Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Cell Rep ; 42(12): 113535, 2023 12 26.
Article in English | MEDLINE | ID: mdl-38060450

ABSTRACT

The phosphoinositide 3-kinase p110α is an essential mediator of insulin signaling and glucose homeostasis. We interrogated the human serine, threonine, and tyrosine kinome to search for novel regulators of p110α and found that the Hippo kinases phosphorylate p110α at T1061, which inhibits its activity. This inhibitory state corresponds to a conformational change of a membrane-binding domain on p110α, which impairs its ability to engage membranes. In human primary hepatocytes, cancer cell lines, and rodent tissues, activation of the Hippo kinases MST1/2 using forskolin or epinephrine is associated with phosphorylation of T1061 and inhibition of p110α, impairment of downstream insulin signaling, and suppression of glycolysis and glycogen synthesis. These changes are abrogated when MST1/2 are genetically deleted or inhibited with small molecules or if the T1061 is mutated to alanine. Our study defines an inhibitory pathway of PI3K signaling and a link between epinephrine and insulin signaling.


Subject(s)
Protein Serine-Threonine Kinases , Humans , Animals , Mice , Cell Line , Mice, Inbred C57BL , Male , Female , Epinephrine/pharmacology , Enzyme Activation/drug effects , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Phosphatidylinositols/chemistry , Phosphatidylinositols/metabolism , Gene Deletion , Colforsin/pharmacology , Insulin/metabolism , Phosphorylation/drug effects , Hippo Signaling Pathway/drug effects , Hippo Signaling Pathway/genetics
2.
Bioorg Med Chem Lett ; 39: 127854, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33631370

ABSTRACT

p300 and CREB-binding protein (CBP) are essential for a multitude of cellular processes. Dysregulation of p300/CBP histone acetyltransferase activity is linked to a broad spectrum of human diseases including cancers. A novel drug-like spirohydantoin (21) has been discovered as a selective orally bioavailable inhibitor of p300/CBP histone acetyltransferase. Lead compound 21 is more potent than the first-in-class lead A-485 in both enzymatic and cellular assays and lacks the off-target inhibition of dopamine and serotonin transporters, that was observed with A-485.


Subject(s)
CREB-Binding Protein/antagonists & inhibitors , Drug Discovery , E1A-Associated p300 Protein/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Hydantoins/pharmacology , Spiro Compounds/pharmacology , Administration, Oral , Biological Availability , CREB-Binding Protein/metabolism , Dose-Response Relationship, Drug , E1A-Associated p300 Protein/metabolism , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/metabolism , Humans , Hydantoins/administration & dosage , Hydantoins/metabolism , Molecular Structure , Spiro Compounds/administration & dosage , Spiro Compounds/metabolism , Structure-Activity Relationship
3.
Nature ; 558(7710): E1, 2018 06.
Article in English | MEDLINE | ID: mdl-29769713

ABSTRACT

In the originally published version of this Letter, the authors Arthur F. Kluge, Michael A. Patane and Ce Wang were inadvertently omitted from the author list. Their affiliations are: I-to-D, Inc., PO Box 6177, Lincoln, Massachusetts 01773, USA (A.F.K.); Mitobridge, Inc. 1030 Massachusetts Avenue, Cambridge, Massachusetts 02139, USA (M.A.P.); and China Novartis Institutes for BioMedical Research, No. 4218 Jinke Road, Zhangjiang Hi-Tech Park, Pudong District, Shanghai 201203, China (C.W.). These authors contributed to the interpretation of results and design of compounds. In addition, author 'Edward A. Kesicki' was misspelled as 'Ed Kesicki'. These errors have been corrected online.

4.
Cell ; 174(1): 231-244.e12, 2018 06 28.
Article in English | MEDLINE | ID: mdl-29804834

ABSTRACT

The acetyltransferases CBP and p300 are multifunctional transcriptional co-activators. Here, we combined quantitative proteomics with CBP/p300-specific catalytic inhibitors, bromodomain inhibitor, and gene knockout to reveal a comprehensive map of regulated acetylation sites and their dynamic turnover rates. CBP/p300 acetylates thousands of sites, including signature histone sites and a multitude of sites on signaling effectors and enhancer-associated transcriptional regulators. Time-resolved acetylome analyses identified a subset of CBP/p300-regulated sites with very rapid (<30 min) acetylation turnover, revealing a dynamic balance between acetylation and deacetylation. Quantification of acetylation, mRNA, and protein abundance after CBP/p300 inhibition reveals a kinetically competent network of gene expression that strictly depends on CBP/p300-catalyzed rapid acetylation. Collectively, our in-depth acetylome analyses reveal systems attributes of CBP/p300 targets, and the resource dataset provides a framework for investigating CBP/p300 functions and for understanding the impact of small-molecule inhibitors targeting its catalytic and bromodomain activities.


Subject(s)
Acetyltransferases/metabolism , p300-CBP Transcription Factors/metabolism , Acetylation/drug effects , Acetyltransferases/antagonists & inhibitors , Animals , Cell Line , Gene Knockout Techniques , Half-Life , Heterocyclic Compounds, 4 or More Rings/chemistry , Heterocyclic Compounds, 4 or More Rings/metabolism , Heterocyclic Compounds, 4 or More Rings/pharmacology , Histones/metabolism , Humans , Isotope Labeling , Kinetics , Mass Spectrometry , Mice , Peptides/analysis , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Signal Transduction , Small Molecule Libraries/chemistry , Small Molecule Libraries/metabolism , Small Molecule Libraries/pharmacology , Transcriptome/drug effects , p300-CBP Transcription Factors/antagonists & inhibitors , p300-CBP Transcription Factors/genetics
5.
ACS Med Chem Lett ; 9(1): 28-33, 2018 Jan 11.
Article in English | MEDLINE | ID: mdl-29348807

ABSTRACT

p300 and its paralog CBP can acetylate histones and other proteins and have been implicated in a number of diseases characterized by aberrant gene activation, such as cancer. A novel, highly selective, orally bioavailable histone acetyltransferase (HAT) domain inhibitor has been identified through virtual ligand screening and subsequent optimization of a unique hydantoin screening hit. Conformational restraint in the form of a spirocyclization followed by substitution with a urea led to a significant improvement in potency. Replacement of the hydantoin moiety with an oxazolidinedione followed by fluoro substitution led to A-485, which exhibits potent cell activity, low clearance, and high oral bioavailability.

6.
PLoS One ; 11(5): e0155209, 2016.
Article in English | MEDLINE | ID: mdl-27171280

ABSTRACT

The 2-aminothiazole series has anti-bacterial activity against the important global pathogen Mycobacterium tuberculosis. We explored the nature of the activity by designing and synthesizing a large number of analogs and testing these for activity against M. tuberculosis, as well as eukaryotic cells. We determined that the C-2 position of the thiazole can accommodate a range of lipophilic substitutions, while both the C-4 position and the thiazole core are sensitive to change. The series has good activity against M. tuberculosis growth with sub-micromolar minimum inhibitory concentrations being achieved. A representative analog was selective for mycobacterial species over other bacteria and was rapidly bactericidal against replicating M. tuberculosis. The mode of action does not appear to involve iron chelation. We conclude that this series has potential for further development as novel anti-tubercular agents.


Subject(s)
Antitubercular Agents/chemical synthesis , Antitubercular Agents/pharmacology , Thiazoles/chemical synthesis , Thiazoles/pharmacology , Animals , Antitubercular Agents/chemistry , Iron Chelating Agents/pharmacology , Microbial Sensitivity Tests , Mycobacterium tuberculosis/drug effects , Structure-Activity Relationship , Thiazoles/chemistry , Vero Cells
7.
Bioorg Med Chem ; 22(24): 6965-79, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25456390

ABSTRACT

The 2,4-diaminoquinazoline class of compounds has previously been identified as an effective inhibitor of Mycobacterium tuberculosis growth. We conducted an extensive evaluation of the series for its potential as a lead candidate for tuberculosis drug discovery. Three segments of the representative molecule N-(4-fluorobenzyl)-2-(piperidin-1-yl)quinazolin-4-amine were examined systematically to explore structure-activity relationships influencing potency. We determined that the benzylic amine at the 4-position, the piperidine at 2-position and the N-1 (but not N-3) are key activity determinants. The 3-deaza analog retained similar activity to the parent molecule. Biological activity was not dependent on iron or carbon source availability. We demonstrated through pharmacokinetic studies in rats that good in vivo compound exposure is achievable. A representative compound demonstrated bactericidal activity against both replicating and non-replicating M. tuberculosis. We isolated and sequenced M. tuberculosis mutants resistant to this compound and observed mutations in Rv3161c, a gene predicted to encode a dioxygenase, suggesting that the compound may act as a pro-drug.


Subject(s)
Antitubercular Agents/chemical synthesis , Antitubercular Agents/pharmacology , Mycobacterium tuberculosis/drug effects , Quinazolines/chemistry , Quinazolines/pharmacology , Animals , Antitubercular Agents/chemistry , Antitubercular Agents/pharmacokinetics , Drug Resistance, Bacterial/drug effects , Half-Life , Microbial Sensitivity Tests , Quinazolines/chemical synthesis , Quinazolines/pharmacokinetics , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
8.
PLoS One ; 8(11): e72786, 2013.
Article in English | MEDLINE | ID: mdl-24244263

ABSTRACT

The enzyme pantothenate synthetase, PanC, is an attractive drug target in Mycobacterium tuberculosis. It is essential for the in vitro growth of M. tuberculosis and for survival of the bacteria in the mouse model of infection. PanC is absent from mammals. We developed an enzyme-based assay to identify inhibitors of PanC, optimized it for high-throughput screening, and tested a large and diverse library of compounds for activity. Two compounds belonging to the same chemical class of 3-biphenyl-4- cyanopyrrole-2-carboxylic acids had activity against the purified recombinant protein, and also inhibited growth of live M. tuberculosis in manner consistent with PanC inhibition. Thus we have identified a new class of PanC inhibitors with whole cell activity that can be further developed.


Subject(s)
Bacterial Proteins/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Mycobacterium tuberculosis/enzymology , Peptide Synthases/antagonists & inhibitors , Tuberculosis/drug therapy , Animals , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Chlorocebus aethiops , Disease Models, Animal , Drug Evaluation, Preclinical , Mice , Microbial Viability/drug effects , Peptide Synthases/chemistry , Peptide Synthases/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Tuberculosis/enzymology , Vero Cells
9.
J Bacteriol ; 194(3): 663-8, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22123255

ABSTRACT

Mycobacterium tuberculosis is a pathogen of major global importance. Validated drug targets are required in order to develop novel therapeutics for drug-resistant strains and to shorten therapy. The Clp protease complexes provide a means for quality control of cellular proteins; the proteolytic activity of ClpP in concert with the ATPase activity of the ClpX/ClpC subunits results in degradation of misfolded or damaged proteins. Thus, the Clp system plays a major role in basic metabolism, as well as in stress responses and pathogenic mechanisms. M. tuberculosis has two ClpP proteolytic subunits. Here we demonstrate that ClpP1 is essential for viability in this organism in culture, since the gene could only be deleted from the chromosome when a second functional copy was provided. Overexpression of clpP1 had no effect on growth in aerobic culture or viability under anaerobic conditions or during nutrient starvation. In contrast, clpP2 overexpression was toxic, suggesting different roles for the two homologs. We synthesized known activators of ClpP protease activity; these acyldepsipeptides (ADEPs) were active against M. tuberculosis. ADEP activity was enhanced by the addition of efflux pump inhibitors, demonstrating that ADEPs gain access to the cell but that export occurs. Taken together, the genetic and chemical validation of ClpP as a drug target leads to new avenues for drug discovery.


Subject(s)
Bacterial Proteins/metabolism , Endopeptidase Clp/metabolism , Mycobacterium tuberculosis/enzymology , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/genetics , Endopeptidase Clp/antagonists & inhibitors , Endopeptidase Clp/genetics , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Bacterial , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/growth & development
10.
11.
Cancer Res ; 65(12): 4987-92, 2005 Jun 15.
Article in English | MEDLINE | ID: mdl-15958537

ABSTRACT

DNA-dependent protein kinase (DNA-PK)-defective severe combined immunodeficient (SCID) mice have a greater sensitivity to ionizing radiation compared with wild-type mice due to deficient repair of DNA double-strand break. SCID cells were therefore studied to determine whether radiosensitization by the specific inhibitor of DNA-PK, IC87361, is eliminated in the absence of functional DNA-PK. IC87361 enhanced radiation sensitivity in wild-type C57BL6 endothelial cells but not in SCID cells. The tumor vascular window model was used to assess IC87361-induced radiosensitization of SCID and wild-type tumor microvasculature. Vascular density was 5% in irradiated SCID host compared with 50% in C57BL6 mice (P < 0.05). IC87361 induced radiosensitization of tumor microvasculature in wild-type mice that resembled the radiosensitive phenotype of tumor vessels in SCID mice. Radiosensitization by IC87361 was eliminated in SCID tumor vasculature, which lack functional DNA-PK. Irradiated LLC and B16F0 tumors implanted into SCID mice showed greater tumor growth delay compared with tumors implanted into either wild-type C57BL6 or nude mice. Furthermore, LLC tumors treated with radiation and IC87361 showed tumor growth delay that was significantly greater than tumors treated with radiation alone (P < 0.01 for 3 Gy alone versus 3 Gy + IC87361). DNA-PK inhibitors induced no cytotoxicity and no toxicity in mouse normal tissues. Mouse models deficient in enzyme activity are useful to assess the specificity of novel kinase inhibitors. DNA-PK is an important target for the development of novel radiation-sensitizing drugs that have little intrinsic cytotoxicity.


Subject(s)
DNA-Binding Proteins/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Radiation-Sensitizing Agents/pharmacology , Animals , Carcinoma, Lewis Lung/blood supply , Carcinoma, Lewis Lung/drug therapy , Carcinoma, Lewis Lung/enzymology , Carcinoma, Lewis Lung/radiotherapy , Cell Growth Processes/drug effects , Cell Growth Processes/radiation effects , Cell Line, Tumor , Combined Modality Therapy , DNA-Activated Protein Kinase , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/radiation effects , Melanoma, Experimental/blood supply , Melanoma, Experimental/drug therapy , Melanoma, Experimental/enzymology , Melanoma, Experimental/radiotherapy , Mice , Mice, Nude , Mice, SCID , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/enzymology , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/radiotherapy
12.
Cancer Res ; 64(14): 4893-9, 2004 Jul 15.
Article in English | MEDLINE | ID: mdl-15256460

ABSTRACT

The phosphatidylinositol 3'-kinase (PI3k)/protein kinase B (PKB/Akt) signal transduction pathway plays a critical role in mediating endothelial cell survival and function during oxidative stress. The role of the PI3k/Akt signaling pathway in promoting cell viability was studied in vascular endothelial cells treated with ionizing radiation. Western blot analysis showed that Akt was rapidly phosphorylated in response to radiation in primary culture endothelial cells (human umbilical vascular endothelial cells) in the absence of serum or growth factors. PI3k consists of p85 and p110 subunits, which play a central upstream role in Akt activation in response to exogenous stimuli. The delta isoform of the p110 subunit is expressed in endothelial cells. We studied the effects of the p110delta specific inhibitor IC486068, which abrogated radiation-induced phosphorylation of Akt. IC486068 enhanced radiation-induced apoptosis in endothelial cells and reduced cell migration and tubule formation of endothelial cells in Matrigel following irradiation. In vivo tumor growth delay was studied in mice with Lewis lung carcinoma and GL261 hind limb tumors. Mice were treated with daily i.p. injections (25 mg/kg) of IC486068 during 6 days of radiation treatment (18 Gy). Combined treatment with IC486068 and radiation significantly reduced tumor volume as compared with either treatment alone. Reduction in vasculature was confirmed using the dorsal skinfold vascular window model. The vascular length density was measured by use of the tumor vascular window model and showed IC486068 significantly enhanced radiation-induced destruction of tumor vasculature as compared with either treatment alone. IC486068 enhances radiation-induced endothelial cytotoxicity, resulting in tumor vascular destruction and tumor control when combined with fractionated radiotherapy in murine tumor models. These findings suggest that p110delta is a therapeutic target to enhance radiation-induced tumor control.


Subject(s)
Enzyme Inhibitors/pharmacology , Neoplasms, Experimental/blood supply , Neoplasms, Experimental/radiotherapy , Phosphoinositide-3 Kinase Inhibitors , Quinazolines/pharmacology , Animals , Catalytic Domain , Cell Movement/drug effects , Cell Survival/drug effects , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Endothelium, Vascular/radiation effects , Humans , Mice , Neoplasms, Experimental/drug therapy , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/radiotherapy , Radiation-Sensitizing Agents/pharmacology
13.
Mol Cancer Ther ; 2(12): 1257-64, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14707266

ABSTRACT

Cancer presents a difficult challenge for oncologists, as there are few therapies that specifically target disease cells. Existing treatment strategies rely heavily on physical and chemical agents that nonspecifically affect DNA metabolism. To improve the effectiveness of these treatments, we have identified a new class of protein kinase inhibitor that targets a major DNA repair pathway. A representative of this class, 1-(2-hydroxy-4-morpholin-4-yl-phenyl)-ethanone, inhibits the DNA-dependent protein kinase (DNA-PK) and differs significantly from previously studied DNA-PK inhibitors both structurally and functionally. DNA-PK participates in the cellular response to and repair of chromosomal DNA double-strand breaks (DSBs). These new selective inhibitors recapitulate the phenotype of DNA-PK defective cell lines including those from SCID mice. These compounds directly inhibit the repair of DNA DSBs and consequently enhance the cytotoxicity of physical and chemical agents that induce DSBs but not other DNA lesions. In contrast to previously studied DNA-PK inhibitors, these compounds appear benign, exhibiting no toxic effects in the absence of DSB-inducing treatments. Most importantly, 1-(2-hydroxy-4-morpholin-4-yl-phenyl)-ethanone synergistically enhances radiation-induced tumor control in a mouse-human xenograft assay. These studies validate DNA-PK as a cancer drug target and suggest a new approach for enhancing the effects of existing cancer therapies.


Subject(s)
Antineoplastic Agents/pharmacology , DNA-Binding Proteins , Enzyme Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , Antineoplastic Agents/therapeutic use , DNA Damage , DNA-Activated Protein Kinase , Enzyme Inhibitors/therapeutic use , HeLa Cells , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Nuclear Proteins , Phenotype
SELECTION OF CITATIONS
SEARCH DETAIL
...