Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters











Database
Language
Publication year range
1.
Elife ; 72018 12 18.
Article in English | MEDLINE | ID: mdl-30561332

ABSTRACT

The postdoctoral community is an essential component of the academic and scientific workforce, but a lack of data about this community has made it difficult to develop policies to address concerns about salaries, working conditions, diversity and career development, and to evaluate the impact of existing policies. Here we present comprehensive survey results from 7,603 postdocs based at 351 US academic and non-academic (e.g. hospital, industry and government lab) institutions in 2016. In addition to demographic and salary information, we present multivariate analyses on factors influencing postdoc career plans and satisfaction with mentorship. We further analyze gender dynamics and expose wage disparities. Academic research positions remain the predominant career choice, although women and US citizens are less likely than their male and non-US citizen counterparts to choose academic research positions. Receiving mentorship training has a significant positive effect on postdoc satisfaction with mentorship. Quality of and satisfaction with postdoc mentorship also appear to heavily influence career choice.


Subject(s)
Career Choice , Gender Identity , Mentors , Research Personnel , Age Distribution , Ethnicity , Female , Humans , Male , Surveys and Questionnaires , United States
2.
PLoS One ; 10(11): e0143523, 2015.
Article in English | MEDLINE | ID: mdl-26618351

ABSTRACT

Soluble guanylyl cyclase (sGC) is a heterodimeric nitric oxide (NO) receptor that produces cyclic GMP. This signaling mechanism is a key component in the cardiovascular system. NO binds to heme in the ß subunit and stimulates the catalytic conversion of GTP to cGMP several hundred fold. Several endogenous factors have been identified that modulate sGC function in vitro and in vivo. In previous work, we determined that protein disulfide isomerase (PDI) interacts with sGC in a redox-dependent manner in vitro and that PDI inhibited NO-stimulated activity in cells. To our knowledge, this was the first report of a physical interaction between sGC and a thiol-redox protein. To characterize this interaction between sGC and PDI, we first identified peptide linkages between sGC and PDI, using a lysine cross-linking reagent and recently developed mass spectrometry analysis. Together with Flag-immunoprecipitation using sGC domain deletions, wild-type (WT) and mutated PDI, regions of sGC involved in this interaction were identified. The observed data were further explored with computational modeling to gain insight into the interaction mechanism between sGC and oxidized PDI. Our results indicate that PDI interacts preferentially with the catalytic domain of sGC, thus providing a mechanism for PDI inhibition of sGC. A model in which PDI interacts with either the α or the ß catalytic domain is proposed.


Subject(s)
Guanylate Cyclase/chemistry , Protein Disulfide-Isomerases/chemistry , Receptors, Cytoplasmic and Nuclear/chemistry , Amino Acid Sequence , Animals , COS Cells , Catalytic Domain , Chlorocebus aethiops , Guanylate Cyclase/metabolism , Molecular Sequence Data , Protein Binding , Protein Disulfide-Isomerases/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Soluble Guanylyl Cyclase
3.
Biochem J ; 452(1): 161-9, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23477350

ABSTRACT

NO binds to the receptor sGC (soluble guanylyl cyclase), stimulating cGMP production. The NO-sGC-cGMP pathway is a key component in the cardiovascular system. Discrepancies in sGC activation and deactivation in vitro compared with in vivo have led to a search for endogenous factors that regulate sGC or assist in cellular localization. In our previous work, which identified Hsp (heat-shock protein) 70 as a modulator of sGC, we determined that PDI (protein disulfide-isomerase) bound to an sGC-affinity matrix. In the present study, we establish and characterize this interaction. Incubation of purified PDI with semi-purified sGC, both reduced and oxidized, resulted in different migration patterns on non-reducing Western blots indicating a redox component to the interaction. In sGC-infected COS-7 cells, transfected FLAG-tagged PDI and PDI CXXS (redox active site 'trap mutant') pulled down sGC. This PDI-sGC complex was resolved by reductant, confirming a redox interaction. PDI inhibited NO-stimulated sGC activity in COS-7 lysates, however, a PDI redox-inactive mutant PDI SXXS did not. Together, these data unveil a novel mechanism of sGC redox modulation via thiol-disulfide exchange. Finally, in SMCs (smooth muscle cells), endogenous PDI and sGC co-localize by in situ proximity ligation assay, which suggests biological relevance. PDI-dependent redox regulation of sGC NO sensitivity may provide a secondary control over vascular homoeostasis.


Subject(s)
Guanylate Cyclase/metabolism , Protein Disulfide-Isomerases/metabolism , Protein Interaction Mapping , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , COS Cells , Chlorocebus aethiops , Enzyme Activation/physiology , Guanylate Cyclase/chemistry , Humans , Mice , Oxidation-Reduction , Protein Binding/physiology , Protein Disulfide-Isomerases/chemistry , Protein Interaction Mapping/methods , Rats , Receptors, Cytoplasmic and Nuclear/chemistry , Soluble Guanylyl Cyclase
4.
PLoS One ; 6(11): e26976, 2011.
Article in English | MEDLINE | ID: mdl-22096512

ABSTRACT

Nitric oxide signals through activation of soluble guanylyl cyclase (sGC), a heme-containing heterodimer. NO binds to the heme domain located in the N-terminal part of the ß subunit of sGC resulting in increased production of cGMP in the catalytic domain located at the C-terminal part of sGC. Little is known about the mechanism by which the NO signaling is propagated from the receptor domain (heme domain) to the effector domain (catalytic domain), in particular events subsequent to the breakage of the bond between the heme iron and Histidine 105 (H105) of the ß subunit. Our modeling of the heme-binding domain as well as previous homologous heme domain structures in different states point to two regions that could be critical for propagation of the NO activation signal. Structure-based mutational analysis of these regions revealed that residues T110 and R116 in the αF helix-ß1 strand, and residues I41 and R40 in the αB-αC loop mediate propagation of activation between the heme domain and the catalytic domain. Biochemical analysis of these heme mutants allows refinement of the map of the residues that are critical for heme stability and propagation of the NO/YC-1 activation signal in sGC.


Subject(s)
Guanylate Cyclase/chemistry , Guanylate Cyclase/metabolism , Heme/metabolism , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Blotting, Western , COS Cells , Catalytic Domain , Chlorocebus aethiops , Enzyme Activation/genetics , Enzyme Activation/physiology , Guanylate Cyclase/genetics , Mutation , Protein Structure, Tertiary , Rats , Receptors, Cytoplasmic and Nuclear/genetics , Soluble Guanylyl Cyclase
5.
Biochemistry ; 50(20): 4291-7, 2011 May 24.
Article in English | MEDLINE | ID: mdl-21491881

ABSTRACT

Nitric oxide (NO) is involved in the physiology and pathophysiology of the cardiovascular and neuronal systems via activation of soluble guanylyl cyclase (sGC), a heme-containing heterodimer. Recent structural studies have allowed a better understanding of the residues that dictate the affinity and binding of NO to the heme and the resulting breakage of the bond between the heme iron and histidine 105 (H105) of the ß subunit of sGC. Still, it is unknown how the breakage of the iron-His bond translates into NO-dependent increased catalysis. Structural studies on homologous H-NOX domains in various states pointed to a role for movement of the H105 containing αF helix. Our modeling of the heme-binding domain highlighted conserved residues in the vicinity of H105 that could potentially regulate the extent to which the αF helix shifts and/or propagate the activation signal once the covalent bond with H105 has been broken. These include a direct interaction of αF helix residue aspartate 102 (D102) with the backbone nitrogen of F120. Mutational analysis of this region points to an essential role of the interactions in the vicinity of H105 for heme stability and identifies D102 as having a key role in NO activation following breakage of the iron-His bond.


Subject(s)
Aspartic Acid/metabolism , Guanylate Cyclase/chemistry , Guanylate Cyclase/metabolism , Heme/chemistry , Heme/metabolism , Nitric Oxide/pharmacology , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/metabolism , Alanine/metabolism , Animals , COS Cells , Chlorocebus aethiops , Enzyme Activation/drug effects , Guanylate Cyclase/genetics , Indazoles/pharmacology , Models, Molecular , Mutagenesis, Site-Directed , Mutation , Protein Structure, Tertiary , Protein Subunits/chemistry , Protein Subunits/metabolism , Rats , Receptors, Cytoplasmic and Nuclear/genetics , Sequence Homology, Amino Acid , Soluble Guanylyl Cyclase
6.
FEBS Lett ; 584(8): 1521-5, 2010 Apr 16.
Article in English | MEDLINE | ID: mdl-20211621

ABSTRACT

Quiescin sulfhydryl oxidase (QSOX) catalyzes formation of disulfide bonds between cysteine residues in substrate proteins. Human QSOX1 is a multi-domain, monomeric enzyme containing a module related to the single-domain sulfhydryl oxidases of the Erv family. A partial QSOX1 crystal structure reveals a single-chain pseudo-dimer mimicking the quaternary structure of Erv enzymes. However, one pseudo-dimer "subunit" has lost its cofactor and catalytic activity. In QSOX evolution, a further concatenation to a member of the protein disulfide isomerase family resulted in an enzyme capable of both disulfide formation and efficient transfer to substrate proteins.


Subject(s)
Oxidoreductases Acting on Sulfur Group Donors/chemistry , Oxidoreductases Acting on Sulfur Group Donors/metabolism , Oxidoreductases/chemistry , Protein Multimerization , Amino Acid Sequence , Catalytic Domain , Crystallography, X-Ray , Humans , Models, Molecular , Oxidoreductases Acting on Sulfur Group Donors/genetics , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Structure, Quaternary
7.
Biochemistry ; 47(17): 4955-63, 2008 Apr 29.
Article in English | MEDLINE | ID: mdl-18393449

ABSTRACT

The flavoprotein quiescin-sulfhydryl oxidase (QSOX) rapidly inserts disulfide bonds into unfolded, reduced proteins with the concomitant reduction of oxygen to hydrogen peroxide. This study reports the first heterologous expression and enzymological characterization of a human QSOX1 isoform. Like QSOX isolated from avian egg white, recombinant HsQSOX1 is highly active toward reduced ribonuclease A (RNase) and dithiothreitol but shows a >100-fold lower k cat/ K m for reduced glutathione. Previous studies on avian QSOX led to a model in which reducing equivalents were proposed to relay through the enzyme from the first thioredoxin domain (C70-C73) to a distal disulfide (C509-C512), then across the dimer interface to the FAD-proximal disulfide (C449-C452), and finally to the FAD. The present work shows that, unlike the native avian enzyme, HsQSOX1 is monomeric. The recombinant expression system enabled construction of the first cysteine mutants for mechanistic dissection of this enzyme family. Activity assays with mutant HsQSOX1 indicated that the conserved distal C509-C512 disulfide is dispensable for the oxidation of reduced RNase or dithiothreitol. The four other cysteine residues chosen for mutagenesis, C70, C73, C449, and C452, are all crucial for efficient oxidation of reduced RNase. C452, of the proximal disulfide, is shown to be the charge-transfer donor to the flavin ring of QSOX, and its partner, C449, is expected to be the interchange thiol, forming a mixed disulfide with C70 in the thioredoxin domain. These data demonstrate that all the internal redox steps occur within the same polypeptide chain of mammalian QSOX and commence with a direct interaction between the reduced thioredoxin domain and the proximal disulfide of the Erv/ALR domain.


Subject(s)
Mutagenesis , Thioredoxins/genetics , Thioredoxins/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Catalysis , Cattle , Cysteine/metabolism , Disulfides/metabolism , Egg Proteins/chemistry , Egg Proteins/genetics , Gene Expression Regulation, Bacterial , Humans , Milk Proteins/chemistry , Milk Proteins/genetics , Molecular Sequence Data , Oxidation-Reduction , Oxidoreductases Acting on Sulfur Group Donors , Protein Folding , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Analysis, Protein , Sequence Homology, Amino Acid , Thioredoxins/chemistry
8.
Biochim Biophys Acta ; 1783(4): 567-77, 2008 Apr.
Article in English | MEDLINE | ID: mdl-17980160

ABSTRACT

The Quiescin-sulfhydryl oxidase (QSOX) family of flavoenzymes catalyzes the direct and facile insertion of disulfide bonds into unfolded reduced proteins with concomitant reduction of oxygen to hydrogen peroxide. This review discusses the chemical mechanism of these enzymes and the involvement of thioredoxin and flavin-binding domains in catalysis. The variability of CxxC motifs in the QSOX family is highlighted and attention is drawn to the steric factors that may promote efficient thiol/disulfide exchange during oxidative protein folding. The varied cellular location of these multi-domain sulfhydryl oxidases is reviewed and potential intracellular and extracellular roles are summarized. Finally, this review identifies important unresolved questions concerning this ancient family of sulfhydryl oxidases.


Subject(s)
Disulfides/metabolism , Thioredoxins/physiology , Amino Acid Sequence , Animals , Humans , Molecular Sequence Data , Oxidation-Reduction , Oxidoreductases Acting on Sulfur Group Donors , Protein Folding
SELECTION OF CITATIONS
SEARCH DETAIL