Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 76
Filter
1.
Nature ; 615(7953): 712-719, 2023 03.
Article in English | MEDLINE | ID: mdl-36922590

ABSTRACT

Mitochondria are critical to the governance of metabolism and bioenergetics in cancer cells1. The mitochondria form highly organized networks, in which their outer and inner membrane structures define their bioenergetic capacity2,3. However, in vivo studies delineating the relationship between the structural organization of mitochondrial networks and their bioenergetic activity have been limited. Here we present an in vivo structural and functional analysis of mitochondrial networks and bioenergetic phenotypes in non-small cell lung cancer (NSCLC) using an integrated platform consisting of positron emission tomography imaging, respirometry and three-dimensional scanning block-face electron microscopy. The diverse bioenergetic phenotypes and metabolic dependencies we identified in NSCLC tumours align with distinct structural organization of mitochondrial networks present. Further, we discovered that mitochondrial networks are organized into distinct compartments within tumour cells. In tumours with high rates of oxidative phosphorylation (OXPHOSHI) and fatty acid oxidation, we identified peri-droplet mitochondrial networks wherein mitochondria contact and surround lipid droplets. By contrast, we discovered that in tumours with low rates of OXPHOS (OXPHOSLO), high glucose flux regulated perinuclear localization of mitochondria, structural remodelling of cristae and mitochondrial respiratory capacity. Our findings suggest that in NSCLC, mitochondrial networks are compartmentalized into distinct subpopulations that govern the bioenergetic capacity of tumours.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Energy Metabolism , Lung Neoplasms , Mitochondria , Humans , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/ultrastructure , Fatty Acids/metabolism , Glucose/metabolism , Lipid Droplets/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/ultrastructure , Microscopy, Electron , Mitochondria/metabolism , Mitochondria/ultrastructure , Oxidative Phosphorylation , Phenotype , Positron-Emission Tomography
2.
J Ethnopharmacol ; 276: 114196, 2021 Aug 10.
Article in English | MEDLINE | ID: mdl-33984457

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Feiyanning (FYN), the Chinese herbal medicine (CHM), has been used to manage non-small cell lung cancer (NSCLC) for the past 23 years. Chemotherapeutic drugs can induce autophagy in cancer cells to protect themselves from death. However, FYN can inhibit the protective autophagy in cancer cells. We investigated the biological mechanisms on the synergistic effects of FYN combined with chemotherapy in lung cancer cells. MATERIALS AND METHODS: We analyzed the effective chemical components for the quality control of FYN using the UPLC-Q-TOF-MS.The cell proliferation ability was detected by the cell counting kit-8 (CCK-8) and colony formation. The cell apoptosis was determined with Flow cytometry. Expression of important differential proteins were detected by western blot. Autophagy structure was observed by TEM (Tansmission electron microscopy). Tandem mCherry-EGFP-LC3B immunofluorescence was used to measure autophagic flux. RESULTS: Both FYN and cisplatin significantly induced apoptosis and inhibited cell proliferation in A549 cells. FYN reduced cell viability and increased apoptotic cell populations less effectively than cisplatin. FYN cooperated with cisplatin suppressed the cell viability, colony formation, as well as increased the cell apoptosis rate, and the expression of cleaved caspase-3 and PARP. FYN inhibited autophagy in A549 cells, which characterized by the decrease of autophagosome formation, lysosomal fusion, LC3B-II accumulation and SQSTM1 degradation, down-regulation of ATG5 and ATG7. Protective autophagy in A549 cells was induced by cisplatin. Suppression of the autophagic response using chloroquine (CQ) which is autophagy inhibitor improved the ability of cisplatin to kill cancer cells, as did FYN combined with cisplatin. CONCLUSION: In summary, we revealed that the synergistic mechanism of FYN and cisplatin is that FYN inhibited the protective autophagy induced by cisplatin in A549 cells.


Subject(s)
Antineoplastic Agents/pharmacology , Autophagy/drug effects , Carcinoma, Non-Small-Cell Lung/drug therapy , Cisplatin/pharmacology , Drugs, Chinese Herbal/pharmacology , Lung Neoplasms/drug therapy , A549 Cells , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/ultrastructure , Cell Proliferation/drug effects , Cell Survival/drug effects , Chloroquine/pharmacology , Drug Synergism , Drugs, Chinese Herbal/chemistry , Humans , Lung Neoplasms/ultrastructure
3.
Cell Oncol (Dordr) ; 44(1): 135-150, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32936421

ABSTRACT

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.


Subject(s)
Apoptosis , Biphenyl Compounds/pharmacology , Carcinoma, Non-Small-Cell Lung/pathology , Lignans/pharmacology , Lung Neoplasms/pathology , Paclitaxel/pharmacology , Animals , Apoptosis/drug effects , Calcium/metabolism , Carcinoma, Non-Small-Cell Lung/ultrastructure , Cell Line, Tumor , Drug Synergism , Endoplasmic Reticulum Stress/drug effects , Humans , Lung Neoplasms/ultrastructure , Male , Mice, Inbred BALB C , Mice, Nude , Microtubule-Associated Proteins/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Proteasome Endopeptidase Complex/metabolism , Ubiquitin-Activating Enzymes/metabolism , Vacuoles/drug effects , Vacuoles/metabolism
4.
Cell Death Dis ; 11(6): 476, 2020 06 19.
Article in English | MEDLINE | ID: mdl-32561752

ABSTRACT

Y-box binding protein 1 (YBX1) is involved in the development of multiple types of tumors. However, the relationship between YBX1 and autophagy in non-small cell lung cancer (NSCLC) remains unclear. In this study, we analyzed the expression and clinical significance of YBX1 and markers of autophagy (LC3I/II) in NSCLC and examined their roles in regulating sensitivity to cisplatin in NSCLC. The retrospective analysis of patients with NSCLC indicated that YBX1 was positively correlated with autophagy. Increased levels of YBX1 or autophagy also observed in NSCLC cells compared with those in 16HBE cells. Compared to the controls, the knockdown of YBX1 expression suppressed autophagy, increased drug sensitivity and promoted apoptosis in response to cisplatin in NSCLC cells by targeting the p110ß promoter and inhibiting p110ß/Vps34/beclin1 signaling pathways. We also demonstrated in an in vivo study that the overexpressed YBX1 effectively increased NSCLC growth and progression and decreased the sensitivity to cisplatin by inducing autophagy in a xenograft tumor model, and these effects were concomitant with the increasing of p110ß and beclin1 expression. Collectively, these results show that YBX1 plays an essential role in autophagy in NSCLC.


Subject(s)
Autophagy , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Cisplatin/urine , Class Ia Phosphatidylinositol 3-Kinase/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Y-Box-Binding Protein 1/metabolism , Aged , Aged, 80 and over , Animals , Apoptosis/drug effects , Autophagy/drug effects , Beclin-1/metabolism , Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/ultrastructure , Cell Line, Tumor , Cell Proliferation/drug effects , Cisplatin/pharmacology , Disease Models, Animal , Female , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/ultrastructure , Male , Mice, Inbred BALB C , Mice, Nude , Microtubule-Associated Proteins/metabolism , Middle Aged
5.
Theranostics ; 9(18): 5134-5148, 2019.
Article in English | MEDLINE | ID: mdl-31410206

ABSTRACT

Rationale: Lung cancer is the leading cause of cancer death worldwide, and treatment options are limited to mainly cytotoxic agents. Here we reveal a novel role of miR-150 in non-small cell lung cancer (NSCLC) development and seek potential new therapeutic targets. Methods: The miR-150-mediated autophagy dysfunction in NSCLC cells were examined using molecular methods in vitro and in vivo. The upstream regulatory element and downstream target of miR-150 were identified in vitro and validated in vivo. Potential therapeutic methods (anti-c-myc or anti-miR-150) were tested in vitro and in vivo. Clinical relevance of the c-myc/miR-150/EPG5 axis in NSCLC was validated in human clinical samples and large genomics database. Results: miR-150 blocked the fusion of autophagosomes and lysosomes through directly repressing EPG5. The miR-150-mediated autophagy defect induced ER stress and increased cellular ROS levels and DNA damage response, and promoted NSCLC cell proliferation and tumor growth. Knockdown of EPG5 promoted NSCLC cell proliferation, and attenuated the effects of miR-150. c-myc gene was identified as a miR-150 transcriptional factor which increased miR-150 accumulation, therefore pharmacologically or genetically inhibiting c-myc/miR-150 expression significantly inhibited NSCLC cell growth in vitro and in vivo. Both c-myc and miR-150 were significantly over-expressed in NSCLC, while EPG5 was down-regulated in NSCLC. Expression levels of these molecules were well correlated, and also well correlated with patient survival. Conclusions: Our findings suggest that c-myc/miR-150/EPG5 mediated dysfunction of autophagy contributes to NSCLC development, which may provide a potential new diagnostic and therapeutic target in NSCLC.


Subject(s)
Autophagy-Related Proteins/metabolism , Autophagy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , MicroRNAs/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Vesicular Transport Proteins/metabolism , Animals , Autophagosomes/metabolism , Autophagosomes/ultrastructure , Base Sequence , Carcinogenesis/genetics , Carcinogenesis/pathology , Carcinoma, Non-Small-Cell Lung/ultrastructure , Cell Line, Tumor , Cell Proliferation/genetics , DNA Damage/genetics , Disease Progression , Endoplasmic Reticulum Stress/genetics , Female , Gene Silencing , Humans , Lung Neoplasms/ultrastructure , Lysosomes/metabolism , Lysosomes/ultrastructure , Membrane Fusion , Mice, Inbred BALB C , Mice, Nude , Prognosis , Reactive Oxygen Species/metabolism , Survival Analysis
6.
Cells ; 8(8)2019 08 20.
Article in English | MEDLINE | ID: mdl-31434250

ABSTRACT

Large tumor-derived Extracellular Vesicles (tdEVs) detected in blood of metastatic prostate, breast, colorectal, and non-small cell lung cancer patients after enrichment for Epithelial Cell Adhesion Molecule (EpCAM) expression and labeling with 4',6-diamidino-2-phenylindole (DAPI), phycoerythrin-conjugated antibodies against Cytokeratins (CK-PE), and allophycocyanin-conjugated antibody against the cluster of differentiation 45 (CD45-APC), are negatively associated with the overall survival of patients. Here, we investigated whether, similarly to tdEVs, leukocyte-derived EVs (ldEVs) could also be detected in EpCAM-enriched blood. Presence of ldEVs and leukocytes in image data sets of EpCAM-enriched samples of 25 healthy individuals and 75 metastatic cancer patients was evaluated using the ACCEPT software. Large ldEVs could indeed be detected, but in contrast to the 20-fold higher frequency of tdEVs as compared to Circulating Tumor Cells (CTCs), ldEVs were present in a 5-fold lower frequency as compared to leukocytes. To evaluate whether these ldEVs pre-exist in the blood or are formed during the CellSearch procedure, the blood of healthy individuals without EpCAM enrichment was labelled with the nuclear dye Hoechst and fluorescently tagged monoclonal antibodies recognizing the leukocyte-specific CD45, platelet-specific CD61, and red blood cell-specific CD235a. Fluorescence microscopy imaging using a similar setup as the CellSearch was performed and demonstrated the presence of a similar population of ldEVs present at a 3-fold lower frequency as compared to leukocytes.


Subject(s)
Adenocarcinoma/pathology , Breast Neoplasms/pathology , Carcinoma, Non-Small-Cell Lung/pathology , Colorectal Neoplasms/pathology , Epithelial Cell Adhesion Molecule/metabolism , Extracellular Vesicles/metabolism , Lung Neoplasms/pathology , Neoplastic Cells, Circulating/pathology , Carcinoma, Non-Small-Cell Lung/ultrastructure , Cell Count , Extracellular Vesicles/pathology , Female , Humans , Leukocytes/metabolism , Lung Neoplasms/ultrastructure
7.
Lung Cancer ; 97: 66-72, 2016 07.
Article in English | MEDLINE | ID: mdl-27237030

ABSTRACT

OBJECTIVES: Regarding the fatality rates stemming from various existing forms of cancers worldwide, lung cancer (LC) is ranked as the main cause of death amongst those who suffer from cancer. Although the epidemiological, clinical, and histological profile of patients with this type of cancer is largely unknown, Brazil has made tremendous efforts to generate data for supporting healthcare policies concerning lung cancer. Taking these factors into account, this study aims to analyse the epidemiological, clinical, and histological profiles of patients with non-small-cell lung cancer (NSCLC) in Brazil. MATERIAL AND METHODS: For this study, a cross-sectional epidemiological study was conducted to nationally analyse patient's data within the cancer hospital registries found in the National Cancer Institute (INCA) and the São Paulo Cancer Foundation (FOSP) between 2000 and 2011. RESULTS: A total of 35,018 patients diagnosed with NSCLC in Brazil between 2000 and 2011 were analysed. The analysis demonstrated the occurrence of an epidemiological shift, related to the most prevalent histological type of NSCLC in the study population from 2003. The shift resulted in a higher percentage of adenocarcinoma (43.3%) over squamous cell carcinoma (36.5%). Additionally, there was a significant increase in both the number of cases of LC in women and in the rates of patients diagnosed with metastatic disease. CONCLUSION: The use of filtered cigarettes since the 60's and the increase in the number of LC cases in women, were one of the causes for the switch in the histological profile of NSCLC in Brazil. Consequently, adenocarcinoma is now the predominant type of cancer detected. Late diagnosis is a hallmark sign.


Subject(s)
Adenocarcinoma/epidemiology , Carcinoma, Non-Small-Cell Lung/epidemiology , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/epidemiology , Lung Neoplasms/pathology , Adenocarcinoma/pathology , Adenocarcinoma/ultrastructure , Adolescent , Adult , Aged , Aged, 80 and over , Brazil/epidemiology , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/ultrastructure , Carcinoma, Squamous Cell/epidemiology , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/ultrastructure , Cross-Sectional Studies , Female , Histology , Humans , Lung Neoplasms/mortality , Lung Neoplasms/ultrastructure , Male , Middle Aged , Neoplasm Staging , Prevalence , Retrospective Studies , Smoking/epidemiology , Survival Rate , Treatment Outcome , Young Adult
8.
Respir Res ; 16: 138, 2015 Nov 09.
Article in English | MEDLINE | ID: mdl-26553068

ABSTRACT

BACKGROUND: The development of chemo-resistance in non-small lung cancer is a major obstacle in treating patients. Hypoxia is a commonly faced microenvironment in solid tumor and suggested to be related to both autophagy and chemo-resistance. METHODS: In this study, we investigated the role of hypoxia-induced autophagy in acquiring chemo-resistance in both cancer cell (A549) and human cancer tissue RESULTS: Hypoxic exposure (1 % O2) of A549 cell stimulated autophagic induction in cancer cells, shown by increase of LC3BI to LC3BII conversion and decrease of p62/sequestosome1 in Western blot, increased GFP-LC puncta in confocal microscopy, and increased number of double-membrane autophagic vacuoles in electron micrographs. Hypoxic exposure also induced resistance of cancer cells to cisplatin, and LC3B siRNA restored the sensitivity of cancer cells to chemotherapy. Furthermore, Human lung cancer tissues that experienced chemotherapy showed increase of LC3BI to LC3BII conversion and decrease of p62/sequestosome1 compared with chemo-naïve cancer tissue in Western blot. CONCLUSION: Autophagy may play an important role in acquiring resistance to chemotherapy in lung cancer and hypoxia related pathway seems to be involved in autophagy induction.


Subject(s)
Antineoplastic Agents/pharmacology , Autophagy/drug effects , Carcinoma, Non-Small-Cell Lung/drug therapy , Cisplatin/pharmacology , Drug Resistance, Neoplasm , Lung Neoplasms/drug therapy , Tumor Microenvironment , Adaptor Proteins, Signal Transducing/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/ultrastructure , Cell Hypoxia , Cell Line, Tumor , Dose-Response Relationship, Drug , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/ultrastructure , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , RNA Interference , Sequestosome-1 Protein , Time Factors , Transfection
9.
Nat Commun ; 6: 8524, 2015 Oct 05.
Article in English | MEDLINE | ID: mdl-26437053

ABSTRACT

Endosomes, lysosomes and related catabolic organelles are a dynamic continuum of vacuolar structures that impact a number of cell physiological processes such as protein/lipid metabolism, nutrient sensing and cell survival. Here we develop a library of ultra-pH-sensitive fluorescent nanoparticles with chemical properties that allow fine-scale, multiplexed, spatio-temporal perturbation and quantification of catabolic organelle maturation at single organelle resolution to support quantitative investigation of these processes in living cells. Deployment in cells allows quantification of the proton accumulation rate in endosomes; illumination of previously unrecognized regulatory mechanisms coupling pH transitions to endosomal coat protein exchange; discovery of distinct pH thresholds required for mTORC1 activation by free amino acids versus proteins; broad-scale characterization of the consequence of endosomal pH transitions on cellular metabolomic profiles; and functionalization of a context-specific metabolic vulnerability in lung cancer cells. Together, these biological applications indicate the robustness and adaptability of this nanotechnology-enabled 'detection and perturbation' strategy.


Subject(s)
Endocytosis , Endosomes/ultrastructure , Fluorescent Dyes , Lysosomes/ultrastructure , Nanoparticles , Amino Acids/metabolism , Apoptosis , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/ultrastructure , Cell Line , Cell Line, Tumor , Endosomes/metabolism , HeLa Cells , Humans , Hydrogen-Ion Concentration , Lung Neoplasms/metabolism , Lung Neoplasms/ultrastructure , Lysosomes/metabolism , Mechanistic Target of Rapamycin Complex 1 , Metabolome , Microscopy, Confocal , Microscopy, Fluorescence , Multiprotein Complexes/metabolism , Organelles/ultrastructure , Proteins/metabolism , TOR Serine-Threonine Kinases/metabolism , Vacuoles/ultrastructure
10.
Biochemistry ; 54(24): 3729-38, 2015 Jun 23.
Article in English | MEDLINE | ID: mdl-26030124

ABSTRACT

The tumor suppressor p53 plays a unique role as a central hub of numerous cell proliferation and apoptotic pathways, and its malfunction due to mutations is a major cause of various malignancies. Therefore, it serves as an attractive target for developing novel anticancer therapeutics. Because of its intrinsically unstable DNA binding domain, p53 unfolds rapidly at physiological temperature. Certain mutants shift the equilibrium toward the unfolded state and yield high-molecular weight, nonfunctional, and cytotoxic ß-sheet-rich aggregates that share tinctorial and conformational similarities with amyloid deposits found in various protein misfolding diseases. Here, we examined the effect of a novel protein assembly modulator, the lysine (Lys)-specific molecular tweezer, CLR01, on different aggregation stages of misfolded mutant p53 in vitro and on the cytotoxicity of the resulting p53 aggregates in cell culture. We found that CLR01 induced rapid formation of ß-sheet-rich, intermediate-size p53 aggregates yet inhibited further p53 aggregation and reduced the cytotoxicity of the resulting aggregates. Our data suggest that aggregation modulators, such as CLR01, could prevent the formation of toxic p53 aggregates.


Subject(s)
Antineoplastic Agents/pharmacology , Bridged-Ring Compounds/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Models, Molecular , Mutation , Organophosphates/pharmacology , Protein Aggregation, Pathological/drug therapy , Tumor Suppressor Protein p53/antagonists & inhibitors , Amino Acid Substitution , Antineoplastic Agents/chemistry , Binding Sites , Bridged-Ring Compounds/chemistry , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/ultrastructure , Cell Line, Tumor , Cell Survival/drug effects , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/ultrastructure , Microscopy, Electron, Transmission , Mutagenesis, Site-Directed , Organophosphates/chemistry , Protein Aggregates/drug effects , Protein Aggregation, Pathological/genetics , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/pathology , Protein Stability/drug effects , Protein Unfolding/drug effects , Proteostasis Deficiencies/drug therapy , Proteostasis Deficiencies/genetics , Proteostasis Deficiencies/metabolism , Proteostasis Deficiencies/pathology , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
11.
Biochim Biophys Acta ; 1853(2): 328-37, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25450981

ABSTRACT

Tumor invasion and metastasis are key aspects of non-small cell lung cancer (NSCLC). During migration, cells undergo mechanical alterations. The mechanical phenotype of breast cancer cells is correlated with aromatase gene expression. We have previously shown that targeting aromatase is a promising strategy for NSCLC. The aim of this study was to examine morphological and mechanical changes of NSCLC cells, upon treatment with aromatase inhibitor and correlate their ability to migrate and invade. In vitro experiments were performed using H23 and A549 NSCLC cell lines and exemestane was used for aromatase inhibition. We demonstrated that exemestane reduced H23 cell migration and invasion and caused changes in cell morphology including increased vacuolar structures and greater pleomorphism. In addition, exemestane changed the distribution of α-tubulin in H23 and A549 cells in a way that might destabilize microtubules polymerization. These effects were associated with increased cell viscosity and decreased elastic shear modulus. Although exemestane caused similar effects in A549 cells regarding viscosity and elastic shear modulus, it did not affect A549 cell migration and caused an increase in invasion. The increased invasion was in line with vimentin perinuclear localization. Our data show that the treatment of NSCLC cells with an aromatase inhibitor not only affects cell migration and invasion but also alters the mechanical properties of the cells. It suggests that the different origin of cancer cells is associated with different morphological characteristics and mechanical behavior.


Subject(s)
Aromatase Inhibitors/pharmacology , Aromatase/metabolism , Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/pathology , Cell Movement/drug effects , Lung Neoplasms/pathology , Rheology/drug effects , Actins/metabolism , Androstadienes/pharmacology , Androstadienes/therapeutic use , Apoptosis/drug effects , Aromatase Inhibitors/therapeutic use , Biomechanical Phenomena/drug effects , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/ultrastructure , Cell Line, Tumor , Cell Shape/drug effects , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Elastic Modulus/drug effects , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/enzymology , Lung Neoplasms/ultrastructure , Neoplasm Invasiveness , Tubulin/metabolism , Vimentin/metabolism
12.
Micron ; 67: 125-131, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25151215

ABSTRACT

INTRODUCTION: In recent years, differences have emerged in the treatment of squamous and non-squamous non-small cell lung carcinomas (NSCLCs). This highlights the importance of accurate histopathologic classification. However, there remains inter-observer disagreement when making diagnoses based on histology. Fractal dimension (FD) is a mathematical measure of irregularity and complexity of shape. We hypothesize that the FD of carcinoma epithelial architecture can assist in differentiating adenocarcinoma (ADC) from squamous cell carcinoma (SCC) of the lung. METHODS: 134 resected (88 ADC and 46 SCC) cases of resected early-stage NSCLC were analyzed. Tissue micro arrays were generated from formalin-fixed paraffin-embedded tissue, stained with pan-cytokeratin, and digitally imaged and the FD of the epithelial structure calculated. Mean FD of ADC and SCC were compared using the independent t-test, partial correlations, and receiver operating characteristic (ROC) analyses. RESULTS: A statistically significant difference (p<0.001) between the mean FD of ADC (M=1.70, SD=0.07) and SCC (M=1.78, SD=0.07) was found. Significance remained (p<0.001) when controlling for several possible confounders. ROC analysis demonstrated an area-under-the-curve of 0.81 (p<0.001). CONCLUSIONS: The epithelial structure FD of NSCLC has potential as a reproducible and automated measure to help subtype NSCLCs into ADC and SCC. With further image analysis algorithm improvements, fractal analysis may be a component in computerized histomorphological assessments of lung cancer and may provide an adjunct test in differentiating NSCLCs.


Subject(s)
Carcinoma, Non-Small-Cell Lung/ultrastructure , Lung Neoplasms/ultrastructure , Respiratory Mucosa/ultrastructure , Adenocarcinoma/ultrastructure , Aged , Carcinoma, Non-Small-Cell Lung/diagnosis , Female , Fractals , Humans , Image Processing, Computer-Assisted , Kaplan-Meier Estimate , Lung Neoplasms/diagnosis , Male , Middle Aged , Sensitivity and Specificity
13.
Biochem Biophys Res Commun ; 437(1): 173-8, 2013 Jul 19.
Article in English | MEDLINE | ID: mdl-23810394

ABSTRACT

BTG3 is identified as a tumor suppressor gene in some malignancies. Btg3-deficient mice display a higher incidence of lung cancer. These results suggest that BTG3 plays an important role in lung tumorigenesis, although the underlying mechanisms are unknown. The BTG3 expression was detected using immunohistochemical staining and our results showed that the expression of BTG3 was reduced in lung cancer compared to benign lung tissues. We identified two BTG3 isoforms present in lung cancer: Full-length BTG3 and BTG3b lacking the 44 amino acids. BTG3 was predominantly expressed in benign lung tissues, whereas its expression was generally undetectable in lung cancer and cancer cell lines. Functional analysis revealed that BTG3 but not BTG3b inhibited lung cancer growth. Our results disclosed an important role of BTG3 in lung tumorigenesis.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Down-Regulation/genetics , Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , Proteins/genetics , Animals , Autophagy/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/ultrastructure , Cell Cycle/genetics , Cell Cycle Proteins , Cell Differentiation/genetics , Cell Proliferation , Gene Expression Profiling , Humans , Immunohistochemistry , Lung Neoplasms/pathology , Lung Neoplasms/ultrastructure , Male , Mice , Mice, Nude , Neoplasm Metastasis , Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
14.
J Pharmacol Sci ; 121(4): 272-81, 2013.
Article in English | MEDLINE | ID: mdl-23603895

ABSTRACT

Genipin, an active constituent of Gardenia fruit, has been reported to show an anti-tumor effect in several cancer cell systems. Here, we demonstrate how genipin exhibits a strong apoptotic cell death effect in human non-small-cell lung cancer H1299 cells. Genipin-mediated decrease in cell viability was observed through apoptosis as demonstrated by induction of a sub-G1 peak through flow cytometry, DNA fragmentation measured by TUNEL assay, and cleavage of poly ADP-ribose-polymerase. During genipin-induced apoptosis, the mitochondrial execution pathway was activated by caspase-9 and -3 activation as examined by a kinetic study, cytochrome c release, and a dose-dependent increase in Bax/Bcl-2 ratio. A search for the downstream pathway reveals that genipin-induced apoptosis was mediated by an increase in phosphorylated p38MAPK expression, which further activated downstream signaling by phosphorylating ATF-2. SB203580, a p38MAPK inhibitor, markedly blocked the formation of TUNEL-positive apoptotic cells in genipin-treated cells. Besides, the interference of p38MAPK inhibited Bax expression and cytochrome c release. Altogether, our observations imply that genipin causes increased levels of Bax in response to p38MAPK signaling, which results in the initiation of mitochondrial death cascade, and therefore it holds promise as a potential chemotherapeutic agent for the treatment of H1299 cells.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/ultrastructure , Iridoids/pharmacology , Mitochondria/pathology , p38 Mitogen-Activated Protein Kinases/physiology , Carcinoma, Non-Small-Cell Lung/drug therapy , Cell Survival/drug effects , Cytochromes c/metabolism , Dose-Response Relationship, Drug , Humans , Molecular Targeted Therapy , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction/physiology , Tumor Cells, Cultured , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
15.
Int J Biochem Cell Biol ; 44(11): 1813-24, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22750415

ABSTRACT

Na(+)/K(+)-ATPase targeted cancer therapy has attracted increasing interests of oncologists in lung cancer field. Although multiple anti-cancer mechanisms of cardiac glycosides as Na(+)/K(+)-ATPase inhibitors are revealed, the role of autophagy and related molecular signaling pathway for the class of compounds in human non-small cell lung cancer (NSCLC) cells has not been systematically examined. We herein investigated the anti-cancer effects of two representative cardiac glycosides, digoxin and ouabain, in A549 and H460 cell lines. Both agents caused significant growth inhibition at nanomolar level. The cardiac glycosides were found to induce moderate G(2)/M arrest but not apoptosis at IC(50) level in the NSCLC cell lines. Moreover, autophagy was markedly induced by both agents, as evidenced by the time- and dose-dependent increase of LC3-II, up-regulation of Atg5 and Beclin1, as well as by the observations through acridine orange staining, transmission electron microscopy and quantification of GFP-LC3 fluorescence. Importantly, AMP-activated protein kinase (AMPK) pathway was activated, resulting in mammalian target of rapamycin (mTOR) deactivation during autophagy induction. Moreover, extracellular-signal-regulated kinase 1/2 (ERK1/2) activation was simultaneously found to be involved in the autophagy regulation. Co-treatment with respective inhibitors or siRNAs could either block the autophagic phenotypes and signals, or significantly increase the cellular viability, indicating the drugs-induced autophagy plays tumor-suppressing role. This work provides first evidence showing that the cardiac glycosides induce autophagy in human NSCLC cells through regulation of both mTOR and ERK1/2 signaling pathways. The autophagy may at least partially account for the growth inhibitory effects of the compounds in human NSCLC cells.


Subject(s)
Autophagy/drug effects , Carcinoma, Non-Small-Cell Lung/pathology , Cardiac Glycosides/pharmacology , Lung Neoplasms/pathology , Signal Transduction/drug effects , AMP-Activated Protein Kinases/metabolism , Androstadienes/pharmacology , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Autophagy-Related Protein 5 , Beclin-1 , Biomarkers/metabolism , Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/ultrastructure , Cell Line, Tumor , Digoxin/pharmacology , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , G2 Phase Cell Cycle Checkpoints/drug effects , Gene Knockdown Techniques , Humans , Inhibitory Concentration 50 , Lung Neoplasms/enzymology , Lung Neoplasms/ultrastructure , MAP Kinase Signaling System/drug effects , Membrane Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Mitosis/drug effects , Ouabain/pharmacology , TOR Serine-Threonine Kinases/metabolism , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/metabolism , Vacuoles/drug effects , Vacuoles/metabolism , Vacuoles/ultrastructure , Wortmannin
16.
Asian Pac J Cancer Prev ; 13(4): 1505-10, 2012.
Article in English | MEDLINE | ID: mdl-22799356

ABSTRACT

To aim of this was to observe emodin-mediated cytotoxicity and its influence on Rad51 and ERCC1 expressionin non-small cell lung cancer (NSCLC). NSCLC cells were cultured in vitro with emodin at various concentrations (0, 25, 50, 75 and 100 µmol/L) for 48 h and the proliferation inhibition rate was determined by the MTT method. Then, NSCLC were treated with emodin (SK-MES-1 40 µmol/L, A549 70 µmol/L) or 20 µmol/L U0126 (an ERK inhibitor) for 48 h, or with various concentrations of emodin for 48 h and the protein and mRNA expressions of ERCC1 and Rad51 were determined by RT-PCR and Western blot assay, respectively. Emodin exerted a suppressive effect on the proliferation of NSCLC in a concentration dependent manner. Protein and mRNA expression of ERCC1 and Rad51 was also significantly decreased with the dose. Vacuolar degeneration was observed in A549 and SK-MES-1 cell lines after emodin treatment by transmission electron microscopy. Emodin may thus inhibited cell proliferation in NSCLC cells by downregulation ERCC1 and Rad51.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , DNA-Binding Proteins/metabolism , Emodin/pharmacology , Endonucleases/metabolism , Plant Extracts/pharmacology , Protein Kinase Inhibitors/pharmacology , Rad51 Recombinase/metabolism , Analysis of Variance , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/ultrastructure , Cell Line, Tumor , Cell Proliferation/drug effects , DNA-Binding Proteins/drug effects , DNA-Binding Proteins/genetics , Down-Regulation , Drugs, Chinese Herbal/pharmacology , Endonucleases/drug effects , Endonucleases/genetics , Gene Expression , Humans , RNA, Messenger/metabolism , Rad51 Recombinase/drug effects , Rad51 Recombinase/genetics , Vacuoles/ultrastructure
17.
Mol Pharm ; 9(1): 196-200, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-22053709

ABSTRACT

The intracellular activity and drug depot characteristics of micrometer-sized hydrogels are described. The hydrogel structure is formed after cellular uptake of a solid polymeric nanoparticle that swells in response to mildly acidic conditions as it transforms from a hydrophobic to a hydrophilic structure. These nanoparticles are rapidly taken up into A549 human non-small cell lung cancer cells with 88.3 ± 0.8% of cells experiencing uptake in 24 h, undergo expansion to release encapsulated drug and can effectively deliver chemotherapeutics in vitro. The anticancer drug paclitaxel was also shown to have a 3- to 4-fold increased affinity for the expanded nanoparticle state, allowing the expansile nanoparticles to act as intracellular drug depots and concentrate the drug locally.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Drug Carriers/administration & dosage , Lung Neoplasms/drug therapy , Nanoparticles/chemistry , Paclitaxel/administration & dosage , Biological Transport , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/ultrastructure , Cell Line, Tumor , Cell Survival/drug effects , Chemical Phenomena , Drug Carriers/chemistry , Drug Carriers/metabolism , Drug Carriers/pharmacology , Drug Compounding , Humans , Hydrogels , Hydrogen-Ion Concentration , Hydrophobic and Hydrophilic Interactions , Lung Neoplasms/metabolism , Lung Neoplasms/ultrastructure , Microscopy, Electron, Scanning , Microscopy, Fluorescence , Nanoparticles/ultrastructure , Osmolar Concentration , Paclitaxel/chemistry , Paclitaxel/metabolism , Paclitaxel/pharmacology , Particle Size , Polymethacrylic Acids/chemistry
19.
J Vis Exp ; (52)2011 Jun 21.
Article in English | MEDLINE | ID: mdl-21712803

ABSTRACT

Non-destructive volume visualization can be achieved only by tomographic techniques, of which the most efficient is the x-ray micro computerized tomography (µCT). High resolution µCT is a very versatile yet accurate (1-2 microns of resolution) technique for 3D examination of ex-vivo biological samples(1, 2). As opposed to electron tomography, the µCT allows the examination of up to 4 cm thick samples. This technique requires only few hours of measurement as compared to weeks in histology. In addition, µCT does not rely on 2D stereologic models, thus it may complement and in some cases can even replace histological methods(3, 4), which are both time consuming and destructive. Sample conditioning and positioning in µCT is straightforward and does not require high vacuum or low temperatures, which may adversely affect the structure. The sample is positioned and rotated 180° or 360°between a microfocused x-ray source and a detector, which includes a scintillator and an accurate CCD camera, For each angle a 2D image is taken, and then the entire volume is reconstructed using one of the different available algorithms(5-7). The 3D resolution increases with the decrease of the rotation step. The present video protocol shows the main steps in preparation, immobilization and positioning of the sample followed by imaging at high resolution.


Subject(s)
Imaging, Three-Dimensional/methods , X-Ray Microtomography/methods , Animals , Carcinoma, Non-Small-Cell Lung/ultrastructure , Embryo, Mammalian/ultrastructure , Femur/cytology , Femur/ultrastructure , Image Processing, Computer-Assisted/methods , Imaging, Three-Dimensional/instrumentation , Lung/ultrastructure , Lung Neoplasms/ultrastructure , Mice , Mice, Inbred C57BL , Rats , X-Ray Microtomography/instrumentation
20.
J Immunol ; 186(6): 3735-44, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21300821

ABSTRACT

1E10 is a murine anti-idiotypic mAb specific for an idiotypic mAb that reacts with NeuGc-containing gangliosides, sulfatides, and Ags expressed in some human tumors. In melanoma, breast, and lung cancer patients, this anti-idiotypic Ab was able to induce a specific Ab response against N-glycosylated gangliosides, attractive targets for cancer immunotherapy as these glycolipids are not naturally expressed in humans. A clinical study with nonsmall cell lung cancer patients showed encouraging clinical benefits. Immunological studies performed in 20 of these patients suggested a correlation between the induction of Abs against NeuGcGM3 and longer survival times. The induced anti-NeuGcGM3 Abs recognized and directly killed tumor cells expressing the Ag, by a mechanism independent of complement activation. In the present work, we show that this cytotoxicity differs from apoptosis because it is temperature independent, no chromatin condensation or caspase 3 induction are detected, and the DNA fragmentation induced has a different pattern than the one characteristic for apoptosis. It is a very quick process and involves cytosqeleton reorganization. The Abs induce cellular swelling and the formation of big membrane lesions that allow the leakage of cytoplasm and the loss of the cell membrane integrity. All of these characteristics resemble a process of oncotic necrosis. To our knowledge, this is the first report of the active induction in cancer patients of NeuGcGM3-specific Abs able to induce complement independent oncotic necrosis to tumor cells. These results contribute to reinforcing the therapeutic potential of anti-idiotypic vaccines and the importance of NeuGcGM3 ganglioside as antitumor target.


Subject(s)
Antibodies, Neoplasm/physiology , Cancer Vaccines/administration & dosage , Carcinoma, Non-Small-Cell Lung/therapy , G(M3) Ganglioside/analogs & derivatives , G(M3) Ganglioside/immunology , Immunoglobulin Idiotypes/physiology , Lung Neoplasms/immunology , Lung Neoplasms/therapy , Animals , Antibodies, Neoplasm/administration & dosage , Antibodies, Neoplasm/biosynthesis , Cancer Vaccines/immunology , Carcinoma, Lewis Lung/immunology , Carcinoma, Lewis Lung/therapy , Carcinoma, Lewis Lung/ultrastructure , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/ultrastructure , Cell Death/immunology , Cell Line, Tumor , Dogs , Horses , Humans , Immunoglobulin G/biosynthesis , Immunoglobulin Idiotypes/administration & dosage , Immunoglobulin M/biosynthesis , Leukemia L1210/immunology , Leukemia L1210/pathology , Leukemia L1210/therapy , Lung Neoplasms/ultrastructure , Mice , Mice, Inbred BALB C , Plasmacytoma/immunology , Plasmacytoma/pathology , Plasmacytoma/therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...