Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
J Biol Chem ; 300(1): 105464, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37979917

ABSTRACT

Neuronal nitric oxide synthase (nNOS) is a homodimeric cytochrome P450-like enzyme that catalyzes the conversion of L-arginine to nitric oxide in the presence of NADPH and molecular oxygen. The binding of calmodulin (CaM) to a linker region between the FAD/FMN-containing reductase domain, and the heme-containing oxygenase domain is needed for electron transfer reactions, reduction of the heme, and NO synthesis. Due to the dynamic nature of the reductase domain and low resolution of available full-length structures, the exact conformation of the CaM-bound active complex during heme reduction is still unresolved. Interestingly, hydrogen-deuterium exchange and mass spectrometry studies revealed interactions of the FMN domain and CaM with the oxygenase domain for iNOS, but not nNOS. This finding prompted us to utilize covalent crosslinking and mass spectrometry to clarify interactions of CaM with nNOS. Specifically, MS-cleavable bifunctional crosslinker disuccinimidyl dibutyric urea was used to identify thirteen unique crosslinks between CaM and nNOS as well as 61 crosslinks within the nNOS. The crosslinks provided evidence for CaM interaction with the oxygenase and reductase domain residues as well as interactions of the FMN domain with the oxygenase dimer. Cryo-EM studies, which gave a high-resolution model of the oxygenase domain, along with crosslink-guided docking provided a model of nNOS that brings the FMN within 15 Å of the heme in support for a more compact conformation than previously observed. These studies also point to the utility of covalent crosslinking and mass spectrometry in capturing transient dynamic conformations that may not be captured by hydrogen-deuterium exchange and mass spectrometry experiments.


Subject(s)
Calmodulin , Cross-Linking Reagents , Models, Molecular , Nitric Oxide Synthase Type I , Calmodulin/metabolism , Heme/metabolism , Mass Spectrometry , Nitric Oxide Synthase Type I/metabolism , Oxygenases/metabolism , Cross-Linking Reagents/chemistry , Calcium/chemistry , Protein Structure, Quaternary , Protein Binding , Cryoelectron Microscopy
2.
J Biol Chem ; 299(2): 102856, 2023 02.
Article in English | MEDLINE | ID: mdl-36596358

ABSTRACT

Heat shock protein 90 (Hsp90) is known to mediate heme insertion and activation of heme-deficient neuronal nitric oxide (NO) synthase (apo-nNOS) in cells by a highly dynamic interaction that has been extremely difficult to study mechanistically with the use of subcellular systems. In that the heme content of many critical hemeproteins is regulated by Hsp90 and the heme chaperone GAPDH, the development of an in vitro system for the study of this chaperone-mediated heme regulation would be extremely useful. Here, we show that use of an antibody-immobilized apo-nNOS led not only to successful assembly of chaperone complexes but the ability to show a clear dependence on Hsp90 and GAPDH for heme-mediated activation of apo-nNOS. The kinetics of binding for Hsp70 and Hsp90, the ATP and K+ dependence, and the absolute requirement for Hsp70 in assembly of Hsp90•apo-nNOS heterocomplexes all point to a similar chaperone machinery to the well-established canonical machine regulating steroid hormone receptors. However, unlike steroid receptors, the use of a purified protein system containing Hsp90, Hsp70, Hsp40, Hop, and p23 is unable to activate apo-nNOS. Thus, heme insertion requires a unique Hsp90-chaperone complex. With this newly developed in vitro system, which recapitulates the cellular process requiring GAPDH as well as Hsp90, further mechanistic studies are now possible to better understand the components of the Hsp90-based chaperone system as well as how this heterocomplex works with GAPDH to regulate nNOS and possibly other hemeproteins.


Subject(s)
Glyceraldehyde-3-Phosphate Dehydrogenases , HSP70 Heat-Shock Proteins , HSP90 Heat-Shock Proteins , Heme , Hemeproteins , Molecular Chaperones , Nitric Oxide Synthase , Heme/chemistry , Hemeproteins/chemistry , Hemeproteins/metabolism , HSP70 Heat-Shock Proteins/chemistry , HSP70 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/chemistry , HSP90 Heat-Shock Proteins/metabolism , Molecular Chaperones/chemistry , Molecular Chaperones/metabolism , Protein Binding , Nitric Oxide Synthase/chemistry , Nitric Oxide Synthase/metabolism , Enzymes, Immobilized , Glyceraldehyde-3-Phosphate Dehydrogenases/chemistry , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Enzyme Activation
3.
Mol Pharmacol ; 98(3): 243-249, 2020 09.
Article in English | MEDLINE | ID: mdl-32591478

ABSTRACT

Heat shock protein (Hsp) 70 modulators are being developed to enhance the removal of toxic proteins in a variety of protein misfolding diseases. In the course of our studies on neuronal nitric oxide synthase (nNOS), a client of the Hsp90 and Hsp70 chaperone system, we have established that inactivation of nNOS by heme or tetrahydrobiopterin (BH4) alteration and loss triggers ubiquitination by the Hsp70-associated E3 ligase c-terminus of Hsp70-interacting protein (CHIP) and subsequent degradation in cells. Although in cells Hsp90 and Hsp70 work together to maintain protein quality control, in this study, we specifically developed an assay to assess the selectivity of the Hsp70:CHIP complex for inactivated nNOS. We developed a highly sensitive ELISA to measure Hsp70:CHIP-dependent nNOS ubiquitination without interference from direct ubiquitination by CHIP, as evidenced by Bcl-2 associated athanogene 1-M completely abolishing ubiquitination. To further validate the assay we demonstrated, JG-98, a rhodocyanin compound that acts on Hsp70 but not its inactive structural analog JG-258, enhances the ubiquitination of nNOS 3-fold. Utilizing this assay, we have shown that the Hsp70:CHIP complex preferentially ubiquitinates heme-deficient nNOS (apo-nNOS) over heme-containing nNOS (holo-nNOS). Moreover, depletion of nNOS-bound BH4 triggers ubiquitination of holo-nNOS by the Hsp70:CHIP complex. Most importantly, JG-98 was shown to enhance the ubiquitination of only dysfunctional nNOS while leaving the native functional nNOS untouched. Thus, the finding that enhancing Hsp70:CHIP-mediated ubiquitination does not affect native proteins has important pharmacological implications. Moreover, development of a facile in vitro method for Hsp70:CHIP-mediated ubiquitination will be beneficial for testing other Hsp70 modulators. SIGNIFICANCE STATEMENT: The heat shock protein 70 (Hsp70):c-terminus of Hsp70-interacting protein (CHIP) complex facilitates the ubiquitination and subsequent degradation of several hundred-client proteins, and activation of Hsp70 has been suggested as a therapeutic strategy to enhance the degradation of disease-causing proteins. The current study shows that the pharmacological activation of Hsp70 enhances the ubiquitination of dysfunctional but not native nNOS, and it suggests that this therapeutic strategy will likely be highly selective.


Subject(s)
HSP70 Heat-Shock Proteins/metabolism , Nitric Oxide Synthase Type I/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , DNA-Binding Proteins/metabolism , Enzyme-Linked Immunosorbent Assay , Humans , Sf9 Cells , Transcription Factors/metabolism , Ubiquitination
4.
Mol Pharmacol ; 94(3): 984-991, 2018 09.
Article in English | MEDLINE | ID: mdl-29941666

ABSTRACT

Several hundred proteins cycle into heterocomplexes with a dimer of the chaperone heat shock protein 90 (Hsp90), regulating their activity and turnover. There are two isoforms of Hsp90, Hsp90α and Hsp90ß, and their relative chaperone activities and composition in these client protein•Hsp90 heterocomplexes has not been determined. Here, we examined the activity of human Hsp90α and Hsp90ß in a purified five-protein chaperone machinery that assembles glucocorticoid receptor (GR)•Hsp90 heterocomplexes to generate high-affinity steroid-binding activity. We found that human Hsp90α and Hsp90ß have equivalent chaperone activities, and when mixed together in this assay, they formed only GR•Hsp90αα and GR•Hsp90ßß homodimers and no GR•Hsp90αß heterodimers. In contrast, GR•Hsp90 heterocomplexes formed in human embryonic kidney (HEK) cells also contain GR•Hsp90αß heterodimers. The formation of GR•Hsp90αß heterodimers in HEK cells probably reflects the longer time permitted for exchange to form Hsp90αß heterodimers in the cell versus in the cell-free assembly conditions. This purified GR-activating chaperone machinery can be used to determine how modifications of Hsp90 affect its chaperone activity. To that effect, we have tested whether the unique phosphorylation of Hsp90α at threonines 5 and 7 that occurs during DNA damage repair affects its chaperone activity. We showed that the phosphomimetic mutant Hsp90α T5/7D has the same intrinsic chaperone activity as wild-type human Hsp90α in activation of GR steroid-binding activity by the five-protein machinery, supporting the conclusion that T5/7 phosphorylation does not affect Hsp90α chaperone activity.


Subject(s)
HSP70 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/metabolism , Molecular Chaperones/metabolism , Protein Multimerization/physiology , Receptors, Glucocorticoid/metabolism , Animals , HEK293 Cells , HSP70 Heat-Shock Proteins/chemistry , HSP90 Heat-Shock Proteins/chemistry , Humans , Mice , Molecular Chaperones/chemistry , Protein Binding/physiology , Receptors, Glucocorticoid/chemistry
5.
Anal Biochem ; 511: 24-6, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27487179

ABSTRACT

The assembly of mutated and wild type monomers into functional heterodimeric hemeproteins has provided important mechanistic insights. As in the case of NO synthase (NOS), the existing methods to make such heterodimeric NOSs are inefficient and labor intensive with typical yields of about 5%. We have found that expression of neuronal NOS heterodimers in insect cells, where we take advantage of an exogenous heme-triggered chaperone-assisted assembly process, provides an approximately 43% yield in heterodimeric NOS. In contrast, in Escherichia coli little heterodimerization occurred. Thus, insect cells are preferred and may represent a valuable method for assembly of other dimeric hemeproteins.


Subject(s)
Hemeproteins/chemistry , Nitric Oxide Synthase Type I/chemistry , Protein Multimerization , Animals , Hemeproteins/genetics , Hemeproteins/metabolism , Humans , Nitric Oxide Synthase Type I/genetics , Nitric Oxide Synthase Type I/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sf9 Cells , Spodoptera
6.
J Biol Chem ; 289(24): 16855-65, 2014 Jun 13.
Article in English | MEDLINE | ID: mdl-24737326

ABSTRACT

Nitric-oxide synthase (NOS) is required in mammals to generate NO for regulating blood pressure, synaptic response, and immune defense. NOS is a large homodimer with well characterized reductase and oxygenase domains that coordinate a multistep, interdomain electron transfer mechanism to oxidize l-arginine and generate NO. Ca(2+)-calmodulin (CaM) binds between the reductase and oxygenase domains to activate NO synthesis. Although NOS has long been proposed to adopt distinct conformations that alternate between interflavin and FMN-heme electron transfer steps, structures of the holoenzyme have remained elusive and the CaM-bound arrangement is unknown. Here we have applied single particle electron microscopy (EM) methods to characterize the full-length of the neuronal isoform (nNOS) complex and determine the structural mechanism of CaM activation. We have identified that nNOS adopts an ensemble of open and closed conformational states and that CaM binding induces a dramatic rearrangement of the reductase domain. Our three-dimensional reconstruction of the intact nNOS-CaM complex reveals a closed conformation and a cross-monomer arrangement with the FMN domain rotated away from the NADPH-FAD center, toward the oxygenase dimer. This work captures, for the first time, the reductase-oxygenase structural arrangement and the CaM-dependent release of the FMN domain that coordinates to drive electron transfer across the domains during catalysis.


Subject(s)
Calmodulin/metabolism , Catalytic Domain , Nitric Oxide Synthase Type I/chemistry , Amino Acid Sequence , Animals , Calmodulin/chemistry , Holoenzymes/chemistry , Holoenzymes/metabolism , Molecular Sequence Data , Nitric Oxide Synthase Type I/metabolism , Protein Binding , Rats
7.
J Biol Chem ; 287(51): 42601-10, 2012 Dec 14.
Article in English | MEDLINE | ID: mdl-23109339

ABSTRACT

Nitric-oxide synthase, a cytochrome P450-like hemoprotein enzyme, catalyzes the synthesis of nitric oxide, a critical signaling molecule in a variety of physiological processes. Our laboratory has discovered that certain drugs suicide-inactivate neuronal nitric-oxide synthase (nNOS) and lead to the preferential ubiquitination of the inactivated nNOS by an Hsp70- and CHIP (C terminus of Hsc70-interacting protein)-dependent process. To further understand the process by which altered nNOS is recognized, ubiquitinated, and proteasomally degraded, we examined the sites of ubiquitination on nNOS. We utilized an in vitro ubiquitination system containing purified E1, E2 (UbcH5a), Hsp70, and CHIP that recapitulates the ability of the cells to selectively recognize and ubiquitinate the altered forms of nNOS. LC-MS/MS analysis of the tryptic peptides obtained from the in vitro ubiquitinated nNOS identified 12 ubiquitination sites. Nine of the sites were within the oxygenase domain and two were in the calmodulin-binding site, which links the oxygenase and reductase domains, and one site was in the reductase domain. Mutational analysis of the lysines in the calmodulin-binding site revealed that Lys-739 is a major site for poly-ubiquitination of nNOS in vitro and regulates, in large part, the CHIP-dependent degradation of nNOS in HEK293 cells, as well as in in vitro studies with fraction II. Elucidating the exact site of ubiquitination is an important step in understanding how chaperones recognize and trigger degradation of nNOS.


Subject(s)
Calmodulin/metabolism , Nitric Oxide Synthase Type I/chemistry , Nitric Oxide Synthase Type I/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Ubiquitination , Amino Acid Sequence , Animals , Binding Sites , Chromatography, Liquid , HEK293 Cells , HSP70 Heat-Shock Proteins/metabolism , Heme/metabolism , Humans , Lysine/metabolism , Mass Spectrometry , Models, Biological , Molecular Sequence Data , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Nitroarginine/pharmacology , Protein Binding , Rats , Stereoisomerism , Substrate Specificity , Ubiquitin-Protein Ligases
8.
Biochemistry ; 50(33): 7146-56, 2011 Aug 23.
Article in English | MEDLINE | ID: mdl-21755988

ABSTRACT

We have reported that heme-dependent activation of apo-neuronal nitric oxide synthase (apo-nNOS) to the active holo-enzyme dimer is dependent upon factors present in reticulocyte lysate and other cytosols. Here, we find that both Hsp70 and thioredoxin are components of the activation system. The apo-nNOS activating activity of reticulocyte lysate is retained in a pool of fractions containing Hsp70 that elute from DE52 prior to Hsp90. All of the activating activity and 20-30% of the Hsp70 elute in the flow-through fraction upon subsequent ATP-agarose chromatography. Apo-nNOS activation by this flow-through fraction is inhibited by pifithrin-µ, a small molecule inhibitor of Hsp70, suggesting that a non-ATP-binding form of Hsp70 is involved in heme-dependent apo-nNOS activation. Previous work has shown that apo-nNOS can be activated by thiol-disulfide exchange, and we show substantial activation with a small molecule dithiol modeled on the active motifs of thioredoxin and protein disulfide isomerase. Further fractionation of the ATP-agarose flow-through on Sephacryl S-300 separates free thioredoxin from apo-nNOS activating activity, Hsp70, and a small amount of thioredoxin, all of which are eluted throughout the macromolecular peak. Incubation of apo-nNOS with the macromolecular fraction in combination either with the thioredoxin-containing fraction or with purified recombinant human thioredoxin restores full heme-dependent activating activity. This supports a model in which Hsp70 binding to apo-nNOS stabilizes an open state of the heme/substrate binding cleft to facilitate thioredoxin access to the active site cysteine that coordinates with heme iron, permitting heme binding and dimerization to the active enzyme.


Subject(s)
Cytosol/metabolism , Disulfides/metabolism , HSP70 Heat-Shock Proteins/metabolism , Heme/metabolism , Nitric Oxide Synthase/metabolism , Sulfhydryl Compounds/metabolism , Thioredoxins/metabolism , Animals , Apoenzymes , Blood Coagulation Factors/metabolism , Dimerization , Enzyme Activation , Humans , Immunoblotting , Nitric Oxide Synthase Type I , Protein Binding , RNA-Binding Proteins , Rabbits , Rats , Reticulocytes , Ribosomal Proteins , Sepharose/analogs & derivatives
9.
J Biol Chem ; 285(44): 33642-51, 2010 Oct 29.
Article in English | MEDLINE | ID: mdl-20729196

ABSTRACT

It is established that suicide inactivation of neuronal nitric-oxide synthase (nNOS) by drugs and other xenobiotics leads to ubiquitination and proteasomal degradation of the enzyme. The exact mechanism is not known, although it is widely thought that the covalent alteration of the active site during inactivation triggers the degradation. A mechanism that involves recognition of the altered nNOS by Hsp70 and its cochaperone CHIP, an E3-ubiquitin ligase, has been proposed. To further address how alterations of the active site trigger ubiquitination of nNOS, we examined a C331A nNOS mutant, which was reported to have impaired ability to bind L-arginine and tetrahydrobiopterin. We show here that C331A nNOS is highly susceptible to ubiquitination by a purified system containing ubiquitinating enzymes and chaperones, by the endogenous ubiquitinating system in reticulocyte lysate fraction II, and by intact HEK293 cells. The involvement of the altered heme cleft in regulating ubiquitination is confirmed by the finding that the slowly reversible inhibitor of nNOS, N(G)-nitro-L-arginine, but not its inactive D-isomer, protects the C331A nNOS from ubiquitination in all these experimental systems. We also show that both Hsp70 and CHIP play a major role in the ubiquitination of C331A nNOS, although Hsp90 protects from ubiquitination. Thus, these studies further strengthen the link between the mobility of the substrate-binding cleft and chaperone-dependent ubiquitination of nNOS. These results support a general model of chaperone-mediated protein quality control and lead to a novel mechanism for substrate stabilization based on nNOS interaction with the chaperone machinery.


Subject(s)
Cytochrome P-450 Enzyme System/chemistry , HSP70 Heat-Shock Proteins/metabolism , Mutation , Nitric Oxide Synthase Type I/genetics , Nitric Oxide Synthase Type I/metabolism , Ubiquitin-Protein Ligases/chemistry , Ubiquitin/chemistry , Catalytic Domain , Cell Line , Chromatin Immunoprecipitation , Heat-Shock Proteins/chemistry , Humans , Ligands , Molecular Chaperones/chemistry , Protein Structure, Tertiary , Subcellular Fractions , Xenobiotics/chemistry
10.
Biophys Chem ; 274: 106590, 2021 07.
Article in English | MEDLINE | ID: mdl-33894563

ABSTRACT

Covalent crosslinking and mass spectrometry techniques hold great potential in the study of multiprotein complexes, but a major challenge is the inability to differentiate intra- and inter- protein crosslinks in homomeric complexes. In the current study we use CYP102A1, a well-characterized homodimeric P450, to examine a subtractive method that utilizes limited crosslinking with disuccinimidyl dibutyric urea (DSBU) and isolation of the monomer, in addition to the crosslinked dimer, to identify inter-monomer crosslinks. The utility of this approach was examined with the use of MS-cleavable crosslinker DSBU and recently published cryo-EM based structures of the CYP102A1 homodimer. Of the 31 unique crosslinks found, 26 could be fit to the reported structures whereas 5 exceeded the spatial constraints. Not only did these crosslinks validate the cryo-EM structure, they point to new conformations of CYP102A1 that bring the flavins in closer proximity to the heme.


Subject(s)
Bacterial Proteins/chemistry , Cross-Linking Reagents/chemistry , Cytochrome P-450 Enzyme System/chemistry , NADPH-Ferrihemoprotein Reductase/chemistry , Mass Spectrometry , Models, Molecular , Protein Binding
11.
Biochemistry ; 48(35): 8483-90, 2009 Sep 08.
Article in English | MEDLINE | ID: mdl-19642705

ABSTRACT

NO production by neuronal nitric oxide synthase (nNOS) requires calmodulin and is enhanced by the chaperone Hsp90, which cycles dynamically with the enzyme. The proteasomal degradation of nNOS is enhanced by suicide inactivation and by treatment with Hsp90 inhibitors, the latter suggesting that dynamic cycling with Hsp90 stabilizes nNOS. Here, we use a purified ubiquitinating system containing CHIP (carboxyl terminus of Hsp70-interacting protein) as the E3 ligase to show that Hsp90 inhibits CHIP-dependent nNOS ubiquitination. Like the established Hsp90 enhancement of NO synthesis, Hsp90 inhibition of nNOS ubiquitination is Ca2+/calmodulin-dependent, suggesting that the same interaction of Hsp90 with the enzyme is responsible for both enhancement of nNOS activity and inhibition of ubiquitination. It is established that CHIP binds to Hsp90 as well as to Hsp70, but we show here the two chaperones have opposing actions on nNOS ubiquitination, with Hsp70 stimulating and Hsp90 inhibiting. We have used two mechanism-based inactivators, guanabenz and NG-amino-L-arginine, to alter the heme/substrate binding cleft and promote nNOS ubiquitination that can be inhibited by Hsp90. We envision that, as nNOS undergoes toxic damage, the heme/substrate binding cleft opens exposing hydrophobic residues as the initial step in unfolding. As long as Hsp90 can form even transient complexes with the opening cleft, ubiquitination by Hsp70-dependent ubiquitin E3 ligases, like CHIP, is inhibited. When unfolding of the cleft progresses to a state that cannot cycle with Hsp90, Hsp70-dependent ubiquitination is unopposed. In this way, the Hsp70/Hsp90 machinery makes the quality control decision for stabilization versus degradation of nNOS.


Subject(s)
Calmodulin/pharmacology , HSP90 Heat-Shock Proteins/metabolism , Nitric Oxide Synthase Type I/metabolism , Ubiquitin/metabolism , Ubiquitination/drug effects , Animals , Cell Line , Cyclization , Dimerization , Humans , Rabbits , Reticulocytes/enzymology , Reticulocytes/metabolism , Signal Transduction , Spodoptera
12.
Brain Res Mol Brain Res ; 142(1): 19-27, 2005 Dec 07.
Article in English | MEDLINE | ID: mdl-16216381

ABSTRACT

Tetrahydrobiopterin is a necessary cofactor for the synthesis of nitric oxide by the hemeprotein enzyme, NO-synthase (NOS). It is widely thought that inadequate levels of tetrahydrobiopterin lead to tissue injury and organ dysfunction due, in part, to formation of superoxide from pterin-deficient NOS. In the course of studies on the ubiquitylation of neuronal NOS (nNOS), we have found that certain substrate analogs, such as N(G)-nitro-L-arginine, stabilize the dimeric form of nNOS and protect the enzyme from ubiquitylation. Since tetrahydrobiopterin is known to bind near heme and confers stability to the active dimeric structure of nNOS, we wondered if the loss of tetrahydrobiopterin could be an endogenous signal for nNOS ubiquitylation and degradation. We show here in HEK293 cells stably transfected with nNOS that depletion of tetrahydrobiopterin by treatment with 2,4-diamino-6-hydroxypyrimidine leads to destabilization of the dimeric form and enhances ubiquitylation of nNOS. Sepiapterin, a precursor to tetrahydrobiopterin in the salvage pathway, completely reverses the effect of 2,4-diamino-6-hydroxypyrimidine on nNOS ubiquitylation. Consistent with that found in cells, the in vitro ubiquitylation of nNOS by reticulocyte proteins decreases when tetrahydrobiopterin is present. Thus, inadequate amounts of tetrahydrobiopterin may lead to a sustained decrease in the steady state level of nNOS that is not readily reversed.


Subject(s)
Biopterins/analogs & derivatives , Nitric Oxide Synthase Type I/metabolism , Ubiquitins/metabolism , Biopterins/metabolism , Biopterins/pharmacology , Blotting, Western/methods , Cell Line , Chromatography, High Pressure Liquid/methods , Dimerization , Dose-Response Relationship, Drug , Drug Interactions , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Heme/metabolism , Humans , Hypoxanthines/pharmacology , Immunoprecipitation/methods , Leupeptins/pharmacology , Nitric Oxide Synthase Type I/chemistry , Pterins/pharmacology , Time Factors
13.
Plant Signal Behav ; 2(2): 129-30, 2007 Mar.
Article in English | MEDLINE | ID: mdl-19704759

ABSTRACT

Cigarette smoking is known to cause a decrease in NO production in man resulting in a variety of pathological effects, including vascular dysfunction. Aqueous extracts of cigarette and cigarette smoke contain chemical inhibitors to NO-synthases, a heme-containing cytochrome P450 enzymes. More recently, it was shown that freshly harvested leaves from the tobacco plant (Nicotiana tabacum, Solanaceae) also contain chemical inhibitors to neuronal NO-synthase (nNOS). Examination of leaves from 32 other plants representing diverse members of the plant kingdom showed that 17 other plants, besides tobacco, contain these chemical inhibitors. Of all these plants, 16 are members of the core asterids flowering plant group and 6 are members of the Solanaceae family. Although the identity of the chemicals is not known, perhaps the closely related plants contain the same or similar compounds that inhibit nNOS. The inhibitory effects are not attributable to nicotine. The discovery of these chemicals and their further characterization may help to explain the loss of nNOS in smokers. In this addendum, we discuss these results in light of the effect of tobacco-derived chemicals in inhibiting P450 cytochromes, as well as our thoughts on how the inactivation of nNOS leads to its selective downregulation through proteolytic degradation.

14.
Phytomedicine ; 14(5): 344-52, 2007 May.
Article in English | MEDLINE | ID: mdl-17084601

ABSTRACT

NO-synthase (NOS) is a heme-containing enzyme that catalyzes the oxidation of L-arginine to nitric oxide, an important cellular signaling molecule. Recently, it was found that aqueous extracts of tobacco cigarettes cause the inactivation of the neuronal isoform of NOS (nNOS) and that this may explain some of the toxicological effects of smoking. Although the exact identity of the chemical inactivator(s) is not known, we wondered if extracts prepared from other plants, including those closely related to tobacco, Nicotiana tabacum (Solanaceae), would similarly inactivate nNOS. We examined 33 plants, representing diverse members of the plant kingdom ranging from whisk fern, Psilotum nudum (Psilotaceae) to tobacco and discovered 18 plants that contain a chemical inactivator(s) of nNOS. Of these plants, 16 are members of the core asterids flowering plant group. Of these asterids, 6 are members of the Solanaceae family, of which tobacco is a member. Based on the phylogenetic relationship of the plants, it is possible that the same chemical or related chemical inactivator(s) exist. This, in turn, may help elucidate the structure of the chemical(s), as well as provide a source of a potentially novel inactivator of nNOS. The alkaloid nicotine can be excluded as putative nNOS inhibitor.


Subject(s)
Enzyme Inhibitors/pharmacology , Nicotiana , Nitric Oxide Synthase Type I/antagonists & inhibitors , Phytotherapy , Plant Extracts/pharmacology , Chromatography, High Pressure Liquid , Enzyme Inhibitors/chemistry , Humans , Inhibitory Concentration 50 , Nitric Oxide Synthase Type I/chemistry , Plant Components, Aerial , Plant Extracts/chemistry
15.
Drug Metab Dispos ; 34(9): 1448-56, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16738031

ABSTRACT

It is established that guanabenz inhibits neuronal nitric-oxide (NO) synthase (nNOS) and causes the enhanced proteasomal degradation of nNOS in vivo. Although the time- and NADPH-dependent inhibition of nNOS has been reported in studies where guanabenz was incubated with crude cytosolic preparations of nNOS, the exact mechanism for inhibition is not known. Moreover, even less is known about how the inhibition of nNOS triggers its proteasomal degradation. In the current study, we show, with the use of purified nNOS, that guanabenz treatment leads to the oxidation of tetrahydrobiopterin and formation of a pterin-depleted nNOS, which is not able to form NO. With the use of 14C-labeled guanabenz, we were unable to detect any guanabenz metabolites or guanabenz-nNOS adducts, indicating that reactive intermediates of guanabenz probably do not play a role in the inhibition. Superoxide dismutase, however, prevents the guanabenz-mediated oxidation of tetrahydrobiopterin and inhibition of nNOS, suggesting the role of superoxide as an intermediate. Studies in rats show that administration of tetrahydrobiopterin prevents the inhibition and loss of penile nNOS due to guanabenz, indicating that the loss of tetrahydrobiopterin plays a major role in the effects of guanabenz in vivo. Our findings are consistent with the destabilization and enhanced degradation of nNOS found after tetrahydrobiopterin depletion. These studies suggest that drug-mediated destabilization and subsequent enhanced degradation of protein targets will likely be an important toxicological consideration.


Subject(s)
Biopterins/analogs & derivatives , Enzyme Inhibitors/pharmacology , Guanabenz/pharmacology , Nitric Oxide Synthase Type I/antagonists & inhibitors , Nitric Oxide Synthase/antagonists & inhibitors , Penis/drug effects , Animals , Biopterins/chemistry , Biopterins/metabolism , Biopterins/pharmacology , Dose-Response Relationship, Drug , Drug Interactions , Enzyme Stability , Male , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type I/chemistry , Nitric Oxide Synthase Type I/metabolism , Oxidation-Reduction , Penis/enzymology , Rats , Rats, Wistar , Superoxide Dismutase/chemistry , Superoxide Dismutase/metabolism , Time Factors , Ubiquitin/metabolism
16.
Chem Res Toxicol ; 18(12): 1927-33, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16359183

ABSTRACT

It is established that aminoguanidine (AG), diaminoguanidine (DAG), and NG-amino-l-arginine (NAA) are metabolism-based inactivators of the three major isoforms of nitric oxide synthase (NOS). In the case of neuronal NOS (nNOS), heme alteration is known to be a major cause of inactivation, although the exact mechanism by which this occurs is not well-understood. We show here by the use of LC/MS/MS techniques that AG, DAG, and NAA are metabolized by nNOS to products with corresponding mass ions at m/z of 45.2, 60.2, and 160.0, respectively. These results are consistent with the loss of a hydrazine moiety from each inactivator. These findings are confirmed by exact mass measurements and comparison to authentic standards in the case of the products for NAA and AG, respectively. Moreover, the major dissociable heme product that was formed during inactivation of nNOS by AG, DAG, and NAA had molecular ions at m/z 660.2, 675.2, and 775.3, respectively. These results are consistent with an adduct of heme and inactivator minus a hydrazine moiety. In support of this, MS/MS studies reveal a fragment ion of heme in each case. With the use of 14C-labeled heme, we also show that in the case of AG, the dissociable heme adduct accounts for approximately one-half of the heme that is altered. In addition, we employ a software-based differential metabolic profiling method by subtracting LC/MS data sets derived from samples that contained nNOS from those that did not contain the enzyme to search for products and substrates in complex reaction mixtures. The metabolic profiling method established in this study can be used as a general tool to search for substrates and products of enzyme systems, including the drug-metabolizing liver microsomal P450 cytochromes. We propose that the metabolism-based inactivation of nNOS by AG, DAG, and NAA occurs through oxidative removal of the hydrazine group and the formation of a radical intermediate that forms stable products after H-atom abstraction or reacts with the heme prosthetic moiety and inactivates nNOS.


Subject(s)
Arginine/analogs & derivatives , Guanidines/metabolism , Heme/metabolism , Nitric Oxide Synthase Type I/metabolism , Arginine/metabolism , Arginine/pharmacology , Cells, Cultured , Enzyme Activation/drug effects , Gas Chromatography-Mass Spectrometry , Guanidines/pharmacology , Heme/chemistry , Models, Chemical , Molecular Structure , Nitric Oxide Synthase Type I/antagonists & inhibitors
17.
Drug Metab Dispos ; 33(1): 131-8, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15470159

ABSTRACT

Smoking causes a dysfunction in endothelial nitric-oxide synthase (eNOS), which is ameliorated, in part, by administration of tetrahydrobiopterin (BH(4)). The exact mechanism by which the nitric oxide deficit occurs is unknown. We have previously shown that aqueous extracts of chemicals in cigarettes (CE) cause the suicide inactivation of neuronal NO synthase (nNOS) by interacting at the substrate-binding site. In the current study, we have found that CE directly inactivates eNOS by a process that is not affected by the natural substrate l-arginine and is distinct from the mechanism of inactivation of nNOS. We discovered that CE causes a time-, concentration-, and NADPH-dependent inactivation of eNOS in an in vitro system containing the purified enzyme, indicating a metabolic component to the inactivation. The CE-treated eNOS but not nNOS was nearly fully reactivated upon incubation with excess BH(4), suggesting that BH(4) depletion is a potential mechanism of inactivation. Moreover, in the presence of CE, eNOS catalyzed the oxidation of BH(4) to dihydrobiopterin and biopterin by a process attenuated by high concentrations of superoxide dismutase but not catalase. We speculate that a redox active component in CE, perhaps a quinone compound, causes oxidative uncoupling of eNOS to form superoxide, which in turn oxidizes BH(4). The discovery of a direct inactivation of eNOS by a compound(s) present in tobacco provides a basis not only for further study of the mechanisms responsible for the biological effects of tobacco but also a search for a potentially novel inactivator of eNOS.


Subject(s)
Biopterins/analogs & derivatives , Biopterins/metabolism , Nitric Oxide Synthase/antagonists & inhibitors , Tars/pharmacology , Animals , Dose-Response Relationship, Drug , Enzyme Inhibitors/isolation & purification , Enzyme Inhibitors/pharmacology , Insecta , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type III , Rats , Smoking/metabolism , Tars/isolation & purification , Time Factors
18.
J Biol Chem ; 277(29): 26021-30, 2002 Jul 19.
Article in English | MEDLINE | ID: mdl-11983704

ABSTRACT

The redox active metal copper is an essential cofactor in critical biological processes such as respiration, iron transport, oxidative stress protection, hormone production, and pigmentation. A widely conserved family of high affinity copper transport proteins (Ctr proteins) mediates copper uptake at the plasma membrane. However, little is known about Ctr protein topology, structure, and the mechanisms by which this class of transporters mediates high affinity copper uptake. In this report, we elucidate the topological orientation of the yeast Ctr1 copper transport protein. We show that a series of clustered methionine residues in the hydrophilic extracellular domain and an MXXXM motif in the second transmembrane domain are important for copper uptake but not for protein sorting and delivery to the cell surface. The conversion of these methionine residues to cysteine, by site-directed mutagenesis, strongly suggests that they coordinate to copper during the process of metal transport. Genetic evidence supports an essential role for cooperativity between monomers for the formation of an active Ctr transport complex. Together, these results support a fundamentally conserved mechanism for high affinity copper uptake through the Ctr proteins in yeast and humans.


Subject(s)
Cation Transport Proteins , Copper/metabolism , Membrane Proteins/metabolism , Saccharomyces cerevisiae Proteins , Amino Acid Sequence , Cell Line , Cell Membrane/metabolism , Copper Transporter 1 , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Methionine/metabolism , Molecular Sequence Data , Mutagenesis, Site-Directed , Oxidation-Reduction , Oxidative Stress , Oxygen Consumption , Peptide Mapping , Protein Conformation , Saccharomyces cerevisiae , Sequence Alignment
19.
J Biol Chem ; 279(29): 30298-306, 2004 Jul 16.
Article in English | MEDLINE | ID: mdl-15133041

ABSTRACT

Iron and copper are redox active metals essential for life. In the budding yeast Saccharomyces cerevisiae, expression of iron and copper genes involved in metal acquisition and utilization is tightly regulated at the transcriptional level. In addition iron and copper metabolism are inextricably linked because of the dependence on copper as a co-factor for iron uptake or mobilization. To further identify genes that function in iron and copper homeostasis, we screened for novel yeast mutants defective for iron limiting growth and thereby identified the CTI6 gene. Cti6 is a PHD finger-containing protein that has been shown to participate in the interaction of the Ssn6-Tup1 co-repressor with the Gcn5-containing SAGA chromatin-remodeling complex. In this report we show that CTI6 mRNA levels are increased under iron-limiting conditions, and that cti6 mutants display a growth defect under conditions of iron deprivation. Furthermore, we demonstrate that Cti6 is a nuclear protein that functionally associates with the Rpd3-Sin3 histone deacetylase complex involved in transcriptional repression. Cti6 demonstrates Rpd3-dependent transcriptional repression, and cti6 mutants exhibit an enhanced silencing of telomeric, rDNA and HMR loci, similar to mutants in genes encoding other Rpd3-Sin3-associated proteins. Microarray experiments with cti6 mutants grown under iron-limiting conditions show a down-regulation of telomeric genes and an up-regulation of Aft1 and Tup1 target genes involved in iron and oxygen regulation. Taken together, these data suggest a specific role for Cti6 in the regulation of gene expression under conditions of iron limitation.


Subject(s)
Carrier Proteins/metabolism , Histone Deacetylases/chemistry , Histone Deacetylases/metabolism , Iron/metabolism , Repressor Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Transcription Factors/metabolism , Amino Acid Motifs , Carrier Proteins/chemistry , Cell Division , Cell Nucleus/metabolism , Copper/metabolism , DNA, Ribosomal/chemistry , Dose-Response Relationship, Drug , Down-Regulation , Gene Silencing , Genetic Vectors , Genome, Fungal , Genotype , Green Fluorescent Proteins , Iron/chemistry , Luminescent Proteins/chemistry , Microscopy, Fluorescence , Models, Genetic , Mutation , Oligonucleotide Array Sequence Analysis , Oxidation-Reduction , Oxygen/metabolism , Protein Structure, Tertiary , RNA/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Transcription, Genetic , Up-Regulation , beta-Galactosidase/metabolism
20.
J Biol Chem ; 279(51): 52970-7, 2004 Dec 17.
Article in English | MEDLINE | ID: mdl-15466472

ABSTRACT

It is established that neuronal nitric-oxide synthase (nNOS) is ubiquitylated and proteasomally degraded. The proteasomal degradation of nNOS is enhanced by suicide inactivation of nNOS or by the inhibition of hsp90, which is a chaperone found in a native complex with nNOS. In the current study, we have examined whether CHIP, a chaperone-dependent E3 ubiquitin-protein isopeptide ligase that is known to ubiquitylate other hsp90-chaperoned proteins, could act as an ubiquitin ligase for nNOS. We found with the use of HEK293T or COS-7 cells and transient transfection methods that CHIP overexpression causes a decrease in immunodetectable levels of nNOS. The extent of the loss of nNOS is dependent on the amount of CHIP cDNA used for transfection. Lactacystin (10 microM), a selective proteasome inhibitor, attenuates the loss of nNOS in part by causing the nNOS to be found in a detergent-insoluble form. Immunoprecipitation of the nNOS and subsequent Western blotting with an anti-ubiquitin IgG shows an increase in nNOS-ubiquitin conjugates because of CHIP. Moreover, incubation of nNOS with a purified system containing an E1 ubiquitin-activating enzyme, an E2 ubiquitin carrier protein conjugating enzyme (UbcH5a), CHIP, glutathione S-transferase-tagged ubiquitin, and an ATP-generating system leads to the ubiquitylation of nNOS. The addition of purified hsp70 and hsp40 to this in vitro system greatly enhances the amount of nNOS-ubiquitin conjugates, suggesting that CHIP is an E3 ligase for nNOS whose action is facilitated by (and possibly requires) its interaction with nNOS-bound hsp70.


Subject(s)
Acetylcysteine/analogs & derivatives , HSP90 Heat-Shock Proteins/metabolism , Nitric Oxide Synthase/metabolism , Ubiquitin-Protein Ligases/physiology , Ubiquitin/metabolism , Acetylcysteine/metabolism , Adenosine Triphosphate/metabolism , Animals , Blotting, Western , COS Cells , Cell Line , Cysteine Proteinase Inhibitors/pharmacology , DNA, Complementary/metabolism , Detergents/pharmacology , Dose-Response Relationship, Drug , Glutathione Transferase/metabolism , HSP70 Heat-Shock Proteins/metabolism , Humans , Immunoglobulin G/chemistry , Immunoprecipitation , Lactones/pharmacology , Macrolides , Nitric Oxide Synthase Type I , Palmitic Acids/metabolism , Proteasome Inhibitors , Protein Structure, Tertiary , Rabbits , Rats , Time Factors , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL