Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Nat Commun ; 12(1): 1222, 2021 02 22.
Article in English | MEDLINE | ID: mdl-33619272

ABSTRACT

Programmed death-ligand 1 is a glycoprotein expressed on antigen presenting cells, hepatocytes, and tumors which upon interaction with programmed death-1, results in inhibition of antigen-specific T cell responses. Here, we report a mechanism of inhibiting programmed death-ligand 1 through small molecule-induced dimerization and internalization. This represents a mechanism of checkpoint inhibition, which differentiates from anti-programmed death-ligand 1 antibodies which function through molecular disruption of the programmed death 1 interaction. Testing of programmed death ligand 1 small molecule inhibition in a humanized mouse model of colorectal cancer results in a significant reduction in tumor size and promotes T cell proliferation. In addition, antigen-specific T and B cell responses from patients with chronic hepatitis B infection are significantly elevated upon programmed death ligand 1 small molecule inhibitor treatment. Taken together, these data identify a mechanism of small molecule-induced programmed death ligand 1 internalization with potential therapeutic implications in oncology and chronic viral infections.


Subject(s)
B7-H1 Antigen/metabolism , Endocytosis , Immune Checkpoint Inhibitors/pharmacology , Small Molecule Libraries/pharmacology , Animals , Antineoplastic Agents/pharmacology , Antiviral Agents/pharmacology , CHO Cells , Cell Proliferation/drug effects , Colorectal Neoplasms/pathology , Cricetulus , Disease Models, Animal , Female , Hepatitis B virus/drug effects , Humans , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/metabolism , Protein Multimerization/drug effects , Small Molecule Libraries/chemistry
2.
Nat Med ; 26(8): 1264-1270, 2020 08.
Article in English | MEDLINE | ID: mdl-32661391

ABSTRACT

Cancer cachexia is a highly prevalent condition associated with poor quality of life and reduced survival1. Tumor-induced perturbations in the endocrine, immune and nervous systems drive anorexia and catabolic changes in adipose tissue and skeletal muscle, hallmarks of cancer cachexia2-4. However, the molecular mechanisms driving cachexia remain poorly defined, and there are currently no approved drugs for the condition. Elevation in circulating growth differentiation factor 15 (GDF15) correlates with cachexia and reduced survival in patients with cancer5-8, and a GDNF family receptor alpha like (GFRAL)-Ret proto-oncogene (RET) signaling complex in brainstem neurons that mediates GDF15-induced weight loss in mice has recently been described9-12. Here we report a therapeutic antagonistic monoclonal antibody, 3P10, that targets GFRAL and inhibits RET signaling by preventing the GDF15-driven interaction of RET with GFRAL on the cell surface. Treatment with 3P10 reverses excessive lipid oxidation in tumor-bearing mice and prevents cancer cachexia, even under calorie-restricted conditions. Mechanistically, activation of the GFRAL-RET pathway induces expression of genes involved in lipid metabolism in adipose tissues, and both peripheral chemical sympathectomy and loss of adipose triglyceride lipase protect mice from GDF15-induced weight loss. These data uncover a peripheral sympathetic axis by which GDF15 elicits a lipolytic response in adipose tissue independently of anorexia, leading to reduced adipose and muscle mass and function in tumor-bearing mice.


Subject(s)
Cachexia/drug therapy , Glial Cell Line-Derived Neurotrophic Factor Receptors/genetics , Growth Differentiation Factor 15/genetics , Multiprotein Complexes/ultrastructure , Neoplasms/drug therapy , Proto-Oncogene Proteins c-ret/genetics , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , Antibodies, Monoclonal , Cachexia/complications , Cachexia/genetics , Cachexia/immunology , Cell Line, Tumor , Crystallography, X-Ray , Glial Cell Line-Derived Neurotrophic Factor Receptors/ultrastructure , Growth Differentiation Factor 15/ultrastructure , Heterografts , Humans , Lipid Peroxidation , Mice , Multiprotein Complexes/genetics , Muscle, Skeletal/drug effects , Muscle, Skeletal/pathology , Neoplasms/complications , Neoplasms/genetics , Neoplasms/immunology , Proto-Oncogene Mas , Proto-Oncogene Proteins c-ret/ultrastructure , Signal Transduction , Weight Loss
3.
Bioorg Med Chem Lett ; 29(17): 2503-2510, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31327531

ABSTRACT

Cancer cells reprogram their metabolism to support growth and to mitigate cellular stressors. The serine synthesis pathway has been identified as a metabolic pathway frequently altered in cancers and there has been considerable interest in developing pharmacological agents to target this pathway. Here, we report a series of indole amides that inhibit human 3-phosphoglycerate dehydrogenase (PHGDH), the enzyme that catalyzes the first committed step of the serine synthesis pathway. Using X-ray crystallography, we show that the indole amides bind the NAD+ pocket of PHGDH. Through structure-based optimization we were able to develop compounds with low nanomolar affinities for PHGDH in an enzymatic IC50 assay. In cellular assays, the most potent compounds inhibited de novo serine synthesis with low micromolar to sub-micromolar activities and these compounds successfully abrogated the proliferation of cancer cells in serine free media. The indole amide series reported here represent an important improvement over previously published PHGDH inhibitors as they are markedly more potent and their mechanism of action is better defined.


Subject(s)
Amides/chemistry , Enzyme Inhibitors/chemistry , Indoles/chemistry , Phosphoglycerate Dehydrogenase/antagonists & inhibitors , Serine/biosynthesis , Amides/metabolism , Amides/pharmacology , Binding Sites , Cell Line, Tumor , Cell Proliferation/drug effects , Crystallography, X-Ray , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Humans , Molecular Dynamics Simulation , Phosphoglycerate Dehydrogenase/metabolism , Protein Structure, Tertiary , Structure-Activity Relationship
4.
J Med Chem ; 61(18): 8202-8211, 2018 09 27.
Article in English | MEDLINE | ID: mdl-30165024

ABSTRACT

BET proteins are key epigenetic regulators that regulate transcription through binding to acetylated lysine (AcLys) residues of histones and transcription factors through bromodomains (BDs). The disruption of this interaction with small molecule bromodomain inhibitors is a promising approach to treat various diseases including cancer, autoimmune and cardiovascular diseases. Covalent inhibitors can potentially offer a more durable target inhibition leading to improved in vivo pharmacology. Here we describe the design of covalent inhibitors of BRD4(BD1) that target a methionine in the binding pocket by attaching an epoxide warhead to a suitably oriented noncovalent inhibitor. Using thermal denaturation, MALDI-TOF mass spectrometry, and an X-ray crystal structure, we demonstrate that these inhibitors selectively form a covalent bond with Met149 in BRD4(BD1) but not other bromodomains and provide durable transcriptional and antiproliferative activity in cell based assays. Covalent targeting of methionine offers a novel approach to drug discovery for BET proteins and other targets.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Design , Drug Discovery , Hematologic Neoplasms/drug therapy , Methionine/chemistry , Nuclear Proteins/antagonists & inhibitors , Transcription Factors/antagonists & inhibitors , Antineoplastic Agents/chemistry , Cell Cycle Proteins , Crystallography, X-Ray , Hematologic Neoplasms/pathology , Humans , Models, Molecular , Molecular Structure , Protein Conformation , Structure-Activity Relationship , Tumor Cells, Cultured
5.
Sci Transl Med ; 10(431)2018 03 07.
Article in English | MEDLINE | ID: mdl-29515000

ABSTRACT

Glycogen synthase kinase 3 (GSK3), a key regulatory kinase in the wingless-type MMTV integration site family (WNT) pathway, is a therapeutic target of interest in many diseases. Although dual GSK3α/ß inhibitors have entered clinical trials, none has successfully translated to clinical application. Mechanism-based toxicities, driven in part by the inhibition of both GSK3 paralogs and subsequent ß-catenin stabilization, are a concern in the translation of this target class because mutations and overexpression of ß-catenin are associated with many cancers. Knockdown of GSK3α or GSK3ß individually does not increase ß-catenin and offers a conceptual resolution to targeting GSK3: paralog-selective inhibition. However, inadequate chemical tools exist. The design of selective adenosine triphosphate (ATP)-competitive inhibitors poses a drug discovery challenge due to the high homology (95% identity and 100% similarity) in this binding domain. Taking advantage of an Asp133→Glu196 "switch" in their kinase hinge, we present a rational design strategy toward the discovery of paralog-selective GSK3 inhibitors. These GSK3α- and GSK3ß-selective inhibitors provide insights into GSK3 targeting in acute myeloid leukemia (AML), where GSK3α was identified as a therapeutic target using genetic approaches. The GSK3α-selective compound BRD0705 inhibits kinase function and does not stabilize ß-catenin, mitigating potential neoplastic concerns. BRD0705 induces myeloid differentiation and impairs colony formation in AML cells, with no apparent effect on normal hematopoietic cells. Moreover, BRD0705 impairs leukemia initiation and prolongs survival in AML mouse models. These studies demonstrate feasibility of paralog-selective GSK3α inhibition, offering a promising therapeutic approach in AML.


Subject(s)
Enzyme Inhibitors/therapeutic use , Glycogen Synthase Kinase 3/antagonists & inhibitors , Leukemia, Myeloid, Acute/drug therapy , Dipeptides/chemistry , Dipeptides/metabolism , Glycogen Synthase Kinase 3/chemistry , Glycogen Synthase Kinase 3/metabolism , Humans , Mutagenesis, Site-Directed , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/chemistry , Protein Isoforms/metabolism , U937 Cells , beta Catenin/genetics , beta Catenin/metabolism
7.
Nature ; 550(7675): 255-259, 2017 10 12.
Article in English | MEDLINE | ID: mdl-28953886

ABSTRACT

Under homeostatic conditions, animals use well-defined hypothalamic neural circuits to help maintain stable body weight, by integrating metabolic and hormonal signals from the periphery to balance food consumption and energy expenditure. In stressed or disease conditions, however, animals use alternative neuronal pathways to adapt to the metabolic challenges of altered energy demand. Recent studies have identified brain areas outside the hypothalamus that are activated under these 'non-homeostatic' conditions, but the molecular nature of the peripheral signals and brain-localized receptors that activate these circuits remains elusive. Here we identify glial cell-derived neurotrophic factor (GDNF) receptor alpha-like (GFRAL) as a brainstem-restricted receptor for growth and differentiation factor 15 (GDF15). GDF15 regulates food intake, energy expenditure and body weight in response to metabolic and toxin-induced stresses; we show that Gfral knockout mice are hyperphagic under stressed conditions and are resistant to chemotherapy-induced anorexia and body weight loss. GDF15 activates GFRAL-expressing neurons localized exclusively in the area postrema and nucleus tractus solitarius of the mouse brainstem. It then triggers the activation of neurons localized within the parabrachial nucleus and central amygdala, which constitute part of the 'emergency circuit' that shapes feeding responses to stressful conditions. GDF15 levels increase in response to tissue stress and injury, and elevated levels are associated with body weight loss in numerous chronic human diseases. By isolating GFRAL as the receptor for GDF15-induced anorexia and weight loss, we identify a mechanistic basis for the non-homeostatic regulation of neural circuitry by a peripheral signal associated with tissue damage and stress. These findings provide opportunities to develop therapeutic agents for the treatment of disorders with altered energy demand.


Subject(s)
Body Weight/physiology , Brain Stem/metabolism , Glial Cell Line-Derived Neurotrophic Factor Receptors/metabolism , Growth Differentiation Factor 15/metabolism , Animals , Brain Stem/cytology , Brain Stem/drug effects , Central Amygdaloid Nucleus/cytology , Central Amygdaloid Nucleus/physiology , Eating/physiology , Energy Metabolism/physiology , Feeding Behavior , Female , Glial Cell Line-Derived Neurotrophic Factor Receptors/deficiency , Glial Cell Line-Derived Neurotrophic Factor Receptors/genetics , Growth Differentiation Factor 15/genetics , Growth Differentiation Factor 15/pharmacology , Homeostasis , Male , Mice , Mice, Knockout , Neurons/drug effects , Neurons/metabolism , Parabrachial Nucleus/cytology , Parabrachial Nucleus/physiology , Stress, Psychological
8.
Mol Genet Metab ; 121(2): 150-156, 2017 06.
Article in English | MEDLINE | ID: mdl-28457719

ABSTRACT

Glycosylasparaginase (GA) is an amidase that cleaves Asn-linked glycoproteins in lysosomes. Deficiency of this enzyme causes accumulation of glycoasparagines in lysosomes of cells, resulting in a genetic condition called aspartylglycosaminuria (AGU). To better understand the mechanism of a disease-causing mutation with a single residue change from a glycine to an aspartic acid, we generated a model mutant enzyme at the corresponding position (named G172D mutant). Here we report a 1.8Å resolution crystal structure of mature G172D mutant and analyzed the reason behind its low hydrolase activity. Comparison of mature G172D and wildtype GA models reveals that the presence of Asp 172 near the catalytic site affects substrate catabolism in mature G172D, making it less efficient in substrate processing. Also recent studies suggest that GA is capable of processing substrates that lack a chitobiose (Glycan, N-acetylchiobios, NAcGlc) moiety, by its exo-hydrolase activity. The mechanism for this type of catalysis is not yet clear. l-Aspartic acid ß-hydroxamate (ß-AHA) is a non-chitobiose substrate that is known to interact with GA. To study the underlying mechanism of non-chitobiose substrate processing, we built a GA-ß-AHA complex structure by comparing to a previously published G172D mutant precursor in complex with a ß-AHA molecule. A hydrolysis mechanism of ß-AHA by GA is proposed based on this complex model.


Subject(s)
Aspartylglucosaminuria/enzymology , Aspartylglucosylaminase/chemistry , Aspartylglucosylaminase/genetics , Disaccharides/metabolism , Mutation , Asparagine/analogs & derivatives , Asparagine/chemistry , Asparagine/metabolism , Aspartylglucosaminuria/metabolism , Aspartylglucosylaminase/metabolism , Biocatalysis , Crystallization , Crystallography, X-Ray , Glycopeptides/metabolism , Humans , Hydrolysis , Lysosomes/metabolism , Models, Molecular , Mutant Proteins/chemistry , Substrate Specificity
9.
Bioorg Med Chem Lett ; 25(14): 2818-23, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-26022843

ABSTRACT

Bromodomains are key transcriptional regulators that are thought to be druggable epigenetic targets for cancer, inflammation, diabetes and cardiovascular therapeutics. Of particular importance is the first of two bromodomains in bromodomain containing 4 protein (BRD4(1)). Protein-ligand docking in BRD4(1) was used to purchase a small, focused screening set of compounds possessing a large variety of core structures. Within this set, a small number of weak hits each contained a dihydroquinoxalinone ring system. We purchased other analogs with this ring system and further validated the new hit series and obtained improvement in binding inhibition. Limited exploration by new analog synthesis showed that the binding inhibition in a FRET assay could be improved to the low µM level making this new core a potential hit-to-lead series. Additionally, the predicted geometries of the initial hit and an improved analog were confirmed by X-ray co-crystallography with BRD4(1).


Subject(s)
Drug Design , Ligands , Nuclear Proteins/antagonists & inhibitors , Transcription Factors/antagonists & inhibitors , Binding Sites , Cell Cycle Proteins , Crystallography, X-Ray , Drug Evaluation, Preclinical , Humans , Molecular Docking Simulation , Nuclear Proteins/metabolism , Protein Binding , Protein Structure, Tertiary , Quinoxalines/chemistry , Quinoxalines/metabolism , Structure-Activity Relationship , Transcription Factors/metabolism
10.
Structure ; 22(12): 1855-1861, 2014 Dec 02.
Article in English | MEDLINE | ID: mdl-25456816

ABSTRACT

Aspartylglucosaminuria (AGU) is a lysosomal storage disease caused by a metabolic disorder of lysosomes to digest Asn-linked glycoproteins. The specific enzyme linked to AGU is a lysosomal hydrolase called glycosylasparaginase. Crystallographic studies revealed that a surface loop blocks the catalytic center of the mature hydrolase. Autoproteolysis is therefore required to remove this P loop and open up the hydrolase center. Nonetheless, AGU mutations result in misprocessing of their precursors and are deficient in hydrolyzing glycoasparagines. To understand the catalytic and structural consequences of AGU mutations, we have characterized two AGU models, one corresponding to a Finnish allele and the other found in a Canadian family. We also report a 2.1 Å resolution structure of the latter AGU model. The current crystallographic study provides a high-resolution structure of an AGU mutant. It reveals substantial conformation changes at the defective autocleavage site of the AGU mutant, which is trapped as an inactive precursor.


Subject(s)
Aspartylglucosaminuria/genetics , Aspartylglucosylaminase/genetics , Point Mutation , Crystallography, X-Ray , Humans , Models, Molecular
11.
Sci Rep ; 1: 186, 2011.
Article in English | MEDLINE | ID: mdl-22355701

ABSTRACT

Endoplasmic reticulum aminopeptidase 1 (ERAP1) is an essential component of the immune system, because it trims peptide precursors and generates the N--restricted epitopes. To examine ERAP1's unique properties of length- and sequence-dependent processing of antigen precursors, we report a 2.3 Å resolution complex structure of the ERAP1 regulatory domain. Our study reveals a binding conformation of ERAP1 to the carboxyl terminus of a peptide, and thus provides direct evidence for the molecular ruler mechanism.


Subject(s)
Aminopeptidases/chemistry , Aminopeptidases/physiology , Allosteric Site , Antigen Presentation , Antigens/chemistry , Binding Sites , Endoplasmic Reticulum/metabolism , Epitopes/chemistry , Histidine/chemistry , Histocompatibility Antigens Class I/chemistry , Humans , Minor Histocompatibility Antigens , Models, Molecular , Peptides/chemistry , Protein Binding , Protein Structure, Tertiary
12.
Article in English | MEDLINE | ID: mdl-20057058

ABSTRACT

The crystal structure of the hypothetical protein YqgQ from Bacillus subtilis has been determined to 2.1 A resolution. The crystals belonged to space group P2(1), with unit-cell parameters a = 51.85, b = 41.25, c = 55.18 A, beta = 113.4 degrees , and contained three protein molecules in the asymmetric unit. The structure was determined by the single-wavelength anomalous dispersion method using selenium-labeled protein and was refined to a final R factor of 24.7% (R(free) = 28.0%). The protein molecule mainly comprises a three-helical bundle. Its putative function is inferred to be single-stranded nucleic acid binding based on sequence and structural homology.


Subject(s)
Bacillus subtilis/chemistry , Bacterial Proteins/chemistry , Amino Acid Sequence , Crystallography, X-Ray , Models, Molecular , Molecular Sequence Data , Sequence Alignment
SELECTION OF CITATIONS
SEARCH DETAIL