Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Artículo en Inglés | MEDLINE | ID: mdl-37683721

RESUMEN

BACKGROUND: Factor XII (FXII) is a multifunctional protease capable of activating thrombotic and inflammatory pathways. FXII has been linked to thrombosis in extracorporeal membrane oxygenation (ECMO), but the role of FXII in ECMO-induced inflammatory complications has not been studied. We used novel gene-targeted FXII- deficient rats to evaluate the role of FXII in ECMO-induced thromboinflammation. METHODS: FXII-deficient (FXII-/-) Sprague-Dawley rats were generated using CRISPR/Cas9. A minimally invasive venoarterial (VA) ECMO model was used to compare wild-type (WT) and FXII-/- rats in 2 separate experimental cohorts: rats placed on ECMO without pharmacologic anticoagulation and rats anticoagulated with argatroban. Rats were maintained on ECMO for 1 hour or until circuit failure occurred. Comparisons were made with unchallenged rats and rats that underwent a sham surgical procedure without ECMO. RESULTS: FXII-/- rats were maintained on ECMO without pharmacologic anticoagulation with low resistance throughout the 1-hour experiment. In contrast, WT rats placed on ECMO without anticoagulation developed thrombotic circuit failure within 10 minutes. Argatroban provided a means to maintain WT and FXII-/- rats on ECMO for the 1-hour time frame without thrombotic complications. Analyses of these rats demonstrated that ECMO resulted in increased neutrophil migration into the liver that was significantly blunted by FXII deficiency. ECMO also resulted in increases in high molecular weight kininogen cleavage and complement activation that were abrogated by genetic deletion of FXII. CONCLUSIONS: FXII initiates hemostatic system activation and key inflammatory sequelae in ECMO, suggesting that therapies targeting FXII could limit both thromboembolism and inopportune inflammatory complications in this setting.

2.
Blood Adv ; 5(22): 4741-4751, 2021 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-34597365

RESUMEN

Extracellular vesicles (EV) have been implicated in diverse biological processes, including intracellular communication, transport of nucleic acids, and regulation of vascular function. Levels of EVs are elevated in cancer, and studies suggest that EV may stimulate thrombosis in patients with cancer through expression of tissue factor. However, limited data also implicate EV in the activation of the contact pathway of coagulation through activation of factor XII (FXII) to FXIIa. To better define the ability of EV to initiate contact activation, we compared the ability of EV derived from different cancer cell lines to activate FXII. EV from all cell lines activated FXII, with those derived from pancreatic and lung cancer cell lines demonstrating the most potent activity. Concordant with the activation of FXII, EV induced the cleavage of high molecular weight kininogen (HK) to cleaved kininogen. We also observed that EVs from patients with cancer stimulated FXII activation and HK cleavage. To define the mechanisms of FXII activation by EV, EV were treated with calf intestinal alkaline phosphatase or Escherichia coli exopolyphosphatase to degrade polyphosphate; this treatment blocked binding of FXII to EVs and the ability of EV to mediate FXII activation. In vivo, EV induced pulmonary thrombosis in wild-type mice, with protection conferred by a deficiency in FXII, HK, or prekallikrein. Moreover, pretreatment of EVs with calf intestinal alkaline phosphatase inhibited their prothrombotic effect. These results indicate that polyphosphate mediates the binding of contact factors to EV and that EV-associated polyphosphate may contribute to the prothrombotic effects of EV in cancer.


Asunto(s)
Vesículas Extracelulares , Neoplasias , Animales , Factor XII , Factor XIIa , Humanos , Ratones , Polifosfatos , Precalicreína
3.
Kidney Int ; 98(4): 897-905, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32763116

RESUMEN

Allogeneic transplants elicit dynamic T cell responses that are modulated by positive and negative co-stimulatory receptors. Understanding mechanisms that intrinsically modulate the immune responses to transplants is vital to develop rational treatment for rejection. Here, we have investigated the impact of programed cell death-1 (PD-1) protein, a negative co-stimulatory receptor, on the rejection of MHC incompatible kidney transplants in mice. T cells were found to rapidly infiltrate the kidneys of A/J mice transplanted to C57BL/6 mice, which peaked at six days and decline by day 14. The T cells primarily encircled tubules with limited infiltration of the tubular epithelium. Lipocalin 2 (LCN2), a marker of tubular injury, also peaked in the urine at day six and then declined. Notably, flow cytometry demonstrated that most of the T cells expressed PD-1 (over 90% of CD8 and about 75% of CD4 cells) at day six. Administration of blocking antibody to PD-L1, the ligand for PD-1, before day six increased T cell infiltrates and urinary LCN2, causing terminal acute rejection. In contrast, blocking PD-1/PD-L1 interactions after day six caused only a transient increase in urinary LCN2. Depleting CD4 and CD8 T cells virtually eliminated LCN2 in the urine in support of T cells injuring tubules. Thus, our data indicate that PD-1/PD-L1 interactions are not just related to chronic antigenic stimulation of T cells but are critical for the regulation of acute T cell responses to renal transplants.


Asunto(s)
Trasplante de Riñón , Receptor de Muerte Celular Programada 1 , Animales , Antígeno B7-H1 , Linfocitos T CD8-positivos , Muerte Celular , Ligandos , Ratones , Ratones Endogámicos C57BL
4.
Biochem Biophys Res Commun ; 495(2): 1775-1781, 2018 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-29229391

RESUMEN

Clusterin is a multifunctional glycoprotein that plays important roles and is up-regulated in liver diseases such as hepatitis and hepatocellular carcinoma. However, little is known about the significance of clusterin in the pathogenesis of non-alcoholic steatohepatitis (NASH). The aim of this study is to examine the role of clusterin in progression of steatohepatitis in mice fed a methionine and choline deficient (MCD) diet. We generated hepatocyte-specific clusterin overexpression (hCLU-tg) mice, and hCLU-tg mice showed lower levels of hepatic triglycerides, less infiltration of macrophages and reduction of TNF-α, activation of Nrf-2 than wild-type littermates fed the MCD diet. Also, sustained clusterin expression in liver ameliorated hepatic fibrogenesis by reducing the activation of hepatic stellate cells by MCD diet. Sustained expression of clusterin in liver functioned as a preconditioning stimulus and prevented MCD diet-induced severe steatohepatitis injury via Nrf2 activation. These results demonstrate a novel function of clusterin as an immune preconditioning regulator in various inflammatory diseases including steatohepatitis.


Asunto(s)
Clusterina/metabolismo , Hepatocitos/metabolismo , Enfermedad del Hígado Graso no Alcohólico/prevención & control , Animales , Deficiencia de Colina/complicaciones , Deficiencia de Colina/metabolismo , Clusterina/genética , Dieta/efectos adversos , Modelos Animales de Enfermedad , Hígado/metabolismo , Hígado/patología , Masculino , Metionina/deficiencia , Ratones , Ratones Transgénicos , Factor 2 Relacionado con NF-E2/metabolismo , Enfermedad del Hígado Graso no Alcohólico/etiología , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Estrés Oxidativo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Regulación hacia Arriba
5.
Biochem Biophys Res Commun ; 482(4): 1407-1412, 2017 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-27965092

RESUMEN

Clusterin is a secretory glycoprotein that is up-regulated in areas of inflammation and under increased levels of oxidative stress. Previously, we demonstrated that clusterin activates NF-κB, and up-regulates the expression of MMP-9 and TNF-α. In this research, we extend our previous findings by reporting that such clusterin-induced macrophage response is mediated via TLR4 signaling. Specifically, we found that TNF-α induced by clusterin was significantly abrogated by pretreatment of TLR4-signaling inhibitors and anti-TLR4 neutralizing antibody. Additionally, a primary culture of macrophages derived from TLR4-signal defective and knockout mice were unresponsive to clusterin, resulting in no TNF-α secretion, whereas macrophages carrying wild-type TLR4 responded to clusterin and induced TNF-α. Moreover, clusterin increased NF-κB promoter activity in HEK-Blue hTLR4 cells, but not in HEK-Blue Null2 cells. To confirm that clusterin elicits TLR4 signal transduction, recombinant clusterin was generated and purified from cell culture. Interestingly, we found that the recombinant clusterin with C-terminal HA-tag induces TNF-α secretion at a significantly lower level compared to an intact form of clusterin without C-terminal HA-tag. Removal of HA-tag from the recombinant clusterin restored its activity, suggesting that C-terminal HA-tag partially masks the domain involved in TLR4 signaling. Furthermore, clusterin enhanced TLR4 mobilization into lipid raft of plasma membrane, and TNF-α and MMP-9 secretion stimulated by clusterin was diminished by pretreatment with methyl-ß-cyclodextrin (MßCD), which was used to disrupt lipid raft. In conclusion, clusterin-induced TNF-α and MMP-9 up-regulation is most likely mediated via TLR4 recruitment into lipid rafts, and these data describe a novel role of clusterin as an endogenous regulator for TLR4 signaling.


Asunto(s)
Clusterina/metabolismo , Macrófagos/metabolismo , Transducción de Señal , Receptor Toll-Like 4/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Humanos , Inflamación , Macrófagos/citología , Masculino , Metaloproteinasa 9 de la Matriz/metabolismo , Microdominios de Membrana/química , Ratones , Ratones Endogámicos C3H , Ratones Noqueados , FN-kappa B/metabolismo , Estrés Oxidativo , Dominios Proteicos , Células RAW 264.7
6.
Biochem Biophys Res Commun ; 445(3): 645-50, 2014 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-24569077

RESUMEN

Clusterin induces the expression of various chemotactic cytokines including tumor necrosis factor-α (TNF-α) in macrophages and is involved in the cell migration. According to the results of this study, clusterin induced the directional migration (chemotaxis) of macrophages based on a checkerboard analysis. The chemotactic activity of clusterin was prevented by pretreatment with pertussis toxin (PTX), indicating that the Gαi/o-protein coupled receptor (GPCR) was involved in the chemotactic response of clusterin. Clusterin-stimulated chemotaxis was abrogated in a dose-dependent manner by pretreatment with gallein (a Gßγ inhibitor), indicating the involvement of Gßγ released from the GPCR. In addition, inhibitors of phospholipase C (PLC, U73122) and phosphoinositide 3-kinase (PI3K, LY294002), the key targets of Gßγ binding and activation, suppressed chemotactic migration by clusterin. The phosphorylation of Akt induced by clusterin was blocked by pretreatment with gallein or LY294002 but not with U73122, indicating that Gßγ released from the PTX-sensitive Gi protein complex activated PLC and PI3K/Akt signaling pathways separately. The activation of cellular MAP kinases was essential in that their inhibitors blocked clusterin-induced chemotaxis, and Gßγ was required for the activation of MAP kinases because gallein reduced their phosphorylations induced by clusterin. In addition, the inflammation-induced migration of macrophages was greatly reduced in clusterin-deficient mice based on a thioglycollate-induced peritonitis model system. These results suggest that clusterin stimulates the chemotactic migration of macrophages through a PTX-sensitive GPCR and Gßγ-dependent pathways and describe a novel role of clusterin as a chemoattractant of monocytes/macrophages, suggesting that clusterin may serve as a molecular bridge between inflammation and its remodeling of related tissue by recruiting immune cells.


Asunto(s)
Quimiotaxis , Clusterina/metabolismo , Macrófagos/citología , Receptores Acoplados a Proteínas G/metabolismo , Animales , Quimiotaxis/efectos de los fármacos , Humanos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Toxina del Pertussis/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Transducción de Señal , Fosfolipasas de Tipo C/antagonistas & inhibidores , Fosfolipasas de Tipo C/metabolismo
7.
Biochem Biophys Res Commun ; 422(1): 200-5, 2012 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-22575505

RESUMEN

Tumor associated macrophages are known to be closely linked with tumor progression and metastasis. On the other hand, clusterin is overexpressed in several tumor types and regarded as a putative tumor-promoting factor due to this overexpression and the subsequent induction of chemoresistance. In our previous study, clusterin was found to induce the expression of matrix metalloproteinase-9 (MMP-9) in macrophages, and MMP-9 is known to be essential for tumor cell migration and invasion via basement membrane breakdown. Because paracrine interactions between tumor cells and surrounding macrophages regulate metastasis, these findings raise the possibility that clusterin promotes the secretion of cytokines in macrophages in addition to MMP-9. Here, we demonstrate that clusterin upregulates the expressions of chemotactic cytokines, that is, monocyte chemotactic protein-1 (MCP-1), macrophage inflammatory protein-1ß (MIP-1ß), regulated upon activation, normal T cell expressed and secreted (RANTES), and tumor necrosis factor-α (TNF-α) in Raw264.7 macrophages. In particular, clusterin stimulated TNF-α secretion via the activations of ERK, JNK, and PI3K/Akt pathways in a time and dose-dependent manner. Furthermore, clusterin-induced TNF-α secretion was found to play a critical role in the chemotactic migration of Raw264.7 macrophages. It was also found that clusterin acts directly as a chemoattractant for macrophages. Together, these results suggest that clusterin stimulates the expression and secretion of TNF-α, which plays a critical role in promoting macrophage chemotaxis, via ERK, JNK, and PI3K/Akt pathways. Collectively, these findings describe a novel function for clusterin as an inducer of TNF-α in macrophages and their chemotactic migration, and suggest that clusterin has a tumor-promoting effect.


Asunto(s)
Quimiotaxis/fisiología , Clusterina/fisiología , Macrófagos Peritoneales/fisiología , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Línea Celular , Quimiotaxis/efectos de los fármacos , Clusterina/farmacología , MAP Quinasa Quinasa 4/biosíntesis , Sistema de Señalización de MAP Quinasas , Macrófagos Peritoneales/efectos de los fármacos , Macrófagos Peritoneales/metabolismo , Ratones , Neoplasias/metabolismo , Neoplasias/patología , Fosfatidilinositol 3-Quinasas/biosíntesis , Proteínas Proto-Oncogénicas c-akt/biosíntesis , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/genética
8.
Biochem Biophys Res Commun ; 420(4): 851-6, 2012 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-22465014

RESUMEN

Clusterin is a disulfide-linked heterodimeric glycoprotein that has been implicated in a variety of biological processes. Its expression has been shown to be elevated during cellular senescence and normal aging, but it is uncertain whether clusterin protects against aging or whether its expression is a consequence of aging. To investigate the functions of clusterin during organismal aging, we established transgenic Drosophila alleles to induce the expression of the secretory form of human clusterin (hClu(S)) using the Gal4/UAS system. hClu(S) protein (~60 kDa) was detected in both adult homogenates and larval hemolymphs of flies ubiquitously overexpressing hClu(S) (da-Gal4>UAS-hClu(S)) and in motoneurons (D42-Gal4>UAS-hClu(S)). Interestingly, the mean lifespans of these hClu(S)-overexpressing flies were significantly greater than those of control flies that exhibited no hClu(S) induction. hClu(S)-overexpressing flies also showed significantly greater tolerance to heat shock, wet starvation, and oxidative stress. Furthermore, amounts of reactive oxygen species (ROS) in whole bodies were significantly lower in hClu(S)-overexpressing flies. In addition, clusterin was found to prevent the inactivation of glutamine synthetase (GS) by metal-catalyzed oxidation (MCO) in vitro, and this protection was only supported by thiol-reducing equivalents, such as, DTT or GSH, and not by ascorbate (a non-thiol MCO system). Furthermore, this protection against GS inactivation by clusterin was abolished by reacting clusterin with N-ethylmaleimide, a sulfhydryl group-modifying agent. Taken together, these results suggest that a disulfide-linked form of clusterin functions as an antioxidant protein via its cysteine sulfhydryl groups to reduce ROS levels and delay the organismal aging in fruit flies.


Asunto(s)
Clusterina/fisiología , Drosophila melanogaster/fisiología , Respuesta al Choque Térmico/genética , Longevidad/genética , Estrés Oxidativo/genética , Animales , Animales Modificados Genéticamente/genética , Animales Modificados Genéticamente/fisiología , Clusterina/genética , Ditiotreitol/farmacología , Proteínas de Drosophila , Drosophila melanogaster/efectos de los fármacos , Drosophila melanogaster/genética , Etilmaleimida/farmacología , Glutamato-Amoníaco Ligasa , Hemolinfa/metabolismo , Humanos , Longevidad/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo
9.
J Leukoc Biol ; 90(4): 761-9, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21742938

RESUMEN

Most solid tumor tissues possess a significant population of macrophages, which are known to be closely linked with tumor progression and metastasis. Clusterin has been reported to be overexpressed in various tumors and to have a tumor-promoting role. As clusterin induction and macrophage infiltration occur concurrently at the tumor site, it raises a possibility that clusterin may regulate the function of macrophages via facilitating ECM remodeling. Here, we demonstrate for the first time the expression of MMP-9 by clusterin in human primary monocytes as well as human and murine macrophage cell lines, THP-1, and Raw264.7. MMP-9 expression was accompanied by increased enzymatic activity, as revealed by gelatin zymography. The MMP-9 activity promoted by clusterin was found to be dependent on the activation of ERK1/2 and PI3K/Akt but not p38 or JNK pathways. Inhibition of PI3K activity did not affect the activation of ERK1/2 and vice versa, indicating that the two pathways were independently operated to stimulate MMP-9 activity. Moreover, clusterin facilitated nuclear translocation of NF-κB p65 along with IκB-α degradation and phosphorylation, which was critical for MMP-9 expression. As NF-κB is a central regulator of inflammation, clusterin may provide a molecular link between inflammation and cancer via up-regulating NF-κB and MMP-9. Collectively, these data highlight a novel role of clusterin as a stimulator for MMP-9 expression in macrophages, which may contribute to the tissue reorganization by serving as a modulator for ECM degradation.


Asunto(s)
Clusterina/metabolismo , Macrófagos/metabolismo , Metaloproteinasa 9 de la Matriz/biosíntesis , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Monocitos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factor de Transcripción ReIA/metabolismo , Animales , Línea Celular , Clusterina/farmacología , Inducción Enzimática , Humanos , MAP Quinasa Quinasa 4/metabolismo , Ratones , Neoplasias/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
10.
Dev Dyn ; 240(3): 605-15, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21290478

RESUMEN

Based on our previous observations that clusterin induction accompanies pancreas regeneration in the rat, we sought to determine if regeneration might be impaired in mice that lacked clusterin. We studied the impact of absent clusterin on morphogenic and functional features of regenerating pancreas. Clusterin induction was accompanied in the regenerating pancreas by a robust development of new lobules with ductules, acini, and endocrine islets in wild type after partial pancreatectomy. In clusterin knock-out mice, however, pancreatectomy resulted in a poor formation of regenerating lobule. In particular, regeneration of beta-cells was also significantly reduced and was associated with persistent hyperglycemia. Duct cells obtained from pancreatectomized clusterin knock-out mice exhibited impaired beta-cell formation in vitro; this was restored by administration of exogenous clusterin. We suggest that clusterin plays a critical role to promote both exocrine and endocrine regeneration following pancreas injury, as well as for in vitro beta-cell regeneration.


Asunto(s)
Clusterina/metabolismo , Páncreas/metabolismo , Páncreas/fisiopatología , Regeneración/fisiología , Animales , Western Blotting , Clusterina/genética , Clusterina/farmacología , Prueba de Tolerancia a la Glucosa , Inmunohistoquímica , Técnicas In Vitro , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Páncreas/citología , Páncreas/cirugía , Pancreatectomía , Reacción en Cadena de la Polimerasa , Regeneración/genética
11.
J Leukoc Biol ; 88(5): 955-63, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20729304

RESUMEN

CLU is a secreted, multifunctional protein implicated in several immunologic and pathologic conditions. As the level of serum CLU was shown to be elevated during inflammatory responses, we questioned if CLU might interact with circulating lymphocytes leading to functional consequences. To assess this possibility directly, mouse splenocytes and purified NK cells were cultured with varying dose of CLU, and its effect on cell proliferation was examined. Our data showed that CLU up-regulated DNA synthesis and expansion of NK cells significantly in response to a suboptimal, but not maximal, dose of IL-2, and CLU alone did not exhibit such effects. This CLU-mediated synergy required the co-presence of CLU at the onset of IL-2 stimulation and needed a continuous presence during the rest of the culture. Importantly, NK cells stimulated with CLU showed increased formation of cell clusters and a CD69 activation receptor, representing a higher cellular activation status compared with those from the control group. Furthermore, these NK cells displayed elevated IFN-γ production upon RMA/S tumor target exposures, implying that CLU regulates not only NK cell expansion but also effector function of NK cells. Collectively, our data present a previously unrecognized function of CLU as a novel regulator of NK cells via providing costimulation required for cell proliferation and IFN-γ secretion. Therefore, the role of CLU on NK cells should be taken into consideration for the previously observed, diverse functions of CLU in chronic inflammatory and autoimmune conditions.


Asunto(s)
Clusterina/farmacología , Interferón gamma/genética , Interleucina-2/farmacología , Células Asesinas Naturales/inmunología , Animales , Western Blotting , División Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Citometría de Flujo , Humanos , Células Asesinas Naturales/citología , Células Asesinas Naturales/efectos de los fármacos , Ratones , Bazo/efectos de los fármacos , Bazo/inmunología
12.
Int J Toxicol ; 28(3): 151-61, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19546254

RESUMEN

2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) has been associated with diabetes in several epidemiological studies. However, the diabetogenic action of TCDD on pancreatic cells is unclear. Here, we investigated the direct toxic effects of TCDD on a rat insulin-secreting beta cell line. We found that TCDD enhances exocytosis of MTT formazan and lysosomal proteins such as beta-hexosaminindase and Lamp-1. This TCDD-induced exocytosis was abrogated by T-type calcium channel blockers (mibefradil, flunarizine) but not by an aryl hydrocarbon receptor antagonist (alpha-naphtoflavone). Indeed, cytosolic calcium levels were increased by TCDD. Furthermore, TCDD stimulated insulin secretion, which was inhibited by flunarizine. Taken together, our results suggest that TCDD-induced calcium influx via T-type channels regulates vesicular trafficking, such as lysosomal and secretory granule exocytosis, and that TCDD might exert adverse effects on beta cells by continuous insulin release followed by beta cell exhaustion. This could contribute to the link between TCDD exposure and the risk of developing diabetes.


Asunto(s)
Canales de Calcio Tipo T/metabolismo , Calcio/metabolismo , Contaminantes Ambientales/toxicidad , Exocitosis/efectos de los fármacos , Células Secretoras de Insulina/efectos de los fármacos , Insulina/metabolismo , Dibenzodioxinas Policloradas/toxicidad , Animales , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Humanos , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Lisosomas/efectos de los fármacos , Lisosomas/enzimología , Ratas , Receptores de Hidrocarburo de Aril/antagonistas & inhibidores , Vías Secretoras/efectos de los fármacos
13.
Neuroreport ; 20(4): 435-9, 2009 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-19218870

RESUMEN

We previously reported that clusterin enhances astrocyte proliferation and extracellular signal-regulated kinase (ERK) activity. It, however, remains largely unknown how clusterin promotes cell growth. Here, we investigate the signaling pathway and related molecules underlying astrocyte proliferation by clusterin. Exogenous clusterin stimulates Ras-dependent Raf-1/mitogen-activated protein kinase kinase (MEK)/ERK activation. Clusterin-induced astrocyte proliferation and ERK1/2 phosphorylation were abrogated by either AG1478 (an inhibitor of epidermal growth factor receptor, EGFR) or EGFR small interfering RNA. Furthermore, clusterin treatment provoked tyrosine phosphorylation of EGFR (pY(1173)), which was also blocked by AG1478. These results suggest that clusterin requires EGFR activation to deliver its mitogenic signal through the Ras/Raf-1/MEK/ERK signaling cascade in astrocytes.


Asunto(s)
Astrocitos/fisiología , Proliferación Celular/efectos de los fármacos , Clusterina/farmacología , Receptores ErbB/metabolismo , Animales , Células Cultivadas , Clusterina/fisiología , Activación Enzimática/efectos de los fármacos , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Humanos , Proteína Quinasa 3 Activada por Mitógenos/efectos de los fármacos , Quinasas de Proteína Quinasa Activadas por Mitógenos/efectos de los fármacos , Mitógenos/farmacología , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-raf/efectos de los fármacos , Quinazolinas , ARN Interferente Pequeño/farmacología , Ratas , Transducción de Señal , Tirfostinos/farmacología
14.
Mol Cells ; 25(2): 305-11, 2008 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-18414006

RESUMEN

After successful clinical application, arginine deiminase (ADI) has been proposed to be a new cancer therapeutic. In the present study, we examined the effect of ADI in combination with ionizing radiation (IR) on MCF-7 cell growth and clonogenic cell death. Cell growth was inhibited by IR in a dose-dependent manner and ADI enhanced the radiosensitivity. ADI itself did not suppress the growth of MCF-7 cells due to the high level of expression of argininosuccinate synthetase (ASS), which convert citrulline, a product of arginine degradation by ADI, to arginine. Previously, it was suggested that ammonia, another product of arginine degradation by ADI, is the main cause of the growth inhibition of irradiated hepatoma cells contaminated with ADI-expressing mycoplasma [van Rijn et al. (2003)]. However, we found that ammonia is not the only factor that enhances radiosensitivity, as enhancement was also observed in the absence of ammonia. In order to identify the enhancing effect, levels of ASS and proteins related to the cell cycle were examined. ASS was unchanged by ADI plus IR, but p21 (a CDK inhibitor) was upregulated and c-Myc downregulated. These findings indicate that changes in the expressions of cell cycle proteins are involved in the enhancement of radiosensitivity by ADI. We suggest that ADI is a potential adjunct to cancer therapy.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Hidrolasas/farmacología , Mycoplasma/enzimología , Tolerancia a Radiación/efectos de los fármacos , Amoníaco/farmacología , Argininosuccinato Sintasa/genética , Argininosuccinato Sintasa/metabolismo , Línea Celular Tumoral , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Humanos , Proteínas Proto-Oncogénicas c-myc/genética , Tolerancia a Radiación/efectos de la radiación , Radiación Ionizante
15.
Orthopedics ; 31(8): 804, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19292415

RESUMEN

The use of BIOLOX delta ceramic (CeramTec AG, Plochingen, Germany) has been increasing. This ceramic prevents cracking by restraining the phase transformation due to the insertion of nano-sized, yttria-stabilized tetragonal zirconia into the alumina matrix. This restrains the progress of cracking through the formation of platelet-like crystal or whiskers due to the addition of an oxide additive. We observed a case of BIOLOX delta ceramic liner (CeramTec AG) rim fracture 4 months postoperatively. Radiographs showed that the ceramic liner was subluxated from the acetabular cup. Scratches on the acetabular cup and femoral neck were seen, and the fracture was visible on the rim of the liner. Under electron microscope, metal particle coatings from the ceramic liner were identified. The ceramic liner, fracture fragments, and adjacent tissues were removed and replaced with a ceramic liner and femoral head of the same size and design. We believe the mechanism of the fracture-dissociation of the ceramic liner in this case is similar to a case of separation of the ceramic liner from the polyethylene shell in a sandwich-type ceramic-ceramic joint. To prevent ceramic liner fracture-dissociation, the diameter of the femoral neck needs to be decreased in a new design, while the diameter of the femoral head needs to be increased to ensure an increase in range of motion.


Asunto(s)
Artroplastia de Reemplazo de Rodilla/efectos adversos , Necrosis de la Cabeza Femoral/cirugía , Luxación de la Cadera/etiología , Luxación de la Cadera/cirugía , Prótesis de la Rodilla/efectos adversos , Falla de Prótesis , Humanos , Masculino , Persona de Mediana Edad , Resultado del Tratamiento
16.
Int J Cancer ; 120(4): 897-905, 2007 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-17096330

RESUMEN

Recently, pegylated arginine deiminase (ADI; EC 3.5.3.6) has been used to treat the patients with hepatocellular carcinoma or melanoma, in which the level of argininosuccinate synthetase (ASS) activity is low or undetectable. The efficacy of its antitumor activity largely depends on the level of intracellular ASS, which enables tumor cells to recycle citrulline to arginine. Thus, we examined the expression levels of ASS in various cancer cells and found that it is low in renal cell carcinoma (RCC) cells, rendering the cells highly sensitive to arginine deprivation by ADI treatment. Immunohistochemical analysis revealed that in biopsy specimens from RCC patients (n = 98), the expression of ASS is highly demonstrated in the epithelium of normal proximal tubule but not seen in tumor cells. Furthermore, RCC cells treated with ADI showed remarkable growth retardation in a dose dependent manner. ADI also exerted in vivo antiproliferative effect on the allografted renal cell carcinoma (RENCA) tumor cells and prolonged the survival of tumor-bearing mice. Histological examination of the tumors revealed that tumor angiogenesis and vascular endothelial growth factor (VEGF) expression were significantly diminished by ADI administration. Therefore, these findings suggest that arginine deprivation by ADI could provide a beneficial strategy for the treatment of RCC in ways of inhibitions of arginine availability and neovascularization.


Asunto(s)
Arginasa/farmacología , Arginina/deficiencia , Argininosuccinato Sintasa/metabolismo , Carcinoma de Células Renales/enzimología , Hidrolasas/metabolismo , Neoplasias Renales/enzimología , Neovascularización Patológica/tratamiento farmacológico , Adulto , Anciano , Animales , Arginasa/química , Carcinoma de Células Renales/patología , Procesos de Crecimiento Celular/efectos de los fármacos , Citrulina/metabolismo , Femenino , Humanos , Neoplasias Renales/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Persona de Mediana Edad , Polietilenglicoles/farmacología , Tasa de Supervivencia , Células Tumorales Cultivadas , Factor A de Crecimiento Endotelial Vascular/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Neuroreport ; 17(18): 1871-5, 2006 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-17179861

RESUMEN

Clusterin, a secretory glycoprotein, has been shown to be up-regulated in the reactive astrocytes in response to brain injury and neurodegenerative diseases, but its function has not been clearly elucidated. In this study, we investigate whether clusterin has growth-stimulatory activity in astrocytes. Suppression of clusterin with antisense oligonucleotide induced growth arrest, whereas transient overexpression of clusterin by cDNA transfection or exogenous treatment with purified clusterin promoted proliferation of the primary astrocytes in culture. This clusterin-stimulated proliferation was abrogated by PD98059, an inhibitor of mitogen-activated protein kinase kinase. These results suggest that clusterin might play an important role in astrogliosis by stimulating the proliferation of astrocytes through activation of the extracellular signal-regulated kinase 1/2 signaling pathway.


Asunto(s)
Astrocitos/fisiología , Proliferación Celular , Clusterina/fisiología , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Animales , Animales Recién Nacidos , Astrocitos/efectos de los fármacos , Western Blotting/métodos , Recuento de Células/métodos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Corteza Cerebral/citología , Clusterina/química , Clusterina/farmacología , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Inhibidores Enzimáticos/farmacología , Flavonoides/farmacología , Citometría de Flujo/métodos , Expresión Génica/efectos de los fármacos , Humanos , Masculino , Oligodesoxirribonucleótidos Antisentido/farmacología , Ratas , Ratas Sprague-Dawley , Timidina/metabolismo , Transfección , Tritio/metabolismo
18.
Exp Cell Res ; 309(2): 305-15, 2005 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-16038898

RESUMEN

Clusterin has been known as a chaperone-like molecule capable of interacting with various proteins. In this study, we show that clusterin interacts with the microtubule-destabilizing stathmin family protein SCLIP by GST pull-down and co-immunoprecipitation assays. Interestingly, SCLIP interacts with 80 kDa mature form of clusterin in the cytosolic fraction of PC12 cells permeabilized by low concentration of a weak nonionic detergent digitonin, but not with intracellular variants of clusterin known as binding isoforms of Ku70 or TGF-beta receptors. Both clusterin and SCLIP are co-localized at the perinuclear region and growth cone of PC12 cells. In addition, we show that the minimal domains for the interaction are mapped to the C-terminal valine-rich region (367-447) of clusterin and the N-terminal palmitoylation and membrane attachment site (1-34) of SCLIP. Finally, we demonstrate that ectopic expression of clusterin in PC12 cells elongates neurite-formation triggered by NGF and induces spontaneous neurite outgrowth even in the absence of NGF. Taken together, these results suggest that the clusterin interacts with SCLIP and the interaction may act as an important modulator during neuronal differentiation.


Asunto(s)
Diferenciación Celular/fisiología , Canales Iónicos/metabolismo , Proteínas de la Membrana/metabolismo , Factores de Crecimiento Nervioso/metabolismo , Neuritas/fisiología , Animales , Citoplasma/metabolismo , Neuronas/citología , Neuronas/fisiología , Células PC12 , Estructura Terciaria de Proteína , Ratas , Canales Catiónicos TRPM
19.
J Ethnopharmacol ; 85(2-3): 283-7, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12639753

RESUMEN

The present study examined the inhibitory effect of aqueous extract from the gall of Rhus chinensis (AEGRC) on alpha-glucosidase activity, an enzyme responsible for digestion of carbohydrate to monosaccharides in the process of intestinal absorption. AEGRC inhibited Bacillus alpha-glucosidase acitvity with an IC(50) of 0.9 micro g/ml. Its inhibition on alpha-glucosidase was determined to be noncompetitive and reversible when the enzyme-substrate mixture was simultaneously treated with AEGRC as an inhibitor. In addition, when it was orally administered to rats with sucrose (2g/kg), AEGRC (250-1000mg/kg) significantly suppressed the increase of blood glucose levels after sucrose loading in a dose dependent manner. These results suggest that AEGRC might exert anti-diabetic effect by suppressing carbohydrate absorption from intestine, and thereby reducing the postprandial increase of blood glucose.


Asunto(s)
Glucemia/metabolismo , Inhibidores Enzimáticos/farmacología , Inhibidores de Glicósido Hidrolasas , Periodo Posprandial/efectos de los fármacos , Rhus/química , Animales , Diálisis , Disacaridasas/metabolismo , Mucosa Intestinal/metabolismo , Intestinos/efectos de los fármacos , Cinética , Masculino , Extractos Vegetales/farmacología , Periodo Posprandial/fisiología , Ratas , Ratas Sprague-Dawley , Sacarosa/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA