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










Publication year range
1.
Cell Chem Biol ; 31(5): 955-961.e4, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38215746

ABSTRACT

NLRP1 is an innate immune receptor that detects pathogen-associated signals, assembles into a multiprotein structure called an inflammasome, and triggers a proinflammatory form of cell death called pyroptosis. We previously discovered that the oxidized, but not the reduced, form of thioredoxin-1 directly binds to NLRP1 and represses inflammasome formation. However, the molecular basis for NLRP1's selective association with only the oxidized form of TRX1 has not yet been established. Here, we leveraged AlphaFold-Multimer, site-directed mutagenesis, thiol-trapping experiments, and mass spectrometry to reveal that a specific cysteine residue (C427 in humans) on NLRP1 forms a transient disulfide bond with oxidized TRX1. Overall, this work demonstrates how NLRP1 monitors the cellular redox state, further illuminating an unexpected connection between the intracellular redox potential and the innate immune system.


Subject(s)
Adaptor Proteins, Signal Transducing , Disulfides , NLR Proteins , Oxidation-Reduction , Thioredoxins , Humans , Disulfides/chemistry , Disulfides/metabolism , Thioredoxins/metabolism , Thioredoxins/chemistry , NLR Proteins/metabolism , NLR Proteins/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/chemistry , HEK293 Cells , Apoptosis Regulatory Proteins/metabolism , Apoptosis Regulatory Proteins/chemistry , Inflammasomes/metabolism , Cysteine/metabolism , Cysteine/chemistry
2.
bioRxiv ; 2023 Sep 28.
Article in English | MEDLINE | ID: mdl-37808697

ABSTRACT

NLRP1 is an innate immune receptor that detects pathogen-associated signals, assembles into a multiprotein structure called an inflammasome, and triggers a proinflammatory form of cell death called pyroptosis. We previously discovered that the oxidized, but not the reduced, form of thioredoxin-1 directly binds to NLRP1 and represses inflammasome formation. However, the molecular basis for NLRP1's selective association with only the oxidized form of TRX1 has not yet been established. Here, we leveraged Alphafold-Multimer, site-directed mutagenesis, thiol-trapping experiments, and mass spectrometry to reveal that a specific cysteine residue (C427 in humans) on NLRP1 forms a transient disulfide bond with oxidized TRX1. Overall, this work demonstrates how NLRP1 monitors the cellular redox state, further illuminating an unexpected connection between the intracellular redox potential and the innate immune system.

3.
J Med Chem ; 66(4): 2589-2607, 2023 02 23.
Article in English | MEDLINE | ID: mdl-36724486

ABSTRACT

Inflammasomes are innate immune signaling platforms that trigger pyroptotic cell death. NLRP1 and CARD8 are related human inflammasomes that detect similar danger signals, but NLRP1 has a higher activation threshold and triggers a more inflammatory form of pyroptosis. Both sense the accumulation of intracellular peptides with Xaa-Pro N-termini, but Xaa-Pro peptides on their own without a second danger signal only activate the CARD8 inflammasome. We recently reported that a dual inhibitor of the Xaa-Pro-cleaving M24B aminopeptidases PEPD and XPNPEP1 called CQ31 selectively activates the CARD8 inflammasome by inducing the build-up of Xaa-Pro peptides. Here, we performed structure-activity relationship studies on CQ31 to develop the optimized dual PEPD/XPNPEP1 inhibitor CQ80 that more effectively induces CARD8 inflammasome activation. We anticipate that CQ80 will become a valuable tool to study the basic biology and therapeutic potential of selective CARD8 inflammasome activation.


Subject(s)
Aminopeptidases , Inflammasomes , Humans , Inflammasomes/metabolism , Aminopeptidases/metabolism , Apoptosis Regulatory Proteins/metabolism , Signal Transduction , Pyroptosis , Neoplasm Proteins/metabolism , CARD Signaling Adaptor Proteins/metabolism
4.
Cell Rep ; 42(1): 111966, 2023 01 31.
Article in English | MEDLINE | ID: mdl-36649710

ABSTRACT

The danger signals that activate the related nucleotide-binding domain leucine-rich repeat pyrin domain-containing 1 (NLRP1) and caspase activation and recruitment domain-containing 8 (CARD8) inflammasomes have not been fully established. We recently reported that the oxidized form of TRX1 binds to NLRP1 and represses inflammasome activation. These findings suggested that intracellular reductive stress, which would reduce oxidized TRX1 and thereby abrogate the NLRP1-TRX1 interaction, is an NLRP1 inflammasome-activating danger signal. However, no agents that induce reductive stress were known to test this premise. Here, we identify and characterize several radical-trapping antioxidants, including JSH-23, that induce reductive stress. We show that these compounds accelerate the proteasome-mediated degradation of the repressive N-terminal fragments of both NLRP1 and CARD8, releasing the inflammasome-forming C-terminal fragments from autoinhibition. Overall, this work validates chemical probes that induce reductive stress and establishes reductive stress as a danger signal sensed by both the NLRP1 and CARD8 inflammasomes.


Subject(s)
Adaptor Proteins, Signal Transducing , Inflammasomes , Inflammasomes/metabolism , Adaptor Proteins, Signal Transducing/metabolism , NLR Proteins/metabolism , Apoptosis Regulatory Proteins/metabolism , Cell Death
5.
Cell Rep ; 42(1): 111965, 2023 01 31.
Article in English | MEDLINE | ID: mdl-36649711

ABSTRACT

NLRP1 and CARD8 are related pattern-recognition receptors (PRRs) that detect intracellular danger signals and form inflammasomes. Both undergo autoproteolysis, generating N-terminal (NT) and C-terminal (CT) fragments. The proteasome-mediated degradation of the NT releases the CT from autoinhibition, but the stimuli that trigger NT degradation have not been fully elucidated. Here, we show that several distinct agents that interfere with protein folding, including aminopeptidase inhibitors, chaperone inhibitors, and inducers of the unfolded protein response, accelerate NT degradation. However, these agents alone do not trigger inflammasome formation because the released CT fragments are physically sequestered by the serine dipeptidase DPP9. We show that DPP9-binding ligands must also be present to disrupt these complexes and allow the CT fragments to oligomerize into inflammasomes. Overall, these results indicate that NLRP1 and CARD8 detect a specific perturbation that induces both protein folding stress and DPP9 ligand accumulation.


Subject(s)
Adaptor Proteins, Signal Transducing , Inflammasomes , Inflammasomes/metabolism , Adaptor Proteins, Signal Transducing/metabolism , NLR Proteins/metabolism , Apoptosis Regulatory Proteins/metabolism , Protein Folding , CARD Signaling Adaptor Proteins/metabolism
6.
Sci Immunol ; 7(77): eabm7200, 2022 11 11.
Article in English | MEDLINE | ID: mdl-36332009

ABSTRACT

The danger signals that activate the NLRP1 inflammasome have not been established. Here, we report that the oxidized, but not the reduced, form of thioredoxin-1 (TRX1) binds to NLRP1. We found that oxidized TRX1 associates with the NACHT-LRR region of NLRP1 in an ATP-dependent process, forming a stable complex that restrains inflammasome activation. Consistent with these findings, patient-derived and ATPase-inactivating mutations in the NACHT-LRR region that cause hyperactive inflammasome formation interfere with TRX1 binding. Overall, this work strongly suggests that reductive stress, the cellular perturbation that will eliminate oxidized TRX1 and abrogate the TRX1-NLRP1 interaction, is a danger signal that activates the NLRP1 inflammasome.


Subject(s)
Inflammasomes , Thioredoxins , Humans , Inflammasomes/metabolism , Thioredoxins/genetics , Thioredoxins/metabolism , Adaptor Proteins, Signal Transducing , NLR Proteins/metabolism
7.
J Biol Chem ; 298(7): 102032, 2022 07.
Article in English | MEDLINE | ID: mdl-35580636

ABSTRACT

CARD8 is a pattern-recognition receptor that forms a caspase-1-activating inflammasome. CARD8 undergoes constitutive autoproteolysis, generating an N-terminal (NT) fragment with a disordered region and a ZU5 domain and a C-terminal (CT) fragment with UPA and CARD domains. Dipeptidyl peptidase 8 and dipeptidyl peptidase 9 inhibitors, including Val-boroPro, accelerate the degradation of the NT fragment via a poorly characterized proteasome-mediated pathway, thereby releasing the inflammatory CT fragment from autoinhibition. Here, we show that the core 20S proteasome, which degrades disordered and misfolded proteins independent of ubiquitin modification, controls activation of the CARD8 inflammasome. In unstressed cells, we discovered that the 20S proteasome degrades just the NT disordered region, leaving behind the folded ZU5, UPA, and CARD domains to act as an inhibitor of inflammasome assembly. However, in Val-boroPro-stressed cells, we show the 20S proteasome degrades the entire NT fragment, perhaps due to ZU5 domain unfolding, freeing the CT fragment from autoinhibition. Taken together, these results show that the susceptibility of the CARD8 NT domain to 20S proteasome-mediated degradation controls inflammasome activation.


Subject(s)
CARD Signaling Adaptor Proteins , Inflammasomes , Proteasome Endopeptidase Complex , CARD Signaling Adaptor Proteins/metabolism , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/antagonists & inhibitors , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/metabolism , Humans , Inflammasomes/metabolism , Neoplasm Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Ubiquitins/metabolism
8.
Nat Chem Biol ; 18(5): 565-574, 2022 05.
Article in English | MEDLINE | ID: mdl-35165443

ABSTRACT

Inflammasomes are multiprotein complexes that sense intracellular danger signals and induce pyroptosis. CARD8 and NLRP1 are related inflammasomes that are repressed by the enzymatic activities and protein structures of the dipeptidyl peptidases 8 and 9 (DPP8/9). Potent DPP8/9 inhibitors such as Val-boroPro (VbP) activate both NLRP1 and CARD8, but chemical probes that selectively activate only one have not been identified. Here we report a small molecule called CQ31 that selectively activates CARD8. CQ31 inhibits the M24B aminopeptidases prolidase (PEPD) and Xaa-Pro aminopeptidase 1 (XPNPEP1), leading to the accumulation of proline-containing peptides that inhibit DPP8/9 and thereby activate CARD8. NLRP1 is distinct from CARD8 in that it directly contacts DPP8/9's active site; these proline-containing peptides, unlike VbP, do not disrupt this repressive interaction and thus do not activate NLRP1. We expect that CQ31 will now become a valuable tool to study CARD8 biology.


Subject(s)
CARD Signaling Adaptor Proteins , Inflammasomes , Aminopeptidases/metabolism , Apoptosis Regulatory Proteins/metabolism , CARD Signaling Adaptor Proteins/metabolism , Neoplasm Proteins , Proline
9.
Cell Rep ; 33(2): 108264, 2020 10 13.
Article in English | MEDLINE | ID: mdl-33053349

ABSTRACT

Several cytosolic pattern-recognition receptors (PRRs) form multiprotein complexes called canonical inflammasomes in response to intracellular danger signals. Canonical inflammasomes recruit and activate caspase-1 (CASP1), which in turn cleaves and activates inflammatory cytokines and gasdermin D (GSDMD), inducing pyroptotic cell death. Inhibitors of the dipeptidyl peptidases DPP8 and DPP9 (DPP8/9) activate both the human NLRP1 and CARD8 inflammasomes. NLRP1 and CARD8 have different N-terminal regions but have similar C-terminal regions that undergo autoproteolysis to generate two non-covalently associated fragments. Here, we show that DPP8/9 inhibition activates a proteasomal degradation pathway that targets disordered and misfolded proteins for destruction. CARD8's N terminus contains a disordered region of ∼160 amino acids that is recognized and destroyed by this degradation pathway, thereby freeing its C-terminal fragment to activate CASP1 and induce pyroptosis. Thus, CARD8 serves as an alarm to signal the activation of a degradation pathway for disordered and misfolded proteins.


Subject(s)
CARD Signaling Adaptor Proteins/chemistry , CARD Signaling Adaptor Proteins/metabolism , Inflammasomes/metabolism , Intrinsically Disordered Proteins/metabolism , Neoplasm Proteins/chemistry , Neoplasm Proteins/metabolism , Animals , Boronic Acids/pharmacology , Dipeptides/pharmacology , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/metabolism , HEK293 Cells , Humans , Lysine/metabolism , Mice , Proteolysis , Proteostasis , RAW 264.7 Cells , THP-1 Cells
10.
Cell Death Dis ; 11(8): 628, 2020 08 14.
Article in English | MEDLINE | ID: mdl-32796818

ABSTRACT

Canonical inflammasomes are innate immune signaling platforms that are formed in response to intracellular pathogen-associated signals and trigger caspase-1-dependent pyroptosis. Inflammasome formation and signaling is thought to mainly occur in myeloid cells, and in particular monocytes and macrophages. Here we show that small molecule inhibitors of dipeptidyl peptidases 8 and 9 (DPP8/9), which activate the related CARD8 and NLRP1 inflammasomes, also activate pyroptosis in human and rodent resting lymphocytes. We found that both CD4+ and CD8+ T cells were particularly sensitive to these inhibitors, although the sensitivity of T cells, like macrophages, varied considerably between species. In human T cells, we show that CARD8 mediates DPP8/9 inhibitor-induced pyroptosis. Intriguingly, although activated human T cells express the key proteins known to be required for CARD8-mediated pyroptosis, these cells were completely resistant to DPP8/9 inhibitors. Overall, these data show that resting lymphoid cells can activate at least one inflammasome, revealing additional cell types and states poised to undergo rapid pyroptotic cell death in response to danger-associated signals.


Subject(s)
CARD Signaling Adaptor Proteins/metabolism , Cell Cycle , Dipeptidases/antagonists & inhibitors , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/antagonists & inhibitors , Inflammasomes/metabolism , Lymphocytes/metabolism , Neoplasm Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , Cell Cycle/drug effects , Cells, Cultured , Dipeptidases/metabolism , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/metabolism , Humans , Lymphocyte Activation/drug effects , Lymphocytes/drug effects , Mice , NLR Proteins , Protease Inhibitors/pharmacology , Pyroptosis/drug effects , Rats
11.
Life Sci Alliance ; 3(3)2020 03.
Article in English | MEDLINE | ID: mdl-32051255

ABSTRACT

Pathogen-related signals induce a number of cytosolic pattern-recognition receptors (PRRs) to form canonical inflammasomes, which activate pro-caspase-1 and trigger pyroptotic cell death. All well-studied inflammasome-forming PRRs oligomerize with the adapter protein ASC (apoptosis-associated speck-like protein containing a CARD) to generate a large structure in the cytosol, which induces the dimerization, autoproteolysis, and activation of the pro-caspase-1 zymogen. However, several PRRs can also directly interact with pro-caspase-1 without ASC, forming smaller "ASC-independent" inflammasomes. It is currently thought that little, if any, pro-caspase-1 autoproteolysis occurs during, and is not required for, ASC-independent inflammasome signaling. Here, we show that the related human PRRs NLRP1 and CARD8 exclusively form ASC-dependent and ASC-independent inflammasomes, respectively, identifying CARD8 as the first canonical inflammasome-forming PRR that does not form an ASC-containing signaling platform. Despite their different structures, we discovered that both the NLRP1 and CARD8 inflammasomes require pro-caspase-1 autoproteolysis between the small and large catalytic subunits to induce pyroptosis. Thus, pro-caspase-1 self-cleavage is a required regulatory step for pyroptosis induced by human canonical inflammasomes.


Subject(s)
Caspase 1/metabolism , Pyroptosis/physiology , Adaptor Proteins, Signal Transducing/metabolism , Apoptosis , Apoptosis Regulatory Proteins/metabolism , CARD Signaling Adaptor Proteins/metabolism , Carrier Proteins/metabolism , HEK293 Cells , Humans , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Signal Transduction , THP-1 Cells
12.
ACS Chem Biol ; 14(11): 2424-2429, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31525884

ABSTRACT

Inflammasomes are multiprotein complexes formed in response to pathogens. NLRP1 and CARD8 are related proteins that form inflammasomes, but the pathogen-associated signal(s) and the molecular mechanisms controlling their activation have not been established. Inhibitors of the serine dipeptidyl peptidases DPP8 and DPP9 (DPP8/9) activate both NLRP1 and CARD8. Interestingly, DPP9 binds directly to NLRP1 and CARD8, and this interaction may contribute to the inhibition of NLRP1. Here, we use activity-based probes, reconstituted inflammasome assays, and mass spectrometry-based proteomics to further investigate the DPP9-CARD8 interaction. We show that the DPP9-CARD8 interaction, unlike the DPP9-NLRP1 interaction, is not disrupted by DPP9 inhibitors or CARD8 mutations that block autoproteolysis. Moreover, wild-type, but not catalytically inactive mutant, DPP9 rescues CARD8-mediated cell death in DPP9 knockout cells. Together, this work reveals that DPP9's catalytic activity and not its binding to CARD8 restrains the CARD8 inflammasome and thus suggests the binding interaction likely serves some other biological purpose.


Subject(s)
CARD Signaling Adaptor Proteins/metabolism , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/metabolism , Inflammasomes/metabolism , Neoplasm Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism , Dipeptidases/metabolism , HEK293 Cells , Humans , Mutation , NLR Proteins , Organofluorophosphonates/metabolism , Protease Inhibitors/metabolism , Protein Binding , Protein Conformation , Signal Transduction
13.
Cell Death Dis ; 10(8): 587, 2019 08 05.
Article in English | MEDLINE | ID: mdl-31383852

ABSTRACT

Intracellular pathogenic structures or activities stimulate the formation of inflammasomes, which recruit and activate caspase-1 and trigger an inflammatory form of cell death called pyroptosis. The well-characterized mammalian inflammasome sensor proteins all detect one specific type of signal, for example double-stranded DNA or bacterial flagellin. Remarkably, NLRP1 was the first protein discovered to form an inflammasome, but the pathogenic signal that NLRP1 detects has not yet been identified. NLRP1 is highly polymorphic, even among inbred rodent strains, and it has been suggested that these diverse NLRP1 alleles may have evolved to detect entirely different stimuli. Intriguingly, inhibitors of the serine proteases DPP8 and DPP9 (DPP8/9) were recently shown to activate human NLRP1, its homolog CARD8, and several mouse NLRP1 alleles. Here, we show now that DPP8/9 inhibitors activate all functional rodent NLRP1 alleles, indicating that DPP8/9 inhibition induces a signal detected by all NLRP1 proteins. Moreover, we discovered that the NLRP1 allele sensitivities to DPP8/9 inhibitor-induced and Toxoplasma gondii-induced pyroptosis are strikingly similar, suggesting that DPP8/9 inhibition phenocopies a key activity of T. gondii. Overall, this work indicates that the highly polymorphic NLRP1 inflammasome indeed senses a specific signal like the other mammalian inflammasomes.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Alleles , Apoptosis Regulatory Proteins/genetics , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Antigens, Bacterial/pharmacology , Apoptosis Regulatory Proteins/metabolism , Bacterial Toxins/pharmacology , Boronic Acids/pharmacology , Dipeptides/pharmacology , Female , HEK293 Cells , Humans , Inflammasomes/drug effects , Inflammasomes/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Nerve Tissue Proteins/metabolism , Pyroptosis/drug effects , RAW 264.7 Cells , Rats , Rats, Sprague-Dawley , Rats, Zucker , Serine Proteinase Inhibitors/pharmacology , Toxoplasma/immunology , Transfection
14.
Science ; 364(6435): 82-85, 2019 04 05.
Article in English | MEDLINE | ID: mdl-30872531

ABSTRACT

Intracellular pathogens and danger signals trigger the formation of inflammasomes, which activate inflammatory caspases and induce pyroptosis. The anthrax lethal factor metalloprotease and small-molecule DPP8/9 inhibitors both activate the NLRP1B inflammasome, but the molecular mechanism of NLRP1B activation is unknown. In this study, we used genome-wide CRISPR-Cas9 knockout screens to identify genes required for NLRP1B-mediated pyroptosis. We discovered that lethal factor induces cell death via the N-end rule proteasomal degradation pathway. Lethal factor directly cleaves NLRP1B, inducing the N-end rule-mediated degradation of the NLRP1B N terminus and freeing the NLRP1B C terminus to activate caspase-1. DPP8/9 inhibitors also induce proteasomal degradation of the NLRP1B N terminus but not via the N-end rule pathway. Thus, N-terminal degradation is the common activation mechanism of this innate immune sensor.


Subject(s)
Antigens, Bacterial/metabolism , Apoptosis Regulatory Proteins/metabolism , Bacterial Toxins/metabolism , Inflammasomes/metabolism , Proteolysis , Pyroptosis/physiology , Animals , Apoptosis Regulatory Proteins/genetics , CRISPR-Cas Systems , Caspase 1/metabolism , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/antagonists & inhibitors , Gene Knockout Techniques , HEK293 Cells , Host-Pathogen Interactions/immunology , Humans , Immunity, Innate , Mice , Proteasome Endopeptidase Complex/metabolism , Pyroptosis/genetics , RAW 264.7 Cells , Serine Proteinase Inhibitors/pharmacology , THP-1 Cells , Ubiquitin-Protein Ligases/genetics
15.
Oncotarget ; 7(15): 20669-79, 2016 Apr 12.
Article in English | MEDLINE | ID: mdl-26942696

ABSTRACT

The Signal Transducer and Activator of Transcription 3 (STAT3) oncogene is a master regulator of many human cancers, and a well-recognized target for therapeutic intervention. A well known STAT3 inhibitor, S3I-201 (NSC 74859), is hypothesized to block STAT3 function in cancer cells by binding the STAT3 SH2 domain and disrupt STAT3 protein complexation events. In this study, liquid chromatography tandem mass spectrometry analysis revealed that STAT3, in the presence of S3I-201, showed a minimum of five specific sites of modification, cysteine's 108, 259, 367, 542, and 687. Moreover, a prepared fluorescently labeled chemical probe of S3I-201 (DB-6-055) revealed that S3I-201 non-specifically and globally alkylated intracellular proteins at concentrations consistent with S3I-201's reported IC50. These data are consistent with the hypothesis that S3I-201 is a sub-optimal probe for interrogating STAT3-related cell biology.


Subject(s)
Alkylating Agents/pharmacology , Benzenesulfonates/pharmacology , Breast Neoplasms/drug therapy , STAT3 Transcription Factor/antagonists & inhibitors , Aminosalicylic Acids/pharmacology , Apoptosis/drug effects , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Female , Humans , Tumor Cells, Cultured
16.
Mol Cancer Ther ; 15(5): 794-805, 2016 05.
Article in English | MEDLINE | ID: mdl-26873728

ABSTRACT

Constitutively activated STAT3 protein has been found to be a key regulator of pancreatic cancer and a target for molecular therapeutic intervention. In this study, PG-S3-001, a small molecule derived from the SH-4-54 class of STAT3 inhibitors, was found to inhibit patient-derived pancreatic cancer cell proliferation in vitro and in vivo in the low micromolar range. PG-S3-001 binds the STAT3 protein potently, Kd = 324 nmol/L by surface plasmon resonance, and showed no effect in a kinome screen (>100 cancer-relevant kinases). In vitro studies demonstrated potent cell killing as well as inhibition of STAT3 activation in pancreatic cancer cells. To better model the tumor and its microenvironment, we utilized three-dimensional (3D) cultures of patient-derived pancreatic cancer cells in the absence and presence of cancer-associated fibroblasts (CAF). In this coculture model, inhibition of tumor growth is maintained following STAT3 inhibition in the presence of CAFs. Confocal microscopy was used to verify tumor cell death following treatment of 3D cocultures with PG-S3-001. The 3D model was predictive of in vivo efficacy as significant tumor growth inhibition was observed upon administration of PG-S3-001. These studies showed that the inhibition of STAT3 was able to impact the survival of tumor cells in a relevant 3D model, as well as in a xenograft model using patient-derived cells. Mol Cancer Ther; 15(5); 794-805. ©2016 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , STAT3 Transcription Factor/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Disease Models, Animal , Female , Humans , Ligands , Male , Models, Molecular , Molecular Conformation , Pancreatic Neoplasms/drug therapy , Phosphorylation , Protein Binding , STAT3 Transcription Factor/chemistry , Signal Transduction/drug effects , Structure-Activity Relationship , Xenograft Model Antitumor Assays , src Homology Domains
17.
ACS Med Chem Lett ; 6(9): 982-6, 2015 Sep 10.
Article in English | MEDLINE | ID: mdl-26396684

ABSTRACT

PTP1B is a master regulator in the insulin and leptin metabolic pathways. Hyper-activated PTP1B results in insulin resistance and is viewed as a key factor in the onset of type II diabetes and obesity. Moreover, inhibition of PTP1B expression in cancer cells dramatically inhibits cell growth in vitro and in vivo. Herein, we report the computationally guided optimization of a salicylic acid-based PTP1B inhibitor 6, identifying new and more potent bidentate PTP1B inhibitors, such as 20h, which exhibited a > 4-fold improvement in activity. In CHO-IR cells, 20f, 20h, and 20j suppressed PTP1B activity and restored insulin receptor phosphorylation levels. Notably, 20f, which displayed a 5-fold selectivity for PTP1B over the closely related PTPσ protein, showed no inhibition of PTP-LAR, PRL2 A/S, MKPX, or papain. Finally, 20i and 20j displayed nanomolar inhibition of PTPσ, representing interesting lead compounds for further investigation.

18.
Expert Opin Ther Pat ; 25(12): 1397-421, 2015.
Article in English | MEDLINE | ID: mdl-26394986

ABSTRACT

INTRODUCTION: The clinical utility of effective direct STAT inhibitors, particularly STAT3 and STAT5, for treating cancer and other diseases is well studied and known. AREAS COVERED: This review will highlight the STAT inhibitor patent literature from 2011 to 2015 inclusive. Emphasis will be placed on inhibitors of the STAT3, STAT5a/b, and STAT1 proteins for cancer treatment. The review will, where suitably investigated, describe the mode and the site of inhibition, list indications that were evaluated, and rank the inhibitor's relative potency among compounds in the same class. The reader will gain an understanding of the diverse set of approaches, used both in academia and industry, to target STAT proteins. EXPERT OPINION: There is still much work to be done to directly target the STAT3 and STAT5 proteins. As yet, there is still no direct STAT3 inhibitor in the clinic. While the SH2 domain remains a popular target for therapeutic intervention, the DNA-binding domain and N-terminal region are now attracting attention as possible sites for inhibition. Multiple putative STAT3 and STAT5 inhibitors have now been patented across a broad spectrum of chemotypes, each with their own advantages and limitations.


Subject(s)
STAT1 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/antagonists & inhibitors , STAT5 Transcription Factor/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , Drug Design , Humans , Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/pathology , Patents as Topic
19.
Bioorg Med Chem ; 21(17): 5618-28, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23810672

ABSTRACT

A focused library of hetero-trisubstituted purines was developed for improving the cell penetrating and biological efficacy of a series of anti-Stat3 protein inhibitors. From this SAR study, lead agent 22e was identified as being a promising inhibitor of MM tumour cells (IC50's <5µM). Surprisingly, biophysical and biochemical characterization proved that 22e was not a Stat3 inhibitor. Initial screening against the kinome, prompted by the purine scaffold's history for targeting ATP binding pockets, suggests possible targeting of the JAK family kinases, as well for ABL1 (nonphosphorylated F317L) and AAK1.


Subject(s)
Adenosine/analogs & derivatives , Antineoplastic Agents/chemistry , Purines/chemistry , STAT3 Transcription Factor/antagonists & inhibitors , Sulfonamides/chemistry , Adenosine/chemical synthesis , Adenosine/chemistry , Adenosine/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Humans , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Phosphorylation/drug effects , Purines/chemical synthesis , Purines/pharmacology , STAT3 Transcription Factor/metabolism , Structure-Activity Relationship , Sulfonamides/chemical synthesis , Sulfonamides/pharmacology
20.
Expert Opin Ther Pat ; 21(1): 65-83, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21114420

ABSTRACT

IMPORTANCE OF THE FIELD: Aberrant activation of signal transducer and activator of transcription (Stat) 3, a member of the STAT family of proteins, is prevalent in numerous human cancers and is now widely recognized as a critical molecular abnormality and a master regulator of tumor processes. Thus, the identification of potent and selective Stat3 inhibitors will have a high commercial potential as anticancer drugs, given the many tumors in which Stat3 is implicated. AREAS COVERED IN THIS REVIEW: This review covers the structures and activities of direct inhibitors of Stat3 protein activity described in the patent literature since the research field's inception in 2001. The patents reviewed include peptide and peptidomimetic compounds, small molecules, oligonucleotides and platinum-based Stat3 inhibitors. WHAT THE READER WILL GAIN: Readers will gain an understanding of how Stat3 protein function has been inhibited by a wide variety of structurally diverse therapeutic compounds. Readers will learn about which classes of patented Stat3 inhibitors are most advanced toward clinical trials, and will be exposed to the proposed mechanisms of inhibition and scope of their application in treating human cancers. TAKE HOME MESSAGE: Numerous groups have shown that in vivo administration of inhibitors of activated Stat3 induce human tumor regression in xenograft models. Indeed, the growing number of preclinical studies in numerous cancer types, as well as the first Phase 0 clinical trial of a Stat3 inhibitor, suggest that Stat3 is a valid and exciting therapeutic target for molecular inhibitors.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , STAT3 Transcription Factor/antagonists & inhibitors , Animals , Drug Delivery Systems , Drug Design , Humans , Neoplasms/physiopathology , Oligonucleotides/pharmacology , Patents as Topic , Peptides/pharmacology , STAT3 Transcription Factor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...