832 results on '"Dd"'
Search Results
2. The Formation of Collagen Hydroxylysine
- Author
-
Van Slyke Dd and Edwin A. Popenoe
- Subjects
chemistry.chemical_classification ,Hydroxylysine ,chemistry.chemical_compound ,chemistry ,Biochemistry ,Lysine ,Cell Biology ,Molecular Biology ,Amino acid - Published
- 1962
- Full Text
- View/download PDF
3. Determination of Carbon and Its Radioactivity
- Author
-
Van Slyke Dd and John Plazin
- Subjects
Range (particle radiation) ,Chemistry ,Analytical chemistry ,chemistry.chemical_element ,Cell Biology ,Biochemistry ,Methane ,chemistry.chemical_compound ,Qualitative analysis ,Transfer operation ,Transfer (computing) ,Carbon dioxide ,Carbon-14 ,Molecular Biology ,Carbon - Abstract
A micromanometric modification of the ninhydrin-CO/sub 2/ method for determining carboxyl CO/sub 2/ of alpha -amino acids is described together with transfer of the CO/sub 2/ to a Bernstein-Ballentine gas counter for counting C/ sup 14/O/sub 2/. The procedure serves for amounts of carboxyl carbon in the range 10 to 30 mu g. When CO/sub 2/ is transferred from a manornetric chamber to a gas counter, a small amount of CO/sub 2/, sufficient to be significant when the sample is less than 2 mg of carbon, may be trapped in the connecting tube between the manometric chamber and the counter. The trapped CO/sub 2/ can be quantitatively washed into the counter by a few milliliters of methane admitted into the mano- metric chamber after the usual transfer operation. (auth)
- Published
- 1960
- Full Text
- View/download PDF
4. APPLICATION OF SENDROY'S IODOMETRIC CHLORIDE TITRATION TO PROTEIN-CONTAINING FLUIDS
- Author
-
Van Slyke Dd and Alma Hiller
- Subjects
Gastric juices ,Chromatography ,Chemistry ,medicine ,Titration ,Cell Biology ,Molecular Biology ,Biochemistry ,Chloride ,medicine.drug - Published
- 1947
- Full Text
- View/download PDF
5. Esterification of cholesterol in high density lipoprotein decreases its ability to support ACTH-stimulated steroidogenesis by rat adrenocortical cells.
- Author
-
Gwynne, JT and Mahaffee, DD
- Abstract
Addition of high density lipoprotein 3 (HDL3) isolated from human plasma of d greater than 1.125 g/ml which had been preincubated for 24 h at 37 degrees C enhanced steroidogenesis by cultured rat adrenal cells only 38% as well as HDL3 isolated from unincubated plasma. Loss of steroidogenic activity due to preincubation was associated with a decrease in the percent HDL3 cholesterol remaining unesterified. Inhibition of lecithin-cholesterol acyltransferase activity by heating (60 degrees C, 1 h) or addition of dithionitrobenzoic acid (1.4 mM) prevented esterification of cholesterol in HDL and also prevented loss of steroidogenic activity. Although incubation of plasma of d greater than 1.125 g/ml prior to isolation caused cholesterol esterification, there was no change in the ratio of total cholesterol to protein in HDL, size and shape of the HDL particle as assayed by measurement of sedimentation velocity, nor affinity for the putative HDL receptor. Addition of unesterified cholesterol to preincubated HDL restored steroidogenic activity. These results indicate that unesterified cholesterol in HDL is preferentially used as substrate for rat adrenal steroidogenesis. The effects of nonlipoprotein serum proteins on HDL action in the adrenal were also examined. The ability of HDL3 to enhance rat adrenal steroidogenesis was not significantly less in serum-free media than in media supplemented with lipoprotein-poor fetal calf serum or human plasma of d greater than 1.21 g/ml, suggesting that rat adrenal uptake of HDL cholesterol does not depend on participation of plasma enzymes or transport proteins.
- Published
- 1987
- Full Text
- View/download PDF
6. Effects of urinary proteins from certain leukemics upon macromolecular synthesis and enzyme levels in bone marrow cultures.
- Author
-
Ross, DD, primary, Groth, DP, additional, and Kinkade, JM, additional
- Published
- 1975
- Full Text
- View/download PDF
7. HSP90 stabilizes visual cycle retinol dehydrogenase 5 in the endoplasmic reticulum by inhibiting its degradation during autophagy.
- Author
-
Jia X, Wang Y, Jiang M, Chen DD, Shang G, Liu B, Xue M, Lang Y, Zhou G, Dong Y, Zhang F, Peng X, and Hu Y
- Abstract
Genetic mutations in retinol dehydrogenase 5 (RDH5), a rate-limiting enzyme of the visual cycle, is associated with nyctalopia, AMD and stationary congenital fundus albipunctatus (FA). A majority of these mutations impair RDH5 protein expression and intracellular localization. However, the regulatory mechanisms underlying RDH5 metabolism remain unclear. Here, we find that RDH5 undergoes degradation via the autophagy-lysosomal pathway, and its stability is regulated by interacting with HSP90. Deletion of HSP90α or HSP90β by CRISPR-Cas9 or inhibition of HSP90 activity by IPI-504 down-regulates RDH5 protein level, but not its mRNA expression, and this downregulation is restored by autophagic inhibitors (3-MA, CQ and Baf-A1) and siRNA of ATG5 or ATG7, but not by the proteasome inhibitor MG132. RDH5 can physically interact with SQSTM1/P62, and this interaction is enhanced in HSP90-deficient cells as well as in CQ-treated cells. Knocking down SQSTM1/P62 by siRNA induces RDH5 protein accumulation. Moreover, HSP90, RDH5 and Calnexin form a complex through intermolecular interactions. Deficiency of HSP90α or HSP90β dissociates RDH5 from Calnexin, and increases RDH5 translocation from the endoplasmic reticulum (ER) to the cytosol. Taken together, we propose that dysfunction of HSP90 leads to RDH5 release from Calnexin in the ER into the cytosol, where it binds to the adaptor SQSTM1/P62 for degradation in the autolysosome. RDH5 is a novel client candidate of HSP90. The downregulation of RDH5 may be responsible for the nyctalopia side effect noted in cancer patients receiving HSP90 inhibitor treatment currently in the clinical trial., Competing Interests: Conflict of interest The authors declare no conflict of interest to the research, authorship, and publication of this article., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
8. G-site residue S67 is involved in the fungicide-degrading activity of a tau class glutathione S-transferase from Carica papaya.
- Author
-
Wang SY, Wang YX, Yue SS, Shi XC, Lu FY, Wu SQ, Herrera-Balandrano DD, and Laborda P
- Subjects
- Fungicides, Industrial metabolism, Mutagenesis, Site-Directed, Plant Proteins chemistry, Plant Proteins genetics, Plant Proteins metabolism, Escherichia coli genetics, Recombinant Proteins genetics, Recombinant Proteins metabolism, Carica enzymology, Carica genetics, Glutathione Transferase metabolism, Glutathione Transferase genetics, Glutathione Transferase chemistry, Thiram metabolism
- Abstract
Thiram is a toxic fungicide extensively used for the management of pathogens in fruits. Although it is known that thiram degrades in plant tissues, the key enzymes involved in this process remain unexplored. In this study, we report that a tau class glutathione S-transferase (GST) from Carica papaya can degrade thiram. This enzyme was easily obtained by heterologous expression in Escherichia coli, showed low promiscuity toward other thiuram disulfides, and catalyzed thiram degradation under physiological reaction conditions. Site-directed mutagenesis indicated that G-site residue S67 shows a key influence for the enzymatic activity toward thiram, while mutation of residue S13, which reduced the GSH oxidase activity, did not significantly affect the thiram-degrading activity. The formation of dimethyl dithiocarbamate, which was subsequently converted into carbon disulfide, and dimethyl dithiocarbamoylsulfenic acid as the thiram degradation products suggested that thiram undergoes an alkaline hydrolysis that involves the rupture of the disulfide bond. Application of the GST selective inhibitor 4-chloro-7-nitro-2,1,3-benzoxadiazole reduced papaya peel thiram-degrading activity by 95%, indicating that this is the main degradation route of thiram in papaya. GST from Carica papaya also catalyzed the degradation of the fungicides chlorothalonil and thiabendazole, with residue S67 showing again a key influence for the enzymatic activity. These results fill an important knowledge gap in understanding the catalytic promiscuity of plant GSTs and reveal new insights into the fate and degradation products of thiram in fruits., Competing Interests: Conflict of interest The authors declare that they have no conflicts of interest with the contents of this article., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
9. Discovery and structural characterization of the D-box, a conserved TonB motif that couples an inner-membrane motor to outer-membrane transport.
- Author
-
Loll PJ, Grasty KC, Shultis DD, Guzman NJ, and Wiener MC
- Subjects
- Biological Transport, Escherichia coli metabolism, Escherichia coli Proteins metabolism, Membrane Transport Proteins metabolism, Protein Binding, Bacterial Proteins chemistry, Bacterial Proteins metabolism, Membrane Proteins chemistry, Membrane Proteins metabolism
- Abstract
Gram-negative bacteria use TonB-dependent transport to take up nutrients from the external environment, employing the Ton complex to import a variety of nutrients that are either scarce or too large to cross the outer membrane unaided. The Ton complex contains an inner-membrane motor (ExbBD) that generates force, as well as nutrient-specific transport proteins on the outer membrane. These two components are coupled by TonB, which transmits the force from the inner to the outer membrane. TonB contains an N-terminus anchored in the inner membrane, a C-terminal domain that binds the outer-membrane transporter, and a proline-rich linker connecting the two. While much is known about the interaction between TonB and outer-membrane transporters, the critical interface between TonB and ExbBD is less well understood. Here, we identify a conserved motif within TonB that we term the D-box, which serves as an attachment point for ExbD. We characterize the interaction between ExbD and the D-box both functionally and structurally, showing that a homodimer of ExbD captures one copy of the D-box peptide via beta-strand recruitment. We additionally show that both the D-box motif and ExbD are conserved in a range of Gram-negative bacteria, including members of the ESKAPE group of pathogens. The ExbD:D-box interaction is likely to represent an important aspect of force transduction between the inner and outer membranes. Given that TonB-dependent transport is an important contributor to virulence, this interaction is an intriguing potential target for novel antibacterial therapies., Competing Interests: Conflict of interest The authors declare that they have no conflicts of interest with the contents of this article. One author is an editorial board member for the Journal of Biological Chemistry and was not involved in the editorial review or the decision to publish this article., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
10. Ketone bodies promote epididymal white adipose expansion to alleviate liver steatosis in response to a ketogenic diet.
- Author
-
Zhao MF, Zhang XG, Tang YP, Zhu YX, Nie HY, Bu DD, Fang L, and Li CJ
- Subjects
- Humans, Lipids, Liver metabolism, PPAR gamma metabolism, Adipose Tissue, White metabolism, Diet, Ketogenic, Fatty Liver metabolism, Ketone Bodies metabolism
- Abstract
Liver can sense the nutrient status and send signals to other organs to regulate overall metabolic homoeostasis. Herein, we demonstrate that ketone bodies act as signals released from the liver that specifically determine the distribution of excess lipid in epididymal white adipose tissue (eWAT) when exposed to a ketogenic diet (KD). An acute KD can immediately result in excess lipid deposition in the liver. Subsequently, the liver sends the ketone body β-hydroxybutyrate (BHB) to regulate white adipose expansion, including adipogenesis and lipogenesis, to alleviate hepatic lipid accumulation. When ketone bodies are depleted by deleting 3-hydroxy-3-methylglutaryl-CoA synthase 2 gene in the liver, the enhanced lipid deposition in eWAT but not in inguinal white adipose tissue is preferentially blocked, while lipid accumulation in liver is not alleviated. Mechanistically, ketone body BHB can significantly decrease lysine acetylation of peroxisome proliferator-activated receptor gamma in eWAT, causing enhanced activity of peroxisome proliferator-activated receptor gamma, the key adipogenic transcription factor. These observations suggest that the liver senses metabolic stress first and sends a corresponding signal, that is, ketone body BHB, to specifically promote eWAT expansion to adapt to metabolic challenges., Competing Interests: Conflict of interest The authors declare that they have no conflicts of interest with the contents of this article., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
11. Drug discovery for heart failure targeting myosin-binding protein C.
- Author
-
Bunch TA, Guhathakurta P, Thompson AR, Lepak VC, Carter AL, Thomas JJ, Thomas DD, and Colson BA
- Subjects
- Humans, Actins metabolism, Myocardium metabolism, Myosins metabolism, Phosphorylation drug effects, Protein Binding drug effects, Drug Evaluation, Preclinical, Biosensing Techniques, Adenosine Triphosphatases metabolism, Muscle, Skeletal metabolism, Recombinant Proteins metabolism, Enzyme Activation drug effects, Fluorescence Resonance Energy Transfer, Drug Discovery methods, Heart Failure drug therapy, Heart Failure metabolism, Small Molecule Libraries pharmacology, Myofibrils drug effects, Carrier Proteins metabolism
- Abstract
Cardiac MyBP-C (cMyBP-C) interacts with actin and myosin to fine-tune cardiac muscle contractility. Phosphorylation of cMyBP-C, which reduces the binding of cMyBP-C to actin and myosin, is often decreased in patients with heart failure (HF) and is cardioprotective in model systems of HF. Therefore, cMyBP-C is a potential target for HF drugs that mimic its phosphorylation and/or perturb its interactions with actin or myosin. We labeled actin with fluorescein-5-maleimide (FMAL) and the C0-C2 fragment of cMyBP-C (cC0-C2) with tetramethylrhodamine (TMR). We performed two complementary high-throughput screens (HTS) on an FDA-approved drug library, to discover small molecules that specifically bind to cMyBP-C and affect its interactions with actin or myosin, using fluorescence lifetime (FLT) detection. We first excited FMAL and detected its FLT, to measure changes in fluorescence resonance energy transfer (FRET) from FMAL (donor) to TMR (acceptor), indicating binding. Using the same samples, we then excited TMR directly, using a longer wavelength laser, to detect the effects of compounds on the environmentally sensitive FLT of TMR, to identify compounds that bind directly to cC0-C2. Secondary assays, performed on selected modulators with the most promising effects in the primary HTS assays, characterized the specificity of these compounds for phosphorylated versus unphosphorylated cC0-C2 and for cC0-C2 versus C1-C2 of fast skeletal muscle (fC1-C2). A subset of identified compounds modulated ATPase activity in cardiac and/or skeletal myofibrils. These assays establish the feasibility of the discovery of small-molecule modulators of the cMyBP-C-actin/myosin interaction, with the ultimate goal of developing therapies for HF., Competing Interests: Conflict of interest D. D. T. holds equity in, and serves as President of, Photonic Pharma LLC. This relationship has been reviewed and managed by the University of Minnesota. The present research is a pre-commercial collaboration between Photonic Pharma, UMN, and the University of Arizona. B. A. C. serves as President of BC Biologics LLC. This relationship has been reviewed and managed by the University of Arizona. BC Biologics had no role in this study. B. A. C. filed a PCT patent application based on this work (patent pending, serial no. PCT/US21/14,142). The other authors declare no competing financial interests., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
12. A detailed kinetic model of glycolysis in Plasmodium falciparum-infected red blood cells for antimalarial drug target identification.
- Author
-
van Niekerk DD, du Toit F, Green K, Palm D, and Snoep JL
- Subjects
- Humans, Acidosis, Lactic, Glucose metabolism, Hypoglycemia, Kinetics, Trophozoites pathogenicity, Trophozoites physiology, Parasite Load, Antimalarials pharmacology, Antimalarials therapeutic use, Antimalarials metabolism, Erythrocytes drug effects, Erythrocytes metabolism, Erythrocytes parasitology, Glycolysis drug effects, Malaria, Falciparum metabolism, Malaria, Falciparum parasitology, Plasmodium falciparum metabolism, Plasmodium falciparum pathogenicity, Plasmodium falciparum physiology, Molecular Targeted Therapy methods, Models, Biological
- Abstract
Upon infection by the malaria parasite Plasmodium falciparum, the glycolytic rate of a red blood cell increases up to 100-fold, possibly contributing to lactic acidosis and hypoglycemia in patients with severe malaria. This dramatic increase in glucose uptake and metabolism was correctly predicted by a newly constructed detailed enzyme kinetic model of glucose metabolism in the trophozoite-infected red blood cell. Subsequently, we expanded the model to simulate an infected red blood cell culture, including the different asexual blood-stage forms of the malaria parasite. The model simulations were in good agreement with experimental data, for which the measured parasitic volume was an important parameter. Upon further analysis of the model, we identified glucose transport as a drug target that would specifically affect infected red blood cells, which was confirmed experimentally with inhibitor titrations. This model can be a first step in constructing a whole-body model for glucose metabolism in malaria patients to evaluate the contribution of the parasite's metabolism to the disease state., Competing Interests: Conflict of interest The authors declare that they have no conflicts of interest with the contents of this article., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
13. Redundant electrostatic interactions between GATOR1 and the Rag GTPase heterodimer drive efficient amino acid sensing in human cells.
- Author
-
Doxsey DD, Tettoni SD, Egri SB, and Shen K
- Subjects
- Humans, Lysosomes metabolism, Nucleotides metabolism, Signal Transduction physiology, Static Electricity, Amino Acids metabolism, Mechanistic Target of Rapamycin Complex 1 metabolism, Monomeric GTP-Binding Proteins metabolism
- Abstract
Cells need to coordinate nutrient availability with their growth and proliferation. In eukaryotic cells, this coordination is mediated by the mechanistic target of the rapamycin complex 1 (mTORC1) pathway. mTORC1 activation is regulated by two GTPase units, the Rag GTPase heterodimer and the Rheb GTPase. The RagA-RagC heterodimer controls the subcellular localization of mTORC1, and its nucleotide loading states are strictly controlled by upstream regulators including amino acid sensors. A critical negative regulator of the Rag GTPase heterodimer is GATOR1. In the absence of amino acids, GATOR1 stimulates GTP hydrolysis by the RagA subunit to turn off mTORC1 signaling. Despite the enzymatic specificity of GATOR1 to RagA, a recent cryo-EM structural model of the human GATOR1-Rag-Ragulator complex reveals an unexpected interface between Depdc5, a subunit of GATOR1, and RagC. Currently, there is no functional characterization of this interface, nor do we know its biological relevance. Here, combining structure-function analysis, enzymatic kinetic measurements, and cell-based signaling assays, we identified a critical electrostatic interaction between Depdc5 and RagC. This interaction is mediated by the positively charged Arg-1407 residue on Depdc5 and a patch of negatively charged residues on the lateral side of RagC. Abrogating this interaction impairs the GAP activity of GATOR1 and cellular response to amino acid withdrawal. Our results reveal how GATOR1 coordinates the nucleotide loading states of the Rag GTPase heterodimer, and thus precisely controls cellular behavior in the absence of amino acids., Competing Interests: Conflict of interest The authors declare that they have no conflicts of interest with the contents of this article., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
14. Disruption of CDK7 signaling leads to catastrophic chromosomal instability coupled with a loss of condensin-mediated chromatin compaction.
- Author
-
Piemonte KM, Webb BM, Bobbitt JR, Majmudar PR, Cuellar-Vite L, Bryson BL, Latina NC, Seachrist DD, and Keri RA
- Subjects
- Mitosis genetics, Chromosomal Instability genetics, Humans, Cell Line, Tumor, Gene Expression Regulation genetics, Cell Cycle Proteins genetics, Cell Cycle Proteins metabolism, Gene Silencing, Chromatin genetics, Chromatin metabolism, Cyclin-Dependent Kinases genetics, Cyclin-Dependent Kinases metabolism, Signal Transduction
- Abstract
Chromatin organization is highly dynamic and modulates DNA replication, transcription, and chromosome segregation. Condensin is essential for chromosome assembly during mitosis and meiosis, as well as maintenance of chromosome structure during interphase. While it is well established that sustained condensin expression is necessary to ensure chromosome stability, the mechanisms that control its expression are not yet known. Herein, we report that disruption of cyclin-dependent kinase 7 (CDK7), the core catalytic subunit of CDK-activating kinase, leads to reduced transcription of several condensin subunits, including structural maintenance of chromosomes 2 (SMC2). Live and static microscopy revealed that inhibiting CDK7 signaling prolongs mitosis and induces chromatin bridge formation, DNA double-strand breaks, and abnormal nuclear features, all of which are indicative of mitotic catastrophe and chromosome instability. Affirming the importance of condensin regulation by CDK7, genetic suppression of the expression of SMC2, a core subunit of this complex, phenocopies CDK7 inhibition. Moreover, analysis of genome-wide chromatin conformation using Hi-C revealed that sustained activity of CDK7 is necessary to maintain chromatin sublooping, a function that is ascribed to condensin. Notably, the regulation of condensin subunit gene expression is independent of superenhancers. Together, these studies reveal a new role for CDK7 in sustaining chromatin configuration by ensuring the expression of condensin genes, including SMC2., Competing Interests: Conflict of interest The authors declare that they have no conflicts of interest with the contents of this article., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
15. A selective nonpeptide somatostatin receptor 5 agonist effectively decreases insulin secretion in hyperinsulinism.
- Author
-
Juliana CA, Chai J, Arroyo P, Rico-Bautista E, Betz SF, and De León DD
- Subjects
- Animals, Child, Humans, Infant, Mice, Adenosine Triphosphate metabolism, Amino Acids metabolism, Glucose metabolism, Hypoglycemia metabolism, Insulin metabolism, Insulin Secretion, Islets of Langerhans metabolism, Mutation, Potassium Channels, Inwardly Rectifying metabolism, Hyperinsulinism drug therapy, Receptors, Somatomedin agonists
- Abstract
Congenital hyperinsulinism (HI), a beta cell disorder most commonly caused by inactivating mutations of beta cell K
ATP channels, results in dysregulated insulin secretion and persistent hypoglycemia. Children with KATP -HI are unresponsive to diazoxide, the only FDA-approved drug for HI, and utility of octreotide, the second-line therapy, is limited because of poor efficacy, desensitization, and somatostatin receptor type 2 (SST2)-mediated side effects. Selective targeting of SST5, an SST receptor associated with potent insulin secretion suppression, presents a new avenue for HI therapy. Here, we determined that CRN02481, a highly selective nonpeptide SST5 agonist, significantly decreased basal and amino acid-stimulated insulin secretion in both Sur1-/- (a model for KATP -HI) and wild-type mouse islets. Oral administration of CRN02481 significantly increased fasting glucose and prevented fasting hypoglycemia compared to vehicle in Sur1-/- mice. During a glucose tolerance test, CRN02481 significantly increased glucose excursion in both WT and Sur1-/- mice compared to the control. CRN02481 also reduced glucose- and tolbutamide-stimulated insulin secretion from healthy, control human islets similar to the effects observed with SS14 and peptide somatostatin analogs. Moreover, CRN02481 significantly decreased glucose- and amino acid-stimulated insulin secretion in islets from two infants with KATP -HI and one with Beckwith-Weideman Syndrome-HI. Taken together, these data demonstrate that a potent and selective SST5 agonist effectively prevents fasting hypoglycemia and suppresses insulin secretion not only in a KATP -HI mouse model but also in healthy human islets and islets from HI patients., Competing Interests: Conflict of interest P. A., E. R. B. and S. F. B work for Crinetic Pharmaceuticals. D. D. D. L. has received consulting fees from Crinetics Pharmaceuticals, Zealand Pharma, Hanmi Pharmaceutical, Eiger Biopharmaceuticals, and Heptares Therapeutics. D. D. D. L. has received research funding from Tiburio Therapeutics, and Twist Pharma for studies not included in this manuscript., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
16. Early-phase drug discovery of β-III-spectrin actin-binding modulators for treatment of spinocerebellar ataxia type 5.
- Author
-
Guhathakurta P, Rebbeck RT, Denha SA, Keller AR, Carter AL, Atang AE, Svensson B, Thomas DD, Hays TS, and Avery AW
- Subjects
- Humans, Drug Discovery, Neurons metabolism, Actins genetics, Actins metabolism, Spectrin metabolism, Spinocerebellar Ataxias drug therapy, Spinocerebellar Ataxias genetics, Spinocerebellar Ataxias metabolism
- Abstract
β-III-Spectrin is a key cytoskeletal protein that localizes to the soma and dendrites of cerebellar Purkinje cells and is required for dendritic arborization and signaling. A spinocerebellar ataxia type 5 L253P mutation in the cytoskeletal protein β-III-spectrin causes high-affinity actin binding. Previously we reported a cell-based fluorescence assay for identification of small-molecule actin-binding modulators of the L253P mutant β-III-spectrin. Here we describe a complementary, in vitro, fluorescence resonance energy transfer (FRET) assay that uses purified L253P β-III-spectrin actin-binding domain (ABD) and F-actin. To validate the assay for high-throughput compatibility, we first confirmed that our 50% FRET signal was responsive to swinholide A, an actin-severing compound, and that this yielded excellent assay quality with a Z' value > 0.77. Second, we screened a 2684-compound library of US Food and Drug Administration-approved drugs. Importantly, the screening identified numerous compounds that decreased FRET between fluorescently labeled L253P ABD and F-actin. The activity and target of multiple Hit compounds were confirmed in orthologous cosedimentation actin-binding assays. Through future medicinal chemistry, the Hit compounds can potentially be developed into a spinocerebellar ataxia type 5-specific therapeutic. Furthermore, our validated FRET-based in vitro high-throughput screening platform is poised for screening large compound libraries for β-III-spectrin ABD modulators., Competing Interests: Conflict of interest D. D. T. holds equity in, and serves as an executive officer for, Photonic Pharma LLC. These relationships have been reviewed and managed by the University of Minnesota. Photonic Pharma had no role in this study, except to provide some instrumentation, as stated in Experimental Procedures., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
17. Next-generation retinoid X receptor agonists increase ATRA signaling in organotypic epithelium cultures and have distinct effects on receptor dynamics.
- Author
-
Melo N, Belyaeva OV, Berger WK, Halasz L, Yu J, Pilli N, Yang Z, Klyuyeva AV, Elmets CA, Atigadda V, Muccio DD, Kane MA, Nagy L, Kedishvili NY, and Renfrow MB
- Subjects
- Humans, Retinoid X Receptors metabolism, Bexarotene, Ligands, Epidermis metabolism, Tetrahydronaphthalenes pharmacology, Tretinoin pharmacology, Tretinoin metabolism
- Abstract
Retinoid X receptors (RXRs) are nuclear transcription factors that partner with other nuclear receptors to regulate numerous physiological processes. Although RXR represents a valid therapeutic target, only a few RXR-specific ligands (rexinoids) have been identified, in part due to the lack of clarity on how rexinoids selectively modulate RXR response. Previously, we showed that rexinoid UAB30 potentiates all-trans-retinoic acid (ATRA) signaling in human keratinocytes, in part by stimulating ATRA biosynthesis. Here, we examined the mechanism of action of next-generation rexinoids UAB110 and UAB111 that are more potent in vitro than UAB30 and the FDA-approved Targretin. Both UAB110 and UAB111 enhanced ATRA signaling in human organotypic epithelium at a 50-fold lower concentration than UAB30. This was consistent with the 2- to 5- fold greater increase in ATRA in organotypic epidermis treated with UAB110/111 versus UAB30. Furthermore, at 0.2 μM, UAB110/111 increased the expression of ATRA genes up to 16-fold stronger than Targretin. The less toxic and more potent UAB110 also induced more changes in differential gene expression than Targretin. Additionally, our hydrogen deuterium exchange mass spectrometry analysis showed that both ligands reduced the dynamics of the ligand-binding pocket but also induced unique dynamic responses that were indicative of higher affinity binding relative to UAB30, especially for Helix 3. UAB110 binding also showed increased dynamics towards the dimer interface through the Helix 8 and Helix 9 regions. These data suggest that UAB110 and UAB111 are potent activators of RXR-RAR signaling pathways but accomplish activation through different molecular responses to ligand binding., Competing Interests: Conflict of interest The authors declare that they have no conflicts of interest with the contents of this article., (Copyright © 2022 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
18. Enhancing interaction of actin and actin-binding domain 1 of dystrophin with modulators: Toward improved gene therapy for Duchenne muscular dystrophy.
- Author
-
Guhathakurta P, Carter AL, Thompson AR, Kurila D, LaFrence J, Zhang L, Trask JR, Vanderheyden B, Muretta JM, Ervasti JM, and Thomas DD
- Subjects
- Humans, Genetic Therapy, Protein Binding drug effects, Small Molecule Libraries chemistry, Small Molecule Libraries metabolism, Small Molecule Libraries pharmacology, Protein Domains, Actins metabolism, Dystrophin genetics, Dystrophin chemistry, Muscular Dystrophy, Duchenne genetics, Muscular Dystrophy, Duchenne therapy
- Abstract
Duchenne muscular dystrophy is a lethal muscle disease, caused by mutations in the gene encoding dystrophin, an actin-binding cytoskeletal protein. Absence of functional dystrophin results in muscle weakness and degeneration, eventually leading to cardiac and respiratory failure. Strategies to replace the missing dystrophin via gene therapy have been intensively pursued. However, the dystrophin gene is too large for current gene therapy approaches. Currently available micro-dystrophin constructs lack the actin-binding domain 2 and show decreased actin-binding affinity in vitro compared to full-length dystrophin. Thus, increasing the actin-binding affinity of micro-dystrophin, using small molecules, could be a beneficial therapeutic approach. Here, we have developed and validated a novel high-throughput screening (HTS) assay to discover small molecules that increase the binding affinity of dystrophin's actin-binding domain 1 (ABD1). We engineered a novel FRET biosensor, consisting of the mClover3, fluorescent protein (donor) attached to the C-terminus of dystrophin ABD1, and Alexa Fluor 568 (acceptor) attached to the C-terminal cysteine of actin. We used this biosensor in small-molecule screening, using a unique high-precision, HTS fluorescence lifetime assay, identifying several compounds from an FDA-approved library that significantly increase the binding between actin and ABD1. This HTS assay establishes feasibility for the discovery of small-molecule modulators of the actin-dystrophin interaction, with the ultimate goal of developing therapies for muscular dystrophy., Competing Interests: Conflict of interest D. D. T. holds equity in and serves as President of Photonic Pharma LLC. This relationship has been reviewed and managed by the University of Minnesota. Photonic Pharma had no role in this study, except to provide instrumentation, as stated in Experimental procedures. J. M. M., J. M. E., and D. D. T. are entitled to royalties from Sarepta Therapeutics, the company sponsoring this research project. This royalty interest has been reviewed and managed according to the University of Minnesota's conflict of interest policies. J. M. E. has received compensation for consulting for Sarepta. This relationship has been reviewed and managed by the University of Minnesota in accordance with its conflict of interest polices. All other authors declare no conflicts of interest with the contents of the article., (Copyright © 2022 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
19. Angiotensin II receptor type 1 blockade regulates Klotho expression to induce TSC2-deficient cell death.
- Author
-
Shrestha S, Adib E, Imani J, Aguiar DJ, Lamattina AM, Tassew DD, Henske EP, Perrella MA, Priolo C, and El-Chemaly S
- Subjects
- Animals, Humans, Female, Tuberous Sclerosis Complex 2 Protein genetics, Cell Death, Receptors, Angiotensin, Mammals, Lymphangioleiomyomatosis drug therapy, Lymphangioleiomyomatosis genetics, Lymphangioleiomyomatosis metabolism, Tuberous Sclerosis genetics, Tuberous Sclerosis metabolism
- Abstract
Lymphangioleiomyomatosis (LAM) is a multisystem disease occurring in women of child-bearing age manifested by uncontrolled proliferation of smooth muscle-like "LAM" cells in the lungs. LAM cells bear loss-of-function mutations in tuberous sclerosis complex (TSC) genes TSC1 and/or TSC2, causing hyperactivation of the proliferation promoting mammalian/mechanistic target of Rapamycin complex 1 pathway. Additionally, LAM-specific active renin-angiotensin system (RAS) has been identified in LAM nodules, suggesting this system potentially contributes to neoplastic properties of LAM cells; however, the role of this renin-angiotensin signaling is unclear. Here, we report that TSC2-deficient cells are sensitive to the blockade of angiotensin II receptor type 1 (Agtr1). We show that treatment of these cells with the AGTR1 inhibitor losartan or silencing of the Agtr1 gene leads to increased cell death in vitro and attenuates tumor progression in vivo. Notably, we found the effect of Agtr1 blockade is specific to TSC2-deficient cells. Mechanistically, we demonstrate that cell death induced by Agtr1 inhibition is mediated by an increased expression of Klotho. In TSC2-deficient cells, we showed overexpression of Klotho or treatment with recombinant (soluble) Klotho mirrored the cytocidal effect of angiotensin blockade. Furthermore, Klotho treatment decreased the phosphorylation of AKT, potentially leading to this cytocidal effect. Conversely, silencing of Klotho rescued TSC2-deficient cells from cell death induced by Agtr1 inhibition. Therefore, we conclude that Agtr1 and Klotho are important for TSC2-deficient cell survival. These findings further illuminate the role of the RAS in LAM and the potential of targeting Agtr1 inhibition in TSC2-deficient cells., Competing Interests: Conflict of interest The authors declare that they have no conflicts of interest with the contents of this article., (Copyright © 2022 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
20. Correction: High cell surface death receptor expression determines type I versus type II signaling.
- Author
-
Meng XW, Peterson KL, Dai H, Schneider P, Lee SH, Zhang JS, Koenig A, Bronk S, Billadeau DD, Gores GJ, and Kaufmann SH
- Published
- 2022
- Full Text
- View/download PDF
21. Disruption of mitochondrial quality control genes promotes caspase-resistant cell survival following apoptotic stimuli.
- Author
-
Kushnareva Y, Moraes V, Suess J, Peters B, Newmeyer DD, and Kuwana T
- Subjects
- Carcinogenesis genetics, Carcinogenesis metabolism, Humans, Mitochondrial Membranes metabolism, Apoptosis physiology, Caspases metabolism, Cell Survival genetics, Mitochondria genetics, Mitochondria metabolism
- Abstract
In cells undergoing cell-intrinsic apoptosis, mitochondrial outer membrane permeabilization (MOMP) typically marks an irreversible step in the cell death process. However, in some cases, a subpopulation of treated cells can exhibit a sublethal response, termed "minority MOMP." In this phenomenon, the affected cells survive, despite a low level of caspase activation and subsequent limited activation of the endonuclease caspase-activated DNase (DNA fragmentation factor subunit beta). Consequently, these cells can experience DNA damage, increasing the probability of oncogenesis. However, little is known about the minority MOMP response. To discover genes that affect the MOMP response in individual cells, we conducted an imaging-based phenotypic siRNA screen. We identified multiple candidate genes whose downregulation increased the heterogeneity of MOMP within single cells, among which were genes related to mitochondrial dynamics and mitophagy that participate in the mitochondrial quality control (MQC) system. Furthermore, to test the hypothesis that functional MQC is important for reducing the frequency of minority MOMP, we developed an assay to measure the clonogenic survival of caspase-engaged cells. We found that cells deficient in various MQC genes were indeed prone to aberrant post-MOMP survival. Our data highlight the important role of proteins involved in mitochondrial dynamics and mitophagy in preventing apoptotic dysregulation and oncogenesis., Competing Interests: Conflict of interest The authors declare that they have no conflicts of interest with the contents of this article., (Copyright © 2022. Published by Elsevier Inc.)
- Published
- 2022
- Full Text
- View/download PDF
22. Cardiac ryanodine receptor N-terminal region biosensors identify novel inhibitors via FRET-based high-throughput screening.
- Author
-
Zhang J, Singh DP, Ko CY, Nikolaienko R, Wong King Yuen SM, Schwarz JA, Treinen LM, Tung CC, Rožman K, Svensson B, Aldrich CC, Zima AV, Thomas DD, Bers DM, Launikonis BS, Van Petegem F, and Cornea RL
- Subjects
- Animals, Calcium metabolism, Fluorescence Resonance Energy Transfer, High-Throughput Screening Assays, Mice, Muscle, Skeletal chemistry, Muscle, Skeletal metabolism, Biosensing Techniques, Ryanodine Receptor Calcium Release Channel analysis, Ryanodine Receptor Calcium Release Channel metabolism
- Abstract
The N-terminal region (NTR) of ryanodine receptor (RyR) channels is critical for the regulation of Ca
2+ release during excitation-contraction (EC) coupling in muscle. The NTR hosts numerous mutations linked to skeletal (RyR1) and cardiac (RyR2) myopathies, highlighting its potential as a therapeutic target. Here, we constructed two biosensors by labeling the mouse RyR2 NTR at domains A, B, and C with FRET pairs. Using fluorescence lifetime (FLT) detection of intramolecular FRET signal, we developed high-throughput screening (HTS) assays with these biosensors to identify small-molecule RyR modulators. We then screened a small validation library and identified several hits. Hits with saturable FRET dose-response profiles and previously unreported effects on RyR were further tested using [3 H]ryanodine binding to isolated sarcoplasmic reticulum vesicles to determine effects on intact RyR opening in its natural membrane. We identified three novel inhibitors of both RyR1 and RyR2 and two RyR1-selective inhibitors effective at nanomolar Ca2+ . Two of these hits activated RyR1 only at micromolar Ca2+ , highlighting them as potential enhancers of excitation-contraction coupling. To determine whether such hits can inhibit RyR leak in muscle, we further focused on one, an FDA-approved natural antibiotic, fusidic acid (FA). In skinned skeletal myofibers and permeabilized cardiomyocytes, FA inhibited RyR leak with no detrimental effect on skeletal myofiber excitation-contraction coupling. However, in intact cardiomyocytes, FA induced arrhythmogenic Ca2+ transients, a cautionary observation for a compound with an otherwise solid safety record. These results indicate that HTS campaigns using the NTR biosensor can identify compounds with therapeutic potential., Competing Interests: Conflict of interest D. D. T. and R. L. C. hold equity in and serve as executive officers for Photonic Pharma LLC. These relationships have been reviewed and managed by the University of Minnesota. Photonic Pharma had no role in this study., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
23. The copper chaperone CCS facilitates copper binding to MEK1/2 to promote kinase activation.
- Author
-
Grasso M, Bond GJ, Kim YJ, Boyd S, Matson Dzebo M, Valenzuela S, Tsang T, Schibrowsky NA, Alwan KB, Blackburn NJ, Burslem GM, Wittung-Stafshede P, Winkler DD, Marmorstein R, and Brady DC
- Subjects
- Cell Line, Enzyme Activation, Humans, Protein Binding, Copper metabolism, MAP Kinase Kinase 1 metabolism, MAP Kinase Kinase 2 metabolism, Molecular Chaperones metabolism
- Abstract
Normal physiology relies on the precise coordination of intracellular signaling pathways that respond to nutrient availability to balance cell growth and cell death. The canonical mitogen-activated protein kinase pathway consists of the RAF-MEK-ERK signaling cascade and represents one of the most well-defined axes within eukaryotic cells to promote cell proliferation, which underscores its frequent mutational activation in human cancers. Our recent studies illuminated a function for the redox-active micronutrient copper (Cu) as an intracellular mediator of signaling by connecting Cu to the amplitude of mitogen-activated protein kinase signaling via a direct interaction between Cu and the kinases MEK1 and MEK2. Given the large quantities of molecules such as glutathione and metallothionein that limit cellular toxicity from free Cu ions, evolutionarily conserved Cu chaperones facilitate efficient delivery of Cu to cuproenzymes. Thus, a dedicated cellular delivery mechanism of Cu to MEK1/2 likely exists. Using surface plasmon resonance and proximity-dependent biotin ligase studies, we report here that the Cu chaperone for superoxide dismutase (CCS) selectively bound to and facilitated Cu transfer to MEK1. Mutants of CCS that disrupt Cu(I) acquisition and exchange or a CCS small-molecule inhibitor were used and resulted in reduced Cu-stimulated MEK1 kinase activity. Our findings indicate that the Cu chaperone CCS provides fidelity within a complex biological system to achieve appropriate installation of Cu within the MEK1 kinase active site that in turn modulates kinase activity and supports the development of novel MEK1/2 inhibitors that target the Cu structural interface or blunt dedicated Cu delivery mechanisms via CCS., Competing Interests: Conflict of interest D. C. B. holds ownership in Merlon Inc. D. C. B. is an inventor on the patent application 20150017261 entitled “Methods of treating and preventing cancer by disrupting the binding of copper in the MAP kinase pathway”. All other authors declare that they have no conflicts of interest with the contents of this article., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
24. Direct detection of the myosin super-relaxed state and interacting-heads motif in solution.
- Author
-
Chu S, Muretta JM, and Thomas DD
- Subjects
- Amino Acid Motifs, Animals, Benzylamines therapeutic use, Cardiomyopathy, Hypertrophic drug therapy, Cardiomyopathy, Hypertrophic metabolism, Cattle, Kinetics, Myosins metabolism, Uracil chemistry, Uracil therapeutic use, Benzylamines chemistry, Fluorescence Resonance Energy Transfer, Myosins chemistry, Uracil analogs & derivatives
- Abstract
The interacting-heads motif (IHM) is a structure of myosin that has been proposed to modulate cardiac output by occluding myosin molecules from undergoing the force-generating cycle. It is hypothesized to be the structural basis for the super-relaxed state (SRX), a low-ATPase kinetic state thought to be cardioprotective. The goal of the present study was to test this hypothesis by determining directly and quantitatively the fractions of myosin in the IHM and SRX under the same conditions in solution. To detect the structural IHM, we used time-resolved fluorescence resonance energy transfer to quantitate two distinct populations. One population was observed at a center distance of 2.0 nm, whereas the other was not detectable by fluorescence resonance energy transfer, implying a distance greater than 4 nm. We confirmed the IHM assignment to the 2.0-nm population by applying the same cross-linking protocol used previously to image the IHM by electron microscopy. Under the same conditions, we also measured the fraction of myosin in the SRX using stopped-flow kinetics. Our results show that the populations of SRX and IHM myosin were similar, unless treated with mavacamten, a drug that recently completed phase III clinical trials to treat hypertrophic cardiomyopathy and is proposed to act by stabilizing both the SRX and IHM. However, we found that mavacamten had a much greater effect on the SRX (55% increase) than on the IHM (4% increase). We conclude that the IHM structure is sufficient but not necessary to produce the SRX kinetic state., Competing Interests: Conflict of interest D. D. T. holds equity in, and serves as an executive officer for, Photonic Pharma LLC. These relationships have been reviewed and managed by the University of Minnesota. Photonic Pharma had no role in this study. All other authors declare that they have no conflicts of interest related to the contents of this article., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
25. TGF-β/activin signaling promotes CDK7 inhibitor resistance in triple-negative breast cancer cells through upregulation of multidrug transporters.
- Author
-
Webb BM, Bryson BL, Williams-Medina E, Bobbitt JR, Seachrist DD, Anstine LJ, and Keri RA
- Subjects
- ATP Binding Cassette Transporter, Subfamily G, Member 2 genetics, Cell Line, Tumor, Cyclin-Dependent Kinases genetics, Cyclin-Dependent Kinases metabolism, Drug Resistance, Neoplasm genetics, Female, Humans, Inhibin-beta Subunits genetics, Neoplasm Proteins genetics, Signal Transduction genetics, Transforming Growth Factor beta genetics, Triple Negative Breast Neoplasms drug therapy, Triple Negative Breast Neoplasms genetics, Triple Negative Breast Neoplasms pathology, Cyclin-Dependent Kinase-Activating Kinase, ATP Binding Cassette Transporter, Subfamily G, Member 2 biosynthesis, Cyclin-Dependent Kinases antagonists & inhibitors, Drug Resistance, Neoplasm drug effects, Gene Expression Regulation, Neoplastic drug effects, Inhibin-beta Subunits metabolism, Neoplasm Proteins biosynthesis, Protein Kinase Inhibitors pharmacology, Signal Transduction drug effects, Transforming Growth Factor beta metabolism, Triple Negative Breast Neoplasms metabolism, Up-Regulation drug effects
- Abstract
Cyclin-dependent kinase 7 (CDK7) is a master regulatory kinase that drives cell cycle progression and stimulates expression of oncogenes in a myriad of cancers. Inhibitors of CDK7 (CDK7i) are currently in clinical trials; however, as with many cancer therapies, patients will most likely experience recurrent disease due to acquired resistance. Identifying targets underlying CDK7i resistance will facilitate prospective development of new therapies that can circumvent such resistance. Here we utilized triple-negative breast cancer as a model to discern mechanisms of resistance as it has been previously shown to be highly responsive to CDK7 inhibitors. After generating cell lines with acquired resistance, high-throughput RNA sequencing revealed significant upregulation of genes associated with efflux pumps and transforming growth factor-beta (TGF-β) signaling pathways. Genetic silencing or pharmacological inhibition of ABCG2, an efflux pump associated with multidrug resistance, resensitized resistant cells to CDK7i, indicating a reliance on these transporters. Expression of activin A (INHBA), a member of the TGF-β family of ligands, was also induced, whereas its intrinsic inhibitor, follistatin (FST), was repressed. In resistant cells, increased phosphorylation of SMAD3, a downstream mediator, confirmed an increase in activin signaling, and phosphorylated SMAD3 directly bound the ABCG2 promoter regulatory region. Finally, pharmacological inhibition of TGF-β/activin receptors or genetic silencing of SMAD4, a transcriptional partner of SMAD3, reversed the upregulation of ABCG2 in resistant cells and phenocopied ABCG2 inhibition. This study reveals that inhibiting the TGF-β/Activin-ABCG2 pathway is a potential avenue for preventing or overcoming resistance to CDK7 inhibitors., Competing Interests: Conflict of interest The authors declare that they have no conflicts of interest with the contents of this article., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
26. Recombinant SARS-CoV-2 envelope protein traffics to the trans-Golgi network following amphipol-mediated delivery into human cells.
- Author
-
Hutchison JM, Capone R, Luu DD, Shah KH, Hadziselimovic A, Van Horn WD, and Sanders CR
- Subjects
- Cell Membrane metabolism, Coronavirus Envelope Proteins genetics, HeLa Cells, Humans, Lipid Bilayers chemistry, Lipid Bilayers metabolism, Lysosomes metabolism, Polymers chemistry, Propylamines chemistry, Protein Transport, Recombinant Proteins genetics, Recombinant Proteins metabolism, Surface-Active Agents chemistry, Cell Membrane drug effects, Coronavirus Envelope Proteins metabolism, Polymers pharmacology, Propylamines pharmacology, Surface-Active Agents pharmacology, trans-Golgi Network metabolism
- Abstract
The severe acute respiratory syndrome coronavirus 2 envelope protein (S2-E) is a conserved membrane protein that is important for coronavirus (CoV) assembly and budding. Here, we describe the recombinant expression and purification of S2-E in amphipol-class amphipathic polymer solutions, which solubilize and stabilize membrane proteins, but do not disrupt membranes. We found that amphipol delivery of S2-E to preformed planar bilayers results in spontaneous membrane integration and formation of viroporin cation channels. Amphipol delivery of the S2-E protein to human cells results in plasma membrane integration, followed by retrograde trafficking to the trans-Golgi network and accumulation in swollen perinuclear lysosomal-associated membrane protein 1-positive vesicles, likely lysosomes. CoV envelope proteins have previously been proposed to manipulate the luminal pH of the trans-Golgi network, which serves as an accumulation station for progeny CoV particles prior to cellular egress via lysosomes. Delivery of S2-E to cells will enable chemical biological approaches for future studies of severe acute respiratory syndrome coronavirus 2 pathogenesis and possibly even development of "Trojan horse" antiviral therapies. Finally, this work also establishes a paradigm for amphipol-mediated delivery of membrane proteins to cells., Competing Interests: Conflict of interest The authors declare that they have no conflicts of interest with the contents of this article., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
27. Cardiac myosin-binding protein C interaction with actin is inhibited by compounds identified in a high-throughput fluorescence lifetime screen.
- Author
-
Bunch TA, Guhathakurta P, Lepak VC, Thompson AR, Kanassatega RS, Wilson A, Thomas DD, and Colson BA
- Subjects
- Actins chemistry, Animals, Biosensing Techniques, Calorimetry, Fluorescence, Fluorescence Resonance Energy Transfer, Humans, Protein Binding, Rabbits, Sarcomeres metabolism, Time Factors, Actins metabolism, Carrier Proteins metabolism, High-Throughput Screening Assays, Myocardium metabolism
- Abstract
Cardiac myosin-binding protein C (cMyBP-C) interacts with actin and myosin to modulate cardiac muscle contractility. These interactions are disfavored by cMyBP-C phosphorylation. Heart failure patients often display decreased cMyBP-C phosphorylation, and phosphorylation in model systems has been shown to be cardioprotective against heart failure. Therefore, cMyBP-C is a potential target for heart failure drugs that mimic phosphorylation or perturb its interactions with actin/myosin. Here we have used a novel fluorescence lifetime-based assay to identify small-molecule inhibitors of actin-cMyBP-C binding. Actin was labeled with a fluorescent dye (Alexa Fluor 568, AF568) near its cMyBP-C binding sites; when combined with the cMyBP-C N-terminal fragment, C0-C2, the fluorescence lifetime of AF568-actin decreases. Using this reduction in lifetime as a readout of actin binding, a high-throughput screen of a 1280-compound library identified three reproducible hit compounds (suramin, NF023, and aurintricarboxylic acid) that reduced C0-C2 binding to actin in the micromolar range. Binding of phosphorylated C0-C2 was also blocked by these compounds. That they specifically block binding was confirmed by an actin-C0-C2 time-resolved FRET (TR-FRET) binding assay. Isothermal titration calorimetry (ITC) and transient phosphorescence anisotropy (TPA) confirmed that these compounds bind to cMyBP-C, but not to actin. TPA results were also consistent with these compounds inhibiting C0-C2 binding to actin. We conclude that the actin-cMyBP-C fluorescence lifetime assay permits detection of pharmacologically active compounds that affect cMyBP-C-actin binding. We now have, for the first time, a validated high-throughput screen focused on cMyBP-C, a regulator of cardiac muscle contractility and known key factor in heart failure., Competing Interests: Conflict of interest D. D. T. holds equity in and serves as President of Photonic Pharma LLC. This relationship has been reviewed and managed by the University of Minnesota. Photonic Pharma had no role in this study, except to provide some instrumentation, as stated in Experimental procedures. B. A. C. filed a PCT patent application based on this work (patent pending, serial no. PCT/US21/14142). The other authors declare no competing financial interests., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
28. Molecular dynamics approach to identification of new OGG1 cancer-associated somatic variants with impaired activity.
- Author
-
Popov AV, Endutkin AV, Yatsenko DD, Yudkina AV, Barmatov AE, Makasheva KA, Raspopova DY, Diatlova EA, and Zharkov DO
- Subjects
- Amino Acid Substitution, Binding Sites, DNA Damage, DNA Glycosylases genetics, DNA Glycosylases metabolism, DNA, Neoplasm genetics, DNA, Neoplasm metabolism, Gene Expression, Guanine chemistry, Guanine metabolism, Humans, Kinetics, Leukemia enzymology, Leukemia genetics, Leukemia pathology, Lung Neoplasms enzymology, Lung Neoplasms genetics, Lung Neoplasms pathology, Molecular Dynamics Simulation, Neoplasm Proteins genetics, Neoplasm Proteins metabolism, Principal Component Analysis, Protein Binding, Protein Conformation, alpha-Helical, Protein Conformation, beta-Strand, Protein Interaction Domains and Motifs, Protein Structure, Tertiary, Small Cell Lung Carcinoma enzymology, Small Cell Lung Carcinoma genetics, Small Cell Lung Carcinoma pathology, DNA Glycosylases chemistry, DNA Repair, DNA, Neoplasm chemistry, Guanine analogs & derivatives, Mutation, Neoplasm Proteins chemistry
- Abstract
DNA of living cells is always exposed to damaging factors. To counteract the consequences of DNA lesions, cells have evolved several DNA repair systems, among which base excision repair is one of the most important systems. Many currently used antitumor drugs act by damaging DNA, and DNA repair often interferes with chemotherapy and radiotherapy in cancer cells. Tumors are usually extremely genetically heterogeneous, often bearing mutations in DNA repair genes. Thus, knowledge of the functionality of cancer-related variants of proteins involved in DNA damage response and repair is of great interest for personalization of cancer therapy. Although computational methods to predict the variant functionality have attracted much attention, at present, they are mostly based on sequence conservation and make little use of modern capabilities in computational analysis of 3D protein structures. We have used molecular dynamics (MD) to model the structures of 20 clinically observed variants of a DNA repair enzyme, 8-oxoguanine DNA glycosylase. In parallel, we have experimentally characterized the activity, thermostability, and DNA binding in a subset of these mutant proteins. Among the analyzed variants of 8-oxoguanine DNA glycosylase, three (I145M, G202C, and V267M) were significantly functionally impaired and were successfully predicted by MD. Alone or in combination with sequence-based methods, MD may be an important functional prediction tool for cancer-related protein variants of unknown significance., Competing Interests: Conflict of interest The authors declare that they have no conflicts of interest with the contents of this article., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
29. Novel drug discovery platform for spinocerebellar ataxia, using fluorescence technology targeting β-III-spectrin.
- Author
-
Rebbeck RT, Andrick AK, Denha SA, Svensson B, Guhathakurta P, Thomas DD, Hays TS, and Avery AW
- Subjects
- Actins chemistry, Actins genetics, Fluorescence Resonance Energy Transfer, Gene Expression, Green Fluorescent Proteins genetics, Green Fluorescent Proteins metabolism, HEK293 Cells, Humans, Kinetics, Luminescent Proteins genetics, Luminescent Proteins metabolism, Marine Toxins pharmacology, Models, Biological, Models, Molecular, Mutation, Neuroprotective Agents pharmacology, Protein Binding drug effects, Protein Conformation, alpha-Helical, Protein Conformation, beta-Strand, Protein Interaction Domains and Motifs, Recombinant Fusion Proteins chemistry, Recombinant Fusion Proteins genetics, Reproducibility of Results, Spectrin chemistry, Spectrin genetics, Spinocerebellar Ataxias genetics, Spinocerebellar Ataxias metabolism, Spinocerebellar Ataxias pathology, Red Fluorescent Protein, Actins metabolism, Binding Sites drug effects, High-Throughput Screening Assays, Recombinant Fusion Proteins metabolism, Spectrin metabolism
- Abstract
Numerous diseases are linked to mutations in the actin-binding domains (ABDs) of conserved cytoskeletal proteins, including β-III-spectrin, α-actinin, filamin, and dystrophin. A β-III-spectrin ABD mutation (L253P) linked to spinocerebellar ataxia type 5 (SCA5) causes a dramatic increase in actin binding. Reducing actin binding of L253P is thus a potential therapeutic approach for SCA5 pathogenesis. Here, we validate a high-throughput screening (HTS) assay to discover potential disrupters of the interaction between the mutant β-III-spectrin ABD and actin in live cells. This assay monitors FRET between fluorescent proteins fused to the mutant ABD and the actin-binding peptide Lifeact, in HEK293-6E cells. Using a specific and high-affinity actin-binding tool compound, swinholide A, we demonstrate HTS compatibility with an excellent Z'-factor of 0.67 ± 0.03. Screening a library of 1280 pharmacologically active compounds in 1536-well plates to determine assay robustness, we demonstrate high reproducibility across plates and across days. We identified nine Hits that reduced FRET between Lifeact and ABD. Four of those Hits were found to reduce Lifeact cosedimentation with actin, thus establishing the potential of our assay for detection of actin-binding modulators. Concurrent to our primary FRET assay, we also developed a high-throughput compatible counter screen to remove undesirable FRET Hits. Using the FRET Hits, we show that our counter screen is sensitive to undesirable compounds that cause cell toxicity or ABD aggregation. Overall, our FRET-based HTS platform sets the stage to screen large compound libraries for modulators of β-III-spectrin, or disease-linked spectrin-related proteins, for therapeutic development., Competing Interests: Conflict of interest D. D. T. holds equity in and serves as an executive officer for Photonic Pharma LLC. These relationships have been reviewed and managed by the University of Minnesota. Photonic Pharma had no role in this study, except to provide some instrumentation, as stated in Experimental Procedures. R. T. R., A. K. A., S. A. D., B. S., P. G., T. S. H., and A. W. A. have no conflict of interest to disclose., (Copyright © 2020 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
30. Mechanistic analysis of actin-binding compounds that affect the kinetics of cardiac myosin-actin interaction.
- Author
-
Roopnarine O and Thomas DD
- Subjects
- Actins drug effects, Adenosine Triphosphatases drug effects, Adenosine Triphosphatases metabolism, Animals, Cardiac Myosins drug effects, Cardiac Myosins physiology, Cattle, Fluorescence, High-Throughput Screening Assays methods, Kinetics, Muscle Contraction physiology, Myosin Subfragments drug effects, Myosin Subfragments metabolism, Myosins drug effects, Myosins metabolism, Physics, Protein Binding, Pyrenes chemistry, Rabbits, Small Molecule Libraries pharmacology, Actins chemistry, Actins metabolism, Cardiac Myosins metabolism
- Abstract
Actin-myosin mediated contractile forces are crucial for many cellular functions, including cell motility, cytokinesis, and muscle contraction. We determined the effects of ten actin-binding compounds on the interaction of cardiac myosin subfragment 1 (S1) with pyrene-labeled F-actin (PFA). These compounds, previously identified from a small-molecule high-throughput screen (HTS), perturb the structural dynamics of actin and the steady-state actin-activated myosin ATPase activity. However, the mechanisms underpinning these perturbations remain unclear. Here we further characterize them by measuring their effects on PFA fluorescence, which is decreased specifically by the strong binding of myosin to actin. We measured these effects under equilibrium and steady-state conditions, and under transient conditions, in stopped-flow experiments following addition of ATP to S1-bound PFA. We observed that these compounds affect early steps of the myosin ATPase cycle to different extents. They increased the association equilibrium constant K
1 for the formation of the strongly bound collision complex, indicating increased ATP affinity for actin-bound myosin, and decreased the rate constant k+2 for subsequent isomerization to the weakly bound ternary complex, thus slowing the strong-to-weak transition that actin-myosin interaction undergoes early in the ATPase cycle. The compounds' effects on actin structure allosterically inhibit the kinetics of the actin-myosin interaction in ways that may be desirable for treatment of hypercontractile forms of cardiomyopathy. This work helps to elucidate the mechanisms of action for these compounds, several of which are currently used therapeutically, and sets the stage for future HTS campaigns that aim to discover new drugs for treatment of heart failure., Competing Interests: Conflict of interest D. D. T. holds equity in, and serves as President of, Photonic Pharma LLC. This relationship has been reviewed and managed by the University of Minnesota. Photonic Pharma had no role in this study. The authors declare no conflicts of interest in regard to this article., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
31. The transmembrane peptide DWORF activates SERCA2a via dual mechanisms.
- Author
-
Li A, Yuen SL, Stroik DR, Kleinboehl E, Cornea RL, and Thomas DD
- Subjects
- Calcium metabolism, Calcium-Binding Proteins metabolism, Cell Line, HEK293 Cells, Heart Failure metabolism, Humans, Myocardium metabolism, Myocytes, Cardiac metabolism, Peptides physiology, Sarcoplasmic Reticulum metabolism, Sarcoplasmic Reticulum Calcium-Transporting ATPases physiology, Peptides metabolism, Sarcoplasmic Reticulum Calcium-Transporting ATPases metabolism
- Abstract
The Ca-ATPase isoform 2a (SERCA2a) pumps cytosolic Ca
2+ into the sarcoplasmic reticulum (SR) of cardiac myocytes, enabling muscle relaxation during diastole. Abnormally high cytosolic [Ca2+ ] is a central factor in heart failure, suggesting that augmentation of SERCA2a Ca2+ transport activity could be a promising therapeutic approach. SERCA2a is inhibited by the protein phospholamban (PLB), and a novel transmembrane peptide, dwarf open reading frame (DWORF), is proposed to enhance SR Ca2+ uptake and myocyte contractility by displacing PLB from binding to SERCA2a. However, establishing DWORF's precise physiological role requires further investigation. In the present study, we developed cell-based FRET biosensor systems that can report on protein-protein interactions and structural changes in SERCA2a complexes with PLB and/or DWORF. To test the hypothesis that DWORF competes with PLB to occupy the SERCA2a-binding site, we transiently transfected DWORF into a stable HEK cell line expressing SERCA2a labeled with a FRET donor and PLB labeled with a FRET acceptor. We observed a significant decrease in FRET efficiency, consistent with a decrease in the fraction of SERCA2a bound to PLB. Surprisingly, we also found that DWORF also activates SERCA's enzymatic activity directly in the absence of PLB at subsaturating calcium levels. Using site-directed mutagenesis, we generated DWORF variants that do not activate SERCA, thus identifying residues P15 and W22 as necessary for functional SERCA2a-DWORF interactions. This work advances our mechanistic understanding of the regulation of SERCA2a by small transmembrane proteins and sets the stage for future therapeutic development in heart failure research., Competing Interests: Conflict of interest D. D. T. and R. L. C. hold equity in, and serve as executive officers of, Photonic Pharma LLC. These relationships have been reviewed and managed by the University of Minnesota in accordance with its conflict-of-interest policies. Photonic Pharma had no role in this study. The authors declare no conflicts of interest with regard to this article., (Published by Elsevier Inc.)- Published
- 2021
- Full Text
- View/download PDF
32. FRET and optical trapping reveal mechanisms of actin activation of the power stroke and phosphate release in myosin V.
- Author
-
Gunther LK, Rohde JA, Tang W, Cirilo JA Jr, Marang CP, Scott BD, Thomas DD, Debold EP, and Yengo CM
- Subjects
- Adenosine Triphosphatases metabolism, Adenosine Triphosphate metabolism, Animals, Chickens, Kinetics, Models, Molecular, Mutation, Myosin Type V genetics, Actins metabolism, Fluorescence Resonance Energy Transfer methods, Myosin Type V metabolism, Optical Imaging methods, Phosphates metabolism
- Abstract
Myosins generate force and motion by precisely coordinating their mechanical and chemical cycles, but the nature and timing of this coordination remains controversial. We utilized a FRET approach to examine the kinetics of structural changes in the force-generating lever arm in myosin V. We directly compared the FRET results with single-molecule mechanical events examined by optical trapping. We introduced a mutation (S217A) in the conserved switch I region of the active site to examine how myosin couples structural changes in the actin- and nucleotide-binding regions with force generation. Specifically, S217A enhanced the maximum rate of lever arm priming (recovery stroke) while slowing ATP hydrolysis, demonstrating that it uncouples these two steps. We determined that the mutation dramatically slows both actin-induced rotation of the lever arm (power stroke) and phosphate release (≥10-fold), whereas our simulations suggest that the maximum rate of both steps is unchanged by the mutation. Time-resolved FRET revealed that the structure of the pre- and post-power stroke conformations and mole fractions of these conformations were not altered by the mutation. Optical trapping results demonstrated that S217A does not dramatically alter unitary displacements or slow the working stroke rate constant, consistent with the mutation disrupting an actin-induced conformational change prior to the power stroke. We propose that communication between the actin- and nucleotide-binding regions of myosin assures a proper actin-binding interface and active site have formed before producing a power stroke. Variability in this coupling is likely crucial for mediating motor-based functions such as muscle contraction and intracellular transport., (Copyright © 2020 © 2020 Gunther et al. Published by Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
33. Actin-binding compounds, previously discovered by FRET-based high-throughput screening, differentially affect skeletal and cardiac muscle.
- Author
-
Guhathakurta P, Phung LA, Prochniewicz E, Lichtenberger S, Wilson A, and Thomas DD
- Subjects
- Animals, Cattle, Drug Evaluation, Preclinical, Fluorescence Resonance Energy Transfer, Rabbits, Actins antagonists & inhibitors, Actins chemistry, Actins metabolism, Muscle, Skeletal metabolism, Myocardium metabolism, Myofibrils metabolism, Myosins chemistry, Myosins metabolism, Small Molecule Libraries
- Abstract
Actin's interactions with myosin and other actin-binding proteins are essential for cellular viability in numerous cell types, including muscle. In a previous high-throughput time-resolved FRET (TR-FRET) screen, we identified a class of compounds that bind to actin and affect actomyosin structure and function. For clinical utility, it is highly desirable to identify compounds that affect skeletal and cardiac muscle differently. Because actin is more highly conserved than myosin and most other muscle proteins, most such efforts have not targeted actin. Nevertheless, in the current study, we tested the specificity of the previously discovered actin-binding compounds for effects on skeletal and cardiac α-actins as well as on skeletal and cardiac myofibrils. We found that a majority of these compounds affected the transition of monomeric G-actin to filamentous F-actin, and that several of these effects were different for skeletal and cardiac actin isoforms. We also found that several of these compounds affected ATPase activity differently in skeletal and cardiac myofibrils. We conclude that these structural and biochemical assays can be used to identify actin-binding compounds that differentially affect skeletal and cardiac muscles. The results of this study set the stage for screening of large chemical libraries for discovery of novel compounds that act therapeutically and specifically on cardiac or skeletal muscle., Competing Interests: Conflict of interest—D. D. T. holds equity in, and serves as President of, Photonic Pharma LLC. This relationship has been reviewed and managed by the University of Minnesota. Photonic Pharma had no role in this study. The authors declare no conflicts of interest in regard to this manuscript., (© 2020 Guhathakurta et al.)
- Published
- 2020
- Full Text
- View/download PDF
34. The transcriptional repressor BCL11A promotes breast cancer metastasis.
- Author
-
Seachrist DD, Hannigan MM, Ingles NN, Webb BM, Weber-Bonk KL, Yu P, Bebek G, Singh S, Sizemore ST, Varadan V, Licatalosi DD, and Keri RA
- Subjects
- Cell Line, Tumor, Disease Progression, Female, Humans, Neoplasm Invasiveness pathology, Neoplasm Metastasis genetics, Neoplasm Metastasis pathology, Triple Negative Breast Neoplasms pathology, Gene Expression Regulation, Neoplastic, Neoplasm Invasiveness genetics, Repressor Proteins genetics, Triple Negative Breast Neoplasms genetics, Up-Regulation
- Abstract
The phenotypes of each breast cancer subtype are defined by their transcriptomes. However, the transcription factors that regulate differential patterns of gene expression that contribute to specific disease outcomes are not well understood. Here, using gene silencing and overexpression approaches, RNA-Seq, and splicing analysis, we report that the transcription factor B-cell leukemia/lymphoma 11A (BCL11A) is highly expressed in triple-negative breast cancer (TNBC) and drives metastatic disease. Moreover, BCL11A promotes cancer cell invasion by suppressing the expression of muscleblind-like splicing regulator 1 ( MBNL1 ), a splicing regulator that suppresses metastasis. This ultimately increases the levels of an alternatively spliced isoform of integrin-α6 ( ITGA6 ), which is associated with worse patient outcomes. These results suggest that BCL11A sustains TNBC cell invasion and metastatic growth by repressing MBNL1-directed splicing of ITGA6 Our findings also indicate that BCL11A lies at the interface of transcription and splicing and promotes aggressive TNBC phenotypes., Competing Interests: Conflict of interest—The authors declare that they have no conflicts of interest with the contents of this article., (© 2020 Seachrist et al.)
- Published
- 2020
- Full Text
- View/download PDF
35. Correction: Bromodomain and extraterminal protein inhibition blocks growth of triple-negative breast cancers through the suppression of aurora kinases.
- Author
-
Sahni JM, Gayle SS, Bonk KLW, Vite LC, Yori JL, Webb B, Ramos EK, Seachrist DD, Landis MD, Chang JC, Bradner JE, and Keri RA
- Published
- 2020
- Full Text
- View/download PDF
36. Leukocyte-associated immunoglobulin-like receptor 1 inhibits T-cell signaling by decreasing protein phosphorylation in the T-cell signaling pathway.
- Author
-
Park JE, Brand DD, Rosloniec EF, Yi AK, Stuart JM, Kang AH, and Myers LK
- Subjects
- Animals, CSK Tyrosine-Protein Kinase metabolism, Cattle, Collagen Type I metabolism, Humans, Jurkat Cells, Mice, Inbred C57BL, Mice, Knockout, Phosphorylation, Phosphotyrosine metabolism, Proto-Oncogene Mas, ZAP-70 Protein-Tyrosine Kinase metabolism, Receptors, Immunologic metabolism, Signal Transduction, T-Lymphocytes metabolism
- Abstract
Multiple observations implicate T-cell dysregulation as a central event in the pathogenesis of rheumatoid arthritis. Here, we investigated mechanisms for suppressing T-cell activation via the inhibitory receptor leukocyte-associated immunoglobulin-like receptor 1 (LAIR-1). To determine how LAIR-1 affects T-cell receptor (TCR) signaling, we compared 1) T cells from LAIR-1-sufficient and -deficient mice, 2) Jurkat cells expressing either LAIR-1 mutants or C-terminal Src kinase (CSK) mutants, and 3) T cells from mice that contain a CSK transgene susceptible to chemical inhibition. Our results indicated that LAIR-1 engagement by collagen or by complement C1q (C1Q, which contains a collagen-like domain) inhibits TCR signaling by decreasing the phosphorylation of key components in the canonical T-cell signaling pathway, including LCK proto-oncogene SRC family tyrosine kinase (LCK), LYN proto-oncogene SRC family tyrosine kinase (LYN), ζ chain of T-cell receptor-associated protein kinase 70 (ZAP-70), and three mitogen-activated protein kinases (extracellular signal-regulated kinase, c-Jun N-terminal kinase 1/2, and p38). The intracellular region of LAIR-1 contains two immunoreceptor tyrosine-based inhibition motifs that are both phosphorylated by LAIR-1 activation, and immunoprecipitation experiments revealed that Tyr-251 in LAIR-1 binds CSK. Using CRISPR/Cas9-mediated genome editing, we demonstrate that CSK is essential for the LAIR-1-induced inhibition of the human TCR signal transduction. T cells from mice that expressed a PP1 analog-sensitive form of CSK (CskAS) corroborated these findings, and we also found that Tyr-251 is critical for LAIR-1's inhibitory function. We propose that LAIR-1 activation may be a strategy for controlling inflammation and may offer a potential therapeutic approach for managing autoimmune diseases.
- Published
- 2020
- Full Text
- View/download PDF
37. Mitochondrial residence of the apoptosis inducer BAX is more important than BAX oligomerization in promoting membrane permeabilization.
- Author
-
Kuwana T, King LE, Cosentino K, Suess J, Garcia-Saez AJ, Gilmore AP, and Newmeyer DD
- Subjects
- Animals, Cells, Cultured, Gene Knockout Techniques, Humans, Mice, Mitochondria genetics, Permeability, Point Mutation, Protein Multimerization, bcl-2-Associated X Protein analysis, bcl-2-Associated X Protein genetics, Apoptosis, Lipid Bilayers metabolism, Mitochondria metabolism, bcl-2-Associated X Protein metabolism
- Abstract
Permeabilization of the mitochondrial outer membrane is a key step in the intrinsic apoptosis pathway, triggered by the release of mitochondrial intermembrane space proteins into the cytoplasm. The BCL-2-associated X apoptosis regulator (BAX) protein critically contributes to this process by forming pores in the mitochondrial outer membrane. However, the relative roles of the mitochondrial residence of BAX and its oligomerization in promoting membrane permeabilization are unclear. To this end, using both cell-free and cellular experimental systems, including membrane permeabilization, size-exclusion chromatography-based oligomer, and retrotranslocation assays, along with confocal microscopy analysis, here we studied two BAX C-terminal variants, T182I and G179P. Neither variant formed large oligomers when activated in liposomes. Nevertheless, the G179P variant could permeabilize liposome membranes, suggesting that large BAX oligomers are not essential for the permeabilization. However, when G179P was transduced into BAX/BCL2 agonist killer (BAK) double-knockout mouse embryonic fibroblasts, its location was solely cytoplasmic, and it then failed to mediate cell death. In contrast, T182I was inefficient in both liposome insertion and permeabilization. Yet, when transduced into cells, BAXT182I resided predominantly on mitochondria, because of its slow retrotranslocation and mediated apoptosis as efficiently as WT BAX. We conclude that BAX's mitochondrial residence in vivo , regulated by both targeting and retrotranslocation, is more significant for its pro-apoptotic activity than its ability to insert and to form higher-order oligomers in model membranes. We propose that this finding should be taken into account when developing drugs that modulate BAX activity., (© 2020 Kuwana et al.)
- Published
- 2020
- Full Text
- View/download PDF
38. The zebrafish NLRP3 inflammasome has functional roles in ASC-dependent interleukin-1β maturation and gasdermin E-mediated pyroptosis.
- Author
-
Li JY, Wang YY, Shao T, Fan DD, Lin AF, Xiang LX, and Shao JZ
- Subjects
- Animals, Caspases metabolism, HEK293 Cells, Humans, Mice, Protein Aggregates, Receptors, Estrogen chemistry, Zebrafish Proteins chemistry, Cytoskeletal Proteins metabolism, Inflammasomes metabolism, Interleukin-1beta metabolism, NLR Family, Pyrin Domain-Containing 3 Protein metabolism, Pyroptosis, Receptors, Estrogen metabolism, Zebrafish metabolism, Zebrafish Proteins metabolism
- Abstract
The NLR family pyrin domain containing 3 (NLRP3) inflammasome is one of the best-characterized inflammasomes in humans and other mammals. However, knowledge about the NLRP3 inflammasome in nonmammalian species remains limited. Here, we report the molecular and functional identification of an NLRP3 homolog ( Dr NLRP3) in a zebrafish ( Danio rerio ) model. We found that Dr NLRP3's overall structural architecture was shared with mammalian NLRP3s. It initiates a classical inflammasome assembly for zebrafish inflammatory caspase ( Dr Caspase-A/-B) activation and interleukin 1β ( Dr IL-1β) maturation in an apoptosis-associated speck-like protein containing a caspase-recruitment domain (ASC)-dependent manner, in which Dr NLRP3 organizes Dr ASC into a filament that recruits Dr Caspase-A/-B by homotypic pyrin domain (PYD)-PYD interactions. Dr Caspase-A/-B activation in the Dr NLRP3 inflammasome occurred in two steps, with Dr Caspase-A being activated first and Dr Caspase-B second. Dr NLRP3 also directly activated full-length Dr Caspase-B and elicited cell pyroptosis in a gasdermin E (GSDME)-dependent but ASC-independent manner. These two events were tightly coordinated by Dr NLRP3 to ensure efficient IL-1β secretion for the initiation of host innate immunity. By knocking down Dr NLRP3 in zebrafish embryos and generating a Dr ASC-knockout ( Dr ASC
-/- ) fish clone, we characterized the function of the Dr NLRP3 inflammasome in anti-bacterial immunity in vivo The results of our study disclosed the origin of the NLRP3 inflammasome in teleost fish, providing a cross-species understanding of the evolutionary history of inflammasomes. Our findings also indicate that the NLRP3 inflammasome may coordinate inflammatory cytokine processing and secretion through a GSDME-mediated pyroptotic pathway, uncovering a previously unrecognized regulatory function of NLRP3 in both inflammation and cell pyroptosis., (© 2020 Li et al.)- Published
- 2020
- Full Text
- View/download PDF
39. DNA polymerase β nucleotide-stabilized template misalignment fidelity depends on local sequence context.
- Author
-
Howard MJ, Cavanaugh NA, Batra VK, Shock DD, Beard WA, and Wilson SH
- Subjects
- Crystallography, X-Ray, DNA chemistry, DNA metabolism, DNA Polymerase beta metabolism, DNA Repair, DNA Replication, Enzyme Activation, Enzyme Stability, Humans, Models, Molecular, Protein Conformation, Protein Domains, DNA Polymerase beta chemistry
- Abstract
DNA polymerase β has two DNA-binding domains that interact with the opposite sides of short DNA gaps. These domains contribute two activities that modify the 5' and 3' margins of gapped DNA during base excision repair. DNA gaps greater than 1 nucleotide (nt) pose an architectural and logistical problem for the two domains to interact with their respective DNA termini. Here, crystallographic and kinetic analyses of 2-nt gap-filling DNA synthesis revealed that the fidelity of DNA synthesis depends on local sequence context. This was due to template dynamics that altered which of the two template nucleotides in the gap served as the coding nucleotide. We observed that, when a purine nucleotide was in the first coding position, DNA synthesis fidelity was similar to that observed with a 1-nt gap. However, when the initial templating nucleotide was a pyrimidine, fidelity was decreased. If the first templating nucleotide was a cytidine, there was a significantly higher probability that the downstream template nucleotide coded for the incoming nucleotide. This dNTP-stabilized misalignment reduced base substitution and frameshift deletion fidelities. A crystal structure of a binary DNA product complex revealed that the cytidine in the first templating site was in an extrahelical position, permitting the downstream template nucleotide to occupy the coding position. These results indicate that DNA polymerase β can induce a strain in the DNA that modulates the position of the coding nucleotide and thereby impacts the identity of the incoming nucleotide. Our findings demonstrate that "correct" DNA synthesis can result in errors when template dynamics induce coding ambiguity.
- Published
- 2020
- Full Text
- View/download PDF
40. Differential and overlapping targets of the transcriptional regulators NRF1, NRF2, and NRF3 in human cells.
- Author
-
Liu P, Kerins MJ, Tian W, Neupane D, Zhang DD, and Ooi A
- Subjects
- Basic-Leucine Zipper Transcription Factors genetics, Cell Line, Humans, NF-E2-Related Factor 1 genetics, NF-E2-Related Factor 2 genetics, Nuclear Respiratory Factor 1, Basic-Leucine Zipper Transcription Factors metabolism, Gene Expression Regulation, NF-E2-Related Factor 1 metabolism, NF-E2-Related Factor 2 metabolism
- Abstract
The nuclear factor (erythroid 2)-like (NRF) transcription factors are a subset of cap'n'collar transcriptional regulators. They consist of three members, NRF1, NRF2, and NRF3, that regulate the expression of genes containing antioxidant-response elements (AREs) in their promoter regions. Although all NRF members regulate ARE-containing genes, each is associated with distinct roles. A comprehensive study of differential and overlapping DNA-binding and transcriptional activities of the NRFs has not yet been conducted. Here, we performed chromatin immunoprecipitation (ChIP)-exo sequencing, an approach that combines ChIP with exonuclease treatment to pinpoint regulatory elements in DNA with high precision, in conjunction with RNA-sequencing to define the transcriptional targets of each NRF member. Our approach, done in three U2OS cell lines, identified 31 genes that were regulated by all three NRF members, 27 that were regulated similarly by all three, and four genes that were differentially regulated by at least one NRF member. We also found genes that were up- or down-regulated by only one NRF member, with 84, 84, and 22 genes that were regulated by NRF1, NRF2, and NRF3, respectively. Analysis of the ARE motifs identified in ChIP peaks revealed that NRF2 prefers binding to AREs flanked by GC-rich regions and that NRF1 prefers AT-rich flanking regions. Thus, sequence preference, likely in combination with upstream signaling events, determines NRF member activation under specific cellular contexts. Our analysis provides a comprehensive description of differential and overlapping gene regulation by the transcriptional regulators NRF1, NRF2, and NRF3., (© 2019 Liu et al.)
- Published
- 2019
- Full Text
- View/download PDF
41. 2'-C-methylated nucleotides terminate virus RNA synthesis by preventing active site closure of the viral RNA-dependent RNA polymerase.
- Author
-
Boehr AK, Arnold JJ, Oh HS, Cameron CE, and Boehr DD
- Subjects
- Base Sequence, Ligands, Methylation, Models, Molecular, RNA, Viral chemistry, RNA, Viral metabolism, Virus Replication genetics, Catalytic Domain, Nucleotides metabolism, RNA, Viral biosynthesis, RNA-Dependent RNA Polymerase chemistry, RNA-Dependent RNA Polymerase metabolism
- Abstract
The 2'-C-methyl ribonucleosides are nucleoside analogs representing an important class of antiviral agents, especially against positive-strand RNA viruses. Their value is highlighted by the highly successful anti-hepatitis C drug sofosbuvir. When appropriately phosphorylated, these nucleotides are successfully incorporated into RNA by the virally encoded RNA-dependent RNA polymerase (RdRp). This activity prevents further RNA extension, but the mechanism is poorly characterized. Previously, we had identified NMR signatures characteristic of formation of RdRp-RNA binary and RdRp-RNA-NTP ternary complexes for the poliovirus RdRp, including an open-to-closed conformational change necessary to prepare the active site for catalysis of phosphoryl transfer. Here we used these observations as a framework for interpreting the effects of 2'-C-methyl adenosine analogs on RNA chain extension in solution-state NMR spectroscopy experiments, enabling us to gain additional mechanistic insights into 2'-C-methyl ribonucleoside-mediated RNA chain termination. Contrary to what has been proposed previously, poliovirus RdRp that was bound to RNA with an incorporated 2'-C-methyl nucleotide could still bind to the next incoming NTP. Our results also indicated that incorporation of the 2'-C-methyl nucleotide does not disrupt RdRp-RNA interactions and does not prevent translocation. Instead, incorporation of the 2'-C-methyl nucleotide blocked closure of the RdRp active site upon binding of the next correct incoming NTP, which prevented further nucleotide addition. We propose that other nucleotide analogs that act as nonobligate chain terminators may operate through a similar mechanism., (© 2019 Boehr et al.)
- Published
- 2019
- Full Text
- View/download PDF
42. Formation of mammalian preribosomes proceeds from intermediate to composed state during ribosome maturation.
- Author
-
Abetov DA, Kiyan VS, Zhylkibayev AA, Sarbassova DA, Alybayev SD, Spooner E, Song MS, Bersimbaev RI, and Sarbassov DD
- Subjects
- Animals, Cell Line, DNA-Binding Proteins metabolism, Humans, Mice, N-Terminal Acetyltransferases metabolism, RNA Processing, Post-Transcriptional, RNA, Ribosomal metabolism, Ribonucleoproteins chemistry, Ribonucleoproteins metabolism, Ribosomal Proteins metabolism, TOR Serine-Threonine Kinases antagonists & inhibitors, TOR Serine-Threonine Kinases metabolism, RNA Precursors metabolism, Ribosomes metabolism
- Abstract
In eukaryotes, ribosome assembly is a rate-limiting step in ribosomal biogenesis that takes place in a distinctive subnuclear organelle, the nucleolus. How ribosomes get assembled at the nucleolar site by forming initial preribosomal complexes remains poorly characterized. In this study, using several human and murine cell lines, we developed a method for isolation of native mammalian preribosomal complexes by lysing cell nuclei through mild sonication. A sucrose gradient fractionation of the nuclear lysate resolved several ribonucleoprotein (RNP) complexes containing rRNAs and ribosomal proteins. Characterization of the RNP complexes with MS-based protein identification and Northern blotting-based rRNA detection approaches identified two types of preribosomes we named here as intermediate preribosomes (IPRibs) and composed preribosome (CPRib). IPRib complexes comprised large preribosomes (105S to 125S in size) containing the rRNA modification factors and premature rRNAs. We further observed that a distinctive CPRib complex consists of an 85S preribosome assembled with mature rRNAs and a ribosomal biogenesis factor, Ly1 antibody-reactive (LYAR), that does not associate with premature rRNAs and rRNA modification factors. rRNA-labeling experiments uncovered that IPRib assembly precedes CPRib complex formation. We also found that formation of the preribosomal complexes is nutrient-dependent because the abundances of IPRib and CPRib decreased substantially when cells were either deprived of amino acids or exposed to an mTOR kinase inhibitor. These findings indicate that preribosomes form via dynamic and nutrient-dependent processing events and progress from an intermediate to a composed state during ribosome maturation., (© 2019 Abetov et al.)
- Published
- 2019
- Full Text
- View/download PDF
43. Protein kinase D and Gβγ mediate sustained nociceptive signaling by biased agonists of protease-activated receptor-2.
- Author
-
Zhao P, Pattison LA, Jensen DD, Jimenez-Vargas NN, Latorre R, Lieu T, Jaramillo JO, Lopez-Lopez C, Poole DP, Vanner SJ, Schmidt BL, and Bunnett NW
- Subjects
- Animals, Cathepsins metabolism, Cell Membrane metabolism, GTP-Binding Protein beta Subunits antagonists & inhibitors, GTP-Binding Protein gamma Subunits antagonists & inhibitors, Ganglia, Spinal cytology, Ganglia, Spinal metabolism, Golgi Apparatus metabolism, HEK293 Cells, Humans, Hyperalgesia metabolism, Hyperalgesia pathology, Hyperalgesia prevention & control, Leukocyte Elastase metabolism, Mice, Mice, Inbred C57BL, Protein Kinase C antagonists & inhibitors, Pyrimidines administration & dosage, Pyrimidines pharmacology, Receptor, PAR-2 agonists, Signal Transduction drug effects, Xanthenes administration & dosage, Xanthenes pharmacology, GTP-Binding Protein beta Subunits metabolism, GTP-Binding Protein gamma Subunits metabolism, Protein Kinase C metabolism, Receptor, PAR-2 metabolism
- Abstract
Proteases sustain hyperexcitability and pain by cleaving protease-activated receptor-2 (PAR
2 ) on nociceptors through distinct mechanisms. Whereas trypsin induces PAR2 coupling to Gαq , Gαs , and β-arrestins, cathepsin-S (CS) and neutrophil elastase (NE) cleave PAR2 at distinct sites and activate it by biased mechanisms that induce coupling to Gαs , but not to Gαq or β-arrestins. Because proteases activate PAR2 by irreversible cleavage, and activated PAR2 is degraded in lysosomes, sustained extracellular protease-mediated signaling requires mobilization of intact PAR2 from the Golgi apparatus or de novo synthesis of new receptors by incompletely understood mechanisms. We found here that trypsin, CS, and NE stimulate PAR2 -dependent activation of protein kinase D (PKD) in the Golgi of HEK293 cells, in which PKD regulates protein trafficking. The proteases stimulated translocation of the PKD activator Gβγ to the Golgi, coinciding with PAR2 mobilization from the Golgi. Proteases also induced translocation of a photoconverted PAR2 -Kaede fusion protein from the Golgi to the plasma membrane of KNRK cells. After incubation of HEK293 cells and dorsal root ganglia neurons with CS, NE, or trypsin, PAR2 responsiveness initially declined, consistent with PAR2 cleavage and desensitization, and then gradually recovered. Inhibitors of PKD, Gβγ, and protein translation inhibited recovery of PAR2 responsiveness. PKD and Gβγ inhibitors also attenuated protease-evoked mechanical allodynia in mice. We conclude that proteases that activate PAR2 by canonical and biased mechanisms stimulate PKD in the Golgi; PAR2 mobilization and de novo synthesis repopulate the cell surface with intact receptors and sustain nociceptive signaling by extracellular proteases., (© 2019 Zhao et al.)- Published
- 2019
- Full Text
- View/download PDF
44. Molecular basis for the faithful replication of 5-methylcytosine and its oxidized forms by DNA polymerase β.
- Author
-
Howard MJ, Foley KG, Shock DD, Batra VK, and Wilson SH
- Subjects
- 5-Methylcytosine chemistry, Catalysis, DNA metabolism, Humans, Kinetics, Oxidation-Reduction, 5-Methylcytosine biosynthesis, DNA Polymerase beta metabolism, DNA Replication
- Abstract
DNA methylation is an epigenetic mark that regulates gene expression in mammals. One method of methylation removal is through ten-eleven translocation-catalyzed oxidation and the base excision repair pathway. The iterative oxidation of 5-methylcytosine catalyzed by ten-eleven translocation enzymes produces three oxidized forms of cytosine: 5-hydroxmethylcytosine, 5-formylcytosine, and 5-carboxycytosine. The effect these modifications have on the efficiency and fidelity of the base excision repair pathway during the repair of opposing base damage, and in particular DNA polymerization, remains to be elucidated. Using kinetic assays, we show that the catalytic efficiency for the incorporation of dGTP catalyzed by human DNA polymerase β is not affected when 5-methylcytosine, 5-hydroxmethylcytosine, and 5-formylcytosine are in the DNA template. In contrast, the catalytic efficiency of dGTP insertion decreases ∼20-fold when 5-carboxycytosine is in the templating position, as compared with unmodified cytosine. However, DNA polymerase fidelity is unaltered when these modifications are in the templating position. Structural analysis reveals that the methyl, hydroxymethyl, and formyl modifications are easily accommodated within the polymerase active site. However, to accommodate the carboxyl modification, the phosphate backbone on the templating nucleotide shifts ∼2.5 Å to avoid a potential steric/repulsive clash. This altered conformation is stabilized by lysine 280, which makes a direct interaction with the carboxyl modification and the phosphate backbone of the templating strand. This work provides the molecular basis for the accommodation of epigenetic base modifications in a polymerase active site and suggests that these modifications are not mutagenically copied during base excision repair.
- Published
- 2019
- Full Text
- View/download PDF
45. Detection of TAR DNA-binding protein 43 (TDP-43) oligomers as initial intermediate species during aggregate formation.
- Author
-
French RL, Grese ZR, Aligireddy H, Dhavale DD, Reeb AN, Kedia N, Kotzbauer PT, Bieschke J, and Ayala YM
- Subjects
- Amyotrophic Lateral Sclerosis metabolism, Biomarkers metabolism, DNA-Binding Proteins genetics, Disulfides metabolism, HEK293 Cells, Humans, Molecular Weight, Mutation, Phase Transition, Protein Folding, RNA-Binding Proteins metabolism, Recombinant Proteins metabolism, Biopolymers metabolism, DNA-Binding Proteins metabolism
- Abstract
Aggregates of the RNA-binding protein TDP-43 (TAR DNA-binding protein) are a hallmark of the overlapping neurodegenerative disorders amyotrophic lateral sclerosis (ALS) and frontotemporal dementia. The process of TDP-43 aggregation remains poorly understood, and whether it includes formation of intermediate complexes is unknown. Here, we analyzed aggregates derived from purified TDP-43 under semidenaturing conditions, identifying distinct oligomeric complexes at the initial time points before the formation of large aggregates. We found that this early oligomerization stage is primarily driven by TDP-43's RNA-binding region. Specific binding to GU-rich RNA strongly inhibited both TDP-43 oligomerization and aggregation, suggesting that RNA interactions are critical for maintaining TDP-43 solubility. Moreover, we analyzed TDP-43 liquid-liquid phase separation and detected similar detergent-resistant oligomers upon maturation of liquid droplets into solid-like fibrils. These results strongly suggest that the oligomers form during the early steps of TDP-43 misfolding. Importantly, the ALS-linked TDP-43 mutations A315T and M337V significantly accelerate aggregation, rapidly decreasing the monomeric population and shortening the oligomeric phase. We also show that aggregates generated from purified TDP-43 seed intracellular aggregation detected by established TDP-43 pathology markers. Remarkably, cytoplasmic aggregate seeding was detected earlier for the A315T and M337V variants and was 50% more widespread than for WT TDP-43 aggregates. We provide evidence for an initial step of TDP-43 self-assembly into intermediate oligomeric complexes, whereby these complexes may provide a scaffold for aggregation. This process is altered by ALS-linked mutations, underscoring the role of perturbations in TDP-43 homeostasis in protein aggregation and ALS-FTD pathogenesis., (© 2019 French et al.)
- Published
- 2019
- Full Text
- View/download PDF
46. Histone deacetylase inhibitors induce complex host responses that contribute to differential potencies of these compounds in HIV reactivation.
- Author
-
Beliakova-Bethell N, Mukim A, White CH, Deshmukh S, Abewe H, Richman DD, and Spina CA
- Subjects
- CD4-Positive T-Lymphocytes pathology, CD4-Positive T-Lymphocytes virology, Female, Gene Expression Regulation drug effects, Humans, Male, Poly (ADP-Ribose) Polymerase-1 biosynthesis, SMARCB1 Protein biosynthesis, Transcription, Genetic drug effects, Virus Latency drug effects, CD4-Positive T-Lymphocytes metabolism, Depsipeptides pharmacology, HIV-1 physiology, Histone Deacetylase Inhibitors pharmacology, Virus Activation drug effects, Vorinostat pharmacology
- Abstract
Histone deacetylase (HDAC) inhibitors (HDACis) have been widely tested in clinical trials for their ability to reverse HIV latency but have yielded only limited success. One HDACi, suberoylanilide hydroxamic acid (SAHA), exhibits off-target effects on host gene expression predicted to interfere with induction of HIV transcription. Romidepsin (RMD) has higher potency and specificity for class I HDACs implicated in maintaining HIV provirus in the latent state. More robust HIV reactivation has indeed been achieved with RMD use ex vivo than with SAHA; however, reduction of viral reservoir size has not been observed in clinical trials. Therefore, using RNA-Seq, we sought to compare the effects of SAHA and RMD on gene expression in primary CD4
+ T cells. Among the genes whose expression was modulated by both HDACi agents, we identified genes previously implicated in HIV latency. Two genes, SMARCB1 and PARP1 , whose modulation by SAHA and RMD is predicted to inhibit HIV reactivation, were evaluated in the major maturation subsets of CD4+ T cells and were consistently either up- or down-regulated by both HDACi compounds. Our results indicate that despite having different potencies and HDAC specificities, SAHA and RMD modulate an overlapping set of genes, implicated in HIV latency regulation. Some of these genes merit exploration as additional targets to improve the therapeutic outcomes of "shock and kill" strategies. The overall complexity of HDACi-induced responses among host genes with predicted stimulatory or inhibitory effects on HIV expression likely contributes to differential HDACi potencies and dictates the outcome of HIV reactivation., (© 2019 by The American Society for Biochemistry and Molecular Biology, Inc.)- Published
- 2019
- Full Text
- View/download PDF
47. The yeast copper chaperone for copper-zinc superoxide dismutase (CCS1) is a multifunctional chaperone promoting all levels of SOD1 maturation.
- Author
-
Boyd SD, Calvo JS, Liu L, Ullrich MS, Skopp A, Meloni G, and Winkler DD
- Subjects
- Catalytic Domain, Disulfides metabolism, Molecular Chaperones chemistry, Protein Binding, Saccharomyces cerevisiae Proteins, Copper metabolism, Molecular Chaperones metabolism, Saccharomyces cerevisiae chemistry, Superoxide Dismutase-1 metabolism
- Abstract
Copper (Cu) is essential for the survival of aerobic organisms through its interaction with molecular oxygen (O
2 ). However, Cu's chemical properties also make it toxic, requiring specific cellular mechanisms for Cu uptake and handling, mediated by Cu chaperones. CCS1, the budding yeast ( S. cerevisiae ) Cu chaperone for Cu-zinc (Zn) superoxide dismutase (SOD1) activates by directly promoting both Cu delivery and disulfide formation in SOD1. The complete mechanistic details of this transaction along with recently proposed molecular chaperone-like functions for CCS1 remain undefined. Here, we present combined structural, spectroscopic, kinetic, and thermodynamic data that suggest a multifunctional chaperoning role(s) for CCS1 during SOD1 activation. We observed that CCS1 preferentially binds a completely immature form of SOD1 and that the SOD1·CCS1 interaction promotes high-affinity Zn(II) binding in SOD1. Conserved aromatic residues within the CCS1 C-terminal domain are integral in these processes. Previously, we have shown that CCS1 delivers Cu(I) to an entry site at the SOD1·CCS1 interface upon binding. We show here that Cu(I) is transferred from CCS1 to the entry site and then to the SOD1 active site by a thermodynamically driven affinity gradient. We also noted that efficient transfer from the entry site to the active site is entirely dependent upon the oxidation of the conserved intrasubunit disulfide bond in SOD1. Our results herein provide a solid foundation for proposing a complete molecular mechanism for CCS1 activity and reclassification as a first-of-its-kind "dual chaperone.", (© 2019 Boyd et al.)- Published
- 2019
- Full Text
- View/download PDF
48. Converter domain mutations in myosin alter structural kinetics and motor function.
- Author
-
Gunther LK, Rohde JA, Tang W, Walton SD, Unrath WC, Trivedi DV, Muretta JM, Thomas DD, and Yengo CM
- Subjects
- Adenosine Diphosphate metabolism, Adenosine Triphosphatases metabolism, Adenosine Triphosphate metabolism, Amino Acid Sequence, Animals, Chickens, Models, Molecular, Myosin Type V genetics, Protein Binding, Protein Conformation, Protein Domains, Sequence Homology, Mechanotransduction, Cellular, Motor Activity, Mutation, Myosin Type V chemistry, Myosin Type V metabolism
- Abstract
Myosins are molecular motors that use a conserved ATPase cycle to generate force. We investigated two mutations in the converter domain of myosin V (R712G and F750L) to examine how altering specific structural transitions in the motor ATPase cycle can impair myosin mechanochemistry. The corresponding mutations in the human β-cardiac myosin gene are associated with hypertrophic and dilated cardiomyopathy, respectively. Despite similar steady-state actin-activated ATPase and unloaded in vitro motility-sliding velocities, both R712G and F750L were less able to overcome frictional loads measured in the loaded motility assay. Transient kinetic analysis and stopped-flow FRET demonstrated that the R712G mutation slowed the maximum ATP hydrolysis and recovery-stroke rate constants, whereas the F750L mutation enhanced these steps. In both mutants, the fast and slow power-stroke as well as actin-activated phosphate release rate constants were not significantly different from WT. Time-resolved FRET experiments revealed that R712G and F750L populate the pre- and post-power-stroke states with similar FRET distance and distance distribution profiles. The R712G mutant increased the mole fraction in the post-power-stroke conformation in the strong actin-binding states, whereas the F750L decreased this population in the actomyosin ADP state. We conclude that mutations in key allosteric pathways can shift the equilibrium and/or alter the activation energy associated with key structural transitions without altering the overall conformation of the pre- and post-power-stroke states. Thus, therapies designed to alter the transition between structural states may be able to rescue the impaired motor function induced by disease mutations., (© 2019 Gunther et al.)
- Published
- 2019
- Full Text
- View/download PDF
49. Parkinson's disease and multiple system atrophy have distinct α-synuclein seed characteristics.
- Author
-
Yamasaki TR, Holmes BB, Furman JL, Dhavale DD, Su BW, Song ES, Cairns NJ, Kotzbauer PT, and Diamond MI
- Subjects
- Brain pathology, HEK293 Cells, Humans, Multiple System Atrophy pathology, Parkinson Disease pathology, Biosensing Techniques methods, Brain metabolism, Multiple System Atrophy metabolism, Parkinson Disease metabolism, alpha-Synuclein analysis
- Abstract
Parkinson's disease (PD) and multiple system atrophy (MSA) are distinct clinical syndromes characterized by the pathological accumulation of α-synuclein (α-syn) protein fibrils in neurons and glial cells. These disorders and other neurodegenerative diseases may progress via prion-like mechanisms. The prion model of propagation predicts the existence of "strains" that link pathological aggregate structure and neuropathology. Prion strains are aggregated conformers that stably propagate in vivo and cause disease with defined incubation times and patterns of neuropathology. Indeed, tau prions have been well defined, and research suggests that both α-syn and β-amyloid may also form strains. However, there is a lack of studies characterizing PD- versus MSA-derived α-syn strains or demonstrating stable propagation of these unique conformers between cells or animals. To fill this gap, we used an assay based on FRET that exploits a HEK293T "biosensor" cell line stably expressing α-syn (A53T)-CFP/YFP fusion proteins to detect α-syn seeds in brain extracts from PD and MSA patients. Both soluble and insoluble fractions of MSA extracts had robust seeding activity, whereas only the insoluble fractions of PD extracts displayed seeding activity. The morphology of MSA-seeded inclusions differed from PD-seeded inclusions. These differences persisted upon propagation of aggregation to second-generation biosensor cells. We conclude that PD and MSA feature α-syn conformers with very distinct biochemical properties that can be transmitted to α-syn monomers in a cell system. These findings are consistent with the idea that distinct α-syn strains underlie PD and MSA and offer possible directions for synucleinopathy diagnosis., (© 2019 Yamasaki et al.)
- Published
- 2019
- Full Text
- View/download PDF
50. Long-chain acyl-CoA synthetase 1 interacts with key proteins that activate and direct fatty acids into niche hepatic pathways.
- Author
-
Young PA, Senkal CE, Suchanek AL, Grevengoed TJ, Lin DD, Zhao L, Crunk AE, Klett EL, Füllekrug J, Obeid LM, and Coleman RA
- Subjects
- Animals, Female, Liver cytology, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Coenzyme A Ligases physiology, Endoplasmic Reticulum metabolism, Fatty Acids metabolism, Liver metabolism, Mitochondria metabolism, Protein Interaction Domains and Motifs
- Abstract
Fatty acid channeling into oxidation or storage modes depends on physiological conditions and hormonal signaling. However, the directionality of this channeling may also depend on the association of each of the five acyl-CoA synthetase isoforms with specific protein partners. Long-chain acyl-CoA synthetases (ACSLs) catalyze the conversion of long-chain fatty acids to fatty acyl-CoAs, which are then either oxidized or used in esterification reactions. In highly oxidative tissues, ACSL1 is located on the outer mitochondrial membrane (OMM) and directs fatty acids into mitochondria for β-oxidation. In the liver, however, about 50% of ACSL1 is located on the endoplasmic reticulum (ER) where its metabolic function is unclear. Because hepatic fatty acid partitioning is likely to require the interaction of ACSL1 with other specific proteins, we used an unbiased protein interaction technique, BioID, to discover ACSL1-binding partners in hepatocytes. We targeted ACSL1 either to the ER or to the OMM of Hepa 1-6 cells as a fusion protein with the Escherichia coli biotin ligase, BirA*. Proteomic analysis identified 98 proteins that specifically interacted with ACSL1 at the ER, 55 at the OMM, and 43 common to both subcellular locations. We found subsets of peroxisomal and lipid droplet proteins, tethering proteins, and vesicle proteins, uncovering a dynamic role for ACSL1 in organelle and lipid droplet interactions. Proteins involved in lipid metabolism were also identified, including acyl-CoA-binding proteins and ceramide synthase isoforms 2 and 5. Our results provide fundamental and detailed insights into protein interaction networks that control fatty acid metabolism., (© 2018 Young et al.)
- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.