Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
J Clin Oncol ; 40(15): 1671-1680, 2022 05 20.
Article in English | MEDLINE | ID: mdl-35180010

ABSTRACT

PURPOSE: Targeting the BCL-XL pathway has demonstrated the ability to overcome Janus kinase inhibitor resistance in preclinical models. This phase II trial investigated the efficacy and safety of adding BCL-XL/BCL-2 inhibitor navitoclax to ruxolitinib therapy in patients with myelofibrosis with progression or suboptimal response to ruxolitinib monotherapy (ClinicalTrials.gov identifier: NCT03222609). METHODS: Thirty-four adult patients with intermediate-/high-risk myelofibrosis who had progression or suboptimal response on stable ruxolitinib dose (≥ 10 mg twice daily) were administered navitoclax at 50 mg once daily starting dose, followed by escalation to a maximum of 300 mg once daily in once in weekly increments (if platelets were ≥ 75 × 109/L). The primary end point was ≥ 35% spleen volume reduction (SVR35) from baseline at week 24. Secondary end points included ≥ 50% reduction in total symptom score (TSS50) from baseline at week 24, hemoglobin improvement, change in bone marrow fibrosis (BMF) grade, and safety. RESULTS: High molecular risk mutations were identified in 58% of patients, and 52% harbored ≥ 3 mutations. SVR35 was achieved by 26.5% of patients at week 24, and by 41%, at any time on study, with an estimated median duration of SVR35 of 13.8 months. TSS50 was achieved by 30% (6 of 20) of patients at week 24, and BMF improved by 1-2 grades in 33% (11 of 33) of evaluable patients. Anemia response was achieved by 64% (7 of 11), including one patient with baseline transfusion dependence. Median overall survival was not reached with a median follow-up of 21.6 months. The most common adverse event was reversible thrombocytopenia without clinically significant bleeding (88%). CONCLUSION: The addition of navitoclax to ruxolitinib in patients with persistent or progressive myelofibrosis resulted in durable SVR35, improved TSS, hemoglobin response, and BMF. Further investigation is underway to qualify the potential for disease modification.


Subject(s)
Primary Myelofibrosis , Adult , Aniline Compounds , Humans , Nitriles/therapeutic use , Primary Myelofibrosis/drug therapy , Pyrazoles/adverse effects , Pyrimidines/therapeutic use , Sulfonamides , Treatment Outcome
2.
CRISPR J ; 2: 230-245, 2019 08.
Article in English | MEDLINE | ID: mdl-31436504

ABSTRACT

Genome-wide CRISPR-Cas9 essentiality screening represents a powerful approach to identify genetic vulnerabilities in cancer cells. Here, we applied this technology and designed a strategy to identify target genes that are synthetic lethal (SL) with von Hippel-Lindau (VHL) tumor suppressor gene. Inactivation of VHL has been frequently found in clear cell renal cell carcinoma. Its SL partners serve as potential drug targets for the development of targeted cancer therapies. We performed parallel genome-wide CRISPR screens in two pairs of isogenic clear cell renal cell carcinoma cell lines that differ only in the VHL status. Comparative analyses of screening results not only confirmed a well-known role for mTOR signaling in renal carcinoma, but also identified DNA damage response and selenocysteine biosynthesis pathways as novel SL targets in VHL-inactivated cancer cells. Follow-up studies provided cellular and mechanistic insights into SL interactions of these pathway genes with the VHL gene. Our CRISPR and RNA-seq datasets provide a rich resource for future investigation of the function of the VHL tumor suppressor protein. Our work demonstrates the efficiency of CRISPR-based synthetic lethality screening in human isogenic cell pairs. Similar strategies could be employed to unveil SL partners with other oncogenic drivers.


Subject(s)
DNA Repair , Selenocysteine/biosynthesis , Signal Transduction , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , CRISPR-Cas Systems , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Cell Line, Tumor , DNA Damage , Gene Editing , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Sequence Analysis, RNA , Von Hippel-Lindau Tumor Suppressor Protein/genetics , von Hippel-Lindau Disease/genetics , von Hippel-Lindau Disease/metabolism
3.
Clin Cancer Res ; 25(21): 6309-6319, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31420359

ABSTRACT

PURPOSE: Bromodomain and extraterminal (BET) proteins play important roles in transcriptional regulation relevant to cancer pathogenesis, and therapeutic targeting/inhibition of BET causes apoptosis of cancer cells in vitro. In this first-in-human study of the pan-BET inhibitor mivebresib (ABBV-075), the safety profile, MTD, and recommended phase II dose (RP2D) were determined in patients with advanced solid tumors. PATIENTS AND METHODS: A 3 + 3 dose escalation for different mivebresib dosing schedules [daily, Monday/Wednesday/Friday (M-W-F), 4 days on/3 off (4/7)] was followed by dose expansion in patients with prostate cancer. Endpoints were safety, tolerability, pharmacokinetics, and preliminary antitumor activity. RESULTS: Seventy-two patients with solid tumors (14% uveal melanoma; 11% colorectal; 11% breast; 8% pancreatic; 7% head/neck; 49% others) were treated with mivebresib during dose escalation, and 12 additional patients with prostate cancer in expansion cohort. Most common treatment-emergent adverse events (TEAE) related to mivebresib were dysgeusia (49%), thrombocytopenia (48%), fatigue (26%), and nausea (25%). Most common grade 3/4 TEAEs related to mivebresib were thrombocytopenia (35%) and anemia (6%). Dose-limiting toxicities included thrombocytopenia (2 mg daily; 4.5 mg M-W-F), gastrointestinal bleed (2 mg daily), hypertension (2-3 mg 4/7), fatigue, decreased appetite, and aspartate aminotransferase elevation (4 mg M-W-F). Of 61 evaluable patients from dose escalation, 26 (43%) had stable disease and 35 (57%) had progressive disease. Median progression-free survival was 1.8 months (95% confidence interval, 1.8-1.9). CONCLUSIONS: On the basis of safety and tolerability, mivebresib RP2D is 1.5 mg for the daily schedule, 2.5 mg for 4/7, and 3 mg for M-W-F. Mivebresib has a tolerable safety profile, and stable disease was observed in some patients with malignant solid tumors.


Subject(s)
Antineoplastic Agents/administration & dosage , Neoplasms/drug therapy , Pyridones/administration & dosage , Sulfonamides/administration & dosage , Adult , Aged , Antineoplastic Agents/adverse effects , Cohort Studies , Drug Administration Schedule , Female , Humans , Male , Maximum Tolerated Dose , Middle Aged , Nausea/chemically induced , Nausea/pathology , Pyridones/adverse effects , Recurrence , Sulfonamides/adverse effects
4.
J Med Chem ; 61(15): 6647-6657, 2018 Aug 09.
Article in English | MEDLINE | ID: mdl-30004704

ABSTRACT

IDH1 plays a critical role in a number of metabolic processes and serves as a key source of cytosolic NADPH under conditions of cellular stress. However, few inhibitors of wild-type IDH1 have been reported. Here we present the discovery and biochemical characterization of two novel inhibitors of wild-type IDH1. In addition, we present the first ligand-bound crystallographic characterization of these novel small molecule IDH1 binding pockets. Importantly, the NADPH competitive α,ß-unsaturated enone 1 makes a unique covalent linkage through active site H315. As few small molecules have been shown to covalently react with histidine residues, these data support the potential utility of an underutilized strategy for reversible covalent small molecule design.


Subject(s)
Drug Discovery , Enzyme Inhibitors/pharmacology , Histidine , Isocitrate Dehydrogenase/antagonists & inhibitors , Isocitrate Dehydrogenase/chemistry , Cell Line, Tumor , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Humans , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , Ligands , Molecular Docking Simulation , Mutation , Protein Conformation , Structure-Activity Relationship
5.
J Biol Chem ; 292(13): 5405-5417, 2017 03 31.
Article in English | MEDLINE | ID: mdl-28188292

ABSTRACT

Cytosolic DNA can elicit beneficial as well as undesirable immune responses. For example, viral or microbial DNA triggers cell-intrinsic immune responses to defend against infections, whereas aberrant cytosolic accumulation of self-DNA results in pathological conditions, such as autoimmunity. Given the importance of these DNA-provoked responses, a better understanding of their molecular mechanisms is needed. Cytosolic DNA engages stimulator of interferon genes (STING) to activate TANK-binding kinase 1 (TBK1), which subsequently phosphorylates the transcription factor interferon regulatory factor 3 (IRF3) to promote interferon expression. Recent studies have reported that additional transcription factors, including nuclear factor κB (NF-κB) and signal transducer and activator of transcription 6 (STAT6), are also activated by cytosolic DNA, suggesting that cytosolic DNA-induced gene expression is orchestrated by multiple factors. Here we show that cytosolic DNA activates STAT3, another member of the STAT family, via an autocrine mechanism involving interferon ß (IFNß) and IL-6. Additionally, we observed a novel cytosolic DNA-induced phosphorylation at serine 754 in the transactivation domain of STAT3. Upon cytosolic DNA stimulation, Ser754 is directly phosphorylated by TBK1 in a STING-dependent manner. Moreover, Ser754 phosphorylation inhibits cytosolic DNA-induced STAT3 transcriptional activity and selectively reduces STAT3 target genes that are up-regulated in response to cytosolic DNA. Taken together, our results suggest that cytosolic DNA-induced STAT3 activation via IFNß and IL-6 is restrained by Ser754 phosphorylation of STAT3. Our findings reveal a new signaling axis downstream of the cytosolic DNA pathway and suggest potential interactions between innate immune responses and STAT3-driven oncogenic pathways.


Subject(s)
DNA/immunology , Protein Serine-Threonine Kinases/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Autocrine Communication , Cell Line , DNA/physiology , Gene Expression Regulation/immunology , Humans , Immunity, Innate , Interferon-beta , Interleukin-6 , Membrane Proteins/metabolism , Phosphorylation
6.
PLoS One ; 7(7): e41494, 2012.
Article in English | MEDLINE | ID: mdl-22859992

ABSTRACT

IKKε and TBK1 are noncanonical IKK family members which regulate inflammatory signaling pathways and also play important roles in oncogenesis. However, few inhibitors of these kinases have been identified. While the substrate specificity of IKKε has recently been described, the substrate specificity of TBK1 is unknown, hindering the development of high-throughput screening technologies for inhibitor identification. Here, we describe the optimal substrate phosphorylation motif for TBK1, and show that it is identical to the phosphorylation motif previously described for IKKε. This information enabled the design of an optimal TBK1/IKKε substrate peptide amenable to high-throughput screening and we assayed a 6,006 compound library that included 4,727 kinase-focused compounds to discover in vitro inhibitors of TBK1 and IKKε. 227 compounds in this library inhibited TBK1 at a concentration of 10 µM, while 57 compounds inhibited IKKε. Together, these data describe a new high-throughput screening assay which will facilitate the discovery of small molecule TBK1/IKKε inhibitors possessing therapeutic potential for both inflammatory diseases and cancer.


Subject(s)
High-Throughput Screening Assays , I-kappa B Kinase/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Protein Serine-Threonine Kinases/antagonists & inhibitors , Amino Acid Motifs , Amino Acid Sequence , Enzyme Assays , HEK293 Cells , Humans , I-kappa B Kinase/chemistry , Kinetics , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Library , Phosphorylation , Protein Serine-Threonine Kinases/chemistry , Small Molecule Libraries , Substrate Specificity
7.
Mol Cell Biol ; 32(13): 2479-89, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22547678

ABSTRACT

Despite their homology, IκB kinase α (IKKα) and IKKß have divergent roles in NF-κB signaling. IKKß strongly activates NF-κB while IKKα can downregulate NF-κB under certain circumstances. Given this, identifying independent substrates for these kinases could help delineate their divergent roles. Peptide substrate array technology followed by bioinformatic screening identified TRAF4 as a substrate for IKKα. Like IKKα, TRAF4 is atypical within its family because it is the only TRAF family member to negatively regulate innate immune signaling. IKKα's phosphorylation of serine-426 on TRAF4 was required for this negative regulation. Binding to the Crohn's disease susceptibility protein, NOD2, is required for TRAF4 phosphorylation and subsequent inhibition of NOD2 signaling. Structurally, serine-426 resides within an exaggerated ß-bulge in TRAF4 that is not present in the other TRAF proteins, and phosphorylation of this site provides a structural basis for the atypical function of TRAF4 and its atypical role in NOD2 signaling.


Subject(s)
I-kappa B Kinase/metabolism , Immunity, Innate , TNF Receptor-Associated Factor 4/metabolism , Amino Acid Sequence , Animals , Base Sequence , Binding Sites , Cell Line , Crohn Disease/genetics , Crohn Disease/immunology , Crohn Disease/metabolism , DNA Primers/genetics , Down-Regulation , HCT116 Cells , HEK293 Cells , Humans , I-kappa B Kinase/genetics , I-kappa B Kinase/immunology , Mice , Models, Molecular , Molecular Sequence Data , NF-kappa B/metabolism , Nod2 Signaling Adaptor Protein/genetics , Nod2 Signaling Adaptor Protein/metabolism , Peptide Library , Phosphorylation , Protein Stability , Protein Structure, Tertiary , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid , Serine/chemistry , Signal Transduction/immunology , Substrate Specificity , TNF Receptor-Associated Factor 4/chemistry , TNF Receptor-Associated Factor 4/immunology
8.
Cancer Res ; 72(13): 3260-9, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22552288

ABSTRACT

The phosphoinositide 3-kinase (PI3K) pathway is one of the most commonly misregulated signaling pathways in human cancers, but its impact on the tumor microenvironment has not been considered as deeply as its autonomous impact on tumor cells. In this study, we show that NF-κB is activated by the two most common PI3K mutations, PIK3CA E545K and H1047R. We found that markers of NF-κB are most strongly upregulated under conditions of growth factor deprivation. Gene expression analysis conducted on cells deprived of growth factors identified the repertoire of genes altered by oncogenic PI3K mutations following growth factor deprivation. This gene set most closely correlated with gene signatures from claudin-low and basal-like breast tumors, subtypes frequently exhibiting constitutive PI3K/Akt activity. An NF-κB-dependent subset of genes driven by oncogenic PI3K mutations was also identified that encoded primarily secreted proteins, suggesting a paracrine role for this gene set. Interestingly, while NF-κB activated by oncogenes such as Ras and EGF receptor leads to cell-autonomous effects, abrogating NF-κB in PI3K-transformed cells did not decrease proliferation or induce apoptosis. However, conditioned media from PI3K mutant-expressing cells led to increased STAT3 activation in recipient THP-1 monocytes or normal epithelial cells in a NF-κB and interleukin-6-dependent manner. Together, our findings describe a PI3K-driven, NF-κB-dependent transcriptional profile that may play a critical role in promoting a microenvironment amenable to tumor progression. These data also indicate that NF-κB plays diverse roles downstream from different oncogenic signaling pathways.


Subject(s)
Cytokines/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Mutation , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/genetics , Blotting, Western , Cell Line, Tumor , Cell Proliferation , Cell Survival , Humans , Oligonucleotide Array Sequence Analysis , Signal Transduction
9.
Mol Cell ; 45(6): 719-30, 2012 Mar 30.
Article in English | MEDLINE | ID: mdl-22342344

ABSTRACT

The IκB kinase (IKK) pathway is an essential mediator of inflammatory, oncogenic, and cell stress pathways. Recently IKK was shown to be essential for autophagy induction in mammalian cells independent of its ability to regulate NF-κB, but the mechanism by which this occurs is unclear. Here we demonstrate that the p85 regulatory subunit of PI3K is an IKK substrate, phosphorylated at S690 in vitro and in vivo in response to cellular starvation. Cells expressing p85 S690A or inhibited for IKK activity exhibit increased Akt activity following cell starvation, demonstrating that p85 phosphorylation is required for starvation-induced PI3K feedback inhibition. S690 is in a conserved region of the p85 cSH2 domain, and IKK-mediated phosphorylation of this site results in decreased affinity for tyrosine-phosphorylated proteins and decreased PI3K membrane localization. Finally, leucine deprivation is shown to be necessary and sufficient for starvation-induced, IKK-mediated p85 phosphorylation and PI3K feedback inhibition.


Subject(s)
I-kappa B Kinase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Starvation/metabolism , Amino Acid Sequence , Amino Acids/metabolism , Animals , Cell Line , Class Ia Phosphatidylinositol 3-Kinase/metabolism , Conserved Sequence , Feedback, Physiological , Fibroblasts/metabolism , Humans , I-kappa B Kinase/genetics , Leucine/metabolism , Mice , Mice, Inbred C57BL , Phosphorylation , Phosphotyrosine/metabolism , src Homology Domains
10.
Biochem J ; 425(2): 445-53, 2009 Dec 23.
Article in English | MEDLINE | ID: mdl-19857203

ABSTRACT

PKNs form a subfamily of the AGC serine/threonine protein kinases, and have a catalytic domain homologous with that of PKC (protein kinase C) in the C-terminal region and three characteristic ACC (antiparallel coiled-coil) domain repeats in the N-terminal region. The preferred peptide phosphorylation motif for PKNs determined by a combinatorial peptide library method was highly similar to that of PKCs within a 10-amino-acid stretch. Previously reported PKN inhibitory compounds also inhibit PKCs to a similar extent, and no PKN selective inhibitors have been commercially available. We have identified a 15-amino-acid peptide inhibitor of PKNs based on amino acids 485-499 of the C-terminal region of the C2-like domain of PKN1. This peptide, designated as PRL, selectively inhibits the kinase activity of all isoforms of PKN (Ki=0.7 muM) towards a peptide substrate, as well as autophosphorylation activity of PKN in vitro, in contrast with PKC. Reversible conjugation by a disulfide bond of a carrier peptide bearing a penetration accelerating sequence to PRL, facilitated the cellular uptake of this peptide and significantly inhibited phosphorylation of tau by PKN1 at the PKN1-specific phosphorylation site in vivo. This peptide may serve as a valuable tool for investigating PKN activation and PKN-mediated responses.


Subject(s)
Peptide Fragments/pharmacology , Protein Kinase C/antagonists & inhibitors , Cell Membrane Permeability , HeLa Cells , Humans , Peptide Fragments/chemical synthesis , Peptide Fragments/pharmacokinetics , Peptide Library , Phosphorylation/drug effects , Protein Isoforms , Substrate Specificity
11.
Biochem J ; 424(1): 47-60, 2009 Oct 23.
Article in English | MEDLINE | ID: mdl-19740074

ABSTRACT

The LRRK2 (leucine-rich repeat protein kinase-2) is mutated in a significant number of Parkinson's disease patients, but little is known about its regulation and function. A common mutation changing Gly2019 to serine enhances catalytic activity, suggesting that small-molecule inhibitors might have utility in treating Parkinson's disease. We employed various approaches to explore the substrate-specificity requirements of LRRK2 and elaborated a peptide substrate termed Nictide, that had 20-fold lower Km and nearly 2-fold higher Vmax than the widely deployed LRRKtide substrate. We demonstrate that LRRK2 has marked preference for phosphorylating threonine over serine. We also observed that several ROCK (Rho kinase) inhibitors such as Y-27632 and H-1152, suppressed LRRK2 with similar potency to which they inhibited ROCK2. In contrast, GSK429286A, a selective ROCK inhibitor, did not significantly inhibit LRRK2. We also identified a mutant LRRK2[A2016T] that was normally active, but resistant to H-1152 and Y-27632, as well as sunitinib, a structurally unrelated multikinase inhibitor that, in contrast with other compounds, suppresses LRRK2, but not ROCK. We have also developed the first sensitive antibody that enables measurement of endogenous LRRK2 protein levels and kinase activity as well as shRNA (short hairpin RNA) methods to reduce LRRK2 expression. Finally, we describe a pharmacological approach to validate whether substrates are phosphorylated by LRRK2 and use this to provide evidence that LRRK2 may not be rate-limiting for the phosphorylation of the proposed substrate moesin. The findings of the present study will aid with the investigation of LRRK2.


Subject(s)
Parkinson Disease/enzymology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Amides/pharmacology , Animals , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Mutation , Parkinson Disease/genetics , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Protein Structure, Secondary , Pyridines/pharmacology , Substrate Specificity , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/chemistry , rho-Associated Kinases/genetics , rho-Associated Kinases/metabolism
12.
Mol Cell ; 34(4): 461-72, 2009 May 14.
Article in English | MEDLINE | ID: mdl-19481526

ABSTRACT

The noncanonical IKK family member IKKepsilon is essential for regulating antiviral signaling pathways and is a recently discovered breast cancer oncoprotein. Although several IKKepsilon targets have been described, direct IKKepsilon substrates necessary for regulating cell transformation have not been identified. Here, we performed a screen for putative IKKepsilon substrates using an unbiased proteomic and bioinformatic approach. Using a positional scanning peptide library assay, we determined the optimal phosphorylation motif for IKKepsilon and used bioinformatic approaches to predict IKKepsilon substrates. Of these potential substrates, serine 418 of the tumor suppressor CYLD was identified as a likely site of IKKepsilon phosphorylation. We confirmed that CYLD is directly phosphorylated by IKKepsilon and that IKKepsilon phosphorylates serine 418 in vivo. Phosphorylation of CYLD at serine 418 decreases its deubiquitinase activity and is necessary for IKKepsilon-driven transformation. Together, these observations define IKKepsilon and CYLD as an oncogene-tumor suppressor network that participates in tumorigenesis.


Subject(s)
Breast Neoplasms , Cell Transformation, Neoplastic , I-kappa B Kinase/metabolism , Tumor Suppressor Proteins/metabolism , Amino Acid Sequence , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Deubiquitinating Enzyme CYLD , Female , Genes, Reporter , Humans , I-kappa B Kinase/genetics , Molecular Sequence Data , NF-kappa B/genetics , NF-kappa B/metabolism , Phosphorylation , Sequence Alignment , Serine/metabolism , Substrate Specificity , TNF Receptor-Associated Factor 2/genetics , TNF Receptor-Associated Factor 2/metabolism , Tumor Suppressor Proteins/genetics
13.
Mol Cell Biol ; 27(21): 7451-61, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17709380

ABSTRACT

Misregulation of NF-kappaB signaling leads to infectious, inflammatory, or autoimmune disorders. IkappaB kinase beta (IKKbeta) is an essential activator of NF-kappaB and is known to phosphorylate the NF-kappaB inhibitor, IkappaBalpha, allowing it to undergo ubiquitin-mediated proteasomal degradation. However, beyond IkappaBalpha, few additional IKKbeta substrates have been identified. Here we utilize a peptide library and bioinformatic approach to predict likely substrates of IKKbeta. This approach predicted Ser381 of the K63 deubiquitinase A20 as a likely site of IKKbeta phosphorylation. While A20 is a known negative regulator of innate immune signaling pathways, the mechanisms regulating the activity of A20 are poorly understood. We show that IKKbeta phosphorylates A20 in vitro and in vivo at serine 381, and we further show that this phosphorylation event increases the ability of A20 to inhibit the NF-kappaB signaling pathway. Phosphorylation of A20 by IKKbeta thus represents part of a novel feedback loop that regulates the duration of NF-kappaB signaling following activation of innate immune signaling pathways.


Subject(s)
Feedback, Physiological , I-kappa B Kinase/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , NF-kappa B/metabolism , Nuclear Proteins/metabolism , Algorithms , Amino Acid Motifs , Amino Acid Sequence , Animals , Antibodies, Phospho-Specific/pharmacology , Cell Line , Cysteine Endopeptidases , Humans , I-kappa B Kinase/chemistry , Lipopolysaccharides/pharmacology , Mice , Models, Biological , Molecular Sequence Data , NF-kappa B/antagonists & inhibitors , Phosphorylation/drug effects , Serine/metabolism , Substrate Specificity/drug effects , Time Factors , Tumor Necrosis Factor alpha-Induced Protein 3
14.
Mol Cell Biol ; 27(17): 6012-25, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17562858

ABSTRACT

K63 polyubiquitin chains spatially and temporally link innate immune signaling effectors such that cytokine release can be coordinated. Crohn's disease is a prototypical inflammatory disorder in which this process may be faulty as the major Crohn's disease-associated protein, NOD2 (nucleotide oligomerization domain 2), regulates the formation of K63-linked polyubiquitin chains on the I kappa kinase (IKK) scaffolding protein, NEMO (NF-kappaB essential modifier). In this work, we study these K63-linked ubiquitin networks to begin to understand the biochemical basis for the signaling cross talk between extracellular pathogen Toll-like receptors (TLRs) and intracellular pathogen NOD receptors. This work shows that TLR signaling requires the same ubiquitination event on NEMO to properly signal through NF-kappaB. This ubiquitination is partially accomplished through the E3 ubiquitin ligase TRAF6. TRAF6 is activated by NOD2, and this activation is lost with a major Crohn's disease-associated NOD2 allele, L1007insC. We further show that TRAF6 and NOD2/RIP2 share the same biochemical machinery (transforming growth factor beta-activated kinase 1 [TAK1]/TAB/Ubc13) to activate NF-kappaB, allowing TLR signaling and NOD2 signaling to synergistically augment cytokine release. These findings suggest a biochemical mechanism for the faulty cytokine balance seen in Crohn's disease.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Nod2 Signaling Adaptor Protein/metabolism , Polyubiquitin/metabolism , Signal Transduction/physiology , Toll-Like Receptors/metabolism , Animals , Crohn Disease/immunology , Enzyme Activation , Genes, Reporter , Humans , Immunity, Innate/physiology , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Knockout , NF-kappa B/genetics , NF-kappa B/metabolism , Nod2 Signaling Adaptor Protein/genetics , Polyubiquitin/genetics , RNA, Small Interfering , Receptor-Interacting Protein Serine-Threonine Kinase 2/genetics , Receptor-Interacting Protein Serine-Threonine Kinase 2/metabolism , TNF Receptor-Associated Factor 6/genetics , TNF Receptor-Associated Factor 6/metabolism , Toll-Like Receptors/genetics
15.
Mol Cell ; 25(5): 689-702, 2007 Mar 09.
Article in English | MEDLINE | ID: mdl-17349956

ABSTRACT

Progression through meiosis in yeast is governed by the cyclin-dependent kinase Cdk1, in concert with a related kinase called Ime2. It remains unclear how these kinases collaborate to meet the unique demands of meiotic progression. We demonstrate that Ime2 and Cdk1 phosphorylate an overlapping substrate set and that the two kinases overlap functionally as inhibitors of the ubiquitin ligase APC(Cdh1) and replication origin licensing. Surprisingly, Ime2 phosphorylates Cdk1 substrates at distinct phosphorylation sites that are highly resistant to dephosphorylation by the phosphatase Cdc14. We propose that Ime2-dependent phosphorylation of a subset of cell-cycle proteins limits the effects of Cdc14 in meiosis.


Subject(s)
CDC2 Protein Kinase/metabolism , Cell Cycle Proteins/metabolism , Evolution, Molecular , Meiosis , Protein Kinases/metabolism , Protein Tyrosine Phosphatases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/enzymology , Active Transport, Cell Nucleus , Amino Acid Sequence , CDC2 Protein Kinase/chemistry , Cell Cycle Proteins/chemistry , Cell Nucleus/metabolism , Chromosomal Proteins, Non-Histone , Consensus Sequence , DNA Replication , Fungal Proteins/metabolism , Intracellular Signaling Peptides and Proteins , Models, Biological , Molecular Sequence Data , Phosphates/metabolism , Phosphoprotein Phosphatases/metabolism , Phosphorylation , Protein Kinases/chemistry , Protein Serine-Threonine Kinases , Saccharomyces cerevisiae Proteins/chemistry , Substrate Specificity
16.
Nat Protoc ; 1(1): 375-9, 2006.
Article in English | MEDLINE | ID: mdl-17406259

ABSTRACT

We describe here a protocol for determining the activity of protein kinases on a large set of peptide substrates. Biotin-tagged peptides are arrayed in multiwell plates and incubated in solution with the kinase of interest and radiolabeled ATP. Reactions are then spotted simultaneously onto a streptavidin membrane, which is washed, dried, and analyzed by autoradiography or phosphor imaging. Differences in the extent of radiolabel incorporation into the various peptide substrates provide a measure of the sequence specificity of the kinase. This approach is a faster, more sensitive, and more generally applicable method for determining kinase phosphorylation motifs than older peptide library screening approaches based on Edman sequencing. The procedure is readily adaptable to other applications that require parallel processing of many kinase reactions, such as screening for small molecule inhibitors. In the format described here, preparation of stock plates prior to running the reactions will require about 4 days. Afterwards, the protocol takes approximately 6 hours to perform.


Subject(s)
Molecular Probe Techniques , Protein Kinases/metabolism , Biotin/analysis , Isotope Labeling/methods , Peptides/metabolism , Solutions , Substrate Specificity
17.
Proc Natl Acad Sci U S A ; 102(23): 8114-9, 2005 Jun 07.
Article in English | MEDLINE | ID: mdl-15928075

ABSTRACT

Kinases catalyze the phosphorylation of proteins, lipids, sugars, nucleosides, and other important cellular metabolites and play key regulatory roles in all aspects of eukaryotic cell physiology. Here, we describe the mining of public databases to collect the sequence information of all identified human kinase genes and the cloning of the corresponding ORFs. We identified 663 genes, 511 encoding protein kinases, and 152 encoding nonprotein kinases. We describe the successful cloning and sequence verification of 270 of these genes. Subcloning of this gene set in mammalian expression vectors and their use in high-throughput cell-based screens allowed the validation of the clones at the level of expression and the identification of previously uncharacterized modulators of the survivin promoter. Moreover, expressions of the kinase genes in bacteria, followed by autophosphorylation assays, identified 21 protein kinases that showed autocatalytic activity. The work described here will facilitate the functional assaying of this important gene family in phenotypic screens and their use in biochemical and structural studies.


Subject(s)
Cloning, Molecular , Computational Biology , Databases, Genetic , Protein Kinases/genetics , Protein Kinases/metabolism , Animals , Biological Assay , Catalysis , Cell Line , Cells/metabolism , Gene Expression Regulation , Genetic Vectors/genetics , Humans , Molecular Sequence Data , Phenotype , Phosphorylation , Plasmids/genetics , Promoter Regions, Genetic/genetics , Protein Kinases/isolation & purification , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Reproducibility of Results
18.
Nat Methods ; 1(1): 27-9, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15782149

ABSTRACT

Selection of target substrates by protein kinases is strongly influenced by the amino acid sequence surrounding the phosphoacceptor site. Identification of the preferred peptide phosphorylation motif for a given kinase permits the production of efficient peptide substrates and greatly simplifies the mapping of phosphorylation sites in protein substrates. Here we describe a combinatorial peptide library method that allows rapid generation of phosphorylation motifs for serine/threonine kinases.


Subject(s)
Microfluidic Analytical Techniques/methods , Peptide Library , Protein Interaction Mapping/methods , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Sequence Analysis, Protein/methods , Amino Acid Motifs , Binding Sites , Enzyme Activation , Phosphorylation , Protein Binding , Protein Serine-Threonine Kinases/analysis , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...