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

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Angew Chem Int Ed Engl ; 63(25): e202405791, 2024 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-38593214

RESUMEN

Herein, we present catalyst-regulated switchable site-selective hydrosilylation of enynes, which are suitable for a wide range of alkyl and aryl substituted polar enynes and exhibit excellent functional group compatibility. Under the optimized conditions, silyl groups can be precisely installed at various positions of 1,3-dienes. While α- and γ-silylation products were obtained under platinum-catalytic systems, ß-silylation products were delivered with [Cp*RuCl]4 as catalyst. This process lead to the formation of 1,3-dienoates with diverse substitutions, which would pose challenges with other methodologies.

2.
Org Biomol Chem ; 21(31): 6307-6311, 2023 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-37492010

RESUMEN

A new method has been successfully developed that offers a facile and reliable approach for synthesizing (E)-2-(1-(methoxyimino)ethyl)-2-phenylbenzofuran-3(2H)-one, providing 28 compounds. This optimized process enables efficient preparation of a wide range of compounds using readily available (E)-1-(benzofuran-2-yl)ethan-1-one O-methyl oxime and iodobenzene, and provides alternative ideas for the structural modification of benzofuran ketones.

3.
Adv Mater ; 36(2): e2306860, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37703533

RESUMEN

Halide perovskites are crystalline semiconductors with exceptional optoelectronic properties, rapidly developing toward large-scale applications. Lead (II) (Pb2+ ) is the core element used to prepare halide perovskites. Pb2+ can displace key 2+ elements, including calcium, zinc and iron, that regulate vital physiological functions. Sn2+ can replace Pb2+ within the perovskite structure and, if accidentally dispersed in the environment, it readily oxidizes to Sn4+ , which is compatible with physiological functions and thus potentially safe. The 3+ salt bismuth (III) (Bi3+ ) is also potentially safe for the same reason and useful to prepare double perovskites. Here, this work studies the biotoxicity of Pb, Sn, and Bi perovskites in mice for the first time. This work analyses histopathology and growth of mice directly exposed to perovskites and investigate the development of their offspring generation. This study provides the screening of organs and key physiological functions targeted by perovskite exposure to design specific studies in mammalians.


Asunto(s)
Compuestos Inorgánicos , Plomo , Titanio , Animales , Ratones , Plomo/toxicidad , Compuestos de Calcio/toxicidad , Óxidos/toxicidad , Mamíferos
4.
J Surg Res ; 182(2): 303-11, 2013 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-23158409

RESUMEN

BACKGROUND: Shikonin, a natural naphthoquinone pigment extracted from the root of Lithospermum erythrorhizon, has shown a variety of pharmacologic properties including anti-inflammatory effect. In the present study, we analyzed the role of shikonin in acute lung injury induced by lipopolysaccharide (LPS) in mice. MATERIALS AND METHODS: Sixty male BALB/C mice were randomly allocated into six groups (n = 10, each): control group, shikonin group (50 mg/kg), LPS group, and three different doses (12.5, 25, and 50 mg/kg) for shikonin-treated groups. Shikonin or vehicle was given with an intragastric administration 1 h before an intratracheal instillation of LPS (5 mg/kg). The severity of pulmonary injury was evaluated 6 h after LPS challenge. RESULTS: Shikonin pretreatment significantly attenuated LPS-induced pulmonary histopathologic changes, alveolar hemorrhage, and neutrophil infiltration. The lung wet-to-dry weight ratios, as the index of pulmonary edema, were markedly decreased by shikonin pretreatment. Moreover, shikonin decreased the productions of the proinflammatory cytokines including tumor necrosis factor alpha and interleukin 1ß and the concentration of total proteins in the bronchoalveolar lavage fluid. Shikonin pretreatment also reduced the concentrations of myeloperoxidase and nitric oxide in lung tissues. In addition, shikonin pretreatment significantly suppressed LPS-induced activation of cyclooxygenase 2 and inducible nitric oxide synthase and the nuclear factor κB DNA-binding activity in lung tissues. CONCLUSIONS: This study indicates that shikonin may have a protective effect against LPS-induced acute lung injury, and the potential mechanism of this action may attribute partly to the inhibition of inducible nitric oxide synthase and cyclooxygenase 2 expression by downregulating nuclear factor κB activation.


Asunto(s)
Lesión Pulmonar Aguda/tratamiento farmacológico , Antiinflamatorios no Esteroideos/uso terapéutico , Medicamentos Herbarios Chinos/uso terapéutico , Naftoquinonas/uso terapéutico , Lesión Pulmonar Aguda/inducido químicamente , Animales , Líquido del Lavado Bronquioalveolar/química , Ciclooxigenasa 2/metabolismo , ADN/metabolismo , Lipopolisacáridos/toxicidad , Pulmón/metabolismo , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos BALB C , FN-kappa B/metabolismo , Óxido Nítrico/análisis , Óxido Nítrico Sintasa de Tipo II/metabolismo , Peroxidasa/metabolismo , Edema Pulmonar/tratamiento farmacológico
5.
Guang Pu Xue Yu Guang Pu Fen Xi ; 33(2): 494-7, 2013 Feb.
Artículo en Zh | MEDLINE | ID: mdl-23697140

RESUMEN

Microcystis aeruginosa is one of the most common species in the algae-bloom events of domestic lakes. Illumination incubator was used to cultivate M. aeruginosa under conditions of different phosphorus concentrations in the laboratory. Spectroscopic data of culture solutions were collected by GER1500 spectrometer under the sunlight. The study focused on the growth rhythm of M. aeruginosa and the characteristics of spectral variation in the culture solutions. The results showed that low phosphorus concentration (< or =10 microg x L(-1)) is a restricting factor for the growth and reproduction of M. aeruginosa. Moreover, the reflections of spectrum from culture solutions of M. aeruginosa showed significant changes along with cultivation period, such as at the wavelengths of 550, 610, 660, 700-710 and 760 nm.


Asunto(s)
Técnicas de Cultivo/métodos , Microcystis/crecimiento & desarrollo , Fósforo/farmacología , Análisis Espectral/métodos , Medios de Cultivo , Microcystis/citología , Fósforo/análisis
6.
Org Lett ; 25(40): 7332-7337, 2023 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-37782771

RESUMEN

We develop a copper-catalyzed disilylation of polysubstituted pent-1-en-4-yn-3-yl acetate derivatives, which in one pot furnishes the bis(silyl)-substituted vinylallenes in moderate yields under mild reaction conditions. The reaction shows broad substrate scope and single stereoselectivity. Besides, we develop a method to decrease the electrocyclization temperature of vinylallenes for the synthesis of methylenecyclobutenes by installing bis(silyl) substituents. And the effect of a silyl substituent on electrocyclization of vinylallenes was studied via DFT computation. The synthetic method features broad substrate scope and excellent stereoselectivity.

7.
Nat Commun ; 14(1): 5048, 2023 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-37598226

RESUMEN

We report a copper-catalyzed ligand-controlled selective 1,2- and 1,4-hydrosilylation of 1,3-enynes, which furnishes enantiomerically enriched propargyl- and 1,2-allenylsilane products in high yields with excellent enantioselectivities (up to 99% ee). This reaction proceeds under mild conditions, shows broad substrate scope for both 1,3-enynes and trihydrosilanes, and displays excellent regioselectivities. Mechanistic studies based on deuterium-labeling reactions and density functional theory (DFT) calculations suggest that allenylcopper is the dominant reactive intermediate under both 1,2- and 1,4-hydrosilylation conditions, and it undergoes metathesis with silanes via selective four-membered or six-membered transition state, depending on the nature of the ligand. The weak interactions between the ligands and the reacting partners are found to be the key controlling factor for the observed regioselectivity switch. The origin of high enantiocontrol in the 1,4-hydrosilylation is also revealed by high level DLPNO-CCSD(T) calculations.

8.
Ann Thorac Surg ; 113(4): e307-e310, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34270974

RESUMEN

For McKeown esophagectomy, hand-sewn and mechanical esophagogastric anastomosis techniques have been improved for constructing esophagogastrostomy. However, postoperative anastomosis-related complication rates remain high in patients undergoing cervical anastomosis. Here, we report an original and reliable hand-sewn cervical tunnel esophagogastric anastomosis technique to maximally reduce cervical anastomotic leakage and stricture rates after McKeown esophagectomy. The key features and innovations of cervical tunnel esophagogastric anastomosis are the right gastroepiploic artery as the center for the esophagogastric anastomosis to reduce ischemia, sufficient width of the anastomotic site for anastomosis without stricture, enfolding of the anastomotic site by the tunnel, and tension- and rotation-free anastomosis.


Asunto(s)
Neoplasias Esofágicas , Esofagectomía , Anastomosis Quirúrgica/métodos , Fuga Anastomótica/etiología , Fuga Anastomótica/prevención & control , Fuga Anastomótica/cirugía , Constricción Patológica/cirugía , Neoplasias Esofágicas/complicaciones , Esofagectomía/métodos , Humanos , Complicaciones Posoperatorias/etiología , Estudios Retrospectivos , Resultado del Tratamiento
9.
Transl Lung Cancer Res ; 11(3): 404-419, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35399566

RESUMEN

Background: The mechanisms involved in the malignant progression of lung adenocarcinoma (LUAD) are still inconclusive. Fibrinogen-like protein 1 (FGL1) and LAG3 are a pair of immune checkpoints that create an inhibitory immune microenvironment in tumors. However, other roles of FGL1 in LUAD have not been extensively studied. Our study aims to explore the role of FGL1 in the malignant progression of LUAD and to provide new therapeutic targets and strategies for LUAD treatment. Methods: Differential gene expression of FGL1 was analyzed using the Gene Expression Profiling Interactive Analysis (GEPIA), Oncomine, UALCAN, and Gene Expression Omnibus (GEO) databases. A pan-cancer analysis was conducted using the Oncomine, TIMER, and UALCAN databases. A total of 140 tumor tissues and paired normal tissues were collected, IHC and immunofluorescence staining were used to explore the expression of FGL1. GeneMANIA database and STRING database were used to analyze gene-gene interaction and protein-protein interaction, respectively. A mutation analysis was conducted using the cBioPortal database, and an immune infiltration analysis was conducted using the TIMER database. A survival analysis was carried out using the GEPIA and PrognoScan database. The knockdown of FGL1 was confirmed by western blot (WB) and immunofluorescence staining. Cell proliferation was tested by cell cycle analysis and real-time cell analysis. RNA sequencing (RNA-seq) was used to explore the differential genes of FGL1 knockdown in LUAD cells. Results: Multiple databases showed that FGL1 was highly expressed in LUAD. The results of IHC indicated that FGL1 was highly expressed in the cytoplasm of LUAD cells. FGL1 was negatively associated with immune infiltration in LUAD. The main mutation of FGL1 is deep deletion, the altered group and high expression group indicated poor prognosis. The downregulation of FGL1 lead to a significantly decreased percentage of PC9 cells in S phase, but had little effect on the proliferation of Jurkat T cells. RNA-seq and GSEA analysis indicated that the differential genes were mainly enriched in MYC-target genes, which suggested that the downregulation of FGL1 inhibited cell proliferation by regulating MYC-target genes. Conclusions: FGL1 exerts in LUAD proliferation in addition to immune regulation. The downregulation of FGL1 inhibits the proliferation of LUAD cells by regulating MYC-target genes. Thus, FGL1 may be a novel therapeutic target in LUAD.

10.
Front Immunol ; 13: 1014053, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36268014

RESUMEN

Rational: Lung cancer is the most common tumor worldwide, with the highest mortality rate and second highest incidence. Immunotherapy is one of the most important treatments for lung adenocarcinoma (LUAD); however, it has relatively low response rate and high incidence of adverse events. Herein, we explored the therapeutic potential of fibrinogen-like protein 1 (FGL1) for LUAD. Methods: Data from GEPIA and ACLBI databases were assessed to explore gene-gene correlations and tumor immune infiltration patterns. A total of 200 patients with LUAD were recruited. FGL1 levels in the serum and cellular supernatant were determined by enzyme-linked immunosorbent assay. In vitro and in vivo experiments were performed to assess the effect FGL1 on the proliferation of LUAD cells. Cocultures were performed to explore the effect of FGL1 knockdown in lung cancer cells on T cells, concerning cytokine secretion and viability. PROMO and hTFtarget databases were used for transcription factor prediction. Quantitative polymerase chain reaction (qPCR), chromatin immunoprecipitation, and dual luciferase reporter assays were performed to validate the identified transcription factor of FGL1. Immunoprecipitation, mass spectrometry and gene ontology analysis were performed to explore the downstream partners of FGL1. Results: FGL1 expression in LUAD was positively associated with PDL1, but not for PD1 expression. Moreover, FGL1 was positively associated with the CD3D expression and negatively associated with FOXP3, S100A9, and TPSB2 within the tumor site. FGL1 promotes the secretion of interleukin-2 by T cells in vitro, simultaneously inducing their apoptosis. Indeed, YY1 is the upstream molecule of FGL1 was found to be transcriptionally regulated by YY1 and to directly by to MYH9 to promote the proliferation of LUAD cells in vitro and in vivo. Conclusions: FGL1 is involved in the immunological and proliferative regulation of LUAD cells by controlling the secretion of important immune-related cytokines via the YY1-FGL1-MYH9 axis. Hence, targeting FGL1 in LUAD may pave the way for the development of new immunotherapies for tackling this malignancy.


Asunto(s)
Adenocarcinoma del Pulmón , Neoplasias Pulmonares , Humanos , Interleucina-2/metabolismo , Regulación Neoplásica de la Expresión Génica , Proliferación Celular/genética , Línea Celular Tumoral , Adenocarcinoma del Pulmón/genética , Adenocarcinoma del Pulmón/patología , Neoplasias Pulmonares/patología , Fibrinógeno/metabolismo , Factores de Transcripción Forkhead/metabolismo , Cadenas Pesadas de Miosina/genética , Cadenas Pesadas de Miosina/metabolismo , Factor de Transcripción YY1/genética , Factor de Transcripción YY1/metabolismo
11.
Biomark Res ; 10(1): 49, 2022 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-35831836

RESUMEN

Immunotherapy has become the major treatment for tumors in clinical practice, but some intractable problems such as the low response rate and high rates of immune-related adverse events still hinder the progress of tumor immunotherapy. Hence, it is essential to explore additional immunotherapy treatment targets. In this review, we focus on the structure, expression and expression-related mechanisms, interactions, biological functions and the progress in preclinical/clinical research of IGSF11 and VISTA in tumors. We cover the progress in recent research with this pair of immune checkpoints in tumor immune regulation, proliferation, immune resistance and predictive prognosis. Both IGSF11 and VISTA are highly expressed in tumors and are modulated by various factors. They co-participate in the functional regulation of immune cells and the inhibition of cytokine production. Besides, in the downregulation of IGSF11 and VISTA, both inhibit the growth of some tumors. Preclinical and clinical trials all emphasize the predictive role of IGSF11 and VISTA in the prognosis of tumors, and that the predictive role of the same gene varies from tumor to tumor. At present, further research is proving the enormous potential of IGSF11 and VISTA in tumors, and especially the role of VISTA in tumor immune resistance. This may prove to be a breakthrough to solve the current clinical immune resistance, and most importantly, since research has focused on VISTA but less on IGSF11, IGSF11 may be the next candidate for tumor immunotherapy.

12.
Cancers (Basel) ; 14(21)2022 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-36358792

RESUMEN

Cancer remains a serious social health problem, and immunotherapy has become the major treatments in tumor treatment. Additionally, improving the efficiency and safety of treatment is necessary. Further, more therapy targets are warranted for future tumor treatments. In this review, in addition to examining the currently recognized role of immune regulation, we focus on the proliferative role of 15 immune checkpoints in various tumors, including PD1, PD-L1, FGL1, CD155, CD47, SIRPα, CD276, IDO1, SIGLEC-15, TIM3, Galectin-9, CD70, CD27, 4-1BBL, and HVEM. We managed to conclude that various immune checkpoints such as PD1/PD-L1, FGL1, CD155, CD47/SIRPα, CD276, and SIGLEC-15 all regulate the cell cycle, and specifically through Cyclin D1 regulation. Furthermore, a variety of signal pathways engage in proliferation regulation, such as P13K, AKT, mTOR, and NK-κB, which are also the most common pathways involved in the regulation of immune checkpoint proliferation. Currently, only PD1/PD-L1, CD47/SIRPα, TIM3/Galectin-9, and CD70/CD27 checkpoints have been shown to interact with each other to regulate tumor proliferation in pairs. However, for other immune checkpoints, the role of their receptors or ligands in tumor proliferation regulation is still unknown, and we consider the enormous potential in this area. An increasing number of studies have validated the various role of immune checkpoints in tumors, and based on this literature review, we found that most of the immune checkpoints play a dual regulatory role in immunity and proliferation. Therefore, the related pathways in proliferation regulation can served the role of therapy targets in tumor therapy. Further, great potential is displayed by IDO1, SIGLEC-15, 4-1BBL, and HVEM in tumor proliferation regulation, which may become novel therapy targets in tumor treatment.

13.
Front Immunol ; 12: 785091, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35111155

RESUMEN

LAG3 is the most promising immune checkpoint next to PD-1 and CTLA-4. High LAG3 and FGL1 expression boosts tumor growth by inhibiting the immune microenvironment. This review comprises four sections presenting the structure/expression, interaction, biological effects, and clinical application of LAG3/FGL1. D1 and D2 of LAG3 and FD of FGL1 are the LAG3-FGL1 interaction domains. LAG3 accumulates on the surface of lymphocytes in various tumors, but is also found in the cytoplasm in non-small cell lung cancer (NSCLC) cells. FGL1 is found in the cytoplasm in NSCLC cells and on the surface of breast cancer cells. The LAG3-FGL1 interaction mechanism remains unclear, and the intracellular signals require elucidation. LAG3/FGL1 activity is associated with immune cell infiltration, proliferation, and secretion. Cytokine production is enhanced when LAG3/FGL1 are co-expressed with PD-1. IMP321 and relatlimab are promising monoclonal antibodies targeting LAG3 in melanoma. The clinical use of anti-FGL1 antibodies has not been reported. Finally, high FGL1 and LAG3 expression induces EGFR-TKI and gefitinib resistance, and anti-PD-1 therapy resistance, respectively. We present a comprehensive overview of the role of LAG3/FGL1 in cancer, suggesting novel anti-tumor therapy strategies.


Asunto(s)
Antígenos CD/inmunología , Fibrinógeno/inmunología , Neoplasias/inmunología , Animales , Antineoplásicos Inmunológicos/inmunología , Humanos , Ligandos , Microambiente Tumoral/inmunología
14.
Front Oncol ; 11: 693321, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34367975

RESUMEN

This study highlights aspects of the latest clinical research conducted on the relationship between immune checkpoints and tumor metastasis. The overview of each immune checkpoint is divided into the following three sections: 1) structure and expression; 2) immune mechanism related to tumor metastasis; and 3) clinical research related to tumor metastasis. This review expands on the immunological mechanisms of 17 immune checkpoints, including TIM-3, CD47, and OX-40L, that mediate tumor metastasis; evidence shows that most of these immune checkpoints are expressed on the surface of T cells, which mainly exert immunomodulatory effects. Additionally, we have summarized the roles of these immune checkpoints in the diagnosis and treatment of metastatic tumors, as these checkpoints are considered common predictors of metastasis in various cancers such as prostate cancer, non-Hodgkin lymphoma, and melanoma. Moreover, certain immune checkpoints can be used in synergy with PD-1 and CTLA-4, along with the implementation of combination therapies such as LIGHT-VTR and anti-PD-1 antibodies. Presently, most monoclonal antibodies generated against immune checkpoints are under investigation as part of ongoing preclinical or clinical trials conducted to evaluate their efficacy and safety to establish a better combination treatment strategy; however, no significant progress has been made regarding monoclonal antibody targeting of CD28, VISTA, or VTCN1. The application of immune checkpoint inhibitors in early stage tumors to prevent tumor metastasis warrants further evidence; the immune-related adverse events should be considered before combination therapy. This review aims to elucidate the mechanisms of immune checkpoint and the clinical progress on their use in metastatic tumors reported over the last 5 years, which may provide insights into the development of novel therapeutic strategies that will assist with the utilization of various immune checkpoint inhibitors.

15.
J Thorac Dis ; 13(2): 918-926, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33717564

RESUMEN

BACKGROUND: The trachea is the uppermost respiratory airway element connecting the larynx to the bronchi Airway reconstructions in humans are often developed from animal models but there is limited knowledge comparing tracheal biomechanics between species. We aimed to assess the structure and biomechanics of porcine, canine, caprine and human airways. METHODS: Tracheas from pigs (n=15), goats (n=9) and canines (n=9) were divided into three groups (4, 6 and 8-ringswhile human left principal brochi (n=12) were divided into two groups (3and-rings). Airway structures were compared using histology and scanning electron microscopy. Biomechanical properties were measured subjecting samples to uniaxial tension and compression, recording the elastic modulus and (tensile and compressive) strengths. RESULTS: The structures of animal tracheal and human bronchia appeared similar. Biomechanical testing revealed that the elastic modulus of 8-ring tracheas was 1,190.48±363.68, 2,572.00±608.19 and 1,771.27±145.54 kPa, for porcine, canine and caprine samples, respectively, while corresponding tensile strengths were 437.63±191.41, 808.38±223.48 and 445.76±44.00 kPa. Comparable measures of anterior-posterior (A-P) compression strengths were 7.94±0.82, 7.54±0.07 and 8.10±1.87 N, respectively, whereas lateral compression strengths were 8.75±0.82, 14.55±2.29 and 11.12±0.40 N. Compression testing of human samples showed significant differences (P<0.05) between the 3-ring (A-P, 1.06±0.02 N; lateral, 0.55±0.06 N) and 5-ring groups (A-P, 1.08±0.64 N; lateral, 2.32±1.95 N). CONCLUSIONS: The tensile and compressive strengths of mammalian airways show positive correlations with the cartilage ring number (length). On the basis of structural and biomechanical comparisons, porcine, canine and caprine species appear suitable models for the study of airway reconstruction in human.

16.
Transl Lung Cancer Res ; 10(12): 4617-4630, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35070765

RESUMEN

BACKGROUND: Primary tracheobronchial neoplasm is rare yet poses a serious threat to life. Due to its low incidence, the immune microenvironment of such tumors remained unclear. This study aimed to clarify the expression of programmed death-ligand 1 (PD-L1) and infiltration of immune cells in primary tracheobronchial neoplasm, which might be useful for guiding treatment and evaluating clinical outcome. METHODS: We assessed retrospectively the expression of PD-L1 and infiltration in cells expressing CD8, CD16, CD68, CD163 and FOXP3 in 21 patients with primary tracheobronchial neoplasm who underwent surgery in Tangdu Hospital from January 2016 to July 2021. The expression of PD-L1 was assessed based on the tumor proportion score system. The density of immune cells was analyzed by automatic image analysis software. RESULTS: In this study, all of 16 participants with adenoid cystic carcinoma (ACC) had no expression of PD-L1, whereas 4/5 (80%) of those with squamous cell carcinomas (SCC) were positive for PD-L1 expression. Compared with ACC, the density of FOXP3+ cells in both the intratumoral region and peritumoral region was higher in SCC (P<0.01). The density of FOXP3+ cells was significantly higher than that of CD8+, CD16+, and CD163+ cells in SCC in the intratumoral region (P<0.01). In contrast, the density of FOXP3+ cells was significantly lower than that of CD8+, CD16+, and CD68+ cells in ACC in both the intratumoral region and peritumoral regions. The density of CD68+ cells was significantly higher than that of CD8+ cells (P<0.05) and CD163+ cells (P<0.01) in ACC in the intratumoral region. Furthermore, the tumors of patients with metastasis more commonly of immune-excluded status, in which the CD8+ cells accumulated in peritumoral region. CONCLUSIONS: This study demonstrated that the expression of PD-L1 in primary tracheobronchial neoplasm was mainly concentrated in patients with SCC. In the immune microenvironment of SCC, FOXP3+ cells were the dominant immune cells, while in the immune microenvironment of ACC, CD68+ cells were the main immune cells. Therefore, the immune microenvironment was significantly different in primary tracheobronchial neoplasm according to histology.

17.
Respir Res ; 11: 33, 2010 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-20302656

RESUMEN

BACKGROUND: Histone deacetylase (HDAC) inhibitors, developed as promising anti-tumor drugs, exhibit their anti-inflammatory properties due to their effects on reduction of inflammatory cytokines. OBJECTIVE: To investigate the protective effect of butyrate, a HDAC inhibitor, on lipopolysaccharide (LPS)-induced acute lung injury (ALI) in mice. METHODS: ALI was induced in Balb/c mice by intratracheally instillation of LPS (1 mg/kg). Before 1 hour of LPS administration, the mice received butyrate (10 mg/kg) orally. The animals in each group were sacrificed at different time point after LPS administration. Pulmonary histological changes were evaluated by hematoxylin-eosin stain and lung wet/dry weight ratios were observed. Concentrations of interleukin (IL)-1beta and tumor necrosis factor (TNF)-alpha in bronchoalveolar lavage fluid (BALF) and concentrations of nitric oxide (NO) and myeloperoxidase (MPO) activity in lung tissue homogenates were measured by enzyme-linked immunosorbent assay (ELISA). Expression of nuclear factor (NF)-kappaB p65 in cytoplasm and nucleus was determined by Western blot analysis respectively. RESULTS: Pretreatment with butyrate led to significant attenuation of LPS induced evident lung histopathological changes, alveolar hemorrhage, and neutrophils infiltration with evidence of reduced MPO activity. The lung wet/dry weight ratios, as an index of lung edema, were reduced by butyrate administration. Butyrate also repressed the production of TNF-alpha, IL-1beta and NO. Furthermore, the expression of NF-kappaB p65 in nucleus was markedly suppressed by butyrate pretreatment. CONCLUSIONS: Butyrate had a protective effect on LPS-induced ALI, which may be related to its effect on suppression of inflammatory cytokines production and NF-kappaB activation.


Asunto(s)
Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/prevención & control , Butiratos/administración & dosificación , Modelos Animales de Enfermedad , Inhibidores de Histona Desacetilasas/administración & dosificación , Lipopolisacáridos , Lesión Pulmonar Aguda/metabolismo , Administración Oral , Animales , Citocinas/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Resultado del Tratamiento
18.
Biosci Biotechnol Biochem ; 74(4): 736-40, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20378992

RESUMEN

The development and progression of esophageal cancer is associated with multiple alterations in the genome, including loss of the tumor suppressor phosphatase and tensin homolog deleted from the chromosome 10 (PTEN) gene. The purpose of this study was to determine the effects of adenovirus-mediated MMAC/PTEN expression on the growth and survival of human esophageal cancer cells in vitro and in vivo. We found that compared to control cells, overexpression of PTEN significantly suppressed growth and induced apoptosis in esophageal cancer cell lines Eca-109 and TE-1 via downregulation of Bcl-2 expression and changes in cell-cycle progression. Adenovirus PTEN also inhibited the growth of subcutaneous tumor xenografts by significantly reducing tumor size in vivo. Thus our results confirm the proposed functional role of MMAC/PTEN as a regulator of esophageal cancer progression in vivo and in vitro. PTEN might be an important biological marker and potential therapeutic target in the treatment of human esophageal cancer.


Asunto(s)
Monoéster Fosfórico Hidrolasas/genética , Monoéster Fosfórico Hidrolasas/fisiología , Adenoviridae/genética , Adenoviridae/metabolismo , Infecciones por Adenoviridae/genética , Infecciones por Adenoviridae/terapia , Animales , Apoptosis/genética , Apoptosis/fisiología , Carcinoma/genética , Carcinoma/terapia , Línea Celular Tumoral , Regulación hacia Abajo , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/terapia , Genes Supresores de Tumor , Humanos , Ratones , Ratones Desnudos , Neoplasias/genética , Neoplasias/terapia , Fosfohidrolasa PTEN , Monoéster Fosfórico Hidrolasas/metabolismo , Distribución Aleatoria
19.
Turk J Gastroenterol ; 31(12): 948-954, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33626010

RESUMEN

BACKGROUND/AIMS: There is no consensus on treatment for cervical esophageal squamous cell carcinoma (ESCC). Our aim is to evaluate the feasibility and outcome of larynx-preserving limited resection with total thoracic esophagectomy and gastric pull-up reconstruction for the treatment of cervical ESCC without tumor involvement of the larynx and hypopharynx. MATERIALS AND METHODS: Retrospective analysis of patients with cervical ESCC who underwent R0 surgical resection from 2006 to 2011 in our center was performed. Kaplan-Meier method was used to calculate the survival time for patients. RESULTS: In total, 74 cervical ESCC patients were enrolled in the study. The mortality rate in 30 days was 8.1%, the total complication rate (at least one) was 47.3%, anastomosis leakage occurrence was 37.8%, mechanical ventilation ratewas12.2%, the rate of normal oral diet within 15 days was 71.6%, and the anastomosis recurrence rate in follow-up was 8.1%. Detailed analysis showed that the anastomosis leakage, pulmonary infection, laryngeal recurrent nerve injury, and chylothorax were the most common complications in surgical patients. Finally, the survival data showed that the median survival time was 31.83 months (95% CI=12.39-51.28 months) and the 3-year and 5-year survival rates were 49.1% and 35.5%, respectively. CONCLUSION: Larynx-preserving limited resection with total thoracic esophagectomy and gastric pull-up reconstruction might be a feasible and effective surgical alternative for the cervical ESCC patients whose tumor does not involve the larynx and hypopharynx.


Asunto(s)
Neoplasias Esofágicas/cirugía , Carcinoma de Células Escamosas de Esófago/cirugía , Esofagectomía/métodos , Laringe/cirugía , Tratamientos Conservadores del Órgano/métodos , Procedimientos de Cirugía Plástica/métodos , Anastomosis Quirúrgica , Neoplasias Esofágicas/patología , Carcinoma de Células Escamosas de Esófago/patología , Esófago/cirugía , Femenino , Humanos , Hipofaringe/patología , Laringe/patología , Masculino , Persona de Mediana Edad , Cuello/cirugía , Estudios Retrospectivos , Estómago/cirugía , Tasa de Supervivencia , Resultado del Tratamiento
20.
Onco Targets Ther ; 12: 10299-10309, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31819514

RESUMEN

PURPOSE: High metastasis is a leading risk factor for the survival of non-small cell lung cancer (NSCLC) and epithelial-mesenchymal transition (EMT) is a vital step of metastasis. The expression of novel oncogene with kinase domain (NOK) has been observed in some human malignancies, including non-small cell lung cancer (NSCLC); however, the biological function of NOK in NSCLC remains unclear. In the study, we explored the function of NOK in NSCLC, with an aim to elucidate the relevant underlying mechanisms. PATIENTS AND METHODS: We investigate the expression of NOK, p-Akt, p-GSK-3ß, E-cadherin and N-cadherin expression by immunohistochemical analysis using tissue microarrays of 72 paired NSCLC samples of cancerous and adjacent normal tissues. The associations between NOK expression and clinicopathological factors, overall survival, other proteins were assessed. Immunofluorescence analysis of NSCLC tissues was performed to study the location of NOK, Akt and GSK-3ß. Up or down-regulated of NOK were conducted in two NSCLC cell lines to analyze its impact on AKT/GSK3ß pathway. RESULTS: Statistical analysis revealed NOK expression increased in NSCLC tissues compared with normal tissues (P<0.05). It also showed that low NOK expression were associated with a higher possibility of non-lymphatic metastasis, an early pN stage and clinical stage (P<0.05). Moreover, NOK expression was positively correlated with the expression of oncogene p-Akt (Thr308), p-GSK-3ß (Ser9) and N-cadherin (P<0.05). Immunofluorescence analysis of NSCLC tissues revealed that NOK is co-located with Akt and GSK-3ß. Further study in NSCLC cell lines revealed that NOK overexpression can activate the AKT/GSK3ß pathway. Conversely, knockdown of NOK can suppress the AKT/GSK3ß pathway. CONCLUSION: Our results suggest that NOK overexpression correlated significantly with lymphatic metastasis, advanced pN and clinical stage in NSCLC. And NOK may promote EMT by activating the AKT/GSK3ß/N-cadherin pathway in NSCLC.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA