499 results on '"Schlaepfer, David D."'
Search Results
2. Tumor FAK orchestrates immunosuppression in ovarian cancer via the CD155/TIGIT axis
- Author
-
Ozmadenci, Duygu, Narayanan, Jayanth S Shankara, Andrew, Jacob, Ojalill, Marjaana, Barrie, Allison M, Jiang, Shulin, Iyer, Samhita, Chen, Xiao Lei, Rose, Michael, Estrada, Valeria, Molinolo, Alfredo, Bertotto, Thomas, Mikulski, Zbigniew, McHale, Michael C, White, Rebekah R, Connolly, Denise C, Pachter, Jonathan A, Kuchroo, Vijay K, Stupack, Dwayne G, and Schlaepfer, David D
- Subjects
Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Immunology ,Rare Diseases ,Cancer ,Ovarian Cancer ,Development of treatments and therapeutic interventions ,5.1 Pharmaceuticals ,Good Health and Well Being ,Animals ,Carcinoma ,Ovarian Epithelial ,Female ,Focal Adhesion Kinase 1 ,Focal Adhesion Protein-Tyrosine Kinases ,Humans ,Immunosuppression Therapy ,Ligands ,Mice ,Ovarian Neoplasms ,Receptors ,Immunologic ,high-grade serous ovarian cancer ,immunotherapy ,FAK ,CD155 ,tertiary lymphoid structures - Abstract
High-grade serous ovarian cancer (HGSOC) is a lethal malignancy characterized by an immunosuppressive tumor microenvironment containing few tumor infiltrating lymphocytes (TILs) and an insensitivity to checkpoint inhibitor immunotherapies. Gains in the PTK2 gene encoding focal adhesion kinase (FAK) at Chr8 q24.3 occur in ∼70% of HGSOC tumors, and elevated FAK messenger RNA (mRNA) levels are associated with poor patient survival. Herein, we show that active FAK, phosphorylated at tyrosine-576 within catalytic domain, is significantly increased in late-stage HGSOC tumors. Active FAK costained with CD155, a checkpoint receptor ligand for TIGIT (T cell immunoreceptor with immunoglobulin and immunoreceptor tyrosine-based inhibitory motif domains), in HGSOC tumors and a selective association between FAK and TIGIT checkpoint ligands were supported by patient transcriptomic database analysis. HGSOC tumors with high FAK expression were associated with low CD3 mRNA levels. Accordingly, late-stage tumors showed elevated active FAK staining and significantly lower levels of CD3+ TILs. Using the KMF (Kras, Myc, FAK) syngeneic ovarian tumor model containing spontaneous PTK2 (FAK) gene gains, the effects of tumor intrinsic genetic or oral small molecule FAK inhibitior (FAKi; VS-4718) were evaluated in vivo. Blocking FAK activity decreased tumor burden, suppressed ascites KMF-associated CD155 levels, and increased peritoneal TILs. The combination of FAKi with blocking TIGIT antibody (1B4) maintained elevated TIL levels and reduced TIGIT+ T regulatory cell levels, prolonged host survival, increased CXCL13 levels, and led to the formation of omental tertiary lymphoid structures. Collectively, our studies support FAK and TIGIT targeting as a rationale immunotherapy combination for HGSOC.
- Published
- 2022
3. Author Correction: Genotype tunes pancreatic ductal adenocarcinoma tissue tension to induce matricellular fibrosis and tumor progression
- Author
-
Laklai, Hanane, Miroshnikova, Yekaterina A., Pickup, Michael W., Collisson, Eric A., Kim, Grace E., Barrett, Alex S., Hill, Ryan C., Lakins, Johnathon N., Schlaepfer, David D., Mouw, Janna K., LeBleu, Valerie S., Roy, Nilotpal, Novitskiy, Sergey V., Johansen, Julia S., Poli, Valeria, Kalluri, Raghu, Iacobuzio-Donahue, Christine A., Wood, Laura D., Hebrok, Matthias, Hansen, Kirk, Moses, Harold L., and Weaver, Valerie M.
- Published
- 2024
- Full Text
- View/download PDF
4. TGFBI remodels adipose metabolism by regulating the Notch-1 signaling pathway
- Author
-
Lee, Seul Gi, Chae, Jongbeom, Woo, Seon Min, Seo, Seung Un, Kim, Ha-Jeong, Kim, Sang-Yeob, Schlaepfer, David D., Kim, In-San, Park, Hee-Sae, Kwon, Taeg Kyu, and Nam, Ju-Ock
- Published
- 2023
- Full Text
- View/download PDF
5. Autophagy gene haploinsufficiency drives chromosome instability, increases migration, and promotes early ovarian tumors.
- Author
-
Delaney, Joe R, Patel, Chandni B, Bapat, Jaidev, Jones, Christian M, Ramos-Zapatero, Maria, Ortell, Katherine K, Tanios, Ralph, Haghighiabyaneh, Mina, Axelrod, Joshua, DeStefano, John W, Tancioni, Isabelle, Schlaepfer, David D, Harismendy, Olivier, La Spada, Albert R, and Stupack, Dwayne G
- Subjects
Cell Line ,Tumor ,Animals ,Mice ,Ovarian Neoplasms ,Chromosomal Instability ,Microtubule-Associated Proteins ,Cell Movement ,Female ,Metabolome ,Haploinsufficiency ,Carcinogenesis ,Beclin-1 ,Ovarian Cancer ,Genetics ,Cancer ,Human Genome ,Breast Cancer ,Rare Diseases ,2.1 Biological and endogenous factors ,Developmental Biology - Abstract
Autophagy, particularly with BECN1, has paradoxically been highlighted as tumor promoting in Ras-driven cancers, but potentially tumor suppressing in breast and ovarian cancers. However, studying the specific role of BECN1 at the genetic level is complicated due to its genomic proximity to BRCA1 on both human (chromosome 17) and murine (chromosome 11) genomes. In human breast and ovarian cancers, the monoallelic deletion of these genes is often co-occurring. To investigate the potential tumor suppressor roles of two of the most commonly deleted autophagy genes in ovarian cancer, BECN1 and MAP1LC3B were knocked-down in atypical (BECN1+/+ and MAP1LC3B+/+) ovarian cancer cells. Ultra-performance liquid chromatography mass-spectrometry metabolomics revealed reduced levels of acetyl-CoA which corresponded with elevated levels of glycerophospholipids and sphingolipids. Migration rates of ovarian cancer cells were increased upon autophagy gene knockdown. Genomic instability was increased, resulting in copy-number alteration patterns which mimicked high grade serous ovarian cancer. We further investigated the causal role of Becn1 haploinsufficiency for oncogenesis in a MISIIR SV40 large T antigen driven spontaneous ovarian cancer mouse model. Tumors were evident earlier among the Becn1+/- mice, and this correlated with an increase in copy-number alterations per chromosome in the Becn1+/- tumors. The results support monoallelic loss of BECN1 as permissive for tumor initiation and potentiating for genomic instability in ovarian cancer.
- Published
- 2020
6. FAK activity sustains intrinsic and acquired ovarian cancer resistance to platinum chemotherapy.
- Author
-
Diaz Osterman, Carlos J, Ozmadenci, Duygu, Kleinschmidt, Elizabeth G, Taylor, Kristin N, Barrie, Allison M, Jiang, Shulin, Bean, Lisa M, Sulzmaier, Florian J, Jean, Christine, Tancioni, Isabelle, Anderson, Kristen, Uryu, Sean, Cordasco, Edward A, Li, Jian, Chen, Xiao Lei, Fu, Guo, Ojalill, Marjaana, Rappu, Pekka, Heino, Jyrki, Mark, Adam M, Xu, Guorong, Fisch, Kathleen M, Kolev, Vihren N, Weaver, David T, Pachter, Jonathan A, Győrffy, Balázs, McHale, Michael T, Connolly, Denise C, Molinolo, Alfredo, Stupack, Dwayne G, and Schlaepfer, David D
- Subjects
Stem Cells ,Animals ,Humans ,Mice ,Ovarian Neoplasms ,Disease Models ,Animal ,Cisplatin ,Platinum ,Proto-Oncogene Proteins c-myc ,Antineoplastic Agents ,Signal Transduction ,Drug Resistance ,Neoplasm ,Female ,Proto-Oncogene Proteins p21(ras) ,Focal Adhesion Kinase 1 ,beta-catenin ,cancer biology ,focal adhesion kinase FAK ,mouse ,ovarian cancer ,platinum chemotherapy ,pluripotency ,tumorspheres ,Rare Diseases ,Genetics ,Stem Cell Research ,Ovarian Cancer ,Cancer ,2.1 Biological and endogenous factors ,Biochemistry and Cell Biology - Abstract
Gene copy number alterations, tumor cell stemness, and the development of platinum chemotherapy resistance contribute to high-grade serous ovarian cancer (HGSOC) recurrence. Stem phenotypes involving Wnt-β-catenin, aldehyde dehydrogenase activities, intrinsic platinum resistance, and tumorsphere formation are here associated with spontaneous gains in Kras, Myc and FAK (KMF) genes in a new aggressive murine model of ovarian cancer. Adhesion-independent FAK signaling sustained KMF and human tumorsphere proliferation as well as resistance to cisplatin cytotoxicity. Platinum-resistant tumorspheres can acquire a dependence on FAK for growth. Accordingly, increased FAK tyrosine phosphorylation was observed within HGSOC patient tumors surviving neo-adjuvant chemotherapy. Combining a FAK inhibitor with platinum overcame chemoresistance and triggered cell apoptosis. FAK transcriptomic analyses across knockout and reconstituted cells identified 135 targets, elevated in HGSOC, that were regulated by FAK activity and β-catenin including Myc, pluripotency and DNA repair genes. These studies reveal an oncogenic FAK signaling role supporting chemoresistance.
- Published
- 2019
7. Nuclear Focal Adhesion Kinase Controls Vascular Smooth Muscle Cell Proliferation and Neointimal Hyperplasia Through GATA4-Mediated Cyclin D1 Transcription
- Author
-
Jeong, Kyuho, Kim, Jung-Hyun, Murphy, James M, Park, Hyeonsoo, Kim, Su-Jeong, Rodriguez, Yelitza AR, Kong, Hyunkyung, Choi, Chungsik, Guan, Jun-Lin, Taylor, Joan M, Lincoln, Thomas M, Gerthoffer, William T, Kim, Jun-Sub, Ahn, Eun-Young Erin, Schlaepfer, David D, and Lim, Ssang-Taek Steve
- Subjects
Genetics ,Cardiovascular ,2.1 Biological and endogenous factors ,Aetiology ,Active Transport ,Cell Nucleus ,Animals ,Cell Nucleus ,Cell Proliferation ,Cells ,Cultured ,Cyclin D1 ,Focal Adhesion Kinase 1 ,GATA4 Transcription Factor ,Hyperplasia ,Mice ,Mice ,Inbred C57BL ,Myocytes ,Smooth Muscle ,Tunica Intima ,atherosclerosis ,cyclin D1 ,FAK ,GATA4 ,hyperplasia ,smooth muscle cell ,vascular remodeling ,Cardiorespiratory Medicine and Haematology ,Clinical Sciences ,Cardiovascular System & Hematology - Abstract
RationaleNeointimal hyperplasia is characterized by excessive accumulation of vascular smooth muscle cells (SMCs) leading to occlusive disorders, such as atherosclerosis and stenosis. Blood vessel injury increases growth factor secretion and matrix synthesis, which promotes SMC proliferation and neointimal hyperplasia via FAK (focal adhesion kinase).ObjectiveTo understand the mechanism of FAK action in SMC proliferation and neointimal hyperplasia.Methods and resultsUsing combined pharmacological FAK catalytic inhibition (VS-4718) and SMC-specific FAK kinase-dead (Myh11-Cre-ERT2) mouse models, we report that FAK regulates SMC proliferation and neointimal hyperplasia in part by governing GATA4- (GATA-binding protein 4) cyclin D1 signaling. Inhibition of FAK catalytic activity facilitates FAK nuclear localization, which is required for proteasome-mediated GATA4 degradation in the cytoplasm. Chromatin immunoprecipitation identified GATA4 binding to the mouse cyclin D1 promoter, and loss of GATA4-mediated cyclin D1 transcription diminished SMC proliferation. Stimulation with platelet-derived growth factor or serum activated FAK and redistributed FAK from the nucleus to cytoplasm, leading to concomitant increase in GATA4 protein and cyclin D1 expression. In a femoral artery wire injury model, increased neointimal hyperplasia was observed in parallel with elevated FAK activity, GATA4 and cyclin D1 expression following injury in control mice, but not in VS-4718-treated and SMC-specific FAK kinase-dead mice. Finally, lentiviral shGATA4 knockdown in the wire injury significantly reduced cyclin D1 expression, SMC proliferation, and neointimal hyperplasia compared with control mice.ConclusionsNuclear enrichment of FAK by inhibition of FAK catalytic activity during vessel injury blocks SMC proliferation and neointimal hyperplasia through regulation of GATA4-mediated cyclin D1 transcription.
- Published
- 2019
8. A Platform of Synthetic Lethal Gene Interaction Networks Reveals that the GNAQ Uveal Melanoma Oncogene Controls the Hippo Pathway through FAK
- Author
-
Feng, Xiaodong, Arang, Nadia, Rigiracciolo, Damiano Cosimo, Lee, Joo Sang, Yeerna, Huwate, Wang, Zhiyong, Lubrano, Simone, Kishore, Ayush, Pachter, Jonathan A, König, Gabriele M, Maggiolini, Marcello, Kostenis, Evi, Schlaepfer, David D, Tamayo, Pablo, Chen, Qianming, Ruppin, Eytan, and Gutkind, J Silvio
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Biotechnology ,Stem Cell Research ,Cancer ,Rare Diseases ,Stem Cell Research - Nonembryonic - Non-Human ,Genetics ,Eye Disease and Disorders of Vision ,2.1 Biological and endogenous factors ,Aetiology ,Animals ,Cell Line ,Tumor ,Cell Proliferation ,Computational Biology ,Focal Adhesion Kinase 1 ,GTP-Binding Protein alpha Subunits ,Gq-G11 ,Genes ,Lethal ,Hippo Signaling Pathway ,Humans ,Melanoma ,Mice ,Neoplasm Transplantation ,Phosphorylation ,Prognosis ,Protein Serine-Threonine Kinases ,Signal Transduction ,Survival Analysis ,Uveal Neoplasms ,FAK ,G protein ,GNA11 ,GNAQ ,Hippo ,MOB1 ,PTK2 ,YAP ,signal transduction ,uveal melanoma ,Protein-Serine-Threonine Kinases ,Neurosciences ,Oncology and Carcinogenesis ,Oncology & Carcinogenesis ,Biochemistry and cell biology ,Oncology and carcinogenesis - Abstract
Activating mutations in GNAQ/GNA11, encoding Gαq G proteins, are initiating oncogenic events in uveal melanoma (UM). However, there are no effective therapies for UM. Using an integrated bioinformatics pipeline, we found that PTK2, encoding focal adhesion kinase (FAK), represents a candidate synthetic lethal gene with GNAQ activation. We show that Gαq activates FAK through TRIO-RhoA non-canonical Gαq-signaling, and genetic ablation or pharmacological inhibition of FAK inhibits UM growth. Analysis of the FAK-regulated transcriptome demonstrated that GNAQ stimulates YAP through FAK. Dissection of the underlying mechanism revealed that FAK regulates YAP by tyrosine phosphorylation of MOB1, inhibiting core Hippo signaling. Our findings establish FAK as a potential therapeutic target for UM and other Gαq-driven pathophysiologies that involve unrestrained YAP function.
- Published
- 2019
9. FAK activity sustains intrinsic and acquired ovarian cancer resistance to platinum chemotherapy
- Author
-
Osterman, Carlos J Diaz, Ozmadenci, Duygu, Kleinschmidt, Elizabeth G, Taylor, Kristin N, Barrie, Allison M, Jiang, Shulin, Bean, Lisa M, Sulzmaier, Florian J, Jean, Christine, Tancioni, Isabelle, Anderson, Kristen, Uryu, Sean, Cordasco, Edward A, Li, Jian, Chen, Xiao Lei, Fu, Guo, Ojalill, Marjaana, Rappu, Pekka, Heino, Jyrki, Mark, Adam M, Xu, Guorong, Fisch, Kathleen M, Kolev, Vihren N, Weaver, David T, Pachter, Jonathan A, Győrffy, Balázs, McHale, Michael T, Connolly, Denise C, Molinolo, Alfredo, Stupack, Dwayne G, and Schlaepfer, David D
- Subjects
Biochemistry and Cell Biology ,Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Biological Sciences ,Ovarian Cancer ,Genetics ,Rare Diseases ,Women's Health ,Cancer ,Clinical Research ,2.1 Biological and endogenous factors ,5.1 Pharmaceuticals ,Animals ,Antineoplastic Agents ,Cisplatin ,Disease Models ,Animal ,Drug Resistance ,Neoplasm ,Female ,Focal Adhesion Kinase 1 ,Humans ,Mice ,Ovarian Neoplasms ,Platinum ,Proto-Oncogene Proteins c-myc ,Proto-Oncogene Proteins p21(ras) ,Signal Transduction ,Stem Cells ,beta-catenin ,cancer biology ,focal adhesion kinase FAK ,mouse ,ovarian cancer ,platinum chemotherapy ,pluripotency ,tumorspheres ,Biological sciences ,Biomedical and clinical sciences ,Health sciences - Abstract
Gene copy number alterations, tumor cell stemness, and the development of platinum chemotherapy resistance contribute to high-grade serous ovarian cancer (HGSOC) recurrence. Stem phenotypes involving Wnt-β-catenin, aldehyde dehydrogenase activities, intrinsic platinum resistance, and tumorsphere formation are here associated with spontaneous gains in Kras, Myc and FAK (KMF) genes in a new aggressive murine model of ovarian cancer. Adhesion-independent FAK signaling sustained KMF and human tumorsphere proliferation as well as resistance to cisplatin cytotoxicity. Platinum-resistant tumorspheres can acquire a dependence on FAK for growth. Accordingly, increased FAK tyrosine phosphorylation was observed within HGSOC patient tumors surviving neo-adjuvant chemotherapy. Combining a FAK inhibitor with platinum overcame chemoresistance and triggered cell apoptosis. FAK transcriptomic analyses across knockout and reconstituted cells identified 135 targets, elevated in HGSOC, that were regulated by FAK activity and β-catenin including Myc, pluripotency and DNA repair genes. These studies reveal an oncogenic FAK signaling role supporting chemoresistance.
- Published
- 2019
10. Adaptive Resistance to Chemotherapy, A Multi–FAK-torial Linkage
- Author
-
Taylor, Kristin N and Schlaepfer, David D
- Subjects
Biochemistry and Cell Biology ,Biomedical and Clinical Sciences ,Biological Sciences ,Immunology ,Oncology and Carcinogenesis ,Colo-Rectal Cancer ,Stem Cell Research - Nonembryonic - Non-Human ,Cancer ,Digestive Diseases ,Genetics ,Stem Cell Research ,5.1 Pharmaceuticals ,Development of treatments and therapeutic interventions ,Aetiology ,2.1 Biological and endogenous factors ,Animals ,Cell Line ,Tumor ,Colorectal Neoplasms ,Focal Adhesion Protein-Tyrosine Kinases ,Melanoma ,Mice ,Mutation ,Proto-Oncogene Proteins B-raf ,Xenograft Model Antitumor Assays ,beta Catenin ,Pharmacology and Pharmaceutical Sciences ,Oncology & Carcinogenesis ,Biochemistry and cell biology ,Oncology and carcinogenesis - Abstract
Oncogenes provide tumor cells with a growth and survival advantage. Directed therapies targeted to oncogenic mutations (such as BRAF V600E) are part of effective late-stage melanoma treatment. However, tumors with BRAF V600E mutations, in approximately 10% of colorectal cancer, are generally treatment-insensitive. Research has identified various "feedback" mechanisms that result in BRAF signal pathway reactivation in response to BRAF inhibition. Herein, we highlight key findings from Chen and colleagues (this issue) showing that integrin-associated focal adhesion kinase (FAK) activation selectively occurs in BRAF V600E-mutant colorectal cancer cells in response to pharmacological BRAF inhibition. FAK activation results in elevated β-catenin protein levels, β-catenin nuclear localization, and increased gene transcription. Small-molecule inhibitors of β-catenin or FAK synergize with vemurafenib BRAF inhibitor to prevent BRAF V600E colorectal cancer cell proliferation in vitro and xenograft tumor growth in mice. This study complements findings linking FAK to β-catenin in intestinal tumorigenesis, resistance to radiotherapy, and cancer stem cell survival. Thus, FAK activation may occur as a frequent tumor cell "adaptive resistance" mechanism. Although FAK (PTK2) is not mutated in most cancers, targeting FAK activity in combinational approaches may limit tumor cell escape mechanisms and enhance durable responses to treatment. Mol Cancer Ther; 17(4); 719-23. ©2018 AACR.
- Published
- 2018
11. Focal adhesion kinase signaling in unexpected places
- Author
-
Kleinschmidt, Elizabeth G and Schlaepfer, David D
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Adherens Junctions ,Animals ,Binding Sites ,Cell Adhesion ,Cell Movement ,Endosomes ,Enzyme Activation ,Extracellular Matrix ,Focal Adhesion Protein-Tyrosine Kinases ,Humans ,Integrins ,Phosphorylation ,Signal Transduction ,Developmental Biology ,Biochemistry and cell biology - Abstract
Focal adhesion kinase (FAK) is a cytoplasmic protein-tyrosine kinase first identified at extracellular matrix and integrin receptor cell adhesion sites and is a key regulator of cell movement. FAK is activated by a variety of stimuli. Herein, we discuss advances in conformational-associated FAK activation and dimerization mechanisms. Additionally, new roles have emerged for FAK signaling at cell adhesions, adherens junctions, endosomes, and the nucleus. In light of these new findings, we review how FAK activation at these sites is connected to the regulation of integrin recycling-activation, vascular permeability, cell survival, and transcriptional regulation, respectively. Studies uncovering FAK signaling connections in unexpected places within cells have yielded important new regulatory insights in cell biology.
- Published
- 2017
12. Haploinsufficiency networks identify targetable patterns of allelic deficiency in low mutation ovarian cancer.
- Author
-
Delaney, Joe Ryan, Patel, Chandni B, Willis, Katelyn McCabe, Haghighiabyaneh, Mina, Axelrod, Joshua, Tancioni, Isabelle, Lu, Dan, Bapat, Jaidev, Young, Shanique, Cadassou, Octavia, Bartakova, Alena, Sheth, Parthiv, Haft, Carley, Hui, Sandra, Saenz, Cheryl, Schlaepfer, David D, Harismendy, Olivier, and Stupack, Dwayne G
- Subjects
Cell Line ,Tumor ,Humans ,Ovarian Neoplasms ,Drug Delivery Systems ,Cell Proliferation ,Mutation ,Alleles ,Autophagy ,Female ,Genes ,Neoplasm ,DNA Copy Number Variations ,Haploinsufficiency ,Proteostasis ,Cell Line ,Tumor ,Genes ,Neoplasm ,Genetics ,Cancer ,Biotechnology ,Rare Diseases ,Ovarian Cancer ,2.1 Biological and endogenous factors - Abstract
Identification of specific oncogenic gene changes has enabled the modern generation of targeted cancer therapeutics. In high-grade serous ovarian cancer (OV), the bulk of genetic changes is not somatic point mutations, but rather somatic copy-number alterations (SCNAs). The impact of SCNAs on tumour biology remains poorly understood. Here we build haploinsufficiency network analyses to identify which SCNA patterns are most disruptive in OV. Of all KEGG pathways (N=187), autophagy is the most significantly disrupted by coincident gene deletions. Compared with 20 other cancer types, OV is most severely disrupted in autophagy and in compensatory proteostasis pathways. Network analysis prioritizes MAP1LC3B (LC3) and BECN1 as most impactful. Knockdown of LC3 and BECN1 expression confers sensitivity to cells undergoing autophagic stress independent of platinum resistance status. The results support the use of pathway network tools to evaluate how the copy-number landscape of a tumour may guide therapy.
- Published
- 2017
13. Targeting FAK in anticancer combination therapies
- Author
-
Dawson, John C., Serrels, Alan, Stupack, Dwayne G., Schlaepfer, David D., and Frame, Margaret C.
- Published
- 2021
- Full Text
- View/download PDF
14. Focal adhesion kinase signaling – tumor vulnerabilities and clinical opportunities.
- Author
-
Schlaepfer, David D., Ojalill, Marjaana, and Stupack, Dwayne G.
- Subjects
- *
FOCAL adhesion kinase , *EXTRACELLULAR matrix proteins , *NUCLEAR matrix , *PROTEIN-tyrosine kinases , *SCAFFOLD proteins , *CELL adhesion , *FOCAL adhesions - Abstract
Focal adhesion kinase (FAK; encoded by PTK2) was discovered over 30 years ago as a cytoplasmic protein tyrosine kinase that is localized to cell adhesion sites, where it is activated by integrin receptor binding to extracellular matrix proteins. FAK is ubiquitously expressed and functions as a signaling scaffold for a variety of proteins at adhesions and in the cell cytoplasm, and with transcription factors in the nucleus. FAK expression and intrinsic activity are essential for mouse development, with molecular connections to cell motility, cell survival and gene expression. Notably, elevated FAK tyrosine phosphorylation is common in tumors, including pancreatic and ovarian cancers, where it is associated with decreased survival. Small molecule and orally available FAK inhibitors show on-target inhibition in tumor and stromal cells with effects on chemotherapy resistance, stromal fibrosis and tumor microenvironment immune function. Herein, we discuss recent insights regarding mechanisms of FAK activation and signaling, its roles as a cytoplasmic and nuclear scaffold, and the tumor-intrinsic and -extrinsic effects of FAK inhibitors. We also discuss results from ongoing and advanced clinical trials targeting FAK in low- and high-grade serous ovarian cancers, where FAK acts as a master regulator of drug resistance. Although FAK is not known to be mutationally activated, preventing FAK activity has revealed multiple tumor vulnerabilities that support expanding clinical combinatorial targeting possibilities. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
15. FAK activity protects nucleostemin in facilitating breast cancer spheroid and tumor growth
- Author
-
Tancioni, Isabelle, Miller, Nichol LG, Uryu, Sean, Lawson, Christine, Jean, Christine, Chen, Xiao Lei, Kleinschmidt, Elizabeth G, and Schlaepfer, David D
- Subjects
Breast Cancer ,Cancer ,Animals ,Breast Neoplasms ,Cell Cycle ,Cell Line ,Tumor ,Cell Proliferation ,Disease Models ,Animal ,Enzyme Activation ,Female ,Focal Adhesion Protein-Tyrosine Kinases ,GTP-Binding Proteins ,Humans ,Mice ,Nuclear Proteins ,Nucleophosmin ,Protein Kinase Inhibitors ,Protein Transport ,Proto-Oncogene Proteins c-akt ,RNA Interference ,RNA ,Messenger ,RNA ,Small Interfering ,Sirolimus ,Spheroids ,Cellular ,Tumor Burden ,Tumor Cells ,Cultured ,Tumor Stem Cell Assay ,Oncology and Carcinogenesis ,Oncology & Carcinogenesis - Abstract
IntroductionFocal adhesion kinase (FAK) controls cell growth and survival downstream of integrin-matrix receptors. Upon adhesion loss or FAK inhibition, FAK can translocate to the nucleus. The nucleolus is a non-membrane nuclear structure that regulates ribosome biogenesis and cell proliferation. Nucleostemin (NS), a nucleolar-localized protein, modulates cell cycle progression, stemness, and three-dimensional tumor spheroid formation. The signaling pathways that regulate NS levels in tumors remain undefined.MethodsHuman breast carcinoma cells were evaluated for growth in culture (adherent and anchorage-independent spheroid) and as orthotopic tumors. FAK signaling was evaluated by pharmacological FAK inhibitor addition (PF-271, IC50~0.1 μM) and by small hairpin RNA (shRNA) knockdown followed by re-expression of FAK wildtype (WT) or a kinase-dead (KD, K454R) FAK point mutant. Immunoblotting was used to evaluate FAK, NS, nucleolar phosphoprotein B23, and nucleolin levels. Total and phosphospecific antibody imunoblotting were used to detect changes in FAK, Akt kinase (Akt also known as protein kinase B), and 4E-binding protein 1 (4E-BP1) phosphorylation, a translation repressor protein and target of the mammalian target of rapamycin (mTOR) complex. Immunohistochemical, co-immunoprecipitation, and cellular fractionation analyses were used to evaluate FAK association with nucleoli.ResultsPharmacological (0.1 μM PF-271) or genetic inhibition of FAK activity prevents MDA-MB-231 and 4T1L breast carcinoma growth as spheroids and as orthotopic tumors. FAK inhibition triggers proteasome-mediated decreased NS levels but no changes in other nucleolar proteins such as B23 (nucleophosmin) or nucleolin. Active FAK was associated with purified nucleoli of anchorage-independent cells and present within nucleoli of human invasive ductal carcinoma tumor samples. FAK co-immunoprecipitated with B23 that binds NS and a complex between FAK, NS, Akt, and mTOR was detected. Constitutively-active Akt kinase promoted tumor spheroid growth, stabilized NS levels, and promoted pS65 4E-BP1 phosphorylation in the presence of inhibited FAK. Rapamycin lowered NS levels and inhibited pS65 4E-BP1 phosphorylation in cells with activated Akt-mTOR signaling.ConclusionsFAK signaling occurs in the nucleolus, active FAK protects NS, and Akt-mTOR pathway regulates NS protein stability needed for breast carcinoma spheroid and tumor growth.
- Published
- 2015
16. Endothelial angiogenic activity and adipose angiogenesis is controlled by extracellular matrix protein TGFBI
- Author
-
Lee, Seul Gi, Kim, Jin Soo, Kim, Ha-Jeong, Schlaepfer, David D., Kim, In-San, and Nam, Ju-Ock
- Published
- 2021
- Full Text
- View/download PDF
17. Force-FAK signaling coupling at individual focal adhesions coordinates mechanosensing and microtissue repair
- Author
-
Zhou, Dennis W., Fernández-Yagüe, Marc A., Holland, Elijah N., García, Andrés F., Castro, Nicolas S., O’Neill, Eric B., Eyckmans, Jeroen, Chen, Christopher S., Fu, Jianping, Schlaepfer, David D., and García, Andrés J.
- Published
- 2021
- Full Text
- View/download PDF
18. FAK Activation Promotes SMC Dedifferentiation via Increased DNA Methylation in Contractile Genes
- Author
-
Jeong, Kyuho, Murphy, James M., Kim, Jung-Hyun, Campbell, Pamela Moore, Park, Hyeonsoo, Rodriguez, Yelitza A.R., Choi, Chung-Sik, Kim, Jun-Sub, Park, Sangwon, Kim, Hyun Joon, Scammell, Jonathan G., Weber, David S., Honkanen, Richard E., Schlaepfer, David D., Ahn, Eun-Young Erin, and Lim, Ssang-Taek Steve
- Published
- 2021
- Full Text
- View/download PDF
19. A strategy to combine pathway-targeted low toxicity drugs in ovarian cancer
- Author
-
Delaney, Joe R, Patel, Chandni, McCabe, Katelyn E, Lu, Dan, Davis, Mitzie-Ann, Tancioni, Isabelle, von Schalscha, Tami, Bartakova, Alena, Haft, Carley, Schlaepfer, David D, and Stupack, Dwayne G
- Subjects
Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Cancer ,Ovarian Cancer ,Orphan Drug ,Rare Diseases ,5.1 Pharmaceuticals ,Development of treatments and therapeutic interventions ,Animals ,Antineoplastic Combined Chemotherapy Protocols ,Autophagy ,Cell Line ,Tumor ,Cell Proliferation ,Dose-Response Relationship ,Drug ,Female ,Humans ,Mice ,Inbred C57BL ,Molecular Targeted Therapy ,Neoplasms ,Cystic ,Mucinous ,and Serous ,Ovarian Neoplasms ,Signal Transduction ,Time Factors ,Tumor Burden ,Xenograft Model Antitumor Assays ,ovarian cancer ,autophagy ,combination therapy ,adverse events ,necramed ,Oncology and carcinogenesis - Abstract
Serous Ovarian Cancers (SOC) are frequently resistant to programmed cell death. However, here we describe that these programmed death-resistant cells are nonetheless sensitive to agents that modulate autophagy. Cytotoxicity is not dependent upon apoptosis, necroptosis, or autophagy resolution. A screen of NCBI yielded more than one dozen FDA-approved agents displaying perturbed autophagy in ovarian cancer. The effects were maximized via combinatorial use of the agents that impinged upon distinct points of autophagy regulation. Autophagosome formation correlated with efficacy in vitro and the most cytotoxic two agents gave similar effects to a pentadrug combination that impinged upon five distinct modulators of autophagy. However, in a complex in vivo SOC system, the pentadrug combination outperformed the best two, leaving trace or no disease and with no evidence of systemic toxicity. Targeting the autophagy pathway in a multi-modal fashion might therefore offer a clinical option for treating recalcitrant SOC.
- Published
- 2015
20. FAK/PYK2 promotes the Wnt/β-catenin pathway and intestinal tumorigenesis by phosphorylating GSK3β.
- Author
-
Gao, Chenxi, Chen, Guangming, Kuan, Shih-Fan, Zhang, Dennis Han, Schlaepfer, David D, and Hu, Jing
- Subjects
Animals ,Mice ,Inbred C57BL ,Humans ,Mice ,Mice ,Nude ,Colorectal Neoplasms ,Glycogen Synthase Kinase 3 ,Gene Expression Regulation ,Female ,Focal Adhesion Kinase 1 ,Focal Adhesion Kinase 2 ,Wnt Signaling Pathway ,Carcinogenesis ,Glycogen Synthase Kinase 3 beta ,FAK ,GSK3 ,PYK2 ,Wnt/b-catenin pathway ,biochemistry ,cell biology ,human ,intestinal tumorigenesis ,mouse ,phosphorylation ,Inbred C57BL ,Nude ,Digestive Diseases ,Cancer ,Colo-Rectal Cancer ,2.1 Biological and endogenous factors ,Biochemistry and Cell Biology - Abstract
Aberrant activation of Wnt/β-catenin signaling plays an unequivocal role in colorectal cancer, but identification of effective Wnt inhibitors for use in cancer remains a tremendous challenge. New insights into the regulation of this pathway could reveal new therapeutic point of intervention, therefore are greatly needed. Here we report a novel FAK/PYK2/GSK3β(Y216)/β-catenin regulation axis: FAK and PYK2, elevated in adenomas in APC(min/+) mice and in human colorectal cancer tissues, functioned redundantly to promote the Wnt/β-catenin pathway by phosphorylating GSK3β(Y216) to reinforce pathway output-β-catenin accumulation and intestinal tumorigenesis. We previously showed that Wnt-induced β-catenin accumulation requires Wnt-induced GSK3β/β-TrCP interaction; the current study revealed that phosphorylation of GSK3β(Y216) was a molecular determinant of GSK3β recruitment of β-TrCP. Pharmacological inhibition of FAK/PYK2 suppressed adenoma formation in APC(min/+) mice accompanied with reduced intestinal levels of phospho-GSK3β(Y216) and β-catenin, indicating that FAK/PYK2/GSK3β(Y216) axis is critical for the activation of Wnt/β-catenin signaling in APC driven intestinal tumorigenesis.
- Published
- 2015
21. Integrin α4 Enhances Metastasis and May Be Associated with Poor Prognosis in MYCNlow Neuroblastoma
- Author
-
Young, Shanique A, McCabe, Katelyn E, Bartakova, Alena, Delaney, Joe, Pizzo, Donald P, Newbury, Robert O, Varner, Judith A, Schlaepfer, David D, and Stupack, Dwayne G
- Subjects
Biochemistry and Cell Biology ,Biomedical and Clinical Sciences ,Biological Sciences ,Pediatric ,Cancer ,Rare Diseases ,Neurosciences ,Pediatric Cancer ,Neuroblastoma ,Development of treatments and therapeutic interventions ,5.1 Pharmaceuticals ,Animals ,Cell Adhesion ,Cell Line ,Tumor ,Cell Movement ,Gene Expression Regulation ,Neoplastic ,Humans ,Integrin alpha4 ,Intercellular Signaling Peptides and Proteins ,Liver Neoplasms ,Lymphatic Metastasis ,Mice ,N-Myc Proto-Oncogene Protein ,Neoplasm Transplantation ,Nervous System Neoplasms ,Nuclear Proteins ,Oncogene Proteins ,Peptides ,Prognosis ,Signal Transduction ,Survival Analysis ,Vascular Cell Adhesion Molecule-1 ,General Science & Technology - Abstract
High-risk neuroblastoma is associated with an overall survival rate of 30-50%. Neuroblastoma-expressed cell adhesion receptors of the integrin family impact cell adhesion, migration, proliferation and survival. Integrin α4 is essential for neural crest cell motility during development, is highly expressed on leukocytes, and is critical for transendothelial migration. Thus, cancer cells that express this receptor may exhibit increased metastatic potential. We show that α4 expression in human and murine neuroblastoma cell lines selectively enhances in vitro interaction with the alternatively spliced connecting segment 1 of fibronectin, as well as vascular cell adhesion molecule-1 and increases migration. Integrin α4 expression enhanced experimental metastasis in a syngeneic tumor model, reconstituting a pattern of organ involvement similar to that seen in patients. Accordingly, antagonism of integrin α4 blocked metastasis, suggesting adhesive function of the integrin is required. However, adhesive function was not sufficient, as mutants of integrin α4 that conserved the matrix-adhesive and promigratory function in vitro were compromised in their metastatic capacity in vivo. Clinically, integrin α4 is more frequently expressed in non-MYNC amplified tumors, and is selectively associated with poor prognosis in this subset of disease. These results reveal an unexpected role for integrin α4 in neuroblastoma dissemination and identify α4 as a potential prognostic indicator and therapeutic target.
- Published
- 2015
22. Integrin α4 Enhances Metastasis and May Be Associated with Poor Prognosis in MYCN-low Neuroblastoma.
- Author
-
Young, Shanique A, McCabe, Katelyn E, Bartakova, Alena, Delaney, Joe, Pizzo, Donald P, Newbury, Robert O, Varner, Judith A, Schlaepfer, David D, and Stupack, Dwayne G
- Subjects
Cell Line ,Tumor ,Animals ,Humans ,Mice ,Neuroblastoma ,Liver Neoplasms ,Nervous System Neoplasms ,Lymphatic Metastasis ,Intercellular Signaling Peptides and Proteins ,Peptides ,Vascular Cell Adhesion Molecule-1 ,Integrin alpha4 ,Oncogene Proteins ,Nuclear Proteins ,Prognosis ,Survival Analysis ,Neoplasm Transplantation ,Cell Adhesion ,Signal Transduction ,Cell Movement ,Gene Expression Regulation ,Neoplastic ,N-Myc Proto-Oncogene Protein ,Cell Line ,Tumor ,Gene Expression Regulation ,Neoplastic ,Rare Diseases ,Cancer ,Neurosciences ,Pediatric Cancer ,Pediatric ,5.1 Pharmaceuticals ,General Science & Technology - Abstract
High-risk neuroblastoma is associated with an overall survival rate of 30-50%. Neuroblastoma-expressed cell adhesion receptors of the integrin family impact cell adhesion, migration, proliferation and survival. Integrin α4 is essential for neural crest cell motility during development, is highly expressed on leukocytes, and is critical for transendothelial migration. Thus, cancer cells that express this receptor may exhibit increased metastatic potential. We show that α4 expression in human and murine neuroblastoma cell lines selectively enhances in vitro interaction with the alternatively spliced connecting segment 1 of fibronectin, as well as vascular cell adhesion molecule-1 and increases migration. Integrin α4 expression enhanced experimental metastasis in a syngeneic tumor model, reconstituting a pattern of organ involvement similar to that seen in patients. Accordingly, antagonism of integrin α4 blocked metastasis, suggesting adhesive function of the integrin is required. However, adhesive function was not sufficient, as mutants of integrin α4 that conserved the matrix-adhesive and promigratory function in vitro were compromised in their metastatic capacity in vivo. Clinically, integrin α4 is more frequently expressed in non-MYNC amplified tumors, and is selectively associated with poor prognosis in this subset of disease. These results reveal an unexpected role for integrin α4 in neuroblastoma dissemination and identify α4 as a potential prognostic indicator and therapeutic target.
- Published
- 2015
23. FAK in cancer: mechanistic findings and clinical applications
- Author
-
Sulzmaier, Florian J, Jean, Christine, and Schlaepfer, David D
- Subjects
Biomedical and Clinical Sciences ,Cancer ,Stem Cell Research ,2.1 Biological and endogenous factors ,Aetiology ,Animals ,Focal Adhesion Kinase 1 ,Humans ,Molecular Targeted Therapy ,Neoplasm Invasiveness ,Neoplasms ,Protein Kinase Inhibitors ,Signal Transduction ,Small Molecule Libraries ,Stromal Cells ,Tumor Microenvironment ,Medical and Health Sciences ,Oncology & Carcinogenesis ,Biomedical and clinical sciences ,Health sciences - Abstract
Focal adhesion kinase (FAK) is a cytoplasmic protein tyrosine kinase that is overexpressed and activated in several advanced-stage solid cancers. FAK promotes tumour progression and metastasis through effects on cancer cells, as well as stromal cells of the tumour microenvironment. The kinase-dependent and kinase-independent functions of FAK control cell movement, invasion, survival, gene expression and cancer stem cell self-renewal. Small molecule FAK inhibitors decrease tumour growth and metastasis in several preclinical models and have initial clinical activity in patients with limited adverse events. In this Review, we discuss FAK signalling effects on both tumour and stromal cell biology that provide rationale and support for future therapeutic opportunities.
- Published
- 2014
24. FAK Inhibition Disrupts a β5 Integrin Signaling Axis Controlling Anchorage-Independent Ovarian Carcinoma Growth
- Author
-
Tancioni, Isabelle, Uryu, Sean, Sulzmaier, Florian J, Shah, Nina R, Lawson, Christine, Miller, Nichol LG, Jean, Christine, Chen, Xiao Lei, Ward, Kristy K, and Schlaepfer, David D
- Subjects
Biochemistry and Cell Biology ,Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Biological Sciences ,Rare Diseases ,Ovarian Cancer ,Cancer ,Aminopyridines ,Animals ,Antineoplastic Agents ,Cell Adhesion ,Cell Line ,Tumor ,Cell Proliferation ,Drug Resistance ,Neoplasm ,Female ,Focal Adhesion Kinase 1 ,Gene Knockdown Techniques ,Humans ,Integrin beta Chains ,Kaplan-Meier Estimate ,Mice ,Nude ,Neoplasm Transplantation ,Neoplasms ,Cystic ,Mucinous ,and Serous ,Osteopontin ,Ovarian Neoplasms ,Signal Transduction ,Pharmacology and Pharmaceutical Sciences ,Oncology & Carcinogenesis ,Biochemistry and cell biology ,Oncology and carcinogenesis - Abstract
Ovarian cancer ascites fluid contains matrix proteins that can impact tumor growth via integrin receptor binding. In human ovarian tumor tissue arrays, we find that activation of the cytoplasmic focal adhesion (FAK) tyrosine kinase parallels increased tumor stage, β5 integrin, and osteopontin matrix staining. Elevated osteopontin, β5 integrin, and FAK mRNA levels are associated with decreased serous ovarian cancer patient survival. FAK remains active within ovarian cancer cells grown as spheroids, and anchorage-independent growth analyses of seven ovarian carcinoma cell lines identified sensitive (HEY, OVCAR8) and resistant (SKOV3-IP, OVCAR10) cells to 0.1 μmol/L FAK inhibitor (VS-4718, formerly PND-1186) treatment. VS-4718 promoted HEY and OVCAR8 G0-G1 cell-cycle arrest followed by cell death, whereas growth of SKOV3-IP and OVCAR10 cells was resistant to 1.0 μmol/L VS-4718. In HEY cells, genetic or pharmacological FAK inhibition prevented tumor growth in mice with corresponding reductions in β5 integrin and osteopontin expression. β5 knockdown reduced HEY cell growth in soft agar, tumor growth in mice, and both FAK Y397 phosphorylation and osteopontin expression in spheroids. FAK inhibitor-resistant (SKOV3-IP, OVCAR10) cells exhibited anchorage-independent Akt S473 phosphorylation, and expression of membrane-targeted and active Akt in sensitive cells (HEY, OVCAR8) increased growth but did not create a FAK inhibitor-resistant phenotype. These results link osteopontin, β5 integrin, and FAK in promoting ovarian tumor progression. β5 integrin expression may serve as a biomarker for serous ovarian carcinoma cells that possess active FAK signaling.
- Published
- 2014
25. Analyses of merlin/NF2 connection to FAK inhibitor responsiveness in serous ovarian cancer
- Author
-
Shah, Nina R, Tancioni, Isabelle, Ward, Kristy K, Lawson, Christine, Chen, Xiao Lei, Jean, Christine, Sulzmaier, Florian J, Uryu, Sean, Miller, Nichol LG, Connolly, Denise C, and Schlaepfer, David D
- Subjects
Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Neurosciences ,Rare Diseases ,Neurofibromatosis ,Ovarian Cancer ,Cancer ,Animals ,Biomarkers ,Tumor ,Cell Line ,Tumor ,Cystadenocarcinoma ,Serous ,Female ,Focal Adhesion Kinase 1 ,Gene Knockdown Techniques ,Humans ,Mice ,Neurofibromin 2 ,Ovarian Neoplasms ,Protein Kinase Inhibitors ,Ovarian cancer ,Focal adhesion kinase ,Merlin ,Tumor ,Biomarker ,Paediatrics and Reproductive Medicine ,Oncology & Carcinogenesis ,Clinical sciences ,Oncology and carcinogenesis ,Reproductive medicine - Abstract
ObjectiveFocal adhesion kinase (FAK) is overexpressed in serous ovarian cancer. Loss of merlin, a product of the neurofibromatosis 2 tumor suppressor gene, is being evaluated as a biomarker for FAK inhibitor sensitivity in mesothelioma. Connections between merlin and FAK in ovarian cancer remain undefined.MethodsNine human and two murine ovarian cancer cell lines were analyzed for growth in the presence of a small molecule FAK inhibitor (PF-271, also termed VS-6062) from 0.1 to 1 μM for 72 h. Merlin was evaluated by immunoblotting and immunostaining of a human ovarian tumor tissue array. Growth of cells was analyzed in an orthotopic tumor model and evaluated in vitro after stable shRNA-mediated merlin knockdown.ResultsGreater than 50% inhibition of OVCAR8, HEY, and ID8-IP ovarian carcinoma cell growth occurred with 0.1 μM PF-271 in anchorage-independent (p
- Published
- 2014
26. Grb2 Promotes Integrin-Induced Focal Adhesion Kinase (FAK) Autophosphorylation and Directs the Phosphorylation of Protein Tyrosine Phosphatase α by the Src-FAK Kinase Complex
- Author
-
Cheng, Suzanne YS, Sun, Guobin, Schlaepfer, David D, and Pallen, Catherine J
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Aetiology ,2.1 Biological and endogenous factors ,Animals ,Cells ,Cultured ,Embryo ,Mammalian ,Fibroblasts ,Focal Adhesion Kinase 1 ,GRB2 Adaptor Protein ,Immunoblotting ,Integrins ,Mice ,Mice ,Knockout ,Paxillin ,Phosphorylation ,Protein Binding ,RNA Interference ,Receptor-Like Protein Tyrosine Phosphatases ,Class 4 ,Signal Transduction ,Tyrosine ,src Homology Domains ,Medical and Health Sciences ,Developmental Biology ,Biological sciences ,Biomedical and clinical sciences ,Health sciences - Abstract
The integrin-activated Src-focal adhesion kinase (FAK) kinase complex phosphorylates PTPα at Tyr789, initiating PTPα-mediated signaling that promotes cell migration. Recruitment of the BCAR3-Cas complex by PTPα-phospho-Tyr789 at focal adhesions is one mechanism of PTPα signaling. The adaptor protein Grb2 is also recruited by PTPα-phospho-Tyr789, although the role of the PTPα-Grb2 complex in integrin signaling is unknown. We show that silencing Grb2 expression in fibroblasts abolishes PTPα-Tyr789 phosphorylation and that this is due to two unexpected actions of Grb2. First, Grb2 promotes integrin-induced autophosphorylation of FAK-Tyr397. This is impaired in Grb2-depleted cells and prohibits FAK activation and formation of the Src-FAK complex. Grb2-depleted cells contain less paxillin, and paxillin overexpression rescues FAK-Tyr397 phosphorylation, suggesting that the FAK-activating action of Grb2 involves paxillin. A second distinct role for Grb2 in PTPα-Tyr789 phosphorylation involves Grb2-mediated coupling of Src-FAK and PTPα. This requires two phosphosites, FAK-Tyr925 and PTPα-Tyr789, for Grb2-Src homology 2 (SH2) binding. We propose that a Grb2 dimer links FAK and PTPα, and this positions active Src-FAK in proximity with other, perhaps integrin-clustered, molecules of PTPα to enable maximal PTPα-Tyr789 phosphorylation. These findings identify Grb2 as a new FAK activator and reveal its essential role in coordinating PTPα tyrosine phosphorylation to enable downstream integrin signaling and migration.
- Published
- 2014
27. Inhibition of endothelial FAK activity prevents tumor metastasis by enhancing barrier function
- Author
-
Jean, Christine, Chen, Xiao Lei, Nam, Ju-Ock, Tancioni, Isabelle, Uryu, Sean, Lawson, Christine, Ward, Kristy K, Walsh, Colin T, Miller, Nichol LG, Ghassemian, Majid, Turowski, Patric, Dejana, Elisabetta, Weis, Sara, Cheresh, David A, and Schlaepfer, David D
- Subjects
Biochemistry and Cell Biology ,Medical Physiology ,Biomedical and Clinical Sciences ,Biological Sciences ,Cancer ,Rare Diseases ,2.1 Biological and endogenous factors ,Aetiology ,Cardiovascular ,Animals ,Antigens ,CD ,Cadherins ,Cell Movement ,Focal Adhesion Protein-Tyrosine Kinases ,HEK293 Cells ,Humans ,Mice ,Mice ,Inbred BALB C ,Neoplasm Metastasis ,Phosphorylation ,Proto-Oncogene Proteins pp60(c-src) ,Signal Transduction ,Vascular Endothelial Growth Factor A ,Vascular Endothelial Growth Factor Receptor-2 ,Medical and Health Sciences ,Developmental Biology ,Biological sciences ,Biomedical and clinical sciences - Abstract
Pharmacological focal adhesion kinase (FAK) inhibition prevents tumor growth and metastasis, via actions on both tumor and stromal cells. In this paper, we show that vascular endothelial cadherin (VEC) tyrosine (Y) 658 is a target of FAK in tumor-associated endothelial cells (ECs). Conditional kinase-dead FAK knockin within ECs inhibited recombinant vascular endothelial growth factor (VEGF-A) and tumor-induced VEC-Y658 phosphorylation in vivo. Adherence of VEGF-expressing tumor cells to ECs triggered FAK-dependent VEC-Y658 phosphorylation. Both FAK inhibition and VEC-Y658F mutation within ECs prevented VEGF-initiated paracellular permeability and tumor cell transmigration across EC barriers. In mice, EC FAK inhibition prevented VEGF-dependent tumor cell extravasation and melanoma dermal to lung metastasis without affecting primary tumor growth. As pharmacological c-Src or FAK inhibition prevents VEGF-stimulated c-Src and FAK translocation to EC adherens junctions, but FAK inhibition does not alter c-Src activation, our experiments identify EC FAK as a key intermediate between c-Src and the regulation of EC barrier function controlling tumor metastasis.
- Published
- 2014
28. Author Correction: Genotype tunes pancreatic ductal adenocarcinoma tissue tension to induce matricellular fibrosis and tumor progression
- Author
-
Laklai, Hanane, primary, Miroshnikova, Yekaterina A., additional, Pickup, Michael W., additional, Collisson, Eric A., additional, Kim, Grace E., additional, Barrett, Alex S., additional, Hill, Ryan C., additional, Lakins, Johnathon N., additional, Schlaepfer, David D., additional, Mouw, Janna K., additional, LeBleu, Valerie S., additional, Roy, Nilotpal, additional, Novitskiy, Sergey V., additional, Johansen, Julia S., additional, Poli, Valeria, additional, Kalluri, Raghu, additional, Iacobuzio-Donahue, Christine A., additional, Wood, Laura D., additional, Hebrok, Matthias, additional, Hansen, Kirk, additional, Moses, Harold L., additional, and Weaver, Valerie M., additional
- Published
- 2023
- Full Text
- View/download PDF
29. A non-canonical role for Rgnef in promoting integrin-stimulated focal adhesion kinase activation
- Author
-
Miller, Nichol LG, Lawson, Christine, Kleinschmidt, Elizabeth G, Tancioni, Isabelle, Uryu, Sean, and Schlaepfer, David D
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Amino Acid Sequence ,Animals ,Cell Adhesion ,Cells ,Cultured ,Enzyme Activation ,Fibroblasts ,Fibronectins ,Focal Adhesion Protein-Tyrosine Kinases ,Integrin beta1 ,Mice ,Mice ,Knockout ,Molecular Sequence Data ,Paxillin ,Phosphorylation ,Protein Binding ,Protein Structure ,Tertiary ,Sequence Alignment ,Signal Transduction ,ras-GRF1 ,Rgnef ,p190RhoGEF ,Arhgef28 ,FAK ,Adhesion ,PH domain ,GEF ,Medical and Health Sciences ,Developmental Biology ,Biochemistry and cell biology - Abstract
Rgnef (also known as p190RhoGEF or ARHGEF28) is a Rho guanine-nucleotide-exchange factor (GEF) that binds focal adhesion kinase (FAK). FAK is recruited to adhesions and activated by integrin receptors binding to matrix proteins, such as fibronectin (FN). Canonical models place Rgnef downstream of integrin-FAK signaling in regulating Rho GTPase activity and cell movement. Herein, we establish a new, upstream role for Rgnef in enhancing FAK localization to early peripheral adhesions and promoting FAK activation upon FN binding. Rgnef-null mouse embryo fibroblasts (MEFs) exhibit defects in adhesion formation, levels of FAK phosphotyrosine (pY)-397 and FAK localization to peripheral adhesions upon re-plating on FN. Rgnef re-expression rescues these defects, but requires Rgnef-FAK binding. A mutation in the Rgnef pleckstrin homology (PH) domain inhibits adhesion formation, FAK localization, and FAK-Y397 and paxillin-Y118 phosphorylation without disrupting the Rgnef-FAK interaction. A GEF-inactive Rgnef mutant rescues FAK-Y397 phosphorylation and early adhesion localization, but not paxillin-Y118 phosphorylation. This suggests that, downstream of FN binding, paxillin-pY118 requires Rgnef GEF activity through a mechanism distinct from adhesion formation and FAK activation. These results support a scaffolding role for Rgnef in FAK localization and activation at early adhesions in a PH-domain-dependent but GEF-activity-independent manner.
- Published
- 2013
30. pHocal adhesion kinase regulation is on a FERM foundation
- Author
-
Lawson, Christine and Schlaepfer, David D
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Animals ,Cation Transport Proteins ,Embryo ,Mammalian ,Female ,Fibroblasts ,Focal Adhesion Kinase 1 ,Focal Adhesions ,Histidine ,Humans ,Sodium-Hydrogen Exchanger 1 ,Sodium-Hydrogen Exchangers ,Medical and Health Sciences ,Developmental Biology ,Biological sciences ,Biomedical and clinical sciences - Abstract
Increases in intracellular pH (pHi) occur upon integrin receptor binding to matrix proteins and in tumor cells. In this issue, Choi et al. (2013. J. Cell Biol. http://dx.doi.org/10.1083/jcb.201308034) show that pHi increase activates FAK by causing deprotonation of histidine 58 in its FERM (band 4.1, ezrin, radixin, moesin) homology domain, which exposes a region important for FAK autophosphorylation. This model of FAK activation could contribute to motility of tumor cells by promoting focal adhesion turnover.
- Published
- 2013
31. Rgnef promotes ovarian tumor progression and confers protection from oxidative stress
- Author
-
Kleinschmidt, Elizabeth G., Miller, Nichol L. G., Ozmadenci, Duygu, Tancioni, Isabelle, Osterman, Carlos Díaz, Barrie, Allison M., Taylor, Kristin N., Ye, Aaron, Jiang, Shulin, Connolly, Denise C., Stupack, Dwayne G., and Schlaepfer, David D.
- Published
- 2019
- Full Text
- View/download PDF
32. Nuclear-localized focal adhesion kinase regulates inflammatory VCAM-1 expression
- Author
-
Lim, Ssang-Taek, Miller, Nichol LG, Chen, Xiao Lei, Tancioni, Isabelle, Walsh, Colin T, Lawson, Christine, Uryu, Sean, Weis, Sara M, Cheresh, David A, and Schlaepfer, David D
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Genetics ,Cardiovascular ,Aetiology ,2.1 Biological and endogenous factors ,Active Transport ,Cell Nucleus ,Animals ,Cell Nucleus ,Cells ,Cultured ,Embryo ,Mammalian ,Enzyme Activation ,Focal Adhesion Kinase 1 ,GATA4 Transcription Factor ,Gene Expression Regulation ,Developmental ,Humans ,Mice ,Mice ,Transgenic ,Tumor Necrosis Factor-alpha ,Ubiquitination ,Vascular Cell Adhesion Molecule-1 ,Medical and Health Sciences ,Developmental Biology ,Biological sciences ,Biomedical and clinical sciences - Abstract
Vascular cell adhesion molecule-1 (VCAM-1) plays important roles in development and inflammation. Tumor necrosis factor-α (TNF-α) and focal adhesion kinase (FAK) are key regulators of inflammatory and integrin-matrix signaling, respectively. Integrin costimulatory signals modulate inflammatory gene expression, but the important control points between these pathways remain unresolved. We report that pharmacological FAK inhibition prevented TNF-α-induced VCAM-1 expression within heart vessel-associated endothelial cells in vivo, and genetic or pharmacological FAK inhibition blocked VCAM-1 expression during development. FAK signaling facilitated TNF-α-induced, mitogen-activated protein kinase activation, and, surprisingly, FAK inhibition resulted in the loss of the GATA4 transcription factor required for TNF-α-induced VCAM-1 production. FAK inhibition also triggered FAK nuclear localization. In the nucleus, the FAK-FERM (band 4.1, ezrin, radixin, moesin homology) domain bound directly to GATA4 and enhanced its CHIP (C terminus of Hsp70-interacting protein) E3 ligase-dependent polyubiquitination and degradation. These studies reveal new developmental and anti-inflammatory roles for kinase-inhibited FAK in limiting VCAM-1 production via nuclear localization and promotion of GATA4 turnover.
- Published
- 2012
33. FAK Signaling Is Critical for ErbB-2/ErbB-3 Receptor Cooperation for Oncogenic Transformation and Invasion
- Author
-
Benlimame, Naciba, He, Qiang, Jie, Su, Xiao, Dingzhang, Xu, Ying Jie, Loignon, Martin, Schlaepfer, David D., and Alaoui-Jamali, Moulay A.
- Published
- 2005
34. Localized Stabilization of Microtubules by Integrin- and FAK-Facilitated Rho Signaling
- Author
-
Palazzo, Alexander F., Eng, Christina H., Schlaepfer, David D., Marcantonio, Eugene E., and Gundersen, Gregg G.
- Published
- 2004
35. Differential Regulation of Cell Motility and Invasion by FAK
- Author
-
Hsia, Datsun A., Mitra, Satyajit K., Hauck, Christof R., Streblow, Daniel N., Nelson, Jay A., Ilic, Dusko, Huang, Shuang, Li, Erguang, Nemerow, Glen R., Leng, Jay, Cheresh, David A., and Schlaepfer, David D.
- Published
- 2003
36. Compensatory role for Pyk2 during angiogenesis in adult mice lacking endothelial cell FAK
- Author
-
Weis, Sara M, Lim, Ssang-Taek, Lutu-Fuga, Kimberly M, Barnes, Leo A, Chen, Xiao Lei, Göthert, Joachim R, Shen, Tang-Long, Guan, Jun-Lin, Schlaepfer, David D, and Cheresh, David A
- Subjects
Biochemistry and Cell Biology ,Medical Physiology ,Biomedical and Clinical Sciences ,Biological Sciences ,2.1 Biological and endogenous factors ,Aetiology ,Cardiovascular ,Animals ,Aorta ,Endothelial Cells ,Focal Adhesion Kinase 1 ,Focal Adhesion Kinase 2 ,Humans ,Integrins ,Mice ,Mice ,Inbred C57BL ,Mice ,Knockout ,Neovascularization ,Physiologic ,Signal Transduction ,Medical and Health Sciences ,Developmental Biology ,Biological sciences ,Biomedical and clinical sciences - Abstract
Focal adhesion kinase (FAK) plays a critical role during vascular development because knockout of FAK in endothelial cells (ECs) is embryonic lethal. Surprisingly, tamoxifen-inducible conditional knockout of FAK in adult blood vessels (inducible EC-specific FAK knockout [i-EC-FAK-KO]) produces no vascular phenotype, and these animals are capable of developing a robust growth factor-induced angiogenic response. Although angiogenesis in wild-type mice is suppressed by pharmacological inhibition of FAK, i-EC-FAK-KO mice are refractory to this treatment, which suggests that adult i-EC-FAK-KO mice develop a compensatory mechanism to bypass the requirement for FAK. Indeed, expression of the FAK-related proline-rich tyrosine kinase 2 (Pyk2) is elevated and phosphorylated in i-EC-FAK-KO blood vessels. In cultured ECs, FAK knockdown leads to increased Pyk2 expression and, surprisingly, FAK kinase inhibition leads to increased Pyk2 phosphorylation. Pyk2 can functionally compensate for the loss of FAK because knockdown or pharmacological inhibition of Pyk2 disrupts angiogenesis in i-EC-FAK-KO mice. These studies reveal the adaptive capacity of ECs to switch to Pyk2-dependent signaling after deletion or kinase inhibition of FAK.
- Published
- 2008
37. Src-Mediated Coupling of Focal Adhesion Kinase to Integrin αvβ5 in Vascular Endothelial Growth Factor Signaling
- Author
-
Eliceiri, Brian P., Puente, Xose X., Hood, John D., Stupack, Dwayne G., Schlaepfer, David D., Huang, Xiaozhu Z., Sheppard, Dean, and Cheresh, David A.
- Published
- 2002
38. Targeting Pyk2 to β1-Integrin-Containing Focal Contacts Rescues Fibronectin-Stimulated Signaling and Haptotactic Motility Defects of Focal Adhesion Kinase-Null Cells
- Author
-
Klingbeil, Candice K., Hauck, Christof R., Hsia, Datsun A., Jones, K. C., Reider, Shannon R., and Schlaepfer, David D.
- Published
- 2001
39. Matrix Survival Signaling: From Fibronectin via Focal Adhesion Kinase to c-Jun NH 2 -Terminal Kinase
- Author
-
Ilić, Dus̆ko, Han, Qin, Hauck, Christof R., Jin, Fang, Kawakatsu, Hisaaki, Schlaepfer, David D., and Damsky, Caroline H.
- Published
- 2000
40. Extracellular Matrix Survival Signals Transduced by Focal Adhesion Kinase Suppress p53-Mediated Apoptosis
- Author
-
Ilić, Duško, Schlaepfer, David D., Dazin, Paul, Aizawa, Shinichi, and Damsky, Caroline H.
- Published
- 1998
41. Abstract 2363: Ovarian tumor-intrinsic FAK-dependent regulation of the matrisome
- Author
-
Ojalill, Marjaana, primary, Zhang, Joanna, additional, Suwal, Ujjwal, additional, Rappu, Pekka, additional, Heino, Jyrki, additional, Connolly, Denise C., additional, Stupack, Dwayne G., additional, and Schlaepfer, David D., additional
- Published
- 2023
- Full Text
- View/download PDF
42. Supplementary Figure 1 from FAK Inhibition Disrupts a β5 Integrin Signaling Axis Controlling Anchorage-Independent Ovarian Carcinoma Growth
- Author
-
Tancioni, Isabelle, primary, Uryu, Sean, primary, Sulzmaier, Florian J., primary, Shah, Nina R., primary, Lawson, Christine, primary, Miller, Nichol L.G., primary, Jean, Christine, primary, Chen, Xiao Lei, primary, Ward, Kristy K., primary, and Schlaepfer, David D., primary
- Published
- 2023
- Full Text
- View/download PDF
43. Supplementary Figure 5 from FAK Inhibition Disrupts a β5 Integrin Signaling Axis Controlling Anchorage-Independent Ovarian Carcinoma Growth
- Author
-
Tancioni, Isabelle, primary, Uryu, Sean, primary, Sulzmaier, Florian J., primary, Shah, Nina R., primary, Lawson, Christine, primary, Miller, Nichol L.G., primary, Jean, Christine, primary, Chen, Xiao Lei, primary, Ward, Kristy K., primary, and Schlaepfer, David D., primary
- Published
- 2023
- Full Text
- View/download PDF
44. Supplementary Table 1 from FAK Inhibition Disrupts a β5 Integrin Signaling Axis Controlling Anchorage-Independent Ovarian Carcinoma Growth
- Author
-
Tancioni, Isabelle, primary, Uryu, Sean, primary, Sulzmaier, Florian J., primary, Shah, Nina R., primary, Lawson, Christine, primary, Miller, Nichol L.G., primary, Jean, Christine, primary, Chen, Xiao Lei, primary, Ward, Kristy K., primary, and Schlaepfer, David D., primary
- Published
- 2023
- Full Text
- View/download PDF
45. Supplementary Figure 2 from FAK Inhibition Disrupts a β5 Integrin Signaling Axis Controlling Anchorage-Independent Ovarian Carcinoma Growth
- Author
-
Tancioni, Isabelle, primary, Uryu, Sean, primary, Sulzmaier, Florian J., primary, Shah, Nina R., primary, Lawson, Christine, primary, Miller, Nichol L.G., primary, Jean, Christine, primary, Chen, Xiao Lei, primary, Ward, Kristy K., primary, and Schlaepfer, David D., primary
- Published
- 2023
- Full Text
- View/download PDF
46. Data from FAK Inhibition Disrupts a β5 Integrin Signaling Axis Controlling Anchorage-Independent Ovarian Carcinoma Growth
- Author
-
Tancioni, Isabelle, primary, Uryu, Sean, primary, Sulzmaier, Florian J., primary, Shah, Nina R., primary, Lawson, Christine, primary, Miller, Nichol L.G., primary, Jean, Christine, primary, Chen, Xiao Lei, primary, Ward, Kristy K., primary, and Schlaepfer, David D., primary
- Published
- 2023
- Full Text
- View/download PDF
47. Supplementary Figure 4 from FAK Inhibition Disrupts a β5 Integrin Signaling Axis Controlling Anchorage-Independent Ovarian Carcinoma Growth
- Author
-
Tancioni, Isabelle, primary, Uryu, Sean, primary, Sulzmaier, Florian J., primary, Shah, Nina R., primary, Lawson, Christine, primary, Miller, Nichol L.G., primary, Jean, Christine, primary, Chen, Xiao Lei, primary, Ward, Kristy K., primary, and Schlaepfer, David D., primary
- Published
- 2023
- Full Text
- View/download PDF
48. Supplementary Table 2 from FAK Inhibition Disrupts a β5 Integrin Signaling Axis Controlling Anchorage-Independent Ovarian Carcinoma Growth
- Author
-
Tancioni, Isabelle, primary, Uryu, Sean, primary, Sulzmaier, Florian J., primary, Shah, Nina R., primary, Lawson, Christine, primary, Miller, Nichol L.G., primary, Jean, Christine, primary, Chen, Xiao Lei, primary, Ward, Kristy K., primary, and Schlaepfer, David D., primary
- Published
- 2023
- Full Text
- View/download PDF
49. Supplementary Figure 3 from FAK Inhibition Disrupts a β5 Integrin Signaling Axis Controlling Anchorage-Independent Ovarian Carcinoma Growth
- Author
-
Tancioni, Isabelle, primary, Uryu, Sean, primary, Sulzmaier, Florian J., primary, Shah, Nina R., primary, Lawson, Christine, primary, Miller, Nichol L.G., primary, Jean, Christine, primary, Chen, Xiao Lei, primary, Ward, Kristy K., primary, and Schlaepfer, David D., primary
- Published
- 2023
- Full Text
- View/download PDF
50. Supplementary Methods from FAK Inhibition Disrupts a β5 Integrin Signaling Axis Controlling Anchorage-Independent Ovarian Carcinoma Growth
- Author
-
Tancioni, Isabelle, primary, Uryu, Sean, primary, Sulzmaier, Florian J., primary, Shah, Nina R., primary, Lawson, Christine, primary, Miller, Nichol L.G., primary, Jean, Christine, primary, Chen, Xiao Lei, primary, Ward, Kristy K., primary, and Schlaepfer, David D., primary
- Published
- 2023
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.