Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Int J Biol Macromol ; 261(Pt 2): 129640, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38262553

RESUMEN

The study aims to fabricate MUF/paraffin microcapsules with lignin nanoparticles (LNPs)/ melamine-urea-formaldehyde (MUF) resin as hybrid shell material with different LNPs addition were synthesized in oil-in-water emulsion stabilized synergistically by styrene/maleic anhydride (SMA) and LNPs. The morphological characterization of LNPs was observed by transmission electron microscopy (TEM). The particle size of LNPs, the mean particle size and ξ potentials of SMA/LNPs mixture at pH =4.5 were investigated by zeta potential measurement. Field emission scanning electron microscopy (FE-SEM), Fourier-transform infrared spectroscopy (FT-IR), X-ray diffraction (XRD), thermogravimetric analyzer (TGA), and differential scanning calorimetry (DSC) were characterized the morphologies, crystallography, chemical component, thermal stability and phase change properties of MUF/paraffin microcapsules with different LNPs addition. The results showed that MUF/paraffin microcapsules were spherical. The LNPs did not influence the chemical structure or crystal type of MUF/paraffin microcapsules. When the LNPs addition was 0.15 g, the melting enthalpy and crystallization enthalpy is respectively 130.03 and 121.92 J/g and the encapsulation efficiency of MicroC-15 is 61.04 %.


Asunto(s)
Lignina , Parafina , Triazinas , Cápsulas/química , Urea , Espectroscopía Infrarroja por Transformada de Fourier , Formaldehído/química
2.
Leukemia ; 37(3): 593-605, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36631623

RESUMEN

Epigenetic regulators are frequently mutated in hematological malignancies including acute myeloid leukemia (AML). Thus, the identification and characterization of novel epigenetic drivers affecting AML biology holds potential to improve our basic understanding of AML and to uncover novel options for therapeutic intervention. To identify novel tumor suppressive epigenetic regulators in AML, we performed an in vivo short hairpin RNA (shRNA) screen in the context of CEBPA mutant AML. This identified the Histone 3 Lysine 4 (H3K4) demethylase KDM5C as a tumor suppressor, and we show that reduced Kdm5c/KDM5C expression results in accelerated growth both in human and murine AML cell lines, as well as in vivo in Cebpa mutant and inv(16) AML mouse models. Mechanistically, we show that KDM5C act as a transcriptional repressor through its demethylase activity at promoters. Specifically, KDM5C knockdown results in globally increased H3K4me3 levels associated with up-regulation of bivalently marked immature genes. This is accompanied by a de-differentiation phenotype that could be reversed by modulating levels of several direct and indirect downstream mediators. Finally, the association of KDM5C levels with long-term disease-free survival of female AML patients emphasizes the clinical relevance of our findings and identifies KDM5C as a novel female-biased tumor suppressor in AML.


Asunto(s)
Histona Demetilasas , Leucemia Mieloide Aguda , Animales , Femenino , Humanos , Ratones , Diferenciación Celular , Línea Celular , Relevancia Clínica , Histona Demetilasas/genética , Leucemia Mieloide Aguda/genética
3.
Int Immunopharmacol ; 111: 109100, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35932614

RESUMEN

Atherosclerosis (AS) has been regarded as an autoimmune disease. However, studies on immunotherapy against AS are limited. We previously found that IgG in AS patients serum binding to alpha 5 and 6 chain of collagen VI (COL6A5 or COL6A6) was significantly higher than that in healthy subjects, here we tried to identify whether they are AS-protective, and tried to develop human antibodies against them. ApoE-/- mice were immunized with COL6A5 or COL6A6 and COL6A6 was found a protective antigen against atherosclerosis. A phage display human single-chain antibody (scFv) library was constructed and COL6A6-specific scFv was obtained, and cloned into a modified pcDNA3 vector to express full-length human antibodies. ApoE-/- mice were fed a high-fat diet (HFD) for 20 weeks and administered three weekly injections of CVI monoclonal antibody (mAb) or isotype control antibody, CVI mAb was found to be able to reduce plaque area by 45 % via aorta oil red O staining. Flowcytometry method predicted that CVI mAb induced monocyte/macrophage polarization from M1 to M2. Furthermore, CVI mAb induced decreases of pro-inflammatory cytokines of MCP-1and IL-1ß, and increases of IL-4 and IL-10 levels in animal serum by using theLuminexassay. Overall, we found a novel atherosclero-related antigen - Collagen VI, and its protective fragment - Collagen VI alpha 6 chain (COL6A6) and proved that humanized antibody against COL6A6 therapy regresses atherosclerosis and induces monocyte/macrophage polarization from M1 to M2 in ApoE-/- mice animal model.


Asunto(s)
Aterosclerosis , Placa Aterosclerótica , Animales , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Aterosclerosis/metabolismo , Colágeno/metabolismo , Modelos Animales de Enfermedad , Humanos , Inflamación , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/metabolismo
4.
Cardiovasc Diabetol ; 21(1): 93, 2022 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-35659300

RESUMEN

OBJECTIVE: Type 2 diabetes mellitus (T2DM) is often accompanied by undiagnosed dyslipidemia. Research on the association of unconventional lipid markers with prediabetes (pre-DM) and T2DM simultaneously is limited in coronary heart disease (CHD) patients. METHODS: This study included 28,476 patients diagnosed with CHD. Their lipid levels, including triglycerides (TG), total cholesterol (TC), high-density lipoprotein cholesterol (HDL-C), and low-density lipoprotein cholesterol (LDL-C), were measured, and non-traditional lipid parameters were calculated. The patients were divided into three groups based on the diabetic status including normoglycemic (NG), pre-DM, and T2DM. Multiple logistic regression was used to compare the association of TG/HDL-C and other non-traditional lipid parameters with pre-DM and T2DM. The tertiles of TG/HDL-C included T1 (TG/HDL-C < 1.10), T2 (1.10 ≤ TG/HDL-C ≤ 1.89) and T3 (TG/HDL-C > 1.89). Low and high TG/HDL-C was defined with sex-specific cutoff points. RESULTS: Multiple logistic regression results showed that the non-traditional lipid parameters, including non-HDL-C, LDL-C/HDL-C, TC/HDL-C, non-HDL-C/HDL-C and TG/HDL-C, were all correlated with the risk of pre-DM and T2DM. Meanwhile TG/HDL-C showed the strongest correlation (odds ratio [OR]: 1.19; 95% confidence interval [CI] 1.16-1.23), (OR: 1.36; 95% CI 1.33-1.39). When dividing TG/HDL-C into tertiles, using T1 as a reference, T3 was observed to have the highest association with both pre-DM and T2DM (OR: 1.60; 95% CI 1.48-1.74), (OR: 2.79; 95% CI 2.60-3.00). High TG/HDL-C was significantly associated with pre-DM and T2DM (OR: 1.69; 95% CI 1.52-1.88), (OR: 2.85; 95% CI 2.60-3.12). The association of TG/HDL-C with T2DM and pre-DM existed across different sex, age, smoking, and drinking statuses. CONCLUSION: Elevated non-traditional lipid parameters were significantly associated with pre-DM and T2DM in CHD patients, especially TG/HDL-C. High TG/HDL-C was the risk factor with a strong correlation with the risk of pre-DM and T2DM.


Asunto(s)
Enfermedad Coronaria , Diabetes Mellitus Tipo 2 , Estado Prediabético , Colesterol , HDL-Colesterol , LDL-Colesterol , Enfermedad Coronaria/complicaciones , Enfermedad Coronaria/diagnóstico , Enfermedad Coronaria/epidemiología , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/diagnóstico , Diabetes Mellitus Tipo 2/epidemiología , Femenino , Humanos , Masculino , Estado Prediabético/complicaciones , Estado Prediabético/diagnóstico , Estado Prediabético/epidemiología , Factores de Riesgo , Triglicéridos
5.
Apoptosis ; 26(11-12): 639-656, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34743246

RESUMEN

Resistance to epidermal growth factor receptor-tyrosin kinase inhibitors (TKIs, e.g. icotinib) remains a major clinical challenge. Non-small cell lung cancer patients with wild-type EGFR and/or K-RAS mutation are primary resistance to EGFR-TKIs. Berberine has been found to have potent anticancer activities via distinct molecular mechanism. In this study, we sought to investigate the therapeutic utility of BBR in combination with icotinib to overcome icotinib resistance in NSCLC cells, and explore the molecular mechanism of synergism of icotinib and BBR to EGFR-resistant NSCLC cells. We used the two EGFR-resistant NSCLC cell lines H460 and H1299 for testing the inhibitory effect of icotinib and/or BBR on them. Moreover, xenograft mouse model was applied for assessing the anti-tumor activities of BBR and icotinib in combination. Results showed that BBR and icotinib have a synergistic inhibitory effect on H460 and H1299 cells through induction of autophagic cell death and apoptosis. Accordingly, the anti-cancer effect of BBR plus icotinib was further confirmed in the NSCLC xenograft mouse models. Combination of BBR and icotinib significantly inhibited the protein expression and the activity of EGFR by inducing autophagic EGFR degradation. BBR plus icotinib resulted in intracellular ROS accumulation, which could mediated autophagy and apoptosis and involved in the suppression of cell migration and invasion. In conclusions, combination application of BBR and icotinib could be an effective strategy to overcome icotinib resistance in the treatment of NSCLC.


Asunto(s)
Muerte Celular Autofágica , Berberina , Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Animales , Apoptosis , Berberina/farmacología , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Línea Celular Tumoral , Éteres Corona , Receptores ErbB/genética , Receptores ErbB/metabolismo , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Ratones , Quinazolinas , Transducción de Señal
6.
Cell Oncol (Dordr) ; 44(1): 135-150, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32936421

RESUMEN

PURPOSE: Paclitaxel is an anticancer drug for the treatment of non-small cell lung cancer (NSCLC). However, drug-resistance remains a major problem. Honokiol is a natural component which has been found to exhibit anti-tumor activity. Paclitaxel and honokiol have been reported to be able to induce paraptosis. The aim of this study was to investigate whether honokiol can reverse paclitaxel resistance by inducing paraptosis in NSCLC cells. METHODS: NSCLC cell lines H1650 (paclitaxel-sensitive), H1299 and H1650/PTX (intrinsic and acquired paclitaxel-resistant, respectively) were used to assess the cytotoxic effects of paclitaxel and honokiol. Light and transmission electron microscopy were performed to detect cytoplasmic vacuolation. In vitro cell viability and clonogenic survival assays, as well as in vivo xenograft assays were conducted to test synergistic killing effects of paclitaxel and honokiol on NSCLC cells. Western blotting, flow cytometry and immunofluorescence were performed to evaluate paraptosis-regulating mechanisms. RESULTS: We found that combination treatment with paclitaxel and honokiol synergistically killed H1650, H1299 and H1650/PTX cells by inducing paraptosis, which is characterized by cytoplasmic vacuolation. Moreover, paclitaxel/honokiol treatment resulted in a significant growth delay in H1299 xenograft tumors that showed extensive cytoplasmic vacuolation. Mechanistically, proteasomal inhibition-mediated endoplasmic reticulum (ER) stress and unfolded protein responses leading to ER dilation, and the disruption of intracellular Ca2+ homeostasis and mitochondrial Ca2+ overload resulting in mitochondrial disfunction, were found to be involved in paclitaxel/honokiol-induced paraptosis. Cellular protein light chain 3 (LC3) may play an important role in paclitaxel/honokiol induced cytoplasmic vacuolation and NSCLC cell death. CONCLUSIONS: Combination of honokiol and paclitaxel may represent a novel strategy for the treatment of paclitaxel-resistant NSCLC.


Asunto(s)
Apoptosis , Compuestos de Bifenilo/farmacología , Carcinoma de Pulmón de Células no Pequeñas/patología , Lignanos/farmacología , Neoplasias Pulmonares/patología , Paclitaxel/farmacología , Animales , Apoptosis/efectos de los fármacos , Calcio/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/ultraestructura , Línea Celular Tumoral , Sinergismo Farmacológico , Estrés del Retículo Endoplásmico/efectos de los fármacos , Humanos , Neoplasias Pulmonares/ultraestructura , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Proteínas Asociadas a Microtúbulos/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Enzimas Activadoras de Ubiquitina/metabolismo , Vacuolas/efectos de los fármacos , Vacuolas/metabolismo
7.
Atherosclerosis ; 280: 99-108, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30500605

RESUMEN

BACKGROUND AND AIMS: It has been well established that ezetimibe blocks cholesterol absorption to prevent the negative effects of a high-fat diet in atherosclerosis. However, the exact mechanism is unknown. Here we use a transgenic zebrafish, which expresses different fluorescent proteins on either endothelial cells or granulocytes and macrophages, to explore the specific mechanism of ezetimibe and its role in reducing atherosclerosis-related hypercholesteremia. METHODS: Zebrafish larvae were exposed to a control diet, high cholesterol diet (HCD) or a HCD with ezetimibe treatment. Both the control diet and high cholesterol diet were mixed with red or green fluorophore labeled cholesteryl ester to trace lipid distribution. Isobaric tags were used for relative and absolute quantification to examine protein expression profiles of zebrafish larvae in the different treatment groups. To knock down Apo A-II and investigate the role of Apo A-II in the anti-atherosclerotic function of ezetimibe, we used morpholinos to target zebrafish Apoa2 mRNA. To confirm ezetimibe regulatory role on Apo A-II expression, siRNA against HNF4, PPARα, and SREBP1 were transfected into HepG2 cells. RESULTS: The results show that ezetimibe increased the expression of Apo A-II but failed to reduce vascular lipid accumulation and macrophage recruitment induced by the HCD diet when Apo A-II was knocked down. Finally, we found that ezetimibe increased the expression of Apo A-II through HNF4 and PPARα transcriptional factors. CONCLUSIONS: Our data indicates that ezetimibe may not only prevents atherosclerosis by inhibiting cholesterol absorption in the intestine, but also by increasing the expression of Apo A-II in hepatocytes, thereby enhancing reverse cholesterol transport and removing excess cholesterol from the periphery.


Asunto(s)
Anticolesterolemiantes/farmacología , Apolipoproteína A-II/sangre , Aterosclerosis/metabolismo , Colesterol/metabolismo , Ezetimiba/farmacología , Animales , Animales Modificados Genéticamente , Dieta Alta en Grasa/efectos adversos , Células Endoteliales/metabolismo , Granulocitos/metabolismo , Células Hep G2 , Factor Nuclear 4 del Hepatocito/metabolismo , Hepatocitos , Humanos , Hipercolesterolemia/metabolismo , Metabolismo de los Lípidos , Lípidos/química , Macrófagos/metabolismo , Microscopía Confocal , PPAR alfa/metabolismo , Células THP-1 , Pez Cebra
8.
Mol Med Rep ; 18(1): 447-454, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29749519

RESUMEN

When bone resorption, aided by the activity of osteoclasts, exceeds bone formation induced by osteoblasts, bone metabolism loses equilibration, which results in the development of bone diseases, including osteoporosis. Pulsed electromagnetic fields (PEMFs) are known to be involved in various biological processes, including cell proliferation, differentiation and apoptosis. However, the exact mechanism of action of osteoclasts remains poorly understood. In the present study, the effects of PEMFs on osteoclast differentiation and associated signaling pathways were systematically investigated in RAW264.7 macrophages. RAW264.7 cells were induced by receptor activator of nuclear factor­κB ligand (RANKL) to obtain osteoclasts in vitro. The results of the present study demonstrated that PEMF exposure decreased osteoclast formation, limited tartrate­resistant acid phosphatase activity, contracted bone resorption area and inhibited osteoclastic specific gene and protein expression. Furthermore, western blot analysis indicated that PEMFs distinctly abolished the upregulation of phosphorylated­protein kinase B (Akt), ­mammalian target of rapamycin (mTOR) and ­ribosome S6 protein kinase (p70S6K) induced by RANKL, which was consistent with the effects of pharmacological inhibitor perifosine and rapamycin. Therefore, the present study suggested that PEMFs reduced osteoclast formation from RAW264.7 macrophages via inhibition of the Akt/mTOR signaling pathway. These findings provided novel insight into the mechanisms through which PEMFs suppress osteoclast differentiation.


Asunto(s)
Diferenciación Celular , Campos Electromagnéticos , Osteoclastos/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Animales , Ratones , Osteoclastos/citología , Ligando RANK/metabolismo , Células RAW 264.7
9.
Sci Rep ; 7(1): 15031, 2017 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-29118324

RESUMEN

The combination of platinum and gemcitabine is one of the standard regimens in the treatment of advanced lung squamous carcinoma (LSC). Resistance to gemcitabine is main barrier to the successful treatment of LSC. In this study, we showed that suppression of the Fanconi anemia (FA) pathway increased the sensitivity of two LSC cell lines SK-MES-1 and KLN205 to gemcitabine. Moreover, we found that the CHK1 pathway and the FA pathway are functionally compensatory in the repair of DNA damage in the LSC cell lines. Inactivation of one of the two pathways led to DNA damage, triggering compensatory activation of other pathway. Furthermore, we demonstrated that FANCD2 depletion combined with CHK1 inhibitor MK-8776 significantly potentiated the cytotoxicity of gemcitabine to the two LSC cell lines, compared to individual FANCD2 depletion or MK-8776 treatment. The enhanced effect of gemcitabine-chemosensitization was accompanied by loss of DNA repair function and accumulation of DNA single strand breaks and double strand breaks, in parallel with obvious increase of caspase-3 dependent apoptosis. Our results indicate that the enhancement effect of FANCD2 depletion combined with CHK1 inhibitor in sensitizing the LCS cells to gemcitabine supports the FA pathway and CHK1 as two therapeutic targets for improvement of anti-tumor regimens in treatment of LSC.


Asunto(s)
Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/antagonistas & inhibidores , Desoxicitidina/análogos & derivados , Proteínas del Grupo de Complementación de la Anemia de Fanconi/metabolismo , Pirazoles/farmacología , Pirimidinas/farmacología , Transducción de Señal/efectos de los fármacos , Células A549 , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/metabolismo , Desoxicitidina/farmacología , Sinergismo Farmacológico , Proteínas del Grupo de Complementación de la Anemia de Fanconi/genética , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Inhibidores de Proteínas Quinasas/farmacología , Interferencia de ARN , Transducción de Señal/genética , Gemcitabina
10.
J Cell Physiol ; 232(12): 3433-3443, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28075014

RESUMEN

It has been demonstrated that REV3, the catalytic subunit of the translesion synthesis (TLS) polymerase ζ, play an important role in DNA damage response (DDR) induced by cisplatin, and Ataxia-telangietasia mutated and Rad-3-related (ATR) knase is a central player in activating cell cycle checkpoint, stabilizing replication forks, regulating DDR, and promoting repair of DNA damage caused by cisplatin. Cancer cells deficient in either one of REV3 and ATR are more sensitive to cisplatin. However, whether co-inhibition of REV3 and ATR can further increase sensitivity of non-small cell lung cancer (NSCLC) cells to cisplatin is not clear. In this study, we show that REV3 knockdown combined with ATR inhibition further enhance cytotoxicity of cisplatin in NSCLC cells, including cisplatin-sensitive and -resistant cell lines, compared to individual knockdown of REV3 or ATR, which are accompanied by markedly caspase-dependent apoptosis response, pronounced DNA damage accumulation and severe impediment of interstrand crosslink (ICL), and double strand break (DSB) repair. Our results suggest that REV3 knockdown synergize strongly with ATR inhibition to significantly increase sensitivity of cisplatin in NSCLC cells by inhibiting ICL and DSB repair. Thus simultaneously targeting REV3 and ATR may represent one approach to overcome cisplatin resistance and improve chemotherapeutic efficacy in NSCLC treatment.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Cisplatino/farmacología , Proteínas de Unión al ADN/metabolismo , ADN Polimerasa Dirigida por ADN/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Células A549 , Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/enzimología , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Roturas del ADN de Doble Cadena , Proteínas de Unión al ADN/genética , ADN Polimerasa Dirigida por ADN/genética , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo , Resistencia a Antineoplásicos , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Interferencia de ARN , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Transfección
11.
J Cell Mol Med ; 21(5): 929-940, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-27995732

RESUMEN

oxLDL peptide vaccine and its antibody adoptive transferring have shown a significantly preventive or therapeutic effect in atherosclerotic animal model. The molecular mechanism behind this is obscure. Here, we report that oxLDL induces MCP-1 release in monocytes/macrophages through their TLR-4 (Toll-like receptor 4) and ERK MAPK pathway and is calcium/potassium channel-dependent. Using blocking antibodies against CD36, TLR-4, SR-AI and LOX-1, only TLR-4 antibody was found to have an inhibitory effect and ERK MAPK-specific inhibitor (PD98059) was found to have a dramatic inhibitory effect compared to inhibitors of other MAPK group members (p38 and JNK MAPKs) on oxLDL-induced MCP-1 release. The release of cytokines and chemokines needs influx of extracellular calcium and imbalance of efflux of potassium. Nifedipine, a voltage-dependent calcium channel (VDCC) inhibitor, and glyburide, an ATP-regulated potassium channel (K+ATP ) inhibitor, inhibit oxLDL-induced MCP-1 release. Potassium efflux and influx counterbalance maintains the negative potential of macrophages to open calcium channels, and our results suggest that oxLDL actually induces the closing of potassium influx channel - inward rectifier channel (Kir ) and ensuing the opening of calcium channel. ERK MAPK inhibitor PD98059 inhibits oxLDL-induced Ca2+ /Kir channel alterations. The interfering of oxLDL-induced MCP-1 release by its monoclonal antibody is through its FcγRIIB (CD32). Using blocking antibodies against FcγRI (CD64), FcγRIIB (CD32) and FcγRIII (CD16), only CD32 blocking antibody was found to reverse the inhibitory effect of oxLDL antibody on oxLDL-induced MCP-1 release. Interestingly, oxLDL antibody specifically inhibits oxLDL-induced ERK MAPK activation and ensuing Ca2+ /Kir channel alterations, and MCP-1 release. Thus, we found a molecular mechanism of oxLDL antibody on inhibition of oxLDL-induced ERK MAPK pathway and consequent MCP-1 release.


Asunto(s)
Anticuerpos/inmunología , Canales de Calcio/metabolismo , Quimiocina CCL2/metabolismo , Lipoproteínas LDL/inmunología , Macrófagos/metabolismo , Monocitos/metabolismo , Canales de Potasio/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Calcio/metabolismo , Humanos , Sistema de Señalización de MAP Quinasas , Macrófagos/efectos de los fármacos , Ratones , Modelos Biológicos , Monocitos/efectos de los fármacos , Células RAW 264.7 , Receptores de IgG/metabolismo , Receptor Toll-Like 4/metabolismo
12.
Int J Cancer ; 139(11): 2553-69, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27529686

RESUMEN

Voltage-gated sodium channels (VGSCs), which are aberrantly expressed in several human cancers, affect cancer cell behavior; however, their role in gastric cancer (GC) and the link between these channels and tumorigenic signaling remain unclear. The aims of this study were to determine the clinicopathological significance and role of the VGSC Nav 1.7 in GC progression and to investigate the associated mechanisms. Here, we report that the SCN9A gene encoding Nav 1.7 was the most abundantly expressed VGSC subtype in GC tissue samples and two GC cell lines (BGC-823 and MKN-28 cells). SCN9A expression levels were also frequently found to be elevated in GC samples compared to nonmalignant tissues by real-time PCR. In the 319 GC specimens evaluated by immunohistochemistry, Nav 1.7 expression was correlated with prognosis, and transporter Na(+) /H(+) exchanger-1 (NHE1) and oncoprotein metastasis-associated in colon cancer-1 (MACC1) expression. Nav 1.7 suppression resulted in reduced voltage-gated sodium currents, decreased NHE1 expression, increased extracellular pH and decreased intracellular pH, and ultimately, reduced invasion and proliferation rates of GC cells and growth of GC xenografts in nude mice. Nav 1.7 inhibition led to reduced MACC1 expression, while MACC1 inhibition resulted in reduced NHE1 expression in vitro and in vivo. Mechanistically, the suppression of Nav 1.7 decreased NF-κB p65 nuclear translocation via p38 activation, thus reducing MACC1 expression. Downregulation of MACC1 decreased c-Jun phosphorylation and subsequently reduced NHE1 expression, whereas the addition of hepatocyte growth factor (HGF), a c-Met physiological ligand, reversed the effect. These results indicate that Nav 1.7 promotes GC progression through MACC1-mediated upregulation of NHE1. Therefore, Nav 1.7 is a potential prognostic marker and/or therapeutic target for GC.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Canal de Sodio Activado por Voltaje NAV1.7/metabolismo , Intercambiadores de Sodio-Hidrógeno/metabolismo , Neoplasias Gástricas/metabolismo , Factores de Transcripción/metabolismo , Animales , Proteínas de Transporte de Catión/biosíntesis , Proteínas de Transporte de Catión/genética , Línea Celular Tumoral , Progresión de la Enfermedad , Femenino , Técnicas de Silenciamiento del Gen , Factor de Crecimiento de Hepatocito/metabolismo , Xenoinjertos , Humanos , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Desnudos , Canal de Sodio Activado por Voltaje NAV1.7/biosíntesis , Canal de Sodio Activado por Voltaje NAV1.7/genética , FN-kappa B/metabolismo , Invasividad Neoplásica , Proteínas Proto-Oncogénicas c-jun/metabolismo , Proteínas Proto-Oncogénicas c-met/metabolismo , Intercambiador 1 de Sodio-Hidrógeno , Intercambiadores de Sodio-Hidrógeno/biosíntesis , Intercambiadores de Sodio-Hidrógeno/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Transactivadores , Regulación hacia Arriba
13.
Am J Chin Med ; 44(5): 1027-41, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27430907

RESUMEN

Inhaled corticosteroids (ICS) are widely used to manage chronic obstructive pulmonary disease (COPD). However, withdrawal of ICS generally causes various adverse effects, warranting careful management of the ICS withdrawal. Pinellia ternata, a traditional Chinese herbal medicine, has been used to treat respiratory diseases in China for centuries. Here, we investigated its role in antagonizing ICS withdrawal-induced side effects, and explored the underlying mechanisms. The rat COPD model was established using a combination of passive cigarette smoking and intratracheal instillation of lipopolysaccharide (LPS). COPD rats were treated with saline or budesonide inhalation, or with budesonide inhalation followed by saline inhalation or Pinellia ternata gavage. The number of goblet cells and the level of mucin 5AC (MUC5AC) were enhanced by budesonide withdrawal. Pinellia ternata treatment significantly blocked these effects. Further, Pinellia ternata treatment reversed budesonide withdrawal-induced increase of interleukin 1[Formula: see text] (IL-1[Formula: see text] and tumor necrosis factor [Formula: see text] (TNF-[Formula: see text]) levels in bronchoalveolar lavage fluid (BALF). Extracellular signal-regulated kinase (ERK), but neither p38 nor c-Jun N-terminal kinase (JNK), was activated by budesonide withdrawal, and the activation was blocked by Pinellia ternata treatment. The MUC5AC expression was positively correlated with goblet cell number, IL-1[Formula: see text] and TNF-[Formula: see text] levels, and ERK activity. Pinellia ternata treatment protected the airway from ICS withdrawal-induced mucus hypersecretion and airway inflammation by inhibiting ERK activation. Pinellia ternata treatment may represent a novel therapeutic strategy to prevent ICS withdrawal-induced side effects in COPD patients.


Asunto(s)
Corticoesteroides/efectos adversos , Medicamentos Herbarios Chinos/administración & dosificación , Moco/efectos de los fármacos , Pinellia/química , Enfermedad Pulmonar Obstructiva Crónica/tratamiento farmacológico , Síndrome de Abstinencia a Sustancias/tratamiento farmacológico , Administración por Inhalación , Corticoesteroides/administración & dosificación , Animales , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/inmunología , Humanos , Interleucina-1/genética , Interleucina-1/inmunología , Pulmón/efectos de los fármacos , Pulmón/inmunología , Masculino , Mucina 5AC/genética , Mucina 5AC/inmunología , Moco/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/inmunología , Ratas , Ratas Sprague-Dawley , Síndrome de Abstinencia a Sustancias/etiología , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología
14.
Cell Oncol (Dordr) ; 39(6): 511-522, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27473273

RESUMEN

PURPOSE: Cisplatin can cause a variety of DNA crosslink lesions including intra-strand and inter-strand crosslinks (ICLs), which are associated with the sensitivity of cancer cells to cisplatin. Here, we aimed to assess the contribution of the Fanconi anemia (FA), homologous recombination (HR) and nucleotide excision repair (NER) pathways to cisplatin resistance in non-small cell lung cancer (NSCLC)-derived cells. METHODS: The expression of FA, HR and NER pathway-associated genes was assessed by RT-qPCR and Western blotting. siRNAs were used to knock down the expression of these genes. CCK-8 and flow cytometry assays were used to assess the viability and apoptotic rate of NSCLC-derived cells, respectively. Immunofluorescence and alkaline comet assays were used to assess the repair of ICLs. RESULTS: We found that acquired cisplatin-resistant NSCLC-derived A549/DR cells exhibited markedly enhanced FA and HR repair pathway capacities compared to its parental A549 cells and another independent NSCLC-derived cell line, Calu-1, which possesses a moderate innate resistance to cisplatin. siRNA-mediated silencing of the FA-associated genes FANCL and RAD18 and the HR-associated genes BRCA1 and BRCA2 significantly potentiated the sensitivity of A549/DR cells to cisplatin compared to A549 and Calu-1 cells, suggesting that the acquired cisplatin resistance in A549/DR cells may be attributed to enhanced FA and HR pathway capacities responsible for ICL repair. Although we found that expression knockdown of the NER-associated genes XPA and ERCC1 sensitized the three NSCLC-derived cell lines to cisplatin, the sensitization effect was more significant in Calu-1 cells than in A549 and A549/DR cells, implying that the innate cisplatin resistance in Calu-1 cells may result from an increased NER activity. CONCLUSIONS: Our results indicate that the functional status of DNA repair pathways determine the sensitivity of NSCLC cells to cisplatin. Direct targeting of the pathway that is involved in cisplatin resistance may be an effective strategy to surmount cisplatin resistance in NSCLC.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/genética , Reparación del ADN/genética , Resistencia a Antineoplásicos/genética , Neoplasias Pulmonares/genética , Antineoplásicos/uso terapéutico , Western Blotting , Línea Celular Tumoral , Cisplatino/uso terapéutico , Ensayo Cometa , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Técnicas de Silenciamiento del Gen , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA