Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 111
Filter
1.
Acta Pharmacol Sin ; 42(1): 108-114, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32398685

ABSTRACT

Glioblastoma (GBM) patients have extremely poor prognoses, and currently no effective treatment available including surgery, radiation, and chemotherapy. MAPK-interacting kinases (MNK1/2) as the downstream of the MAPK-signaling pathway regulate protein synthesis in normal and tumor cells. Research has shown that targeting MNKs may be an effective strategy to treat GBM. In this study we investigated the antitumor activity of osimertinib, an FDA-approved epidermal growth factor receptor (EGFR) inhibitor, against patient-derived primary GBM cells. Using high-throughput screening approach, we screened the entire panel of FDA-approved drugs against primary cancer cells derived from glioblastoma patients, found that osimertinib (3 µM) suppressed the proliferation of a subset (10/22) of EGFR-negative GBM cells (>50% growth inhibition). We detected the gene expression difference between osimertinib-sensitive and -resistant cells, found that osimertinib-sensitive GBM cells displayed activated MAPK-signaling pathway. We further showed that osimertinib potently inhibited the MNK kinase activities with IC50 values of 324 nM and 48.6 nM, respectively, against MNK1 and MNK2 kinases; osimertinib (0.3-3 µM) dose-dependently suppressed the phosphorylation of eukaryotic translation initiation factor 4E (eIF4E). In GBM patient-derived xenografts mice, oral administration of osimertinib (40 mg· kg-1 ·d-1, for 18 days) significantly suppressed the tumor growth (TGI = 74.5%) and inhibited eIF4E phosphorylation in tumor cells. Given the fact that osimertinib could cross the blood-brain barrier and its toxicity was well tolerated in patients, our results suggest that osimertinib could be a new and effective drug candidate for the EGFR-negative GBM patients.


Subject(s)
Acrylamides/therapeutic use , Aniline Compounds/therapeutic use , Antineoplastic Agents/therapeutic use , Glioblastoma/drug therapy , MAP Kinase Signaling System/drug effects , Protein Kinase Inhibitors/therapeutic use , Adolescent , Adult , Aged , Animals , Cell Proliferation/drug effects , Cells, Cultured , Child , ErbB Receptors/deficiency , Eukaryotic Initiation Factor-4E/chemistry , Eukaryotic Initiation Factor-4E/metabolism , Female , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Male , Mice , Middle Aged , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , Xenograft Model Antitumor Assays , Young Adult
2.
Biochem Pharmacol ; 178: 114095, 2020 08.
Article in English | MEDLINE | ID: mdl-32535106

ABSTRACT

Several different mechanisms are implicated in the resistance of lung cancer cells to epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKIs), and only few have been functionally investigated. Here, using genetically knocked out EGFR and TKI-resistant lung cancer cells, we show that loss of wild-type EGFR attenuates cell proliferation, migration and 3D-spheroid formation, whereas loss of mutant EGFR or resistance to TKIs reinforces those processes. Consistently, disruption of wild-type EGFR leads to suppression of HER2/HER3, while mutant EGFR ablation or resistance to TKIs increases HER2/HER3 expression, compensating for EGFR loss. Furthermore, HER2/HER3 nuclear translocation mediates overexpression of cyclin D1, leading to tumor cell survival and drug resistance. Cyclin D1/CDK4/6 inhibition resensitizes erlotinib-resistant (ER) cells to erlotinib. Analysis of cyclin D1 expression in patients with non-small cell lung carcinoma (NSCLC) showed that its expression is negatively associated with overall survival and disease-free survival. Our results provide biological and mechanistic insights into targeting EGFR and TKI resistance.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Cyclin D1/genetics , Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , Receptor, ErbB-2/genetics , Receptor, ErbB-3/genetics , Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cyclin D1/metabolism , Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/genetics , Cyclin-Dependent Kinase 6/metabolism , Drug Resistance, Neoplasm/genetics , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , ErbB Receptors/deficiency , ErbB Receptors/genetics , Erlotinib Hydrochloride/therapeutic use , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Protein Kinase Inhibitors/therapeutic use , Receptor, ErbB-2/metabolism , Receptor, ErbB-3/metabolism , Signal Transduction , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Survival Analysis , Transcriptional Activation
3.
J Dermatol ; 47(6): 663-668, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32250467

ABSTRACT

Epidermal growth factor receptor (EGFR) is a transmembrane glycoprotein with tyrosine-kinase signaling activity, involved in many cellular functions including cell growth and differentiation. Germ line loss-of-function mutations in EGFR lead to a severe neonatal skin disorder (Online Mendelian Inheritance in Man #131550). We report 18 premature Roma children from 16 families with birthweights ranging 440-1470 g and multisystem diseases due to the homozygous mutation c.1283G˃A (p.Gly428Asp) in EGFR. They presented with thin, translucent, fragile skin (14/15), skin desquamation (10/17), ichthyosis (9/17), recurrent skin infections and sepsis (9/12), nephromegaly (10/16) and congenital heart defects (7/17). Their prognosis was poor, and all died before the age of 6 months except one 13-year-old boy with a severe skin disorder, dentinogenesis imperfecta, Fanconi-like syndrome and secondary hyperaldosteronism. Management of ion and water imbalances and extremely demanding skin care may improve the unfavorable outcome of such patients.


Subject(s)
Dentinogenesis Imperfecta/mortality , Heart Defects, Congenital/mortality , Ichthyosis/mortality , Kidney Diseases/mortality , Roma/genetics , Adolescent , Child , Child, Preschool , Czech Republic/epidemiology , Dentinogenesis Imperfecta/diagnosis , Dentinogenesis Imperfecta/genetics , ErbB Receptors/deficiency , ErbB Receptors/genetics , Heart Defects, Congenital/diagnosis , Heart Defects, Congenital/genetics , Homozygote , Humans , Ichthyosis/diagnosis , Ichthyosis/genetics , Infant , Infant, Newborn , Infant, Premature , Infant, Very Low Birth Weight , Kidney Diseases/congenital , Kidney Diseases/diagnosis , Kidney Diseases/genetics , Loss of Function Mutation , Severity of Illness Index , Slovakia/epidemiology , Syndrome , Exome Sequencing
5.
Sci Rep ; 10(1): 453, 2020 01 16.
Article in English | MEDLINE | ID: mdl-31949213

ABSTRACT

Mature microRNAs are short non-coding RNA sequences which upon incorporation into the RISC ribonucleoprotein complex, play a crucial role in regulation of gene expression. However, miRNAs can exist within the cell also as free molecules fulfilling their biological activity. Therefore, it is emerging that in addition to sequence even the structure adopted by mature miRNAs might play an important role to reach the target. Indeed, we analysed by several spectroscopic techniques the secondary structures of two artificial miRNAs selected by computational tool (miR-Synth) as best candidates to silence c-MET and EGFR genes and of two endogenous miRNAs (miR-15a and miR-15b) having the same seed region, but different biological activity. Our results demonstrate that both endogenous and artificial miRNAs can arrange in several 3D-structures which affect their activity and selectivity toward the targets.


Subject(s)
MicroRNAs/chemistry , MicroRNAs/genetics , Base Sequence , ErbB Receptors/deficiency , ErbB Receptors/genetics , Gene Silencing , Nucleic Acid Conformation , Proto-Oncogene Proteins c-met/deficiency , Proto-Oncogene Proteins c-met/genetics , Sequence Analysis, RNA
6.
Sci Transl Med ; 11(522)2019 12 11.
Article in English | MEDLINE | ID: mdl-31826981

ABSTRACT

Epidermal growth factor receptor (EGFR)-targeted anticancer therapy induces stigmatizing skin toxicities affecting patients' quality of life and therapy adherence. The lack of mechanistic details underlying these adverse events hampers their management. We found that EGFR/ERK signaling is required in LRIG1-positive stem cells during de novo hair eruption to secure barrier integrity and prevent the invasion of commensal microbiota and inflammatory skin disease. EGFR-deficient epidermis is permissive for microbiota outgrowth and displays an atopic-like TH2-dominated signature. The opening of the follicular ostia during hair eruption allows invasion of commensal microbiota into the hair follicle, initiating an additional TH1 and TH17 response culminating in chronic folliculitis. Restoration of epidermal ERK signaling via prophylactic FGF7 treatment or transgenic SOS expression rescues the barrier defect in the absence of EGFR, highlighting a therapeutic anchor point. These data reveal that commensal skin microbiota provoke atopic-like inflammatory skin diseases by invading into the follicular opening of erupting hair.


Subject(s)
Antineoplastic Agents/adverse effects , ErbB Receptors/antagonists & inhibitors , Hair/pathology , Microbiota , Skin/microbiology , Animals , Epidermis/pathology , ErbB Receptors/deficiency , Fibroblast Growth Factor 7/metabolism , Humans , Inflammation/pathology , Keratinocytes/pathology , MAP Kinase Signaling System , Membrane Glycoproteins/metabolism , Mice , Nerve Tissue Proteins/metabolism , Skin/pathology
7.
Eur J Med Chem ; 172: 36-47, 2019 Jun 15.
Article in English | MEDLINE | ID: mdl-30939352

ABSTRACT

A series of novel 2,4-disubstituted quinazolines were synthesized and evaluated for their anti-tumor activity against five human cancer cells (MDA-MB-231, MCF-7, PC-3, HGC-27 and MGC-803) using MTT assay. Among them, compound 9n showed the most potent cytotoxicity against breast cancer cells. Compound 9n also significantly inhibited the colony formation and migration of MDA-MB-231 and MCF-7 cells. Meanwhile, compound 9n induced cell cycle arrest at G1 phase and cell apoptosis, as well as increased accumulation of intracellular ROS. Furthermore, compound 9n exerted anti-tumor effects in vitro via decreasing the expression of anti-apoptotic protein Bcl-2 and increasing the pro-apoptotic protein Bax and p53. Mechanistically, compound 9n markedly decreased p-EGFR and p-PI3K expression, which revealed that compound 9n targeted breast cancer cells via interfering with EGFR-PI3K signaling pathway. Molecular docking suggested that compound 9n could indeed bind into the active pocket of EGFR. All the findings suggest that compound 9n might be a valuable lead compound for anti-tumor agents targeting breast cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , ErbB Receptors/deficiency , ErbB Receptors/metabolism , Female , Humans , Molecular Docking Simulation , Molecular Structure , Phosphatidylinositol 3-Kinases/deficiency , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Quinazolines/chemical synthesis , Quinazolines/chemistry , Structure-Activity Relationship
8.
Glia ; 67(6): 1036-1046, 2019 06.
Article in English | MEDLINE | ID: mdl-30637799

ABSTRACT

We recently discovered a novel role for neuregulin-1 (Nrg1) signaling in mediating spontaneous regenerative processes and functional repair after spinal cord injury (SCI). We revealed that Nrg1 is the molecular signal responsible for spontaneous functional remyelination of dorsal column axons by peripheral nervous system (PNS)-like Schwann cells after SCI. Here, we investigate whether Nrg1/ErbB signaling controls the unusual transformation of centrally derived progenitor cells into these functional myelinating Schwann cells after SCI using a fate-mapping/lineage tracing approach. Specific ablation of Nrg1-ErbB receptors in central platelet-derived growth factor receptor alpha (PDGFRα)-derived lineage cells (using PDGFRαCreERT2/Tomato-red reporter mice crossed with ErbB3fl/fl/ErbB4fl/fl mice) led to a dramatic reduction in P0-positive remyelination in the dorsal columns following spinal contusion injury. Central myelination, assessed by Olig2 and proteolipid protein expression, was unchanged. Loss of ErbB signaling in PDGFRα lineage cells also significantly impacted the degree of spontaneous locomotor recovery after SCI, particularly in tests dependent on proprioception. These data have important implications, namely (a) cells from the PDGFRα-expressing progenitor lineage (which are presumably oligodendrocyte progenitor cells, OPCs) can differentiate into remyelinating PNS-like Schwann cells after traumatic SCI, (b) this process is controlled by ErbB tyrosine kinase signaling, and (c) this endogenous repair mechanism has significant consequences for functional recovery after SCI. Thus, ErbB tyrosine kinase receptor signaling directly controls the transformation of OPCs from the PDGFRα-expressing lineage into PNS-like functional remyelinating Schwann cells after SCI.


Subject(s)
ErbB Receptors/deficiency , Oligodendrocyte Precursor Cells/metabolism , Recovery of Function/physiology , Remyelination/physiology , Signal Transduction/physiology , Spinal Cord Injuries/metabolism , Animals , ErbB Receptors/genetics , Mice , Mice, Transgenic , Schwann Cells/metabolism , Spinal Cord Injuries/pathology
9.
J Cancer Res Clin Oncol ; 144(8): 1413-1422, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29797219

ABSTRACT

PURPOSE: AZD9291 is an irreversible, small-molecule inhibitor which has potency against mutant EGFR- and T790M-resistant mutation. Despite the encouraging efficacy in clinical, the acquired resistance will finally occur. Further study will need to be done to identify the acquired resistance mechanisms and determine the next treatment. METHODS: We established an AZD9291-resistant cell line (HCC827/AZDR) from parental HCC827 cell line through stepwise pulsed selection of AZD9291. The expression of EGFR and its downstream pathways were determined by western blot analysis or immunofluorescence assay. The sensitivity to indicated agents were evaluated by MTS. RESULTS: Compared with parental HCC827 cells, the HCC827/AZDR cells showed high resistance to AZD9291 and other EGFR-TKIs, and exhibited a mesenchymal-like phenotype. Almost complete loss of EGFR expression was observed in HCC827/AZDR cells. But the activation of downstream pathway, MAPK signaling, was found in HCC827/AZDR cells even in the presence of AZD9291. Inhibition of MAPK signaling had no effect on cell viability of HCC827/AZDR and could not reverse AZD9291 resistance because of the subsequent activation of AKT signaling. When treated with the combination of AKT and MAPK inhibitor, HCC827/AZDR showed remarkable growth inhibition. CONCLUSIONS: Loss of EGFR could be proposed as a potential acquired resistance mechanism of AZD9291 in EGFR-mutant NSCLC cells with an EMT phenotype. Despite the loss of EGFR, the activation of MAPK pathway which had crosstalk with AKT pathway could maintain the proliferation and survival of resistant cells. Blocking MAPK and AKT signaling may be a potential therapeutic strategy following AZD9291 resistance.


Subject(s)
Acrylamides/pharmacology , Aniline Compounds/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/enzymology , Lung Neoplasms/drug therapy , Lung Neoplasms/enzymology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Drug Resistance, Neoplasm , Epithelial-Mesenchymal Transition , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/deficiency , ErbB Receptors/genetics , Exons , Gene Deletion , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism
10.
Arterioscler Thromb Vasc Biol ; 38(1): 114-119, 2018 01.
Article in English | MEDLINE | ID: mdl-29191921

ABSTRACT

OBJECTIVE: To determine the consequences of specific inhibition of EGFR (epidermal growth factor receptor) in myeloid cells in atherosclerosis development. APPROACH AND RESULTS: Atherosclerotic lesion size was significantly reduced in irradiated Ldlr-/- mice reconstituted with LysMCre+Egfrlox/lox bone marrow, compared with chimeric Ldlr-/- mice reconstituted with LysMCre-Egfrlox/lox bone marrow, after 4 (-43%; P<0.05), 7 (-34%; P<0.05), and 12 weeks (-54%; P<0.001) of high-fat diet. Reduction of lesion size was associated with marked reduction in macrophage accumulation and necrotic core size. Specific deletion of Egfr in myeloid cells reduced TNF-α (tumor necrosis factor-α) and IL (interleukin)-6 production by stimulated macrophages but had no effect on IL-10 and IL-12p70 secretion. Finally, we found that myeloid deletion of Egfr limited cytoskeletal rearrangements and also lipid uptake by macrophages through a downregulation of the scavenger receptor CD36 (cluster of differentiation 36). CONCLUSIONS: Gene deletion of Egfr in myeloid cells limits IL-6 and TNF-α production, lipid uptake, and consecutively reduces atherosclerosis development.


Subject(s)
Atherosclerosis/prevention & control , ErbB Receptors/deficiency , Gene Deletion , Macrophages/metabolism , Animals , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Bone Marrow Transplantation , CD36 Antigens/metabolism , Cytoskeleton/metabolism , Cytoskeleton/pathology , Diet, High-Fat , Disease Models, Animal , ErbB Receptors/genetics , Interleukin-6/metabolism , Macrophages/pathology , Macrophages/transplantation , Male , Mice, Knockout , Necrosis , Plaque, Atherosclerotic , Receptors, LDL/deficiency , Receptors, LDL/genetics , Tumor Necrosis Factor-alpha/metabolism , Whole-Body Irradiation
11.
Acta Physiol (Oxf) ; 222(3)2018 03.
Article in English | MEDLINE | ID: mdl-29152859

ABSTRACT

AIM: In vivo vascular smooth muscle cell (VSMC) EGF receptor (EGFR) contributes to acute angiotensin II (AII) effects on vascular tone and blood pressure. The ubiquitously expressed EGFR has been implicated in vascular remodelling preceding end-organ damage by pharmacological inhibition, and AII signalling in cultured vascular cells is partly EGFR-dependent. However, the role of VSMC-EGFR in vivo during AII-induced pathophysiological processes is not known. METHODS: This study assesses the in vivo relevance of VSMC-EGFR during chronic AII challenge without further stressors, using a mouse model with inducible, VSMC-specific EGFR knock out (VSMC-EGFR-KO). In these mice functional and structural vascular, renal and cardiac effects or biomarkers were investigated in vivo and ex vivo. RESULTS: Vascular smooth muscle cell-EGFR-KO prevented AII-induced media hypertrophy of mesenteric arteries, renal arterioles and the aorta, VSMC ERK1/2-phosphorylation as well as the impairment of vascular compliance. Furthermore, induction of vascular fibrosis, creatinineamia, renal interstitial fibrosis as well as the increase in fractional water excretion was prevented. AII-induced increase in systolic blood pressure was mitigated. By contrast, endothelial dysfunction, induction of vascular inflammatory marker mRNA and albuminuria were not inhibited. Cardiac and cardiomyocyte hypertrophy were also not prevented by VSMC-EGFR-KO. CONCLUSION: Vascular smooth muscle cell-EGFRs are relevant for pathological AII action in vivo. Our data show in vivo and ex vivo the necessity of VSMC-EGFR for AII-induced structural and functional vascular remodelling, not including endothelial dysfunction. Hereby, VSMC-EGFR gains importance for complete AII-induced renal end-organ damage succeeding vascular remodelling.


Subject(s)
Angiotensin II/pharmacology , ErbB Receptors/deficiency , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Vascular Remodeling/drug effects , Vascular Stiffness/drug effects , Animals , Hypertrophy , Mice , Mice, Knockout , Muscle, Smooth, Vascular/pathology , Tunica Media/pathology
12.
J Virol ; 91(12)2017 06 15.
Article in English | MEDLINE | ID: mdl-28381567

ABSTRACT

The Cbl E3 ligase has been linked to the down-modulation of surface signaling responses by inducing internalization of surface receptors. The adaptor protein CIN85 is a partner of Cbl that augments many of these interactions. Previously, an interaction was demonstrated between ICP0 and CIN85, which results in the removal of epidermal growth factor receptor (EGFR) from the surface of the infected cells with a concomitant attenuation of EGFR signaling. Here, we examined whether Cbl mediates the removal of the herpes simplex virus 1 (HSV-1) entry receptor Nectin-1 from the surface of infected cells. We found the following: (i) that Cbl, Nectin-1, and the viral glycoprotein D (gD) form a complex in infected cells; (ii) that during infection Nectin-1 is removed from the surface of the infected cells but is retained on the surface of cells that have been depleted of Cbl; and (iii) that in cells infected with a ΔICP0 mutant virus, Nectin-1 remained on the cell surface. Thus, Cbl is necessary but not sufficient for the removal of Nectin-1 from the cell surface. In addition, we observed that in Cbl-depleted cells there was enhanced entry after infection. These cells were susceptible to secondary infections by HSV-1. Viral entry in CIN85-depleted cells was only moderately enhanced compared to that in the Cbl-depleted cells, suggesting that the Cbl-Nectin-1 interaction is likely the key to the downregulation of surface Nectin-1. The removal of the HSV-1 entry receptor Nectin-1 from the surface of the infected cells may be part of the strategy of the virus to efficiently spread to uninfected cells.IMPORTANCE The Cbl E3 ligase suppresses surface signaling responses by inducing internalization of surface components. The targets of Cbl include such components as immune system receptors, growth factor receptors, adhesion, and cell-to-cell contact molecules. The immediate early protein ICP0 of herpes simplex virus 1 (HSV-1) interacts with CIN85, an adaptor protein that augments Cbl functions. The consequence of this interaction is the removal of the epidermal growth factor receptor (EGFR) from the surface of the infected cells with concomitant suppression of the EGF ligand signaling. The viral entry receptor Nectin-1 is also internalized during HSV-1 infection in a Cbl-dependent mechanism, and that increases the opportunity of the virus to spread to uninfected cells. The diversion of the Cbl/CIN85 endocytic machinery may be a strategy utilized by the virus to alter the cell surface pattern to prevent detrimental host responses.


Subject(s)
Cell Adhesion Molecules/metabolism , Herpesvirus 1, Human/physiology , Proto-Oncogene Proteins c-cbl/metabolism , Virus Internalization , Adaptor Proteins, Signal Transducing/metabolism , Cell Adhesion Molecules/deficiency , Cell Adhesion Molecules/genetics , Endocytosis , ErbB Receptors/deficiency , ErbB Receptors/genetics , ErbB Receptors/metabolism , HEK293 Cells , Hep G2 Cells , Herpesvirus 1, Human/genetics , Humans , Immediate-Early Proteins/metabolism , Nectins , Proto-Oncogene Proteins c-cbl/genetics , Signal Transduction , Ubiquitin-Protein Ligases/metabolism , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism
13.
Zhongguo Fei Ai Za Zhi ; 20(3): 143-148, 2017 Mar 20.
Article in Chinese | MEDLINE | ID: mdl-28302215

ABSTRACT

BACKGROUND: Targeting the mutations and amplifications in the epidermal growth factor receptor (EGFR) gene has curative effects on cancers of the lung, oral cavity, and gastrointestinal system. However, a systemic immune inflammation is an adverse effect of this therapeutic strategy. In this study, we aimed to identify the possible changes in the tumor microenvironment that contribute to the anti-cancer activity of EGFR inhibition. METHODS: Squamous-cell cancers were induced by the syngeneic transplantation of either EGFR-null or wild-type mouse primary keratinocytes that had been transduced with an oncogenic H-ras retrovirus. The mice were treated with gefinitib. Then, flow cytometric was used to detect the ratio of T cells and the expression of programmed cell death receptor 1 (PD-1). RT-PCR was used to detect the expression of cytokines and chemokines. RESULTS: Tumors that formed from EGFR-null keratinocytes were smaller, had fewer infiltrating FoxP3+ Treg cells, lower Foxp3 RNA, and lower percentage of PD-1 positive CD4 cells than those formed from wild-type keratinocytes. These results indicated that tumor cells can autonomously regulate the tumor microenvironment. Hosts with wild-type cancers and that were treated with gefitinib for 1 week tended to have smaller tumors. The treated mice in the short-term pharmacological model tended to have reduced FoxP3+ cells and FoxP3 RNA in the tumor microenvironment, as well as a substantially increased ratio of IL-1A/IL-1RA transcripts. These results suggested that the brief systemic inhibition of EGFR signaling alters the immune environment of the targeted cancer. CONCLUSIONS: The autonomous (genetic) or systemic (pharmacologic) inhibition of EGFR signaling in tumor cells reduces tumor growth and Treg infiltration in the tumor microenvironment. An EGFR-dependent Treg function supports the growth of squamous cancers. Therefore, Treg is a target in the therapeutic strategy of EGFR inhibition.


Subject(s)
Carcinoma, Squamous Cell/immunology , ErbB Receptors/metabolism , Gene Expression Regulation, Neoplastic , Interleukin-1alpha/metabolism , Lung Neoplasms/immunology , T-Lymphocytes, Regulatory/immunology , Tumor Microenvironment/immunology , Animals , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Transformation, Neoplastic , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/deficiency , ErbB Receptors/genetics , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/immunology , Gene Knockout Techniques , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , RNA, Messenger/genetics , RNA, Messenger/metabolism , T-Lymphocytes, Regulatory/drug effects , Tumor Microenvironment/genetics
14.
Pathol Oncol Res ; 23(3): 673-677, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28025786

ABSTRACT

Genetic variability in KRAS and EGFR predicts response to cetuximab in irinotecan refractory colorectal cancer. Whether these markers or others remain predictive in combination biologic therapies including bevacizumab is unknown. We identified predictive biomarkers from patients with irinotecan refractory metastatic colorectal cancer treated with cetuximab plus bevacizumab. Patients who received cetuximab plus bevacizumab for irinotecan refractory colorectal cancer in either of two Phase II trials conducted were identified. Tumor tissue was available for 33 patients. Genomic DNA was extracted and used for mutational analysis of KRAS, BRAF, and p53 genes. Fluorescence in situ hybridization was performed to assess EGFR copy number. The status of single genes and various combinations were tested for association with response. Seven of 33 patients responded to treatment. KRAS mutations were found in 14/33 cases, and 0 responded to treatment (p = 0.01). EGFR gene amplification was seen in 3/33 of tumors and in every case was associated with response to treatment (p < 0.001). TP53 and BRAF mutations were found in 18/33 and 0/33 tumors, respectively, and there were no associations with response to either gene. EGFR gene amplification and KRAS mutations are predictive markers for patients receiving combination biologic therapy of cetuximab plus bevacizumab for metastatic colorectal cancer. One marker or the other is present in the tumor of half of all patients allowing treatment response to be predicted with a high degree of certainty. The role for molecular markers in combination biologic therapy seems promising.


Subject(s)
Colorectal Neoplasms/genetics , ErbB Receptors/deficiency , Gene Amplification/genetics , Mutation/genetics , ras Proteins/genetics , Adult , Aged , Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bevacizumab/administration & dosage , Biomarkers, Tumor/genetics , Camptothecin/administration & dosage , Camptothecin/analogs & derivatives , Cetuximab/administration & dosage , Colorectal Neoplasms/drug therapy , DNA Mutational Analysis/methods , Female , Genes, p53/genetics , Humans , Irinotecan , Male , Middle Aged
15.
Sci Rep ; 6: 38347, 2016 12 02.
Article in English | MEDLINE | ID: mdl-27910913

ABSTRACT

Hepatoblastoma (HBL), the most common childhood liver cancer is cured with surgical resection after chemotherapy or with liver transplantation if local invasion and multifocality preclude resection. However, variable survival rates of 60-80% and debilitating chemotherapy sequelae argue for more informed treatment selection, which is not possible by grading the Wnt-ß-catenin over activity present in most HBL tumors. A hypothesis-generating whole transcriptome analysis shows that HBL tumors removed at transplantation are enriched most for cancer signaling pathways which depend predominantly on epidermal growth factor (EGF) signaling, and to a lesser extent, on aberrant Wnt-ß-catenin signaling. We therefore evaluated whether EGFR, ASAP1, ERBB2 and ERBB4, which signal downstream after ligation of EGF, and which show aberrant expression in several other invasive cancers, would also predict HBL tumor invasiveness. Immunohistochemistry of HBL tumors (n = 60), which are histologically heterogeneous, shows that compared with well-differentiated fetal cells, less differentiated embryonal and undifferentiated small cells (SCU) progressively lose EGFR and ASAP1 expression. This trend is exaggerated in unresectable, locally invasive or metastatic tumors, in which embryonal tumor cells are EGFR-negative, while SCU cells are EGFR-negative and ASAP1-negative. Loss of EGFR-ASAP1 signaling characterizes undifferentiated and invasive HBL. EGFR-expressing HBL tumors present novel therapeutic targeting opportunities.


Subject(s)
Adaptor Proteins, Signal Transducing/deficiency , ErbB Receptors/deficiency , Gene Expression Regulation, Neoplastic , Hepatoblastoma/genetics , Liver Neoplasms/genetics , Lung Neoplasms/genetics , Adaptor Proteins, Signal Transducing/genetics , Adolescent , Child , Child, Preschool , Epidermal Growth Factor/genetics , Epidermal Growth Factor/metabolism , ErbB Receptors/genetics , Female , Hepatoblastoma/mortality , Hepatoblastoma/secondary , Hepatoblastoma/surgery , Humans , Infant , Liver Neoplasms/mortality , Liver Neoplasms/pathology , Liver Neoplasms/surgery , Liver Transplantation , Lung Neoplasms/mortality , Lung Neoplasms/secondary , Lung Neoplasms/surgery , Male , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Receptor, ErbB-4/genetics , Receptor, ErbB-4/metabolism , Survival Analysis , Transcriptome , Wnt Signaling Pathway , beta Catenin/genetics , beta Catenin/metabolism
16.
Proc Natl Acad Sci U S A ; 113(50): 14360-14365, 2016 12 13.
Article in English | MEDLINE | ID: mdl-27911782

ABSTRACT

Osteoarthritis (OA) is the most common joint disease, characterized by progressive destruction of the articular cartilage. The surface of joint cartilage is the first defensive and affected site of OA, but our knowledge of genesis and homeostasis of this superficial zone is scarce. EGFR signaling is important for tissue homeostasis. Immunostaining revealed that its activity is mostly dominant in the superficial layer of healthy cartilage but greatly diminished when OA initiates. To evaluate the role of EGFR signaling in the articular cartilage, we studied a cartilage-specific Egfr-deficient (CKO) mouse model (Col2-Cre EgfrWa5/flox). These mice developed early cartilage degeneration at 6 mo of age. By 2 mo of age, although their gross cartilage morphology appears normal, CKO mice had a drastically reduced number of superficial chondrocytes and decreased lubricant secretion at the surface. Using superficial chondrocyte and cartilage explant cultures, we demonstrated that EGFR signaling is critical for maintaining the number and properties of superficial chondrocytes, promoting chondrogenic proteoglycan 4 (Prg4) expression, and stimulating the lubrication function of the cartilage surface. In addition, EGFR deficiency greatly disorganized collagen fibrils in articular cartilage and strikingly reduced cartilage surface modulus. After surgical induction of OA at 3 mo of age, CKO mice quickly developed the most severe OA phenotype, including a complete loss of cartilage, extremely high surface modulus, subchondral bone plate thickening, and elevated joint pain. Taken together, our studies establish EGFR signaling as an important regulator of the superficial layer during articular cartilage development and OA initiation.


Subject(s)
Arthritis, Experimental/metabolism , Cartilage, Articular/metabolism , ErbB Receptors/metabolism , Osteoarthritis/metabolism , Animals , Arthritis, Experimental/pathology , Arthritis, Experimental/prevention & control , Cells, Cultured , Chondrocytes/metabolism , Chondrocytes/pathology , Chondrogenesis , ErbB Receptors/deficiency , ErbB Receptors/genetics , Humans , Male , Mice , Mice, Knockout , Osteoarthritis/pathology , Osteoarthritis/prevention & control , Proteoglycans/metabolism , Signal Transduction
17.
Clin Cancer Res ; 22(24): 6010-6020, 2016 Dec 15.
Article in English | MEDLINE | ID: mdl-27281561

ABSTRACT

PURPOSE: Genotype-directed therapy is the standard of care for advanced non-small cell lung cancer (NSCLC), but obtaining tumor tissue for genotyping remains a challenge. Circulating tumor cell (CTC) or cell-free DNA (cfDNA) analysis may allow for noninvasive evaluation. This prospective trial evaluated CTCs and cfDNA in EGFR-mutant NSCLC patients treated with erlotinib until progression. EXPERIMENTAL DESIGN: EGFR-mutant NSCLC patients were enrolled in a phase II trial of erlotinib. Blood was collected at baseline, every 2 months on study, and at disease progression. Plasma genotyping was performed by droplet digital PCR for EGFR19del, L858R, and T790M. CTCs were isolated by CellSave, enumerated, and analyzed by immunofluorescence for CD45 and pan-cytokeratin and EGFR and MET FISH were also performed. Rebiopsy was performed at disease progression. RESULTS: Sixty patients were enrolled; 44 patients discontinued therapy for disease progression. Rebiopsy occurred in 35 of 44 patients (80%), with paired CTC/cfDNA analysis in 41 of 44 samples at baseline and 36 of 44 samples at progression. T790M was identified in 23 of 35 (66%) tissue biopsies and 9 of 39 (23%) cfDNA samples. CTC analysis at progression identified MET amplification in 3 samples in which tissue analysis could not be performed. cfDNA analysis identified T790M in 2 samples in which rebiopsy was not possible. At diagnosis, high levels of cfDNA but not high levels of CTCs correlated with progression-free survival. CONCLUSIONS: cfDNA and CTCs are complementary, noninvasive assays for evaluation of acquired resistance to first-line EGFR TKIs and may expand the number of patients in whom actionable genetic information can be obtained at acquired resistance. Serial cfDNA monitoring may offer greater clinical utility than serial monitoring of CTCs. Clin Cancer Res; 22(24); 6010-20. ©2016 AACR.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Cell-Free Nucleic Acids/drug effects , ErbB Receptors/deficiency , Erlotinib Hydrochloride/therapeutic use , Lung Neoplasms/drug therapy , Neoplastic Cells, Circulating/drug effects , Adult , Aged , Aged, 80 and over , Carcinoma, Non-Small-Cell Lung/blood , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell-Free Nucleic Acids/genetics , DNA, Neoplasm/drug effects , DNA, Neoplasm/genetics , Disease Progression , Disease-Free Survival , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Female , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Middle Aged , Mutation/genetics , Neoplastic Cells, Circulating/pathology , Prospective Studies , Protein Kinase Inhibitors/therapeutic use
18.
J Proteome Res ; 15(5): 1402-17, 2016 05 06.
Article in English | MEDLINE | ID: mdl-27089454

ABSTRACT

Keratinocyte-specific deletion of ADAM17 in mice impairs terminal differentiation of keratinocytes leading to severe epidermal barrier defects. Mice deficient for ADAM17 in keratinocytes phenocopy mice with a keratinocyte-specific deletion of epidermal growth factor receptor (EGFR), which highlights the role of ADAM17 as a "ligand sheddase" of EGFR ligands. In this study, we aim for the first proteomic/degradomic approach to characterize the disruption of the ADAM17-EGFR signaling axis and its consequences for epidermal barrier formation. Proteomic profiling of the epidermal proteome of mice deficient for either ADAM17 or EGFR in keratinocytes at postnatal days 3 and 10 revealed highly similar protein alterations for ADAM17 and EGFR deficiency. These include massive proteome alterations of structural and regulatory components important for barrier formation such as transglutaminases, involucrin, filaggrin, and filaggrin-2. Cleavage site analysis using terminal amine isotopic labeling of substrates revealed increased proteolytic processing of S100 fused-type proteins including filaggrin-2. Alterations in proteolytic processing are supported by altered abundance of numerous proteases upon keratinocyte-specific Adam17 or Egfr deletion, among them kallikreins, cathepsins, and their inhibitors. This study highlights the essential role of proteolytic processing for maintenance of a functional epidermal barrier. Furthermore, it suggests that most defects in formation of the postnatal epidermal barrier upon keratinocyte-specific ADAM17 deletion are mediated via EGFR.


Subject(s)
ADAM17 Protein/deficiency , ErbB Receptors/deficiency , Keratinocytes , Proteome/metabolism , ADAM17 Protein/genetics , Animals , Epidermis/pathology , ErbB Receptors/genetics , Gene Deletion , Mice , Proteolysis , Proteome/analysis
19.
Biochim Biophys Acta ; 1863(7 Pt A): 1519-33, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27012600

ABSTRACT

Besides their importance for the vascular tone, vascular smooth muscle cells (VSMC) also contribute to pathophysiological vessel alterations. Various G-protein coupled receptor ligands involved in vascular dysfunction and remodeling can transactivate the epidermal growth factor receptor (EGFR) of VSMC, yet the importance of EGFR transactivation for the VSMC phenotype is incompletely understood. The aims of this study were (i) to characterize further the importance of the VSMC-EGFR for proliferation, migration and marker gene expression for inflammation, fibrosis and reactive oxygen species (ROS) homeostasis and (ii) to test the hypothesis that vasoactive substances (endothelin-1, phenylephrine, thrombin, vasopressin and ATP) rely differentially on the EGFR with respect to the abovementioned phenotypic alterations. In primary, aortic VSMC from mice without conditional deletion of the EGFR, proliferation, migration, marker gene expression (inflammation, fibrosis and ROS homeostasis) and cell signaling (ERK 1/2, intracellular calcium) were analyzed. VSMC-EGFR loss reduced collective cell migration and single cell migration probability, while no difference between the genotypes in single cell velocity, chemotaxis or marker gene expression could be observed under control conditions. EGF promoted proliferation, collective cell migration, chemokinesis and chemotaxis and leads to a proinflammatory gene expression profile in wildtype but not in knockout VSMC. Comparing the impact of five vasoactive substances (all reported to transactivate EGFR and all leading to an EGFR dependent increase in ERK1/2 phosphorylation), we demonstrate that the importance of EGFR for their action is substance-dependent and most apparent for crowd migration but plays a minor role for gene expression regulation.


Subject(s)
Cell Movement , ErbB Receptors/metabolism , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , Adenosine Triphosphate/pharmacology , Animals , Cell Movement/drug effects , Cell Proliferation , Cells, Cultured , Endothelin-1/pharmacology , Enzyme Activation , Epidermal Growth Factor/pharmacology , ErbB Receptors/agonists , ErbB Receptors/deficiency , ErbB Receptors/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibrosis , Gene Expression Regulation , Genotype , Inflammation/genetics , Inflammation/metabolism , Ligands , Mice, Knockout , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Oxidative Stress , Phenotype , Phenylephrine/pharmacology , Primary Cell Culture , Signal Transduction , Thrombin/pharmacology , Time Factors , Vasopressins/pharmacology
20.
Clin Sci (Lond) ; 130(1): 19-33, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26438881

ABSTRACT

Epi dermal growth factor (EGF) receptor (EGFR) is activated by its canonical ligands and transactivated by various vasoactive substances, e.g. angiotensin II (Ang II). Vascular EGFR has been proposed to be involved in vascular tissue homoeostasis and remodelling. Thus, most studies have focused on its role during long-term vascular changes whereas the relevance for acute regulation of vascular function in vivo and ex vivo is insufficiently understood. To investigate the postnatal role of VSMCs (vascular smooth muscle cells) EGFR in vivo and ex vivo, we generated a mouse model with cell-specific and inducible deletion of VSMC EGFR and studied the effect on basal blood pressure, acute pressure response to, among others, Ang II in vivo as well as ex vivo, cardiovascular tissue homoeostasis and vessel morphometry in male mice. In knockout (KO) animals, systolic, diastolic and mean blood pressures were reduced compared with wild-type (WT). Furthermore, Ang II-induced pressure load was lower in KO animals, as was Ang II-induced force development and extracellular-signal-regulated kinase 1 and 2 (ERK1/2) phosphorylation in aortic rings from KO animals. By contrast, we observed no difference in force development during application of serotonin, KCl, endothelin-1 or endothelin-1-induced pressure load in KO animals. In addition, nitric oxide (NO)-mediated vasodilation was not affected. Heart weight (HW) increase and up-regulation of aortic and cardiac expression of Ccl2 (chemoattractant protein-2) and serpinE1 (plasminogen activator inhibitor 1) during the transition from 4- to 10-months of age were prevented by VSMC EGFR KO. We conclude that VSMC EGFR is involved in basal blood pressure homoeostasis and acute pressure response to Ang II, and thereby contributes to maturation-related remodelling.


Subject(s)
Angiotensin II , Blood Pressure , ErbB Receptors/deficiency , Hypertension/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Age Factors , Animals , Blood Pressure/drug effects , Cardiomegaly/chemically induced , Cardiomegaly/genetics , Cardiomegaly/metabolism , Cardiomegaly/physiopathology , Chemokine CCL2/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , ErbB Receptors/genetics , Gene Deletion , Humans , Hypertension/chemically induced , Hypertension/genetics , Hypertension/physiopathology , Hypertension/prevention & control , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiopathology , Myocytes, Smooth Muscle/drug effects , Phosphorylation , Plasminogen Activator Inhibitor 1/metabolism , Signal Transduction , Time Factors , Vascular Remodeling , Vasoconstrictor Agents/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...