Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
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
11.
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
14.
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
15.
ACS Chem Biol ; 14(5): 1051-1062, 2019 05 17.
Article in English | MEDLINE | ID: mdl-30951276

ABSTRACT

Obesity and rheumatic disease are mechanistically linked via chronic inflammation. The orphan receptor TREM-1 (triggering receptor expressed on myeloid cells-1) is a potent amplifier of proinflammatory and noninfectious immune responses. Here, we show that the pan modulator SR1903 effectively blocks TREM-1 activation. SR1903 emerged from a chemical series of potent RORγ inverse agonists, although unlike close structural analogues, it has modest agonist activity on LXR and weak repressive activity (inverse agonism) of PPARγ, three receptors that play essential roles in inflammation and metabolism. The anti-inflammatory and antidiabetic efficacy of this unique modulator in collagen-induced arthritis and diet-induced obesity mouse models is demonstrated. Interestingly, in the context of obesity, SR1903 aided in the maintenance of the thymic homeostasis unlike selective RORγ inverse agonists. SR1903 was well-tolerated following chronic administration, and combined, these data suggest that it may represent a viable strategy for treatment of both metabolic and inflammatory disease. More importantly, the ability of SR1903 to block LPS signaling suggests the potential utility of this unique polypharmacological modulator for treatment of innate immune response disorders.


Subject(s)
Biphenyl Compounds/pharmacology , Inflammation/metabolism , Piperazines/pharmacology , Polypharmacology , Propanols/pharmacology , Receptors, Cytoplasmic and Nuclear/drug effects , Animals , Arthritis, Experimental/drug therapy , Biphenyl Compounds/therapeutic use , Diet , Disease Models, Animal , Drug Inverse Agonism , Ligands , Macrophages/drug effects , Macrophages/metabolism , Mice , Nuclear Receptor Subfamily 1, Group F, Member 3/agonists , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Obesity/drug therapy , Obesity/etiology , PPAR gamma/agonists , PPAR gamma/metabolism , Piperazines/therapeutic use , Propanols/therapeutic use , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction , Triggering Receptor Expressed on Myeloid Cells-1/metabolism
16.
J Biol Chem ; 294(3): 953-967, 2019 01 18.
Article in English | MEDLINE | ID: mdl-30478170

ABSTRACT

AMP-activated protein kinase (AMPK) is an attractive therapeutic target for managing metabolic diseases. A class of pharmacological activators, including Merck 991, binds the AMPK ADaM site, which forms the interaction surface between the kinase domain (KD) of the α-subunit and the carbohydrate-binding module (CBM) of the ß-subunit. Here, we report the development of two new 991-derivative compounds, R734 and R739, which potently activate AMPK in a variety of cell types, including ß2-specific skeletal muscle cells. Surprisingly, we found that they have only minor effects on direct kinase activity of the recombinant α1ß2γ1 isoform yet robustly enhance protection against activation loop dephosphorylation. This mode of activation is reminiscent of that of ADP, which activates AMPK by binding to the nucleotide-binding sites in the γ-subunit, more than 60 Å away from the ADaM site. To understand the mechanisms of full and partial AMPK activation, we determined the crystal structures of fully active phosphorylated AMPK α1ß1γ1 bound to AMP and R734/R739 as well as partially active nonphosphorylated AMPK bound to R734 and AMP and phosphorylated AMPK bound to R734 in the absence of added nucleotides at <3-Å resolution. These structures and associated analyses identified a novel conformational state of the AMPK autoinhibitory domain associated with partial kinase activity and provide new insights into phosphorylation-dependent activation loop stabilization in AMPK.


Subject(s)
AMP-Activated Protein Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/chemistry , Enzyme Activators/chemistry , AMP-Activated Protein Kinases/metabolism , Catalytic Domain , Hep G2 Cells , Humans , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
17.
Nat Commun ; 9(1): 5366, 2018 12 18.
Article in English | MEDLINE | ID: mdl-30560918

ABSTRACT

Retinoic acid inducible gene-I (RIG-I) ensures immune surveillance of viral RNAs bearing a 5'-triphosphate (5'ppp) moiety. Mutations in RIG-I (C268F and E373A) lead to impaired ATPase activity, thereby driving hyperactive signaling associated with autoimmune diseases. Here we report, using hydrogen/deuterium exchange, mechanistic models for dysregulated RIG-I proofreading that ultimately result in the improper recognition of cellular RNAs bearing 7-methylguanosine and N1-2'-O-methylation (Cap1) on the 5' end. Cap1-RNA compromises its ability to stabilize RIG-I helicase and blunts caspase activation and recruitment domains (CARD) partial opening by threefold. RIG-I H830A mutation restores Cap1-helicase engagement as well as CARDs partial opening event to a level comparable to that of 5'ppp. However, E373A RIG-I locks the receptor in an ATP-bound state, resulting in enhanced Cap1-helicase engagement and a sequential CARDs stimulation. C268F mutation renders a more tethered ring architecture and results in constitutive CARDs signaling in an ATP-independent manner.


Subject(s)
Autoimmunity/genetics , DEAD Box Protein 58/genetics , Immunity, Innate/genetics , RNA Caps/immunology , RNA, Double-Stranded/immunology , Adenosine Triphosphatases/metabolism , Caspase Activation and Recruitment Domain/immunology , DEAD Box Protein 58/chemistry , DEAD Box Protein 58/immunology , DEAD Box Protein 58/metabolism , Deuterium Exchange Measurement/methods , Gain of Function Mutation , Guanosine/analogs & derivatives , Guanosine/chemistry , Guanosine/immunology , Guanosine/metabolism , Interferon-Induced Helicase, IFIH1/immunology , Interferon-Induced Helicase, IFIH1/metabolism , Mass Spectrometry/methods , Methylation , Models, Molecular , Mutagenesis, Site-Directed , Protein Binding/genetics , Protein Binding/immunology , RNA Caps/chemistry , RNA Caps/metabolism , RNA, Double-Stranded/chemistry , RNA, Double-Stranded/metabolism , RNA, Viral/immunology , Receptors, Immunologic , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Signal Transduction/genetics , Signal Transduction/immunology
18.
Cell ; 175(7): 1756-1768.e17, 2018 12 13.
Article in English | MEDLINE | ID: mdl-30550785

ABSTRACT

Irisin is secreted by muscle, increases with exercise, and mediates certain favorable effects of physical activity. In particular, irisin has been shown to have beneficial effects in adipose tissues, brain, and bone. However, the skeletal response to exercise is less clear, and the receptor for irisin has not been identified. Here we show that irisin binds to proteins of the αV class of integrins, and biophysical studies identify interacting surfaces between irisin and αV/ß5 integrin. Chemical inhibition of the αV integrins blocks signaling and function by irisin in osteocytes and fat cells. Irisin increases both osteocytic survival and production of sclerostin, a local modulator of bone remodeling. Genetic ablation of FNDC5 (or irisin) completely blocks osteocytic osteolysis induced by ovariectomy, preventing bone loss and supporting an important role of irisin in skeletal remodeling. Identification of the irisin receptor should greatly facilitate our understanding of irisin's function in exercise and human health.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/metabolism , Bone Remodeling , Fibronectins/metabolism , Integrin alphaV/metabolism , Osteocytes/metabolism , Osteolysis/metabolism , Adipocytes/pathology , Animals , Cell Line, Tumor , Female , Fibronectins/genetics , HEK293 Cells , Humans , Integrin alphaV/genetics , Mice , Osteocytes/pathology , Osteolysis/genetics
19.
Elife ; 72018 12 12.
Article in English | MEDLINE | ID: mdl-30540250

ABSTRACT

Signaling by the G-protein-coupled receptors (GPCRs) plays fundamental role in a vast number of essential physiological functions. Precise control of GPCR signaling requires action of regulators of G protein signaling (RGS) proteins that deactivate heterotrimeric G proteins. RGS proteins are elaborately regulated and comprise multiple domains and subunits, yet structural organization of these assemblies is poorly understood. Here, we report a crystal structure and dynamics analyses of the multisubunit complex of RGS7, a major regulator of neuronal signaling with key roles in controlling a number of drug target GPCRs and links to neuropsychiatric disease, metabolism, and cancer. The crystal structure in combination with molecular dynamics and mass spectrometry analyses reveals unique organizational features of the complex and long-range conformational changes imposed by its constituent subunits during allosteric modulation. Notably, several intermolecular interfaces in the complex work in synergy to provide coordinated modulation of this key GPCR regulator.


Subject(s)
Carrier Proteins/chemistry , GTP-Binding Protein beta Subunits/chemistry , GTP-Binding Proteins/metabolism , Molecular Dynamics Simulation , Neurons/metabolism , RGS Proteins/chemistry , Amino Acid Sequence , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Crystallography, X-Ray , Deuterium Exchange Measurement , GTP-Binding Protein beta Subunits/genetics , GTP-Binding Protein beta Subunits/metabolism , Humans , Intracellular Signaling Peptides and Proteins , Mass Spectrometry , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Protein Conformation , Protein Multimerization , RGS Proteins/genetics , RGS Proteins/metabolism , Sequence Homology, Amino Acid , Sf9 Cells , Spodoptera
20.
Cell ; 175(1): 212-223.e17, 2018 09 20.
Article in English | MEDLINE | ID: mdl-30241607

ABSTRACT

CRISPR-Cas endonucleases directed against foreign nucleic acids mediate prokaryotic adaptive immunity and have been tailored for broad genetic engineering applications. Type VI-D CRISPR systems contain the smallest known family of single effector Cas enzymes, and their signature Cas13d ribonuclease employs guide RNAs to cleave matching target RNAs. To understand the molecular basis for Cas13d function and explain its compact molecular architecture, we resolved cryoelectron microscopy structures of Cas13d-guide RNA binary complex and Cas13d-guide-target RNA ternary complex to 3.4 and 3.3 Å resolution, respectively. Furthermore, a 6.5 Å reconstruction of apo Cas13d combined with hydrogen-deuterium exchange revealed conformational dynamics that have implications for RNA scanning. These structures, together with biochemical and cellular characterization, provide insights into its RNA-guided, RNA-targeting mechanism and delineate a blueprint for the rational design of improved transcriptome engineering technologies.


Subject(s)
CRISPR-Cas Systems/genetics , RNA, Guide, Kinetoplastida/physiology , Ribonucleases/physiology , CRISPR-Cas Systems/physiology , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Cryoelectron Microscopy/methods , Endonucleases/metabolism , HEK293 Cells , Humans , Molecular Conformation , RNA/genetics , RNA, Guide, Kinetoplastida/genetics , RNA, Guide, Kinetoplastida/ultrastructure , Ribonucleases/metabolism , Ribonucleases/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...