Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 244
Filter
1.
Phys Chem Chem Phys ; 26(25): 17481-17488, 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38887991

ABSTRACT

This study utilizes molecular dynamics simulations aided with multiple walker parallel bias metadynamics to investigate the TCF unbinding mechanism from the ß-catenin interface. The results, consistent with experimental binding affinity calculations, unveil a folding-assisted unbinding mechanism.


Subject(s)
Molecular Dynamics Simulation , Protein Binding , Protein Folding , beta Catenin , beta Catenin/metabolism , beta Catenin/chemistry , Humans , Thermodynamics , TCF Transcription Factors/metabolism , TCF Transcription Factors/chemistry
2.
Biophys Chem ; 307: 107197, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38335808

ABSTRACT

BCL9 is a key protein in Wnt signaling pathway. It acts as a transcriptional co-activator to ß-catenin, and dysregulation in this pathway leads to tumor growth. Inhibiting such a protein-protein interaction is considered as a therapeutic challenge. The interaction between ß-catenin and BCL9 is facilitated by a 23-residue helical domain from BCL9 and a hydrophobic groove of ß-catenin. To prevent this interaction, a peptide that mimics the alpha-helical domain of BCL9 can be designed. Stapling is considered a successful strategy in the pursuit of designing such peptides in which amino acids side are stitched together using chemical moieties. Among the various types of cross-linkers, triazole is the most rapid and effective one synthesized via click reaction. However, the underlying interactions behind maintaining the secondary structure of stapled peptides remain less explored. In the current work, we employed the molecular dynamics simulation to study the conformational behavior of the experimentally synthesized single and double triazole stapled BCL9 peptide. Upon the addition of a triazole staple, there is a significant reduction in the conformational space of BCL9. The helical character of the stapled peptide increases with an increase in separation between the triazole cross-linkers. Also, we encompassed the Replica Exchange with Solute Tempering (REST2) simulation to validate the high-temperature response of the stapled peptide. From REST2, the PCA and t-SNE show the reduction in distinct cluster formation on the addition of triazole staple. Our study infers further development of these triazole-stapled BCL9 peptides into effective inhibitors to target the interaction between ß-catenin and BCL9.


Subject(s)
Triazoles , beta Catenin , beta Catenin/chemistry , beta Catenin/metabolism , Triazoles/pharmacology , Neoplasm Proteins/chemistry , Neoplasm Proteins/metabolism , Transcription Factors/metabolism , Peptides/chemistry , Protein Structure, Secondary
3.
J Virol ; 97(10): e0124123, 2023 10 31.
Article in English | MEDLINE | ID: mdl-37772824

ABSTRACT

IMPORTANCE: CD34+ hematopoietic progenitor cells (HPCs) are an important cellular reservoir for latent human cytomegalovirus (HCMV). Several HCMV genes are expressed during latency that are involved with the maintenance of the viral genome in CD34+ HPC. However, little is known about the process of viral reactivation in these cells. Here, we describe a viral protein, pUL8, and its interaction and stabilization with members of the Wnt/ß-catenin pathway as an important component of viral reactivation. We further define that pUL8 and ß-catenin interact with DVL2 via a PDZ-binding domain, and loss of UL8 interaction with ß-catenin-DVL2 restricts viral reactivation. Our findings will be instrumental in understanding the molecular processes involved in HCMV reactivation in order to design new antiviral therapeutics.


Subject(s)
Antigens, CD34 , Cytomegalovirus , Dishevelled Proteins , Hematopoietic Stem Cells , Viral Proteins , Virus Activation , beta Catenin , Humans , Antigens, CD34/metabolism , beta Catenin/chemistry , beta Catenin/metabolism , Cytomegalovirus/genetics , Cytomegalovirus/physiology , Dishevelled Proteins/chemistry , Dishevelled Proteins/metabolism , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/virology , PDZ Domains , Viral Proteins/chemistry , Viral Proteins/metabolism , Virus Latency/genetics
4.
Acta Pharmacol Sin ; 44(5): 1038-1050, 2023 May.
Article in English | MEDLINE | ID: mdl-36470978

ABSTRACT

Renal interstitial fibrosis is the common pathological process of various chronic kidney diseases to end-stage renal disease. Inhibition of fibroblast activation attenuates renal interstitial fibrosis. Our previous studies show that poricoic acid A (PAA) isolated from Poria cocos is a potent anti-fibrotic agent. In the present study we investigated the effects of PAA on renal fibroblast activation and interstitial fibrosis and the underlying mechanisms. Renal interstitial fibrosis was induced in rats or mice by unilateral ureteral obstruction (UUO). UUO rats were administered PAA (10 mg·kg-1·d-1, i.g.) for 1 or 2 weeks. An in vitro model of renal fibrosis was established in normal renal kidney fibroblasts (NRK-49F cells) treated with TGF-ß1. We showed that PAA treatment rescued Sirt3 expression, and significantly attenuated renal fibroblast activation and interstitial fibrosis in both the in vivo and in vitro models. In TGF-ß1-treated NRK-49F cells, we demonstrated that Sirt3 deacetylated ß-catenin (a key transcription factor of fibroblast activation) and then accelerated its ubiquitin-dependent degradation, thus suppressing the protein expression and promoter activity of pro-fibrotic downstream target genes (twist, snail1, MMP-7 and PAI-1) to alleviate fibroblast activation; the lysine-49 (K49) of ß-catenin was responsible for Sirt3-mediated ß-catenin deacetylation. In molecular docking analysis, we found the potential interaction of Sirt3 and PAA. In both in vivo and in vitro models, pharmacological activation of Sirt3 by PAA significantly suppressed renal fibroblast activation via facilitating ß-catenin K49 deacetylation. In UUO mice and NRK-49F cells, Sirt3 overexpression enhanced the anti-fibrotic effect of PAA, whereas Sirt3 knockdown weakened the effect. Taken together, PAA attenuates renal fibroblast activation and interstitial fibrosis by upregulating Sirt3 and inducing ß-catenin K49 deacetylation, highlighting Sirt3 functions as a promising therapeutic target of renal fibroblast activation and interstitial fibrosis.


Subject(s)
Kidney Diseases , Sirtuin 3 , Triterpenes , beta Catenin , Animals , Mice , Rats , beta Catenin/chemistry , beta Catenin/metabolism , Fibroblasts , Fibrosis/drug therapy , Fibrosis/pathology , Kidney/pathology , Kidney Diseases/drug therapy , Kidney Diseases/pathology , Molecular Docking Simulation , Signal Transduction , Sirtuin 3/drug effects , Sirtuin 3/metabolism , Transforming Growth Factor beta1/metabolism , Ureteral Obstruction/complications , Ureteral Obstruction/drug therapy , Ureteral Obstruction/metabolism , Triterpenes/pharmacology , Triterpenes/therapeutic use
5.
Bioorg Chem ; 130: 106234, 2023 01.
Article in English | MEDLINE | ID: mdl-36375353

ABSTRACT

Ras protein has been considered a fascinating target for anticancer therapy because its malfunction is closely related to cancer. However, Ras has been considered undruggable because of the failure to regulate its malfunction by controlling the Ras activation mechanism. Recently, Lumakras targeting the G12C mutation was approved, and therapeutic interest in Ras for anticancer therapy has been rejuvenated. Here, we present a series of compounds that inhibit Ras via a unique mechanism of action that exploits the relationship between the Wnt/ß-catenin pathway and Ras. KYA1797K (1) binds to axin to stabilize the ß-catenin destruction complex that causes the phosphorylation and subsequent degradation of Ras, similar to canonical ß-catenin regulation. Based on the chemical structure of 1, we performed a structural optimization and identified 3-(2-hydroxyethyl)-5-((6-(4-nitrophenyl)pyridin-2-yl)methylene)thiazolidine-2,4-dione (13d) as the most potent compound. 13d displayed antitumor effects in a colorectal cancer model with enhanced inhibition activity on Ras. The results of this study suggest that the further development of 13d could contribute to the development of Ras inhibitors with novel mechanisms of action.


Subject(s)
Colorectal Neoplasms , beta Catenin , ras Proteins , Humans , Axin Protein/chemistry , Axin Protein/genetics , Axin Protein/metabolism , beta Catenin/chemistry , beta Catenin/drug effects , Colorectal Neoplasms/drug therapy , ras Proteins/drug effects , ras Proteins/metabolism , Wnt Signaling Pathway
6.
Development ; 149(3)2022 02 01.
Article in English | MEDLINE | ID: mdl-35156681

ABSTRACT

Axolotls are an important model organism for multiple types of regeneration, including functional spinal cord regeneration. Remarkably, axolotls can repair their spinal cord after a small lesion injury and can also regenerate their entire tail following amputation. Several classical signaling pathways that are used during development are reactivated during regeneration, but how this is regulated remains a mystery. We have previously identified miR-200a as a key factor that promotes successful spinal cord regeneration. Here, using RNA-seq analysis, we discovered that the inhibition of miR-200a results in an upregulation of the classical mesodermal marker brachyury in spinal cord cells after injury. However, these cells still express the neural stem cell marker sox2. In vivo cell tracking allowed us to determine that these cells can give rise to cells of both the neural and mesoderm lineage. Additionally, we found that miR-200a can directly regulate brachyury via a seed sequence in the 3'UTR of the gene. Our data indicate that miR-200a represses mesodermal cell fate after a small lesion injury in the spinal cord when only glial cells and neurons need to be replaced.


Subject(s)
MicroRNAs/metabolism , Spinal Cord Regeneration/genetics , Spinal Cord/metabolism , 3' Untranslated Regions , Ambystoma mexicanum/metabolism , Animals , Antagomirs/metabolism , Cell Differentiation , Fetal Proteins/genetics , Fetal Proteins/metabolism , Mesoderm/cytology , Mesoderm/metabolism , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neuroglia/cytology , Neuroglia/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Spinal Cord/cytology , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology , Stem Cells/cytology , Stem Cells/metabolism , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Tail/physiology , Wnt Signaling Pathway , beta Catenin/antagonists & inhibitors , beta Catenin/chemistry , beta Catenin/metabolism
7.
Bioengineered ; 13(1): 1388-1398, 2022 01.
Article in English | MEDLINE | ID: mdl-35000526

ABSTRACT

Diabetes mellitus (DM) can be implicated in the perturbations of vascular integrity and the dysfunction of angiogenesis. Chitosan has the advantage of promoting the vascular endothelial cell proliferation. However, the molecular mechanism of action in the promotion of wound healing by chitosan derivatives is still debated. In the current study, DM with chronic wound (CW) model rats were prepared and treated with chitosan. Vascular endothelial cells isolated from granulation tissues were conducted by RNA sequencing. Two thousand three hundred and sixteen genes were up-regulated, while 1,864 genes were down-regulated after chitosan treatment compared to CW group. Here, we observed that caveolin 1 (CAV1) was highly expressed induced by chitosan. Furthermore, we observed that CAV1 knockdown could compromise the activation of Wnt pathway by reduction of ß-catenin in rat aortic endothelial cells (RAOECs) and brain endothelium four cells (RBE4s). Moreover, we determined a direct interaction between CAV1 and ß-catenin by IP assay. The C-terminus of CAV1 and ß-catenin (24 to 586 amino acids) contributed to the interaction of these two proteins. Finally, the protein docking analysis indicated that the fragments of ß-catenin (253-261 'FYAITTLHN' and 292-303 'KFLAITTDCLQI') might have affected the structure by CAV1 and facilitated the resistance to degradation. Taken together, our study demonstrates that chitosan can up-regulate CAV1 expression, and CAV1 can interact with ß-catenin for promotion of canonical Wnt signaling pathway activity. Our results deepens the molecular mechanism of the Wnt pathway in vascular endothelial cells and is beneficial to developing new targets to assist in enhancing the pharmacological effect of chitosan on wound healing and angiogenesis against DM.


Subject(s)
Caveolin 1/genetics , Chitosan/administration & dosage , Diabetes Complications/drug therapy , Wnt Signaling Pathway/drug effects , Wound Healing/drug effects , beta Catenin/metabolism , Animals , Binding Sites , Caveolin 1/chemistry , Caveolin 1/metabolism , Cell Line , Chitosan/pharmacology , Diabetes Complications/genetics , Diabetes Complications/metabolism , Disease Models, Animal , Gene Expression Profiling , Gene Expression Regulation/drug effects , Indenes , Male , Molecular Docking Simulation , Protein Binding , Rats , Sequence Analysis, RNA , Sulfonamides , beta Catenin/chemistry , beta Catenin/genetics
8.
Cancer Res Commun ; 2(4): 233-245, 2022 04.
Article in English | MEDLINE | ID: mdl-36873622

ABSTRACT

The catalytic enzymes tankyrase 1 and 2 (TNKS1/2) alter protein turnover by poly-ADP-ribosylating target proteins, which earmark them for degradation by the ubiquitin-proteasomal system. Prominent targets of the catalytic activity of TNKS1/2 include AXIN proteins, resulting in TNKS1/2 being attractive biotargets for addressing of oncogenic WNT/ß-catenin signaling. Although several potent small molecules have been developed to inhibit TNKS1/2, there are currently no TNKS1/2 inhibitors available in clinical practice. The development of tankyrase inhibitors has mainly been disadvantaged by concerns over biotarget-dependent intestinal toxicity and a deficient therapeutic window. Here we show that the novel, potent, and selective 1,2,4-triazole-based TNKS1/2 inhibitor OM-153 reduces WNT/ß-catenin signaling and tumor progression in COLO 320DM colon carcinoma xenografts upon oral administration of 0.33-10 mg/kg twice daily. In addition, OM-153 potentiates anti-programmed cell death protein 1 (anti-PD-1) immune checkpoint inhibition and antitumor effect in a B16-F10 mouse melanoma model. A 28-day repeated dose mouse toxicity study documents body weight loss, intestinal damage, and tubular damage in the kidney after oral-twice daily administration of 100 mg/kg. In contrast, mice treated oral-twice daily with 10 mg/kg show an intact intestinal architecture and no atypical histopathologic changes in other organs. In addition, clinical biochemistry and hematologic analyses do not identify changes indicating substantial toxicity. The results demonstrate OM-153-mediated antitumor effects and a therapeutic window in a colon carcinoma mouse model ranging from 0.33 to at least 10 mg/kg, and provide a framework for using OM-153 for further preclinical evaluations. Significance: This study uncovers the effectiveness and therapeutic window for a novel tankyrase inhibitor in mouse tumor models.


Subject(s)
Carcinoma , Colonic Neoplasms , Tankyrases , Humans , Mice , Animals , beta Catenin/chemistry , Colonic Neoplasms/drug therapy , Wnt Signaling Pathway
9.
mBio ; 12(6): e0279221, 2021 12 21.
Article in English | MEDLINE | ID: mdl-34749529

ABSTRACT

Under pathological conditions like herpes simplex virus 1 (HSV-1) infection, host-pathogen interactions lead to major reconstruction of the host protein network, which contributes to the dysregulation of signaling pathways and disease onset. Of note is the upregulation of a multifunctional host protein, heparanase (HPSE), following infection, which serves as a mediator in HSV-1 replication. In this study, we identify a novel function of HPSE and highlight it as a key regulator of ß-catenin signal transduction. The regulatory role of HPSE on the activation, nuclear translocation, and signal transduction of ß-catenin disrupts cellular homeostasis and establishes a pathogenic environment that promotes viral replication. Under normal physiological conditions, ß-catenin is bound to a group of proteins, referred to as the destruction complex, and targeted for ubiquitination and, ultimately, degradation. We show that virus-induced upregulation of HPSE leads to the activation of Akt and subsequent stabilization and activation of ß-catenin through (i) the release of ß-catenin from the destruction complex, and (ii) direct phosphorylation of ß-catenin at Ser552. This study also provides an in-depth characterization of the proviral role of ß-catenin signaling during HSV-1 replication using physiologically relevant cell lines and in vivo models of ocular infection. Furthermore, pharmacological inhibitors of this pathway generated a robust antiviral state against multiple laboratory and clinical strains of HSV-1. Collectively, our findings assign a novel regulatory role to HPSE as a driver of ß-catenin signaling in HSV-1 infection. IMPORTANCE Heparanase (HPSE) and ß-catenin have independently been implicated in regulating key pathophysiological processes, including neovascularization, angiogenesis, and inflammation; however, the relationship between the two proteins has remained elusive thus far. For that reason, characterizing this relationship is crucial and can lead to the development of novel therapeutics. For HSV-1 specifically, current antivirals are not able to abolish the virus from the host, leaving patients susceptible to episodes of viral reactivation. Identifying a host-based intervention can provide a better alternative with enhanced efficacy and sustained relief.


Subject(s)
Glucuronidase/metabolism , Herpes Simplex/enzymology , Herpesvirus 1, Human/physiology , Proto-Oncogene Proteins c-akt/metabolism , beta Catenin/metabolism , Amino Acid Motifs , Cell Line , Glucuronidase/genetics , Herpes Simplex/genetics , Herpes Simplex/metabolism , Herpes Simplex/virology , Herpesvirus 1, Human/genetics , Host-Pathogen Interactions , Humans , Phosphorylation , Proto-Oncogene Proteins c-akt/genetics , Virus Activation , Virus Replication , Wnt Signaling Pathway , beta Catenin/chemistry , beta Catenin/genetics
10.
J Biol Chem ; 297(5): 101305, 2021 11.
Article in English | MEDLINE | ID: mdl-34656562

ABSTRACT

CEACAM1-LF, a homotypic cell adhesion adhesion molecule, transduces intracellular signals via a 72 amino acid cytoplasmic domain that contains two immunoreceptor tyrosine-based inhibitory motifs (ITIMs) and a binding site for ß-catenin. Phosphorylation of Ser503 by PKC in rodent CEACAM1 was shown to affect bile acid transport or hepatosteatosis via the level of ITIM phosphorylation, but the phosphorylation of the equivalent residue in human CEACAM1 (Ser508) was unclear. Here we studied this analogous phosphorylation by NMR analysis of the 15N labeled cytoplasmic domain peptide. Incubation with a variety of Ser/Thr kinases revealed phosphorylation of Ser508 by GSK3bß but not by PKC. The lack of phosphorylation by PKC is likely due to evolutionary sequence changes between the rodent and human genes. Phosphorylation site assignment by mass spectrometry and NMR revealed phosphorylation of Ser472, Ser461 and Ser512 by PKA, of which Ser512 is part of a conserved consensus site for GSK3ß binding. We showed here that only after phosphorylation of Ser512 by PKA was GSK3ß able to phosphorylate Ser508. Phosphorylation of Ser512 by PKA promoted a tight association with the armadillo repeat domain of ß-catenin at an extended region spanning the ITIMs of CEACAM1. The kinetics of phosphorylation of the ITIMs by Src, as well dephosphorylation by SHP2, were affected by the presence of Ser508/512 phosphorylation, suggesting that PKA and GSK3ß may regulate the signal transduction activity of human CEACAM1-LF. The interaction of CEACAM1-LF with ß-catenin promoted by PKA is suggestive of a tight association between the two ITIMs of CEACAM1-LF.


Subject(s)
Antigens, CD/chemistry , Cell Adhesion Molecules/chemistry , Cyclic AMP-Dependent Protein Kinases/chemistry , Glycogen Synthase Kinase 3 beta/chemistry , beta Catenin/chemistry , Antigens, CD/genetics , Antigens, CD/metabolism , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Glycogen Synthase Kinase 3 beta/genetics , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Protein Binding , beta Catenin/genetics , beta Catenin/metabolism
11.
Proc Natl Acad Sci U S A ; 118(37)2021 09 14.
Article in English | MEDLINE | ID: mdl-34504002

ABSTRACT

Intrinsically disordered proteins often form dynamic complexes with their ligands. Yet, the speed and amplitude of these motions are hidden in classical binding kinetics. Here, we directly measure the dynamics in an exceptionally mobile, high-affinity complex. We show that the disordered tail of the cell adhesion protein E-cadherin dynamically samples a large surface area of the protooncogene ß-catenin. Single-molecule experiments and molecular simulations resolve these motions with high resolution in space and time. Contacts break and form within hundreds of microseconds without a dissociation of the complex. The energy landscape of this complex is rugged with many small barriers (3 to 4 kBT) and reconciles specificity, high affinity, and extreme disorder. A few persistent contacts provide specificity, whereas unspecific interactions boost affinity.


Subject(s)
Antigens, CD/chemistry , Cadherins/chemistry , Intrinsically Disordered Proteins/chemistry , Protein Folding , beta Catenin/chemistry , Antigens, CD/metabolism , Cadherins/metabolism , Diffusion , Humans , Intrinsically Disordered Proteins/metabolism , Kinetics , Ligands , Molecular Dynamics Simulation , Protein Conformation , beta Catenin/metabolism
12.
Fish Shellfish Immunol ; 118: 228-240, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34284111

ABSTRACT

ß-catenin is a structural protein that makes the cell-cell connection in adherence junctions. Besides the structural functions, it also plays a role as a central transducer of the canonical Wnt signaling cascade, regulating nearly four hundred genes related to various cellular processes. Recently the immune functions of ß-catenin during pathogenic invasion have gained more attention. In the present study, we elucidated the immune function of fish ß-catenin by identifying and characterizing the ß-catenin homolog (PhCatß) from redlip mullet, Planiliza haematocheila. The complete open reading frame of PhCatß consists of 2352 bp, which encodes a putative ß-catenin homolog (molecular weight: 85.7 kDa). Multiple sequence alignment analysis revealed that ß-catenin is highly conserved in vertebrates. Phylogenetic reconstruction demonstrated the close evolutionary relationship between PhCatß and other fish ß-catenin counterparts. The tissue distribution analysis showed the highest mRNA expression of PhCatß in heart tissues of the redlip mullet under normal physiological conditions. While in response to pathogenic stress, the PhCatß transcription level was dramatically increased in the spleen and gill tissues. The overexpression of PhCatß stimulated M2 polarization and cell proliferation of murine RAW 264.7 macrophage. In fish cells, the overexpression of PhCatß resulted in a significant upregulation of antiviral gene transcription and vice versa. Moreover, the overexpression of PhCatß could inhibit the replication of VHSV in FHM cells. Our results strongly suggest that PhCatß plays a role in macrophage activation and antiviral immune response in redlip mullet.


Subject(s)
Antiviral Agents , Cytosol , Fish Proteins , Macrophage Activation , Smegmamorpha , beta Catenin , Animals , Antiviral Agents/chemistry , Antiviral Agents/immunology , Antiviral Agents/metabolism , Evolution, Molecular , Fish Proteins/chemistry , Fish Proteins/genetics , Fish Proteins/immunology , Fish Proteins/metabolism , Gene Expression Profiling , Macrophage Activation/drug effects , Macrophages/drug effects , Mice , Organ Specificity , Phylogeny , RAW 264.7 Cells , Smegmamorpha/classification , Smegmamorpha/genetics , beta Catenin/chemistry , beta Catenin/genetics , beta Catenin/immunology , beta Catenin/metabolism
13.
Biomolecules ; 11(6)2021 06 01.
Article in English | MEDLINE | ID: mdl-34206070

ABSTRACT

The Wnt signalling pathway plays an important role in cell proliferation, differentiation, and fate decisions in embryonic development and the maintenance of adult tissues. The twelve armadillo (ARM) repeat-containing protein ß-catenin acts as the signal transducer in this pathway. Here, we investigated the interaction between ß-catenin and the intrinsically disordered transcription factor TCF7L2, comprising a very long nanomolar-affinity interface of approximately 4800 Å2 that spans ten of the twelve ARM repeats of ß-catenin. First, a fluorescence reporter system for the interaction was engineered and used to determine the kinetic rate constants for the association and dissociation. The association kinetics of TCF7L2 and ß-catenin were monophasic and rapid (7.3 ± 0.1 × 107 M-1·s-1), whereas dissociation was biphasic and slow (5.7 ± 0.4 × 10-4 s-1, 15.2 ± 2.8 × 10-4 s-1). This reporter system was then combined with site-directed mutagenesis to investigate the striking variability in the conformation adopted by TCF7L2 in the three different crystal structures of the TCF7L2-ß-catenin complex. We found that the mutation had very little effect on the association kinetics, indicating that most interactions form after the rate-limiting barrier for association. Mutations of the N- and C-terminal subdomains of TCF7L2 that adopt relatively fixed conformations in the crystal structures had large effects on the dissociation kinetics, whereas the mutation of the labile sub-domain connecting them had negligible effect. These results point to a two-site avidity mechanism of binding with the linker region forming a "fuzzy" complex involving transient contacts that are not site-specific. Strikingly, the two mutations in the N-terminal subdomain that had the largest effects on the dissociation kinetics showed two additional phases, indicating partial flux through an alternative dissociation pathway that is inaccessible to the wild type. The results presented here provide insights into the kinetics of the molecular recognition of a long intrinsically disordered region with an elongated repeat-protein surface, a process found to involve parallel routes with sequential steps in each.


Subject(s)
Intrinsically Disordered Proteins/chemistry , Transcription Factor 7-Like 2 Protein/chemistry , beta Catenin/chemistry , Crystallography, X-Ray , Humans , Intrinsically Disordered Proteins/genetics , Mutagenesis, Site-Directed , Protein Structure, Quaternary , Transcription Factor 7-Like 2 Protein/genetics , beta Catenin/genetics
14.
Chem Commun (Camb) ; 57(46): 5658-5661, 2021 Jun 08.
Article in English | MEDLINE | ID: mdl-33972980

ABSTRACT

In this report, we revise the structure for a previously reported synthetic product proposed to be the 1R,2S-cannabidiol epoxide and reassign it as cannabielsoin using anisotropic NMR and synthetic chemistry methods. These results provide a direct link to the first known biological target and function of cannabielsoin.


Subject(s)
Cannabidiol/analogs & derivatives , Wnt Proteins/chemistry , beta Catenin/chemistry , Anisotropy , Cannabidiol/analysis , Magnetic Resonance Spectroscopy , Molecular Conformation
15.
J Med Chem ; 64(8): 4257-4288, 2021 04 22.
Article in English | MEDLINE | ID: mdl-33822624

ABSTRACT

Canonical WNT signaling is an important developmental pathway that has attracted increased attention for anticancer drug discovery. From the production and secretion of WNT ligands, their binding to membrane receptors, and the ß-catenin destruction complex to the expansive ß-catenin transcriptional complex, multiple components have been investigated as drug targets to modulate WNT signaling. Significant progress in developing WNT inhibitors such as porcupine inhibitors, tankyrase inhibitors, ß-catenin/coactivators, protein-protein interaction inhibitors, casein kinase modulators, DVL inhibitors, and dCTPP1 inhibitors has been made, with several candidates (e.g., LGK-974, PRI-724, and ETC-159) in human clinical trials. Herein we summarize recent progress in the drug discovery and development of small-molecule inhibitors targeting the canonical WNT pathway, focusing on their specific target proteins, in vitro and in vivo activities, physicochemical properties, and therapeutic potential. The relevant opportunities and challenges toward maintaining the balance between efficacy and toxicity in effectively targeting this pathway are also highlighted.


Subject(s)
Small Molecule Libraries/pharmacology , Wnt Proteins/metabolism , Wnt Signaling Pathway/drug effects , Animals , Binding Sites , Drug Discovery , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Humans , Molecular Dynamics Simulation , Neoplasms/drug therapy , Peptides/chemistry , Peptides/metabolism , Protein Interaction Maps/drug effects , Small Molecule Libraries/chemistry , Small Molecule Libraries/metabolism , Small Molecule Libraries/therapeutic use , TCF Transcription Factors/chemistry , TCF Transcription Factors/metabolism , Tankyrases/antagonists & inhibitors , Tankyrases/metabolism , Wnt Proteins/chemistry , beta Catenin/chemistry , beta Catenin/metabolism
16.
Nat Commun ; 12(1): 1368, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33649334

ABSTRACT

The homeostasis of the gut epithelium relies upon continuous renewal and proliferation of crypt-resident intestinal epithelial stem cells (IESCs). Wnt/ß-catenin signaling is required for IESC maintenance, however, it remains unclear how this pathway selectively governs the identity and proliferative decisions of IESCs. Here, we took advantage of knock-in mice harboring transgenic ß-catenin alleles with mutations that specifically impair the recruitment of N- or C-terminal transcriptional co-factors. We show that C-terminally-recruited transcriptional co-factors of ß-catenin act as all-or-nothing regulators of Wnt-target gene expression. Blocking their interactions with ß-catenin rapidly induces loss of IESCs and intestinal homeostasis. Conversely, N-terminally recruited co-factors fine-tune ß-catenin's transcriptional output to ensure proper self-renewal and proliferative behaviour of IESCs. Impairment of N-terminal interactions triggers transient hyperproliferation of IESCs, eventually resulting in exhaustion of the self-renewing stem cell pool. IESC mis-differentiation, accompanied by unfolded protein response stress and immune infiltration, results in a process resembling aberrant "villisation" of intestinal crypts. Our data suggest that IESC-specific Wnt/ß-catenin output requires selective modulation of gene expression by transcriptional co-factors.


Subject(s)
Intestinal Mucosa/cytology , Stem Cells/metabolism , Transcription Factors/metabolism , Transcription, Genetic , beta Catenin/chemistry , beta Catenin/metabolism , Algorithms , Animals , Base Sequence , Cell Differentiation , Cell Proliferation , Chromatin/metabolism , Chromatin Assembly and Disassembly , Homeostasis , Hyperplasia , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Mutant Proteins/metabolism , Mutation/genetics , Organoids/metabolism , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction
17.
Arch Biochem Biophys ; 698: 108722, 2021 02 15.
Article in English | MEDLINE | ID: mdl-33321112

ABSTRACT

ß-Catenin, a key transcriptional factor involved in the canonical Wnt signaling pathway, is regulated by a cascade of phosphorylations and plays a major role in the progression of triple-negative breast cancer (TNBC). However, the phosphorylation induced conformational changes in a ß-Catenin is still poorly understood. Hence, we adopted a conventional molecular dynamics approach to study phosphorylations present in a sequence motif Ser 552 675 and Tyr670 of the ß-Catenin domain and analyzed in terms of structural transitions, bond formation, and folding-misfolding conformations. Our results unveil the ß-Catenin linear motif 549-555 (RRTSMGG) of armadillo repeats domain prefers order to disorder state. In contrast, helix C associated with 670-678 (YKKRLSVEL) motif prefers disorder to order upon phosphorylation of Ser 552 675 and Tyr670. In addition, the crucial secondary structural transition from α-helix to coil induced by phospho Ser552 and phospho Tyr670 of ß-Catenin ARM domain connecting helix C modifies conformational diversity and binding affinities of the complex interaction in functional regulation significantly. Moreover, the post phosphorylation disrupted the hydrogen bond interactions (Ser552-Arg549, Arg550-Asp546 and Ser675-Lys672) and abolished the residual alliance with hydrophobic interactions (Tyr670-Leu674) that easily interrupt in secondary structure packing as well as folding conformations connecting ARM and helix C (R10, 12 & R1C) compared to unphosphorylation. Our integrated computational analysis may help in shedding light on understanding the induced folding and unfolding pattern due to motif phosphorylations. Overall, our results provide an atomistic structural description of the way phosphorylation facilitates conformational and dynamic changes in ß-Catenin, a fundamental molecular switch mechanism in triple-negative breast cancer pathogenesis.


Subject(s)
Protein Processing, Post-Translational , beta Catenin/metabolism , Humans , Molecular Dynamics Simulation , Phosphorylation , Protein Conformation , Protein Domains , Serine/chemistry , Tyrosine/chemistry , beta Catenin/chemistry
18.
FEBS J ; 288(10): 3261-3284, 2021 05.
Article in English | MEDLINE | ID: mdl-33284517

ABSTRACT

The transcription factor forkhead box protein P2 (FOXP2) is a highly conserved key regulator of embryonal development. The molecular mechanisms of how FOXP2 regulates embryonal development, however, remain elusive. Using RNA sequencing, we identified the Wnt signaling pathway as key target of FOXP2-dependent transcriptional regulation. Using cell-based assays, we show that FOXP2 transcriptional activity is regulated by the Wnt coregulator ß-catenin and that ß-catenin contacts multiple regions within FOXP2. Using nuclear magnetic resonance spectroscopy, we uncovered the molecular details of these interactions. ß-catenin contacts a disordered FOXP2 region with α-helical propensity via its folded armadillo domain, whereas the intrinsically disordered ß-catenin N terminus and C terminus bind to the conserved FOXP2 DNA-binding domain. Using RNA sequencing, we confirmed that ß-catenin indeed regulates transcriptional activity of FOXP2 and that the FOXP2 α-helical motif acts as a key regulatory element of FOXP2 transcriptional activity. Taken together, our findings provide first insight into novel regulatory interactions and help to understand the intricate mechanisms of FOXP2 function and (mis)-regulation in embryonal development and human diseases. DATABASE: Expression data are available in the GEO database under the accession number GSE138938.


Subject(s)
Forkhead Transcription Factors/chemistry , Gene Expression Regulation, Developmental , Transcription, Genetic , Wnt Signaling Pathway/genetics , beta Catenin/chemistry , Amino Acid Sequence , Animals , Binding Sites , Cell Line, Tumor , Cloning, Molecular , Embryo, Mammalian , Escherichia coli/genetics , Escherichia coli/metabolism , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Profiling , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Humans , Models, Molecular , Osteoblasts/cytology , Osteoblasts/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , beta Catenin/genetics , beta Catenin/metabolism
19.
Nat Commun ; 11(1): 5762, 2020 11 13.
Article in English | MEDLINE | ID: mdl-33188184

ABSTRACT

Occurrence of Colorectal cancer (CRC) is relevant with gut microbiota. However, role of IRF3, a key signaling mediator in innate immune sensing, has been barely investigated in CRC. Here, we unexpectedly found that the IRF3 deficient mice are hyper-susceptible to the development of intestinal tumor in AOM/DSS and Apcmin/+ models. Genetic ablation of IRF3 profoundly promotes the proliferation of intestinal epithelial cells via aberrantly activating Wnt signaling. Mechanically, IRF3 in resting state robustly associates with the active ß-catenin in the cytoplasm, thus preventing its nuclear translocation and cell proliferation, which can be relieved upon microbe-induced activation of IRF3. In accordance, the survival of CRC is clinically correlated with the expression level of IRF3. Therefore, our study identifies IRF3 as a negative regulator of the Wnt/ß-catenin pathway and a potential prognosis marker for Wnt-related tumorigenesis, and describes an intriguing link between gut microbiota and CRC via the IRF3-ß-catenin axis.


Subject(s)
Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Nucleus/metabolism , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/prevention & control , Interferon Regulatory Factor-3/metabolism , beta Catenin/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Enterocytes/metabolism , Enterocytes/pathology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Ki-67 Antigen/metabolism , Mice, Inbred C57BL , Protein Binding , Protein Domains , Protein Transport , Survival Analysis , Wnt Signaling Pathway , beta Catenin/chemistry
20.
Chem Biodivers ; 17(11): e2000654, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33016000

ABSTRACT

Colorectal cancer (CRC) is one of the most common malignant tumors in the world. In this study, the Caco-2 in vitro cell model was used to study the effect and mechanism of Aronia melanocarpa (Michx.) Elliott anthocyanins (AMA) on colon cancer. The experimental results showed that the binding energy of anthocyanins on ß-catenin was in the range of -5.92 to 4.95 kcal/mol, with good low energy parameters and binding positions. AMA can inhibit cell proliferation and cause cell cycle arrest. RT-PCR and Western blot results showed that AMA can reduce cytoplasmic ß-catenin and inhibit the expression of related proteins in Wnt/ß-catenin signaling pathway. This study revealed the AMA inhibitory effect and mechanism of malignant biological behavior of Caco-2 cells, in order to provide theoretical basis for the prevention and treatment of colon cancer by Aronia melanocarpa (Michx.) Elliott.


Subject(s)
Anthocyanins/chemistry , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Photinia/chemistry , Wnt Signaling Pathway/drug effects , Anthocyanins/isolation & purification , Anthocyanins/metabolism , Anthocyanins/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/isolation & purification , Antineoplastic Agents/metabolism , Binding Sites , Caco-2 Cells , Cell Cycle Checkpoints/drug effects , Cell Survival/drug effects , Down-Regulation/drug effects , Fruit/chemistry , Fruit/metabolism , Humans , Molecular Docking Simulation , Photinia/metabolism , Superoxide Dismutase/metabolism , Survivin/genetics , Survivin/metabolism , beta Catenin/chemistry , beta Catenin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...