Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
J Cell Sci ; 135(20)2022 10 15.
Article in English | MEDLINE | ID: mdl-36239192

ABSTRACT

Focal adhesion kinase (FAK; also known as PTK2) was discovered three decades ago and is now recognised as a key player in the regulation of cell-matrix adhesion and mesenchymal cell migration. Although it is essential during development, FAK also drives invasive cancer progression and metastasis. On a structural level, the basic building blocks of FAK have been described for some time. However, a picture of how FAK integrates into larger assemblies in various cellular environments, including one of its main cellular locations, the focal adhesion (FA) complex, is only beginning to emerge. Nano-resolution data from cellular studies, as well as atomic structures from reconstituted systems, have provided first insights, but also point to challenges that remain for obtaining a full structural understanding of how FAK is integrated in the FA complex and the structural changes occurring at different stages of FA maturation. In this Review, we discuss the known structural features of FAK, the interactions with its partners within the FA environment on the cell membrane and propose how its initial assembly in nascent FAs might change during FA maturation under force.


Subject(s)
Focal Adhesions , Cell Adhesion , Cell Movement , Focal Adhesion Kinase 1/metabolism , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Focal Adhesions/metabolism , Phosphorylation
2.
Proc Natl Acad Sci U S A ; 119(14): e2114397119, 2022 04 05.
Article in English | MEDLINE | ID: mdl-35312342

ABSTRACT

SignificanceIn the dynamic environment of the airways, where SARS-CoV-2 infections are initiated by binding to human host receptor ACE2, mechanical stability of the viral attachment is a crucial fitness advantage. Using single-molecule force spectroscopy techniques, we mimic the effect of coughing and sneezing, thereby testing the force stability of SARS-CoV-2 RBD:ACE2 interaction under physiological conditions. Our results reveal a higher force stability of SARS-CoV-2 binding to ACE2 compared to SARS-CoV-1, causing a possible fitness advantage. Our assay is sensitive to blocking agents preventing RBD:ACE2 bond formation. It will thus provide a powerful approach to investigate the modes of action of neutralizing antibodies and other agents designed to block RBD binding to ACE2 that are currently developed as potential COVID-19 therapeutics.


Subject(s)
Angiotensin-Converting Enzyme 2/metabolism , COVID-19/metabolism , COVID-19/virology , Host-Pathogen Interactions , SARS-CoV-2/physiology , Angiotensin-Converting Enzyme 2/chemistry , COVID-19/diagnosis , Disease Susceptibility , Humans , Protein Binding
3.
J Med Chem ; 65(2): 1585-1607, 2022 01 27.
Article in English | MEDLINE | ID: mdl-34978799

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease without any effective treatment. Protein TDP-43 is a pathological hallmark of ALS in both sporadic and familiar patients. Post-translational modifications of TDP-43 promote its aggregation in the cytoplasm. Tau-Tubulin kinase (TTBK1) phosphorylates TDP-43 in cellular and animal models; thus, TTBK1 inhibitors emerge as a promising therapeutic strategy for ALS. The design, synthesis, biological evaluation, kinase-ligand complex structure determination, and molecular modeling studies confirmed novel pyrrolopyrimidine derivatives as valuable inhibitors for further development. Moreover, compound 29 revealed good brain penetration in vivo and was able to reduce TDP-43 phosphorylation not only in cell cultures but also in the spinal cord of transgenic TDP-43 mice. A shift to M2 anti-inflammatory microglia was also demonstrated in vivo. Both these activities led to motor neuron preservation in mice, proposing pyrrolopyrimidine 29 as a valuable lead compound for future ALS therapy.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , Brain/drug effects , DNA-Binding Proteins/metabolism , Inflammation/drug therapy , Macrophages/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , Brain/metabolism , Case-Control Studies , Humans , Inflammation/metabolism , Inflammation/pathology , Macrophages/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Transgenic , Phosphorylation , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacokinetics , Rats , Rats, Wistar , Spinal Cord/drug effects , Spinal Cord/metabolism , Tissue Distribution
4.
Cancer Res ; 81(21): 5438-5450, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34417202

ABSTRACT

Despite the approval of several multikinase inhibitors that target SRC and the overwhelming evidence of the role of SRC in the progression and resistance mechanisms of many solid malignancies, inhibition of its kinase activity has thus far failed to improve patient outcomes. Here we report the small molecule eCF506 locks SRC in its native inactive conformation, thereby inhibiting both enzymatic and scaffolding functions that prevent phosphorylation and complex formation with its partner FAK. This mechanism of action resulted in highly potent and selective pathway inhibition in culture and in vivo. Treatment with eCF506 resulted in increased antitumor efficacy and tolerability in syngeneic murine cancer models, demonstrating significant therapeutic advantages over existing SRC/ABL inhibitors. Therefore, this mode of inhibiting SRC could lead to improved treatment of SRC-associated disorders. SIGNIFICANCE: Small molecule-mediated inhibition of SRC impairing both catalytic and scaffolding functions confers increased anticancer properties and tolerability compared with other SRC/ABL inhibitors.


Subject(s)
Bone Neoplasms/drug therapy , Breast Neoplasms/drug therapy , Focal Adhesion Kinase 1/antagonists & inhibitors , Piperidines/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-abl/antagonists & inhibitors , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Small Molecule Libraries/pharmacology , src-Family Kinases/antagonists & inhibitors , Animals , Apoptosis , Bone Neoplasms/metabolism , Bone Neoplasms/secondary , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Protein Conformation , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , src-Family Kinases/chemistry , src-Family Kinases/metabolism
5.
FASEB J ; 35(8): e21815, 2021 08.
Article in English | MEDLINE | ID: mdl-34314064

ABSTRACT

The Src homology 2 containing inositol 5-phosphatase 2 (SHIP2) is a large multidomain enzyme that catalyzes the dephosphorylation of the phospholipid phosphatidylinositol 3,4,5-triphosphate (PI(3,4,5)P3 ) to form PI(3,4)P2 . PI(3,4,5)P3 is a key lipid second messenger controlling the recruitment of signaling proteins to the plasma membrane, thereby regulating a plethora of cellular events, including proliferation, growth, apoptosis, and cytoskeletal rearrangements. SHIP2, alongside PI3K and PTEN, regulates PI(3,4,5)P3 levels at the plasma membrane and has been heavily implicated in serious diseases such as cancer and type 2 diabetes; however, many aspects of its regulation mechanism remain elusive. We recently reported an activating effect of the SHIP2 C2 domain and here we describe an additional layer of regulation via the pleckstrin homology-related (PHR) domain. We show a phosphoinositide-induced transition to a high activity state of the enzyme that increases phosphatase activity up to 10-15 fold. We further show that PI(3,4)P2 directly interacts with the PHR domain to trigger this allosteric activation. Modeling of the PHR-phosphatase-C2 region of SHIP2 on the membrane suggests no major inter-domain interactions with the PHR domain, but close contacts between the two linkers offer a possible path of allosteric communication. Together, our data show that the PHR domain acts as an allosteric module regulating the catalytic activity of SHIP2 in response to specific phosphoinositide levels in the cell membrane.


Subject(s)
Cell Membrane/metabolism , Phosphatidylinositol Phosphates/metabolism , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases , Pleckstrin Homology Domains , Allosteric Regulation , Biocatalysis , Humans , Models, Molecular , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/chemistry , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/metabolism
6.
EMBO J ; 40(14): e106871, 2021 07 15.
Article in English | MEDLINE | ID: mdl-34124795

ABSTRACT

Low-density lipoprotein (LDL)-cholesterol delivery from late endosomes to the plasma membrane regulates focal adhesion dynamics and cell migration, but the mechanisms controlling it are poorly characterized. Here, we employed auxin-inducible rapid degradation of oxysterol-binding protein-related protein 2 (ORP2/OSBPL2) to show that endogenous ORP2 mediates the transfer of LDL-derived cholesterol from late endosomes to focal adhesion kinase (FAK)-/integrin-positive recycling endosomes in human cells. In vitro, cholesterol enhances membrane association of FAK to PI(4,5)P2 -containing lipid bilayers. In cells, ORP2 stimulates FAK activation and PI(4,5)P2 generation in endomembranes, enhancing cell adhesion. Moreover, ORP2 increases PI(4,5)P2 in NPC1-containing late endosomes in a FAK-dependent manner, controlling their tubulovesicular trafficking. Together, these results provide evidence that ORP2 controls FAK activation and LDL-cholesterol plasma membrane delivery by promoting bidirectional cholesterol/PI(4,5)P2 exchange between late and recycling endosomes.


Subject(s)
Biological Transport/physiology , Cholesterol, LDL/metabolism , Endosomes/metabolism , Focal Adhesion Kinase 1/metabolism , Phosphatidylinositol Phosphates/metabolism , Receptors, Steroid/metabolism , Cell Adhesion/physiology , Cell Line, Tumor , Cell Membrane/metabolism , Cell Movement/physiology , Humans
7.
J Med Chem ; 63(21): 12707-12724, 2020 11 12.
Article in English | MEDLINE | ID: mdl-33119295

ABSTRACT

Human malignant glioblastoma (GBM) is a highly invasive and lethal brain tumor. Targeting of integrin downstream signaling mediators in GBM such as focal adhesion kinase (FAK) seems reasonable and recently demonstrated promising results in early clinical studies. Herein, we report the structure-guided development of a series of covalent inhibitors of FAK. These new compounds displayed highly potent inhibitory potency against FAK enzymatic activity with IC50 values in the nanomolar range. Several inhibitors retarded tumor cell growth as assessed by a cell viability assay in multiple human glioblastoma cell lines. They also significantly reduced the rate of U-87 cell migration and delayed the cell cycle progression by stopping cells in the G2/M phase. Furthermore, these inhibitors showed a potent decrease of autophosphorylation of FAK in glioblastoma cells and its downstream effectors Akt and Erk as well as nuclear factor-κB. These data demonstrated that these inhibitors may have the potential to offer a promising new targeted therapy for human glioblastomas.


Subject(s)
Drug Design , Focal Adhesion Protein-Tyrosine Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/chemical synthesis , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Focal Adhesion Protein-Tyrosine Kinases/metabolism , G2 Phase Cell Cycle Checkpoints/drug effects , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , NF-kappa B/metabolism , Phosphorylation/drug effects , Protein Binding , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Structure-Activity Relationship
8.
EMBO J ; 39(19): e104743, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32779739

ABSTRACT

Focal adhesion kinase (FAK) is a key component of the membrane proximal signaling layer in focal adhesion complexes, regulating important cellular processes, including cell migration, proliferation, and survival. In the cytosol, FAK adopts an autoinhibited state but is activated upon recruitment into focal adhesions, yet how this occurs or what induces structural changes is unknown. Here, we employ cryo-electron microscopy to reveal how FAK associates with lipid membranes and how membrane interactions unlock FAK autoinhibition to promote activation. Intriguingly, initial binding of FAK to the membrane causes steric clashes that release the kinase domain from autoinhibition, allowing it to undergo a large conformational change and interact itself with the membrane in an orientation that places the active site toward the membrane. In this conformation, the autophosphorylation site is exposed and multiple interfaces align to promote FAK oligomerization on the membrane. We show that interfaces responsible for initial dimerization and membrane attachment are essential for FAK autophosphorylation and resulting cellular activity including cancer cell invasion, while stable FAK oligomerization appears to be needed for optimal cancer cell proliferation in an anchorage-independent manner. Together, our data provide structural details of a key membrane bound state of FAK that is primed for efficient autophosphorylation and activation, hence revealing the critical event in integrin mediated FAK activation and signaling at focal adhesions.


Subject(s)
Avian Proteins/chemistry , Focal Adhesion Protein-Tyrosine Kinases/chemistry , Membranes/chemistry , Protein Multimerization , Animals , Avian Proteins/metabolism , Chickens , Enzyme Activation , Focal Adhesion Protein-Tyrosine Kinases/metabolism , HEK293 Cells , Humans , Membranes/enzymology , Structure-Activity Relationship
9.
Int J Mol Sci ; 21(15)2020 Aug 01.
Article in English | MEDLINE | ID: mdl-32752284

ABSTRACT

The composition and organization of the plasma membrane play important functional and regulatory roles in integrin signaling, which direct many physiological and pathological processes, such as development, wound healing, immunity, thrombosis, and cancer metastasis. Membranes are comprised of regions that are thick or thin owing to spontaneous partitioning of long-chain saturated lipids from short-chain polyunsaturated lipids into domains defined as ordered and liquid-disorder domains, respectively. Liquid-ordered domains are typically 100 nm in diameter and sometimes referred to as lipid rafts. We posit that integrin ß senses membrane thickness and that mechanical force on the membrane regulates integrin activation through membrane thinning. This review examines what we know about the nature and mechanism of the interaction of integrins with the plasma membrane and its effects on regulating integrins and its binding partners.


Subject(s)
Cell Membrane/metabolism , Focal Adhesions/metabolism , Integrins/metabolism , Membrane Microdomains/metabolism , Signal Transduction , Cell Adhesion , Humans , Membrane Lipids/metabolism , Membrane Proteins/metabolism , Protein Binding
10.
Biomolecules ; 10(2)2020 01 24.
Article in English | MEDLINE | ID: mdl-31991559

ABSTRACT

Focal adhesion kinase (FAK) is a non-receptor tyrosine kinase with key roles in the regulation of cell adhesion migration, proliferation and survival. In cancer FAK is a major driver of invasion and metastasis and its upregulation is associated with poor patient prognosis. FAK is autoinhibited in the cytosol, but activated upon localisation into a protein complex, known as focal adhesion complex. This complex forms upon cell adhesion to the extracellular matrix (ECM) at the cytoplasmic side of the plasma membrane at sites of ECM attachment. FAK is anchored to the complex via multiple sites, including direct interactions with specific membrane lipids and connector proteins that attach focal adhesions to the actin cytoskeleton. In migrating cells, the contraction of actomyosin stress fibres attached to the focal adhesion complex apply a force to the complex, which is likely transmitted to the FAK protein, causing stretching of the FAK molecule. In this review we discuss the current knowledge of the FAK structure and how specific structural features are involved in the regulation of FAK signalling. We focus on two major regulatory mechanisms known to contribute to FAK activation, namely interactions with membrane lipids and stretching forces applied to FAK, and discuss how they might induce structural changes that facilitate FAK activation.


Subject(s)
Cell Adhesion/genetics , Focal Adhesion Kinase 1/genetics , Focal Adhesions/genetics , Mechanotransduction, Cellular/genetics , Actin Cytoskeleton/genetics , Actin Cytoskeleton/ultrastructure , Cell Movement , Focal Adhesion Kinase 1/ultrastructure , Focal Adhesions/ultrastructure , Humans , Membranes/ultrastructure , Phosphorylation , Signal Transduction/genetics
11.
Mol Cancer Ther ; 19(2): 637-649, 2020 02.
Article in English | MEDLINE | ID: mdl-31784455

ABSTRACT

We mutated the focal adhesion kinase (FAK) catalytic domain to inhibit binding of the chaperone Cdc37 and ATP, mimicking the actions of a FAK kinase inhibitor. We reexpressed mutant and wild-type FAK in squamous cell carcinoma (SCC) cells from which endogenous FAK had been deleted, genetically fixing one axis of a FAK inhibitor combination high-content phenotypic screen to discover drugs that may synergize with FAK inhibitors. Histone deacetylase (HDAC) inhibitors represented the major class of compounds that potently induced multiparametric phenotypic changes when FAK was rendered kinase-defective or inhibited pharmacologically in SCC cells. Combined FAK and HDAC inhibitors arrest proliferation and induce apoptosis in a subset of cancer cell lines in vitro and efficiently inhibit their growth as tumors in vivo Mechanistically, HDAC inhibitors potentiate inhibitor-induced FAK inactivation and impair FAK-associated nuclear YAP in sensitive cancer cell lines. Here, we report the discovery of a new, clinically actionable, synergistic combination between FAK and HDAC inhibitors.


Subject(s)
Focal Adhesion Protein-Tyrosine Kinases/therapeutic use , Histone Deacetylase Inhibitors/therapeutic use , Neoplasms/drug therapy , Animals , Cell Proliferation , Drug Synergism , Humans , Mice , Signal Transduction
12.
Proc Natl Acad Sci U S A ; 116(14): 6766-6774, 2019 04 02.
Article in English | MEDLINE | ID: mdl-30877242

ABSTRACT

Focal adhesion kinase (FAK) is a key signaling molecule regulating cell adhesion, migration, and survival. FAK localizes into focal adhesion complexes formed at the cytoplasmic side of cell attachment to the ECM and is activated after force generation via actomyosin fibers attached to this complex. The mechanism of translating mechanical force into a biochemical signal is not understood, and it is not clear whether FAK is activated directly by force or downstream to the force signal. We use experimental and computational single-molecule force spectroscopy to probe the mechanical properties of FAK and examine whether force can trigger activation by inducing conformational changes in FAK. By comparison with an open and active mutant of FAK, we are able to assign mechanoactivation to an initial rupture event in the low-force range. This activation event occurs before FAK unfolding at forces within the native range in focal adhesions. We are also able to assign all subsequent peaks in the force landscape to partial unfolding of FAK modules. We show that binding of ATP stabilizes the kinase domain, thereby altering the unfolding hierarchy. Using all-atom molecular dynamics simulations, we identify intermediates along the unfolding pathway, which provide buffering to allow extension of FAK in focal adhesions without compromising functionality. Our findings strongly support that forces in focal adhesions applied to FAK via known interactions can induce conformational changes, which in turn, trigger focal adhesion signaling.


Subject(s)
Adenosine Triphosphate/chemistry , Avian Proteins/chemistry , Focal Adhesion Protein-Tyrosine Kinases/chemistry , Molecular Dynamics Simulation , Protein Unfolding , Adenosine Triphosphate/metabolism , Animals , Avian Proteins/genetics , Avian Proteins/metabolism , Chickens , Enzyme Activation , Focal Adhesion Protein-Tyrosine Kinases/genetics , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Focal Adhesions/enzymology , Focal Adhesions/genetics , Mechanotransduction, Cellular/genetics , Protein Domains , Structure-Activity Relationship
13.
ACS Chem Biol ; 13(8): 2067-2073, 2018 08 17.
Article in English | MEDLINE | ID: mdl-29897729

ABSTRACT

Focal Adhesion Kinase signaling pathway and its functions have been involved in the development and aggressiveness of tumor malignancy, it then presents a promising cancer therapeutic target. Several reversible FAK inhibitors have been developed and are being conducted in clinical trials. On the other hand, irreversible covalent inhibitors would bring many desirable pharmacological features including high potency and increased duration of action. Herein we report the structure-guided development of the first highly potent and irreversible inhibitor of the FAK kinase. This inhibitor showed a very potent decrease of autophosphorylation of FAK in squamous cell carcinoma. A cocrystal structure of the FAK kinase domain in complex with this compound revealed the inhibitor binding mode within the ATP binding site and confirmed the covalent linkage between the targeted Cys427 of the protein and the inhibitor.


Subject(s)
Drug Design , Focal Adhesion Protein-Tyrosine Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Amino Acid Sequence , Animals , Binding Sites/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Focal Adhesion Protein-Tyrosine Kinases/chemistry , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Humans , Models, Molecular , Neoplasms/drug therapy , Phosphorylation/drug effects , Protein Kinase Inhibitors/chemical synthesis , Sequence Alignment
14.
Clin Cancer Res ; 24(15): 3550-3559, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29588308

ABSTRACT

Purpose: Despite the wide use of antiangiogenic drugs in the clinical setting, predictive biomarkers of response to these drugs are still unknown.Experimental Design: We applied whole-exome sequencing of matched germline and basal plasma cell-free DNA samples (WES-cfDNA) on a RAS/BRAF/PIK3CA wild-type metastatic colorectal cancer patient with primary resistance to standard treatment regimens, including inhibitors to the VEGF:VEGFR2 pathway. We performed extensive functional experiments, including ectopic expression of VEGFR2 mutants in different cell lines, kinase and drug sensitivity assays, and cell- and patient-derived xenografts.Results: WES-cfDNA yielded a 77% concordance rate with tumor exome sequencing and enabled the identification of the KDR/VEGFR2 L840F clonal, somatic mutation as the cause of therapy refractoriness in our patient. In addition, we found that 1% to 3% of samples from cancer sequencing projects harbor KDR somatic mutations located in protein residues frequently mutated in other cancer-relevant kinases, such as EGFR, ABL1, and ALK. Our in vitro and in vivo functional assays confirmed that L840F causes strong resistance to antiangiogenic drugs, whereas the KDR hot-spot mutant R1032Q confers sensitivity to strong VEGFR2 inhibitors. Moreover, we showed that the D717V, G800D, G800R, L840F, G843D, S925F, R1022Q, R1032Q, and S1100F VEGFR2 mutants promote tumor growth in mice.Conclusions: Our study supports WES-cfDNA as a powerful platform for portraying the somatic mutation landscape of cancer and discovery of new resistance mechanisms to cancer therapies. Importantly, we discovered that VEGFR2 is somatically mutated across tumor types and that VEGFR2 mutants can be oncogenic and control sensitivity/resistance to antiangiogenic drugs. Clin Cancer Res; 24(15); 3550-9. ©2018 AACR.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Colorectal Neoplasms/genetics , Neovascularization, Pathologic/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics , Anaplastic Lymphoma Kinase/genetics , Angiogenesis Inhibitors/adverse effects , Animals , Biomarkers, Tumor/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Cell-Free Nucleic Acids/blood , Cell-Free Nucleic Acids/genetics , Class I Phosphatidylinositol 3-Kinases/genetics , Colorectal Neoplasms/blood , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , ErbB Receptors/genetics , Exome/genetics , Female , Heterografts , Humans , Mice , Mutation , Neovascularization, Pathologic/blood , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Protein Conformation/drug effects , Proto-Oncogene Proteins c-abl/genetics , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-2/blood , Vascular Endothelial Growth Factor Receptor-2/chemistry , Exome Sequencing
15.
Elife ; 62017 08 09.
Article in English | MEDLINE | ID: mdl-28792888

ABSTRACT

SH2-containing-inositol-5-phosphatases (SHIPs) dephosphorylate the 5-phosphate of phosphatidylinositol-3,4,5-trisphosphate (PI(3,4,5)P3) and play important roles in regulating the PI3K/Akt pathway in physiology and disease. Aiming to uncover interdomain regulatory mechanisms in SHIP2, we determined crystal structures containing the 5-phosphatase and a proximal region adopting a C2 fold. This reveals an extensive interface between the two domains, which results in significant structural changes in the phosphatase domain. Both the phosphatase and C2 domains bind phosphatidylserine lipids, which likely helps to position the active site towards its substrate. Although located distant to the active site, the C2 domain greatly enhances catalytic turnover. Employing molecular dynamics, mutagenesis and cell biology, we identify two distinct allosteric signaling pathways, emanating from hydrophobic or polar interdomain interactions, differentially affecting lipid chain or headgroup moieties of PI(3,4,5)P3. Together, this study reveals details of multilayered C2-mediated effects important for SHIP2 activity and points towards interesting new possibilities for therapeutic interventions.


Subject(s)
Phosphatidylinositol Phosphates/metabolism , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/chemistry , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/metabolism , Catalytic Domain , Crystallography, X-Ray , DNA Mutational Analysis , Humans , Models, Molecular , Molecular Dynamics Simulation , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/genetics , Phosphatidylserines/metabolism , Protein Binding , Protein Conformation , Protein Domains
16.
Bioorg Med Chem Lett ; 27(8): 1727-1730, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28284808

ABSTRACT

A series of 1,3,5-triazinic inhibitors of focal adhesion kinase (FAK) has recently been shown to exert antiangiogenic activity against HUVEC cells and anticancer efficacy against several cancer cell lines. In this report, we designed and synthesized a series of new compounds containing a 1,2,4-triazine core as novel scaffold for FAK inhibitors. These compounds displayed 10-7M IC50 values, and the best one showed IC50 value of 0.23µM against FAK enzymatic activity. Among them, several inhibitors potently inhibited the proliferation of glioblastoma (U-87MG) and colon (HCT-116) cancer cell lines. Docking of compound 10 into the active site of the FAK kinase was performed to explore its potential binding mode.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Focal Adhesion Protein-Tyrosine Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Triazines/chemistry , Triazines/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Design , Drug Screening Assays, Antitumor , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Humans , Molecular Docking Simulation , Neoplasms/drug therapy , Structure-Activity Relationship
17.
Structure ; 24(8): 1223-1225, 2016 08 02.
Article in English | MEDLINE | ID: mdl-27486913

ABSTRACT

Focal adhesion kinase (FAK) has known signaling roles in cytoplasmic adhesion structures, but was recently shown to act as a transcriptional regulator in the nucleus. In this issue of Structure, Cardoso et al. (2016) report that mechanical forces translocate FAK to the nucleus of cardiomyocytes, and provide structural insights into how FAK interacts with the MEF2 transcription factor to control cardiac hypertrophy.


Subject(s)
Cardiomegaly , Focal Adhesion Protein-Tyrosine Kinases/chemistry , Cell Nucleus , Cells, Cultured , Humans , Protein-Tyrosine Kinases , Signal Transduction
18.
Protein J ; 35(3): 225-30, 2016 06.
Article in English | MEDLINE | ID: mdl-27170292

ABSTRACT

The Src homology 2 containing inositol 5-phosphatase 2 (SHIP2) catalyses the dephosphorylation of the phospholipid phosphatidylinositol 3,4,5-triphosphate (PI(3,4,5)P3) to form PI(3,4)P2. PI(3,4,5)P3 is a key lipid second messenger, which can recruit signalling proteins to the plasma membrane and subsequently initiate numerous downstream signalling pathways responsible for the regulation of a plethora of cellular events such as proliferation, growth, apoptosis and cytoskeletal rearrangements. SHIP2 has been heavily implicated with several serious diseases such as cancer and type 2 diabetes but its regulation remains poorly understood. In order to gain insight into the mechanisms of SHIP2 regulation, a fragment of human SHIP2 containing the phosphatase domain and a region proposed to resemble a C2 domain was crystallized. Currently, no structural information is available on the putative C2-related domain or its relative position with respect to the phosphatase domain. Initial crystals were polycrystalline, but were optimized to obtain diffraction data to a resolution of 2.1 Å. Diffraction data analysis revealed a P212121 space group with unit cell parameters a = 136.04 Å, b = 175.84 Å, c = 176.89 Å. The Matthews coefficient is 2.54 Å(3) Da(-1) corresponding to 8 molecules in the asymmetric unit with a solvent content of 51.7 %.


Subject(s)
Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/chemistry , Cloning, Molecular , Crystallography, X-Ray , Escherichia coli/genetics , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/genetics , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/isolation & purification , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/metabolism , Protein Domains , Signal Transduction
19.
J Biol Chem ; 290(41): 24975-85, 2015 Oct 09.
Article in English | MEDLINE | ID: mdl-26286748

ABSTRACT

Protein kinase B (PKB/Akt) is an important mediator of signals that control various cellular processes including cell survival, growth, proliferation, and metabolism. PKB promotes these processes by phosphorylating many cellular targets, which trigger distinct downstream signaling events. However, how PKB is able to selectively target its substrates to induce specific cellular functions remains elusive. Here we perform a systematic study to dissect mechanisms that regulate intrinsic kinase activity versus mechanisms that specifically regulate activity toward specific substrates. We demonstrate that activation loop phosphorylation and the C-terminal hydrophobic motif are essential for high PKB activity in general. On the other hand, we identify membrane targeting, which for decades has been regarded as an essential step in PKB activation, as a mechanism mainly affecting substrate selectivity. Further, we show that PKB activity in cells can be triggered independently of PI3K by initial hydrophobic motif phosphorylation, presumably through a mechanism analogous to other AGC kinases. Importantly, different modes of PKB activation result in phosphorylation of distinct downstream targets. Our data indicate that specific mechanisms have evolved for signaling nodes, like PKB, to select between various downstream events. Targeting such mechanisms selectively could facilitate the development of therapeutics that might limit toxic side effects.


Subject(s)
Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Amino Acid Motifs , Amino Acid Sequence , Biocatalysis , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/metabolism , DNA Damage , Enzyme Activation/drug effects , Humans , Hydrophobic and Hydrophilic Interactions , Insulin-Like Growth Factor I/pharmacology , Phosphatidylinositol Phosphates/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/chemistry , Signal Transduction/drug effects , Substrate Specificity , Threonine/metabolism
20.
Biophys J ; 108(3): 698-705, 2015 Feb 03.
Article in English | MEDLINE | ID: mdl-25650936

ABSTRACT

Focal adhesion kinase (FAK) is a nonreceptor tyrosine kinase that regulates cell signaling, proliferation, migration, and development. A major mechanism of regulation of FAK activity is an intramolecular autoinhibitory interaction between two of its domains--the catalytic and FERM domains. Upon cell adhesion to the extracellular matrix, FAK is being translocated toward focal adhesion sites and activated. Interactions of FAK with phosphoinositide phosphatidylinsositol-4,5-bis-phosphate (PIP2) are required to activate FAK. However, the molecular mechanism of the activation remains poorly understood. Recent fluorescence resonance energy transfer experiments revealed a closure of the FERM-kinase interface upon ATP binding, which is reversed upon additional binding of PIP2. Here, we addressed the allosteric regulation of FAK by performing all-atom molecular-dynamics simulations of a FAK fragment containing the catalytic and FERM domains, and comparing the dynamics in the absence or presence of ATP and PIP2. As a major conformational change, we observe a closing and opening motion upon ATP and additional PIP2 binding, respectively, in good agreement with the fluorescence resonance energy transfer experiments. To reveal how the binding of the regulatory PIP2 to the FERM F2 lobe is transduced to the very distant F1/N-lobe interface, we employed force distribution analysis. We identified a network of mainly charged residue-residue interactions spanning from the PIP2 binding site to the distant interface between the kinase and FERM domains, comprising candidate residues for mutagenesis to validate the predicted mechanism of FAK activation.


Subject(s)
Adenosine Triphosphate/pharmacology , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Inositol Phosphates/pharmacology , Allosteric Regulation/drug effects , Animals , Focal Adhesion Protein-Tyrosine Kinases/chemistry , Molecular Dynamics Simulation , Principal Component Analysis , Protein Conformation , Thermodynamics
SELECTION OF CITATIONS
SEARCH DETAIL