Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Adv Sci (Weinh) ; 10(17): e2204378, 2023 06.
Article in English | MEDLINE | ID: mdl-37097643

ABSTRACT

Immune checkpoint inhibitor (ICI) clinically benefits cancer treatment. However, the ICI responses are only achieved in a subset of patients, and the underlying mechanisms of the limited response remain unclear. 160 patients with non-small cell lung cancer treated with anti-programmed cell death protein-1 (anti-PD-1) or anti-programmed death ligand-1 (anti-PD-L1) are analyzed to understand the early determinants of response to ICI. It is observed that high levels of intracellular adhesion molecule-1 (ICAM-1) in tumors and plasma of patients are associated with prolonged survival. Further reverse translational studies using murine syngeneic tumor models reveal that soluble ICAM-1 (sICAM-1) is a key molecule that increases the efficacy of anti-PD-1 via activation of cytotoxic T cells. Moreover, chemokine (CXC motif) ligand 13 (CXCL13) in tumors and plasma is correlated with the level of ICAM-1 and ICI efficacy, suggesting that CXCL13 might be involved in the ICAM-1-mediated anti-tumor pathway. Using sICAM-1 alone and in combination with anti-PD-1 enhances anti-tumor efficacy in anti-PD-1-responsive tumors in murine models. Notably, combinatorial therapy with sICAM-1 and anti-PD-1 converts anti-PD-1-resistant tumors to responsive ones in a preclinical study. These findings provide a new immunotherapeutic strategy for treating cancers using ICAM-1.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Mice , Animals , CD8-Positive T-Lymphocytes , Carcinoma, Non-Small-Cell Lung/drug therapy , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Intercellular Adhesion Molecule-1
2.
Microbiome ; 10(1): 188, 2022 11 04.
Article in English | MEDLINE | ID: mdl-36333752

ABSTRACT

BACKGROUND: Comparisons of the gut microbiome of lean and obese humans have revealed that obesity is associated with the gut microbiome plus changes in numerous environmental factors, including high-fat diet (HFD). Here, we report that two species of Bifidobacterium are crucial to controlling metabolic parameters in the Korean population. RESULTS: Based on gut microbial analysis from 99 Korean individuals, we observed the abundance of Bifidobacterium longum and Bifidobacterium bifidum was markedly reduced in individuals with increased visceral adipose tissue (VAT), body mass index (BMI), blood triglyceride (TG), and fatty liver. Bacterial transcriptomic analysis revealed that carbohydrate/nucleoside metabolic processes of Bifidobacterium longum and Bifidobacterium bifidum were associated with protecting against diet-induced obesity. Oral treatment of specific commercial Bifidobacterium longum and Bifidobacterium bifidum enhanced bile acid signaling contributing to potentiate oxidative phosphorylation (OXPHOS) in adipose tissues, leading to reduction of body weight gain and improvement in hepatic steatosis and glucose homeostasis. Bifidobacterium longum or Bifidobacterium bifidum manipulated intestinal sterol biosynthetic processes to protect against diet-induced obesity in germ-free mice. CONCLUSIONS: Our findings support the notion that treatment of carbohydrate/nucleoside metabolic processes-enriched Bifidobacterium longum and Bifidobacterium bifidum would be a novel therapeutic strategy for reprograming the host metabolic homeostasis to protect against metabolic syndromes, including diet-induced obesity. Video Abstract.


Subject(s)
Bifidobacterium longum , Bifidobacterium , Humans , Mice , Animals , Bifidobacterium/metabolism , Nucleosides/metabolism , Nucleosides/therapeutic use , Oxidative Phosphorylation , Obesity/microbiology , Diet, High-Fat/adverse effects , Adipose Tissue, White/metabolism
3.
Nat Microbiol ; 6(3): 277-288, 2021 03.
Article in English | MEDLINE | ID: mdl-33432149

ABSTRACT

The gut microbiome can influence the development of tumours and the efficacy of cancer therapeutics1-5; however, the multi-omics characteristics of antitumour bacterial strains have not been fully elucidated. In this study, we integrated metagenomics, genomics and transcriptomics of bacteria, and analyses of mouse intestinal transcriptome and serum metabolome data to reveal an additional mechanism by which bacteria determine the efficacy of cancer therapeutics. In gut microbiome analyses of 96 samples from patients with non-small-cell lung cancer, Bifidobacterium bifidum was abundant in patients responsive to therapy. However, when we treated syngeneic mouse tumours with commercial strains of B. bifidum to establish relevance for potential therapeutic uses, only specific B. bifidum strains reduced tumour burden synergistically with PD-1 blockade or oxaliplatin treatment by eliciting an antitumour host immune response. In mice, these strains induced tuning of the immunological background by potentiating the production of interferon-γ, probably through the enhanced biosynthesis of immune-stimulating molecules and metabolites.


Subject(s)
Bifidobacterium bifidum/physiology , Immune Checkpoint Inhibitors/therapeutic use , Probiotics/therapeutic use , Tumor Burden/drug effects , Animals , Bifidobacterium bifidum/classification , Bifidobacterium bifidum/genetics , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/microbiology , Carcinoma, Non-Small-Cell Lung/pathology , Drug Therapy, Combination , Gastrointestinal Microbiome , Humans , Interferon-gamma/genetics , Interferon-gamma/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/microbiology , Lung Neoplasms/pathology , Metabolome/drug effects , Mice , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Probiotics/administration & dosage , Species Specificity , Transcriptome/drug effects , Tryptophan/metabolism
4.
Nat Biomed Eng ; 2(4): 239-253, 2018 04.
Article in English | MEDLINE | ID: mdl-30936439

ABSTRACT

The accumulation of genetic and epigenetic alterations in cancer cells rewires cellular signalling pathways through changes in the patterns of protein-protein interactions (PPIs). Understanding these patterns may facilitate the design of tailored cancer therapies. Here, we show that single-molecule pull-down and co-immunoprecipitation techniques can be used to characterize signalling complexes of the human epidermal growth-factor receptor (HER) family in specific cancers. By analysing cancer-specific signalling phenotypes, including post-translational modifications and PPIs with downstream interactions, we found that activating mutations of the epidermal growth-factor receptor (EGFR) gene led to the formation of large protein complexes surrounding mutant EGFR proteins and to a reduction in the dependency of mutant EGFR signalling on phosphotyrosine residues, and that the strength of HER-family PPIs is correlated with the strength of the dependence of breast and lung adenocarcinoma cells on HER-family signalling pathways. Furthermore, using co-immunoprecipitation profiling to screen for EGFR-dependent cancers, we identified non-small-cell lung cancers that respond to an EGFR-targeted inhibitor. Our approach might help predict responses to targeted cancer therapies, particularly for cancers that lack actionable genomic mutations.


Subject(s)
ErbB Receptors/metabolism , Molecular Diagnostic Techniques/methods , Neoplasms/diagnosis , Protein Interaction Maps/physiology , Signal Transduction/physiology , Animals , Cell Line, Tumor , Female , Humans , Mice , Mice, SCID , Middle Aged , Neoplasms/metabolism
5.
Mol Cancer Ther ; 15(7): 1627-36, 2016 07.
Article in English | MEDLINE | ID: mdl-27196768

ABSTRACT

Although treatment of BRAF V600E-mutant non-small cell lung cancer (NSCLC(V600E)) with GSK2118436 has shown an encouraging efficacy, most patients develop resistance. To investigate the mechanisms of acquired resistance to GSK2118436 in NSCLC(V600E), we established GSK2118436-resistant (GSR) cells by exposing MV522 NSCLC(V600E) to increasing GSK2118436 concentrations. GSR cells displayed activated EGFR-RAS-CRAF signaling with upregulated EGFR ligands and sustained activation of ERK1/2, but not MEK1/2, in the presence of GSK2118436. Treatment of GSR cells with GSK2118436 enhanced EGFR-mediated RAS activity, leading to the formation of BRAF-CRAF dimers and transactivation of CRAF. Interestingly, sustained activation of ERK1/2 was partly dependent on receptor-interacting protein kinase-2 (RIP2) activity, but not on MEK1/2 activity. Combined BRAF and EGFR inhibition blocked reactivation of ERK signaling and improved efficacy in vitro and in vivo Our findings support the evaluation of combined BRAF and EGFR inhibition in NSCLC(V600E) with acquired resistance to BRAF inhibitors. Mol Cancer Ther; 15(7); 1627-36. ©2016 AACR.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Drug Resistance, Neoplasm/genetics , ErbB Receptors/metabolism , Imidazoles/pharmacology , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , MAP Kinase Signaling System , Mutation , Oximes/pharmacology , Proto-Oncogene Proteins B-raf/genetics , Amino Acid Substitution , Animals , Antineoplastic Agents/pharmacology , Autocrine Communication , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Survival/drug effects , Codon , DNA Mutational Analysis , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Mice , Models, Biological , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Receptor-Interacting Protein Serine-Threonine Kinase 2/metabolism , Xenograft Model Antitumor Assays , ras Proteins/metabolism
6.
PLoS One ; 10(9): e0137678, 2015.
Article in English | MEDLINE | ID: mdl-26366867

ABSTRACT

Glioblastoma multiforme (GBM) is the most aggressive type of brain tumor, and the prognosis remains poor. Rearrangement of ROS1 gene, which was shown to have an oncogenic potential, was previously discovered in GBM cell lines. In this pilot study, we aimed to identify the incidence of ROS1 rearrangement in GBM patient tissues to explore novel biomarkers for therapeutic strategy. Formalin-fixed and paraffin-embedded (FFPE) tissue sections from 109 patients with GBM were screened for ROS1 rearrangement by anti-ROS immunohistochemistry (IHC) and ROS1 break-apart fluorescent in situ hybridization (FISH) assays. O6-methylguanine-DNA methyltransferase (MGMT) gene promoter methylation and Isocitrate dehydrogenase 1 (IDH1) mutation status were also assessed. All samples were interpreted by two experienced pathologists who were blinded to the clinical data. A total of 109 samples were collected and all samples were examined for ROS1 rearrangement by IHC and FISH assays, and none was found to harbor ROS1 rearrangement. MGMT gene methylation was found in 42 (39.2%) cases, and IDH1 mutation was found in 6 (5.5%) cases. In this study, ROS1 rearrangement was not identified in GBM patients, and thus it is difficult to classify ROS1 rearrangement as a novel molecular subset in GBM patients for now.


Subject(s)
Gene Rearrangement , Glioblastoma/genetics , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins/genetics , Adult , Aged , Aged, 80 and over , Biomarkers/metabolism , DNA Methylation , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Female , Glioblastoma/diagnosis , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Isocitrate Dehydrogenase/genetics , Male , Middle Aged , Multivariate Analysis , Pilot Projects , Prognosis , Promoter Regions, Genetic , Tumor Suppressor Proteins/genetics
7.
Mol Cancer Ther ; 14(10): 2238-48, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26208525

ABSTRACT

RET rearrangement is a newly identified oncogenic mutation in lung adenocarcinoma (LADC). Activity of dovitinib (TKI258), a potent inhibitor of FGFR, VEGFR, and PDGFR, in RET-rearranged LADC has not been reported. The aims of the study are to explore antitumor effects and mechanisms of acquired resistance of dovitinib in RET-rearranged LADC. Using structural modeling and in vitro analysis, we demonstrated that dovitinib induced cell-cycle arrest at G0-G1 phase and apoptosis by selective inhibition of RET kinase activity and ERK1/2 signaling in RET-rearranged LC-2/ad cells. Strong antitumor effect of dovitinib was observed in an LC-2/ad tumor xenograft model. To identify the acquired resistance mechanisms to dovitinib, LC-2/ad cells were exposed to increasing concentrations of dovitinib to generate LC-2/ad DR cells. Gene-set enrichment analysis of gene expression and phosphor-kinase revealed that Src, a central gene in focal adhesion, was activated in LC-2/ad DR cells. Saracatinib, an src kinase inhibitor, suppressed ERK1/2 phosphorylation and growth of LC-2/ad DR cells. Taken together, these findings suggest that dovitinib can be a potential therapeutic option for RET-rearranged LADC, in which acquired resistance to dovitinib can be overcome by targeting Src.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents/pharmacology , Benzimidazoles/pharmacology , Drug Resistance, Neoplasm , Lung Neoplasms/drug therapy , Proto-Oncogene Proteins c-ret/antagonists & inhibitors , Quinolones/pharmacology , Adenocarcinoma/enzymology , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Animals , Antineoplastic Agents/chemistry , Apoptosis , Benzimidazoles/chemistry , Catalytic Domain , Cell Cycle Checkpoints , Cell Line, Tumor , Enzyme Activation , HEK293 Cells , Humans , Inhibitory Concentration 50 , Lung Neoplasms/enzymology , Lung Neoplasms/pathology , Mice, Nude , Models, Molecular , Protein Kinase Inhibitors , Proto-Oncogene Proteins c-ret/chemistry , Quinolones/chemistry , Tumor Burden/drug effects , Xenograft Model Antitumor Assays , src-Family Kinases/metabolism
8.
J Biomed Biotechnol ; 2012: 170958, 2012.
Article in English | MEDLINE | ID: mdl-22778542

ABSTRACT

Aminoamide local anesthetics induce vasoconstriction in vivo and in vitro. The goals of this in vitro study were to investigate the potency of local anesthetic-induced vasoconstriction and to identify the physicochemical property (octanol/buffer partition coefficient, pKa, molecular weight, or potency) of local anesthetics that determines their potency in inducing isolated rat aortic ring contraction. Cumulative concentration-response curves to local anesthetics (levobupivacaine, ropivacaine, lidocaine, and mepivacaine) were obtained from isolated rat aorta. Regression analyses were performed to determine the relationship between the reported physicochemical properties of local anesthetics and the local anesthetic concentration that produced 50% (ED(50)) of the local anesthetic-induced maximum vasoconstriction. We determined the order of potency (ED(50)) of vasoconstriction among local anesthetics to be levobupivacaine > ropivacaine > lidocaine > mepivacaine. The relative importance of the independent variables that affect the vasoconstriction potency is octanol/buffer partition coefficient > potency > pKa > molecular weight. The ED(50) in endothelium-denuded aorta negatively correlated with the octanol/buffer partition coefficient of local anesthetics (r(2) = 0.9563; P < 0.001). The potency of the vasoconstriction in the endothelium-denuded aorta induced by local anesthetics is determined primarily by lipid solubility and, in part, by other physicochemical properties including potency and pKa.


Subject(s)
Amides/pharmacology , Anesthetics, Local/pharmacology , Vasoconstrictor Agents/pharmacology , Amides/chemistry , Anesthetics, Local/chemistry , Animals , Aorta/drug effects , Dose-Response Relationship, Drug , Male , Molecular Weight , Octanols/chemistry , Rats , Rats, Sprague-Dawley , Regression Analysis , Solubility , Vasoconstriction/drug effects , Vasoconstrictor Agents/chemistry
9.
Biochem Biophys Res Commun ; 336(4): 1164-71, 2005 Nov 04.
Article in English | MEDLINE | ID: mdl-16171786

ABSTRACT

Genomic instability and apoptosis evasion are hallmarks of cancer, but the molecular mechanisms governing these processes remain elusive. Here, we found that survivin, a member of the apoptosis-inhibiting gene family, and aurora B kinase, a chromosomal passenger protein, were co-overexpressed in the various glioblastoma cell lines and tumors. Notably, exogenous introduction of the aurora B in human BJ cells was shown to decrease cell growth and increase the senescence-associated beta-galactosidase activity by activation of p53 tumor suppressor. However, aurora B overexpression failed to inhibit cell proliferation in BJ and U87MG cells transduced with dominant-negative p53 as well as in p53(-/-) mouse astrocytes. Aurora B was shown to increase centrosome amplification in the p53(-/-) astrocytes. Survivin was shown to induce anchorage-independent growth and inhibit anti-proliferation and drug-sensitive apoptosis caused by aurora B. Overexpression of both survivin and aurora B further accelerated the proliferation of BJ cells. Taken together, the present study indicates that survivin should accelerate tumorigenesis by inhibiting the anti-proliferative effect of p53 tumor suppressor that is activated by aurora B in normal and glioblastoma cells containing intact p53.


Subject(s)
Apoptosis/physiology , Microtubule-Associated Proteins/physiology , Neoplasm Proteins/physiology , Neoplasms/metabolism , Protein Serine-Threonine Kinases/physiology , Tumor Suppressor Protein p53/antagonists & inhibitors , Animals , Astrocytes/metabolism , Aurora Kinase B , Aurora Kinases , Brain Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation , Centrosome/metabolism , Contact Inhibition/physiology , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Enzyme Activation , Genes, Reporter , Glioblastoma/metabolism , Humans , Inhibitor of Apoptosis Proteins , Mice , Microtubule-Associated Proteins/biosynthesis , Neoplasm Proteins/biosynthesis , Phosphorylation , Protein Serine-Threonine Kinases/biosynthesis , Survivin , Tumor Suppressor Protein p53/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...