Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Respir Res ; 24(1): 304, 2023 Dec 06.
Article in English | MEDLINE | ID: mdl-38053045

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a progressive and fatal lung disease with a poor prognosis. Current/available clinical prediction tools have limited sensitivity and accuracy when evaluating clinical outcomes of IPF. Research has shown that focal adhesion kinase (FAK), produced by the protein tyrosine kinase 2 (PTK2) gene, is crucial in IPF development. FAK activation is a characteristic of lesional fibroblasts; Thus, FAK may be a valuable therapeutic target or prognostic biomarker for IPF. This study aimed to create a gene signature based on PTK2-associated genes and microarray data from blood cells to predict disease prognosis in patients with IPF. PTK2 levels were found to be higher in lung tissues of IPF patients compared to healthy controls, and PTK2 inhibitor Defactinib was found to reduce TGFß-induced FAK activation and increase α-smooth muscle actin. Although the blood PTK2 levels were higher in IPF patients, blood PTK level alone could not predict IPF prognosis. From 196 PTK2-associated genes, 11 genes were prioritized to create a gene signature (PTK2 molecular signature) and a risk score system using univariate and multivariate Cox regression analysis. Patients were divided into high-risk and low-risk groups using PTK2 molecular signature. Patients in the high-risk group experienced decreased survival rates compared to patients in the low-risk group across all discovery and validation cohorts. Further functional enrichment and immune cell proportion analyses revealed that the PTK2 molecular signature strongly reflected the activation levels of immune pathways and immune cells. These findings suggested that PTK2 is a molecular target of IPF and the PTK2 molecular signature is an effective IPF prognostic biomarker.


Subject(s)
Idiopathic Pulmonary Fibrosis , Humans , Focal Adhesion Kinase 1/genetics , Focal Adhesion Kinase 1/metabolism , Idiopathic Pulmonary Fibrosis/diagnosis , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/metabolism , Lung/metabolism , Prognosis , Biomarkers/metabolism
2.
Chem Biol Drug Des ; 95(6): 584-599, 2020 06.
Article in English | MEDLINE | ID: mdl-32080977

ABSTRACT

The N-terminal FERM domain of focal adhesion kinase (FAK) contributes to FAK scaffolding and interacts with HER2, an oncogene and receptor tyrosine kinase. The interaction between HER2 and FAK drives resistance to FAK-kinase domain inhibitors through FAK Y397 transphosphorylation and FAK re-activation upon inhibition. As such, FAK FERM remains an attractive drug discovery target. In this report, we detail an alternative approach to targeting FAK through virtual screening-based discovery of chemical probes that target FAK FERM. We validated the binding interface between HER2 and FAK using site-directed mutagenesis and GST pull-down experiments. We assessed the ligandability of key-binding residues of HER2 and FAK utilizing computational tools. We developed a virtual screening method to screen ~200,000 compounds against the FAK FERM domain, identifying 20 virtual chemical probes. We performed GST pull-down screening on these compounds, discovering two hits, VS4 and VS14, with nanomolar IC50 s in disrupting HER2-FAK. We performed further testing, including molecular docking, immunofluorescence, phosphorylation, and cellular invasion assays to evaluate the compounds' biological effects. One probe, VS14, was identified with the ability to block both auto- and transphosphorylation of Y397. In all, these studies identify two new probes that target FAK FERM, enabling future investigation of this domain.


Subject(s)
Focal Adhesion Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor, ErbB-2/metabolism , Small Molecule Libraries/chemistry , Amino Acid Sequence , FERM Domains , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Humans , Ligands , Molecular Docking Simulation , Mutagenesis , Neoplasms , Phosphorylation , Protein Binding , Signal Transduction , Small Molecule Libraries/pharmacology
3.
SLAS Discov ; 25(1): 21-32, 2020 01.
Article in English | MEDLINE | ID: mdl-31513463

ABSTRACT

Focal adhesion kinase (FAK) is a promising cancer drug target due to its massive overexpression in multiple solid tumors and its critical role in the integration of signals that control proliferation, invasion, apoptosis, and metastasis. Previous FAK drug discovery and high-throughput screening have exclusively focused on the identification of inhibitors that target the kinase domain of FAK. Because FAK is both a kinase and scaffolding protein, the development of novel screening assays that detect inhibitors of FAK protein-protein interactions remains a critical need. In this report, we describe the development of a high-throughput fluorescence polarization (FP) screening assay that measures the interactions between FAK and paxillin, a focal adhesion-associated protein. We designed a tetramethylrhodamine (TAMRA)-tagged paxillin peptide based on the paxillin LD2 motif that binds to the focal adhesion targeting (FAT) domain with significant dynamic range, specificity, variability, stability, and a Z'-factor suitable for high-throughput screening. In addition, we performed a pilot screen of 1593 compounds using this FP assay, showing its feasibility for high-throughput drug screening. Finally, we identified three compounds that show dose-dependent competition of FAT-paxillin binding. This assay represents the first described high-throughput screening assay for FAK scaffold inhibitors and can accelerate drug discovery efforts for this promising drug target.


Subject(s)
Drug Discovery , Fluorescence Polarization , Focal Adhesion Kinase 1/metabolism , High-Throughput Screening Assays , Paxillin/metabolism , Protein Binding/drug effects , Drug Discovery/methods , Fluorescence Polarization/methods , Focal Adhesion Kinase 1/chemistry , Humans , Models, Molecular , Molecular Conformation , Paxillin/chemistry , Protein Interaction Domains and Motifs , Structure-Activity Relationship
4.
Molecules ; 24(18)2019 Sep 14.
Article in English | MEDLINE | ID: mdl-31540099

ABSTRACT

The Focal Adhesion Targeting (FAT) domain of Focal Adhesion Kinase (FAK) is a promising drug target since FAK is overexpressed in many malignancies and promotes cancer cell metastasis. The FAT domain serves as a scaffolding protein, and its interaction with the protein paxillin localizes FAK to focal adhesions. Various studies have highlighted the importance of FAT-paxillin binding in tumor growth, cell invasion, and metastasis. Targeting this interaction through high-throughput screening (HTS) provides a challenge due to the large and complex binding interface. In this report, we describe a novel approach to targeting FAT through fragment-based drug discovery (FBDD). We developed two fragment-based screening assays-a primary SPR assay and a secondary heteronuclear single quantum coherence nuclear magnetic resonance (HSQC-NMR) assay. For SPR, we designed an AviTag construct, optimized SPR buffer conditions, and created mutant controls. For NMR, resonance backbone assignments of the human FAT domain were obtained for the HSQC assay. A 189-compound fragment library from Enamine was screened through our primary SPR assay to demonstrate the feasibility of a FAT-FBDD pipeline, with 19 initial hit compounds. A final total of 11 validated hits were identified after secondary screening on NMR. This screening pipeline is the first FBDD screen of the FAT domain reported and represents a valid method for further drug discovery efforts on this difficult target.


Subject(s)
Focal Adhesion Kinase 1/chemistry , Focal Adhesions/chemistry , Nuclear Magnetic Resonance, Biomolecular , Surface Plasmon Resonance , Drug Discovery , Focal Adhesion Kinase 1/genetics , Focal Adhesions/genetics , Humans , Paxillin/chemistry , Paxillin/genetics , Protein Domains
5.
BMC Mol Cell Biol ; 20(1): 10, 2019 05 20.
Article in English | MEDLINE | ID: mdl-31109284

ABSTRACT

BACKGROUND: Focal Adhesion Kinase (FAK) is a major cancer drug target that is involved in numerous aspects of tumor progression and survival. While multiple research groups have developed ATP-competitive small molecule inhibitors that target the kinase enzyme, recent attention has been focused on the FAK FERM (Band 4.1, Ezrin, Radixin, Moesin) domain that contains key residue Y397 and contributes to many protein-protein interactions. Previous x-ray crystal structures of the FAK FERM domain gave conflicting results on the structure of the Y397 region and therefore the overall druggability. RESULTS: Here, we report the identification of a higher resolution crystal structure of the avian FAK FERM domain that shows conformational differences in Y397 and surrounding residues in the F1 lobe. In addition, we resolve the residues of the Src SH3 binding site, an area of the FERM domain that has previously shown limited electron density. CONCLUSIONS: These crystallographic data suggest that the Y397 region is highly dynamic and question the druggability of a putative pocket on the F1 lobe. In addition, new electron density data around the Src SH3 binding site provide structural insight on the FAK-Src activation cascade through a putative auto-inhibitory conformation.


Subject(s)
Avian Proteins/chemistry , Crystallization , FERM Domains , Focal Adhesion Protein-Tyrosine Kinases/chemistry , Proto-Oncogene Proteins pp60(c-src)/chemistry , Tyrosine/chemistry , src Homology Domains , Animals , Binding Sites , Computer Simulation , Crystallography, X-Ray , Cytoskeletal Proteins/chemistry , Dimerization , Drug Discovery/methods , Focal Adhesion Protein-Tyrosine Kinases/antagonists & inhibitors , Membrane Proteins/chemistry , Microfilament Proteins/chemistry , Phosphorylation , Protein Binding , Protein Structure, Secondary
6.
Mol Cancer Ther ; 15(12): 3028-3039, 2016 12.
Article in English | MEDLINE | ID: mdl-27638858

ABSTRACT

Focal adhesion kinase (FAK) is a major drug target in cancer and current inhibitors targeted to the ATP-binding pocket of the kinase domain have entered clinical trials. However, preliminary results have shown limited single-agent efficacy in patients. Despite these unfavorable data, the molecular mechanisms that drive intrinsic and acquired resistance to FAK-kinase inhibitors are largely unknown. We have demonstrated that receptor tyrosine kinases (RTK) can directly bypass FAK-kinase inhibition in cancer cells through phosphorylation of FAK's critical tyrosine 397 (Y397). We also showed that HER2 forms a direct protein-protein interaction with the FAK-FERM-F1 lobe, promoting direct phosphorylation of Y397. In addition, FAK-kinase inhibition induced two forms of compensatory RTK reprogramming: (i) the rapid phosphorylation and activation of RTK signaling pathways in RTKHigh cells and (ii) the long-term acquisition of RTKs novel to the parental cell line in RTKLow cells. Finally, HER2 +: cancer cells displayed resistance to FAK-kinase inhibition in 3D growth assays using a HER2 isogenic system and HER2+ cancer cell lines. Our data indicate a novel drug resistance mechanism to FAK-kinase inhibitors whereby HER2 and other RTKs can rescue and maintain FAK activation (pY397) even in the presence of FAK-kinase inhibition. These data may have important ramifications for existing clinical trials of FAK inhibitors and suggest that individual tumor stratification by RTK expression would be important to predict patient response to FAK-kinase inhibitors. Mol Cancer Ther; 15(12); 3028-39. ©2016 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Focal Adhesion Protein-Tyrosine Kinases/antagonists & inhibitors , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Protein Kinase Inhibitors/pharmacology , Receptor Protein-Tyrosine Kinases/metabolism , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Movement/genetics , ErbB Receptors/chemistry , ErbB Receptors/metabolism , Focal Adhesion Protein-Tyrosine Kinases/chemistry , Focal Adhesion Protein-Tyrosine Kinases/genetics , Gene Knockout Techniques , Humans , Phosphorylation , Protein Binding , Protein Kinase Inhibitors/chemistry , Receptor Protein-Tyrosine Kinases/chemistry , Receptor, ErbB-2/chemistry , Receptor, ErbB-2/metabolism , Signal Transduction/drug effects
7.
Free Radic Biol Med ; 90: 261-71, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26627937

ABSTRACT

We have previously reported that neem limonoids (neem) induce multiple cancer cell death pathways. Here we dissect the underlying mechanisms of neem-induced apoptotic cell death in cancer. We observed that neem-induced caspase activation does not require Bax/Bak channel-mediated mitochondrial outer membrane permeabilization, permeability transition pore, and mitochondrial fragmentation. Neem enhanced mitochondrial DNA and mitochondrial biomass. While oxidative phosphorylation (OXPHOS) Complex-I activity was decreased, the activities of other OXPHOS complexes including Complex-II and -IV were unaltered. Increased reactive oxygen species (ROS) levels were associated with an increase in mitochondrial biomass and apoptosis upon neem exposure. Complex-I deficiency due to the loss of Ndufa1-encoded MWFE protein inhibited neem-induced caspase activation and apoptosis, but cell death induction was enhanced. Complex II-deficiency due to the loss of succinate dehydrogenase complex subunit C (SDHC) robustly decreased caspase activation, apoptosis, and cell death. Additionally, the ablation of Complexes-I, -III, -IV, and -V together did not inhibit caspase activation. Together, we demonstrate that neem limonoids target OXPHOS system to induce cancer cell death, which does not require upregulation or activation of proapoptotic Bcl-2 family proteins.


Subject(s)
Apoptosis/drug effects , Azadirachta/chemistry , Limonins/pharmacology , Neoplasms/pathology , Oxidative Phosphorylation , Caspases/metabolism , Cyclin-Dependent Kinase Inhibitor p21/physiology , DNA, Mitochondrial/analysis , Dynamins , Electron Transport Complex I/physiology , GTP Phosphohydrolases/analysis , HCT116 Cells , Humans , Microtubule-Associated Proteins/analysis , Mitochondrial Membrane Transport Proteins/physiology , Mitochondrial Permeability Transition Pore , Mitochondrial Proteins/analysis , Neoplasms/drug therapy , Tumor Suppressor Protein p53/physiology
8.
PLoS One ; 8(9): e74688, 2013.
Article in English | MEDLINE | ID: mdl-24086362

ABSTRACT

Reduction or depletion of mitochondrial DNA (mtDNA) has been associated with cancer progression. Although imbalanced mtDNA content is known to occur in prostate cancer, differences in mtDNA content between African American (AA) and Caucasian American (CA) men are not defined. We provide the first evidence that tumors in AA men possess reduced level of mtDNA compared to CA men. The median tumor mtDNA content was reduced in AA men. mtDNA content was also reduced in normal prostate tissues of AA men compared to CA men, suggesting a possible predisposition to cancer in AA men. mtDNA content was also reduced in benign prostatic hyperplasia (BPH) tissue from AA men. Tumor and BPH tissues from patients ≥ 60 years of age possess reduced mtDNA content compared to patients <60 years of age. In addition, mtDNA content was higher in normal tissues from patients with malignant T3 stage disease compared to patients with T2 stage disease. mtDNA levels in matched normal prostate tissues were nearly doubled in Gleason grade of >7 compared to ≤ 7, whereas reduced mtDNA content was observed in tumors of Gleason grade >7 compared to ≤ 7. Together, our data suggest that AA men possess lower mtDNA levels in normal and tumor tissues compared to CA men, which could contribute to higher risk and more aggressive prostate cancer in AA men.


Subject(s)
Black or African American/genetics , DNA, Mitochondrial/metabolism , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/genetics , Aged , Demography , Humans , Male , Middle Aged , Neoplasm Grading , Neoplasm Staging , Prognosis , Prostate/metabolism , Prostate/pathology , Prostate-Specific Antigen/metabolism , Prostatic Hyperplasia/pathology , White People/genetics
9.
Sci Signal ; 6(268): pe10, 2013 Mar 26.
Article in English | MEDLINE | ID: mdl-23532331

ABSTRACT

Focal adhesion kinase (FAK) is emerging as a promising cancer target because it is highly expressed at both the transcriptional and translational level in cancer and is involved in many aspects of tumor growth, invasion, and metastasis. Existing FAK-based therapeutics focus on inhibiting the kinase's catalytic function and not the large scaffold it creates that includes many oncogenic receptor tyrosine kinases and tumor suppressor proteins. Targeting the FAK scaffold is a feasible and promising approach for developing highly specific therapeutics that disrupt FAK signaling pathways in cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Delivery Systems/methods , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Models, Biological , Neoplasms/drug therapy , Neoplasms/metabolism , Protein Binding/drug effects , Humans , Protein Structure, Tertiary
10.
Carcinogenesis ; 33(11): 2199-207, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22915764

ABSTRACT

Azadirachta indica, commonly known as neem, has a wide range of medicinal properties. Neem extracts and its purified products have been examined for induction of apoptosis in multiple cancer cell types; however, its underlying mechanisms remain undefined. We show that neem oil (i.e., neem), which contains majority of neem limonoids including azadirachtin, induced apoptotic and autophagic cell death. Gene silencing demonstrated that caspase cascade was initiated by the activation of caspase-9, whereas caspase-8 was also activated late during neem-induced apoptosis. Pretreatment of cancer cells with pan caspase inhibitor, z-VAD inhibited activities of both initiator caspases (e.g., caspase-8 and -9) and executioner caspase-3. Neem induced the release of cytochrome c and apoptosis-inducing factor (AIF) from mitochondria, suggesting the involvement of both caspase-dependent and AIF-mediated apoptosis. p21 deficiency caused an increase in caspase activities at lower doses of neem, whereas p53 deficiency did not modulate neem-induced caspase activation. Additionally, neem treatment resulted in the accumulation of LC3-II in cancer cells, suggesting the involvement of autophagy in neem-induced cancer cell death. Low doses of autophagy inhibitors (i.e., 3-methyladenine and LY294002) did not prevent accumulation of neem-induced LC3-II in cancer cells. Silencing of ATG5 or Beclin-1 further enhanced neem-induced cell death. Phosphoinositide 3-kinase (PI3K) or autophagy inhibitors increased neem-induced caspase-3 activation and inhibition of caspases enhanced neem-induced autophagy. Together, for the first time, we demonstrate that neem induces caspase-dependent and AIF-mediated apoptosis, and autophagy in cancer cells.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Glycerides/chemistry , Limonins/pharmacology , Terpenes/chemistry , Tumor Suppressor Protein p53/metabolism , Adenine/analogs & derivatives , Adenine/pharmacology , Apoptosis Inducing Factor/metabolism , Apoptosis Regulatory Proteins/metabolism , Beclin-1 , Blotting, Western , Caspases/chemistry , Caspases/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cytochromes c/metabolism , Fluorescent Antibody Technique , Humans , Insecticides/pharmacology , Membrane Proteins/metabolism , Mitochondria/drug effects , Mitochondria/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...