Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 51
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
Cell Res ; 34(9): 648-660, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39103523

RÉSUMÉ

While lysine methylation is well-known for regulating gene expression transcriptionally, its implications in translation have been largely uncharted. Trimethylation at lysine 22 (K22me3) on RPL40, a core ribosomal protein located in the GTPase activation center, was first reported 27 years ago. Yet, its methyltransferase and role in translation remain unexplored. Here, we report that SMYD5 has robust in vitro activity toward RPL40 K22 and primarily catalyzes RPL40 K22me3 in cells. The loss of SMYD5 and RPL40 K22me3 leads to reduced translation output and disturbed elongation as evidenced by increased ribosome collisions. SMYD5 and RPL40 K22me3 are upregulated in hepatocellular carcinoma (HCC) and negatively correlated with patient prognosis. Depleting SMYD5 renders HCC cells hypersensitive to mTOR inhibition in both 2D and 3D cultures. Additionally, the loss of SMYD5 markedly inhibits HCC development and growth in both genetically engineered mouse and patient-derived xenograft (PDX) models, with the inhibitory effect in the PDX model further enhanced by concurrent mTOR suppression. Our findings reveal a novel role of the SMYD5 and RPL40 K22me3 axis in translation elongation and highlight the therapeutic potential of targeting SMYD5 in HCC, particularly with concurrent mTOR inhibition. This work also conceptually broadens the understanding of lysine methylation, extending its significance from transcriptional regulation to translational control.


Sujet(s)
Carcinome hépatocellulaire , Histone-lysine N-methyltransferase , Tumeurs du foie , Lysine , Methyltransferases , Protéines ribosomiques , Animaux , Humains , Souris , Carcinome hépatocellulaire/métabolisme , Carcinome hépatocellulaire/anatomopathologie , Carcinome hépatocellulaire/génétique , Lignée cellulaire tumorale , Histone-lysine N-methyltransferase/métabolisme , Histone-lysine N-methyltransferase/génétique , Tumeurs du foie/métabolisme , Tumeurs du foie/anatomopathologie , Tumeurs du foie/génétique , Lysine/métabolisme , Méthylation , Souris nude , Biosynthèse des protéines , Protéines ribosomiques/métabolisme , Protéines ribosomiques/génétique , Methyltransferases/génétique , Methyltransferases/métabolisme
2.
Public Health Nurs ; 41(3): 476-486, 2024.
Article de Anglais | MEDLINE | ID: mdl-38468509

RÉSUMÉ

BACKGROUND: Hepatocellular carcinoma (HCC) is the sixth most common cancer worldwide and the third leading cause of cancer mortality. HCC has high morbidity, high mortality, and low survival rates. Screening is one of the most significant methods of lowering incidence and death while also increasing survival. OBJECTIVES: The aim of this study was to identify the facilitators and barriers to participation in HCC screening among high-risk populations. METHODS: A comprehensive and systematic search was undertaken in PubMed, Web of Science, MEDLINE, EMBACE, EBSCOhost and the Cochrane Library. A combination of synonyms of the keywords including HCC, screening, factors and adherence were used for searching. Studies addressing the facilitators and barriers to HCC screening compliance in at-risk individuals were included. Data were synthesized using Review Manager version 5.4. A random/fixed effects model meta-analysis was performed to estimate the pooled data and expressed with odds ratio (OR) and 95% confidence interval (CI). RESULTS: A total of seven articles met the inclusion criteria. Qualitative (n = 1) and quantitative (n = 6) studies using various types of surgery were conducted. The most commonly mentioned barriers were insufficient knowledge and awareness of HCC screening, unawareness of the necessity for early detection of HCC and lack of physician recommendation. A meta-analysis of seven studies showed that individuals with a family history of HCC increased screening uptake by nearly three times (OR: 2.69, 95% CI: 1.93, 3.75). Other most frequently reported facilitators include age, education level, and perceived risk et al. CONCLUSIONS: Many barriers to HCC screening were found. Meanwhile, this review points out that improving the awareness of high-risk populations toward HCC screening is expected to enhance compliance, thereby promoting early diagnosis of liver cancer, reducing mortality, and alleviating the burden of HCC.


Sujet(s)
Carcinome hépatocellulaire , Dépistage précoce du cancer , Tumeurs du foie , Humains , Carcinome hépatocellulaire/diagnostic , Tumeurs du foie/diagnostic , Observance par le patient/statistiques et données numériques , Dépistage de masse/méthodes , Facteurs de risque
3.
Bioorg Chem ; 139: 106726, 2023 10.
Article de Anglais | MEDLINE | ID: mdl-37451145

RÉSUMÉ

O-GlcNAcylation is a specific type of post-translational glycosylation modification, which is regulated by two enzymes, O-GlcNAc transferase (OGT) and O-GlcNAcase (OGA). Aberrant overexpression of OGT is associated with the development of many solid tumors. In this study, we have developed and optimized a sensitive Homogeneous Time-Resolved Fluorescence (HTRF) assay then identified a novel OGT inhibitor CDDO (also called Bardoxolone) through a high-throughput screening (HTS) based on HTRF assay. Further characterization suggested that CDDO is an effective OGT inhibitor with an IC50 value of 6.56 ± 1.69 µM. CPMG-NMR analysis confirmed that CDDO is a direct binder of OGT with a binding affinity (Kd) of approximately 1.7 µM determined by the MST analysis. Moreover, HDX-MS analysis indicated that CDDO binds to the TPR domain and N-Terminal domain of OGT, which was further confirmed by the enzymatic competition experiments as the binding of CDDO to OGT was not affected by the catalytic site binding inhibitor OSMI-4. Our docking modeling analysis further predicted the possible interactions between CDDO and OGT, providing informative molecular basis for further optimization of the inhibitor in the future. Together, our results suggested CDDO is a new inhibitor of OGT with a distinct binding pocket from the reported OGT inhibitors. Our work paved a new direction for developing OGT inhibitors driven by novel mechanisms.


Sujet(s)
Tests de criblage à haut débit , Maturation post-traductionnelle des protéines , Glycosylation
4.
Bioorg Med Chem ; 84: 117262, 2023 04 15.
Article de Anglais | MEDLINE | ID: mdl-37018878

RÉSUMÉ

Autophagy related 4B (ATG4B) which regulates autophagy by promoting the formation of autophagosome through reversible modification of LC3, is closely related to cancer cell growth and drug resistance, and therefore is an attractive therapeutic target. Recently, ATG4B inhibitors have been reported, yet with drawbacks including weak potency. To discover more promising ATG4B inhibitors, we developed a high-throughput screening (HTS) assay and identified a new ATG4B inhibitor named DC-ATG4in. DC-ATG4in directly binds to ATG4B and inhibits its enzyme activity with an IC50 of 3.08 ± 0.47 µM. We further confirmed that DC-ATG4in is an autophagy inhibitor and blocks autophagy induced by Sorafenib in Hepatocellular Carcinoma (HCC) cells. More importantly, combination of DC-ATG4in with Sorafenib synergized the cancer cell killing effect and proliferation inhibition activities on HCC cells. Our data suggested that inactivation of autophagy via ATG4B inhibition may be a viable strategy to sensitize existing targeted therapy such as Sorafenib in the future.


Sujet(s)
Protéines associées à l'autophagie , Autophagie , Sorafénib , Humains , Autophagie/effets des médicaments et des substances chimiques , Protéines associées à l'autophagie/antagonistes et inhibiteurs , Protéines associées à l'autophagie/métabolisme , Carcinome hépatocellulaire/traitement médicamenteux , Cysteine endopeptidases/métabolisme , Tumeurs du foie/traitement médicamenteux , Sorafénib/pharmacologie , Sorafénib/usage thérapeutique
5.
Adv Cancer Res ; 158: 387-421, 2023.
Article de Anglais | MEDLINE | ID: mdl-36990537

RÉSUMÉ

Oncogenic transcription activation is associated with tumor development and resistance derived from chemotherapy or target therapy. The super elongation complex (SEC) is an important complex regulating gene transcription and expression in metazoans closely related to physiological activities. In normal transcriptional regulation, SEC can trigger promoter escape, limit proteolytic degradation of transcription elongation factors and increase the synthesis of RNA polymerase II (POL II), and regulate many normal human genes to stimulate RNA elongation. Dysregulation of SEC accompanied by multiple transcription factors in cancer promotes rapid transcription of oncogenes and induce cancer development. In this review, we summarized recent progress in understanding the mechanisms of SEC in regulating normal transcription, and importantly its roles in cancer development. We also highlighted the discovery of SEC complex target related inhibitors and their potential applications in cancer treatment.


Sujet(s)
Tumeurs , Facteur B d'élongation transcriptionnelle positive , Humains , Facteur B d'élongation transcriptionnelle positive/génétique , Facteur B d'élongation transcriptionnelle positive/métabolisme , Transcription génétique , Facteurs d'élongation transcriptionnelle/génétique , Facteurs d'élongation transcriptionnelle/métabolisme , RNA polymerase II/génétique , RNA polymerase II/métabolisme , Tumeurs/traitement médicamenteux , Tumeurs/génétique
6.
J Med Chem ; 65(16): 11034-11057, 2022 08 25.
Article de Anglais | MEDLINE | ID: mdl-35925880

RÉSUMÉ

Aberrant hyperactivation of cyclins results in carcinogenesis and therapy resistance in cancers. Direct degradation of the specific cyclin or cyclin-dependent kinase (CDK)-cyclin complex by small-molecule degraders remains a great challenge. Here, we applied the first application of hydrophobic tagging to induce degradation of CDK9-cyclin T1 heterodimer, which is required to keep productive transcription of oncogenes in cancers. LL-K9-3 was identified as a potent small-molecule degrader of CDK9-cyclin T1. Quantitative and time-resolved proteome profiling exhibited LL-K9-3 induced selective and synchronous degradation of CDK9 and cyclin T1. The expressions of androgen receptor (AR) and cMyc were reduced by LL-K9-3 in 22RV1 cells. LL-K9-3 exhibited enhanced anti-proliferative and pro-apoptotic effects compared with its parental CDK9 inhibitor SNS032 and suppressed downstream signaling of CDK9 and AR more effectively than SNS032. Moreover, LL-K9-3 inhibited AR and Myc-driven oncogenic transcriptional programs and exerted stronger inhibitory effects on several intrinsic target genes of AR than the monomeric CDK9 PROTAC (Thal-SNS032).


Sujet(s)
Kinase-9 cycline-dépendante , Tumeurs de la prostate , Noyau de la cellule/métabolisme , Cycline T/génétique , Cycline T/métabolisme , Kinases cyclines-dépendantes/métabolisme , Cyclines/métabolisme , Humains , Mâle
7.
Opt Lett ; 47(10): 2390-2393, 2022 May 15.
Article de Anglais | MEDLINE | ID: mdl-35561358

RÉSUMÉ

This paper studies the use of MUltiple SIgnal Classification (MUSIC) as a super-resolution algorithm to improve demodulation results for intrinsic Fabry-Perot interferometer (IFPI) sensor arrays. Through distinction between noise and signal subspaces in an observation matrix, this paper shows that a 38-fold improvement in the full width at half maximum (FWHM) estimation of IFPI optical path differences (OPD) can be achieved using this algorithm. Based on this improved method, this paper demonstrates that a tunable laser with a 1.3-nm tuning range can achieve the same sensor demodulation performance as a tunable laser with a 50-nm tuning range if a conventional Fourier transform-based algorithm is used. This paper presents a new approach to analyzing optical signals produced by multiple multiplexed interferometers with similar OPDs with potential applications for both single-mode and multiple-mode devices.

8.
J Med Chem ; 65(7): 5317-5333, 2022 04 14.
Article de Anglais | MEDLINE | ID: mdl-35352560

RÉSUMÉ

Polycomb Repressive Complex 2 (PRC2) plays an important role in transcriptional regulation during animal development and in cell differentiation, and alteration of PRC2 activity has been associated with cancer. On a molecular level, PRC2 catalyzes methylation of histone H3 lysine 27 (H3K27), resulting in mono-, di-, or trimethylated forms of H3K27, of which the trimethylated form H3K27me3 leads to transcriptional repression of polycomb target genes. Previously, we have shown that binding of the low-molecular-weight compound EED226 to the H3K27me3 binding pocket of the regulatory subunit EED can effectively inhibit PRC2 activity in cells and reduce tumor growth in mouse xenograft models. Here, we report the stepwise optimization of the tool compound EED226 toward the potent and selective EED inhibitor MAK683 (compound 22) and its subsequent preclinical characterization. Based on a balanced PK/PD profile, efficacy, and mitigated risk of forming reactive metabolites, MAK683 has been selected for clinical development.


Sujet(s)
Histone , Tumeurs , Animaux , Antienzymes , Histone/métabolisme , Humains , Méthylation , Souris , Tumeurs/traitement médicamenteux , Complexe répresseur Polycomb-2
9.
Acta Pharmacol Sin ; 43(2): 470-482, 2022 Feb.
Article de Anglais | MEDLINE | ID: mdl-33850276

RÉSUMÉ

Aerobic glycolysis, also known as the Warburg effect, is a hallmark of cancer cell glucose metabolism and plays a crucial role in the activation of various types of immune cells. Glyceraldehyde 3-phosphate dehydrogenase (GAPDH) catalyzes the conversion of D-glyceraldehyde 3-phosphate to D-glycerate 1,3-bisphosphate in the 6th critical step in glycolysis. GAPDH exerts metabolic flux control during aerobic glycolysis and therefore is an attractive therapeutic target for cancer and autoimmune diseases. Recently, GAPDH inhibitors were reported to function through common suicide inactivation by covalent binding to the cysteine catalytic residue of GAPDH. Herein, by developing a high-throughput enzymatic screening assay, we discovered that the natural product 1,2,3,4,6-penta-O-galloyl-ß-D-glucopyranose (PGG) is an inhibitor of GAPDH with Ki = 0.5 µM. PGG blocks GAPDH activity by a reversible and NAD+ and Pi competitive mechanism, suggesting that it represents a novel class of GAPDH inhibitors. In-depth hydrogen deuterium exchange mass spectrometry (HDX-MS) analysis revealed that PGG binds to a region that disrupts NAD+ and inorganic phosphate binding, resulting in a distal conformational change at the GAPDH tetramer interface. In addition, structural modeling analysis indicated that PGG probably reversibly binds to the center pocket of GAPDH. Moreover, PGG inhibits LPS-stimulated macrophage activation by specific downregulation of GAPDH-dependent glucose consumption and lactate production. In summary, PGG represents a novel class of GAPDH inhibitors that probably reversibly binds to the center pocket of GAPDH. Our study sheds new light on factors for designing a more potent and specific inhibitor of GAPDH for future therapeutic applications.


Sujet(s)
Glyceraldehyde 3-phosphate dehydrogenases/antagonistes et inhibiteurs , Tanins hydrolysables/pharmacologie , Animaux , Évaluation préclinique de médicament/méthodes , Glucose/métabolisme , Glyceraldehyde 3-phosphate dehydrogenase (phosphorylating)/antagonistes et inhibiteurs , Humains , Échange hydrogène/deutérium couplé à la spectrométrie de masse , Acide lactique/métabolisme , Spectroscopie par résonance magnétique , Mâle , Souris , Souris de lignée C57BL , Composés organométalliques , Réaction de polymérisation en chaine en temps réel
10.
Nucleic Acids Res ; 50(2): e9, 2022 01 25.
Article de Anglais | MEDLINE | ID: mdl-34718755

RÉSUMÉ

Epigenetic therapy has significant potential for cancer treatment. However, few small potent molecules have been identified against DNA or RNA modification regulatory proteins. Current approaches for activity detection of DNA/RNA methyltransferases and demethylases are time-consuming and labor-intensive, making it difficult to subject them to high-throughput screening. Here, we developed a fluorescence polarization-based 'High-Throughput Methyl Reading' (HTMR) assay to implement large-scale compound screening for DNA/RNA methyltransferases and demethylases-DNMTs, TETs, ALKBH5 and METTL3/METTL14. This assay is simple to perform in a mix-and-read manner by adding the methyl-binding proteins MBD1 or YTHDF1. The proteins can be used to distinguish FAM-labelled substrates or product oligonucleotides with different methylation statuses catalyzed by enzymes. Therefore, the extent of the enzymatic reactions can be coupled with the variation of FP binding signals. Furthermore, this assay can be effectively used to conduct a cofactor competition study. Based on the assay, we identified two natural products as candidate compounds for DNMT1 and ALKBH5. In summary, this study outlines a powerful homogeneous approach for high-throughput screening and evaluating enzymatic activity for DNA/RNA methyltransferases and demethylases that is cheap, easy, quick, and highly sensitive.


Sujet(s)
DNA modification methylases/métabolisme , Découverte de médicament/méthodes , Tests de criblage à haut débit/méthodes , Methyltransferases/métabolisme , Oxidoreductases, (N-demethylating)/métabolisme , Protéines de transport/métabolisme , Méthylation de l'ADN , DNA modification methylases/antagonistes et inhibiteurs , Antienzymes/pharmacologie , Tests de criblage à haut débit/normes , Humains , Methyltransferases/antagonistes et inhibiteurs , Nucléotides/métabolisme , Oxidoreductases, (N-demethylating)/antagonistes et inhibiteurs , ARN/métabolisme
11.
J Med Chem ; 64(20): 15111-15125, 2021 10 28.
Article de Anglais | MEDLINE | ID: mdl-34668699

RÉSUMÉ

Post-translational modification (PTM) on protein plays important roles in the regulation of cellular function and disease pathogenesis. The systematic analysis of PTM dynamics presents great opportunities to enlarge the target space by PTM allosteric regulation. Here, we presented a framework by integrating the sequence, structural topology, and particular dynamics features to characterize the functional context and druggabilities of PTMs in the well-known kinase family. The machine learning models with these biophysical features could successfully predict PTMs. On the other hand, PTMs were identified to be significantly enriched in the reported allosteric pockets and the allosteric potential of PTM pockets were thus proposed through these biophysical features. In the end, the covalent inhibitor DC-Srci-6668 targeting the PTM pocket in c-Src kinase was identified, which inhibited the phosphorylation and locked c-Src in the inactive state. Our findings represent a crucial step toward PTM-inspired drug design in the kinase family.


Sujet(s)
CSK tyrosine-protein kinase/antagonistes et inhibiteurs , Conception de médicament , Inhibiteurs de protéines kinases/pharmacologie , CSK tyrosine-protein kinase/métabolisme , Relation dose-effet des médicaments , Humains , Apprentissage machine , Modèles moléculaires , Structure moléculaire , Inhibiteurs de protéines kinases/synthèse chimique , Inhibiteurs de protéines kinases/composition chimique , Maturation post-traductionnelle des protéines , Relation structure-activité
12.
J Med Chem ; 64(12): 8194-8207, 2021 06 24.
Article de Anglais | MEDLINE | ID: mdl-34077206

RÉSUMÉ

Disruption of EZH2-embryonic ectoderm development (EED) protein-protein interaction (PPI) is a new promising cancer therapeutic strategy. We have previously reported the discovery of astemizole, a small-molecule inhibitor targeting the EZH2-EED PPI. Herein, we report the cocrystal structure of EED in complex with astemizole at 2.15 Å. The structure elucidates the detailed binding mode of astemizole to EED and provides a structure-guided design for the discovery of a novel EZH2-EED interaction inhibitor, DC-PRC2in-01, with an affinity Kd of 4.56 µM. DC-PRC2in-01 destabilizes the PRC2 complex, thereby leading to the degradation of PRC2 core proteins and the decrease of global H3K27me3 levels in cancer cells. The proliferation of PRC2-driven lymphomas cells is effectively inhibited, and the cell cycle is arrested in the G0/G1 phase. Together, these data demonstrate that DC-PRC2in-01 could be an effective chemical probe for investigating the PRC2-related physiology and pathology and providing a promising chemical scaffold for further development.


Sujet(s)
Astémizole/analogues et dérivés , Astémizole/pharmacologie , Protéine-2 homologue de l'activateur de Zeste/antagonistes et inhibiteurs , Antienzymes/pharmacologie , Complexe répresseur Polycomb-2/antagonistes et inhibiteurs , Liaison aux protéines/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Repositionnement des médicaments , Protéine-2 homologue de l'activateur de Zeste/métabolisme , Antienzymes/synthèse chimique , Humains , Simulation de docking moléculaire , Structure moléculaire , Complexe répresseur Polycomb-2/métabolisme , Relation structure-activité
13.
Bioorg Med Chem ; 34: 116054, 2021 03 15.
Article de Anglais | MEDLINE | ID: mdl-33571875

RÉSUMÉ

Tumor suppressor p53-binding protein 1 (53BP1), a tantem tudor domain (TTD) protein, takes part in DNA Damage Repair (DDR) pathways through the specific recognition of lysine methylation on histones. The dysregulation of 53BP1 is closely related to the development of many diseases including cancer. Moreover, recent studies found that deficiency of 53BP1 could increase the efficiency of precise CRISPR/Cas9 genome editing. Thus, discovery of inhibitor is beneficial to the study of biological functions of 53BP1 and the application of CRISPR/Cas9 genome editing. UNC2170 and its derivatives have been reported as 53BP1 targeted small molecular inhibitors with modest activities. Hence, to discover better 53BP1 inhibitors, we conducted an AlphaScreen assay based high-throughput screening (HTS) and identified a novel and effective 53BP1-TTD inhibitor DP308 which disrupts the binding between 53BP1 and H4K20me2 peptide with an IC50 value of 1.69 ± 0.73 µM. Both Microscale Themophoresis (MST) and Surface Plasmon Resonance (SPR) assays confirmed the direct binding between DP308 and 53BP1-TTD protein with binding affinity (Kd) of about 2.7 µM. Molecular docking studies further suggested that DP308 possibly occupies the H4K20me2 binding pocket of the 53BP1-TTD aromatic cage. These results demonstrated that DP308 is a promising small molecule inhibitor for further optimization towards a more potent chemical probe of 53BP1. Additionally, it could be a potential valuable tool for applying to gene editing therapy by increasing the efficiency of CRISPR/Cas9 genome editing.


Sujet(s)
Découverte de médicament/méthodes , Canal potassique ERG1/métabolisme , Récepteurs couplés aux protéines G/agonistes , Protéine-1 liant le suppresseur de tumeur p53/antagonistes et inhibiteurs , Animaux , Cellules CHO , Cricetinae , Cricetulus , Canal potassique ERG1/génétique , Régulation de l'expression des gènes , Tests de criblage à haut débit , Hypoglycémiants/composition chimique , Hypoglycémiants/pharmacologie , Microsomes du foie/métabolisme , Modèles moléculaires , Structure moléculaire , Techniques de patch-clamp , Rats
14.
Med Res Rev ; 41(3): 1701-1750, 2021 05.
Article de Anglais | MEDLINE | ID: mdl-33355944

RÉSUMÉ

Modern drug design aims to discover novel lead compounds with attractable chemical profiles to enable further exploration of the intersection of chemical space and biological space. Identification of small molecules with good ligand efficiency, high activity, and selectivity is crucial toward developing effective and safe drugs. However, the intersection is one of the most challenging tasks in the pharmaceutical industry, as chemical space is almost infinity and continuous, whereas the biological space is very limited and discrete. This bottleneck potentially limits the discovery of molecules with desirable properties for lead optimization. Herein, we present a new direction leveraging posttranslational modification (PTM) protein isoforms target space to inspire drug design termed as "Post-translational Modification Inspired Drug Design (PTMI-DD)." PTMI-DD aims to extend the intersections of chemical space and biological space. We further rationalized and highlighted the importance of PTM protein isoforms and their roles in various diseases and biological functions. We then laid out a few directions to elaborate the PTMI-DD in drug design including discovering covalent binding inhibitors mimicking PTMs, targeting PTM protein isoforms with distinctive binding sites from that of wild-type counterpart, targeting protein-protein interactions involving PTMs, and hijacking protein degeneration by ubiquitination for PTM protein isoforms. These directions will lead to a significant expansion of the biological space and/or increase the tractability of compounds, primarily due to precisely targeting PTM protein isoforms or complexes which are highly relevant to biological functions. Importantly, this new avenue will further enrich the personalized treatment opportunity through precision medicine targeting PTM isoforms.


Sujet(s)
Conception de médicament , Maturation post-traductionnelle des protéines , Humains , Isoformes de protéines , Ubiquitination
15.
Opt Express ; 28(14): 20225-20235, 2020 Jul 06.
Article de Anglais | MEDLINE | ID: mdl-32680087

RÉSUMÉ

This paper presents a method of using femtosecond laser inscribed nanograting as low-loss- and high-temperature-stable in-fiber reflectors. By introducing a pair of nanograting inside the core of a single-mode optical fiber, an intrinsic Fabry-Perot interferometer can be created for high-temperature sensing applications. The morphology of the nanograting inscribed in fiber cores was engineered by tuning the fabrication conditions to achieve a high fringe visibility of 0.49 and low insertion loss of 0.002 dB per sensor. Using a white light interferometry demodulation algorithm, we demonstrate the temperature sensitivity, cross-talk, and spatial multiplexability of sensor arrays. Both the sensor performance and stability were studied from room temperature to 1000°C with cyclic heating and cooling. Our results demonstrate a femtosecond direct laser writing technique capable of producing highly multiplexable in-fiber intrinsic Fabry-Perot interferometer sensor devices with high fringe contrast, high sensitivity, and low-loss for application in harsh environmental conditions.

16.
Pharmacol Ther ; 204: 107406, 2019 12.
Article de Anglais | MEDLINE | ID: mdl-31521697

RÉSUMÉ

Epigenetics has emerged as an extremely exciting fast-growing area of biomedical research in post genome era. Epigenetic dysfunction is tightly related with various diseases such as cancer and aging related degeneration, potentiating epigenetics modulators as important therapeutics targets. Indeed, inhibitors of histone deacetylase and DNA methyltransferase have been approved for treating blood tumor malignancies, whereas inhibitors of histone methyltransferase and histone acetyl-lysine recognizer bromodomain are in clinical stage. However, it remains a great challenge to discover potent and selective inhibitors by targeting catalytic site, as the same subfamily of epigenetic enzymes often share high sequence identity and very conserved catalytic core pocket. It is well known that epigenetic modifications are usually carried out by multi-protein complexes, and activation of catalytic subunit is often tightly regulated by other interactive protein component, especially in disease conditions. Therefore, it is not unusual that epigenetic complex machinery may exhibit allosteric regulation site induced by protein-protein interactions. Targeting allosteric site emerges as a compelling alternative strategy to develop epigenetic drugs with enhanced druggability and pharmacological profiles. In this review, we highlight recent progress in the development of allosteric inhibitors for epigenetic complexes through targeting protein-protein interactions. We also summarized the status of clinical applications of those inhibitors. Finally, we provide perspectives of future novel allosteric epigenetic machinery modulators emerging from otherwise undruggable single protein target.


Sujet(s)
Régulation allostérique/effets des médicaments et des substances chimiques , Antienzymes/pharmacologie , Antienzymes/usage thérapeutique , Épigenèse génétique/effets des médicaments et des substances chimiques , Thérapie moléculaire ciblée/méthodes , Motifs et domaines d'intéraction protéique/effets des médicaments et des substances chimiques , Animaux , Humains
17.
SLAS Discov ; 23(9): 930-940, 2018 10.
Article de Anglais | MEDLINE | ID: mdl-29562800

RÉSUMÉ

Ubiquitin-like with PHD and RING finger domains 1 (UHRF1) is a multidomain protein that plays a critical role in maintaining DNA methylation patterns through concurrent recognition of hemimethylated DNA and histone marks by various domains, and recruitment of DNA methyltransferase 1 (DNMT1). UHRF1 is overexpressed in various cancers, including breast cancer. The tandem tudor domain (TTD) of UHRF1 specifically and tightly binds to histone H3 di- or trimethylated at lysine 9 (H3K9me2 or H3K9me3, respectively), and this binding is essential for UHRF1 function. We developed an H3K9me3 peptide displacement assay, which was used to screen a library of 44,000 compounds for small molecules that disrupt the UHRF1-H3K9me3 interaction. This screen resulted in the identification of NV01, which bound to UHRF1-TTD with a Kd value of 5 µM. The structure of UHRF1-TTD in complex with NV01 confirmed binding to the H3K9me3-binding pocket. Limited structure-based optimization of NV01 led to the discovery of NV03 (Kd of 2.4 µM). These well-characterized small-molecule antagonists of the UHRF1-H3K9me2/3 interaction could be valuable starting chemical matter for developing more potent and cell-active probes toward further characterizing UHRF1 function, with possible applications as anticancer therapeutics.


Sujet(s)
Protéines liant les séquences stimulatrices de type CCAAT/composition chimique , Découverte de médicament/méthodes , Histone/composition chimique , Liaison aux protéines/effets des médicaments et des substances chimiques , Domaine Tudor , Sites de fixation , Dosage biologique/méthodes , Protéines liant les séquences stimulatrices de type CCAAT/génétique , Protéines liant les séquences stimulatrices de type CCAAT/métabolisme , Histone/métabolisme , Humains , Conformation moléculaire , Simulation de docking moléculaire , Simulation de dynamique moléculaire , Structure moléculaire , Mutagenèse dirigée , Bibliothèques de petites molécules , Relation structure-activité , Ubiquitin-protein ligases
18.
ACS Chem Biol ; 13(3): 772-781, 2018 03 16.
Article de Anglais | MEDLINE | ID: mdl-29381856

RÉSUMÉ

DNA methyltransferase-1 (DNMT1) plays a crucial role in the maintenance of genomic methylation patterns. The crystal structure of DNMT1 was determined in two different states in which the helix that follows the catalytic loop was either kinked (designated helix-kinked) or well folded (designated helix-straight state). Here, we show that the proper structural transition between these two states is required for DNMT1 activity. The mutations of N1248A and R1279D, which did not affect interactions between DNMT1 and substrates or cofactors, allosterically reduced enzymatic activities in vitro by decreasing kcat/ Km for AdoMet. The crystallographic data combined with molecular dynamic (MD) simulations indicated that the N1248A and R1279D mutants bias the catalytic helix to either the kinked or straight conformation. In addition, genetic complementation assays for the two mutants suggested that disturbing the conformational transition reduced DNMT1 activity in cells, which could act additively with existing DNMT inhibitors to decrease DNA methylation. Collectively, our studies provide molecular insights into conformational changes of the catalytic helix, which is essential for DNMT1 catalytic activity, and thus aid in better understanding the relationship between DNMT1 dynamic switching and enzymatic activity.


Sujet(s)
DNA (Cytosine-5-)-methyltransferase 1/composition chimique , Simulation de dynamique moléculaire , Animaux , Domaine catalytique , Cristallographie aux rayons X , DNA (Cytosine-5-)-methyltransferase 1/génétique , Méthylation de l'ADN/génétique , Humains , Mutation , Conformation des protéines
19.
J Med Chem ; 61(3): 1204-1217, 2018 02 08.
Article de Anglais | MEDLINE | ID: mdl-29244490

RÉSUMÉ

PRMT3 catalyzes the asymmetric dimethylation of arginine residues of various proteins. It is crucial for maturation of ribosomes and has been implicated in several diseases. We recently disclosed a highly potent, selective, and cell-active allosteric inhibitor of PRMT3, compound 4. Here, we report comprehensive structure-activity relationship studies that target the allosteric binding site of PRMT3. We conducted design, synthesis, and evaluation of novel compounds in biochemical, selectivity, and cellular assays that culminated in the discovery of 4 and other highly potent (IC50 values: ∼10-36 nM), selective, and cell-active allosteric inhibitors of PRMT3 (compounds 29, 30, 36, and 37). In addition, we generated compounds that are very close analogs of these potent inhibitors but displayed drastically reduced potency as negative controls (compounds 49-51). These inhibitors and negative controls are valuable chemical tools for the biomedical community to further investigate biological functions and disease associations of PRMT3.


Sujet(s)
Conception de médicament , Protein-arginine N-methyltransferases/métabolisme , Régulation allostérique/effets des médicaments et des substances chimiques , Composés hétérocycliques bicycliques/composition chimique , Composés hétérocycliques bicycliques/pharmacologie , Cellules HEK293 , Humains , Liaison hydrogène , Concentration inhibitrice 50 , Modèles moléculaires , Conformation des protéines , Protein-arginine N-methyltransferases/composition chimique , Relation structure-activité
20.
Oncotarget ; 8(49): 85085-85101, 2017 Oct 17.
Article de Anglais | MEDLINE | ID: mdl-29156705

RÉSUMÉ

LSD1 (Lysine Specific Demethylase1)/KDM1A (Lysine Demethylase 1A), a flavin adenine dinucleotide (FAD)-dependent histone H3K4/K9 demethylase, sustains oncogenic potential of leukemia stem cells in primary human leukemia cells. However, the pro-differentiation and anti-proliferation effects of LSD1 inhibition in acute myeloid leukemia (AML) are not yet fully understood. Here, we report that small hairpin RNA (shRNA) mediated LSD1 inhibition causes a remarkable transcriptional activation of myeloid lineage marker genes (CD11b/ITGAM and CD86), reduction of cell proliferation and decrease of clonogenic ability of human AML cells. Cell surface expression of CD11b and CD86 is significantly and dynamically increased in human AML cells upon sustained LSD1 inhibition. Chromatin immunoprecipitation and quantitative PCR (ChIP-qPCR) analyses of histone marks revealed that there is a specific increase of H3K4me2 modification and an accompanied increase of H3K4me3 modification at the respective CD11b and CD86 promoter region, whereas the global H3K4me2 level remains constant. Consistently, inhibition of LSD1 in vivo significantly blocks tumor growth and induces a prominent increase of CD11b and CD86. Taken together, our results demonstrate the anti-tumor properties of LSD1 inhibition on human AML cell line and mouse xenograft model. Our findings provide mechanistic insights into the LSD1 functions in controlling both differentiation and proliferation in AML.

SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE