Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Proc Natl Acad Sci U S A ; 117(40): 24802-24812, 2020 10 06.
Article in English | MEDLINE | ID: mdl-32958664

ABSTRACT

The oligoadenylate synthetase (OAS)-RNase L system is an IFN-inducible antiviral pathway activated by viral infection. Viral double-stranded (ds) RNA activates OAS isoforms that synthesize the second messenger 2-5A, which binds and activates the pseudokinase-endoribonuclease RNase L. In cells, OAS activation is tamped down by ADAR1, an adenosine deaminase that destabilizes dsRNA. Mutation of ADAR1 is one cause of Aicardi-Goutières syndrome (AGS), an interferonopathy in children. ADAR1 deficiency in human cells can lead to RNase L activation and subsequent cell death. To evaluate RNase L as a possible therapeutic target for AGS, we sought to identify small-molecule inhibitors of RNase L. A 500-compound library of protein kinase inhibitors was screened for modulators of RNase L activity in vitro. We identified ellagic acid (EA) as a hit with 10-fold higher selectivity against RNase L compared with its nearest paralog, IRE1. SAR analysis identified valoneic acid dilactone (VAL) as a superior inhibitor of RNase L, with 100-fold selectivity over IRE1. Mechanism-of-action analysis indicated that EA and VAL do not bind to the pseudokinase domain of RNase L despite acting as ATP competitive inhibitors of the protein kinase CK2. VAL is nontoxic and functional in cells, although with a 1,000-fold decrease in potency, as measured by RNA cleavage activity in response to treatment with dsRNA activator or by rescue of cell lethality resulting from self dsRNA induced by ADAR1 deficiency. These studies lay the foundation for understanding novel modes of regulating RNase L function using small-molecule inhibitors and avenues of therapeutic potential.


Subject(s)
Adenosine Deaminase/deficiency , Autoimmune Diseases of the Nervous System/enzymology , Endoribonucleases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Nervous System Malformations/enzymology , Phenol/pharmacology , 2',5'-Oligoadenylate Synthetase/genetics , 2',5'-Oligoadenylate Synthetase/metabolism , Adenine Nucleotides/metabolism , Adenosine Deaminase/genetics , Autoimmune Diseases of the Nervous System/genetics , Autoimmune Diseases of the Nervous System/physiopathology , Cell Death/drug effects , Endoribonucleases/genetics , Endoribonucleases/metabolism , Enzyme Inhibitors/chemistry , Humans , Nervous System Malformations/genetics , Nervous System Malformations/physiopathology , Oligoribonucleotides/metabolism , Phenol/chemistry , RNA-Binding Proteins/genetics
2.
Biochem J ; 477(17): 3387-3399, 2020 09 18.
Article in English | MEDLINE | ID: mdl-32830849

ABSTRACT

The pseudokinase (PK) RNase L is a functional ribonuclease and plays important roles in human innate immunity. The ribonuclease activity of RNase L can be regulated by the kinase inhibitor sunitinib. The combined use of oncolytic virus and sunitinib has been shown to exert synergistic effects in anticancer therapy. In this study, we aimed to uncover the mechanism of action through which sunitinib inhibits RNase L. We solved the crystal structures of RNase L in complex with sunitinib and its analogs toceranib and SU11652. Our results showed that sunitinib bound to the ATP-binding pocket of RNase L. Unexpectedly, the αA helix linking the ankyrin repeat-domain and the PK domain affected the binding mode of sunitinib and resulted in an unusual flipped orientation relative to other structures in PDB. Molecular dynamics simulations and dynamic light scattering results support that the binding of sunitinib in the PK domain destabilized the dimer conformation of RNase L and allosterically inhibited its ribonuclease activity. Our study suggested that dimer destabilization could be an effective strategy for the discovery of RNase L inhibitors and that targeting the ATP-binding pocket in the PK domain of RNase L was an efficient approach for modulating its ribonuclease activity.


Subject(s)
Endoribonucleases , Protein Multimerization , Sunitinib/chemistry , Crystallography, X-Ray , Endoribonucleases/antagonists & inhibitors , Endoribonucleases/chemistry , Humans , Protein Conformation, alpha-Helical , Protein Domains
3.
Cancer Sci ; 106(5): 512-21, 2015 May.
Article in English | MEDLINE | ID: mdl-25664501

ABSTRACT

Bispecific antibodies play an important role in immunotherapy. They have received intense interest from pharmaceutical enterprises. The first antibody drug, OKT3 (muromonab-CD3), showed great performance in clinical treatment. We have successfully developed a single-chain variable fragment (ScFv) combination of anti-CD3 ScFv and anti-CD138 ScFv with the hIgG1 Fc (hIgFc) sequence. The novel bispecific T-cell engager (BiTE) with an additional hIgFc (BiTE-hIgFc, STL001) can target T cells, natural killer cells, and multiple myeloma cells (RPMI-8226 or U266). In addition, BiTE-hIgFc (STL001) has nanomolar-level affinity to recombinant human CD138 protein and shows more potent antitumor activity against RPMI-8226 cells than that of separate aCD3-ScFv-hIgFc and aCD138-ScFv-hIgFc, or the isotype mAb in vitro or in vivo.


Subject(s)
Antibodies, Bispecific/immunology , Immunotherapy/methods , Multiple Myeloma/therapy , Single-Chain Antibodies/immunology , Amino Acid Sequence , Animals , Antibodies, Bispecific/genetics , Antibodies, Bispecific/metabolism , Antigens, CD/metabolism , Antigens, Differentiation, T-Lymphocyte/metabolism , CD3 Complex/genetics , CD3 Complex/immunology , CD3 Complex/metabolism , Cell Line, Tumor , Humans , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Interleukin-2 Receptor alpha Subunit/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lectins, C-Type/metabolism , Mice, SCID , Molecular Sequence Data , Multiple Myeloma/immunology , Receptors, IgG/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Single-Chain Antibodies/genetics , Syndecan-1/genetics , Syndecan-1/immunology , Syndecan-1/metabolism , T-Lymphocytes/immunology , Xenograft Model Antitumor Assays
4.
J Med Chem ; 65(2): 1445-1457, 2022 01 27.
Article in English | MEDLINE | ID: mdl-34841869

ABSTRACT

The pseudokinase-endoribonuclease RNase L plays important roles in antiviral innate immunity and is also implicated in many other cellular activities. The inhibition of RNase L showed therapeutic potential for Aicardi-Goutières syndrome (AGS). Thus, RNase L is a promising drug target. In this study, using an enzyme assay and NMR screening, we discovered 13 inhibitory fragments against RNase L. Cocrystal structures of RNase L separately complexed with two different fragments were determined in which both fragments bound to the ATP-binding pocket of the pseudokinase domain. Myricetin, vitexin, and hyperoside, three natural products sharing similar scaffolds with the fragment AC40357, demonstrated a potent inhibitory activity in vitro. In addition, myricetin has a promising cellular inhibitory activity. A cocrystal structure of RNase L with myricetin provided a structural basis for inhibitor design by allosterically modulating the ribonuclease activity. Our findings demonstrate that fragment screening can lead to the discovery of natural product inhibitors of RNase L.


Subject(s)
Biological Products/pharmacology , Drug Discovery , Endoribonucleases/antagonists & inhibitors , High-Throughput Screening Assays/methods , Small Molecule Libraries/pharmacology , Humans
5.
Nat Commun ; 12(1): 488, 2021 01 20.
Article in English | MEDLINE | ID: mdl-33473130

ABSTRACT

SARS-CoV-2 is the pathogen responsible for the COVID-19 pandemic. The SARS-CoV-2 papain-like cysteine protease (PLpro) has been implicated in playing important roles in virus maturation, dysregulation of host inflammation, and antiviral immune responses. The multiple functions of PLpro render it a promising drug target. Therefore, we screened a library of approved drugs and also examined available inhibitors against PLpro. Inhibitor GRL0617 showed a promising in vitro IC50 of 2.1 µM and an effective antiviral inhibition in cell-based assays. The co-crystal structure of SARS-CoV-2 PLproC111S in complex with GRL0617 indicates that GRL0617 is a non-covalent inhibitor and it resides in the ubiquitin-specific proteases (USP) domain of PLpro. NMR data indicate that GRL0617 blocks the binding of ISG15 C-terminus to PLpro. Using truncated ISG15 mutants, we show that the C-terminus of ISG15 plays a dominant role in binding PLpro. Structural analysis reveals that the ISG15 C-terminus binding pocket in PLpro contributes a disproportionately large portion of binding energy, thus this pocket is a hot spot for antiviral drug discovery targeting PLpro.


Subject(s)
Antiviral Agents/chemistry , Antiviral Agents/pharmacology , COVID-19 Drug Treatment , Coronavirus 3C Proteases/chemistry , SARS-CoV-2/drug effects , COVID-19/metabolism , COVID-19/virology , Coronavirus 3C Proteases/genetics , Coronavirus 3C Proteases/metabolism , Cytokines/metabolism , Drug Discovery , Drug Interactions , HEK293 Cells , High-Throughput Screening Assays , Humans , Inhibitory Concentration 50 , Models, Molecular , Pandemics , Protein Conformation , SARS-CoV-2/genetics , SARS-CoV-2/metabolism , Ubiquitins/metabolism
6.
Int J Biol Macromol ; 188: 137-146, 2021 Oct 01.
Article in English | MEDLINE | ID: mdl-34364941

ABSTRACT

COVID-19 is a disease caused by SARS-CoV-2, which has led to more than 4 million deaths worldwide. As a result, there is a worldwide effort to develop specific drugs for targeting COVID-19. Papain-like protease (PLpro) is an attractive drug target because it has multiple essential functions involved in processing viral proteins, including viral genome replication and removal of post-translational ubiquitination modifications. Here, we established two assays for screening PLpro inhibitors according to protease and anti-ISGylation activities, respectively. Application of the two screening techniques to the library of clinically approved drugs led to the discovery of tanshinone IIA sulfonate sodium and chloroxine with their IC50 values of lower than 10 µM. These two compounds were found to directly interact with PLpro and their molecular mechanisms of binding were illustrated by docking and molecular dynamics simulations. The results highlight the usefulness of the two developed screening techniques for locating PLpro inhibitors.


Subject(s)
Antiviral Agents/pharmacology , COVID-19 Drug Treatment , Coronavirus Papain-Like Proteases/antagonists & inhibitors , Coronavirus Protease Inhibitors/pharmacology , Drug Repositioning , SARS-CoV-2/enzymology , Antiviral Agents/chemistry , Binding Sites , Chloroquinolinols/chemistry , Chloroquinolinols/pharmacology , Coronavirus Papain-Like Proteases/genetics , Coronavirus Papain-Like Proteases/isolation & purification , Coronavirus Protease Inhibitors/chemistry , High-Throughput Screening Assays/methods , Humans , Molecular Docking Simulation , Molecular Dynamics Simulation , Phenanthrenes/chemistry , Phenanthrenes/pharmacology , SARS-CoV-2/drug effects
7.
Oncotarget ; 7(23): 34070-83, 2016 Jun 07.
Article in English | MEDLINE | ID: mdl-27083001

ABSTRACT

Various CD7-targeting immunotoxins have been tested for its potential in treating CD7+ malignant patients but none of those immunotoxins was approved clinically because of lacking enough efficacy and safety. Here we successfully constructed the monovalent and bivalent CD7 nanobody-based immunotoxins PG001 and PG002, both conjugated with a truncated derivative of Pseudomonas exotoxin A respectively. The prokaryotic system expressed immunotoxins not only maintained their binding specificity for CD7-positive cells with a Kd of 16.74 nM and 3.6 nM for PG001 and PG002 respectively, but also efficiently promoted antigen-restricted apoptosis of the CD7-positive leukemia cell lines Jurkat and CEM, and primary T-cell acute lymphoblastic leukemia (T-ALL) and acute myeloid leukemia (AML) cells with an in vitro cytotoxic activity (EC50) in the range of 23-30 pM for PG002. In NOD/SCID mice transplanted with CEM cells, PG001 and PG002 prevented engraftment of the cells and markedly prolonged mouse survival. Owing to the efficient antigen-restricted anti-leukemic activity of PG002, this CD7 nanobody-based immunotoxin exhibited a superior anti-CD7 positive malignancies activity than previously reported immunotoxins, and may represent a promising therapeutic strategy in treating CD7-positive leukemia and lymphoma, which still remain a significant clinical challenge.


Subject(s)
Antineoplastic Agents/pharmacology , Immunotoxins/pharmacology , Single-Domain Antibodies/pharmacology , Animals , Antigens, CD7 , Apoptosis/drug effects , Cell Line, Tumor , Humans , Leukemia-Lymphoma, Adult T-Cell , Mice , Mice, Inbred NOD , Mice, SCID , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
Cancer Cell ; 29(5): 639-652, 2016 05 09.
Article in English | MEDLINE | ID: mdl-27132469

ABSTRACT

Proteasome inhibitors have revolutionized outcomes in multiple myeloma, but they are used empirically, and primary and secondary resistance are emerging problems. We have identified TJP1 as a determinant of plasma cell proteasome inhibitor susceptibility. TJP1 suppressed expression of the catalytically active immunoproteasome subunits LMP7 and LMP2, decreased proteasome activity, and enhanced proteasome inhibitor sensitivity in vitro and in vivo. This occurred through TJP1-mediated suppression of EGFR/JAK1/STAT3 signaling, which modulated LMP7 and LMP2 levels. In the clinic, high TJP1 expression in patient myeloma cells was associated with a significantly higher likelihood of responding to bortezomib and a longer response duration, supporting the use of TJP1 as a biomarker to identify patients most likely to benefit from proteasome inhibitors.


Subject(s)
ErbB Receptors/metabolism , Janus Kinase 1/metabolism , Multiple Myeloma/drug therapy , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/therapeutic use , STAT3 Transcription Factor/metabolism , Zonula Occludens-1 Protein/metabolism , Animals , Antineoplastic Agents/pharmacology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Bortezomib/pharmacology , Bortezomib/therapeutic use , Cell Line, Tumor , Cysteine Endopeptidases/metabolism , Disease-Free Survival , Erlotinib Hydrochloride/pharmacology , Erlotinib Hydrochloride/therapeutic use , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice, SCID , Multiple Myeloma/genetics , Multiple Myeloma/metabolism , Proteasome Inhibitors/pharmacology , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Xenograft Model Antitumor Assays/methods , Zonula Occludens-1 Protein/genetics
9.
PLoS One ; 10(11): e0142901, 2015.
Article in English | MEDLINE | ID: mdl-26565401

ABSTRACT

Impairments in mitochondrial energy metabolism are thought to be involved in many neurodegenerative diseases. The mitochondrial inhibitor 3-nitropropionic acid (3-NP) induces striatal pathology mimicking neurodegeneration in vivo. Previous studies showed that 3-NP also triggered autophagy activation and apoptosis. In this study, we focused on the high-mobility group box 1 (HMGB1) protein, which is important in oxidative stress signaling as well as in autophagy and apoptosis, to explore whether the mechanisms of autophagy and apoptosis in neurodegenerative diseases are associated with metabolic impairment. To elucidate the role of HMGB1 in striatal degeneration, we investigated the impact of HMGB1 on autophagy activation and cell death induced by 3-NP. We intoxicated rat striata with 3-NP by stereotaxic injection and analyzed changes in expression HMGB1, proapoptotic proteins caspase-3 and phospho-c-Jun amino-terminal kinases (p-JNK). 3-NP-induced elevations in p-JNK, cleaved caspase-3, and autophagic marker LC3-II as well as reduction in SQSTM1 (p62), were significantly reduced by the HMGB1 inhibitor glycyrrhizin. Glycyrrhizin also significantly inhibited 3-NP-induced striatal damage. Neuronal death was replicated by exposing primary striatal neurons in culture to 3-NP. It was clear that HMGB1 was important for basal autophagy which was shown by rescue of cells through HMGB1 targeting shRNA approach.3-NP also induced the expression of HMGB1, p-JNK, and LC3-II in striatal neurons, and p-JNK expression was significantly reduced by shRNA knockdown of HMGB1, an effect that was reversed by exogenously increased expression of HMGB1. These results suggest that HMGB1 plays important roles in signaling for both autophagy and apoptosis in neurodegeneration induced by mitochondrial dysfunction.


Subject(s)
Apoptosis , Autophagy , Corpus Striatum/physiopathology , HMGB1 Protein/genetics , Mitochondria/pathology , Neurodegenerative Diseases/genetics , Animals , Caspase 3/metabolism , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Glycyrrhizic Acid/chemistry , Heat-Shock Proteins/metabolism , Lentivirus , MAP Kinase Kinase 4/metabolism , Neurodegenerative Diseases/metabolism , Neurons/metabolism , Nitro Compounds/chemistry , Oxidative Stress , Propionates/chemistry , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Sequestosome-1 Protein , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL