Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cancers (Basel) ; 13(21)2021 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-34771464

RESUMEN

Breast implant-associated anaplastic large-cell lymphoma (BIA-ALCL) is a distinct malignancy associated with textured breast implants. We investigated whether bacteria could trigger the activation and multiplication of BIA-ALCL cells in vitro. BIA-ALCL patient-derived BIA-ALCL tumor cells, BIA-ALCL cell lines, cutaneous ALCL cell lines, an immortal T-cell line (MT-4), and peripheral blood mononuclear cells (PBMC) from BIA-ALCL, capsular contracture, and primary augmentation patients were studied. Cells were subjected to various mitogenic stimulation assays including plant phytohemagglutinin (PHA), Gram-negative bacterial lipopolysaccharide (LPS), Staphylococcal superantigens enterotoxin A (SEA), toxic shock syndrome toxin-1 (TSST-1), or sterilized implant shells. Patient-derived BIA-ALCL tumor cells and BIA-ALCL cell lines showed a unique response to LPS stimulation. This response was dampened significantly in the presence of a Toll-like receptor 4 (TLR4) inhibitor peptide. In contrast, cutaneous ALCL cells, MT-4, and PBMC cells from all patients responded significantly more to PHA, SEA, and TSST-1 than to LPS. Breast implant shells of all surface grades alone did not produce a proliferative response of BIA-ALCL cells, indicating the breast implant does not act as a pro-inflammatory stimulant. These findings indicate a possible novel pathway for LPS to promote BIA-ALCL cell proliferation via a TLR4 receptor-mediated bacterial transformation of T-cells into malignancy.

3.
Prostate Cancer ; 2015: 285193, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25802762

RESUMEN

Background. New nontoxic targeted approaches are needed for patients with castrate resistant prostate cancer (CRPC). Our preclinical studies show that activated T cells (ATC) armed with anti-CD3 x anti-Her2 bispecific antibody (Her2Bi) kill prostate cancer cells lines, induce a Th1 cytokine pattern upon engagement of tumor cells, prevent the development of prostate tumors, and retard tumor growth in immunodeficient mice. These studies provided strong rationale for our phase I dose-escalation pilot study to test ATC armed with Her2Bi (aATC) for safety in men with CRPC. Methods. Seven of 8 men with CRPC were evaluable after receiving two infusions per week for 4 weeks. The men received 2.5, 5 or 10 × 10(9) aATC per infusion with low dose interleukin-2 and granulocyte-macrophage colony stimulating factor. Results. There were no dose limiting toxicities, and there was 1 partial responder and 3 of 7 patients had significant decreases in their PSA levels and pain scores. Immune evaluations of peripheral blood mononuclear cells in 2 patients before and after immunotherapy showed increases in IFN-γ EliSpot responses and Th1 serum cytokines. Conclusions. These results provide a strong rationale for developing phase II trials to determine whether aATC are effective for treating CRPC.

4.
Clin Cancer Res ; 21(10): 2305-14, 2015 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-25688159

RESUMEN

PURPOSE: This study reports a phase I immunotherapy trial in 23 women with metastatic breast cancer consisting of eight infusions of anti-CD3 × anti-HER2 bispecific antibody (HER2Bi) armed anti-CD3-activated T cells (ATC) in combination with low-dose IL-2 and granulocyte-macrophage colony-stimulating factor to determine safety, maximum tolerated dose (MTD), technical feasibility, T-cell trafficking, immune responses, time to progression, and overall survival (OS). EXPERIMENTAL DESIGN: ATC were expanded from leukapheresis product using IL2 and anti-CD3 monoclonal antibody and armed with HER2Bi. In 3+3 dose escalation design, groups of 3 patients received 5, 10, 20, or 40 × 10(9) armed ATC (aATC) per infusion. RESULTS: There were no dose-limiting toxicities and the MTD was not defined. It was technically feasible to grow 160 × 10(9) ATC from a single leukapheresis. aATC persisted in the blood for weeks and trafficked to tumors. Infusions of aATC induced anti-breast cancer responses and increases in immunokines. At 14.5 weeks after enrollment, 13 of 22 (59.1%) evaluable patients had stable disease and 9 of 22 (40.9%) had progressive disease. The median OS was 36.2 months for all patients, 57.4 months for HER2 3+ patients, and 27.4 months for HER2 0-2+ patients. CONCLUSIONS: Targeting HER2(+) and HER2(-) tumors with aATC infusions induced antitumor responses, increases in Th1 cytokines, and IL12 serum levels that suggest that aATC infusions vaccinated patients against their own tumors. These results provide a strong rationale for conducting phase II trials.


Asunto(s)
Neoplasias de la Mama/terapia , Inmunoterapia , Linfocitos T/trasplante , Adulto , Anciano , Anticuerpos Biespecíficos/administración & dosificación , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/administración & dosificación , Humanos , Interleucina-2/administración & dosificación , Estimación de Kaplan-Meier , Dosis Máxima Tolerada , Persona de Mediana Edad , Estadificación de Neoplasias , Linfocitos T/inmunología
5.
Transfusion ; 49(8): 1569-79, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19413726

RESUMEN

BACKGROUND: Observational studies have reported that patients transfused with red blood cells (RBCs) have a worse clinical outcome than untransfused patients and that storage age of RBCs at the time of transfusion may be an independent predictor of this adverse clinical outcome. STUDY DESIGN AND METHODS: Eight RBC concentrates in additive solution were studied over an 8-week storage period. The RBC supernatant was ultracentrifuged to concentrate microvesicles (RCMVs). RCMVs were studied by flow cytometry to identify phosphatidylserine (PS)-expressing RCMVs and in a thromboelastograph (TEG) using a modified assay to detect a thrombin facilitation effect. RESULTS: For all products, the percentage of RCMVs that exhibited PS expression on Day 1 was 50 ± 13%, which increased with storage, and on Day 31 was 90 ± 4%. After 31 days, four of the eight products showed a thrombin facilitation effect as evidenced by a shortening of the TEG reaction (R) time of 1.3 ± 1.1 minutes, which persisted to Day 41. Data are the mean ± 1 SD. This TEG R shortening effect was neutralized by annexin V. No such effect was observed on, or before, Day 21. CONCLUSIONS: Some stored RBCs release RCMVs, which express PS and are capable of facilitating thrombin generation in vitro. This provides a possible mechanism by which stored RBCs could promote adverse thrombotic or inflammatory effects.


Asunto(s)
Eritrocitos/citología , Eritrocitos/metabolismo , Refrigeración , Trombina/biosíntesis , Femenino , Citometría de Flujo/métodos , Humanos , Masculino , Fosfatidilserinas/biosíntesis , Tromboelastografía/métodos , Factores de Tiempo
6.
J Cell Mol Med ; 13(9B): 3632-43, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18681908

RESUMEN

SIRT1 is a member of a highly conserved gene family (sirtuins) encoding nicotinamide adenine dinucleotide (NAD)(+)-dependent deacetylases, originally found to deacetylate histones leading to increased DNA stability and prolonged survival in yeast and higher organisms, including mammals. SIRT1 has been found to function as a deacetylase for numerous protein targets involved in various cellular pathways, including stress responses, apoptosis and axonal degeneration. However, the role of SIRT1 in ultraviolet (UV) signalling pathways remains unknown. Using cell culture and Western blot analysis in this study we found that SIRT1 is expressed in cultured human skin keratinocytes. Both UV radiation and H(2)O(2), two major inducers of skin cell damage, down-regulate SIRT1 in a time- and dose-dependent manner. We observed that reactive oxygen species-mediated JNK activation is involved in this SIRT1 down-regulation. SIRT1 activator, resveratrol, which has been considered as an important antioxidant, protects against UV- and H(2)O(2)-induced cell death, whereas SIRT inhibitors such as sirtinol and nicotinamide enhance cell death. Activation of SIRT1 negatively regulates UV- and H(2)O(2)-induced p53 acetylation, because nicotinamide and sirtinol as well as SIRT1 siRNA enhance UV- and H(2)O(2)-induced p53 acetylation, whereas SIRT1 activator resveratrol inhibits it. We also found that SIRT1 is involved in UV-induced AMP-activated protein kinase (AMPK) and downstream acetyl-CoA carboxylase (ACC), phosphofructose kinase-2 (PFK-2) phosphorylation. Collectively, our data provide new insights into understanding of the molecular mechanisms of UV-induced skin aging, suggesting that SIRT1 activators such as resveratrol could serve as new anti-skin aging agents.


Asunto(s)
Células Cultivadas/metabolismo , Regulación de la Expresión Génica , Peróxido de Hidrógeno/farmacología , MAP Quinasa Quinasa 4/metabolismo , Sirtuina 1/metabolismo , Piel/citología , Proteína p53 Supresora de Tumor/metabolismo , Animales , Muerte Celular , Proliferación Celular , Relación Dosis-Respuesta a Droga , Humanos , Ratones , Especies Reactivas de Oxígeno
7.
Cell Signal ; 20(10): 1830-8, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18644433

RESUMEN

Ultraviolet radiation (UV) induces apoptosis and functional maturation in skin dendritic cells (DCs). However, the molecular mechanisms through which UV activates DCs have not been thoroughly investigated. In this study, we examined the mechanisms of activation and apoptosis of DCs after UV irradiation by focusing on epidermal growth factor receptor (EGFR). Our previous studies have demonstrated that in addition to cognate ligands, EGFR is also activated by UVB irradiation in cultured human skin keratinocytes in vitro and in human skin in vivo. We found for the first time in this study that UV also induces EGFR activation in cultured mouse skin DCs (XS 106 cell line) as well as mouse monocyte-derived dendritic cells (MoDCs). Pharmacological inhibition of EGFR tyrosine kinase significantly inhibits UV-induced ERK, p38, and JNK MAP kinases, and their effectors, transcription factors c-Fos and c-Jun. Inhibition of EGFR also suppresses UV-induced activation of PI3K/AKT/mTOR/S6K and NF-kappaB signal transduction pathways. Our data demonstrated that UV induces LKB1/AMPK pathway, also dependent on EGFR trans-activation. We further observed that MAPK, LKB1/AMPK, PI3K/AKT/mTOR/S6K as well as NF-kappaB activation are impaired in EGFR-/- cells compared to wide type MEF cells after UV radiation. Taken together, we conclude that UV induces multiple signaling pathways mediated by EGFR trans-activation leading to possible maturation, apoptosis and survival, and EGFR activation protects against UV-induced apoptosis in cultured mouse dendritic cells.


Asunto(s)
Apoptosis/efectos de la radiación , Células Dendríticas/citología , Células Dendríticas/enzimología , Receptores ErbB/metabolismo , Piel/citología , Rayos Ultravioleta , Proteínas Quinasas Activadas por AMP , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular , Células Cultivadas , Células Dendríticas/efectos de la radiación , Activación Enzimática/efectos de la radiación , Receptores ErbB/genética , Factores Eucarióticos de Iniciación , Humanos , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Modelos Biológicos , Complejos Multienzimáticos/metabolismo , FN-kappa B/metabolismo , Fosfoproteínas/metabolismo , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Quinasas S6 Ribosómicas/metabolismo , Transducción de Señal/efectos de la radiación , Piel/enzimología , Serina-Treonina Quinasas TOR , Activación Transcripcional/efectos de la radiación
8.
Lung ; 186(3): 145-150, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18401642

RESUMEN

Increased circulating endothelial cells (CECs) have been observed in patients with vascular injury associated with acute myocardial infarction, pulmonary hypertension, and congestive heart failure. Decreased circulating endothelial progenitor cells (EPCs) have been observed in patients with risk factors for cardiovascular disease. Obstructive sleep apnea (OSA) is associated with increased risk of cardiovascular disease and endothelial dysfunction. Subjects were recruited from patients referred for overnight polysomnograms; 17 subjects had OSA and 10 control subjects did not have OSA. All subjects lacked vascular disease and risk factors for vascular disease. Peripheral blood was obtained from fasting subjects in the morning, following sleep studies. CECs and EPCs were quantified using magnetic bead separation with UV epifluorescence microscopy and flow cytometry immunophenotyping, respectively. Cell counts and demographic variables were compared using unpaired t tests. Regression analysis was performed comparing cell counts with the apnea-hypopnea index (AHI) and nadir SaO(2). Subjects with OSA and controls did not differ significantly in terms of age and body mass index. Subjects with OSA had higher AHI, lower nadir SaO(2), and greater sleepiness (Epworth Sleepiness Scale scores). There were no significant differences in CEC (7.0+/-1.5 vs. 4.9+/-0.9, p>0.05) or EPC (1077+/-318 vs. 853+/-176, p>0.05) between controls and OSA cases, respectively. In this small study, we found no differences in CECs or circulating EPCs between patients with OSA and controls. OSA may not be associated with these markers of vascular endothelial cell injury in patients with no concomitant vascular disease.


Asunto(s)
Células Endoteliales/patología , Endotelio Vascular/patología , Apnea Obstructiva del Sueño/sangre , Células Madre/patología , Recuento de Células , Humanos , Persona de Mediana Edad , Neovascularización Patológica , Polisomnografía , Pronóstico , Estudios Prospectivos , Apnea Obstructiva del Sueño/patología , Apnea Obstructiva del Sueño/fisiopatología
9.
J Ocul Pharmacol Ther ; 24(2): 164-74, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18341430

RESUMEN

Ultraviolet (UV) radiation is related to cataract formation. The dynamics of matrix proteins play crucial roles in cell proliferation, cell migration, and the remodeling of lens capsule and, possibly, cataract formation. However, the change of dynamics of matrix proteins, such as collagens, in lens cells in response to UV radiation has not been investigated. Using cultured human lens epithelial cells, we, for the first time, demonstrate that UV radiation induces a decrease of collagen type I in a time- and dose-dependent manner. Hydrogen peroxide (H(2)O(2)) also induces a collagen type I decrease in a similar pattern. We observed that UV and H(2)O(2) induce JNK and its downstream component, c-Jun, activation in both a time- and dose-dependent manner. The pharmacologic inhibitor of JNK or JNKi inhibits UV-induced JNK and c-Jun activation and attenuates a UV-induced decrease of collagen type I. Quercetin, a well known antioxidant, also protects against a UV- and H(2)O(2)-induced decrease of collagen type I in a dose-dependent manner. Quercetin inhibits UV- and H(2)O(2)-induced JNK and c-Jun activation. Collectively, we conclude that quercetin attenuates both a UV- and H(2)O(2)-induced decrease of collagen type I via the inhibiting of JNK/c-Jun activity. Understanding the cellular-signaling pathways involved in the UV- and H(2)O(2)-induced decrease of collagen type I may reveal potential therapeutic targets for the UV-induced cataract.


Asunto(s)
Antioxidantes/farmacología , Colágeno Tipo I/efectos de los fármacos , Cristalino/efectos de los fármacos , Quercetina/farmacología , Antioxidantes/administración & dosificación , Catarata/etiología , Catarata/prevención & control , Línea Celular , Colágeno Tipo I/metabolismo , Relación Dosis-Respuesta a Droga , Relación Dosis-Respuesta en la Radiación , Sistemas de Liberación de Medicamentos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Humanos , Peróxido de Hidrógeno/toxicidad , Proteínas Quinasas JNK Activadas por Mitógenos/efectos de los fármacos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Cristalino/citología , Quercetina/administración & dosificación , Factores de Tiempo , Rayos Ultravioleta/efectos adversos
10.
Cancer Chemother Pharmacol ; 62(5): 857-65, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18214481

RESUMEN

OBJECTIVE: Aquaporin (AQP) water channels are expressed in high-grade tumor cells of different tissue origins. Based on the involvement of AQPs in angiogenesis and cell migration as well as our previous studies which show that AQP3 is involved in human skin fibroblasts cell migration, in this study, we investigated whether AQP3 is expressed in cultured human ovarian cancer cell line CaOV3 cells, and whether AQP3 expression in these cells enhances cell migration and metastatic potential. METHODS: Cultured CaOV3 cells were treated with EGF and/or various reagents and subjected to cell migration assay by phagokinetic track mobility assay or biochemical analysis for expression or activation of proteins by SDS-PAGE/Western blot analysis. RESULTS: In this study, we demonstrate that AQP3 is expressed in CaOV3 cells. EGF induces CaOV3 migration and up-regulates AQP3 expression. EGF-induced cell migration is inhibited by specific AQP3 siRNA knockdown or AQP3 water transport inhibitor CuSO4 and NiCl2. We also find that curcumin, a well known anti-ovarian cancer drug, down-regulates AQP3 expression and reduces cell migration in CaOV3, and the effects of curcumin are mediated, at least in part, by its inhibitory effects on EGFR and downstream AKT/ERK activation. CONCLUSIONS: Collectively, our results provide evidence for AQP3-facilitated ovarian cancer cell migration, suggesting a novel function for AQP3 expression in high-grade tumors. The results that curcumin inhibits EGF-induced up-regulation of AQP3 and cell migration, provide a new explanation for the anticancer potential of curcumin.


Asunto(s)
Antineoplásicos/farmacología , Acuaporina 3/biosíntesis , Curcumina/farmacología , Factor de Crecimiento Epidérmico/antagonistas & inhibidores , Neoplasias Ováricas/tratamiento farmacológico , Acuaporina 3/antagonistas & inhibidores , Western Blotting , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Sulfato de Cobre/farmacología , Factor de Crecimiento Epidérmico/farmacología , Receptores ErbB/antagonistas & inhibidores , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Níquel/farmacología , Neoplasias Ováricas/patología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Interferencia de ARN , Regulación hacia Arriba/efectos de los fármacos
11.
J Cell Physiol ; 215(2): 506-16, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18064629

RESUMEN

One of the major characteristics of human skin photoaging induced by ultraviolet (UV) radiation is the dehydration of the skin. Water movement across plasma membrane occurs via diffusion through lipid bilayer and via aquaporins (AQPs). We find that UV induces aquaporin-3 (AQP3) down-regulation in human skin keratinocytes. MEK/ERK inhibitors PD98059 and U0126 inhibit UV-induced down-regulation of AQP3. Antioxidant N-acetyl-L-cysteine or NAC blocks UV-induced MEK/ERK activation and down-regulation of AQP3. All-trans retinoic acid or atRA, while alone inducing AQP3 expression, attenuates UV-induced down-regulation of AQP3 and water permeability. Using special inhibitors, we find that activation of EGFR and inhibition on ERK activation are involved in atRA's protective effects against UV-induced AQP3 down-regulation. Using specific AQP3's water transport inhibitors and siRNA knockdown, we observe that AQP3 is involved in cell migration and in vitro wound healing. UV-induced AQP3 down-regulation results in reduced water permeability, decreased cell migration, and delayed wound healing, which are attenuated by atRA pretreatment. We conclude that atRA protects against UV-induced down-regulation AQP3 and decrease in water permeability, reduction in cell migration and delayed in vitro wound healing via trans-activation of EGFR and inhibition on ROS-mediated MEK/ERK pathway. This novel finding provides evidence to support possible involvement of AQP3 in UV induced skin dehydration.


Asunto(s)
Acuaporina 3/metabolismo , Queratinocitos/metabolismo , Queratinocitos/efectos de la radiación , Tretinoina/farmacología , Agua/metabolismo , Acetilcisteína/farmacología , Antioxidantes/farmacología , Permeabilidad de la Membrana Celular/efectos de los fármacos , Permeabilidad de la Membrana Celular/efectos de la radiación , Movimiento Celular/fisiología , Células Cultivadas , Regulación hacia Abajo/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Genes erbB-1 , Humanos , Peróxido de Hidrógeno/farmacología , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Oxidantes/farmacología , Factores de Tiempo , Activación Transcripcional/fisiología , Rayos Ultravioleta , Cicatrización de Heridas/fisiología
12.
Wound Repair Regen ; 15(6): 856-65, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-18028134

RESUMEN

Bone marrow-derived cells have long been regarded to play a crucial role in the homeostasis of skin. We have previously described the clinical benefit of directly applying autologous bone marrow aspirate and cultured bone marrow cells to recalcitrant chronic skin wounds. The initial response to treatment appears to be vascular in nature with the formation of new blood vessels. The difficulty in consistently growing adequate numbers of cells for delivery to patients was, however, a limiting factor. Here, in a subsequent protocol, we describe an improved bone marrow culture system yielding a reliable growth of bone marrow cells and leading to a greater clinical response. Cells expressing markers of endothelial progenitors including CD133, CD146, and particularly CD14 are enhanced in these cultures. CD14-isolated cells produced colonies in endothelial cell assays and sprouting in matrigel assays. Angiogenic cytokines, including angiogenin, epithelial neutrophil-activating protein-78, growth-regulated oncogene, growth-regulated oncogene-alpha, Interleukin-8, CXC16, and monocyte chemoattractant protein-1, were found to be elevated in these cultures. Administration of improved culture cells to patients with chronic wounds present for >1 year lead to an enhanced clinical response.


Asunto(s)
Trasplante de Médula Ósea/métodos , Cicatrización de Heridas/fisiología , Heridas y Lesiones/terapia , Administración Tópica , Células Cultivadas , Enfermedad Crónica , Citocinas/metabolismo , Humanos , Inyecciones , Apósitos Oclusivos , Piel/patología , Trasplante Autólogo , Resultado del Tratamiento
13.
Transfusion ; 47(7): 1154-61, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17581149

RESUMEN

BACKGROUND: Current FDA regulations allow the prestorage pooling of whole blood-derived platelet concentrates (PCs) of identical ABO type in a recently cleared platelet (PLT) pooling bag (Acrodose, Pall Medical). It is unclear how ABO-mixed PC pools would store if pooled before storage. STUDY DESIGN AND METHODS: Pools consisting of ABO-identical PLTs and mixed A and O PLTs in varying proportions were evaluated on Days 1, 5, and 7 of storage with measures of the PLT storage lesion, lymphocyte activation, and activation of the complement and coagulation system. Data were analyzed by analysis of variance. RESULTS: Pools did not differ on Day 7 in pH (p = 0.63), hypotonic shock response (p = 0.25), extent of shape change (p = 0.26), morphology score (p = 0.18), or white cell count (p = 0.79), but surface P-selectin expression was more evident in the ABO-mixed pools (p = 0.02). Small microscopic clumps of PLTs were observed in all pools, but were more prominent in the ABO-mixed pools (p < 0.01). PLT counts, however, did not differ between pools (p = 0.93), indicating that only a small proportion of PLTs were clumped. Surface A-antigen expression was proportional to the number of A PCs in each pool and did not vary between study days. Anti-A(1) titers were either unchanged or decreased by one dilution. Complement and coagulation activation markers did not differ between pools. CONCLUSION: Pooling A and O PCs is associated with evidence of increased microscopic PLT clumping and activation, but these differences are not exacerbated with 7-day storage. Other major measures of PLT quality do not differ, and there was no evidence of a mixed lymphocyte reaction.


Asunto(s)
Sistema del Grupo Sanguíneo ABO , Plaquetas , Conservación de la Sangre/métodos , Forma de la Célula , Humanos , Activación Plaquetaria , Adhesividad Plaquetaria , Pruebas de Función Plaquetaria , Factores de Tiempo
14.
J Cell Physiol ; 212(1): 252-63, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17301957

RESUMEN

Ultraviolet radiation (UV) induces cell damages leading to skin photoaging and skin cancer. ATP-sensitive potassium (K(ATP)) channel openers (KCOs) have been shown to exert significant myocardial preservation and neuroprotection in vitro and in vivo, and yet the potential role of those KCOs in protection against UV-induced skin cell damage is unknown. We investigated the effects of pinacidil and diazoxide, two classical KCOs, on UV-induced cell death using cultured human keratinocytes (HaCat cells). Here, we demonstrated for the first time that Kir 6.1, Kir 6.2 and SUR2 subunits of K(ATP) channels are functionally expressed in HaCaT cells and both non-selective K(ATP) channel opener pinacidil and mitoK(ATP) (mitochondrial K(ATP)) channel opener diazoxide attenuated UV-induced keratinocytes cell death. The protective effects were abolished by both non-selective K(ATP) channel blocker glibenclamide and selective mitoK(ATP) channel blocker 5-hydroxydecanoate (5-HD). Also, activation of K(ATP) channel with pinacidil or diazoxide resulted in suppressive effects on UV-induced MAPK activation and reactive oxygen species (ROS) production. Unexpectedly, we found that the level of intracellular ROS was slightly elevated in HaCaT cells when treated with pinacidil or diazoxide alone. Furthermore, UV-induced mitochondrial membrane potential loss, cytochrome c release and ultimately apoptotic cell death were also inhibited by preconditioning with pinacidil and diazoxide, and their effects were reversed by glibenclamide and 5-HD. Taken together, we contend that mitoK(ATP) is likely to contribute the protection against UV-induced keratinocytes cell damage. Our findings suggest that K(ATP) openers such as pinacidil and diazoxide may be utilized to prevent from UV-induced skin aging.


Asunto(s)
Adenosina Trifosfato/metabolismo , Queratinocitos/citología , Queratinocitos/efectos de la radiación , Canales de Potasio de Rectificación Interna/metabolismo , Rayos Ultravioleta/efectos adversos , Diazóxido/farmacología , Relación Dosis-Respuesta a Droga , Relación Dosis-Respuesta en la Radiación , Humanos , Queratinocitos/efectos de los fármacos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Pinacidilo/farmacología , Especies Reactivas de Oxígeno/metabolismo , Factores de Tiempo , Vasodilatadores/farmacología
15.
Int J Oncol ; 29(4): 1003-11, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16964397

RESUMEN

Ovarian cancer remains the leading cause of fatality among all gynecologic cancers, although promising therapies are in the making. It has been speculated that metastasis is critical for ovarian cancer, and yet the molecular mechanisms of metastasis in ovarian cancer are poorly understood. Growth factors have been proven to play important roles in cell migration associated with metastasis, and inhibition of growth factor receptors and their distinct cell signaling pathways has been intensively studied, and yet the uncovered interaction or crosstalk among various growth factor receptors complicates this otherwise promising approach. We investigated the crosstalk between EGFR and TrkB, both of which have been known to be important in cell survival and migration in response to EGF and BDNF. Our results showed that both EGF and BDNF induced cell migration and cell proliferation in cultured human ovarian cancer cells (Caov3 cell line). EGF and BDNF transactivated TrkB and EGFR respectively, and activated downstream cell survival components such as Akt. EGFR and TrkB kinase inhibitors inhibited EGF- and BDNF-induced TrkB and EGFR activation and Akt phosphorylation, and cell proliferation and migration. Using EGFR knockout cells, we further demonstrated that EGFR is required for EGF-induced cell migration. Collectively, our data indicate that EGFR and TrkB crosstalk each other in response to EGF and BDNF, leading to cell survival pathway activation in ovarian cancer cells. Our data suggest that a combination of inhibitors of both receptors with cell survival pathway inhibitors would provide a better outcome in the clinical treatment of ovarian cancer.


Asunto(s)
Receptores ErbB/metabolismo , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Receptor trkB/metabolismo , Factor Neurotrófico Derivado del Encéfalo/antagonistas & inhibidores , Factor Neurotrófico Derivado del Encéfalo/farmacología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Factor de Crecimiento Epidérmico/antagonistas & inhibidores , Factor de Crecimiento Epidérmico/farmacología , Receptores ErbB/agonistas , Receptores ErbB/genética , Femenino , Humanos , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Proto-Oncogénicas c-akt/agonistas , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Receptor trkB/agonistas , Receptor trkB/genética , Activación Transcripcional , Células Tumorales Cultivadas
16.
Oncol Rep ; 16(4): 907-13, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16969513

RESUMEN

The molecular and cellular mechanism of the development of pancreatic cancer is under constant and intensive study, and yet the cure is still out of reach. While surgical treatment is optional, conventional chemotherapy or chemo-radiotherapy remains the best choice. Among others, paclitaxel is proven to be a popular and, to a certain extent, effective chemotherapy agent. We proposed that the combination of paclitaxel and membrane permeable ceramide would enhance the fatality of cancer cells, and reported that the combination increased cell death of both head and neck and leukemic cancer cells. In this study, we treated pancreatic cancer cells (L3.6 cells) with paclitaxel and ceramide at the concentrations of clinical relevance, and treatment was then followed up with an investigation of the molecular mechanism of the synergism of paclitaxel and ceramide. The results of Western blot analysis indicated that the combo synergistically induced ERK and JNK phosphorylation, but not p38 and Akt phosphorylation. We also found that the combination (combo) induced EGFR phosphorylation in a synergistic manner. Furthermore, we observed that paclitaxel, ceramide, or combo-induced EGFR phosphorylation was inhibited by EGFR inhibitor, PD153035, while paclitaxel, ceramide, or combo-induced JNK and ERK phosphorylation was blocked by EGFR inhibitor, PD153035 and ERK inhibitor, U126. Taken together, our results demonstrated that the combination of paclitaxel and ceramide synergistically induced pancreatic cancer cell death through differential activation of EGFR-mediated MAP kinases. EGFR and ERK inhibitors may further enhance the paclitaxel and ceramide effect.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Ceramidas/metabolismo , Receptores ErbB/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Paclitaxel/farmacología , Neoplasias Pancreáticas/metabolismo , Apoptosis , Muerte Celular , Línea Celular Tumoral , Activación Enzimática , Humanos , Proteínas Inhibidoras de la Apoptosis , Sistema de Señalización de MAP Quinasas , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas de Neoplasias/metabolismo , Fosforilación , Survivin , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
17.
Biochem J ; 400(2): 225-34, 2006 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-16848764

RESUMEN

AQP3 (aquaporin-3), known as an integral membrane channel in epidermal keratinocytes, facilitates water and glycerol movement into and out of the skin. Here, we demonstrate that AQP3 is also expressed in cultured human skin fibroblasts, which under normal wound healing processes migrate from surrounding tissues to close the wound. EGF (epidermal growth factor), which induced fibroblast migration, also induced AQP3 expression in a time- and dose-dependent manner. CuSO4 and NiCl2, previously known as AQP3 water transport inhibitors, as well as two other bivalent heavy metals Mn2+ and Co2+, inhibited EGF-induced cell migration in human skin fibroblasts. AQP3 knockdown by small interfering RNA inhibited EGF-induced AQP3 expression and cell migration. Furthermore, an EGFR (EGF receptor) kinase inhibitor, PD153035, blocked EGF-induced AQP3 expression and cell migration. MEK [MAPK (mitogen-activated protein kinase)/ERK (extracellular-signal-regulated kinase) kinase]/ERK inhibitor U0126 and PI3K (phosphoinositide 3-kinase) inhibitor LY294002 also inhibited EGF-induced AQP3 expression and cell migration. Collectively, our findings show for the first time that AQP3 is expressed in human skin fibroblasts and that EGF induces AQP3 expression via EGFR, PI3K and ERK signal transduction pathways. We have provided evidence for a novel role of AQP3 in human skin fibroblast cell migration, which occurs during normal wound healing.


Asunto(s)
Acuaporina 3/biosíntesis , Movimiento Celular/fisiología , Receptores ErbB/metabolismo , Piel/citología , Piel/metabolismo , Acuaporina 3/genética , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Interacciones Farmacológicas , Factor de Crecimiento Epidérmico/antagonistas & inhibidores , Factor de Crecimiento Epidérmico/farmacología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/enzimología , Fibroblastos/metabolismo , Humanos , Queratinocitos/citología , Queratinocitos/efectos de los fármacos , Queratinocitos/enzimología , Queratinocitos/metabolismo , Níquel/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Interferencia de ARN , Transducción de Señal/efectos de los fármacos , Piel/efectos de los fármacos , Piel/enzimología , Sulfatos/farmacología , Cicatrización de Heridas/efectos de los fármacos , Cicatrización de Heridas/fisiología
18.
Int J Mol Med ; 18(2): 381-7, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16820949

RESUMEN

Ultraviolet radiation-induced cataract has been believed to be associated with degradation of cellular components. We report that, in cultured human lens epithelial cells, UV radiation analogous to H2O2 treatment down-regulates desmosomal protein desmoglein-2. UV radiation induces generation of reactive oxygen species and transiently activates epidermal growth factor receptor, which in turn induces translocation of Rac2 and NADPH oxidase activity. Collectively, our data demonstrate that UV-induced desmoglein-2 down-regulation is mediated reactive oxygen species which are generated through EGFR activation and Rac2/NADPH oxidase activation, suggesting that antioxidants may be applied for protection against UV-induced cataract.


Asunto(s)
Desmogleína 2/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/efectos de la radiación , Cristalino , NADPH Oxidasas/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Animales , Línea Celular , Desmogleína 2/genética , Regulación hacia Abajo , Inducción Enzimática , Células Epiteliales/citología , Receptores ErbB/metabolismo , Humanos , Peróxido de Hidrógeno/metabolismo , Cristalino/citología , Cristalino/efectos de la radiación , Oxidantes/metabolismo , Fosforilación , Especies Reactivas de Oxígeno/metabolismo , Rayos Ultravioleta , Proteína RCA2 de Unión a GTP
19.
Int J Oncol ; 29(1): 269-78, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16773209

RESUMEN

Trichostatin A (TSA), a hydroxamate-type inhibitor of mammalian histone deacetylases, is emerging as one of a potentially new class of anticancer agents. TSA is known to act by promoting the acetylation of histones, leading to uncoiling of chromatin and activation of a variety of genes implicated in the regulation of cell survival, proliferation, differentiation, and apoptosis. In addition, there is an increasing appreciation of the fact that TSA may act through mechanisms other than induction of histone acetylation. Accumulated experimental data indicate that TSA activates phosphatidyl inositol-3-kinase (PI3K)/AKT signaling. Using human ovarian cancer cell line Caov3 cells, we observed that TSA induced cell death in a time- and dose-dependent manner and also inhibited cell migration. TSA transiently activated EGFR tyrosine phosphorylation and AKT activation in a time- and dose-dependent manner, which had been inhibited by EGFR inhibitor PD153035 and PI3 kinase inhibitor LY294002. We also observed that TSA transiently induced survivin expression that had been inhibited by PD153035 and LY294002, suggesting that TSA-induced survivin expression is mediated by EGFR/PI3 kinase pathway. Combination of EGFR inhibitor 153035 or PI3 kinase inhibitor LY294002 with TSA enhanced TSA-induced cell death and TSA reduction of cell migration. Collectively, our data demonstrate that TSA transiently activated EGFR/PI3K/AKT cell survival pathway, leading to expression of survivin. Inhibition of this pathway enhanced TSA-induced cell death and inhibited cell migration. Our data suggest that combination of EGFR/PI3K/AKT cell survival pathway inhibitors with TSA be a better approach to ovarian cancer treatment.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Receptores ErbB/metabolismo , Ácidos Hidroxámicos/farmacología , Neoplasias Ováricas/enzimología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Acetilación/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Cromonas/farmacología , Relación Dosis-Respuesta a Droga , Receptores ErbB/efectos de los fármacos , Femenino , Inhibidores de Histona Desacetilasas , Histona Desacetilasas/metabolismo , Histonas/metabolismo , Humanos , Proteínas Inhibidoras de la Apoptosis , Proteínas Asociadas a Microtúbulos/metabolismo , Morfolinas/farmacología , Proteínas de Neoplasias/metabolismo , Neoplasias Ováricas/patología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas/farmacología , Quinazolinas/farmacología , Survivin , Factores de Tiempo
20.
Transfusion ; 46(5): 835-40, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16686852

RESUMEN

BACKGROUND: Prestorage pooling of platelet (PLT)-rich plasma (PRP)-derived PLT concentrates (PCs) and storage in either plasma (PS) or an additive solution (AS) is logistically feasible and would result in a product similar to buffy-coat or apheresis PLTs. STUDY DESIGN AND METHODS: On Day 0, PS PRP PCs were pooled with a sterile connecting device into a new 1.3-L storage container (ELX, PALL Medical). AS-PCs were prepared by addition of a new low-pH glucose-containing AS to the PLT sediment. AS-PCs were pooled into a 1.3-L ELX bag containing four tablets of NaHCO3. PC pools were sampled on Days 1, 5, and 7. RESULTS: PS pools containing 5 units had a mean PLT yield similar to the AS pools (39 x 10(10) +/- 3 x 10(10) vs. 37 x 10(10) +/- 6 x 10(10); p = 0.25). All pools had WBC counts of less than 1 x 10(6). pH and HCO3 decreased in PS pools with storage, but either increased or remained constant in the AS pools. On Day 7, no differences were seen in morphology score or extent of shape change. Hypotonic shock response was better preserved in the plasma pools (71 +/- 12% vs. 56 +/- 13%, p < 0.01); however, surface P-selectin was expressed less in the AS pools (6 +/- 4% vs. 18 +/- 10%, p < 0.01). CONCLUSION: Manufacture and storage of PRP-PCs in pools either in plasma or in a glucose-containing AS in this new container are feasible, and there is good preservation of PLT quality to Day 7.


Asunto(s)
Plaquetas , Conservación de la Sangre , Plasma , Plaquetas/citología , Conservación de la Sangre/instrumentación , Glucosa/química , Humanos , Plasma/citología , Soluciones , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...