Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Mol Cancer Ther ; 23(1): 56-67, 2024 Jan 03.
Article in English | MEDLINE | ID: mdl-37703580

ABSTRACT

Triple-negative breast cancer (TNBC) represents the most lethal and treatment-resistant breast cancer subtype with limited treatment options. We previously identified a protein complex unique to TNBC composed of the gap junction protein connexin 26 (Cx26), the pluripotency transcription factor NANOG, and focal adhesion kinase (FAK). We sought to determine whether a peptide mimetic of the interaction region of Cx26 attenuated tumor growth in preclinical models. We designed peptides based on Cx26 juxtamembrane domains and performed binding experiments with NANOG and FAK using surface plasmon resonance. Binding studies revealed that the Cx26 C-terminal tail and intracellular loop bound to NANOG and FAK with submicromolar-to-micromolar affinity and that a 5-amino acid sequence in the C-terminal tail of Cx26 (RYCSG) was sufficient for binding. Peptides with high affinity were engineered with a cell-penetrating antennapedia sequence and assessed in functional assays including cell proliferation, tumorsphere formation, and in vivo tumor growth, and downstream signaling changes were measured. The cell-penetrating Cx26 peptide (aCx26-pep) disrupted self-renewal while reducing nuclear FAK and NANOG and inhibiting NANOG target gene expression in TNBC cells but not luminal mammary epithelial cells. In vivo, aCx26-pep reduced tumor growth and proliferation and induced cell death. Here, we provide proof-of-concept that a Cx26 peptide-based strategy inhibits growth and alters NANOG activity specifically in TNBC, indicating the therapeutic potential of this targeting approach.


Subject(s)
Cell-Penetrating Peptides , Connexin 26 , Focal Adhesion Kinase 1 , Nanog Homeobox Protein , Triple Negative Breast Neoplasms , Triple Negative Breast Neoplasms/therapy , Nanog Homeobox Protein/antagonists & inhibitors , Humans , Animals , Mice , Cell Line, Tumor , Connexin 26/chemistry , Connexin 26/therapeutic use , Focal Adhesion Kinase 1/antagonists & inhibitors , Cell-Penetrating Peptides/chemistry , Cell-Penetrating Peptides/therapeutic use
2.
Sci Rep ; 13(1): 9166, 2023 06 06.
Article in English | MEDLINE | ID: mdl-37280310

ABSTRACT

A growing body of evidence suggests that oxysterols such as 25-hydroxycholesterol (25HC) are biologically active and involved in many physiological and pathological processes. Our previous study demonstrated that 25HC induces an innate immune response during viral infections by activating the integrin-focal adhesion kinase (FAK) pathway. 25HC produced the proinflammatory response by binding directly to integrins at a novel binding site (site II) and triggering the production of proinflammatory mediators such as tumor necrosis factor-α (TNF) and interleukin-6 (IL-6). 24-(S)-hydroxycholesterol (24HC), a structural isomer of 25HC, plays a critical role in cholesterol homeostasis in the human brain and is implicated in multiple inflammatory conditions, including Alzheimer's disease. However, whether 24HC can induce a proinflammatory response like 25HC in non-neuronal cells has not been studied and remains unknown. The aim of this study was to examine whether 24HC produces such an immune response using in silico and in vitro experiments. Our results indicate that despite being a structural isomer of 25HC, 24HC binds at site II in a distinct binding mode, engages in varied residue interactions, and produces significant conformational changes in the specificity-determining loop (SDL). In addition, our surface plasmon resonance (SPR) study reveals that 24HC could directly bind to integrin αvß3, with a binding affinity three-fold lower than 25HC. Furthermore, our in vitro studies with macrophages support the involvement of FAK and NFκB signaling pathways in triggering 24HC-mediated production of TNF. Thus, we have identified 24HC as another oxysterol that binds to integrin αvß3 and promotes a proinflammatory response via the integrin-FAK-NFκB pathway.


Subject(s)
Hydroxycholesterols , Integrin alphaVbeta3 , Computer Simulation , Humans , Integrin alphaVbeta3/chemistry , Integrin alphaVbeta3/metabolism , Hydroxycholesterols/chemistry , Hydroxycholesterols/metabolism , Inflammation/metabolism , Signal Transduction , Macrophages/metabolism , Models, Molecular , Thermodynamics , Protein Conformation , Surface Plasmon Resonance , Cholesterol 24-Hydroxylase/metabolism
3.
J Struct Biol ; 214(2): 107856, 2022 06.
Article in English | MEDLINE | ID: mdl-35427781

ABSTRACT

INTRODUCTION: The central C4 and C5 domains (C4C5) of cardiac myosin binding protein C (cMyBPC) contain a flexible interdomain linker and a cardiac-isoform specific loop. However, their importance in the functional regulation of cMyBPC has not been extensively studied. METHODS AND RESULTS: We expressed recombinant C4C5 proteins with deleted linker and loop regions and performed biophysical experiments to determine each of their structural and dynamic roles. We show that the linker and C5 loop regions modulate the secondary structure and thermal stability of C4C5. Furthermore, we provide evidence through extended molecular dynamics simulations and principle component analyses that C4C5 can adopt a completely bent or latched conformation. The simulation trajectory and interaction network analyses reveal that the completely bent conformation of C4C5 exhibits a specific pattern of residue-level interactions. Therefore, we propose a "hinge-and-latch" mechanism where the linker allows a great degree of flexibility and bending, while the loop aids in achieving a completely bent and latched conformation. Although this may be one of many bent positions that C4C5 can adopt, we illustrate for the first time in molecular detail that this type of large scale conformational change can occur in the central domains of cMyBPC. CONCLUSIONS: Our hinge-and-latch mechanism demonstrates that the linker and loop regions participate in dynamic modulation of cMyBPC's motion and global conformation. These structural and dynamic features may contribute to muscle isoform-specific regulation of actomyosin activity, and have potential implications regarding its ability to propagate or retract cMyBPC's regulatory N-terminal domains.


Subject(s)
Actin Cytoskeleton , Molecular Dynamics Simulation , Actin Cytoskeleton/chemistry , Protein Conformation , Protein Structure, Secondary
4.
Arterioscler Thromb Vasc Biol ; 40(1): 206-219, 2020 01.
Article in English | MEDLINE | ID: mdl-31645128

ABSTRACT

OBJECTIVE: Oxidized phospholipids (OxPL), such as the oxidized derivatives of 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphorylcholine, 1-palmitoyl-2-(5-oxovaleroyl)-sn-glycero-3-phosphorylcholine, and 1-palmitoyl-2-glutaroyl-sn-glycero-3-phosphorylcholine, have been shown to be the principal biologically active components of minimally oxidized LDL (low-density lipoprotein). The role of OxPL in cardiovascular diseases is well recognized, including activation of inflammation within vascular cells. Atherosclerotic Apoe-/- mice fed a high-fat diet develop antibodies to OxPL, and hybridoma B-cell lines producing natural anti-OxPL autoantibodies have been successfully generated and characterized. However, as yet, no studies have been reported demonstrating that treatment with OxPL neutralizing antibodies can be used to prevent or reverse advanced atherosclerosis. Approach and Results: Here, using a screening against 1-palmitoyl-2-(5-oxovaleroyl)-sn-glycero-3-phosphorylcholine/1-palmitoyl-2-glutaroyl-sn-glycero-3-phosphorylcholine, we generated a novel IgM autoantibody, 10C12, from the spleens of Apoe-/- mice fed a long-term Western diet, that demonstrated potent OxPL neutralizing activity in vitro and the ability to inhibit macrophage accumulation within arteries of Apoe-/- mice fed a Western diet for 4 weeks. Of interest, 10C12 failed to inhibit atherosclerosis progression in Apoe-/- mice treated between 18 and 26 weeks of Western diet feeding likely due at least in part to high levels of endogenous anti-OxPL antibodies. However, 10C12 treatment caused a 40% decrease in lipid accumulation within aortas of secreted IgM deficient, sIgM-/-Apoe-/-, mice fed a low-fat diet, when the antibody was administrated between 32-40 weeks of age. CONCLUSIONS: Taken together, these results provide direct evidence showing that treatment with a single autoimmune anti-OxPL IgM antibody during advanced disease stages can have an atheroprotective outcome.


Subject(s)
Atherosclerosis/diet therapy , Autoantibodies/immunology , Diet, Fat-Restricted/methods , Diet, Western , Immunoglobulin M/immunology , Animals , Apolipoproteins E/metabolism , Atherosclerosis/immunology , Atherosclerosis/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Humans , Immunohistochemistry , Male , Mice , Oxidation-Reduction
5.
ACS Sens ; 4(8): 1980-1985, 2019 08 23.
Article in English | MEDLINE | ID: mdl-31309821

ABSTRACT

Nature's great repository provides nucleic acids and amino acids as the fundamental elements of life. Inspired by the programmability of nucleic acids, DNA nanotechnology has been extensively developed based on the strand displacement reaction of nucleic acids. In comparison with nucleic acids, amino acids possess higher programmability and more functionalities owing to the diversity of the amino acid unit. However, the design of the peptide-based bimolecular cascade is still limited. We herein describe a peptide-based strand displacement reaction, which was granted with a specific biological function by addition of a functional domain onto the coiled-coil peptide based displacement substrate. The displacement substrate was specifically designed to response to Tau protein based on a well-established Tau inhibition sequence. We demonstrated that the kinetics of the designed displacement reaction can be dynamically tuned through blocking the toehold region to prevent migration. A nanomolar Tau detection linear range was achieved through the designed displacement reaction within a rapid turnaround time of 30 min. We also presented the capability of the peptide strand displacement based sensing system operating in real human biological samples and its excellent orthogonality on response to irrelevant biological components. We envision that this will be of especially high utility for the development of next-generation biotechnology.


Subject(s)
Biosensing Techniques , DNA/chemistry , Electrochemical Techniques , Peptides/chemistry , tau Proteins/chemistry , Electrodes , Gold/chemistry , Sulfur/chemistry
6.
Nat Commun ; 10(1): 1482, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30931941

ABSTRACT

Integrins are components of cell-matrix adhesions, and function as scaffolds for various signal transduction pathways. So far no lipid ligand for integrin has been reported. Here we show that a lipid, oxysterol 25-hydroxycholesterol (25HC), directly binds to α5ß1 and αvß3 integrins to activate integrin-focal adhesion kinase (FAK) signaling. Treatment of macrophages and epithelial cells with 25HC results in an increase in activated αvß3 integrin in podosome and focal adhesion matrix adhesion sites. Moreover, activation of pattern recognition receptor on macrophages induces secretion of 25HC, triggering integrin signaling and the production of proinflammatory cytokines such as TNF and IL-6. Thus, the lipid molecule 25HC is a physiologically relevant activator of integrins and is involved in positively regulating proinflammatory responses. Our data suggest that extracellular 25HC links innate immune inflammatory response with integrin signaling.


Subject(s)
Focal Adhesion Protein-Tyrosine Kinases/metabolism , Hydroxycholesterols/metabolism , Immunity, Innate/immunology , Integrin alpha5beta1/immunology , Integrin alphaVbeta3/immunology , Macrophages/immunology , Animals , Focal Adhesions , Inflammation , Integrin alpha5beta1/metabolism , Integrin alphaVbeta3/metabolism , Interleukin-6/immunology , Macrophages/metabolism , Mice , Mice, Knockout , Receptors, Pattern Recognition/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/immunology
7.
PLoS One ; 11(2): e0148905, 2016.
Article in English | MEDLINE | ID: mdl-26901772

ABSTRACT

Inflammatory agonists differentially activate gene expression of the chemokine family of proteins in endothelial cells (EC). TNF is a weak inducer of the chemokine CXCL11, while TNF and IFN-γ costimulation results in potent CXCL11 induction. The molecular mechanisms underlying TNF plus IFN-γ-mediated CXCL11 induction are not fully understood. We have previously reported that the protein arginine methyltransferase PRMT5 catalyzes symmetrical dimethylation of the NF-κB subunit p65 in EC at multiple arginine residues. Methylation of Arg30 and Arg35 on p65 is critical for TNF induction of CXCL10 in EC. Here we show that PRMT5-mediated methylation of p65 at Arg174 is required for induction of CXCL11 when EC are costimulated with TNF and IFN-γ. Knockdown of PRMT5 by RNAi reduced CXCL11 mRNA and protein levels in costimulated cells. Reconstitution of p65 Arg174Ala or Arg174Lys mutants into EC that were depleted of endogenous p65 blunted TNF plus IFN-γ-mediated CXCL11 induction. Mass spectrometric analyses showed that p65 Arg174 arginine methylation is enhanced by TNF plus IFN-γ costimulation, and is catalyzed by PRMT5. Chromatin immunoprecipitation assays (ChIP) demonstrated that PRMT5 is necessary for p65 association with the CXCL11 promoter in response to TNF plus IFN-γ. Further, reconstitution of p65 Arg174Lys mutant in EC abrogated this p65 association with the CXCL11 promoter. Finally, ChIP and Re-ChIP assays revealed that symmetrical dimethylarginine-containing proteins complexed with the CXCL11 promoter were diminished in p65 Arg174Lys-reconstituted EC stimulated with TNF and IFN-γ. In total, these results indicate that PRMT5-mediated p65 methylation at Arg174 is essential for TNF plus IFN-γ-mediated CXCL11 gene induction. We therefore suggest that the use of recently developed small molecule inhibitors of PRMT5 may present a therapeutic approach to moderating chronic inflammatory pathologies.


Subject(s)
Chemokine CXCL11/genetics , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Gene Expression Regulation/drug effects , Interferon-gamma/pharmacology , Protein-Arginine N-Methyltransferases/metabolism , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Cells, Cultured , Gene Knockdown Techniques , Human Umbilical Vein Endothelial Cells , Humans , Methylation , Mutation , Promoter Regions, Genetic , Protein-Arginine N-Methyltransferases/genetics
8.
J Biol Chem ; 289(22): 15328-39, 2014 May 30.
Article in English | MEDLINE | ID: mdl-24753255

ABSTRACT

The chemokine CXCL10/IP-10 facilitates recruitment of Th1-type leukocytes to inflammatory sites. In this study, we show that the arginine methyltransferase PRMT5 is critical for CXCL10 transcription in TNF-α-activated human endothelial cells (EC). We found that depletion of PRMT5 results in significantly reduced levels of CXCL10 mRNA, demonstrating a positive role for PRMT5 in CXCL10 induction. Chromatin immunoprecipitation experiments revealed the presence of the symmetrical dimethylarginine modification catalyzed by PRMT5 associated with the CXCL10 promoter in response to TNF-α. However, symmetrical dimethylarginine-modified proteins were not detected at the promoter in the absence of PRMT5, indicating that PRMT5 is essential for methylation to occur. Furthermore, NF-κB p65, a critical driver of TNF-α-mediated CXCL10 induction, was determined to be methylated at arginine residues. Crucially, RNAi-mediated PRMT5 depletion abrogated p65 methylation and CXCL10 promoter binding. Mass spectrometric analysis in EC identified five dimethylated arginine residues in p65, four of which are uncharacterized in the literature. Expression of Arg-to-Lys point mutants of p65 demonstrated that both Arg-30 and Arg-35 must be dimethylated to achieve full CXCL10 expression. In conclusion, we have identified previously uncharacterized p65 post-translational modifications critical for CXCL10 induction.


Subject(s)
Chemokine CXCL10/genetics , Endothelial Cells/physiology , Protein-Arginine N-Methyltransferases/metabolism , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/metabolism , Arginine/metabolism , Chemokine CXCL10/metabolism , Endothelial Cells/cytology , Humans , Inflammation/genetics , Inflammation/metabolism , Methylation , Primary Cell Culture , Promoter Regions, Genetic/physiology , Protein Processing, Post-Translational/physiology , Protein-Arginine N-Methyltransferases/genetics , Transcription, Genetic/physiology
9.
Mol Cell Biol ; 32(7): 1202-13, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22269951

ABSTRACT

The induction of proinflammatory proteins in stimulated endothelial cells (EC) requires activation of multiple transcription programs. The homeobox transcription factor HOXA9 has an important regulatory role in cytokine induction of the EC-leukocyte adhesion molecules (ELAM) E-selectin and vascular cell adhesion molecule 1 (VCAM-1). However, the mechanism underlying stimulus-dependent activation of HOXA9 is completely unknown. Here, we elucidate the molecular mechanism of HOXA9 activation by tumor necrosis factor alpha (TNF-α) and show an unexpected requirement for arginine methylation by protein arginine methyltransferase 5 (PRMT5). PRMT5 was identified as a TNF-α-dependent binding partner of HOXA9 by mass spectrometry. Small interfering RNA (siRNA)-mediated depletion of PRMT5 abrogated stimulus-dependent HOXA9 methylation with concomitant loss in E-selectin or VCAM-1 induction. Chromatin immunoprecipitation analysis revealed that PRMT5 is recruited to the E-selectin promoter following transient HOXA9 binding to its cognate recognition sequence. PRMT5 induces symmetric dimethylation of Arg140 on HOXA9, an event essential for E-selectin induction. In summary, PRMT5 is a critical coactivator component in a newly defined, HOXA9-containing transcription complex. Moreover, stimulus-dependent methylation of HOXA9 is essential for ELAM expression during the EC inflammatory response.


Subject(s)
E-Selectin/genetics , Gene Expression Regulation , Homeodomain Proteins/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Nuclear Proteins/metabolism , Vascular Cell Adhesion Molecule-1/genetics , E-Selectin/immunology , Homeodomain Proteins/genetics , Homeodomain Proteins/immunology , Human Umbilical Vein Endothelial Cells/immunology , Humans , Methylation , Nuclear Proteins/genetics , Nuclear Proteins/immunology , Promoter Regions, Genetic , Tumor Necrosis Factor-alpha/immunology , Vascular Cell Adhesion Molecule-1/immunology
10.
Am J Physiol Cell Physiol ; 298(4): C952-60, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20032511

ABSTRACT

Arterial thrombosis is a common disease leading to severe ischemia beyond the obstructing thrombus. Additionally, endothelial dysfunction at the site of thrombosis can be rescued by l-arginine supplementation or arginase blockade in several animal models. Exposure of rat aortic endothelial cells (RAECs) to thrombin upregulates arginase I mRNA and protein levels. In this study, we further investigated the molecular mechanism of thrombin-induced arginase changes in endothelial cells. Thrombin strikingly increased arginase I promoter and enzyme activity in primary cultured RAECs. Using different deletion and point mutations of the promoter, we demonstrated that the activating protein-1 (AP-1) consensus site located at -3,157 bp in the arginase I promoter was a thrombin-responsive element. Electrophoretic mobility shift assay and chromatin immunoprecipitation assay further confirmed that upon thrombin stimulation, c-Jun and activating transcription factor-2 (ATF-2) bound to the AP-1 site, which initiated the transactivation. Moreover, loss-of-function studies using small interfering RNA confirmed that recruitment of these two transcription factors to the AP-1 site was required for thrombin-induced arginase upregulation. In the course of defining the signaling pathway leading to the activation of AP-1 by thrombin, we found thrombin-induced phosphorylation of stress-activated protein kinase/c-Jun-NH(2)-terminal kinase (SAPK/JNK or JNK1/2/3) and p38 mitogen-activated protein kinase, which were followed by the phosphorylation of both c-Jun and ATF-2. These findings reveal the basis for thrombin induction of endothelial arginase I and indicate that arginase inhibition may be an attractive therapeutic alternative in the setting of arterial thrombosis and its associated endothelial dysfunction.


Subject(s)
Arginase/metabolism , Endothelial Cells/enzymology , Transcription Factor AP-1/metabolism , Activating Transcription Factor 2/genetics , Activating Transcription Factor 2/metabolism , Animals , Aorta/cytology , Arginase/genetics , Cells, Cultured , Endothelial Cells/cytology , Mitogen-Activated Protein Kinases/metabolism , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-jun/metabolism , RNA Interference , Rats , Signal Transduction/physiology , Thrombin/genetics , Thrombin/metabolism , Transcription Factor AP-1/genetics
11.
Mol Cell Biol ; 27(12): 4207-16, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17452460

ABSTRACT

The homeobox gene HOXA9 has recently been shown to be an important regulator of endothelial cell (EC) differentiation and activation in addition to its role in embryonic development and hematopoiesis. In this report, we have determined that the EC-leukocyte adhesion molecule E-selectin is a key target for HOXA9. The depletion of HOXA9 protein in ECs resulted in a significant and specific decrease in tumor necrosis factor alpha (TNF-alpha)-induced E-selectin gene expression. In addition, HOXA9 specifically activated the E-selectin gene promoter in ECs. Progressive deletional analyses together with site-specific mutagenesis of the E-selectin promoter indicated that the Abd-B-like HOX DNA-binding motif, CAATTTTATTAA, located in the proximal region spanning bp -210 to -221 upstream of the transcription start site was crucial for the promoter induction by HOXA9. Both HOXA9 in EC nuclear extract and recombinant HOXA9 protein bound to this sequence in vitro. Moreover, we showed that HOXA9 binds temporally, in a TNF-alpha-dependent manner, to the region containing this Abd-B-like element in vivo. We have thus identified a novel and functionally critical cis-regulatory element for TNF-alpha-mediated transient expression of the E-selectin gene. Further, we provide evidence that HOXA9 acts as an obligate proinflammatory factor by mediating cytokine induction of E-selectin.


Subject(s)
E-Selectin/metabolism , Endothelial Cells/metabolism , Genes, Homeobox , Homeodomain Proteins/metabolism , Transcriptional Activation , Binding Sites , Cell Culture Techniques , Cells, Cultured , DNA Mutational Analysis , E-Selectin/genetics , Endothelium, Vascular/cytology , Gene Expression Regulation , Genes, Reporter , Homeodomain Proteins/genetics , Humans , Luciferases/metabolism , Promoter Regions, Genetic , Protein Binding , Recombinant Proteins/metabolism , Transcription, Genetic , Transfection , Tumor Necrosis Factor-alpha/metabolism , Umbilical Veins/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...