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

Bases de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Biochem Biophys Res Commun ; 650: 73-80, 2023 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-36773342

RESUMEN

Intracellular lipid droplet (LD) generation is the primary site of energy storage, which is necessary for physiological homeostasis but is related to pathological metabolic disorders. Lipid metabolism is critical for maintaining innate and adaptive immunity; however, it is mainly undefined in peripheral immune cells. Flow cytometry-based immune profiling in healthy peripheral blood cells showed significant original generation of LDs in dendritic cells (DCs, CD3-CD19-CD56-CD11+), monocytes (CD3-CD19-CD56-CD14+), natural killer cells (NK, CD3-CD19-CD56+), and B cells (CD3-CD19+). CD36, a common scavenger receptor of lipids, was also highly expressed in LD-accumulated DCs and monocytes. Following short-term treatment with oxidized LDL (oxLDL) in an experimental ex vivo model, CD14+ monocytes showed an effective increase in LD generation, but there were no alterations in the immune cell populations. Furthermore, oxLDL-treated CD14+ monocytes displayed CD36 expression. However, oxLDL-primed CD14+ monocytes showed a blockade in the uptake of extra oxLDL, even while expressing increased CD36, indicating a defect in lipid clearance. Exogenous treatment with oxLDL caused monocyte type 1 polarization accompanied by increased LD accumulation and CD36 expression. This study describes a method to monitor LD generation and CD36 expression in peripheral immune cells and identified an immunomodulatory effect of oxLDL on monocytes by tilting them towards type 1 polarization.


Asunto(s)
Dislipidemias , Monocitos , Humanos , Monocitos/metabolismo , Gotas Lipídicas/metabolismo , Lipoproteínas LDL/metabolismo , Receptores Depuradores/metabolismo , Antígenos CD36/metabolismo , Dislipidemias/metabolismo
2.
Int J Mol Sci ; 23(9)2022 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-35563060

RESUMEN

Glycogen synthase kinase-3 (GSK-3), a serine/threonine kinase, is a vital glycogen synthase regulator controlling glycogen synthesis, glucose metabolism, and insulin signaling. GSK-3 is widely expressed in different types of cells, and its abundant roles in cellular bioregulation have been speculated. Abnormal GSK-3 activation and inactivation may affect its original bioactivity. Moreover, active and inactive GSK-3 can regulate several cytosolic factors and modulate their diverse cellular functional roles. Studies in experimental liver disease models have illustrated the possible pathological role of GSK-3 in facilitating acute hepatic injury. Pharmacologically targeting GSK-3 is therefore suggested as a therapeutic strategy for liver protection. Furthermore, while the signaling transduction of GSK-3 facilitates proinflammatory interferon (IFN)-γ in vitro and in vivo, the blockade of GSK-3 can be protective, as shown by an IFN-γ-induced immune hepatitis model. In this study, we explored the possible regulation of GSK-3 and the potential relevance of GSK-3 blockade in IFN-γ-mediated immune hepatitis.


Asunto(s)
Glucógeno Sintasa Quinasa 3 , Hepatitis , Interferón gamma , Animales , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Hepatitis/inmunología , Interferón gamma/farmacología , Ratones , Proteínas Serina-Treonina Quinasas , Transducción de Señal
3.
Radiology ; 301(3): 735-740, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34807772

RESUMEN

History A 55-year-old woman without systemic underlying disease, such as diabetes mellitus, inflammatory bowel disease, autoimmune disease, or chronic kidney disease, presented with generalized dull abdominal pain of 1-week duration. She had ingested herbal medicine for physical conditioning for several years. Laboratory findings, including biochemistry, electrolyte levels, and complete blood count, were all within normal limits, except for elevated serum C-reactive protein level (7.719 mg/dL; normal range, <1 mg/dL). The patient underwent initial evaluation with conventional abdominal radiography. She underwent subsequent evaluation with noncontrast CT of the abdomen and colonoscopy.


Asunto(s)
Colitis/complicaciones , Colitis/diagnóstico por imagen , Tomografía Computarizada por Rayos X/métodos , Calcificación Vascular/complicaciones , Calcificación Vascular/diagnóstico por imagen , Dolor Abdominal/etiología , Anticoagulantes/uso terapéutico , Colitis/tratamiento farmacológico , Colon/diagnóstico por imagen , Femenino , Humanos , Mucosa Intestinal/diagnóstico por imagen , Persona de Mediana Edad , Radiografía Abdominal , Calcificación Vascular/tratamiento farmacológico , Warfarina/uso terapéutico
4.
Radiology ; 300(2): 481-483, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34310227

RESUMEN

History A 55-year-old woman without systemic underlying disease, such as diabetes mellitus, inflammatory bowel disease, autoimmune disease, or chronic kidney disease, presented with generalized dull abdominal pain of 1-week duration. She had ingested herbal medicine for physical conditioning for several years. Laboratory findings, including biochemistry, electrolyte levels, and complete blood count, were all within normal limits, except for elevated serum C-reactive protein level (7.719 mg/dL; normal range,<1 mg/dL). The patient underwent initial evaluation with conventional abdominal radiography (Fig 1). She underwent subsequent evaluation with noncontrast CT of the abdomen (Figs 2, 3) and colonoscopy (Fig 4).

5.
J Cell Physiol ; 234(9): 16003-16010, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-30767202

RESUMEN

Fractionated ionizing radiation (FIR) is a radiotherapy regimen that is regularly performed as part of lung cancer treatment. In contrast to the growth inhibition caused by DNA damage, immunomodulation in post-irradiated cancer cells is not well documented. Interferon (IFN)-γ confers anticancer activity by triggering both growth inhibition and cytotoxicity. This study investigated the priming effects of FIR with immunomodulation on the anticancer IFN-γ. Cell morphology, cell growth, and cytotoxicity were observed in FIR-treated A549 lung adenocarcinoma. Induction of p53 and epithelial-mesenchymal transition (EMT) were monitored. Following FIR, activation of IFN-γ signaling pathways were detected. FIR caused changes in cell morphology, inhibited cell growth, and induced cytotoxicity. While p53 was induced by FIR, no epithelial-mesenchymal transition could be found. Following IFN-γ stimulation, FIR-induced p53-associated cell cytotoxicity was significantly enhanced. Additionally, FIR increased the downstream response to IFN-γ by facilitating IFN-γ-induced signal transducer and activator of transcription 1 (STAT1) signaling without affecting the receptor expression. FIR-facilitated STAT1 activation through the mechanism involving mitogen-activated protein kinase activation and Src-homology 2 domain-containing tyrosine phosphatase 2 inactivation. These results demonstrate the FIR-facilitated IFN-γ signaling and its anticancer activity.

6.
J Biomed Sci ; 24(1): 10, 2017 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-28143527

RESUMEN

Immune interferon (IFN), also known as IFN-γ, promotes not only immunomodulation but also antimicrobial and anticancer activity. After IFN-γ binds to the complex of IFN-γ receptor (IFNGR) 1-IFNGR2 and subsequently activates its downstream signaling pathways, IFN-γ immediately causes transcriptional stimulation of a variety of genes that are principally involved in its biological activities. Regarding IFN-γ-dependent immunosurveillance, IFN-γ can directly suppress tumorigenesis and infection and/or can modulate the immunological status in both cancer cells and infected cells. Regarding the anticancer effects of IFN-γ, cancer cells develop strategies to escape from IFN-γ-dependent cancer immunosurveillance. Immune evasion, including the recruitment of immunosuppressive cells, secretion of immunosuppressive factors, and suppression of cytotoxic T lymphocyte responses, is speculated to be elicited by the oncogenic microenvironment. All of these events effectively downregulate IFN-γ-expressing cells and IFN-γ production. In addition to these extrinsic pathways, cancer cells may develop cellular tolerance that manifests as hyporesponsiveness to IFN-γ stimulation. This review discusses the potential escape mechanisms from IFN-γ-dependent immunosurveillance in tumorigenesis.


Asunto(s)
Interferón gamma/inmunología , Neoplasias/inmunología , Escape del Tumor/inmunología , Microambiente Tumoral/inmunología , Animales , Humanos , Neoplasias/patología
7.
J Virol ; 89(14): 7028-37, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25926657

RESUMEN

UNLABELLED: Enterovirus 71 (EV71) infection causes severe mortality involving multiple possible mechanisms, including cytokine storm, brain stem encephalitis, and fulminant pulmonary edema. Gamma interferon (IFN-γ) may confer anti-EV71 activity; however, the claim that disease severity is highly correlated to an increase in IFN-γ is controversial and would indicate an immune escape initiated by EV71. This study, investigating the role of IFN-γ in EV71 infection using a murine model, showed that IFN-γ was elevated. Moreover, IFN-γ receptor-deficient mice showed higher mortality rates and more severe disease progression with slower viral clearance than wild-type mice. In vitro results showed that IFN-γ pretreatment reduced EV71 yield, whereas EV71 infection caused IFN-γ resistance with attenuated IFN-γ signaling in IFN regulatory factor 1 (IRF1) gene transactivation. To study the immunoediting ability of EV71 proteins in IFN-γ signaling, 11 viral proteins were stably expressed in cells without cytotoxicity; however, viral proteins 2A and 3D blocked IFN-γ-induced IRF1 transactivation following a loss of signal transducer and activator of transcription 1 (STAT1) nuclear translocation. Viral 3D attenuated IFN-γ signaling accompanied by a STAT1 decrease without interfering with IFN-γ receptor expression. Restoration of STAT1 or blocking 3D activity was able to rescue IFN-γ signaling. Interestingly, viral 2A attenuated IFN-γ signaling using another mechanism by reducing the serine phosphorylation of STAT1 following the inactivation of extracellular signal-regulated kinase without affecting STAT1 expression. These results demonstrate the anti-EV71 ability of IFN-γ and the immunoediting ability by EV71 2A and 3D, which attenuate IFN-γ signaling through different mechanisms. IMPORTANCE: Immunosurveillance by gamma interferon (IFN-γ) may confer anti-enterovirus 71 (anti-EV71) activity; however, the claim that disease severity is highly correlated to an increase in IFN-γ is controversial and would indicate an immune escape initiated by EV71. IFN-γ receptor-deficient mice showed higher mortality and more severe disease progression, indicating the anti-EV71 property of IFN-γ. However, EV71 infection caused cellular insusceptibility in response to IFN-γ stimulation. We used an in vitro system with viral protein expression to explore the novel IFN-γ inhibitory properties of the EV71 2A and 3D proteins through the different mechanisms. According to this study, targeting either 2A or 3D pharmacologically and/or genetically may sustain a cellular susceptibility in response to IFN-γ, particularly for IFN-γ-mediated anti-EV71 activity.


Asunto(s)
Enterovirus Humano A/inmunología , Infecciones por Enterovirus/inmunología , Interacciones Huésped-Patógeno , Interferón gamma/antagonistas & inhibidores , Transducción de Señal , Proteínas Virales/metabolismo , Animales , Línea Celular , Modelos Animales de Enfermedad , Humanos , Ratones Endogámicos C57BL , Ratones Noqueados
8.
J Immunol ; 193(8): 4149-58, 2014 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-25225672

RESUMEN

Helicobacter pylori infection not only induces gastric inflammation but also increases the risk of gastric tumorigenesis. IFN-γ has antimicrobial effects; however, H. pylori infection elevates IFN-γ-mediated gastric inflammation and may suppress IFN-γ signaling as a strategy to avoid immune destruction through an as-yet-unknown mechanism. This study was aimed at investigating the mechanism of H. pylori-induced IFN-γ resistance. Postinfection of viable H. pylori decreased IFN-γ-activated signal transducers and activators of transcription 1 and IFN regulatory factor 1 not only in human gastric epithelial MKN45 and AZ-521 but also in human monocytic U937 cells. H. pylori caused an increase in the C-terminal tyrosine phosphorylation of Src homology-2 domain-containing phosphatase (SHP) 2. Pharmacologically and genetically inhibiting SHP2 reversed H. pylori-induced IFN-γ resistance. In contrast to a clinically isolated H. pylori strain HP238, the cytotoxin-associated gene A (CagA) isogenic mutant strain HP238(CagAm) failed to induce IFN-γ resistance, indicating that CagA regulates this effect. Notably, HP238 and HP238(CagAm) differently caused SHP2 phosphorylation; however, imaging and biochemical analyses demonstrated CagA-mediated membrane-associated binding with phosphorylated SHP2. CagA-independent generation of reactive oxygen species (ROS) contributed to H. pylori-induced SHP2 phosphorylation; however, ROS/SHP2 mediated IFN-γ resistance in a CagA-regulated manner. This finding not only provides an alternative mechanism for how CagA and ROS coregulate SHP2 activation but may also explain their roles in H. pylori-induced IFN-γ resistance.


Asunto(s)
Antígenos Bacterianos/inmunología , Proteínas Bacterianas/inmunología , Infecciones por Helicobacter/inmunología , Helicobacter pylori/inmunología , Interferón gamma/inmunología , Proteína Tirosina Fosfatasa no Receptora Tipo 11/inmunología , Animales , Antígenos Bacterianos/genética , Proteínas Bacterianas/genética , Carcinogénesis , Línea Celular Tumoral , Mucosa Gástrica/inmunología , Mucosa Gástrica/microbiología , Humanos , Inflamación/inmunología , Inflamación/microbiología , Factor 1 Regulador del Interferón/metabolismo , Interferón gamma/antagonistas & inhibidores , Ratones , Ratones Endogámicos C57BL , Fosforilación , Proteína Tirosina Fosfatasa no Receptora Tipo 11/antagonistas & inhibidores , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Interferencia de ARN , ARN Interferente Pequeño , Especies Reactivas de Oxígeno/metabolismo , Factor de Transcripción STAT1/metabolismo , Células U937
9.
Biochem Biophys Res Commun ; 466(3): 578-84, 2015 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-26385178

RESUMEN

Src homology-2 domain-containing phosphatase (SHP) 2, an oncogenic phosphatase, inhibits type II immune interferon (IFN)-γ signaling by subverting signal transducers and activators of transcription 1 tyrosine phosphorylation and activation. For cancer immunoediting, this study aimed to investigate the decrease of phosphatase and tensin homolog deleted on chromosome 10 (PTEN), a tumor suppressor protein, leading to cellular impairment of IFN-γ signaling. In comparison with human lung adenocarcinoma A549 cells, the natural PTEN loss in another human lung adenocarcinoma line, PC14PE6/AS2 cells, presents reduced responsiveness in IFN-γ-induced IFN regulatory factor 1 activation and CD54 expression. Artificially silencing PTEN expression in A549 cells also caused cells to be unresponsive to IFN-γ without affecting IFN-γ receptor expression. IFN-γ-induced inhibition of cell proliferation and cytotoxicity were demonstrated in A549 cells but were defective in PC14PE6/AS2 cells and in PTEN-deficient A549 cells. Aberrant activation of SHP2 by ROS was specifically shown in PC14PE6/AS2 cells and PTEN-deficient A549 cells. Inhibiting ROS and SHP2 rescued cellular responses to IFN-γ-induced cytotoxicity and inhibition of cell proliferation in PC14PE6/AS2 cells. These results demonstrate that a decrease in PTEN facilitates ROS/SHP2 signaling, causing lung cancer cells to become unresponsive to IFN-γ.


Asunto(s)
Adenocarcinoma/metabolismo , Interferón gamma/metabolismo , Neoplasias Pulmonares/metabolismo , Fosfohidrolasa PTEN/deficiencia , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Adenocarcinoma/inmunología , Adenocarcinoma/patología , Adenocarcinoma del Pulmón , Línea Celular Tumoral , Proliferación Celular , Activación Enzimática , Técnicas de Silenciamiento del Gen , Humanos , Interferón gamma/inmunología , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/patología , Fosfohidrolasa PTEN/antagonistas & inhibidores , Fosfohidrolasa PTEN/genética , Interferencia de ARN , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Escape del Tumor
10.
Mediators Inflamm ; 2015: 274025, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26199460

RESUMEN

Infection with dengue virus (DENV) causes an increase in proinflammatory responses, such as nitric oxide (NO) generation and TNF-α expression; however, the molecular mechanism underlying this inflammatory activation remains undefined, although the activation of the transcription factor NF-κB is generally involved. In addition to TNF-α production in DENV-infected murine macrophage RAW264.7 cells, inducible NO synthase was transcriptionally and posttranslationally elevated and accompanied by NO generation. NF-κB is known to be activated by DENV infection. Pharmacologically inhibiting NF-κB activation abolishes iNOS/NO biosynthesis and TNF-α production. With inhibition, the potential role of NF-κB in oxidative signaling regulation was prevented during DENV infection. Heat-inactivated DENV failed to cause the identified inflammatory responses. Pharmacological inhibition of TLR3 partly decreased NF-κB activation; however, it effectively abolished inducible iNOS/NO biosynthesis but did not inhibit TNF-α production. In contrast to TLR3, viral protein NS2B3 also independently contributed to NF-κB activation to regulate TNF-α production. These results show the distinct pathways for NF-κB activation caused by DENV infection individually for the regulation of iNOS/NO and TNF-α expression.


Asunto(s)
Virus del Dengue/patogenicidad , Macrófagos/metabolismo , Macrófagos/virología , FN-kappa B/fisiología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Ácidos Cafeicos/farmacología , Línea Celular , Macrófagos/efectos de los fármacos , Ratones , Óxido Nítrico/metabolismo , Alcohol Feniletílico/análogos & derivados , Alcohol Feniletílico/farmacología , Transducción de Señal/efectos de los fármacos
11.
Cell Commun Signal ; 12: 69, 2014 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-25398317

RESUMEN

BACKGROUND: Increased activity or expression of integrin-linked kinase (ILK), which regulates cell adhesion, migration, and proliferation, leads to oncogenesis. We identified the molecular basis for the regulation of ILK and its alternative role in conferring ERK1/2/NF-κB-mediated growth advantages to gastric cancer cells. RESULTS: Inhibiting ILK with short hairpin RNA or T315, a putative ILK inhibitor, abolished NF-κB-mediated the growth in the human gastric cancer cells AGS, SNU-1, MKN45, and GES-1. ILK stimulated Ras activity to activate the c-Raf/MEK1/2/ERK1/2/ribosomal S6 kinase/inhibitor of κBα/NF-κB signaling by facilitating the formation of the IQ motif-containing GTPase-activating protein 1 (IQGAP1)-Ras complex. Forced enzymatic ILK expression promoted cell growth by facilitating ERK1/2/NF-κB signaling. PI3K activation or decreased PTEN expression prolonged ERK1/2 activation by protecting ILK from proteasome-mediated degradation. C-terminus of heat shock cognate 70 interacting protein, an HSP90-associated E3 ubiquitin ligase, mediated ILK ubiquitination to control PI3K- and HSP90-regulated ILK stabilization and signaling. In addition to cell growth, the identified pathway promoted cell migration and reduced the sensitivity of gastric cancer cells to the anticancer agents 5-fluorouracil and cisplatin. Additionally, exogenous administration of EGF as well as overexpression of EGFR triggered ILK- and IQGAP1-regulated ERK1/2/NF-κB activation, cell growth, and migration. CONCLUSION: An increase in ILK non-canonically promotes ERK1/2/NF-κB activation and leads to the growth of gastric cancer cells.


Asunto(s)
Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , FN-kappa B/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Neoplasias Gástricas/metabolismo , Animales , Apoptosis , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Supervivencia Celular , Humanos , Masculino , Ratones Endogámicos BALB C , Fosfohidrolasa PTEN/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Cicatrización de Heridas , Proteínas Activadoras de ras GTPasa/metabolismo , Proteínas ras/metabolismo
12.
Dent Mater ; 40(6): 958-965, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38729780

RESUMEN

OBJECTIVE: To investigate the feasibility of optical coherence tomography (OCT)-based digital image correlation (DIC) analysis and to identify the experimental parameters for measurements of polymerization shrinkage. METHODS: Class I cavities were prepared on bovine incisors and filled with Filtek Z350XT Flowable (Z350F). One OCT image of the polymerized restoration was processed to generate virtually displaced images. In addition, the tooth specimen was physically moved under OCT scanning. A DIC software analyzed these virtual and physical transformation sets and assessed the effects of subset sizes on accuracy. The refractive index of unpolymerized and polymerized Z350F was measured via OCT images. Finally, different particles (70-80 µm glass beads, 150-212 µm glass beads, and 75-150 µm zirconia powder) were added to Z350F to inspect the analyzing quality. RESULTS: The analyses revealed a high correlation (>99.99%) for virtual movements within 131 pixels (639 µm) and low errors (<5.21%) within a 10-µm physical movement. A subset size of 51 × 51 pixels demonstrated the convergence of correlation coefficients and calculation time. The refractive index of Z350F did not change significantly after polymerization. Adding glass beads or zirconia particles caused light reflection or shielding in OCT images, whereas blank Z350F produced the best DIC analysis results. SIGNIFICANCE: The OCT-based DIC analysis with the experimental conditions is feasible in measuring polymerization shrinkage of RBC restorations. The subset size in the DIC analysis should be identified to optimize the analysis conditions and results. Uses of hyper- or hypo-reflective particles is not recommended in this method.


Asunto(s)
Resinas Compuestas , Polimerizacion , Tomografía de Coherencia Óptica , Tomografía de Coherencia Óptica/métodos , Animales , Bovinos , Resinas Compuestas/química , Circonio/química , Estudios de Factibilidad , Incisivo/diagnóstico por imagen , Ensayo de Materiales , Procesamiento de Imagen Asistido por Computador/métodos , Técnicas In Vitro , Preparación de la Cavidad Dental/métodos , Propiedades de Superficie , Refractometría , Restauración Dental Permanente
13.
Dent Mater ; 39(5): 513-521, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37031095

RESUMEN

OBJECTIVES: To investigate the shrinkage-induced damage at the composite-tooth interface by finite element analysis (FEA) using the cohesive zone model (CZM). METHODS: Axisymmetric models of Class I restorations were created to illustrate the interfacial damage around composite resin restorations of different dimensions, with polymerization shrinkage modeled analogously to thermal shrinkage. The damage to the adhesive interface was determined using a CZM based on the fracture strength and fracture energy. To show the effects of damage, conventional models with perfectly bonded composite resin restorations were created as controls. RESULTS: The results indicated interfacial damage at the butt-joint cavosurface margin, dentinoenamel junction, and internal line angle. The percentage of damaged interfacial area was found to increase with decreasing diameter for restorations of the same height. For a given diameter, the damage was more severe for restorations of greater depth. The effects of the damage were further illustrated in the model with a restoration of 2-mm diameter and height. The interfacial damage occurred primarily at the internal line angle (83.3 % of all the damaged interfacial area), leading to local stress relief (from 18.3 MPa to 12.8 MPa), but also higher stress at the damage fronts. Greater local shrinkage was found in composites adjacent to the damage. SIGNIFICANCE: The damage mechanics-based CZM is an essential refinement of the FEA to predict interfacial damage and its implications. The extent of damage was found to be greater around restorations with smaller diameters and greater depths. The entire simulation is available via an open-source platform to facilitate further applications in adhesive dentistry.


Asunto(s)
Resinas Compuestas , Restauración Dental Permanente , Restauración Dental Permanente/métodos , Ensayo de Materiales , Análisis de Elementos Finitos , Polimerizacion
14.
Life Sci ; 321: 121615, 2023 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-37001403

RESUMEN

AIMS: Actinomycin (Act) D, a polypeptide antibiotic, is used clinically to inhibit the growth of malignant tumors. Act D binds to DNA at the transcription initiation complex to prevent the elongation of RNA. Act D causes DNA damage, growth inhibition, and cell death. Myeloid cell leukemia (Mcl-1) is an anti-apoptotic Bcl-2 family member protein, and the present study explored the effects and molecular mechanism of Act D-induced Mcl-1 downregulation. MAIN METHODS: Human adenocarcinoma A549 cells were used to check the cytotoxic signaling pathways of Act D, particularly in apoptotic mechanism, in a cell-based study approach. Specific blockers targeting the apoptotic factors were examined for their possible roles. KEY FINDINGS: We found that Act D caused cell growth inhibition and apoptosis. Propidium iodide-based flow cytometric analysis and immunostaining confirmed cell apoptosis. Treatment with Act D caused DNA damage, followed by p53-independent cell death. Western blotting showed a significant decrease in Mcl-1 expression, mitochondrial transmembrane potential loss, and caspase-9/caspase-3 cascade activation. The proteasome inhibitor MG132 reversed Act D-induced Mcl-1 downregulation. However, pharmacological inhibition of glycogen synthase kinase-3, p53 expression, ER stress, autophagy, and vesicle acidification, which are Mcl-1-regulating signaling pathways, did not rescue these effects. Notably, Cullin-Ring E3 ligase partially mediated Mcl-1 downregulation. Administration of transforming growth factor-ß induced mesenchymal cell differentiation, but Act D still decreased Mcl-1 and caused cell apoptosis. SIGNIFICANCE: All of these data show a potential pro-apoptotic effect for Act D by facilitating Mcl-1 uncanonical downregulation.


Asunto(s)
Leucemia , Neoplasias Pulmonares , Humanos , Dactinomicina/farmacología , Dactinomicina/metabolismo , Regulación hacia Abajo , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Antibacterianos/farmacología , Proteína p53 Supresora de Tumor/metabolismo , Línea Celular Tumoral , Neoplasias Pulmonares/metabolismo , Apoptosis , Células Mieloides , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo
15.
Biomed Pharmacother ; 160: 114397, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36796279

RESUMEN

As an alternative strategy for cancer treatment, the combination of cancer nanomedicine and immunotherapy is promising with regard to efficacy and safety; however, precise modulation of the activation of antitumor immunity remains challenging. Therefore, the aim of the present study was to describe an intelligent nanocomposite polymer immunomodulator, drug-free polypyrrole-polyethyleneimine nanozyme (PPY-PEI NZ), which responds to the B-cell lymphoma tumor microenvironment, for precision cancer immunotherapy. Earlier engulfment of PPY-PEI NZs in an endocytosis-dependent manner resulted in rapid binding in four different types of B-cell lymphoma cells. The PPY-PEI NZ effectively suppressed B cell colony-like growth in vitro accompanied by cytotoxicity via apoptosis induction. During PPY-PEI NZ-induced cell death, mitochondrial swelling, loss of mitochondrial transmembrane potential (MTP), downregulation of antiapoptotic proteins, and caspase-dependent apoptosis were observed. Deregulated AKT and ERK signaling contributed to glycogen synthase kinase-3-regulated cell apoptosis following deregulation of Mcl-1 and MTP loss. Additionally, PPY-PEI NZs induced lysosomal membrane permeabilization while inhibiting endosomal acidification, partly protecting cells from lysosomal apoptosis. PPY-PEI NZs selectively bound and eliminated exogenous malignant B cells in a mixed culture system with healthy leukocytes ex vivo. While PPY-PEI NZs showed no cytotoxicity in wild-type mice, they provided long-term and efficient inhibition of the growth of B-cell lymphoma-driven nodules in a subcutaneous xenograft model. This study explores a potential PPY-PEI NZ-based anticancer agent against B-cell lymphoma.


Asunto(s)
Antineoplásicos , Linfoma de Células B , Linfoma , Humanos , Animales , Ratones , Polietileneimina/farmacología , Polímeros , Pirroles , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Linfoma de Células B/tratamiento farmacológico , Línea Celular Tumoral , Microambiente Tumoral
16.
J Pharmacol Exp Ther ; 343(1): 125-33, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22773863

RESUMEN

Glycogen synthase kinase-3 (GSK-3) facilitates interferon (IFN)-γ signaling. Because IFN-γ is involved in inflammatory skin diseases, such as psoriasis, the aim of this study was to investigate the pathogenic role of GSK-3 in 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced IFN-γ-mediated ear skin inflammation. TPA (3 µg per ear) induced acute skin inflammation in the ears of C57BL/6 mice, including edema, infiltration of granulocytes but not T cells, and IFN-γ receptor 1-mediated deregulation of intercellular adhesion molecule 1 (CD54). TPA/IFN-γ induced GSK-3 activation, which in turn activated signal transducer and activator of transcription 1. Inhibiting GSK-3 pharmacologically, by administering 6-bromoindirubin-3'-oxime (1.5 µg per ear), and genetically, with lentiviral-based short-hairpin RNA, reduced TPA-induced acute skin inflammation but not T-cell infiltration. It is noteworthy that inhibiting GSK-3 decreased TPA-induced IFN-γ production and the nuclear translocation of T-box transcription factor Tbx21, a transcription factor of IFN-γ, in CD3-positive T cells. In chronic TPA-induced skin inflammation, inhibiting GSK-3 attenuated epidermis hyperproliferation and dermis angiogenesis. These results demonstrate the dual role of GSK-3 in TPA-induced skin inflammation that is not only to facilitate IFN-γ signaling but also to regulate IFN-γ production. Inhibiting GSK-3 may be a potential treatment strategy for preventing such effects.


Asunto(s)
Dermatitis/tratamiento farmacológico , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Interferón gamma/fisiología , Acetato de Tetradecanoilforbol/antagonistas & inhibidores , Acetato de Tetradecanoilforbol/toxicidad , Animales , Línea Celular , Dermatitis/enzimología , Glucógeno Sintasa Quinasa 3/metabolismo , Humanos , Indoles/farmacología , Indoles/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oximas/farmacología , Oximas/uso terapéutico , Receptores de Interferón/deficiencia , Receptor de Interferón gamma
17.
FASEB J ; 25(10): 3661-73, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21705667

RESUMEN

Inactivation of glycogen synthase kinase (GSK)-3 has been implicated in cancer progression. Previously, we showed an abundance of inactive GSK-3 in the human chronic myeloid leukemia (CML) cell line. CML is a hematopoietic malignancy caused by an oncogenic Bcr-Abl tyrosine kinase. In Bcr-Abl signaling, the role of GSK-3 is not well defined. Here, we report that enforced expression of constitutively active GSK-3 reduced proliferation and increased Bcr-Abl inhibition-induced apoptosis by nearly 1-fold. Bcr-Abl inhibition activated GSK-3 and GSK-3-dependent apoptosis. Inactivation of GSK-3 by Bcr-Abl activity is, therefore, confirmed. To reactivate GSK-3, we used glucosylceramide synthase (GCS) inhibitor PDMP to accumulate endogenous ceramide, a tumor-suppressor sphingolipid and a potent GSK-3 activator. We found that either PDMP or silence of GCS increased Bcr-Abl inhibition-induced GSK-3 activation and apoptosis. Furthermore, PDMP sensitized the most clinical problematic drug-resistant CML T315I mutant to Bcr-Abl inhibitor GNF-2-, imatinib-, or nilotinib-induced apoptosis by >5-fold. Combining PDMP and GNF-2 eliminated transplanted-CML-T315I-mutants in vivo and dose dependently sensitized primary cells from CML T315I patients to GNF-2-induced proliferation inhibition and apoptosis. The synergistic efficacy was Bcr-Abl restricted and correlated to increased intracellular ceramide levels and acted through GSK-3-mediated apoptosis. This study suggests a feasible novel anti-CML strategy by accumulating endogenous ceramide to reactivate GSK-3 and abrogate drug resistance.


Asunto(s)
Apoptosis/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Genes abl , Glucosiltransferasas/antagonistas & inhibidores , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Morfolinas/farmacología , 1-Desoxinojirimicina/análogos & derivados , 1-Desoxinojirimicina/farmacología , Animales , Apoptosis/fisiología , Línea Celular Tumoral , Ceramidas/metabolismo , Relación Dosis-Respuesta a Droga , Resistencia a Antineoplásicos , Femenino , Genes abl/efectos de los fármacos , Genes abl/fisiología , Glucógeno Sintasa Quinasa 3/metabolismo , Humanos , Inmunoglobulina G , Melfalán , Ratones , Ratones SCID , Mutación , Neoplasias Experimentales , Pirimidinas , Trasplante Heterólogo
18.
Anesthesiology ; 116(4): 868-81, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22334036

RESUMEN

BACKGROUND: Overdose propofol treatment with a prolong time causes injury to multiple cell types; however, its molecular mechanisms remain unclear. Activation of glycogen synthase kinase (GSK)-3ß is proapoptotic under death stimuli. The authors therefore hypothesize that propofol overdose induces macrophage apoptosis through GSK-3ß. METHODS: Phagocytic analysis by uptake of Staphylococcus aureus showed the effects of propofol overdose on murine macrophages RAW264.7 and BV2 and primary human neutrophils in vitro. The authors further investigated cell apoptosis in vitro and in vivo, lysosomal membrane permeabilization, and the loss of mitochondrial transmembrane potential (MTP) by propidium iodide, annexin V, acridine orange, and rhodamine 123 staining, respectively. Protein analysis identified activation of apoptotic signals, and pharmacologic inhibition and genetic knockdown using lentiviral-based short hairpin RNA were further used to clarify their roles. RESULTS: A high dose of propofol caused phagocytic inhibition and apoptosis in vitro for 24 h (25 µg/ml, in triplicate) and in vivo for 6 h (10 mg/kg/h, n = 5 for each group). Propofol induced lysosomal membrane permeabilization and MTP loss while stabilizing MTP and inhibiting caspase protected cells from mitochondrial apoptosis. Lysosomal cathepsin B was required for propofol-induced lysosomal membrane permeabilization, MTP loss, and apoptosis. Propofol decreased antiapoptotic Bcl-2 family proteins and then caused proapoptotic Bcl-2-associated X protein (Bax) activation. Propofol-activated GSK-3ß and inhibiting GSK-3ß prevented Mcl-1 destabilization, MTP loss, and lysosomal/mitochondrial apoptosis. Forced expression of Mcl-1 prevented the apoptotic effects of propofol. Decreased Akt was important for GSK-3ß activation caused by propofol. CONCLUSIONS: These results suggest an essential role of GSK-3ß in propofol-induced lysosomal/mitochondrial apoptosis.


Asunto(s)
Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Glucógeno Sintasa Quinasa 3/fisiología , Lisosomas/efectos de los fármacos , Macrófagos/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Propofol/farmacología , Anestésicos/farmacología , Animales , Línea Celular , Glucógeno Sintasa Quinasa 3 beta , Células Hep G2 , Humanos , Lisosomas/metabolismo , Macrófagos/citología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Mitocondrias/enzimología , Mitocondrias/fisiología
19.
Cancer Lett ; 539: 215712, 2022 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-35490920

RESUMEN

The epithelial-to-mesenchymal transition (EMT) is involved in cancer metastasis; nevertheless, interferon (IFN)-γ induces anticancer activities by causing cell growth suppression, cytotoxicity, and migration inhibition. Regarding the poor response to exogenously administered IFN-γ as anticancer therapy, it was hypothesized that malignant cells may acquire a means of escaping from IFN-γ immunosurveillance, likely through an EMT-related process. A genomic analysis of human lung cancers revealed a negative link between the EMT and IFN-γ signaling, while compared to human lung adenocarcinoma A549 cells, IFN-γ-hyporesponsive AS2 cells exhibited mesenchymal characteristics. Chemically, physically, and genetically engineered EMT attenuated IFN-γ-induced IFN regulatory factor 1 transactivation. Poststimulation of transforming growth factor-ß induced the EMT and also selectively retarded IFN-γ-responsive gene expression as well as IFN-γ-induced signal transducer and activator of transcription 1 activation, major histocompatibility complex I, and CD54 expression, cell migration/invasion inhibition, and direct/indirect cytotoxicity. Without changes in IFN-γ receptors, excessive oxidative activation of Src homology-2 containing phosphatase 2 (SHP2) in cells undergoing the EMT primarily caused cellular hyporesponsiveness to IFN-γ signaling and cytotoxicity, while combining an SHP2 inhibitor or antioxidant sensitized EMT-associated AS2 and mesenchymal A549 cells to IFN-γ-induced priming effects on tumor necrosis factor-related apoptosis-inducing ligand cytotoxicity. In cell line-derived xenograft models, combined treatment with IFN-γ and an SHP2 inhibitor induced enhanced anticancer activities. These results imply that EMT-associated SHP2 activation inhibits IFN-γ signaling, facilitating lung cancer cell escape from IFN-γ immunosurveillance.


Asunto(s)
Interferón gamma , Neoplasias Pulmonares , Línea Celular Tumoral , Movimiento Celular/inmunología , Transición Epitelial-Mesenquimal/inmunología , Humanos , Vigilancia Inmunológica , Interferón gamma/inmunología , Interferón gamma/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/patología
20.
J Inflamm Res ; 15: 1717-1728, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35282270

RESUMEN

Background: Infection with dengue virus (DENV) causes hematological complications in dengue diseases characterized by thrombocytopenia accompanied by macrophage activation syndrome and hemophagocytosis in fatal patients. Methods: In this study, we investigate the undefined mechanisms underlying the progression of thrombocytopenia caused by thrombophagocytosis based on an ex vivo whole-blood co-culture model of DENV infection for mimicking the acute febrile phase of infection. Results: In this model, complete blood count test showed a decrease in monocytes (p < 0.01), but not neutrophils nor other white blood cells, accompanied by a low thrombocyte count (p < 0.01) in DENV infection with a positive correlation (r = 0.636, p < 0.05). Furthermore, DENV exposure caused significant thrombophagocytosis in mononuclear cells (p < 0.05). Abnormal production of tumor necrosis factor (TNF)-α was highly associated with induction of thrombophagocytosis (r = 0.758, p < 0.01), decreased monocytes (r = -0.758, p < 0.01), and decreased thrombocyte (r = -0.728, p < 0.01). Neutralizing TNF-α considerably (p < 0.05) reversed such DENV-induced effects and was further validated by immunostaining-based flow cytometry analysis on mononuclear CD14 positive monocytes. Exogenous administration of TNF-α effectively caused thrombophagocytosis accompanied by decreased monocytes and thrombocytes, probably causing monocyte activation. Conclusion: These results demonstrate the potential pathogenesis of thrombocytopenia caused by TNF-α-induced thrombophagocytosis in monocytes during DENV infection.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA