Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
J Med Chem ; 67(10): 8043-8059, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38730324

ABSTRACT

Discoidin domain receptor 1 (DDR1) is a potential target for cancer drug discovery. Although several DDR1 kinase inhibitors have been developed, recent studies have revealed the critical roles of the noncatalytic functions of DDR1 in tumor progression, metastasis, and immune exclusion. Degradation of DDR1 presents an opportunity to block its noncatalytic functions. Here, we report the discovery of the DDR1 degrader LLC355 by employing autophagosome-tethering compound technology. Compound LLC355 efficiently degraded DDR1 protein with a DC50 value of 150.8 nM in non-small cell lung cancer NCI-H23 cells. Mechanistic studies revealed compound LLC355 to induce DDR1 degradation via lysosome-mediated autophagy. Importantly, compound LLC355 potently suppressed cancer cell tumorigenicity, migration, and invasion and significantly outperformed the corresponding inhibitor 1. These results underline the therapeutic advantage of targeting the noncatalytic function of DDR1 over inhibition of its kinase activity.


Subject(s)
Autophagy , Discoidin Domain Receptor 1 , Humans , Discoidin Domain Receptor 1/metabolism , Discoidin Domain Receptor 1/antagonists & inhibitors , Autophagy/drug effects , Cell Line, Tumor , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Animals , Drug Discovery , Cell Movement/drug effects , Proteolysis/drug effects , Structure-Activity Relationship , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/chemical synthesis , Cell Proliferation/drug effects , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/metabolism
2.
Bioorg Chem ; 148: 107456, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38761706

ABSTRACT

The targeting of cyclin-dependent kinase 7 (CDK7) has become a highly desirable therapeutic approach in the field of oncology due to its dual role in regulating essential biological processes, encompassing cell cycle progression and transcriptional control. We have previously identified a highly selective thieno[3,2-d]pyrimidine-based CDK7 inhibitor with demonstrated efficacy and safety in animal model. In this study, we sought to optimize the thieno[3,2-d]pyrimidine core to discover a novel series of CDK7 inhibitors with improved potency and pharmacokinetic (PK) properties. Through extensive structure-activity relationship (SAR) studies, compound 20 has emerged as the lead candidate due to its potent inhibitory activity against CDK7 and remarkable efficacy on MDA-MB-453 cells, a representative triple negative breast cancer (TNBC) cell line. Furthermore, 20 has demonstrated favorable oral bioavailability and exhibited highly desirable pharmacokinetic (PK) properties, making it a promising lead candidate for further structural optimization.


Subject(s)
Antineoplastic Agents , Cyclin-Dependent Kinase-Activating Kinase , Cyclin-Dependent Kinases , Drug Design , Protein Kinase Inhibitors , Pyrimidines , Pyrimidines/chemistry , Pyrimidines/chemical synthesis , Pyrimidines/pharmacology , Pyrimidines/pharmacokinetics , Humans , Structure-Activity Relationship , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/metabolism , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacokinetics , Molecular Structure , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Cell Line, Tumor , Rats
3.
J Med Chem ; 67(9): 6938-6951, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38687638

ABSTRACT

Nuclear receptor-binding SET domain-containing 2 (NSD2), a methyltransferase that primarily installs the dimethyl mark on lysine 36 of histone 3 (H3K36me2), has been recognized as a promising therapeutic target against cancer. However, existing NSD2 inhibitors suffer from low activity or inferior selectivity, and none of them can simultaneously remove the methyltransferase activity and chromatin binding function of NSD2. Herein we report the discovery of a novel NSD2 degrader LLC0424 by leveraging the proteolysis-targeting chimera technology. LLC0424 potently degraded NSD2 protein with a DC50 value of 20 nM and a Dmax value of 96% in acute lymphoblastic leukemia (ALL) RPMI-8402 cells. Mechanistic studies revealed LLC0424 to selectively induce NSD2 degradation in a cereblon- and proteasome-dependent fashion. LLC0424 also caused continuous downregulation of H3K36me2 and growth inhibition of ALL cell lines with NSD2 mutation. Importantly, intravenous or intraperitoneal injection of LLC0424 showed potent NSD2 degradation in vivo.


Subject(s)
Histone-Lysine N-Methyltransferase , Proteolysis , Humans , Histone-Lysine N-Methyltransferase/metabolism , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Proteolysis/drug effects , Animals , Cell Line, Tumor , Mice , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Drug Discovery , Proteasome Endopeptidase Complex/metabolism , Structure-Activity Relationship , Ubiquitin-Protein Ligases/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Histones/metabolism , Cell Proliferation/drug effects
4.
J Med Chem ; 67(8): 6099-6118, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38586950

ABSTRACT

The duality of function (cell cycle regulation and gene transcription) of cyclin-dependent kinase 7 (CDK7) makes it an attractive oncology target and the discovery of CDK7 inhibitors has been a long-term pursuit by academia and pharmaceutical companies. However, achieving selective leading compounds is still difficult owing to the similarities among the ATP binding pocket. Herein, we detail the design and synthesis of a series of macrocyclic derivatives with pyrazolo[1,5-a]-1,3,5-triazine core structure as potent and selective CDK7 inhibitors. The diverse manners of macrocyclization led to distinguished selectivity profiles of the CDK family. Molecular dynamics (MD) simulation explained the binding difference between 15- and 16-membered macrocyclic compounds. Further optimization generated compound 37 exhibiting good CDK7 inhibitory activity and high selectivity over other CDKs. This work clearly demonstrated macrocyclization is a versatile method to finely tune the selectivity profile of small molecules and MD simulation can be a valuable tool in prioritizing designs of the macrocycle.


Subject(s)
Cyclin-Dependent Kinases , Drug Design , Macrocyclic Compounds , Molecular Dynamics Simulation , Protein Kinase Inhibitors , Macrocyclic Compounds/pharmacology , Macrocyclic Compounds/chemical synthesis , Macrocyclic Compounds/chemistry , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/metabolism , Humans , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Structure-Activity Relationship , Cyclin-Dependent Kinase-Activating Kinase
5.
Bioorg Med Chem ; 103: 117661, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38489998

ABSTRACT

Son of sevenless homolog 1 (SOS1) plays a pivotal role as a molecular switch in the conversion of GDP-bound inactive KRAS to its active GTP-bound form, making SOS1 a promising therapeutic target for KRAS-driven cancers. While the most advanced SOS1 inhibitor has processed to phase I clinical trial, the exploration of novel SOS1 targeting strategies with distinct modes of action remains required. By employing proteolysis targeting chimera (PROTAC) technology, we obtained a series of new SOS1 degraders. The representative compound LHF418 potently induced SOS1 degradation with a DC50 value of 209.4 nM and a Dmax value of over 80 %. Mechanistic studies have illuminated that compound LHF418 induced the formation of ternary complex involving SOS1-PROTAC-cereblon (CRBN) and triggered SOS1 protein degradation in a CRBN- and proteasome-dependent manner. In addition, compound LHF418 effectively inhibited KRAS-RAF-ERK signalling, leading to the suppression of colony formation in KRAS-driven cancer cells. Overall, compound LHF418 represents a new lead compound in the developing novel and potent therapy for the treatment of KRAS-driven cancers.


Subject(s)
Proteolysis Targeting Chimera , Proto-Oncogene Proteins p21(ras) , Cell Line, Tumor , Proteolysis , Proto-Oncogene Proteins p21(ras)/genetics , Signal Transduction
6.
Eur J Med Chem ; 269: 116310, 2024 Apr 05.
Article in English | MEDLINE | ID: mdl-38479166

ABSTRACT

Hematopoietic progenitor kinase 1 (HPK1) is a negative regulator of T-cell receptor signaling. While HPK1 is considered as a promising target for cancer immunotherapy, no small-molecule HPK1 inhibitors have been approved for cancer treatment. Herein, we report the discovery of a series of new HPK1 inhibitors with a 5-aminopyrido[2,3-d]pyrimidin-7(8H)-one scaffold. The most potent compound 9f inhibited HPK1 kinase activity with an IC50 of 0.32 nM in the time-resolved fluorescence resonance energy transfer (TR-FRET) assays, while displayed reasonable selectivity in a panel of 416 kinases. Cellular engagement of HPK1 by compound 9f was confirmed through the nano-bioluminescence resonance energy transfer (Nano-BRET) experiments. Compound 9f effectively reduced the phosphorylation of the downstream protein SLP-76 in primary peripheral blood mononuclear cells (PBMCs) and human T lymphocytic leukemia Jurkat cells. Compound 9f also enhanced the IL-2 and IFN-γ secretion in PBMCs. Furthermore, the binding mode of compound 9f with HPK1 was confirmed by the resolved cocrystal structure. Taken together, this study provides HPK1 inhibitors with a novel scaffold and clear binding mode for further development of HPK1-targeted therapeutic agents.


Subject(s)
Leukocytes, Mononuclear , Protein Serine-Threonine Kinases , Humans , Leukocytes, Mononuclear/metabolism , Signal Transduction , Phosphorylation
7.
Acta Pharmacol Sin ; 45(1): 1-22, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37704745

ABSTRACT

Epilepsy is a prevalent neurological disorder characterized by unprovoked seizures. γ-Aminobutyric acid (GABA) serves as the primary fast inhibitory neurotransmitter in the brain, and GABA binding to the GABAA receptor (GABAAR) regulates Cl- and bicarbonate (HCO3-) influx or efflux through the channel pore, leading to GABAergic inhibition or excitation, respectively. The neuron-specific K+-Cl- cotransporter 2 (KCC2) is essential for maintaining a low intracellular Cl- concentration, ensuring GABAAR-mediated inhibition. Impaired KCC2 function results in GABAergic excitation associated with epileptic activity. Loss-of-function mutations and altered expression of KCC2 lead to elevated [Cl-]i and compromised synaptic inhibition, contributing to epilepsy pathogenesis in human patients. KCC2 antagonism studies demonstrate the necessity of limiting neuronal hyperexcitability within the brain, as reduced KCC2 functioning leads to seizure activity. Strategies focusing on direct (enhancing KCC2 activation) and indirect KCC2 modulation (altering KCC2 phosphorylation and transcription) have proven effective in attenuating seizure severity and exhibiting anti-convulsant properties. These findings highlight KCC2 as a promising therapeutic target for treating epilepsy. Recent advances in understanding KCC2 regulatory mechanisms, particularly via signaling pathways such as WNK, PKC, BDNF, and its receptor TrkB, have led to the discovery of novel small molecules that modulate KCC2. Inhibiting WNK kinase or utilizing newly discovered KCC2 agonists has demonstrated KCC2 activation and seizure attenuation in animal models. This review discusses the role of KCC2 in epilepsy and evaluates its potential as a drug target for epilepsy treatment by exploring various strategies to regulate KCC2 activity.


Subject(s)
Epilepsy , Symporters , Animals , Humans , K Cl- Cotransporters , Symporters/metabolism , Epilepsy/drug therapy , Epilepsy/metabolism , Neurons/metabolism , gamma-Aminobutyric Acid/metabolism , Seizures
8.
Eur J Med Chem ; 264: 115974, 2024 Jan 15.
Article in English | MEDLINE | ID: mdl-38007910

ABSTRACT

Fms-like tyrosine kinase 3 (FLT3) has been validated as a therapeutic target for acute myeloid leukemia (AML). While a number of FLT3 kinase inhibitors have been approved for AML treatment, the clinical data revealed that they cannot achieve complete and sustained suppression of FLT3 signaling at the tolerated dose. Here we report a series of new, potent and selective FLT3 proteolysis targeting chimera degraders. The optimal compound LWY713 potently induced the degradation of FLT3 with a DC50 value of 0.64 nM and a Dmax value of 94.8% in AML MV4-11 cells with FLT3-internal tandem duplication (ITD) mutation. Mechanistic studies demonstrated that LWY713 selectively induced FLT3 degradation in a cereblon- and proteasome-dependent manner. LWY713 potently inhibited FLT3 signaling, suppressed cell proliferation, and induced cell G0/G1-phase arrest and apoptosis in MV4-11 cells. Importantly, LWY713 displayed potent in vivo antitumor activity in MV4-11 xenograft models.


Subject(s)
Leukemia, Myeloid, Acute , fms-Like Tyrosine Kinase 3 , Humans , fms-Like Tyrosine Kinase 3/genetics , Cell Line, Tumor , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Mutation , Cell Proliferation , Apoptosis , Leukemia, Myeloid, Acute/pathology
9.
J Med Chem ; 66(17): 12432-12445, 2023 09 14.
Article in English | MEDLINE | ID: mdl-37605297

ABSTRACT

The phosphoinositide kinase PIKfyve has emerged as a new potential therapeutic target in various cancers. However, limited clinical progress has been achieved with PIKfyve inhibitors. Here, we report the discovery of a first-in-class PIKfyve degrader 12d (PIK5-12d) by employing the proteolysis-targeting chimera approach. PIK5-12d potently degraded PIKfyve protein with a DC50 value of 1.48 nM and a Dmax value of 97.7% in prostate cancer VCaP cells. Mechanistic studies revealed that it selectively induced PIKfyve degradation in a VHL- and proteasome-dependent manner. PIKfyve degradation by PIK5-12d caused massive cytoplasmic vacuolization and blocked autophagic flux in multiple prostate cancer cell lines. Importantly, PIK5-12d was more effective in suppressing the growth of prostate cancer cells than the parent inhibitor and exerted prolonged inhibition of downstream signaling. Further, intraperitoneal administration of PIK5-12d exhibited potent PIKfyve degradation and suppressed tumor proliferation in vivo. Overall, PIK5-12d is a valuable chemical tool for exploring PIKfyve-based targeted therapy.


Subject(s)
Prostatic Neoplasms , Humans , Male , Autophagy , Cell Line , Cytoplasm , Lipids , Prostatic Neoplasms/drug therapy
10.
Cell Rep ; 42(7): 112750, 2023 07 25.
Article in English | MEDLINE | ID: mdl-37421623

ABSTRACT

The present study examines whether there is a mechanism beyond the current concept of post-translational modifications to regulate the function of a protein. A small gas molecule, hydrogen sulfide (H2S), was found to bind at active-site copper of Cu/Zn-SOD using a series of methods including radiolabeled binding assay, X-ray absorption near-edge structure (XANES), and crystallography. Such an H2S binding enhanced the electrostatic forces to guide the negatively charged substrate superoxide radicals to the catalytic copper ion, changed the geometry and energy of the frontier molecular orbitals of the active site, and subsequently facilitated the transfer of an electron from the superoxide radical to the catalytic copper ion and the breakage of the copper-His61 bridge. The physiological relevance of such an H2S effect was also examined in both in vitro and in vivo models where the cardioprotective effects of H2S were dependent on Cu/Zn-SOD.


Subject(s)
Copper , Hydrogen Sulfide , Copper/metabolism , Superoxide Dismutase/metabolism , Catalytic Domain , Superoxides , Zinc/metabolism
11.
J Med Chem ; 66(3): 1873-1891, 2023 02 09.
Article in English | MEDLINE | ID: mdl-36695404

ABSTRACT

AXL kinase is heavily involved in tumorigenesis, metastasis, and drug resistance of many cancers, and several AXL inhibitors are in clinical investigations. Recent studies demonstrated that the N-terminal distal region of AXL plays more important roles in cell invasiveness than its C-terminal kinase domain. Therefore, degradation of AXL may present a novel superior therapeutic approach than the kinase inhibitor therapy. Herein, we report the discovery of a series of new AXL PROTAC degraders. One representative compound 6n potently depletes AXL with a DC50 value of 5 nM in MDA-MB-231 TNBC cells. It also demonstrates significantly improved potencies against the AXL signaling activation, cell proliferation, migration and invasion of TNBC cells comparing with the corresponding kinase inhibitor. Moreover, the compound exhibits promising therapeutic potential both in patient-derived organoids and a xenograft mouse model of MDA-MB-231 cells.


Subject(s)
Receptor Protein-Tyrosine Kinases , Triple Negative Breast Neoplasms , Humans , Mice , Animals , Triple Negative Breast Neoplasms/pathology , Proto-Oncogene Proteins/metabolism , Cell Line, Tumor , Signal Transduction , Cell Proliferation , Disease Models, Animal
12.
J Med Chem ; 65(16): 11066-11083, 2022 08 25.
Article in English | MEDLINE | ID: mdl-35938508

ABSTRACT

Selective degradation of the cyclin-dependent kinases 12 and 13 (CDK12/13) presents a novel therapeutic opportunity for triple-negative breast cancer (TNBC), but there is still a lack of dual CDK12/13 degraders. Here, we report the discovery of the first series of highly potent and selective dual CDK12/13 degraders by employing the proteolysis-targeting chimera (PROTAC) technology. The optimal compound 7f effectively degraded CDK12 and CDK13 with DC50 values of 2.2 and 2.1 nM, respectively, in MDA-MB-231 breast cancer cells. Global proteomic profiling demonstrated the target selectivity of 7f. In vitro, 7f suppressed expression of core DNA damage response (DDR) genes in a time- and dose-dependent manner. Further, 7f markedly inhibited proliferation of multiple TNBC cell lines including MFM223, with an IC50 value of 47 nM. Importantly, 7f displayed a significantly improved antiproliferative activity compared to the structurally similar inhibitor 4, suggesting the potential advantage of a CDK12/13 degrader for TNBC targeted therapy.


Subject(s)
CDC2 Protein Kinase , Cyclin-Dependent Kinases , Triple Negative Breast Neoplasms , Humans , CDC2 Protein Kinase/antagonists & inhibitors , Cell Line, Tumor , Cyclin-Dependent Kinases/antagonists & inhibitors , Proteolysis , Proteomics , Triple Negative Breast Neoplasms/drug therapy
13.
Bioorg Med Chem Lett ; 64: 128683, 2022 05 15.
Article in English | MEDLINE | ID: mdl-35307569

ABSTRACT

Photoinduced drug release can reduce systemic side effects by releasing active drugs with high spatiotemporal accuracy, representing a promising strategy for precise cancer therapy. Here we designed and synthesized a novel photocaged B-RafV600E inhibitor 2, which, upon UV irradiation, could release a potent B-RafV600E inhibitor 1. Accordingly, once activated by the UV light, compound 2 could potently inhibit the proliferation of melanoma cells bearing B-RafV600E mutant while sparing melanoma cells expressing wild-type B-Raf, and could dose-dependently suppress the activation of the MAPK signaling pathway. Notably, the UV-mediated active component release and the resulting antiproliferative effects of compound 2 could be recapitulated when exposed to the sunlight, greatly enhancing its practicality. This photocaged B-RafV600E inhibitor 2 might serve as a novel therapeutic agent toward precise melanoma treatment.


Subject(s)
Melanoma , Cell Line, Tumor , Cell Proliferation , Humans , MAP Kinase Signaling System , Melanoma/drug therapy , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf , Signal Transduction
14.
J Med Chem ; 65(3): 1735-1748, 2022 02 10.
Article in English | MEDLINE | ID: mdl-35000385

ABSTRACT

Protein kinases have been highly fruitful targets for cancer drug discovery in the past two decades, while most of these drugs bind to the "adenosine triphosphate (ATP)-site" and inhibit kinase catalytic activity. Recently, accumulated evidence suggests that kinases possess functions beyond catalysis through their scaffolds, and the scaffolding functions could play critical roles in multiple cellular signaling and cell fate controls. Small molecules modulating the noncatalytic functions of kinases are rarely reported but emerge as new promising therapeutic strategies for various diseases. Herein, we summarize the characterized noncatalytic functions of kinases, and highlight the recent progress on developing small-molecule modulators of the noncatalytic functions of kinases. Mechanisms and characteristics of different kinds of modulators are also discussed. It is also speculated that targeting the noncatalytic functions would represent a new direction for kinase-based drug discovery.


Subject(s)
Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Protein Kinases/metabolism , Allosteric Regulation/drug effects , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Drug Discovery , Humans , Neoplasms/drug therapy , Protein Binding/drug effects , Protein Kinase Inhibitors/therapeutic use , Signal Transduction/drug effects , Transcription, Genetic/drug effects
15.
Bioengineered ; 13(2): 1975-1987, 2022 02.
Article in English | MEDLINE | ID: mdl-34898382

ABSTRACT

Bacterial peritonitis is a severe disease that diagnosis remains challenging for clinicians. Measuring biomarkers might be a rapid diagnostic method. The objective of this study was to analyze and evaluate the dynamic changes in HIF-1α concentration in serum exosomes during bacterial peritonitis. The pre-clinical application value of serum exosomal HIF-1α was evaluated via imipenem and cilastatin sodium (ICS) intervention in the bacterial peritonitis model. The new colorimetric method to quantitate dynamic expression changes of HIF-1α in serum exosomes during bacterial peritonitis was established by our team via using the gold seed-coated with aptamer-functionalized Au @ Au core-shell peroxidase mimic. The typical inflammatory cytokines of bacterial peritonitis were also measured. Following intramuscular administration with ICS, In-Vivo Xtreme imaging system was used to visualize abdominal infection extent. Meanwhile, HIF-1α concentration in rat serum exosomes and pro-inflammatory factors levels in serum were detected. The serum typical inflammatory cytokines levels were elevated in GFP-labeled E.coli induced bacterial peritonitis. The serum exosomal HIF-1α levels clearly increased at 12 h, reached the peak during 24-48 h, and then gradually decreased at 72 h. Following intramuscular administration with ICS, the abdominal infection extent, HIF-1α concentration in serum exosomes, and the serum pro-inflammatory factors levels were reduced at 24 h in GFP-labeled E. coli induced bacterial peritonitis model. The serum exosomal HIF-1α can be used as a biomarker in the early stage of bacterial peritonitis, which might provide the basic research in the pre-clinical for further predicting and monitoring the pathological process of bacterial peritonitis.


Subject(s)
Bacterial Infections/blood , Hypoxia-Inducible Factor 1, alpha Subunit/blood , Peritonitis/blood , Animals , Biomarkers/blood , Female , Male , Mice , Mice, Inbred BALB C , Rats , Rats, Sprague-Dawley
16.
Enzyme Microb Technol ; 147: 109786, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33992408

ABSTRACT

Xylanases degrade xylan to valuable end products. In our previous study, the alkaline xylanase S7-xyl from Bacillus halodurans S7 was engineered by rational design and the best mutant xylanase 254RL1 exhibited 3.4-fold improvements in specific activity at pH 9.0. Further research found that the enzyme activity at pH 6.0 was almost 2-fold than that at pH 9.0. To elucidate the reason of enhanced performance of 254RL1 at decreased pH optimum, we determined the X-ray crystal structure of 254RL1 at 2.21 Å resolution. The structural analysis revealed that the mutations enlarged the opening of the access tunnel and shortened the tunnel. Moreover, the mutations changed the hydrogen bond network around the catalytic residue and decreased the pKa value of acid-base catalyst E159 which reduced the pH optimum of the xylanase. The result provided the basis for the acid-alkaline engineering of the glycoside hydrolases.


Subject(s)
Bacillus , Endo-1,4-beta Xylanases , Bacillus/genetics , Endo-1,4-beta Xylanases/genetics , Hydrogen Bonding , Hydrogen-Ion Concentration
17.
Mikrochim Acta ; 187(1): 61, 2019 12 18.
Article in English | MEDLINE | ID: mdl-31853650

ABSTRACT

An ultra-sensitive method is described here for the determination of HIF-1α (an early biomarker for myocardial infarction) in circulating exosomes in serum. Gold nanospheres were functionalized with a HIF-1α-binding aptamer via sulfydryl chemistry. The apt-AuNP-coated gold seeds were grown by seed-mediated growth, and this significantly increased the peroxidase-mimicking property the nanoparticles. A chromogenic system composed of 3,3'5,5'-tetramethylbenzidine and hydrogen peroxide was used. Absorbance at 652 nm increases linearly in the 0.3 to 200 ng L-1 HIF-1α concentration range, and the limit of detection is 0.2 ng L-1. The method was tested by analyzing rat serum from isoproterenol (ISO)-induced myocardial infarction. It allows HIF-1α to be directly determined in a 25 µL sample without preconcentration. The assay is not interfered by the polydispersity of exosomes released under either health and disease conditions. Graphical abstractGold nanospheres were functionalized with a HIF-1α-binding aptamer via sulfydryl chemistry. Nanosized gold seed particles were then modified with the functionalized gold nanospheres, and this strongly increases the peroxidase-mimicking activity of the nanomaterial. By using the tetramethylbenzidine/H2O2 chromogenic system, the absorbance at 652 nm increases linearly in the 0.3 to 200 ng L-1 HIF-1α concentration range.


Subject(s)
Aptamers, Nucleotide/chemistry , Colorimetry , Exosomes/chemistry , Gold/chemistry , Hypoxia-Inducible Factor 1, alpha Subunit/blood , Peroxidase/chemistry , Animals , Aptamers, Nucleotide/metabolism , Biomarkers/blood , Biomarkers/metabolism , Enzyme-Linked Immunosorbent Assay , Exosomes/metabolism , Gold/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Myocardial Infarction/blood , Myocardial Infarction/metabolism , Particle Size , Peroxidase/metabolism , Rats , Rats, Sprague-Dawley , Surface Properties
18.
Mitochondrion ; 26: 7-18, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26593335

ABSTRACT

PURPOSE: Mitochondrial dysfunction is a prominent feature of ischemia heart disease but the underlying mechanism of dynamics (fusion/fission) is still unclear. Here we investigated a novel function and underlying mechanism of Rg1 on an in vitro cardiomyocyte model of hypoxia/reoxygenation (H/R). METHODS: Cellular cytotoxicity was evaluated by MTT, mitochondrial viable staining, and cardiac marker detection. Mitochondrial function was evaluated by ATP content measurement, MMP determination, ROS, OCR and ECAR assay. Mitochondrial dynamics was investigated by Live-cell imaging with time-lapse fluorescence microscopy and morphological features were evaluated by the high-content image analysis. Mitochondrial fusion and fission-related proteins, GDH were determined by Western blot, RT-PCR and immunofluorescence. RESULTS: Rg1 moderated GDH dysregulation and then protected against H/R-induced cellular damage and mitochondrial dysfunction in a dose-dependent manner. Rg1 significantly increased mitochondrial length, reduced the number of cells with fragmented mitochondria and up-regulated the MFN2 expression finally leading to preventing the imbalance of mitochondrial dynamics following H/R. Knock-down of MFN2 by specific siRNA completely abolished the ability of Rg1 to cell survival by H/R. CONCLUSION: Rg1 through modulation of GDH and MFN2 maintained mitochondrial dynamics that resulted in protection against H/R-induced cardiomyocyte injury. All these results put forward a new protective mechanism of Rg1 on the therapeutic potential in cardiac I/R disorders.


Subject(s)
Glutamate Dehydrogenase/metabolism , Membrane Proteins/metabolism , Mitochondria, Heart/metabolism , Mitochondrial Proteins/metabolism , Myocardial Ischemia/metabolism , Myocytes, Cardiac/metabolism , Phosphoprotein Phosphatases/metabolism , Animals , Cell Line , GTP Phosphohydrolases , Glutamate Dehydrogenase/genetics , Membrane Proteins/genetics , Mitochondria, Heart/genetics , Mitochondria, Heart/pathology , Mitochondrial Proteins/genetics , Myocardial Ischemia/pathology , Myocytes, Cardiac/pathology , Phosphoprotein Phosphatases/genetics , Rats
19.
Curr Pharm Des ; 20(8): 1259-67, 2014.
Article in English | MEDLINE | ID: mdl-23713777

ABSTRACT

Mutant p53 could have either a dominant negative effect or a gain of function to interfere with p53's ability to maintain genomic stability. In the present study, we screened for TP53 mutations in hepatocellular carcinoma (HCC) samples from 202 Chinese patients, followed by analysis of transcriptional and apoptotic activities of 21 p53 mutants with or without wild-type p53 present. We identified a new point mutation p.P72A, and a mutation (p.E294SfsX51) with a record long frameshift. We found TP53 mutations in HCC bear mutants without a dominant wild-type p53 inhibition on p21 transcription at a higher frequency. We found an anti-correlation for p53 WT/mutant heterotetramer to activate p21 and BAX transcription, i.e., at given p53 WT/mutant concentration, the fold increase p21 transcription is proportional to the fold of decreasing BAX transcription. Our kinetic model reproduced the trend in the experimental observation and confirmed that the p53 WT-dimer/mutant- heterotetramer is the major species to confer the differential activation of p21 and BAX transcription. p53 may have different binding modes on p21 and BAX, most likely resulting from the combinational effects of core domain binding and C-terminal mediation. Our study demonstrated that p53 mutants interfere with the ability of WT p53 to maintain genomic stability.


Subject(s)
Genetic Therapy , Genomic Instability , Liver Neoplasms/genetics , Tumor Suppressor Protein p53/genetics , Apoptosis/genetics , Blotting, Western , Cell Culture Techniques , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/genetics , DNA/genetics , Humans , Liver Neoplasms/pathology , Liver Neoplasms/therapy , Models, Genetic , Plasmids , Point Mutation , Transcription, Genetic , bcl-2-Associated X Protein/genetics
20.
Plant Sci ; 210: 141-50, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23849121

ABSTRACT

Granule-bound starch synthase 1 (GBSS1) is responsible for amylose synthesis in cereals, and this enzyme is regulated at the transcriptional and post-transcriptional levels. In this study, we show that GBSS1 from Oryza sativa L. (OsGBSS1) can form oligomers in rice endosperm, and oligomerized OsGBSS1 exhibits much higher specific enzymatic activity than the monomer. A monomer-oligomer transition equilibrium for OsGBSS1 occurs in the endosperm during development. Redox potential is a key factor affecting the oligomer percentage as well as the enzymatic activity of OsGBSS1. Adenosine diphosphate glucose, the direct donor of glucose, also impacts OsGBSS1 oligomerization in a concentration-dependent manner. OsGBSS1 oligomerization is influenced by phosphorylation status, which was strongly enhanced by Mitogen-activated protein kinase (MAPK) and ATP treatment and was sharply weakened by protein phosphatase (PPase) treatment. The activity of OsGBSS1 affects the ratio of amylose to amylopectin and therefore the eating quality of rice. Understanding the regulation of OsGBSS1 activity may lead to the improvement of rice eating quality.


Subject(s)
Gene Expression Regulation, Plant , Oryza/enzymology , Starch Synthase/metabolism , Starch/metabolism , Endosperm , NADP , Oryza/genetics , Phosphorylation , Plant Proteins/genetics , Plant Proteins/metabolism , Plants, Genetically Modified , Protein Multimerization , Recombinant Proteins , Starch Synthase/genetics , Starch Synthase/isolation & purification , Two-Hybrid System Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...