Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 80(2): 263-278.e7, 2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-33022274

RESUMEN

Cancer metastasis accounts for the major cause of cancer-related deaths. How disseminated cancer cells cope with hostile microenvironments in secondary site for full-blown metastasis is largely unknown. Here, we show that AMPK (AMP-activated protein kinase), activated in mouse metastasis models, drives pyruvate dehydrogenase complex (PDHc) activation to maintain TCA cycle (tricarboxylic acid cycle) and promotes cancer metastasis by adapting cancer cells to metabolic and oxidative stresses. This AMPK-PDHc axis is activated in advanced breast cancer and predicts poor metastasis-free survival. Mechanistically, AMPK localizes in the mitochondrial matrix and phosphorylates the catalytic alpha subunit of PDHc (PDHA) on two residues S295 and S314, which activates the enzymatic activity of PDHc and alleviates an inhibitory phosphorylation by PDHKs, respectively. Importantly, these phosphorylation events mediate PDHc function in cancer metastasis. Our study reveals that AMPK-mediated PDHA phosphorylation drives PDHc activation and TCA cycle to empower cancer cells adaptation to metastatic microenvironments for metastasis.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/patología , Ciclo del Ácido Cítrico , Complejo Piruvato Deshidrogenasa/metabolismo , Animales , Dominio Catalítico , Línea Celular Tumoral , Supervivencia Celular , Activación Enzimática , Femenino , Humanos , Ratones Endogámicos BALB C , Ratones Desnudos , Metástasis de la Neoplasia , Fosforilación , Fosfoserina/metabolismo , Transducción de Señal , Estrés Fisiológico , Análisis de Supervivencia
2.
Mol Cell ; 58(6): 989-1000, 2015 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-26051179

RESUMEN

The regulation of RagA(GTP) is important for amino-acid-induced mTORC1 activation. Although GATOR1 complex has been identified as a negative regulator for mTORC1 by hydrolyzing RagA(GTP), how GATOR1 is recruited to RagA to attenuate mTORC1 signaling remains unclear. Moreover, how mTORC1 signaling is terminated upon amino acid stimulation is also unknown. We show that the recruitment of GATOR1 to RagA is induced by amino acids in an mTORC1-dependent manner. Skp2 E3 ligase drives K63-linked ubiquitination of RagA, which facilitates GATOR1 recruitment and RagA(GTP) hydrolysis, thereby providing a negative feedback loop to attenuate mTORC1 lysosomal recruitment and prevent mTORC1 hyperactivation. We further demonstrate that Skp2 promotes autophagy but inhibits cell size and cilia growth through RagA ubiquitination and mTORC1 inhibition. We thereby propose a negative feedback whereby Skp2-mediated RagA ubiquitination recruits GATOR1 to restrict mTORC1 signaling upon sustained amino acid stimulation, which serves a critical mechanism to maintain proper cellular functions.


Asunto(s)
Aminoácidos/farmacología , Proteínas de Unión al GTP Monoméricas/metabolismo , Complejos Multiproteicos/metabolismo , Proteínas Quinasas Asociadas a Fase-S/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Autofagia/genética , Línea Celular Tumoral , Activación Enzimática/efectos de los fármacos , Retroalimentación Fisiológica/efectos de los fármacos , Guanosina Trifosfato/metabolismo , Células HEK293 , Humanos , Immunoblotting , Lisina/metabolismo , Lisosomas/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Ratones Noqueados , Microscopía Confocal , Modelos Biológicos , Células 3T3 NIH , Unión Proteica/efectos de los fármacos , Interferencia de ARN , Proteínas Quinasas Asociadas a Fase-S/genética , Ubiquitinación/efectos de los fármacos
3.
Cell Tissue Bank ; 23(3): 417-440, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35000046

RESUMEN

The application of 3D printing technologies fields for biological tissues, organs, and cells in the context of medical and biotechnology applications requires a significant amount of innovation in a narrow printability range. 3D bioprinting is one such way of addressing critical design challenges in tissue engineering. In a more general sense, 3D printing has become essential in customized implant designing, faithful reproduction of microenvironmental niches, sustainable development of implants, in the capacity to address issues of effective cellular integration, and long-term stability of the cellular constructs in tissue engineering. This review covers various aspects of 3D bioprinting, describes the current state-of-the-art solutions for all aforementioned critical issues, and includes various illustrative representations of technologies supporting the development of phases of 3D bioprinting. It also demonstrates several bio-inks and their properties crucial for being used for 3D printing applications. The review focus on bringing together different examples and current trends in tissue engineering applications, including bone, cartilage, muscles, neuron, skin, esophagus, trachea, tympanic membrane, cornea, blood vessel, immune system, and tumor models utilizing 3D printing technology and to provide an outlook of the future potentials and barriers.


Asunto(s)
Bioimpresión , Huesos , Tinta , Impresión Tridimensional , Ingeniería de Tejidos , Andamios del Tejido
4.
JBMR Plus ; 8(5): ziae031, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38606146

RESUMEN

Nonunion resulting from early bone resorption is common after bone transplantation surgery. In these patients, instability or osteoporosis causes hyperactive catabolism relative to anabolism, leading to graft resorption instead of fusion. Systemic zoledronate administration inhibits osteoclastogenesis and is widely used to prevent osteoporosis; however, evidence on local zoledronate application is controversial due to osteoblast cytotoxicity, uncontrolled dosing regimens, and local release methods. We investigated the effects of zolendronate on osteoclastogenesis and osteogenesis and explored the corresponding signaling pathways. In vitro cytotoxicity and differentiation of MC3T3E1 cells, rat bone marrow stromal cells (BMSCs) and preosteoclasts (RAW264.7 cells) were evaluated with different zolendronate concentrations. In vivo bone regeneration ability was tested by transplanting different concentrations of zolendronate with ß-tricalcium phosphate (TCP) bone substitute into rat femoral critical-sized bone defects. In vitro, zolendronate concentrations below 2.5 × 10-7 M did not compromise viability in the three cell lines and did not promote osteogenic differentiation in MC3T3E1 cells and BMSCs. In RAW264.7 cells, zoledronate inhibited extracellular regulated protein kinases and c-Jun n-terminal kinase signaling, downregulating c-Fos and NFATc1 expression, with reduced expression of fusion-related dendritic cell­specific transmembrane protein and osteoclast-specific Ctsk and tartrate-resistant acid phosphatase (. In vivo, histological staining revealed increased osteoid formation and neovascularization and reduced fibrotic tissue with 500 µM and 2000 µM zolendronate. More osteoclasts were found in the normal saline group after 6 weeks, and sequential osteoclast formation occurred after zoledronate treatment, indicating inhibition of bone resorption during early callus formation without inhibition of late-stage bone remodeling. In vivo, soaking ß-TCP artificial bone with 500 µM or 2000 µM zoledronate is a promising approach for bone regeneration, with potential applications in bone transplantation.

5.
J Mater Chem B ; 12(26): 6394-6409, 2024 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-38855886

RESUMEN

This study develops a composite bone graft of CaO-MgO-SiO2 glass-ceramic and CaSO4 [abbreviated as (CMS)3-x(CS)x] via the sponge replication technique with weight fractions of x = 0, 1, 1.5, 2, and 3. The (CMS)1.5(CS)1.5 composite displays a superior degradability and, a suitable compressive strength of ∼3 MPa, and excellent cell proliferation and differentiation. The in vivo rat femur test in the hybrid-pore (CMS)1.5(CS)1.5 composite granules achieves a higher rate of bone formation, which is ∼2.7 times better than that of the commercial HAP/ß-TCP at 12 weeks. Improved expressions of osteocyte and mature osteocyte marker genes, namely (Spp1, Dmp1, and Fgf23), were observed in the (CMS)1.5(CS)1.5 group, indicating a faster differentiation into mature bone tissue. The ions release of (CMS)1.5(CS)1.5 through the ERK1/2 signaling pathway promotes osteogenic differentiation. The high bone generation rate can be attributed to faster active ions release and modified surface topography. This work highlights an excellent bone graft candidate for clinical applications in orthopedic surgery.


Asunto(s)
Cerámica , Osteogénesis , Cerámica/química , Animales , Osteogénesis/efectos de los fármacos , Ratas , Diferenciación Celular/efectos de los fármacos , Compuestos de Calcio/química , Ratas Sprague-Dawley , Proliferación Celular/efectos de los fármacos , Óxidos/química , Dióxido de Silicio/química , Masculino , Sustitutos de Huesos/química , Sustitutos de Huesos/farmacología , Trasplante Óseo/métodos , Óxido de Magnesio/química , Propiedades de Superficie , Fémur
6.
J Biol Chem ; 287(39): 32512-24, 2012 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-22859294

RESUMEN

Annexin A2 (ANXA2) overexpression is required for cancer cell proliferation; however, the molecular mechanisms underlying ANXA2-mediated regulation of the cell cycle are still unknown. ANXA2 is highly expressed in non-small cell lung cancer (NSCLC) and is positively correlated with a poor prognosis. NSCLC A549 cells lacking ANXA2 exhibited defects in tumor growth in vivo and in cell proliferation in vitro without cytotoxicity. ANXA2 knockdown induced cell cycle arrest at G(2) phase. Unexpectedly, ANXA2 silencing increased the expression of p53 and its downstream genes, which resulted in p53-dependent and -independent G(2) arrest. Aberrant JNK inactivation, which was observed in ANXA2-deficient cells, inhibited cell proliferation following G(2) arrest. A lack of ANXA2 caused a loss of JNK-regulated c-Jun expression, resulting in an increase in p53 transcription. These results demonstrate a novel role for ANXA2 in NSCLC cell proliferation by facilitating the cell cycle partly through the regulation of p53 via JNK/c-Jun.


Asunto(s)
Anexina A2/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Puntos de Control de la Fase G2 del Ciclo Celular , Silenciador del Gen , Neoplasias Pulmonares/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Anexina A2/genética , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Técnicas de Silenciamiento del Gen , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , MAP Quinasa Quinasa 4/genética , MAP Quinasa Quinasa 4/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteínas Proto-Oncogénicas c-jun/genética , Proteínas Proto-Oncogénicas c-jun/metabolismo , Transcripción Genética/genética , Proteína p53 Supresora de Tumor/genética
7.
Sci Total Environ ; 904: 166911, 2023 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-37689187

RESUMEN

Atmospheric fine particulate matter (PM2.5) is a human health risk factor, but its ambient concentration depends on both precursor emissions and meteorology. While emission reductions are used to set PM2.5-related health policies, the effect of meteorology is often overlooked. To explore this aspect, we examined PM2.5 interannual variability (IAV) associated with meteorological parameters using the long-term simulation from the Community Earth System Model (CESM1), a global climate-chemistry model, with fixed emissions. The results are subsequently contrasted with the MERRA-2 reanalysis dataset, which inherently considers emission and meteorology effects. Over continental East Asia, the CESM1 domain-average PM2.5 IAV is 6.7 %, mainly attributed to humidity, precipitation, and ventilation variation. The grid-cell PM2.5 IAVs over southern East China are larger, up to 12 % due to the more substantial influence of El Niño-induced meteorological anomalies. Under such climate extreme, sub-regional PM2.5 concentration may occasionally exceed WHO air quality guideline levels despite the compliance of the long-term mean. The simulated PM2.5 IAV over continental East Asia is ~25 % of that derived from the MERRA-2 data, which highlights the influence of both emission and meteorology-driven variations and trends inherent in the latter. Although emission-driven variability is significant to PM2.5 IAV, in remote areas downwind of major source regions in East Asia, North America, and Western Europe, the MERRA-2 data revealed that meteorological variations contributed more to PM2.5 IAV than emission variations. Thus, when setting policies for complying with the WHO PM2.5-related air quality guideline levels, the highest annual PM2.5 associated with climate extremes should be considered instead of that based on average climate conditions.

8.
Spine J ; 23(2): 315-324, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36058516

RESUMEN

BACKGROUND CONTEXT: Pedicle screw-rod assembly procedures following pedicle screw insertion include contouring and placing rods into screw tulips, introducing set screws into the tulip along the screw thread, applying a counter-torque holder and tightening all the set screws clockwise. Even if an appropriate pedicle screw is implanted, screw dislodgement after tightening of the tulip and set screw is not uncommon. Pedicle wall violation resulting from excessive rotational force due to inadequate use of a counter-torque holder might be the reason. However, the strain change in the pedicle during tulip-set screw tightening and the role of counter-torque have never been investigated. PURPOSE: This study determined differences in the strain change in the outer and inner pedicle walls during tulip-set screw tightening; additionally, the influence of counter-torque on pedicle wall violation was elucidated. STUDY DESIGN: A controlled biomechanical study; the strain values of outer and inner pedicle walls in cadaveric porcine L4-L5 vertebrae during tulip-set screw tightening with or without a counter-torque holder were measured. METHODS: Twelve L4-L5 fresh-frozen porcine lumbar vertebrae were implanted with screw-rod constructs; the set screw was randomly locked into the tulip in the right L5, right L4, left L5 and left L4 testing groups. The maximal values from eight strain gauges (P-R-O: outer cortex of right pedicle in proximal vertebra; P-R-I: inner cortex of right pedicle in proximal vertebra; D-R-O: outer cortex of right pedicle in distal vertebra; D-R-I: inner cortex of right pedicle in distal vertebra; P-L-O: outer cortex of left pedicle in proximal vertebra; P-L-I: inner cortex of left pedicle in proximal vertebra; D-L-O: outer cortex of left pedicle in distal vertebra; D-L-I: outer cortex of left pedicle in proximal vertebra) for each specimen during tightening to 12 Nm were measured. RESULTS: The maximal strain values of the ipsilateral strain gauges in all testing groups were almost significantly higher when a counter-torque holder was not used than when one was used. The strain values in the adjacent pedicle of specimens without a counter-torque holder were significantly increased: P-R-O and P-R-I in the right L5 group; D-R-I in the right L4 group; P-L-I and P-L-O in the left L5 group; D-L-O and D-L-I in the left L4 group. CONCLUSIONS: The constraint effect of counter-torque during tulip-set screw tightening is necessary. Clockwise rotational force with a fragile lateral pedicle wall suggests that caution is required when using a counter-torque holder to tighten the right L5 and left L4 constructs. CLINICAL SIGNIFICANCE: A counter-torque holder is important during tulip-set screw tightening; improper use may lead to adjacent pedicle wall violation, sequentially resulting in pedicle screw loosening.


Asunto(s)
Tornillos Pediculares , Fusión Vertebral , Animales , Fenómenos Biomecánicos , Vértebras Lumbares/cirugía , Fusión Vertebral/métodos , Porcinos , Torque
9.
J Cell Physiol ; 227(6): 2775-87, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21928315

RESUMEN

Annexin A2 (p36) is usually present together with its natural ligand p11 as a heterotetramer complex, which has multiple biological functions depending on its cellular localization. However, the detailed mechanism of annexin A2 translocation and its physiological role in inflammation remain unclear. Here, we show that IFN-γ stimulation enhances surface translocation of annexin A2 on lung epithelial cells. While total annexin A2 protein remains unchanged, the expression of p11 is upregulated via the IFN-γ-activated JAK2/STAT1 signal pathway. Notably, IFN-γ-induced p11 expression is required for annexin A2 translocation to the cell surface. Since annexin A2 lacks a signal peptide for surface translocation by the classical endoplasmic reticulum-Golgi route, its mode of trafficking remains unclear. We observed that p11-dependent surface translocation of annexin A2 is associated with the exosomal secretion pathway. The IFN-γ-induced increase of annexin A2 in the exosomes is blocked in p11-silenced cells. Furthermore, IFN-γ-induced surface expression of annexin A2 mediates phagocytosis of apoptotic cells by lung epithelial cells. These findings provide insights into the surface translocation mechanism of annexin A2 and illustrate a pivotal function of surface annexin A2 in the phagocytic response to IFN-γ.


Asunto(s)
Anexina A2/metabolismo , Células Epiteliales/metabolismo , Exosomas/metabolismo , Interferón gamma/metabolismo , Pulmón/metabolismo , Fagocitosis , Proteínas S100/metabolismo , Anexina A2/genética , Membrana Celular/metabolismo , Técnicas de Cocultivo , Células Epiteliales/inmunología , Células Epiteliales/patología , Células HEK293 , Humanos , Janus Quinasa 2/metabolismo , Células Jurkat , Pulmón/inmunología , Pulmón/patología , Transporte de Proteínas , Interferencia de ARN , Proteínas S100/genética , Factor de Transcripción STAT1/metabolismo , Transducción de Señal , Factores de Tiempo , Transfección , Regulación hacia Arriba
10.
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
11.
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
12.
Membranes (Basel) ; 12(3)2022 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-35323807

RESUMEN

N-(2-hydroxy) propyl-3-trimethylammonium chitosan chloride (HTCC) is a type of quaternary ammonium chitosan derivative with an antibacterial activity superior to the pristine chitosan, but its electrospinnability is limited. In this study, polyvinyl alcohol (PVA) was blended with HTCC to improve the electrospinnability of nanofibers. The electrospinning of PVA-HTCC nanofiber membranes was optimized in terms of structural stability and antimicrobial performance. Based on scanning electron microscopic analysis, the morphology and diameter of the produced nanofibers were influenced by the applied voltage, flow rate of the feed solution, and weight ratio of the polymer blend. An increase in the HTCC content decreased the average nanofiber diameter. The maximum water solubility of the PVA-HTCC nanofibers reached the maximum value of 70.92% at 12 h and 25 °C. The antibacterial activity of PVA-HTCC nanofiber membranes against Escherichia coli was ~90%, which is significantly higher than that of PVA-chitosan nanofiber membrane. Moreover, the antibacterial efficiency of PVA-HTCC nanofiber membranes remained unaffected after 5 cycles of antibacterial treatment. The good antibacterial performance and biocompatibility of PVA-HTCC nanofiber membrane makes them attractive for biomedical and biochemical applications that necessitate sterile conditions.

13.
J Biol Chem ; 285(37): 28715-22, 2010 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-20592027

RESUMEN

Autophagy is regulated for IFN-gamma-mediated antimicrobial efficacy; however, its molecular effects for IFN-gamma signaling are largely unknown. Here, we show that autophagy facilitates IFN-gamma-activated Jak2-STAT1. IFN-gamma induces autophagy in wild-type but not in autophagy protein 5 (Atg5(-/-))-deficient mouse embryonic fibroblasts (MEFs), and, autophagy-dependently, IFN-gamma induces IFN regulatory factor 1 and cellular inflammatory responses. Pharmacologically inhibiting autophagy using 3-methyladenine, a known inhibitor of class III phosphatidylinositol 3-kinase, confirms these effects. Either Atg5(-/-) or Atg7(-/-) MEFs are, independent of changes in IFN-gamma receptor expression, resistant to IFN-gamma-activated Jak2-STAT1, which suggests that autophagy is important for IFN-gamma signal transduction. Lentivirus-based short hairpin RNA for Atg5 knockdown confirmed the importance of autophagy for IFN-gamma-activated STAT1. Without autophagy, reactive oxygen species increase and cause SHP2 (Src homology-2 domain-containing phosphatase 2)-regulated STAT1 inactivation. Inhibiting SHP2 reversed both cellular inflammation and the IFN-gamma-induced activation of STAT1 in Atg5(-/-) MEFs. Our study provides evidence that there is a link between autophagy and both IFN-gamma signaling and cellular inflammation and that autophagy, because it inhibits the expression of reactive oxygen species and SHP2, is pivotal for Jak2-STAT1 activation.


Asunto(s)
Autofagia/fisiología , Fibroblastos/metabolismo , Interferón gamma/metabolismo , Janus Quinasa 2/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Factor de Transcripción STAT1/metabolismo , Adenina/análogos & derivados , Adenina/farmacología , Animales , Proteína 5 Relacionada con la Autofagia , Proteína 7 Relacionada con la Autofagia , Células Cultivadas , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Inhibidores Enzimáticos/farmacología , Fibroblastos/citología , Técnicas de Silenciamiento del Gen , Inflamación/genética , Inflamación/metabolismo , Interferón gamma/genética , Janus Quinasa 2/genética , Lentivirus , Ratones , Proteínas Asociadas a Microtúbulos/genética , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Factor de Transcripción STAT1/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
14.
Biochem Biophys Res Commun ; 412(2): 334-40, 2011 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-21821001

RESUMEN

Galectin-3 is regulated for cancer cell survival and apoptosis depending upon the cell type and stimulus. We investigated a glycogen synthase kinase (GSK)-3ß/galectin-3-regulated mechanism used by leukemia cells to escape from apoptotic stimuli. Galectin-3 expression was time- and transcription-dependently deregulated in K562 chronic myeloid leukemia cells stimulated for apoptosis by cisplatin (a platinum-based chemotherapy drug), sphingolipid ceramide analog C(2)-ceramide, and LY294002 (a phosphatidylinositol 3-kinase inhibitor). Notably, galectin-3 was upregulated in survival cells. Forced galectin-3 expression caused resistance to apoptosis, whereas knockdown galectin-3 expression increased susceptibility to apoptosis. Sub-cellular distribution of inducible galectin-3 was mitochondria-specific. Apoptotic stimuli decreased pro-survival Bcl-2 family protein expression (especially Mcl-1), whereas galectin-3 overexpression reversed but it was enhanced by a galectin-3 expression knockdown. Under apoptotic stimulation, GSK-3ß was activated after Akt was inactivated and GSK-3ß was inhibited-either pharmacologically or using short hairpin RNA to abolish galectin-3, increase apoptosis, and inhibit colony formation-which suggests a pro-survival role for GSK-3ß. We found that GSK-3ß upregulated galectin-3 and stabilized anti-apoptotic Bcl-2 family proteins, which is important for the escape of leukemia cells from apoptotic stimuli.


Asunto(s)
Apoptosis , Resistencia a Antineoplásicos , Galectina 3/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Línea Celular Tumoral , Supervivencia Celular , Cromonas/farmacología , Galectina 3/genética , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3 beta , Humanos , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Mitocondrias/metabolismo , Morfolinas/farmacología , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Tiadiazoles/farmacología , Transcripción Genética , Proteína bcl-X/metabolismo
15.
J Immunol ; 183(2): 856-64, 2009 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-19542364

RESUMEN

Glycogen synthase kinase-3beta (GSK-3beta)-modulated IFN-gamma-induced inflammation has been reported; however, the mechanism that activates GSK-3beta and the effects of activation remain unclear. Inhibiting GSK-3beta decreased IFN-gamma-induced inflammation. IFN-gamma treatment rapidly activated GSK-3beta via neutral sphingomyelinase- and okadaic acid-sensitive phosphatase-regulated dephosphorylation at Ser(9), and proline-rich tyrosine kinase 2 (Pyk2)-regulated phosphorylation at Tyr(216). Pyk2 was activated through phosphatidylcholine-specific phospholipase C (PC-PLC)-, protein kinase C (PKC)-, and Src-regulated pathways. The activation of PC-PLC, Pyk2, and GSK-3beta was potentially regulated by IFN-gamma receptor 2-associated Jak2, but it was independent of IFN-gamma receptor 1. Furthermore, Jak2/PC-PLC/PKC/cytosolic phospholipase A(2) positively regulated neutral sphingomyelinase. Inhibiting GSK-3beta activated Src homology-2 domain-containing phosphatase 2 (SHP2), thereby preventing STAT1 activation in the late stage of IFN-gamma stimulation. All these results showed that activated GSK-3beta synergistically affected IFN-gamma-induced STAT1 activation by inhibiting SHP2.


Asunto(s)
Glucógeno Sintasa Quinasa 3/fisiología , Interferón gamma/farmacología , Proteína Tirosina Fosfatasa no Receptora Tipo 11/antagonistas & inhibidores , Factor de Transcripción STAT1/metabolismo , Animales , Quinasa 2 de Adhesión Focal/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Inflamación/tratamiento farmacológico , Janus Quinasa 2/metabolismo , Ratones , Fosforilación , Receptores de Interferón/metabolismo , Transducción de Señal , Fosfolipasas de Tipo C/metabolismo
16.
Biomed Mater ; 16(5)2021 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-34410226

RESUMEN

Poly(methyl methacrylate) (PMMA) has been widely used in orthopedic applications, but bone ingrowth and toxic monomer release are drawback of this material. Particle reinforcement with osteoconductive substitute, such as calcium sulfate (CaSO4), is one of the solutions used to modify PMMA bone cement. The current study investigated the mechanical, chemical and biological properties of CaSO4-augmented bone cement. Mechanical strength was measured by a material testing machine. The concentration of methyl methacrylate (MMA) monomer from the various formulations of PMMA mixed with CaSO4was measured by ultra-performance liquid chromatography (UPLC). CCK-8 assay and ALP assay were performed to evaluate cytotoxicity of released MMA monomer and cell differentiation. The attachment of cells to CaSO4-augmented bone cement discs was observed by confocal and scanning electron microscopy, and surface topography was also evaluated by atomic force microscopy. The results revealed that increased CaSO4weight ratios led to compromised mechanical strength and increased MMA monomer release. Cell density and cell differentiation on CaSO4-augmented bone cement discs were decreased at CaSO4weight ratios above 10%. In addition, the presence of micropores on the surface and surface roughness were both increased for PMMA composite discs containing higher levels of CaSO4. These results demonstrated that fewer MC3T3-E1 cells on the surface of CaSO4-PMMA composites was correlated to increased MMA monomer release, micropore number and surface roughness. In summary, the augmentation of a higher proportion of CaSO4(>10 wt. %) to PMMA did not promote the biological properties of traditional PMMA bone cement.


Asunto(s)
Cementos para Huesos , Sulfato de Calcio , Adhesión Celular/efectos de los fármacos , Polimetil Metacrilato , Animales , Cementos para Huesos/química , Cementos para Huesos/farmacología , Cementos para Huesos/toxicidad , Sulfato de Calcio/química , Sulfato de Calcio/farmacología , Sulfato de Calcio/toxicidad , Línea Celular , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ratones , Osteoblastos/efectos de los fármacos , Polimetil Metacrilato/química , Polimetil Metacrilato/farmacología , Polimetil Metacrilato/toxicidad , Propiedades de Superficie
17.
Int J Biol Macromol ; 181: 508-520, 2021 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-33775766

RESUMEN

This study aimed to develop a novel electrospun polyacrylonitrile (PAN) nanofiber membrane with the enhanced antibacterial property. The PAN nanofiber membrane was first subjected to alkaline hydrolysis treatment, and the treated membrane was subsequently grafted with chitosan (CS) to obtain a CS-modified nanofiber membrane (P-COOH-CS). The modified membrane was then coupled with different dye molecules to form P-COOH-CS-Dye membranes. Lastly, poly(hexamethylene biguanide) hydrochloride (PHMB) was immobilized on the modified membrane to produce P-COOH-CS-Dye-PHMB. Physical characterization studies were conducted on all the synthesized nanofiber membranes. The antibacterial efficacies of nanofiber membranes prepared under different synthesis conditions were evaluated systematically. Under the optimum synthesis conditions, P-COOH-CS-Dye-PHMB was highly effective in disinfecting a high concentration of Escherichia coli, with an antibacterial efficacy of approximately 100%. Additionally, the P-COOH-CS-Dye-PHMB exhibited an outstanding wash durability as its antibacterial efficacy was only reduced in the range of 5%-7% even after 5 repeated cycles of treatment. Overall, the experimental results of this study suggested that the P-COOH-CS-Dye-PHMB is a promising antibacterial nanofiber membrane that can be adopted in the food, pharmaceutical, and textile industries.


Asunto(s)
Antibacterianos/farmacología , Biguanidas/farmacología , Quitosano/química , Colorantes/química , Membranas Artificiales , Nanofibras/química , Antibacterianos/síntesis química , Antibacterianos/química , Biguanidas/síntesis química , Biguanidas/química , Escherichia coli/efectos de los fármacos , Concentración de Iones de Hidrógeno , Cinética , Pruebas de Sensibilidad Microbiana , Espectroscopía Infrarroja por Transformada de Fourier
18.
Carbohydr Polym ; 262: 117910, 2021 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-33838797

RESUMEN

N-[(2-hydroxyl-3-trimethylammonium) propyl] chitosan chloride (HTCC), which is a type of chitosan derivative with quaternary ammonium groups, possesses a higher antibacterial activity as compared to the pristine chitosan. The nanofiber membranes made of HTCC are attractive for applications demanding for antibacterial function. However, the hydrophilic nature of HTCC makes it unsuitable for electrospinning of nanofibers. Hence, biodegradable polyvinyl alcohol (PVA) was proposed as an additive to improve the electrospinnability of HTCC. In this work, PVA/HTCC nanofiber membrane was crosslinked with the blocked diisocyanate (BI) to enhance the stability of nanofiber membrane in water. Microbiological assessments showed that the PVA/HTCC/BI nanofiber membranes possessed a good antibacterial efficacy (∼100 %) against E. coli. Moreover, the biocompatibility of PVA/HTCC/BI nanofiber membrane was proven by the cytotoxicity test on mouse fibroblasts. These promising results indicated that the PVA/HTCC/BI nanofiber membrane can be a promising material for food packaging and as a potential wound dressing for skin regeneration.


Asunto(s)
Antibacterianos/farmacología , Quitosano/química , Isocianatos/química , Nanofibras/química , Alcohol Polivinílico/química , Animales , Antibacterianos/química , Vendajes , Línea Celular , Quitosano/análogos & derivados , Reactivos de Enlaces Cruzados/química , Escherichia coli/efectos de los fármacos , Embalaje de Alimentos/métodos , Interacciones Hidrofóbicas e Hidrofílicas , Ratones , Compuestos de Amonio Cuaternario/química
19.
Mater Sci Eng C Mater Biol Appl ; 124: 112060, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33947554

RESUMEN

This work reports a new CaO-MgO-SiO2 (CMS) bioactive glass-ceramic, using ZrO2 as a nucleus to modulate the ratios of glass and ceramic phases as a function of sintering temperature. Mg-rich bioactive CMS glass-ceramics exhibit advantages regarding mechanical strength (flexural strength ~190 MPa and compressive strength ~555 MPa), in-vitro and in-vivo biocompatibilities, and bone ingrowth. The high mechanical strengths could be attributed to the CaMgSi2O6 glass-ceramic and lower porosity. X-ray absorption spectra indicate an increased SiO covalent bond via the development of CaMgSi2O6 glass-ceramics. From the in-vitro cytotoxicity and BMSC differentiation assays, the CMS samples sintered above 800 °C exhibited better cell attachment and differentiation, possibly due to structural stability, appropriate pore, and ion release to boost osteogenesis. Compared to hydroxyapatite (HA) ceramics, the CMS glass-ceramics display higher mechanical strengths, biocompatibility, and osteoconductivity. An in-vivo experiment demonstrated a fine bone-ingrowth profile around the CMS implant. This study may further the application of CMS glass-ceramics in bone implants.


Asunto(s)
Óxido de Magnesio , Dióxido de Silicio , Cerámica , Durapatita , Vidrio
20.
BMC Immunol ; 11: 50, 2010 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-20939898

RESUMEN

BACKGROUND: Patients infected with Vibrio vulnificus (V. vulnificus) show severe inflammatory responses characterised by the upregulation of proinflammatory cytokines. Macrophage migration inhibitory factor (MIF), an upstream proinflammatory regulator, increases the inflammation caused by sepsis. Whether MIF regulates responses to V. vulnificus infection and the actual mechanism by which V. vulnificus initiates these MIF-modulated proinflammatory cytokines remain unclear. RESULTS: MIF increased inflammation during V. vulnificus infection in vivo. In V. vulnificus-infected mice, MIF was produced earlier than tumour necrosis factor (TNF)-α and interleukin (IL)-6 and was expressed in a time-dependent manner. ISO-1 ((S, R)-3-(4-hydroxyphenyl)-4,5-dihydro-5-isoxazole acetic acid methyl ester), a small-molecule inhibitor of MIF, significantly decreased IL-6, IL-8, and TNF-α production in a time- and dose-dependent manner in human peripheral blood cells infected with V. vulnificus. The induction of IL-6, IL-8, and TNF-α production by V. vulnificus infection was mediated via the NF-κB- and p38 MAPK-regulated pathways but not via the Akt pathway. ISO-1-treated human peripheral blood cells showed lower V. vulnificus-induced NF-κB activation, IL-6 mRNA expression, and IκB phosphorylation, but they did not show lower p38 MAPK activation. CONCLUSIONS: We conclude that MIF regulates V. vulnificus-induced IL-6 production via NF-κB activation and that p38 MAPK activation in V. vulnificus infection is not MIF dependent.


Asunto(s)
Interleucina-6/biosíntesis , Oxidorreductasas Intramoleculares/metabolismo , Leucocitos Mononucleares/metabolismo , Factores Inhibidores de la Migración de Macrófagos/metabolismo , FN-kappa B/metabolismo , Vibriosis/inmunología , Vibrio vulnificus/inmunología , Animales , Células Cultivadas , Femenino , Humanos , Interleucina-6/genética , Interleucina-6/metabolismo , Oxidorreductasas Intramoleculares/antagonistas & inhibidores , Oxidorreductasas Intramoleculares/genética , Oxidorreductasas Intramoleculares/inmunología , Isoxazoles/farmacología , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/patología , Leucocitos Mononucleares/virología , Factores Inhibidores de la Migración de Macrófagos/antagonistas & inhibidores , Factores Inhibidores de la Migración de Macrófagos/genética , Factores Inhibidores de la Migración de Macrófagos/inmunología , Ratones , Ratones Endogámicos BALB C , FN-kappa B/inmunología , Activación Transcripcional/efectos de los fármacos , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Vibriosis/genética , Vibriosis/metabolismo , Vibrio vulnificus/patogenicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA