Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 79
Filter
1.
Proc Natl Acad Sci U S A ; 119(17): e2117065119, 2022 04 26.
Article in English | MEDLINE | ID: mdl-35467979

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.


Subject(s)
Ovarian Neoplasms , Animals , Carcinoma, Ovarian Epithelial , Female , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , Humans , Immunosuppression Therapy , Ligands , Mice , Ovarian Neoplasms/pathology , Receptors, Immunologic/metabolism
2.
Circ Res ; 129(12): e215-e233, 2021 12 03.
Article in English | MEDLINE | ID: mdl-34702049

ABSTRACT

RATIONALE: Vascular smooth muscle cells (SMCs) exhibit remarkable plasticity and can undergo dedifferentiation upon pathological stimuli associated with disease and interventions. OBJECTIVE: Although epigenetic changes are critical in SMC phenotype switching, a fundamental regulator that governs the epigenetic machineries regulating the fate of SMC phenotype has not been elucidated. METHODS AND RESULTS: Using SMCs, mouse models, and human atherosclerosis specimens, we found that FAK (focal adhesion kinase) activation elicits SMC dedifferentiation by stabilizing DNMT3A (DNA methyltransferase 3A). FAK in SMCs is activated in the cytoplasm upon serum stimulation in vitro or vessel injury and active FAK prevents DNMT3A from nuclear FAK-mediated degradation. However, pharmacological or genetic FAK catalytic inhibition forced FAK nuclear localization, which reduced DNMT3A protein via enhanced ubiquitination and proteasomal degradation. Reduced DNMT3A protein led to DNA hypomethylation in contractile gene promoters, which increased SMC contractile protein expression. RNA-sequencing identified SMC contractile genes as a foremost upregulated group by FAK inhibition from injured femoral artery samples compared with vehicle group. DNMT3A knockdown in injured arteries reduced DNA methylation and enhanced contractile gene expression supports the notion that nuclear FAK-mediated DNMT3A degradation via E3 ligase TRAF6 (TNF [tumor necrosis factor] receptor-associated factor 6) drives differentiation of SMCs. Furthermore, we observed that SMCs of human atherosclerotic lesions exhibited decreased nuclear FAK, which was associated with increased DNMT3A levels and decreased contractile gene expression. CONCLUSIONS: This study reveals that nuclear FAK induced by FAK catalytic inhibition specifically suppresses DNMT3A expression in injured vessels resulting in maintaining SMC differentiation by promoting the contractile gene expression. Thus, FAK inhibitors may provide a new treatment option to block SMC phenotypic switching during vascular remodeling and atherosclerosis.


Subject(s)
Cell Dedifferentiation , Contractile Proteins/genetics , DNA Methylation , Focal Adhesion Kinase 1/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Animals , Cells, Cultured , Contractile Proteins/metabolism , DNA Methyltransferase 3A/genetics , DNA Methyltransferase 3A/metabolism , Focal Adhesion Kinase 1/genetics , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Inbred C57BL , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/physiology , Proteolysis , Ubiquitination , Up-Regulation
3.
PLoS Genet ; 16(1): e1008558, 2020 01.
Article in English | MEDLINE | ID: mdl-31923184

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.


Subject(s)
Beclin-1/genetics , Chromosomal Instability , Haploinsufficiency , Microtubule-Associated Proteins/genetics , Ovarian Neoplasms/genetics , Animals , Carcinogenesis/genetics , Cell Line, Tumor , Cell Movement , Female , Metabolome , Mice , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology
4.
Circ Res ; 125(2): 152-166, 2019 07 05.
Article in English | MEDLINE | ID: mdl-31096851

ABSTRACT

RATIONALE: Neointimal 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). OBJECTIVE: To understand the mechanism of FAK action in SMC proliferation and neointimal hyperplasia. METHODS AND RESULTS: Using 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. CONCLUSIONS: Nuclear 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.


Subject(s)
Cell Proliferation , Cyclin D1/metabolism , Focal Adhesion Kinase 1/metabolism , GATA4 Transcription Factor/metabolism , Myocytes, Smooth Muscle/metabolism , Tunica Intima/metabolism , Active Transport, Cell Nucleus , Animals , Cell Nucleus/metabolism , Cells, Cultured , Cyclin D1/genetics , Focal Adhesion Kinase 1/antagonists & inhibitors , Hyperplasia/metabolism , Mice , Mice, Inbred C57BL , Myocytes, Smooth Muscle/physiology , Tunica Intima/pathology
5.
J Cell Sci ; 129(8): 1580-91, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26906414

ABSTRACT

Oxidized low-density lipoprotein (oxLDL) accumulates early in atherosclerosis and promotes endothelial nuclear factor κB (NF-κB) activation, proinflammatory gene expression and monocyte adhesion. Like for other atherogenic factors, oxLDL-induced proinflammatory responses requires integrin-dependent focal adhesion kinase (FAK, also known as PTK2) signaling; however, the mechanism by which FAK mediates oxLDL-dependent NF-κB signaling has yet to be revealed. We now show that oxLDL induces NF-κB activation and VCAM-1 expression through FAK-dependent IκB kinase ß (IKKß, also known as IKBKB) activation. We further identify FAK-dependent activation of p90 ribosomal S6 kinase family proteins (RSK) as a crucial mediator of oxLDL-dependent IKKß and NF-κB signaling, as inhibiting RSK blocks oxLDL-induced IKKß and NF-κB activation, VCAM-1 expression and monocyte adhesion. Finally, transgenic mice containing a kinase-dead mutation in FAK specifically in the endothelial cells show reduced RSK activity, decreased VCAM-1 expression and reduced macrophage accumulation in regions of early atherosclerosis. Taken together, our data elucidates a new mechanism whereby oxLDL-induced endothelial FAK signaling drives an ERK-RSK pathway to activate IKKß and NF-κB signaling and proinflammatory gene expression.


Subject(s)
Atherosclerosis/metabolism , Endothelial Cells/physiology , Focal Adhesion Kinase 1/metabolism , I-kappa B Kinase/metabolism , Lipoproteins, LDL/metabolism , NF-kappa B/metabolism , Vascular Cell Adhesion Molecule-1/metabolism , Animals , Cell Adhesion , Focal Adhesion Kinase 1/genetics , Gene Expression Regulation , HEK293 Cells , Humans , I-kappa B Kinase/genetics , Macrophages/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , NF-kappa B/genetics , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Signal Transduction , Vascular Cell Adhesion Molecule-1/genetics
6.
J Biol Chem ; 290(24): 15197-209, 2015 Jun 12.
Article in English | MEDLINE | ID: mdl-25922072

ABSTRACT

The guanine nucleotide exchange factor Rgnef (also known as ArhGEF28 or p190RhoGEF) promotes colon carcinoma cell motility and tumor progression via interaction with focal adhesion kinase (FAK). Mechanisms of Rgnef activation downstream of integrin or G protein-coupled receptors remain undefined. In the absence of a recognized G protein signaling homology domain in Rgnef, no proximal linkage to G proteins was known. Utilizing multiple methods, we have identified Rgnef as a new effector for Gα13 downstream of gastrin and the type 2 cholecystokinin receptor. In DLD-1 colon carcinoma cells depleted of Gα13, gastrin-induced FAK Tyr(P)-397 and paxillin Tyr(P)-31 phosphorylation were reduced. RhoA GTP binding and promoter activity were increased by Rgnef in combination with active Gα13. Rgnef co-immunoprecipitated with activated Gα13Q226L but not Gα12Q229L. The Rgnef C-terminal (CT, 1279-1582) region was sufficient for co-immunoprecipitation, and Rgnef-CT exogenous expression prevented Gα13-stimulated SRE activity. A domain at the C terminus of the protein close to the FAK binding domain is necessary to bind to Gα13. Point mutations of Rgnef-CT residues disrupt association with active Gα13 but not Gαq. These results show that Rgnef functions as an effector of Gα13 signaling and that this linkage may mediate FAK activation in DLD-1 colon carcinoma cells.


Subject(s)
Colonic Neoplasms/metabolism , GTP-Binding Protein alpha Subunits, G12-G13/physiology , Gastrins/physiology , Guanine Nucleotide Exchange Factors/metabolism , Cell Line, Tumor , Colonic Neoplasms/pathology , Focal Adhesion Protein-Tyrosine Kinases/metabolism , HEK293 Cells , Humans , Paxillin/chemistry , Paxillin/metabolism , Phosphorylation , Receptor, Cholecystokinin B/metabolism , Rho Guanine Nucleotide Exchange Factors/chemistry , Rho Guanine Nucleotide Exchange Factors/metabolism , Tyrosine/metabolism
7.
Breast Cancer Res ; 17: 47, 2015 Mar 28.
Article in English | MEDLINE | ID: mdl-25880415

ABSTRACT

INTRODUCTION: Focal 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. METHODS: Human 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. RESULTS: Pharmacological (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. CONCLUSIONS: FAK 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.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Focal Adhesion Protein-Tyrosine Kinases/metabolism , GTP-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Animals , Breast Neoplasms/genetics , Cell Cycle/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Enzyme Activation , Female , Focal Adhesion Protein-Tyrosine Kinases/antagonists & inhibitors , Focal Adhesion Protein-Tyrosine Kinases/genetics , Humans , Mice , Nucleophosmin , Protein Kinase Inhibitors/pharmacology , Protein Transport , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Sirolimus/pharmacology , Spheroids, Cellular , Tumor Burden/drug effects , Tumor Burden/genetics , Tumor Cells, Cultured , Tumor Stem Cell Assay
8.
J Cell Sci ; 126(Pt 21): 5074-85, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24006257

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.


Subject(s)
Focal Adhesion Protein-Tyrosine Kinases/metabolism , Integrin beta1/metabolism , ras-GRF1/metabolism , Amino Acid Sequence , Animals , Cell Adhesion , Cells, Cultured , Enzyme Activation , Fibroblasts/cytology , Fibroblasts/enzymology , Fibroblasts/metabolism , Fibronectins/metabolism , Focal Adhesion Protein-Tyrosine Kinases/chemistry , Focal Adhesion Protein-Tyrosine Kinases/genetics , Integrin beta1/genetics , Mice , Mice, Knockout , Molecular Sequence Data , Paxillin/genetics , Paxillin/metabolism , Phosphorylation , Protein Binding , Protein Structure, Tertiary , Sequence Alignment , Signal Transduction , ras-GRF1/chemistry , ras-GRF1/genetics
9.
J Cell Sci ; 125(Pt 24): 5960-73, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23077174

ABSTRACT

Transmembrane 4 L six family member 5 (TM4SF5) plays an important role in cell migration, and focal adhesion kinase (FAK) activity is essential for homeostatic and pathological migration of adherent cells. However, it is unclear how TM4SF5 signaling mediates the activation of cellular migration machinery, and how FAK is activated during cell adhesion. Here, we showed that direct and adhesion-dependent binding of TM4SF5 to FAK causes a structural alteration that may release the inhibitory intramolecular interaction in FAK. In turn, this may activate FAK at the cell's leading edge, to promote migration/invasion and in vivo metastasis. TM4SF5-mediated FAK activation occurred during integrin-mediated cell adhesion. TM4SF5 was localized at the leading edge of the cells, together with FAK and actin-organizing molecules, indicating a signaling link between TM4SF5/FAK and actin reorganization machinery. Impaired interactions between TM4SF5 and FAK resulted in an attenuated FAK phosphorylation (the signaling link to actin organization machinery) and the metastatic potential. Our findings demonstrate that TM4SF5 directly binds to and activates FAK in an adhesion-dependent manner, to regulate cell migration and invasion, suggesting that TM4SF5 is a promising target in the treatment of metastatic cancer.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Focal Adhesion Kinase 1/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Tetraspanins/genetics , Amino Acid Sequence , Animals , Carcinoma, Hepatocellular/enzymology , Cell Adhesion/physiology , Cell Movement/physiology , Enzyme Activation , Female , Heterografts , Humans , Liver Neoplasms/enzymology , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Neoplasm Metastasis , Phosphorylation , Signal Transduction , Tetraspanins/metabolism
10.
Gynecol Oncol ; 134(1): 104-11, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24786638

ABSTRACT

OBJECTIVE: Focal 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. METHODS: Nine 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. RESULTS: Greater than 50% inhibition of OVCAR8, HEY, and ID8-IP ovarian carcinoma cell growth occurred with 0.1 µM PF-271 in anchorage-independent (p<0.001) but not in adherent culture conditions. PF-271-mediated reduction in FAK Y397 phosphorylation occurred independently of growth inhibition. Suspended growth of OVCAR3, OVCAR10, IGROV1, IGROV1-IP, SKOV3, SKOV3-IP, A2780, and 5009-MOVCAR was not affected by 0.1 µM PF-271. Merlin expression did not correlate with serous ovarian tumor grade or stage. PF-271 (30 mg/kg, BID) did not inhibit 5009-MOVCAR tumor growth and merlin knockdown in SKOV3-IP and OVCAR10 cells did not alter suspended cell growth upon PF-271 addition. CONCLUSIONS: Differential responsiveness to FAK inhibitor treatment was observed. Intrinsic low merlin protein level correlated with PF-271-mediated anchorage-independent growth inhibition, but reduction in merlin expression did not induce sensitivity to FAK inhibition. Merlin levels may be useful for patient stratification in FAK inhibitor trials.


Subject(s)
Cystadenocarcinoma, Serous/drug therapy , Focal Adhesion Kinase 1/antagonists & inhibitors , Neurofibromin 2/metabolism , Ovarian Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Animals , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Cystadenocarcinoma, Serous/enzymology , Cystadenocarcinoma, Serous/metabolism , Female , Focal Adhesion Kinase 1/metabolism , Gene Knockdown Techniques , Humans , Mice , Neurofibromin 2/genetics , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/metabolism
11.
Biomaterials ; 308: 122542, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38547833

ABSTRACT

Focal adhesions (FAs) are nanoscale complexes containing clustered integrin receptors and intracellular structural and signaling proteins that function as principal sites of mechanotransduction in part via promoting the nuclear translocation and activation of the transcriptional coactivator yes-associated protein (YAP). Knockdown of FA proteins such as focal adhesion kinase (FAK), talin, and vinculin can prevent YAP nuclear localization. However, the mechanism(s) of action remain poorly understood. Herein, we investigated the role of different functional domains in vinculin, talin, and FAK in regulating YAP nuclear localization. Using genetic or pharmacological inhibition of fibroblasts and human mesenchymal stem cells (hMSCs) adhering to deformable substrates, we find that disruption of vinculin-talin binding versus talin-FAK binding reduces YAP nuclear localization and transcriptional activity via different mechanisms. Disruption of vinculin-talin binding or knockdown of talin-1 reduces nuclear size, traction forces, and YAP nuclear localization. In contrast, disruption of the talin binding site on FAK or elimination of FAK catalytic activity did not alter nuclear size yet still prevented YAP nuclear localization and activity. These data support both nuclear tension-dependent and independent models for matrix stiffness-regulated YAP nuclear localization. Our results highlight the importance of vinculin-talin-FAK interactions at FAs of adherent cells, controlling YAP nuclear localization and activity.


Subject(s)
Cell Nucleus , Mechanotransduction, Cellular , Talin , Vinculin , YAP-Signaling Proteins , Talin/metabolism , Vinculin/metabolism , Humans , Cell Nucleus/metabolism , YAP-Signaling Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Transcription Factors/metabolism , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Animals , Focal Adhesions/metabolism , Mice , Fibroblasts/metabolism , Focal Adhesion Kinase 1/metabolism , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Protein Binding
12.
Curr Opin Cell Biol ; 18(5): 516-23, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16919435

ABSTRACT

Integrins can alter cellular behavior through the recruitment and activation of signaling proteins such as non-receptor tyrosine kinases including focal adhesion kinase (FAK) and c-Src that form a dual kinase complex. The FAK-Src complex binds to and can phosphorylate various adaptor proteins such as p130Cas and paxillin. In normal cells, multiple integrin-regulated linkages exist to activate FAK or Src. Activated FAK-Src functions to promote cell motility, cell cycle progression and cell survival. Recent studies have found that the FAK-Src complex is activated in many tumor cells and generates signals leading to tumor growth and metastasis. As both FAK and Src catalytic activities are important in promoting VEGF-associated tumor angiogenesis and protease-associated tumor metastasis, support is growing that FAK and Src may be therapeutically relevant targets in the inhibition of tumor progression.


Subject(s)
Focal Adhesion Protein-Tyrosine Kinases/metabolism , Integrins/metabolism , Neoplasms/metabolism , Signal Transduction/physiology , src-Family Kinases/metabolism , Animals , Cell Movement/physiology , Cell Survival , Disease Progression , Enzyme Activation , Neoplasms/pathology
13.
Exp Mol Med ; 55(3): 520-531, 2023 03.
Article in English | MEDLINE | ID: mdl-36854775

ABSTRACT

Extracellular matrix proteins are associated with metabolically healthy adipose tissue and regulate inflammation, fibrosis, angiogenesis, and subsequent metabolic deterioration. In this study, we demonstrated that transforming growth factor-beta (TGFBI), an extracellular matrix (ECM) component, plays an important role in adipose metabolism and browning during high-fat diet-induced obesity. TGFBI KO mice were resistant to adipose tissue hypertrophy, liver steatosis, and insulin resistance. Furthermore, adipose tissue from TGFBI KO mice contained a large population of CD11b+ and CD206+ M2 macrophages, which possibly control adipokine secretion through paracrine mechanisms. Mechanistically, we showed that inhibiting TGFBI-stimulated release of adipsin by Notch-1-dependent signaling resulted in adipocyte browning. TGFBI was physiologically bound to Notch-1 and stimulated its activation in adipocytes. Our findings revealed a novel protective effect of TGFBI deficiency in obesity that is realized via the activation of the Notch-1 signaling pathway.


Subject(s)
Insulin Resistance , Transforming Growth Factor beta , Mice , Animals , Transforming Growth Factor beta/metabolism , Obesity/metabolism , Adipose Tissue/metabolism , Adipocytes/metabolism , Signal Transduction , Diet, High-Fat/adverse effects , Mice, Inbred C57BL , Adipose Tissue, White/metabolism
14.
J Biol Chem ; 285(3): 1743-53, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-19880522

ABSTRACT

Pyk2 is a cytoplasmic tyrosine kinase related to focal adhesion kinase (FAK). Compensatory Pyk2 expression occurs upon FAK loss in mice. However, the impact of Pyk2 up-regulation remains unclear. Previous studies showed that nuclear-localized FAK promotes cell proliferation and survival through FAK FERM domain-enhanced p53 tumor suppressor degradation (Lim, S. T., Chen, X. L., Lim, Y., Hanson, D. A., Vo, T. T., Howerton, K., Larocque, N., Fisher, S. J., Schlaepfer, D. D., and Ilic, D. (2008) Mol. Cell 29, 9-22). Here, we show that FAK knockdown triggered p53 activation and G(1) cell cycle arrest in human umbilical vein endothelial cells after 4 days. However, by 7 days elevated Pyk2 expression occurred with a reduction in p53 levels and the release of the G(1) block under conditions of continued FAK knockdown. To determine whether Pyk2 regulates p53, experiments were performed in FAK(-/-)p21(-/-) mouse embryo fibroblasts expressing endogenous Pyk2 and in ID8 ovarian carcinoma cells expressing both Pyk2 and FAK. In both cell lines, Pyk2 knockdown increased p53 levels and inhibited cell proliferation associated with G(1) cell cycle arrest. Pyk2 FERM domain re-expression was sufficient to reduce p53 levels and promote increased BrdUrd incorporation. Pyk2 FERM promoted Mdm2-dependent p53 ubiquitination. Pyk2 FERM effects on p53 were blocked by proteasomal inhibition or mutational-inactivation of Pyk2 FERM nuclear localization. Staurosporine stress of ID8 cells promoted endogenous Pyk2 nuclear accumulation and enhanced Pyk2 binding to p53. Pyk2 knockdown potentiated ID8 cell death upon staurosporine addition. Moreover, Pyk2 FERM expression in human fibroblasts upon FAK knockdown prevented cisplatin-mediated apoptosis. Our studies demonstrate that nuclear Pyk2 functions to limit p53 levels, thus facilitating cell growth and survival in a kinase-independent manner.


Subject(s)
Adaptation, Physiological , Focal Adhesion Kinase 2/metabolism , Tumor Suppressor Protein p53/metabolism , Active Transport, Cell Nucleus , Animals , Bromodeoxyuridine/metabolism , Cell Cycle/genetics , Cell Line, Tumor , Cell Proliferation , Cell Survival/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Focal Adhesion Kinase 2/chemistry , Focal Adhesion Kinase 2/deficiency , Focal Adhesion Kinase 2/genetics , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Mice , Mutation , Protein Structure, Tertiary , Proto-Oncogene Proteins c-mdm2/metabolism , Staurosporine/pharmacology , Stress, Physiological/drug effects , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics
15.
J Biol Chem ; 285(28): 21526-36, 2010 Jul 09.
Article in English | MEDLINE | ID: mdl-20442405

ABSTRACT

Focal adhesion kinase (FAK) associates with both integrins and growth factor receptors in the control of cell motility and survival. Loss of FAK during mouse development results in lethality at embryonic day 8.5 (E8.5) and a block in cell proliferation. Because FAK serves as both a scaffold and signaling protein, gene knock-outs do not provide mechanistic insights in distinguishing between these modes of FAK function. To determine the role of FAK activity during development, a knock-in point mutation (lysine 454 to arginine (R454)) within the catalytic domain was introduced by homologous recombination. Homozygous FAK(R454/R454) mutation was lethal at E9.5 with defects in blood vessel formation as determined by lack of yolk sac primary capillary plexus formation and disorganized endothelial cell patterning in FAK(R454/R454) embryos. In contrast to the inability of embryonic FAK(-/-) cells to proliferate ex vivo, primary FAK(R454/R454) mouse embryo fibroblasts (MEFs) were established from E8.5 embryos. R454 MEFs exhibited no difference in cell growth compared with normal MEFs, and R454 FAK localized to focal adhesions but was not phosphorylated at Tyr-397. In E8.5 embryos and primary MEFs, FAK R454 mutation resulted in decreased c-Src Tyr-416 phosphorylation. R454 MEFs exhibited enhanced focal adhesion formation, decreased migration, and defects in cell polarity. Within immortalized MEFs, FAK activity was required for fibronectin-stimulated FAK-p190RhoGAP association and p190RhoGAP tyrosine phosphorylation linked to decreased RhoA GTPase activity, focal adhesion turnover, and directional motility. Our results establish that intrinsic FAK activity is essential for developmental processes controlling blood vessel formation and cell motility-polarity but not cell proliferation. This work supports the use of FAK inhibitors to disrupt neovascularization.


Subject(s)
Blood Vessels/enzymology , Focal Adhesion Protein-Tyrosine Kinases/genetics , Focal Adhesion Protein-Tyrosine Kinases/physiology , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Mutation , Animals , Blood Vessels/metabolism , Cell Movement , Cell Proliferation , Fibronectins/metabolism , GTPase-Activating Proteins/metabolism , Golgi Apparatus/metabolism , Homozygote , Mice , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Recombination, Genetic , Repressor Proteins/metabolism
16.
PLoS Pathog ; 5(2): e1000304, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19229316

ABSTRACT

While most chemokine receptors fail to cross the chemokine class boundary with respect to the ligands that they bind, the human cytomegalovirus (HCMV)-encoded chemokine receptor US28 binds multiple CC-chemokines and the CX(3)C-chemokine Fractalkine. US28 binding to CC-chemokines is both necessary and sufficient to induce vascular smooth muscle cell (SMC) migration in response to HCMV infection. However, the function of Fractalkine binding to US28 is unknown. In this report, we demonstrate that Fractalkine binding to US28 not only induces migration of macrophages but also acts to inhibit RANTES-mediated SMC migration. Similarly, RANTES inhibits Fractalkine-mediated US28 migration in macrophages. While US28 binding of both RANTES and Fractalkine activate FAK and ERK-1/2, RANTES signals through Galpha12 and Fractalkine through Galphaq. These findings represent the first example of differential chemotactic signaling via a multiple chemokine family binding receptor that results in migration of two different cell types. Additionally, the demonstration that US28-mediated chemotaxis is both ligand-specific and cell type-specific has important implications in the role of US28 in HCMV pathogenesis.


Subject(s)
Cell Movement/physiology , Chemokine CCL5/metabolism , Chemokine CX3CL1/metabolism , Receptors, Chemokine/metabolism , Viral Proteins/metabolism , Animals , Cell Line , Chemokines, CC/metabolism , Cytomegalovirus/metabolism , Focal Adhesion Protein-Tyrosine Kinases/metabolism , GTP-Binding Proteins/metabolism , Humans , Macrophages/metabolism , Muscle, Smooth/cytology , Muscle, Smooth/metabolism , Rats , Receptors, Chemokine/genetics , Signal Transduction , Stress Fibers/metabolism , Viral Proteins/genetics
17.
Nat Rev Cancer ; 21(5): 313-324, 2021 05.
Article in English | MEDLINE | ID: mdl-33731845

ABSTRACT

Focal adhesion kinase (FAK) is both a non-receptor tyrosine kinase and an adaptor protein that primarily regulates adhesion signalling and cell migration, but FAK can also promote cell survival in response to stress. FAK is commonly overexpressed in cancer and is considered a high-value druggable target, with multiple FAK inhibitors currently in development. Evidence suggests that in the clinical setting, FAK targeting will be most effective in combination with other agents so as to reverse failure of chemotherapies or targeted therapies and enhance efficacy of immune-based treatments of solid tumours. Here, we discuss the recent preclinical evidence that implicates FAK in anticancer therapeutic resistance, leading to the view that FAK inhibitors will have their greatest utility as combination therapies in selected patient populations.


Subject(s)
Focal Adhesion Protein-Tyrosine Kinases/antagonists & inhibitors , Molecular Targeted Therapy , Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , Animals , Antineoplastic Combined Chemotherapy Protocols , Humans , Neoplasms/enzymology , Neoplasms/pathology
18.
Sci Rep ; 11(1): 9644, 2021 05 06.
Article in English | MEDLINE | ID: mdl-33958649

ABSTRACT

Several studies have suggested that extracellular matrix (ECM) remodeling and the microenvironment are tightly associated with adipogenesis and adipose angiogenesis. In the present study, we demonstrated that transforming growth factor-beta induced (TGFBI) suppresses angiogenesis stimulated by adipocyte-conditioned medium (Ad-CM), both in vitro and in vivo. TGFBI knockout (KO) mice exhibited increased numbers of blood vessels in adipose tissue, and blood vessels from these mice showed enhanced infiltration into Matrigel containing Ad-CM. The treatment of Ad-CM-stimulated SVEC-10 endothelial cells with TGFBI protein reduced migration and tube-forming activity. TGFBI protein suppressed the activation of the Src and extracellular signaling-related kinase signaling pathways of these SVEC-10 endothelial cells. Our findings indicated that TGFBI inhibited adipose angiogenesis by suppressing the activation of Src and ERK signaling pathways, possibly because of the stimulation of the angiogenic activity of endothelial cells.


Subject(s)
Adipose Tissue/blood supply , Endothelium, Vascular/metabolism , Extracellular Matrix Proteins/metabolism , Neovascularization, Physiologic , Transforming Growth Factor beta/metabolism , Adipose Tissue/metabolism , Animals , Capillaries/growth & development , Capillaries/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
19.
Nat Commun ; 12(1): 2359, 2021 04 21.
Article in English | MEDLINE | ID: mdl-33883558

ABSTRACT

How adhesive forces are transduced and integrated into biochemical signals at focal adhesions (FAs) is poorly understood. Using cells adhering to deformable micropillar arrays, we demonstrate that traction force and FAK localization as well as traction force and Y397-FAK phosphorylation are linearly coupled at individual FAs on stiff, but not soft, substrates. Similarly, FAK phosphorylation increases linearly with external forces applied to FAs using magnetic beads. This mechanosignaling coupling requires actomyosin contractility, talin-FAK binding, and full-length vinculin that binds talin and actin. Using an in vitro 3D biomimetic wound healing model, we show that force-FAK signaling coupling coordinates cell migration and tissue-scale forces to promote microtissue repair. A simple kinetic binding model of talin-FAK interactions under force can recapitulate the experimental observations. This study provides insights on how talin and vinculin convert forces into FAK signaling events regulating cell migration and tissue repair.


Subject(s)
Focal Adhesion Kinase 1/metabolism , Focal Adhesions/metabolism , Models, Biological , Actomyosin/metabolism , Animals , Biomechanical Phenomena , Biomimetics , Cell Movement/physiology , Cells, Cultured , Fibroblasts/metabolism , Focal Adhesion Kinase 1/deficiency , Focal Adhesion Kinase 1/genetics , Mechanotransduction, Cellular , Mice , Mice, Knockout , Phosphorylation , RNA, Small Interfering/genetics , Signal Transduction , Talin/antagonists & inhibitors , Talin/genetics , Talin/metabolism , Wound Healing/physiology
20.
J Cell Biol ; 171(3): 505-16, 2005 Nov 07.
Article in English | MEDLINE | ID: mdl-16275754

ABSTRACT

The overexpression of members of the ErbB tyrosine kinase receptor family has been associated with cancer progression. We demonstrate that focal adhesion kinase (FAK) is essential for oncogenic transformation and cell invasion that is induced by ErbB-2 and -3 receptor signaling. ErbB-2/3 overexpression in FAK-deficient cells fails to promote cell transformation and rescue chemotaxis deficiency. Restoration of FAK rescues both oncogenic transformation and invasion that is induced by ErbB-2/3 in vitro and in vivo. In contrast, the inhibition of FAK in FAK-proficient invasive cancer cells prevented cell invasion and metastasis formation. The activation of ErbB-2/3 regulates FAK phosphorylation at Tyr-397, -861, and -925. ErbB-induced oncogenic transformation correlates with the ability of FAK to restore ErbB-2/3-induced mitogen-activated protein kinase (MAPK) activation; the inhibition of MAPK prevented oncogenic transformation. In contrast, the inhibition of Src but not MAPK prevented ErbB-FAK-induced chemotaxis. In migratory cells, activated ErbB-2/3 receptors colocalize with activated FAK at cell protrusions. This colocalization requires intact FAK. In summary, distinct FAK signaling has an essential function in ErbB-induced oncogenesis and invasiveness.


Subject(s)
Cell Transformation, Neoplastic , Focal Adhesion Protein-Tyrosine Kinases/physiology , Neoplasm Invasiveness , Receptor, ErbB-2/physiology , Receptor, ErbB-3/physiology , Animals , Cell Line , Cell Line, Tumor , Chemotaxis , Enzyme Activation , Focal Adhesion Protein-Tyrosine Kinases/genetics , Humans , MAP Kinase Signaling System/physiology , Mice , Mice, Knockout , Mice, SCID , Neoplasm Metastasis , Phosphorylation , Phosphotyrosine/metabolism , RNA, Small Interfering/genetics , Receptor, ErbB-2/genetics , Receptor, ErbB-3/genetics , Signal Transduction , src-Family Kinases/physiology
SELECTION OF CITATIONS
SEARCH DETAIL