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










Publication year range
1.
J Biol Chem ; 299(12): 105440, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37949227

ABSTRACT

In enterobacteria such as Escherichia coli, the general stress response is mediated by σs, the stationary phase dissociable promoter specificity subunit of RNA polymerase. σs is degraded by ClpXP during active growth in a process dependent on the RssB adaptor, which is thought to be stimulated by the phosphorylation of a conserved aspartate in its N-terminal receiver domain. Here we present the crystal structure of full-length RssB bound to a beryllofluoride phosphomimic. Compared to the structure of RssB bound to the IraD anti-adaptor, our new RssB structure with bound beryllofluoride reveals conformational differences and coil-to-helix transitions in the C-terminal region of the RssB receiver domain and in the interdomain segmented helical linker. These are accompanied by masking of the α4-ß5-α5 (4-5-5) "signaling" face of the RssB receiver domain by its C-terminal domain. Critically, using hydrogen-deuterium exchange mass spectrometry, we identify σs-binding determinants on the 4-5-5 face, implying that this surface needs to be unmasked to effect an interdomain interface switch and enable full σs engagement and hand-off to ClpXP. In activated receiver domains, the 4-5-5 face is often the locus of intermolecular interactions, but its masking by intramolecular contacts upon phosphorylation is unusual, emphasizing that RssB is a response regulator that undergoes atypical regulation.


Subject(s)
DNA-Binding Proteins , Endopeptidase Clp , Escherichia coli Proteins , Escherichia coli , Proteolysis , Sigma Factor , Transcription Factors , Crystallography, X-Ray , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Endopeptidase Clp/chemistry , Endopeptidase Clp/metabolism , Enzyme Activation , Escherichia coli/chemistry , Escherichia coli/enzymology , Escherichia coli/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Hydrogen Deuterium Exchange-Mass Spectrometry , Phosphorylation , Protein Domains , Sigma Factor/chemistry , Sigma Factor/metabolism , Transcription Factors/chemistry , Transcription Factors/metabolism
2.
Sci Signal ; 16(795): eadd9539, 2023 07 25.
Article in English | MEDLINE | ID: mdl-37490546

ABSTRACT

Precise synapse formation is essential for normal functioning of the nervous system. Retinal photoreceptors establish selective contacts with bipolar cells, aligning the neurotransmitter release apparatus with postsynaptic signaling cascades. This involves transsynaptic assembly between the dystroglycan-dystrophin complex on the photoreceptor and the orphan receptor GPR179 on the bipolar cell, which is mediated by the extracellular matrix protein pikachurin (also known as EGFLAM). This complex plays a critical role in the synaptic organization of photoreceptors and signal transmission, and mutations affecting its components cause blinding disorders in humans. Here, we investigated the structural organization and molecular mechanisms by which pikachurin orchestrates transsynaptic assembly and solved structures of the human pikachurin domains by x-ray crystallography and of the GPR179-pikachurin complex by single-particle, cryo-electron microscopy. The structures reveal molecular recognition principles of pikachurin by the Cache domains of GPR179 and show how the interaction is involved in the transsynaptic alignment of the signaling machinery. Together, these data provide a structural basis for understanding the synaptic organization of photoreceptors and ocular pathology.


Subject(s)
Extracellular Matrix Proteins , Synapses , Humans , Carrier Proteins/metabolism , Cryoelectron Microscopy , Extracellular Matrix Proteins/metabolism , Photoreceptor Cells/metabolism , Synapses/metabolism
3.
Plant Commun ; 4(6): 100639, 2023 Nov 13.
Article in English | MEDLINE | ID: mdl-37322867

ABSTRACT

Jasmonates (JAs) are plant hormones with crucial roles in development and stress resilience. They activate MYC transcription factors by mediating the proteolysis of MYC inhibitors called JAZ proteins. In the absence of JA, JAZ proteins bind and inhibit MYC through the assembly of MYC-JAZ-Novel Interactor of JAZ (NINJA)-TPL repressor complexes. However, JAZ and NINJA are predicted to be largely intrinsically unstructured, which has precluded their experimental structure determination. Through a combination of biochemical, mutational, and biophysical analyses and AlphaFold-derived ColabFold modeling, we characterized JAZ-JAZ and JAZ-NINJA interactions and generated models with detailed, high-confidence domain interfaces. We demonstrate that JAZ, NINJA, and MYC interface domains are dynamic in isolation and become stabilized in a stepwise order upon complex assembly. By contrast, most JAZ and NINJA regions outside of the interfaces remain highly dynamic and cannot be modeled in a single conformation. Our data indicate that the small JAZ Zinc finger expressed in Inflorescence Meristem (ZIM) motif mediates JAZ-JAZ and JAZ-NINJA interactions through separate surfaces, and our data further suggest that NINJA modulates JAZ dimerization. This study advances our understanding of JA signaling by providing insights into the dynamics, interactions, and structure of the JAZ-NINJA core of the JA repressor complex.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Arabidopsis Proteins/metabolism , Arabidopsis/genetics , Arabidopsis/metabolism , Repressor Proteins/genetics , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Cyclopentanes/metabolism
4.
J Immunother Cancer ; 10(11)2022 11.
Article in English | MEDLINE | ID: mdl-36442911

ABSTRACT

BACKGROUND: Despite numerous therapeutic options, safe and curative therapy is unavailable for most patients with chronic lymphocytic leukemia (CLL). A drawback of current therapies such as the anti-CD20 monoclonal antibody (mAb) rituximab is the elimination of all healthy B cells, resulting in impaired humoral immunity. We previously reported the identification of a patient-derived, CLL-binding mAb, JML-1, and identified sialic acid-binding immunoglobulin-like lectin-6 (Siglec-6) as the target of JML-1. Although little is known about Siglec-6, it appears to be an attractive target for cancer immunotherapy due to its absence on most healthy cells and tissues. METHODS: We used a target-specific approach to mine for additional patient-derived anti-Siglec-6 mAbs. To assess the therapeutic utility of targeting Siglec-6 in the context of CLL, T cell-recruiting bispecific antibodies (T-biAbs) that bind to Siglec-6 and CD3 were engineered into single-chain variable fragment-Fc and dual-affinity retargeting (DART)-Fc constructs. T-biAbs were evaluated for their activity in vitro, ex vivo, and in vivo. RESULTS: We discovered the anti-Siglec-6 mAbs RC-1 and RC-2, which bind with higher affinity than JML-1 yet maintain similar specificity. Both JML-1 and RC-1 T-biAbs were effective at activating T cells and killing Siglec-6+ target cells. The RC-1 clone in the DART-Fc format was the most potent T-biAb tested and was the only anti-Siglec-6 T-biAb that eliminated Siglec-6+ primary CLL cells via autologous T cells at pathological T-to-CLL cell ratios. Tested at healthy T-to-B cell ratios, it also eliminated a Siglec-6+ fraction of primary B cells from healthy donors. The subpicomolar potency of the DART-Fc format was attributed to the reduction in the length and flexibility of the cytolytic synapse. Furthermore, the RC-1 T-biAb was effective at clearing MEC1 CLL cells in vivo and demonstrated a circulatory half-life of over 7 days. CONCLUSION: Siglec-6-targeting T-biAbs are highly potent and specific for eliminating Siglec-6+ leukemic and healthy B cells while sparing Siglec-6- healthy B cells, suggesting a unique treatment strategy for CLL with diminished suppression of humoral immunity. Our data corroborate reports that T-biAb efficacy is dependent on synapse geometry and reveal that synapse architecture can be tuned via antibody engineering. Our fully human anti-Siglec-6 antibodies and T-biAbs have potential for cancer immunotherapy. TRIAL REGISTRATION NUMBER: NCT00923507.


Subject(s)
Leukemia, Lymphocytic, Chronic, B-Cell , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , T-Lymphocytes , B-Lymphocytes , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Immunotherapy
5.
Science ; 378(6619): 549-553, 2022 11 04.
Article in English | MEDLINE | ID: mdl-36378961

ABSTRACT

Cereblon (CRBN) is a ubiquitin ligase (E3) substrate receptor protein co-opted by CRBN E3 ligase modulatory drug (CELMoD) agents that target therapeutically relevant proteins for degradation. Prior crystallographic studies defined the drug-binding site within CRBN's thalidomide-binding domain (TBD), but the allostery of drug-induced neosubstrate binding remains unclear. We performed cryo-electron microscopy analyses of the DNA damage-binding protein 1 (DDB1)-CRBN apo complex and compared these structures with DDB1-CRBN in the presence of CELMoD compounds alone and complexed with neosubstrates. Association of CELMoD compounds to the TBD is necessary and sufficient for triggering CRBN allosteric rearrangement from an open conformation to the canonical closed conformation. The neosubstrate Ikaros only stably associates with the closed CRBN conformation, illustrating the importance of allostery for CELMoD compound efficacy and informing structure-guided design strategies to improve therapeutic efficacy.


Subject(s)
Adaptor Proteins, Signal Transducing , Ubiquitin-Protein Ligases , Adaptor Proteins, Signal Transducing/chemistry , Cryoelectron Microscopy , Thalidomide/chemistry , Ubiquitin-Protein Ligases/chemistry , Protein Domains , Allosteric Regulation
6.
Science ; 375(6576): 86-91, 2022 Jan 07.
Article in English | MEDLINE | ID: mdl-34793198

ABSTRACT

GPR158 is an orphan G protein­coupled receptor (GPCR) highly expressed in the brain, where it controls synapse formation and function. GPR158 has also been implicated in depression, carcinogenesis, and cognition. However, the structural organization and signaling mechanisms of GPR158 are largely unknown. We used single-particle cryo­electron microscopy (cryo-EM) to determine the structures of human GPR158 alone and bound to an RGS signaling complex. The structures reveal a homodimeric organization stabilized by a pair of phospholipids and the presence of an extracellular Cache domain, an unusual ligand-binding domain in GPCRs. We further demonstrate the structural basis of GPR158 coupling to RGS7-Gß5. Together, these results provide insights into the unusual biology of orphan receptors and the formation of GPCR-RGS complexes.


Subject(s)
GTP-Binding Protein beta Subunits/chemistry , RGS Proteins/chemistry , Receptors, G-Protein-Coupled/chemistry , Binding Sites , Cryoelectron Microscopy , GTP-Binding Protein beta Subunits/metabolism , Humans , Ligands , Models, Molecular , Phospholipids/chemistry , Protein Binding , Protein Conformation , Protein Conformation, alpha-Helical , Protein Domains , Protein Multimerization , Protein Subunits/chemistry , RGS Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction
7.
Nat Commun ; 12(1): 5451, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34521837

ABSTRACT

Circularized nandiscs (cNDs) exhibit superb monodispersity and have the potential to transform functional and structural studies of membrane proteins. In particular, cNDs can stabilize large patches of lipid bilayers for the reconstitution of complex membrane biochemical reactions, enabling the capture of crucial intermediates involved in synaptic transmission and viral entry. However, previous methods for building cNDs require multiple steps and suffer from low yields. We herein introduce a simple, one-step approach to ease the construction of cNDs using the SpyCatcher-SpyTag technology. This approach increases the yield of cNDs by over 10-fold and is able to rapidly generates cNDs with diameters ranging from 11 to over 100 nm. We demonstrate the utility of these cNDs for mechanistic interrogations of vesicle fusion and protein-lipid interactions that are unattainable using small nanodiscs. Together, the remarkable performance of SpyCatcher-SpyTag in nanodisc circularization paves the way for the use of cNDs in membrane biochemistry and structural biology.


Subject(s)
Lipid Bilayers/chemistry , Membrane Proteins/genetics , Nanostructures/chemistry , Peptides/genetics , Protein Engineering/methods , Cell Engineering/methods , Extracellular Vesicles/chemistry , Extracellular Vesicles/metabolism , Fluorescein-5-isothiocyanate/chemistry , Fluorescent Dyes/chemistry , Humans , Lipid Bilayers/metabolism , Membrane Fusion , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Nanostructures/ultrastructure , Oxadiazoles/chemistry , Particle Size , Peptides/chemistry , Peptides/metabolism , Phospholipids/chemistry , Phospholipids/metabolism , Plasmids/chemistry , Plasmids/metabolism , Staining and Labeling/methods
8.
J Mol Biol ; 433(22): 167258, 2021 11 05.
Article in English | MEDLINE | ID: mdl-34547329

ABSTRACT

The retinoic acid receptor-related orphan receptor γ (RORγ) is a ligand-dependent transcription factor of the nuclear receptor super family that underpins metabolic activity, immune function, and cancer progression. Despite being a valuable drug target in health and disease, our understanding of the ligand-dependent activities of RORγ is far from complete. Like most nuclear receptors, RORγ must recruit coregulatory protein to enact the RORγ target gene program. To date, a majority of structural studies have been focused exclusively on the RORγ ligand-binding domain and the ligand-dependent recruitment of small peptide segments of coregulators. Herein, we examine the ligand-dependent assembly of full length RORγ:coregulator complexes on cognate DNA response elements using structural proteomics and small angle x-ray scattering. The results from our studies suggest that RORγ becomes elongated upon DNA recognition, preventing long range interdomain crosstalk. We also determined that the DNA binding domain adopts a sequence-specific conformation, and that coregulatory protein may be able to 'sense' the ligand- and DNA-bound status of RORγ. We propose a model where ligand-dependent coregulator recruitment may be influenced by the sequence of the DNA to which RORγ is bound. Overall, the efforts described herein will illuminate important aspects of full length RORγ and monomeric orphan nuclear receptor target gene regulation through DNA-dependent conformational changes.


Subject(s)
Nuclear Receptor Coactivator 3/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/chemistry , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Response Elements , Animals , Binding Sites , DNA/metabolism , Female , Gene Expression Regulation , Humans , Mass Spectrometry/methods , Mice, Inbred BALB C , Nuclear Receptor Coactivator 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Protein Conformation , Scattering, Small Angle , X-Ray Diffraction
9.
Science ; 373(6553): 413-419, 2021 07 23.
Article in English | MEDLINE | ID: mdl-34437114

ABSTRACT

Adenosine monophosphate (AMP)-activated protein kinase (AMPK) regulates metabolism in response to the cellular energy states. Under energy stress, AMP stabilizes the active AMPK conformation, in which the kinase activation loop (AL) is protected from protein phosphatases, thus keeping the AL in its active, phosphorylated state. At low AMP:ATP (adenosine triphosphate) ratios, ATP inhibits AMPK by increasing AL dynamics and accessibility. We developed conformation-specific antibodies to trap ATP-bound AMPK in a fully inactive, dynamic state and determined its structure at 3.5-angstrom resolution using cryo-electron microscopy. A 180° rotation and 100-angstrom displacement of the kinase domain fully exposes the AL. On the basis of the structure and supporting biophysical data, we propose a multistep mechanism explaining how adenine nucleotides and pharmacological agonists modulate AMPK activity by altering AL phosphorylation and accessibility.


Subject(s)
AMP-Activated Protein Kinases/chemistry , AMP-Activated Protein Kinases/immunology , AMP-Activated Protein Kinases/metabolism , Adenosine Monophosphate/metabolism , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/metabolism , Cryoelectron Microscopy , Humans , Immunoglobulin Fab Fragments , Models, Molecular , Phosphorylation , Protein Conformation , Protein Domains , Protein Engineering
10.
Proc Natl Acad Sci U S A ; 118(35)2021 08 31.
Article in English | MEDLINE | ID: mdl-34452998

ABSTRACT

Efforts to improve estrogen receptor-α (ER)-targeted therapies in breast cancer have relied upon a single mechanism, with ligands having a single side chain on the ligand core that extends outward to determine antagonism of breast cancer growth. Here, we describe inhibitors with two ER-targeting moieties, one of which uses an alternate structural mechanism to generate full antagonism, freeing the side chain to independently determine other critical properties of the ligands. By combining two molecular targeting approaches into a single ER ligand, we have generated antiestrogens that function through new mechanisms and structural paradigms to achieve antagonism. These dual-mechanism ER inhibitors (DMERIs) cause alternate, noncanonical structural perturbations of the receptor ligand-binding domain (LBD) to antagonize proliferation in ER-positive breast cancer cells and in allele-specific resistance models. Our structural analyses with DMERIs highlight marked differences from current standard-of-care, single-mechanism antiestrogens. These findings uncover an enhanced flexibility of the ER LBD through which it can access nonconsensus conformational modes in response to DMERI binding, broadly and effectively suppressing ER activity.


Subject(s)
Breast Neoplasms/drug therapy , Estrogen Antagonists/chemistry , Estrogen Antagonists/pharmacology , Estrogen Receptor alpha/antagonists & inhibitors , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Crystallography, X-Ray , Female , Humans , Protein Binding , Protein Conformation , Structure-Activity Relationship , Tumor Cells, Cultured
11.
Bioorg Med Chem ; 42: 116246, 2021 07 15.
Article in English | MEDLINE | ID: mdl-34130216

ABSTRACT

We report the discovery of a fluorescent small molecule probe. This probe exhibits an emission increase in the presence of the oncoprotein MYC that can be attenuated by a competing inhibitor. Hydrogen-deuterium exchange mass spectrometry analysis, rationalized by induced-fit docking, suggests it binds to the "coiled-coil" region of the leucine zipper domain. Point mutations of this site produced functional MYC constructs resistant to inhibition in an oncogenic transformation assay by compounds that displace the probe. Utilizing this probe, we have developed a high-throughput assay to identify MYC inhibitor scaffolds. Screening of a diversity library (N = 1408, 384-well) and a library of pharmacologically active compounds (N = 1280, 1536-well) yielded molecules with greater drug-like properties than the probe. One lead is a potent inhibitor of oncogenic transformation and is specific for MYC relative to resistant mutants and transformation-inducing oncogenes. This method is simple, inexpensive, and does not require protein modification, DNA binding, or the dimer partner MAX. This assay presents an opportunity for MYC inhibition researchers to discover unique scaffolds.


Subject(s)
Drug Development , Fluorescent Dyes/pharmacology , High-Throughput Screening Assays , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Binding Sites/drug effects , Dose-Response Relationship, Drug , Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/chemistry , Humans , Molecular Structure , Proto-Oncogene Proteins c-myc/metabolism , Structure-Activity Relationship
12.
Nat Commun ; 12(1): 3239, 2021 05 28.
Article in English | MEDLINE | ID: mdl-34050165

ABSTRACT

The human mitochondrial AAA+ protein LONP1 is a critical quality control protease involved in regulating diverse aspects of mitochondrial biology including proteostasis, electron transport chain activity, and mitochondrial transcription. As such, genetic or aging-associated imbalances in LONP1 activity are implicated in pathologic mitochondrial dysfunction associated with numerous human diseases. Despite this importance, the molecular basis for LONP1-dependent proteolytic activity remains poorly defined. Here, we solved cryo-electron microscopy structures of human LONP1 to reveal the underlying molecular mechanisms governing substrate proteolysis. We show that, like bacterial Lon, human LONP1 adopts both an open and closed spiral staircase orientation dictated by the presence of substrate and nucleotide. Unlike bacterial Lon, human LONP1 contains a second spiral staircase within its ATPase domain that engages substrate as it is translocated toward the proteolytic chamber. Intriguingly, and in contrast to its bacterial ortholog, substrate binding within the central ATPase channel of LONP1 alone is insufficient to induce the activated conformation of the protease domains. To successfully induce the active protease conformation in substrate-bound LONP1, substrate binding within the protease active site is necessary, which we demonstrate by adding bortezomib, a peptidomimetic active site inhibitor of LONP1. These results suggest LONP1 can decouple ATPase and protease activities depending on whether AAA+ or both AAA+ and protease domains bind substrate. Importantly, our structures provide a molecular framework to define the critical importance of LONP1 in regulating mitochondrial proteostasis in health and disease.


Subject(s)
ATP-Dependent Proteases/ultrastructure , Mitochondrial Proteins/ultrastructure , ATP-Dependent Proteases/antagonists & inhibitors , ATP-Dependent Proteases/genetics , ATP-Dependent Proteases/metabolism , Adenosine Triphosphate/metabolism , Aging/metabolism , Bortezomib/pharmacology , Catalytic Domain/drug effects , Cryoelectron Microscopy , Enzyme Assays , Humans , Hydrolysis , Mitochondria/metabolism , Mitochondrial Proteins/antagonists & inhibitors , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Models, Molecular , Oxidation-Reduction , Protein Binding/drug effects , Protein Domains/genetics , Proteolysis , Proteostasis , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Recombinant Proteins/ultrastructure
13.
Proc Natl Acad Sci U S A ; 118(13)2021 03 30.
Article in English | MEDLINE | ID: mdl-33753480

ABSTRACT

Through dominant mutations, aminoacyl-tRNA synthetases constitute the largest protein family linked to Charcot-Marie-Tooth disease (CMT). An example is CMT subtype 2N (CMT2N), caused by individual mutations spread out in AlaRS, including three in the aminoacylation domain, thereby suggesting a role for a tRNA-charging defect. However, here we found that two are aminoacylation defective but that the most widely distributed R329H is normal as a purified protein in vitro and in unfractionated patient cell samples. Remarkably, in contrast to wild-type (WT) AlaRS, all three mutant proteins gained the ability to interact with neuropilin 1 (Nrp1), the receptor previously linked to CMT pathogenesis in GlyRS. The aberrant AlaRS-Nrp1 interaction is further confirmed in patient samples carrying the R329H mutation. However, CMT2N mutations outside the aminoacylation domain do not induce the Nrp1 interaction. Detailed biochemical and biophysical investigations, including X-ray crystallography, small-angle X-ray scattering, hydrogen-deuterium exchange (HDX), switchSENSE hydrodynamic diameter determinations, and protease digestions reveal a mutation-induced structural loosening of the aminoacylation domain that correlates with the Nrp1 interaction. The b1b2 domains of Nrp1 are responsible for the interaction with R329H AlaRS. The results suggest Nrp1 is more broadly associated with CMT-associated members of the tRNA synthetase family. Moreover, we revealed a distinct structural loosening effect induced by a mutation in the editing domain and a lack of conformational impact with C-Ala domain mutations, indicating mutations in the same protein may cause neuropathy through different mechanisms. Our results show that, as with other CMT-associated tRNA synthetases, aminoacylation per se is not relevant to the pathology.


Subject(s)
Alanine-tRNA Ligase/metabolism , Charcot-Marie-Tooth Disease/genetics , Neuropilin-1/metabolism , Alanine-tRNA Ligase/chemistry , Alanine-tRNA Ligase/genetics , Aminoacylation/genetics , Cells, Cultured , Charcot-Marie-Tooth Disease/blood , Crystallography, X-Ray , Deuterium Exchange Measurement , Humans , Lymphocytes , Mutation , Neuropilin-1/genetics , Primary Cell Culture , Protein Binding/genetics , Protein Domains/genetics , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Recombinant Proteins/ultrastructure , Scattering, Small Angle
14.
J Med Chem ; 64(2): 980-990, 2021 01 28.
Article in English | MEDLINE | ID: mdl-33434430

ABSTRACT

Despite a myriad of available pharmacotherapies for the treatment of type 2 diabetes (T2D), challenges still exist in achieving glycemic control. Several novel glucose-lowering strategies are currently under clinical investigation, highlighting the need for more robust treatments. Previously, we have shown that suppressing peroxisome proliferator-activated receptor gamma coactivator 1-alpha activity with a small molecule (SR18292, 16) can reduce glucose release from hepatocytes and ameliorate hyperglycemia in diabetic mouse models. Despite structural similarities in 16 to known ß-blockers, detailed structure-activity relationship studies described herein have led to the identification of analogues lacking ß-adrenergic activity that still maintain the ability to suppress glucagon-induced glucose release from hepatocytes and ameliorate hyperglycemia in diabetic mouse models. Hence, these compounds exert their biological effects in a mechanism that does not include adrenergic signaling. These probe molecules may lead to a new therapeutic approach to treat T2D either as a single agent or in combination therapy.


Subject(s)
Glucagon/antagonists & inhibitors , Gluconeogenesis/drug effects , Hypoglycemic Agents/pharmacology , Indoles/pharmacology , Propanols/pharmacology , Adipocytes, Brown/drug effects , Adipocytes, Brown/metabolism , Animals , Diabetes Mellitus, Type 2/drug therapy , Hepatocytes/drug effects , Hepatocytes/metabolism , Hypoglycemic Agents/chemistry , Indoles/chemistry , Lipolysis/drug effects , Liver Glycogen/metabolism , Male , Mice , Mice, Inbred C57BL , PPAR gamma/drug effects , Propanols/chemistry , Receptors, Adrenergic, beta/drug effects , Signal Transduction/drug effects , Structure-Activity Relationship , Sympathetic Nervous System/drug effects
15.
Anal Chem ; 92(16): 11018-11028, 2020 08 18.
Article in English | MEDLINE | ID: mdl-32658454

ABSTRACT

Hydrogen/Deuterium Exchange (HDX) coupled with Mass Spectrometry (HDX-MS) is a sensitive and robust method to probe protein conformational changes and protein-ligand interactions. HDX-MS relies on successful proteolytic digestion of target proteins under acidic conditions to localize perturbations in exchange behavior to protein structure. The ability of the protease to produce small peptides and overlapping fragments and provide sufficient coverage of the protein sequence is essential for localizing regions of interest. While the acid protease pepsin has been the enzyme of choice for HDX-MS studies, recently, it was shown that aspartic proteases from carnivorous pitcher plants of the genus Nepenthes are active under low-pH conditions and cleave at basic residues that are "forbidden" in peptic digests. In this report, we describe the utility of one of these enzymes, Nepenthesin II (NepII), in a HDX-MS workflow. A systematic and statistical analysis of data from 11 proteins (6391 amino acid residues) digested with immobilized porcine pepsin or NepII under conditions compatible with HDX-MS was performed to examine protease cleavage specificities. The cleavage of pepsin was most influenced by the amino acid residue at position P1. Phe, Leu, and Met are favored residues, each with a cleavage probability of greater than 40%. His, Lys, Arg, or Pro residues prohibit cleavage when found at the P1 position. In contrast, NepII offers advantageous cleavage to all basic residues and produces shortened peptides that could improve the spatial resolution in HDX-MS studies.


Subject(s)
Enzymes, Immobilized/chemistry , Pepsin A/chemistry , Proteolysis , Animals , Biocatalysis , Deuterium/chemistry , Deuterium Exchange Measurement , Mass Spectrometry , Sarraceniaceae/enzymology , Substrate Specificity , Swine
16.
Nat Commun ; 10(1): 5825, 2019 12 20.
Article in English | MEDLINE | ID: mdl-31862968

ABSTRACT

The repressive states of nuclear receptors (i.e., apo or bound to antagonists or inverse agonists) are poorly defined, despite the fact that nuclear receptors are a major drug target. Most ligand bound structures of nuclear receptors, including peroxisome proliferator-activated receptor γ (PPARγ), are similar to the apo structure. Here we use NMR, accelerated molecular dynamics and hydrogen-deuterium exchange mass spectrometry to define the PPARγ structural ensemble. We find that the helix 3 charge clamp positioning varies widely in apo and is stabilized by efficacious ligand binding. We also reveal a previously undescribed mechanism for inverse agonism involving an omega loop to helix switch which induces disruption of a tripartite salt-bridge network. We demonstrate that ligand binding can induce multiple structurally distinct repressive states. One state recruits peptides from two different corepressors, while another recruits just one, providing structural evidence of ligand bias in a nuclear receptor.


Subject(s)
Co-Repressor Proteins/metabolism , PPAR gamma/metabolism , Peptides/metabolism , Anilides/pharmacology , Benzamides/pharmacology , Binding Sites/drug effects , Binding Sites/genetics , Hydrogen Deuterium Exchange-Mass Spectrometry , Ligands , Magnetic Resonance Spectroscopy , Molecular Dynamics Simulation , Mutagenesis, Site-Directed , PPAR gamma/agonists , PPAR gamma/antagonists & inhibitors , PPAR gamma/ultrastructure , Protein Conformation, alpha-Helical/drug effects , Protein Conformation, alpha-Helical/genetics , Pyridines/pharmacology , Rosiglitazone/pharmacology
17.
Science ; 366(6470): 1255-1259, 2019 12 06.
Article in English | MEDLINE | ID: mdl-31806816

ABSTRACT

Enzyme-catalyzed reactions have begun to transform pharmaceutical manufacturing, offering levels of selectivity and tunability that can dramatically improve chemical synthesis. Combining enzymatic reactions into multistep biocatalytic cascades brings additional benefits. Cascades avoid the waste generated by purification of intermediates. They also allow reactions to be linked together to overcome an unfavorable equilibrium or avoid the accumulation of unstable or inhibitory intermediates. We report an in vitro biocatalytic cascade synthesis of the investigational HIV treatment islatravir. Five enzymes were engineered through directed evolution to act on non-natural substrates. These were combined with four auxiliary enzymes to construct islatravir from simple building blocks in a three-step biocatalytic cascade. The overall synthesis requires fewer than half the number of steps of the previously reported routes.


Subject(s)
Biocatalysis , Deoxyadenosines/chemistry , Reverse Transcriptase Inhibitors/chemistry , Biotechnology/methods , Pharmaceutical Preparations/chemical synthesis , Stereoisomerism
20.
Elife ; 82019 06 07.
Article in English | MEDLINE | ID: mdl-31172947

ABSTRACT

Members of the nuclear receptor (NR) superfamily regulate both physiological and pathophysiological processes ranging from development and metabolism to inflammation and cancer. Synthetic small molecules targeting NRs are often deployed as therapeutics to correct aberrant NR signaling or as chemical probes to explore the role of the receptor in physiology. Nearly half of NRs do not have specific cognate ligands (termed orphan NRs) and it's unclear if they possess ligand dependent activities. Here we demonstrate that ligand-dependent action of the orphan RORγ can be defined by selectively disrupting putative endogenous-but not synthetic-ligand binding. Furthermore, the characterization of a library of RORγ modulators reveals that structural dynamics of the receptor assessed by HDX-MS correlate with activity in biochemical and cell-based assays. These findings, corroborated with X-ray co-crystallography and site-directed mutagenesis, collectively reveal the structural determinants of RORγ activation, which is critical for designing RORγ agonists for cancer immunotherapy.


Subject(s)
Hydrogen Deuterium Exchange-Mass Spectrometry , Nuclear Receptor Subfamily 1, Group F, Member 3/agonists , Nuclear Receptor Subfamily 1, Group F, Member 3/chemistry , HEK293 Cells , Humans , Ligands , Models, Molecular , Nuclear Receptor Subfamily 1, Group F, Member 3/antagonists & inhibitors , Protein Binding , Static Electricity
SELECTION OF CITATIONS
SEARCH DETAIL
...