Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
J Med Chem ; 67(7): 5758-5782, 2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38511649

ABSTRACT

Eukaryotic translation initiation factor 2B (eIF2B) is a key component of the integrated stress response (ISR), which regulates protein synthesis and stress granule formation in response to cellular insult. Modulation of the ISR has been proposed as a therapeutic strategy for treatment of neurodegenerative diseases such as vanishing white matter (VWM) disease and amyotrophic lateral sclerosis (ALS) based on its ability to improve cellular homeostasis and prevent neuronal degeneration. Herein, we report the small-molecule discovery campaign that identified potent, selective, and CNS-penetrant eIF2B activators using both structure- and ligand-based drug design. These discovery efforts culminated in the identification of DNL343, which demonstrated a desirable preclinical drug profile, including a long half-life and high oral bioavailability across preclinical species. DNL343 was progressed into clinical studies and is currently undergoing evaluation in late-stage clinical trials for ALS.


Subject(s)
Amyotrophic Lateral Sclerosis , Leukoencephalopathies , Neurodegenerative Diseases , Humans , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/metabolism , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/metabolism , Mutation , Eukaryotic Initiation Factor-2B/genetics , Eukaryotic Initiation Factor-2B/metabolism , Brain/metabolism , Leukoencephalopathies/metabolism
2.
Sci Rep ; 11(1): 12900, 2021 06 18.
Article in English | MEDLINE | ID: mdl-34145320

ABSTRACT

Variants in the leucine-rich repeat kinase 2 (LRRK2) gene are associated with increased risk for familial and sporadic Parkinson's disease (PD). Pathogenic variants in LRRK2, including the common variant G2019S, result in increased LRRK2 kinase activity, supporting the therapeutic potential of LRRK2 kinase inhibitors for PD. To better understand the role of LRRK2 in disease and to support the clinical development of LRRK2 inhibitors, quantitative and high-throughput assays to measure LRRK2 levels and activity are needed. We developed and applied such assays to measure the levels of LRRK2 as well as the phosphorylation of LRRK2 itself or one of its substrates, Rab10 (pT73 Rab10). We observed increased LRRK2 activity in various cellular models of disease, including iPSC-derived microglia, as well as in human subjects carrying the disease-linked variant LRRK2 G2019S. Capitalizing on the high-throughput and sensitive nature of these assays, we detected a significant reduction in LRRK2 activity in subjects carrying missense variants in LRRK2 associated with reduced disease risk. Finally, we optimized these assays to enable analysis of LRRK2 activity following inhibition in human peripheral blood mononuclear cells (PBMCs) and whole blood, demonstrating their potential utility as biomarkers to assess changes in LRRK2 expression and activity in the clinic.


Subject(s)
Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , rab GTP-Binding Proteins/metabolism , Animals , Biomarkers , Enzyme Activation , Enzyme Assays/methods , Enzyme Assays/standards , Gene Expression , High-Throughput Screening Assays , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Leukocytes, Mononuclear/metabolism , Mice , Neuroglia/metabolism , rab GTP-Binding Proteins/genetics
3.
Nat Med ; 21(5): 457-66, 2015 May.
Article in English | MEDLINE | ID: mdl-25849135

ABSTRACT

A common key regulator of oncogenic signaling pathways in multiple tumor types is the unique isomerase Pin1. However, available Pin1 inhibitors lack the required specificity and potency for inhibiting Pin1 function in vivo. By using mechanism-based screening, here we find that all-trans retinoic acid (ATRA)--a therapy for acute promyelocytic leukemia (APL) that is considered the first example of targeted therapy in cancer, but whose drug target remains elusive--inhibits and degrades active Pin1 selectively in cancer cells by directly binding to the substrate phosphate- and proline-binding pockets in the Pin1 active site. ATRA-induced Pin1 ablation degrades the protein encoded by the fusion oncogene PML-RARA and treats APL in APL cell and animal models as well as in human patients. ATRA-induced Pin1 ablation also potently inhibits triple-negative breast cancer cell growth in human cells and in animal models by acting on many Pin1 substrate oncogenes and tumor suppressors. Thus, ATRA simultaneously blocks multiple Pin1-regulated cancer-driving pathways, an attractive property for treating aggressive and drug-resistant tumors.


Subject(s)
Breast Neoplasms/metabolism , Gene Expression Regulation, Leukemic , Gene Expression Regulation, Neoplastic , Leukemia, Promyelocytic, Acute/metabolism , Peptidylprolyl Isomerase/genetics , Tretinoin/metabolism , Animals , Antineoplastic Agents/chemistry , Breast Neoplasms/genetics , Catalysis , Catalytic Domain , Cell Line, Tumor , Dose-Response Relationship, Drug , Female , Fibroblasts/metabolism , HEK293 Cells , Humans , Leukemia, Promyelocytic, Acute/genetics , MCF-7 Cells , Mice , Mice, Inbred BALB C , Mice, Knockout , NIMA-Interacting Peptidylprolyl Isomerase , Neoplasm Transplantation , Phosphates/chemistry , Phosphorylation , Proline/chemistry , Triple Negative Breast Neoplasms/metabolism
4.
Mol Syst Biol ; 11(3): 797, 2015 Mar 26.
Article in English | MEDLINE | ID: mdl-25814555

ABSTRACT

Drugs that inhibit RAF/MEK signaling, such as vemurafenib, elicit profound but often temporary anti-tumor responses in patients with BRAF(V) (600E) melanoma. Adaptive responses to RAF/MEK inhibition occur on a timescale of hours to days, involve homeostatic responses that reactivate MAP kinase signaling and compensatory mitogenic pathways, and attenuate the anti-tumor effects of RAF/MEK inhibitors. We profile adaptive responses across a panel of melanoma cell lines using multiplex biochemical measurement, single-cell assays, and statistical modeling and show that adaptation involves at least six signaling cascades that act to reduce drug potency (IC50) and maximal effect (i.e., Emax â‰ª 1). Among these cascades, we identify a role for JNK/c-Jun signaling in vemurafenib adaptation and show that RAF and JNK inhibitors synergize in cell killing. This arises because JNK inhibition prevents a subset of cells in a cycling population from becoming quiescent upon vemurafenib treatment, thereby reducing drug Emax. Our findings demonstrate the breadth and diversity of adaptive responses to RAF/MEK inhibition and a means to identify which steps in a signaling cascade are most predictive of phenotypic response.


Subject(s)
Drug Resistance, Neoplasm , Indoles/pharmacology , Melanoma/genetics , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/genetics , Sulfonamides/pharmacology , Apoptosis , Cell Line, Tumor , Cell Survival/drug effects , Drug Synergism , Humans , MAP Kinase Signaling System , Melanoma/drug therapy , Mutation , Vemurafenib
5.
J Biomol Screen ; 19(6): 947-58, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24518064

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV) has a causative role in several human malignancies, especially in immunocompromised hosts. KSHV latently infects tumor cells and persists as an extrachromosomal episome (plasmid). KSHV latency-associated nuclear antigen (LANA) mediates KSHV episome persistence. LANA binds specific KSHV sequence to replicate viral DNA. In addition, LANA tethers KSHV genomes to mitotic chromosomes to efficiently segregate episomes to daughter nuclei after mitosis. N-terminal LANA (N-LANA) binds histones H2A and H2B to attach to chromosomes. Currently, there are no specific inhibitors of KSHV latent infection. To enable high-throughput screening (HTS) of inhibitors of N-LANA binding to nucleosomes, here we develop, miniaturize, and validate a fluorescence polarization (FP) assay that detects fluorophore-labeled N-LANA peptide binding to nucleosomes. We also miniaturize a counterscreen to identify DNA intercalators that nonspecifically inhibit N-LANA binding to nucleosomes, and also develop an enzyme-linked immunosorbent assay to assess N-LANA binding to nucleosomes in the absence of fluorescence. HTS of libraries containing more than 350,000 compounds identified multiple compounds that inhibited N-LANA binding to nucleosomes. No compounds survived all counterscreens, however. More complex small-molecule libraries will likely be necessary to identify specific inhibitors of N-LANA binding to histones H2A and H2B; these assays should prove useful for future screens.


Subject(s)
Antiviral Agents/chemistry , Herpesvirus 8, Human/drug effects , Nuclear Proteins/antagonists & inhibitors , Nucleosomes/chemistry , Animals , Antigens/chemistry , Antigens, Viral/chemistry , Cell Survival , Chickens , Drug Design , Enzyme-Linked Immunosorbent Assay , Erythrocytes/virology , Fluorescence Polarization , Fluorescent Dyes/chemistry , Glutathione Transferase/metabolism , HeLa Cells , High-Throughput Screening Assays , Histones/chemistry , Humans , Mitosis , Nuclear Proteins/chemistry , Plasmids/chemistry , Protein Domains , Spectrometry, Fluorescence
6.
Proc Natl Acad Sci U S A ; 108(8): 3383-8, 2011 Feb 22.
Article in English | MEDLINE | ID: mdl-21289279

ABSTRACT

Considerable evidence indicates that the general blockade of protein synthesis prevents both the initial consolidation and the postretrieval reconsolidation of long-term memories. These findings come largely from studies of drugs that block ribosomal function, so as to globally interfere with both cap-dependent and -independent forms of translation. Here we show that intra-amygdala microinfusions of 4EGI-1, a small molecule inhibitor of cap-dependent translation that selectively disrupts the interaction between eukaryotic initiation factors (eIF) 4E and 4G, attenuates fear memory consolidation but not reconsolidation. Using a combination of behavioral and biochemical techniques, we provide both in vitro and in vivo evidence that the eIF4E-eIF4G complex is more stringently required for plasticity induced by initial learning than for that triggered by reactivation of an existing memory.


Subject(s)
Eukaryotic Initiation Factor-4E/metabolism , Eukaryotic Initiation Factor-4G/metabolism , Memory, Long-Term , Protein Synthesis Inhibitors/pharmacology , Amygdala , Animals , Eukaryotic Initiation Factor-4G/antagonists & inhibitors , Male , Neuronal Plasticity , Protein Binding/drug effects , Protein Biosynthesis/physiology , Rats , Rats, Sprague-Dawley
7.
Curr Protoc Chem Biol ; 3(1): 39-52, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-23836588

ABSTRACT

The in-cell western (ICW) technique is a cell-based immunoassay method for quantitative measurement of protein expression or phosphorylation levels that can be used for both small molecule and siRNA screening. The method involves growth of cells in microplates, fixation, permeabilization, and staining with specific antibodies and/or cell labeling dyes. ICW assays take advantage of the properties of near-infrared dyes to achieve higher signal-to-noise ratios than are possible for methods utilizing fluorophores in the visible range of the spectrum, and typically involve measurements using two fluorescent channels: one to measure levels of the target of interest, and one to measure total cell number for normalization. The ICW method is readily adaptable to high-throughput format and has been successfully used with a variety of targets and cell lines. The protocols in this unit describe an ICW procedure for quantitative measurement of rpS6-phosphorylation as an endpoint for monitoring mTORC1 signaling in HeLa cells. This assay can be used for small molecule or siRNA screening, and with modification is adaptable to other cell lines and targets. Curr. Protoc. Chem. Biol. 3:39-52 © 2011 by John Wiley & Sons, Inc.

8.
Assay Drug Dev Technol ; 8(2): 186-99, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20085456

ABSTRACT

The mTORC1 pathway is a central regulator of cell growth, and defective mTORC1 regulation plays a causative role in a variety of human diseases, including cancer, tumor syndromes such as the tuberous sclerosis complex (TSC) and lymphangioleiomyomatosis (LAM), and metabolic diseases such as diabetes and obesity. Given the importance of mTORC1 signaling in these diseases, there has been significant interest in developing screening methods suitable for identifying inhibitors of mTORC1 activation. To this end, we have developed a high-throughput, cell-based assay for the detection of rpS6-phosphorylation as a measure of mTORC1 signaling. This assay takes advantage of the "In Cell Western" (ICW) technique using the Aerius infrared imaging system (LI-COR Biosciences). The ICW procedure involves fixation and immunostaining of cells in a manner similar to standard immunofluorescence methods but takes advantage of secondary antibodies conjugated to infrared-excitable fluorophores for quantitative detection by the Aerius scanner. In addition, the cells are stained with an infrared-excitable succinimidyl ester dye, which covalently modifies free amine groups in fixed cells and provides a quantitative measure of cell number. We present validation data and pilot screens in a 384-well format demonstrating that this assay provides a statistically robust method for both small molecule and siRNA screening approaches designed to identify inhibitors of mTORC1 signaling.


Subject(s)
RNA, Small Interfering/pharmacology , Signal Transduction/drug effects , Transcription Factors/antagonists & inhibitors , Amines/chemistry , Amines/radiation effects , Antibody Specificity , Blotting, Western , Cell Count , Cell Survival , Drug Evaluation, Preclinical , Endpoint Determination , Fluorescent Antibody Technique, Indirect , HeLa Cells , Humans , Infrared Rays , Mechanistic Target of Rapamycin Complex 1 , Multiprotein Complexes , Proteins , Reproducibility of Results , Small Molecule Libraries , TOR Serine-Threonine Kinases , Transfection
9.
Curr Protoc Chem Biol ; 1(1): 1-15, 2009 Dec 01.
Article in English | MEDLINE | ID: mdl-23839960

ABSTRACT

The technique of fluorescence polarization (FP) is based on the observation that when a fluorescently labeled molecule is excited by polarized light, it emits light with a degree of polarization that is inversely proportional to the rate of molecular rotation. This property of fluorescence can be used to measure the interaction of a small labeled ligand with a larger protein and provides a basis for direct and competition binding assays. FP assays are readily adaptable to a high-throughput format, have been used successfully in screens directed against a wide range of targets, and are particularly valuable in screening for inhibitors of protein-protein and protein-nucleic acid interactions when a small binding epitope can be identified for one of the partners. The protocols in this article describe a general procedure for development of FP assays to monitor binding of such a peptide or oligonucleotide to a protein of interest. Curr. Protoc. Chem Biol. 1:1-15. © 2009 by John Wiley & Sons, Inc.

10.
Cell ; 128(2): 257-67, 2007 Jan 26.
Article in English | MEDLINE | ID: mdl-17254965

ABSTRACT

Assembly of the eIF4E/eIF4G complex has a central role in the regulation of gene expression at the level of translation initiation. This complex is regulated by the 4E-BPs, which compete with eIF4G for binding to eIF4E and which have tumor-suppressor activity. To pharmacologically mimic 4E-BP function we developed a high-throughput screening assay for identifying small-molecule inhibitors of the eIF4E/eIF4G interaction. The most potent compound identified, 4EGI-1, binds eIF4E, disrupts eIF4E/eIF4G association, and inhibits cap-dependent translation but not initiation factor-independent translation. While 4EGI-1 displaces eIF4G from eIF4E, it effectively enhances 4E-BP1 association both in vitro and in cells. 4EGI-1 inhibits cellular expression of oncogenic proteins encoded by weak mRNAs, exhibits activity against multiple cancer cell lines, and appears to have a preferential effect on transformed versus nontransformed cells. The identification of this compound provides a new tool for studying translational control and establishes a possible new strategy for cancer therapy.


Subject(s)
Antineoplastic Agents/isolation & purification , Antineoplastic Agents/pharmacology , Cell Transformation, Neoplastic/drug effects , Eukaryotic Initiation Factor-4E/drug effects , Eukaryotic Initiation Factor-4G/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Nitro Compounds/isolation & purification , Nitro Compounds/pharmacology , Thiazoles/isolation & purification , Thiazoles/pharmacology , Animals , Antineoplastic Agents/chemistry , Cell Line, Transformed , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Drug Evaluation, Preclinical/methods , Eukaryotic Initiation Factor-4E/genetics , Eukaryotic Initiation Factor-4E/metabolism , Eukaryotic Initiation Factor-4G/genetics , Eukaryotic Initiation Factor-4G/metabolism , Feedback, Physiological/drug effects , Feedback, Physiological/physiology , Fluorescence Polarization Immunoassay/methods , Gene Expression Regulation, Neoplastic/genetics , Humans , Hydrazones , Jurkat Cells , Mice , Models, Molecular , Nitro Compounds/chemistry , Oncogenes/drug effects , Oncogenes/genetics , Peptide Fragments/genetics , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Protein Binding/drug effects , Protein Binding/genetics , Protein Biosynthesis/drug effects , Protein Biosynthesis/genetics , RNA, Messenger/drug effects , RNA, Messenger/genetics , Thiazoles/chemistry
11.
Cell ; 115(6): 739-50, 2003 Dec 12.
Article in English | MEDLINE | ID: mdl-14675538

ABSTRACT

The eukaryotic initiation factor 4G (eIF4G) is the core of a multicomponent switch controlling gene expression at the level of translation initiation. It interacts with the small ribosomal subunit interacting protein, eIF3, and the eIF4E/cap-mRNA complex in order to load the ribosome onto mRNA during cap-dependent translation. We describe the solution structure of the complex between yeast eIF4E/cap and eIF4G (393-490). Binding triggers a coupled folding transition of eIF4G (393-490) and the eIF4E N terminus resulting in a molecular bracelet whereby eIF4G (393-490) forms a right-handed helical ring that wraps around the N terminus of eIF4E. Cofolding allosterically enhances association of eIF4E with the cap and is required for maintenance of optimal growth and polysome distributions in vivo. Our data explain how mRNA, eIF4E, and eIF4G exists as a stable mRNP that may facilitate multiple rounds of ribosomal loading during translation initiation, a key determinant in the overall rate of protein synthesis.


Subject(s)
Eukaryotic Initiation Factor-4E/metabolism , Eukaryotic Initiation Factor-4G/metabolism , Protein Biosynthesis/genetics , RNA Caps/metabolism , Ribosomes/metabolism , Binding Sites/physiology , Eukaryotic Initiation Factor-4E/genetics , Eukaryotic Initiation Factor-4G/genetics , Guanosine Diphosphate/metabolism , Macromolecular Substances , Models, Molecular , Molecular Conformation , Nucleic Acid Conformation , Protein Conformation , RNA Caps/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ribonucleoproteins/metabolism , Ribosomes/genetics , Saccharomyces cerevisiae
SELECTION OF CITATIONS
SEARCH DETAIL
...