from D21 and D42 RBCs in AS-5 also increased endothelial surface expression of ICAM-1. D42 supernatants and lipids also caused coprecipitation of ß-arrestin-1 with BLT2, protein kinase C (PKC)ßI , and PKCδ and served as the first event in a two-event rodent model of ALI. CONCLUSION: Lipids that accumulate during RBC storage activate endothelium and predispose to ALI, which may explain some of the adverse events associated with the transfusion of critically injured patients.


Asunto(s)
Conservación de la Sangre/métodos , Eritrocitos/citología , Lípidos/farmacología , Pulmón/irrigación sanguínea , Proteína Quinasa C/metabolismo , Receptores de Leucotrieno B4/metabolismo , Lesión Pulmonar Aguda/etiología , Medios de Cultivo Condicionados/farmacología , Células Endoteliales/metabolismo , Activación Enzimática , Transfusión de Eritrocitos/efectos adversos , Humanos , Microvasos/citología , Neumonía/etiología
4.
J Leukoc Biol ; 101(1): 261-273, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27531930

RESUMEN

Lysophosphatidylcholines (lysoPCs) are effective polymorphonuclear neutrophil (PMN) priming agents implicated in transfusion-related acute lung injury (TRALI). LysoPCs cause ligation of the G2A receptor, cytosolic Ca2+ flux, and activation of Hck. We hypothesize that lysoPCs induce Hck-dependent activation of protein kinase C (PKC), resulting in phosphorylation and membrane translocation of 47 kDa phagocyte oxidase protein (p47phox). PMNs, human or murine, were primed with lysoPCs and were smeared onto slides and examined by digital microscopy or separated into subcellular fractions or whole-cell lysates. Proteins were immunoprecipitated or separated by polyacrylamide gel electrophoresis and immunoblotted for proteins of interest. Wild-type (WT) and PKCγ knockout (KO) mice were used in a 2-event model of TRALI. LysoPCs induced Hck coprecipitation with PKCδ and PKCγ and the PKCδ:PKCγ complex also had a fluorescence resonance energy transfer (FRET)+ interaction with lipid rafts and Wiskott-Aldrich syndrome protein family verprolin-homologous protein 2 (WAVE2). PKCγ then coprecipitated with p47phox Immunoblotting, immunoprecipitation (IP), specific inhibitors, intracellular depletion of PKC isoforms, and PMNs from PKCγ KO mice demonstrated that Hck elicited activation/Tyr phosphorylation (Tyr311 and Tyr525) of PKCδ, which became Thr phosphorylated (Thr507). Activated PKCδ then caused activation of PKCγ, both by Tyr phosphorylation (Τyr514) and Ser phosphorylation, which induced phosphorylation and membrane translocation of p47phox In PKCγ KO PMNs, lysoPCs induced Hck translocation but did not evidence a FRET+ interaction between PKCδ and PKCγ nor prime PMNs. In WT mice, lysoPCs served as the second event in a 2-event in vivo model of TRALI but did not induce TRALI in PKCγ KO mice. We conclude that lysoPCs prime PMNs through Hck-dependent activation of PKCδ, which stimulates PKCγ, resulting in translocation of phosphorylated p47phox.


Asunto(s)
Membrana Celular/metabolismo , Lisofosfatidilcolinas/farmacología , NADPH Oxidasas/metabolismo , Neutrófilos/metabolismo , Proteína Quinasa C-delta/metabolismo , Proteína Quinasa C/metabolismo , Proteínas Proto-Oncogénicas c-hck/metabolismo , Animales , Calcio/metabolismo , Membrana Celular/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Humanos , Lesión Pulmonar/patología , Ratones , Ratones Noqueados , Neutrófilos/efectos de los fármacos , Fosforilación/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Transporte de Proteínas/efectos de los fármacos , Proteínas Recombinantes/farmacología
5.
Genes Cancer ; 7(3-4): 125-35, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27382436

RESUMEN

Hepatoblastoma is the most common liver cancer in children, accounting for over 65% of all childhood liver malignancies. Hepatoblastoma is distinct from adult liver cancer in that it is not associated with hepatitis virus infection, cirrhosis, or other underlying liver pathology. The paucity of appropriate cell and animal models has been hampering the mechanistic understanding of hepatoblastoma pathogenesis. Consequently, there is no molecularly targeted therapy for hepatoblastoma. To gain insight into cytokine signaling in hepatoblastoma, we employed mass spectrometry to analyze the proteins secreted from Hep293TT hepatoblastoma cell line we established and identified the specific secretion of fibroblast growth factor 19 (FGF19), a growth factor for liver cells. We determined that silencing FGF19 by shRNAs or neutralizing secreted FGF19 by anti-FGF19 antibody inhibits the proliferation of hepatoblastoma cells. Furthermore, blocking FGF19 signaling by an FGF receptor kinase inhibitor suppressed hepatoblastoma growth. RNA expression analysis in hepatoblastoma tumors revealed that the high expression of FGF19 signaling pathway components as well as the low expression of FGF19 signaling repression targets correlates with the aggressiveness of the tumors. These results suggest the role of FGF19 as autocrine growth factor for hepatoblastoma.

6.
Exp Gerontol ; 73: 1-4, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26547052

RESUMEN

Cellular senescence has been proposed to play critical roles in tumor suppression and organismal aging, but the molecular mechanism of senescence remains incompletely understood. Here we report that a putative lysosomal carbohydrate efflux transporter, Spinster, induces cellular senescence in human primary fibroblasts. Administration of d-galactose synergistically enhanced Spinster-induced senescence and this synergism required the transporter activity of Spinster. Intracellular d-galactose is metabolized to galactose-1-phosphate by galactokinase. Galactokinase-deficient fibroblasts, which accumulate intracellular d-galactose, displayed increased baseline senescence. Senescence of galactokinase-deficient fibroblasts was further enhanced by d-galactose administration and was diminished by restoration of wild-type galactokinase expression. Silencing galactokinase in normal fibroblasts also induced senescence. These results suggest a role for intracellular galactose in the induction of cellular senescence.


Asunto(s)
Senescencia Celular/fisiología , Galactosa/fisiología , Proteínas Adaptadoras Transductoras de Señales/farmacología , Proteínas Adaptadoras Transductoras de Señales/fisiología , Células Cultivadas , Senescencia Celular/efectos de los fármacos , Sinergismo Farmacológico , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Galactoquinasa/deficiencia , Galactoquinasa/fisiología , Galactosa/farmacología , Humanos , Lisosomas/metabolismo , Proteínas de la Membrana/farmacología , Proteínas de la Membrana/fisiología
7.
Genes Cancer ; 6(11-12): 452-61, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26807198

RESUMEN

Ewing sarcoma is a cancer of bone and soft tissue in children that is characterized by a chromosomal translocation involving EWS and an Ets family transcription factor, most commonly FLI-1. The EWS-FLI-1 fusion oncogene is widely believed to play a central role in Ewing sarcoma. The EWS-FLI-1 gene product regulates the expression of a number of genes important for cancer progression, can transform mouse cells such as NIH3T3 and C3H10T1/2, and is necessary for proliferation and tumorigenicity of Ewing sarcoma cells, suggesting that EWS-FLI-1 is the causative oncogene. However, a variety of evidence also suggest that EWS-FLI-1 alone cannot fully explain the Ewing sarcomagenesis. Here we report that FLI-1-EWS, a fusion gene reciprocal to EWS-FLI-1, is frequently expressed in Ewing sarcoma. We present evidence suggesting that endogenous FLI-1-EWS is required for Ewing sarcoma growth and that FLI-1-EWS cooperates with EWS-FLI-1 in human mesenchymal stem cells, putative cells of origin of Ewing sarcoma, through abrogation of the proliferation arrest induced by EWS- FLI-1.

8.
J Proteome Res ; 13(8): 3783-91, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24999758

RESUMEN

Ewing sarcoma is a cancer of bone and soft tissue in children that is characterized by a chromosomal translocation involving EWS and an Ets family transcription factor, most commonly Fli-1. EWS-Fli-1 fusion accounts for 85% of cases. The growth and survival of Ewing sarcoma cells are critically dependent on EWS-Fli-1. A large body of evidence has established that EWS-Fli-1 functions as a DNA-binding transcription factor that regulates the expression of a number of genes important for cell proliferation and transformation. However, little is known about the biochemical properties of the EWS-Fli-1 protein. We undertook a series of proteomic analyses to dissect the EWS-Fli-1 interactome. Employing a proximity-dependent biotinylation technique, BioID, we identified cation-independent mannose 6-phosphate receptor (CIMPR) as a protein located in the vicinity of EWS-Fli-1 within a cell. CIMPR is a cargo that mediates the delivery of lysosomal hydrolases from the trans-Golgi network to the endosome, which are subsequently transferred to the lysosomes. Further molecular cell biological analyses uncovered a role for lysosomes in the turnover of the EWS-Fli-1 protein. We demonstrate that an mTORC1 active-site inhibitor, torin 1, which stimulates the TFEB-lysosome pathway, can induce the degradation of EWS-Fli-1, suggesting a potential therapeutic approach to target EWS-Fli-1 for degradation.


Asunto(s)
Lisosomas/metabolismo , Proteínas de Fusión Oncogénica/fisiología , Proteómica , Proteína Proto-Oncogénica c-fli-1/fisiología , Proteína EWS de Unión a ARN/fisiología , Sarcoma de Ewing/tratamiento farmacológico , Biotinilación , Dominio Catalítico , Línea Celular Tumoral , Proliferación Celular , Transformación Celular Neoplásica/genética , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Células HeLa , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina , Complejos Multiproteicos/metabolismo , Proteoma/metabolismo , Sarcoma de Ewing/patología , Serina-Treonina Quinasas TOR/metabolismo , Espectrometría de Masas en Tándem , Factores de Transcripción/metabolismo , Red trans-Golgi/metabolismo
9.
Mol Cell Biol ; 32(21): 4388-99, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22927647

RESUMEN

Cellular senescence has emerged as a critical tumor suppressive mechanism in recent years, but relatively little is known about how senescence occurs. Here, we report that secreted Frizzled-related protein 1 (SFRP1), a secreted antagonist of Wnt signaling, is oversecreted upon cellular senescence caused by DNA damage or oxidative stress. SFRP1 is necessary for stress-induced senescence caused by these factors and is sufficient for the induction of senescence phenotypes. We present evidence suggesting that SFRP1 functions as a secreted mediator of senescence through inhibition of Wnt signaling and activation of the retinoblastoma (Rb) pathway and that cancer-associated SFRP1 mutants are defective for senescence induction.


Asunto(s)
Senescencia Celular , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas de la Membrana/metabolismo , Proteína de Retinoblastoma/metabolismo , Proteínas Wnt/antagonistas & inhibidores , Vía de Señalización Wnt , Línea Celular Tumoral , Proliferación Celular , Daño del ADN , Fibroblastos , Regulación Neoplásica de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Proteínas de la Membrana/genética , Mutación , Estrés Oxidativo , Regiones Promotoras Genéticas , Interferencia de ARN , ARN Interferente Pequeño , Transducción de Señal , Proteínas Wnt/genética
10.
Proc Natl Acad Sci U S A ; 109(30): 12052-7, 2012 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-22778398

RESUMEN

Cellular senescence is widely believed to play a key role in tumor suppression, but the molecular pathways that regulate senescence are only incompletely understood. By using a secretome proteomics approach, we identified insulin-like growth factor binding protein 3 (IGFBP3) as a secreted mediator of breast cancer senescence upon chemotherapeutic drug treatment. The senescence-inducing activity of IGFBP3 is inhibited by tissue-type plasminogen activator-mediated proteolysis, which is counteracted by plasminogen activator inhibitor 1 (PAI-1), another secreted mediator of senescence. We demonstrate that IGFBP3 is a critical downstream target of PAI-1-induced senescence. These results suggest a role for an extracellular cascade of secreted proteins in the regulation of cellular senescence.


Asunto(s)
Senescencia Celular/fisiología , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Neoplasias/tratamiento farmacológico , Inhibidor 1 de Activador Plasminogénico/metabolismo , Proteolisis/efectos de los fármacos , Estrés Fisiológico/fisiología , Activador de Tejido Plasminógeno/farmacología , Análisis de Varianza , Línea Celular Tumoral , Medios de Cultivo/química , Cartilla de ADN/genética , Doxorrubicina/farmacología , Ensayo de Inmunoadsorción Enzimática , Técnica del Anticuerpo Fluorescente , Humanos , Immunoblotting , Inmunohistoquímica , Neoplasias/metabolismo , Neoplasias/fisiopatología , Proteómica/métodos , ARN Interferente Pequeño/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Activador de Tejido Plasminógeno/metabolismo , Células Tumorales Cultivadas , beta-Galactosidasa
11.
Transfusion ; 51(12): 2549-54, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21615744

RESUMEN

BACKGROUND: Lipids accumulate during the storage of red blood cells (RBCs), prime neutrophils (PMNs), and have been implicated in transfusion-related acute lung injury (TRALI). These lipids are composed of two classes: nonpolar lipids and lysophosphatidylcholines based on their retention time on separation by high-pressure liquid chromatography. Prestorage leukoreduction significantly decreases white blood cell and platelet contamination of RBCs; therefore, it is hypothesized that prestorage leukoreduction changes the classes of lipids that accumulate during storage, and these lipids prime PMNs and induce acute lung injury (ALI) as the second event in a two-event in vivo model. STUDY DESIGN AND METHODS: RBC units were divided: 50% was leukoreduced (LR-RBCs), stored, and sampled on Day 1 and at the end of storage, Day 42. Priming activity was evaluated on isolated PMNs, and the purified lipids from Day 1 or Day 42 were used as the second event in the in vivo model. RESULTS: The plasma and lipids from RBCs and LR-RBCs primed PMNs, and the LR-RBC activity decreased with longer storage. Unlike RBCs, nonpolar lipids comprised the PMN-priming activity from stored LR-RBCs. Mass spectroscopy identified these lipids as arachidonic acid and 5-, 12-, and 15-hydroxyeicsotetranoic acid. At concentrations from Day 42, but not Day 1, three of four of these lipids individually, and the mixture, primed PMNs. The mixture also caused ALI as the second event in a two-event model of TRALI. CONCLUSION: We conclude that the nonpolar lipids that accumulate during LR-RBC storage may represent the agents responsible for antibody-negative TRALI.


Asunto(s)
Lesión Pulmonar Aguda/etiología , Ácido Araquidónico/análisis , Conservación de la Sangre/efectos adversos , Eritrocitos/química , Ácidos Hidroxieicosatetraenoicos/análisis , Leucocitos Mononucleares/química , Ácido Araquidónico/metabolismo , Transfusión de Eritrocitos/efectos adversos , Eritrocitos/citología , Eritrocitos/metabolismo , Femenino , Humanos , Ácidos Hidroxieicosatetraenoicos/metabolismo , Leucaféresis/métodos , Leucocitos Mononucleares/metabolismo , Masculino , Espectrometría de Masas , Plasma/química , Plasma/metabolismo , Factores de Tiempo
12.
Shock ; 35(3): 240-4, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20926984

RESUMEN

Leukotrienes are proinflammatory lipid mediators, derived from arachidonic acid via 5-lipoxygenase (5-LO). Leukotriene B4 (LTB4) is an effective polymorphonuclear neutrophil (PMN) chemoattractant, as well as being a major product of PMN priming. Leukotriene B4 is rapidly metabolized into products that are thought to be inactive, and little is known about the effects of LTB4 on the pulmonary endothelium. We hypothesize that LTB4 and its metabolites are effective PMN priming agents and cause proinflammatory activation of pulmonary endothelial cells. Isolated PMNs were primed (5 min, 37°C) with serial concentrations 10 to 10 M of LTB4 and its metabolites: 6-trans-LTB4, 20-OH-LTB4, and 20-COOH-LTB4, and then activated with fMLP. Primary human pulmonary microvascular endothelial cells (HMVECs) were incubated with these lipids (6 h, 37°C, 5% CO2), and intercellular adhesion molecule 1 was measured by flow cytometry. Polymorphonuclear neutrophil adhesion was measured by myeloperoxidase assays, and to ensure that these reactions were specific to the LTB4 receptors, BLT1 and BLT2 were antagonized with CP105,696 (BLT1) or silenced with siRNA (BLT1 and BLT2). Leukotriene B4 and its metabolites primed PMNs over a wide range of concentrations, depending on the specific metabolite. In addition, at high concentrations these lipids also caused increases in the surface expression of intercellular adhesion molecule 1 on HMVECs and induced HMVEC-mediated adhesion of PMNs. Silencing of BLT2 abrogated HMVEC activation, and blockade of BLT1 inhibited the observed PMN priming activity. We conclude that LTB4 and its ω-oxidation and nonenzymatic metabolites prime PMNs over a range of concentrations and activate HMVECs. These data have expanded the repertoire of causative agents in acute lung injury and postinjury multiple organ failure.


Asunto(s)
Células Endoteliales/metabolismo , Neutrófilos/enzimología , Neutrófilos/metabolismo , Oxidorreductasas/metabolismo , Células Cultivadas , Cromatografía Líquida de Alta Presión , Células Endoteliales/inmunología , Humanos , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/metabolismo , Leucotrieno B4/genética , Leucotrieno B4/metabolismo , Pulmón/citología , Pulmón/metabolismo , Neutrófilos/inmunología , Interferencia de ARN , Receptores de Leucotrieno B4/genética , Receptores de Leucotrieno B4/metabolismo
13.
J Neurotrauma ; 26(11): 2071-82, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19469685

RESUMEN

Abstract Physical training in experimental animals can improve locomotor activity via the regulation of spinal neural circuitry or peripheral nerve regeneration. Here we investigated the effects of treadmill training (TMT) on regenerative responses of the corticospinal tract (CST) after contusive spinal cord injury (SCI). One week after injury of the low thoracic spinal cord, rats were given TMT or sedentary treatment for 1-4 weeks. Anterograde tracing of descending CST axons revealed that TMT enhanced collateral arborization of CST axons surrounding the injury cavity and promoted extension into the caudal spinal cord. The number of oligodendrocytes in the vicinity of the injury cavity was significantly increased at 2 or 4 weeks after TMT compared to sedentary controls. The data further showed that TMT increased phosphorylation of Erk1/2 in the motor cortex as well as the spinal cord injury area, and inhibition of Erk1/2 activity by administration of the MEK1 inhibitors PD98059 and U0126 reduced collateral outgrowth of descending CST axons in TMT animals. TMT for 2-4 weeks significantly improved behavioral scores as assessed by the Basso-Beattie-Bresnahan scale, as well as on motor function and gridwalk testing. Our data imply that Erk1/2 may be an important mediator for transmitting signals from the injury site to the cell body, and further suggest that activation of the Erk1/2 signaling pathway may be involved in enhanced outgrowth of CST axons after TMT.


Asunto(s)
Axones/fisiología , Terapia por Ejercicio , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Regeneración Nerviosa/fisiología , Traumatismos de la Médula Espinal/rehabilitación , Animales , Axones/patología , Western Blotting , Encéfalo/enzimología , Activación Enzimática/fisiología , Inmunohistoquímica , Masculino , Condicionamiento Físico Animal/fisiología , Tractos Piramidales/fisiología , Ratas , Ratas Sprague-Dawley , Transducción de Señal/fisiología , Traumatismos de la Médula Espinal/enzimología , Traumatismos de la Médula Espinal/patología
14.
J Immunol ; 180(12): 8192-203, 2008 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-18523285

RESUMEN

Neutrophils (polymorphonuclear leukocytes, PMNs) are vital to innate immunity and receive proinflammatory signals that activate G protein-coupled receptors (GPCRs). Because GPCRs transduce signals through clathrin-mediated endocytosis (CME), we hypothesized that platelet-activating factor (PAF), an effective chemoattractant that primes the PMN oxidase, would signal through CME, specifically via dynamin-2 activation and endosomal formation resulting in membrane translocation of cytosolic phagocyte oxidase (phox) proteins. PMNs were incubated with buffer or 2 muM PAF for 1-3 min, and in some cases activated with PMA, and O(2)(-) was measured, whole-cell lysates and subcellular fractions were prepared, or the PMNs were fixed onto slides for digital or electron microscopy. PAF caused activation of dynamin-2, resulting in endosomal formation that required PI3K and contained early endosomal Ag-1 (EEA-1) and Rab5a. The apoptosis signal-regulating kinase-1/MAPK kinase-3/p38 MAPK signalosome assembled on Rab5a and phosphorylated EEA-1 and Rab GDP dissociation inhibitor, with the latter causing Rab5a activation. Electron microscopy demonstrated that PAF caused two distinct sites for activation of p38 MAPK. EEA-1 provided a scaffold for recruitment of the p40(phox)-p67(phox) complex and PI3K-dependent Akt1 phosphorylation of these two phox proteins. PAF induced membrane translocation of p40(phox)-p67(phox) localizing to gp91(phox), which was PI3K-, but not p47(phox)-, dependent. In conclusion, PAF transduces signals through CME, and such GPCR signaling may allow for pharmacological manipulation of these cells to decrease PMN-mediated acute organ injury.


Asunto(s)
Membrana Celular/metabolismo , Endosomas/metabolismo , Neutrófilos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfoproteínas/metabolismo , Factor de Activación Plaquetaria/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Proteínas de Unión al GTP rab5/metabolismo , Membrana Celular/enzimología , Dinamina II/metabolismo , Endosomas/enzimología , Activación Enzimática/fisiología , Transferencia Resonante de Energía de Fluorescencia , Humanos , Ligandos , Sistema de Señalización de MAP Quinasas/fisiología , Neutrófilos/enzimología , Factor de Activación Plaquetaria/metabolismo , Glicoproteínas de Membrana Plaquetaria/metabolismo , Transporte de Proteínas/fisiología , Receptores Acoplados a Proteínas G/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología
15.
Mol Cell Biol ; 28(15): 4819-28, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18505829

RESUMEN

Transcription of the catalytic subunit of telomerase (hTERT) in keratinocytes can be induced by human papillomavirus type 16 (HPV16) E6/E6AP ubiquitin ligase through degradation of the repressor, NFX1-91. Here, we demonstrate that NFX1-91 interacts with the corepressor complex mSin3A/histone deacetylase (HDAC) at the hTERT promoter. By degrading NFX1-91, E6/E6AP changes the chromatin structure at the hTERT promoter as indicated by enhanced acetylation of histones H3 and H4 as well as dimethylation of H3K4. Knockdown of NFX1-91 by short hairpin RNA (shRNA) mimics the effect of E6 and leads to acetylation of histones H3 and H4. Conversely, knockdown of E6AP by shRNA suppresses histone acetylation at the hTERT promoter. These data demonstrate that targeted degradation of NFX1-91 by E6/E6AP dissociates the mSin3A/HDAC complex from the hTERT promoter and induces hTERT transcription.


Asunto(s)
Histona Desacetilasas/metabolismo , Queratinocitos/enzimología , Queratinocitos/metabolismo , Proteínas Represoras/metabolismo , Telomerasa/genética , Transcripción Genética , Acetilación , Células Cultivadas , Cromatina/metabolismo , Activación Enzimática , Semivida , Histonas/metabolismo , Humanos , Inmunoprecipitación , Modelos Biológicos , Proteínas Mutantes/metabolismo , Proteínas Oncogénicas Virales/metabolismo , Regiones Promotoras Genéticas/genética , Unión Proteica , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteínas Represoras/química , Complejo Correpresor Histona Desacetilasa y Sin3 , Ubiquitina-Proteína Ligasas/metabolismo
16.
Cell Calcium ; 34(6): 445-55, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14572803

RESUMEN

Chemoattractant priming and activation of PMNs results in changes in cytosolic Ca2+ concentration, tyrosine kinase activity, and gene expression. We hypothesize that the initial signaling for the activation of a 105kDa protein (Rel-1) requires Ca2+-dependent tyrosine phosphorylation. A rapid and time-dependent tyrosine phosphorylation of Rel-1 occurred following formyl-Met-Leu-Phe (fMLP) stimulation of human PMNs at concentrations that primed or activated the NADPH oxidase (10(-9) to 10(-6)M), becoming maximal after 30s. Pretreatment with pertussis toxin (Ptx) or tyrosine kinase inhibitors abrogated this phosphorylation and inhibited fMLP activation of the oxidase. The fMLP concentrations employed also caused a rapid increase in cytosolic Ca2+ but chelation negated the effects, including the cytosolic Ca2+ flux, oxidase activation, and the tyrosine phosphorylation of Rel-1. Conversely, chelation of extracellular Ca2+ decreased the fMLP-mediated Ca2+ flux, had no affect on the oxidase, and augmented tyrosine phosphorylation of Rel-1. Phosphorylation of Rel-1 was inhibited when PMNs were preincubated with a p38 MAP kinase (MAPK) inhibitor (SB203580). In addition, fMLP elicited rapid activation of p38 MAPK which was abrogated by chelation of cytosolic Ca2+. Thus, fMLP concentrations that prime or activate the oxidase cause a rapid Ca2+-dependent tyrosine phosphorylation of Rel-1 involving p38 MAPK activation.


Asunto(s)
Calcio/metabolismo , N-Formilmetionina Leucil-Fenilalanina/farmacología , Neutrófilos/metabolismo , Factores de Transcripción/metabolismo , Tirosina/metabolismo , Citosol , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Expresión Génica , Humanos , Proteínas I-kappa B/metabolismo , Imidazoles/farmacología , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Peso Molecular , N-Formilmetionina Leucil-Fenilalanina/antagonistas & inhibidores , NADPH Oxidasas/efectos de los fármacos , NADPH Oxidasas/metabolismo , FN-kappa B/metabolismo , Neutrófilos/efectos de los fármacos , Toxina del Pertussis/farmacología , Fosforilación/efectos de los fármacos , Proteínas Tirosina Quinasas/metabolismo , Proteínas Tirosina Quinasas/farmacología , Piridinas/farmacología , Factores de Transcripción/genética , Proteínas Quinasas p38 Activadas por Mitógenos
17.
J Leukoc Biol ; 73(4): 511-24, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12660226

RESUMEN

A mixture of lysophosphatidylcholines (lyso-PCs) are generated during blood storage and are etiologic in models of acute lung injury. We hypothesize that lyso-PCs stimulate polymorphonuclear neutrophils (PMNs) through Ca(2)(+)-dependent signaling. The lyso-PC mix (0.45-14.5 micro M) and the individual lyso-PCs primed formyl-Met-Leu-Phe (fMLP) activation of the oxidase (1.8- to 15.7-fold and 1.7- to 14.8-fold; P<0.05). Labeled lyso-PCs demonstrated a membrane association with PMNs and caused rapid increases in cytosolic Ca(2)(+). Receptor desensitization studies implicated a common receptor or a family of receptors for the observed lyso-PC-mediated changes in PMN priming, and cytosolic Ca(2)(+) functions were pertussis toxin-sensitive. Lyso-PCs caused rapid serine phosphorylation of a 68-kD protein but did not activate mitogen-activated protein kinases or cause changes in tyrosine phosphorylation. With respect to alterations in PMN function, lyso-PCs caused PMN adherence, increased expression of CD11b and the fMLP receptor, reduced chemotaxis, provoked changes in morphology, elicited degranulation, and augmented fMLP-induced azurophilic degranulation (P<0.05). Cytosolic Ca(2)(+) chelation inhibited lyso-PC-mediated priming of the oxidase, CD11b surface expression, changes in PMN morphology, and serine phosphorylation of the 68-kD protein. In conclusion, lyso-PCs affect multiple PMN functions in a Ca(2)(+)-dependent manner that involves the activation of a pertussis toxin-sensitive G-protein.


Asunto(s)
Calcio/metabolismo , Lisofosfatidilcolinas/farmacología , NADPH Oxidasas/metabolismo , Neutrófilos/fisiología , Receptores Acoplados a Proteínas G , Antígenos CD11/metabolismo , Señalización del Calcio , Adhesión Celular/efectos de los fármacos , Quimiotaxis/efectos de los fármacos , Citosol , Activación Enzimática , Humanos , Péptidos y Proteínas de Señalización Intercelular , Lactoferrina/metabolismo , Lisofosfatidilcolinas/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/metabolismo , N-Formilmetionina Leucil-Fenilalanina , Neutrófilos/efectos de los fármacos , Elastasa Pancreática/metabolismo , Peroxidasa/metabolismo , Toxina del Pertussis/farmacología , Fosforilación/efectos de los fármacos , Glicoproteínas de Membrana Plaquetaria/antagonistas & inhibidores , Receptores de Superficie Celular/antagonistas & inhibidores , Receptores de Formil Péptido , Receptores Inmunológicos/metabolismo , Receptores de Péptidos/metabolismo , Serina/metabolismo , Tirosina/metabolismo
18.
Blood ; 101(2): 454-62, 2003 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-12393667

RESUMEN

Transfusion-related acute lung injury (TRALI) is a life-threatening complication of hemotherapy. We report a series of 90 TRALI reactions in 81 patients secondary to transfusion with whole blood platelets (72 reactions), apheresis platelets (2), packed red cells (15), and plasma (1). The overall prevalence was 1 in 1120 cellular components. To examine the epidemiology of TRALI, we completed a nested case-control study of the first 46 patients with TRALI compared with 225 controls who had received transfusions. We then completed a prospective analysis of possible biologic response modifiers responsible for 51 of the TRALI cases, including human leukocyte antigen (HLA) class I, class II, and granulocyte antibodies in donors and neutrophil (PMN) priming activity in the plasma of the implicated units and recipients. Two groups were at risk: patients with hematologic malignancies (P <.0004) and patients with cardiac disease (P <.0006). TRALI was associated with older platelets (P =.014). In the prospective study, antileukocyte antibodies were found in only 3.6% of cases. The implicated blood components had greater PMN priming activity than controls (P <.05), and compared with pretransfusion samples, TRALI patients' plasma demonstrated increases in both interleukin 6 (IL-6) and lipid (neutral lipids and lysophosphatidylcholines) priming activity (P <.05). We conclude that TRALI may be more frequent than previously recognized and that patient susceptibility, product age, and increased levels of bioactive lipids in components may predispose patients to TRALI. TRALI, like the acute respiratory distress syndrome, may be a 2-event phenomenon with both recipient predisposition and factors in the stored units playing major roles.


Asunto(s)
Síndrome de Dificultad Respiratoria/etiología , Reacción a la Transfusión , Distribución por Edad , Estudios de Casos y Controles , Granulocitos , Antígenos HLA/inmunología , Humanos , Factores Inmunológicos/sangre , Isoanticuerpos/sangre , Isoantígenos/inmunología , N-Formilmetionina Leucil-Fenilalanina/farmacología , Neutrófilos/efectos de los fármacos , Estudios Prospectivos , Síndrome de Dificultad Respiratoria/epidemiología , Medición de Riesgo
19.
Am J Physiol Cell Physiol ; 283(6): C1592-603, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12388071

RESUMEN

Lysophosphatidylcholines (lyso-PCs), generated during blood storage, are etiologic in a two-insult, sepsis-based model of transfusion-related acute lung injury (TRALI). Individually, endotoxin (LPS) and lyso-PCs prime but do not activate neutrophils (PMNs). We hypothesized that priming of PMNs alters their reactivity such that a second priming agent causes PMN activation and endothelial cell damage. PMNs were primed or not with LPS and then treated with lyso-PCs, and oxidase activation and elastase release were measured. For coculture experiments, activation of human pulmonary microvascular endothelial cells (HMVECs) was assessed by ICAM-1 expression and chemokine release. HMVECs were stimulated or not with LPS, PMNs were added, cells were incubated with lyso-PCs, and the number of viable HMVECs was counted. Lyso-PCs activated LPS-primed PMNs. HMVEC activation resulted in increased ICAM-1 and release of ENA-78, GRO alpha, and IL-8. PMN-mediated HMVEC damage was dependent on LPS activation of HMVECs, chemokine release, PMN adhesion, and lyso-PC activation of the oxidase. In conclusion, sequential exposure of PMNs to priming agents activates the microbicidal arsenal, and PMN-mediated HMVEC damage was the result of two insults: HMVEC activation and PMN oxidase assembly.


Asunto(s)
Quimiocinas CXC , Endotelio Vascular/patología , Endotelio Vascular/fisiopatología , Interleucina-8/análogos & derivados , Neutrófilos/fisiología , Circulación Pulmonar , Adhesión Celular , Recuento de Células , Células Cultivadas , Quimiocina CXCL1 , Quimiocina CXCL5 , Quimiocinas/metabolismo , Factores Quimiotácticos/metabolismo , Técnicas de Cocultivo , Activación Enzimática , Humanos , Molécula 1 de Adhesión Intercelular/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Interleucina-8/metabolismo , Lipopolisacáridos/farmacología , Lisofosfatidilcolinas/farmacología , Microcirculación , Activación Neutrófila , Neutrófilos/efectos de los fármacos , Oxidorreductasas/metabolismo , Elastasa Pancreática/metabolismo
20.
Am J Physiol Cell Physiol ; 283(4): C1102-13, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12225974

RESUMEN

Secretory phospholipase A(2) (sPLA(2)) produces lipids that stimulate polymorphonuclear neutrophils (PMNs). With the discovery of sPLA(2) receptors (sPLA(2)-R), we hypothesize that sPLA(2) stimulates PMNs through a receptor. Scatchard analysis was used to determine the presence of a sPLA(2) ligand. Lysates were probed with an antibody to the M-type sPLA(2)-R, and the immunoreactivity was localized. PMNs were treated with active and inactive (+EGTA) sPLA(2) (1-100 units of enzyme activity/ml, types IA, IB, and IIA), and elastase release and PMN adhesion were measured. PMNs incubated with inactive, FITC-linked sPLA(2)-IB, but not sPLA(2)-IA, demonstrated the presence of a sPLA(2)-R with saturation at 2.77 fM and a K(d) of 167 pM. sPLA(2)-R immunoreactivity was present at 185 kDa and localized to the membrane. Inactive sPLA(2)-IB activated p38 MAPK, and p38 MAPK inhibition attenuated elastase release. Active sPLA(2)-IA caused elastase release, but inactive type IA did not. sPLA(2)-IB stimulated elastase release independent of activity; inactive sPLA(2)-IIA partially stimulated PMNs. sPLA(2)-IB and sPLA(2)-IIA caused PMN adhesion. We conclude that PMNs contain a membrane M-type sPLA(2)-R that activates p38 MAPK.


Asunto(s)
Elastasa de Leucocito/metabolismo , Neutrófilos/metabolismo , Receptores de Superficie Celular/fisiología , Unión Competitiva/efectos de los fármacos , Calcio/metabolismo , Adhesión Celular/fisiología , Membrana Celular/metabolismo , Separación Celular , Quelantes/metabolismo , Quelantes/farmacología , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Citometría de Flujo , Colorantes Fluorescentes , Fosfolipasas A2 Grupo IB , Fosfolipasas A2 Grupo II , Humanos , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Neutrófilos/efectos de los fármacos , Fosfolipasas A/antagonistas & inhibidores , Fosfolipasas A/farmacología , Receptores de Fosfolipasa A2 , Proteínas Quinasas p38 Activadas por Mitógenos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...