Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Life Sci Alliance ; 6(12)2023 12.
Article in English | MEDLINE | ID: mdl-37793775

ABSTRACT

The Dam1 complex is essential for mitotic progression across evolutionarily divergent fungi. Upon analyzing amino acid (aa) sequences of Dad2, a Dam1 complex subunit, we identified a conserved 10-aa-long Dad2 signature sequence (DSS). An arginine residue (R126) in the DSS is essential for viability in Saccharomyces cerevisiae that possesses point centromeres. The corresponding arginine residues are functionally important but not essential for viability in Candida albicans and Cryptococcus neoformans; both carry several kilobases long regional centromeres. The purified recombinant Dam1 complex containing either Dad2ΔDSS or Dad2R126A failed to bind microtubules (MTs) or form any visible rings like the WT complex. Intriguingly, functional analysis revealed that the requirement of the conserved arginine residue for chromosome biorientation and mitotic progression reduced with increasing centromere length. We propose that plasticity of the invariant arginine of Dad2 in organisms with regional centromeres is achieved by conditional elevation of the kinetochore protein(s) to enable multiple kinetochore MTs to bind to each chromosome. The capacity of a chromosome to bind multiple kinetochore MTs may mask the deleterious effects of such lethal mutations.


Subject(s)
Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Cell Cycle Proteins/genetics , Microtubules/genetics , Microtubules/metabolism , Centromere/genetics , Centromere/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Arginine/genetics , Arginine/metabolism
2.
Immunol Cell Biol ; 99(4): 373-391, 2021 04.
Article in English | MEDLINE | ID: mdl-33131099

ABSTRACT

Viperin is an interferon-inducible protein that is pivotal for eliciting an effective immune response against an array of diverse viral pathogens. Here we describe a mechanism of viperin's broad antiviral activity by demonstrating the protein's ability to synergistically enhance the innate immune dsDNA signaling pathway to limit viral infection. Viperin co-localized with the key signaling molecules of the innate immune dsDNA sensing pathway, STING and TBK1; binding directly to STING and inducing enhanced K63-linked polyubiquitination of TBK1. Subsequent analysis identified viperin's necessity to bind the cytosolic iron-sulfur assembly component 2A, to prolong its enhancement of the type-I interferon response to aberrant dsDNA. Here we show that viperin facilitates the formation of a signaling enhanceosome, to coordinate efficient signal transduction following activation of the dsDNA signaling pathway, which results in an enhanced antiviral state. We also provide evidence for viperin's radical SAM enzymatic activity to self-limit its immunomodulatory functions. These data further define viperin's role as a positive regulator of innate immune signaling, offering a mechanism of viperin's broad antiviral capacity.


Subject(s)
Interferon Type I , DNA , Protein Binding , Proteins/metabolism , Signal Transduction
3.
J Biol Chem ; 295(50): 17227-17240, 2020 12 11.
Article in English | MEDLINE | ID: mdl-33051205

ABSTRACT

Heme oxygenase-2 (HO2) and -1 (HO1) catalyze heme degradation to biliverdin, CO, and iron, forming an essential link in the heme metabolism network. Tight regulation of the cellular levels and catalytic activities of HO1 and HO2 is important for maintaining heme homeostasis. HO1 expression is transcriptionally regulated; however, HO2 expression is constitutive. How the cellular levels and activity of HO2 are regulated remains unclear. Here, we elucidate the mechanism of post-translational regulation of cellular HO2 levels by heme. We find that, under heme-deficient conditions, HO2 is destabilized and targeted for degradation, suggesting that heme plays a direct role in HO2 regulation. HO2 has three heme binding sites: one at its catalytic site and the others at its two heme regulatory motifs (HRMs). We report that, in contrast to other HRM-containing proteins, the cellular protein level and degradation rate of HO2 are independent of heme binding to the HRMs. Rather, under heme deficiency, loss of heme binding to the catalytic site destabilizes HO2. Consistently, an HO2 catalytic site variant that is unable to bind heme exhibits a constant low protein level and an enhanced protein degradation rate compared with the WT HO2. Finally, HO2 is degraded by the lysosome through chaperone-mediated autophagy, distinct from other HRM-containing proteins and HO1, which are degraded by the proteasome. These results reveal a novel aspect of HO2 regulation and deepen our understanding of HO2's role in maintaining heme homeostasis, paving the way for future investigation into HO2's pathophysiological role in heme deficiency response.


Subject(s)
Heme Oxygenase (Decyclizing)/metabolism , Heme/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Catalytic Domain , Enzyme Stability , HEK293 Cells , Heme/genetics , Heme Oxygenase (Decyclizing)/genetics , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Humans , Proteasome Endopeptidase Complex/genetics
4.
Nat Commun ; 11(1): 2728, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32483114

ABSTRACT

The Pseudomonas putida phenol-responsive regulator DmpR is a bacterial enhancer binding protein (bEBP) from the AAA+ ATPase family. Even though it was discovered more than two decades ago and has been widely used for aromatic hydrocarbon sensing, the activation mechanism of DmpR has remained elusive. Here, we show that phenol-bound DmpR forms a tetramer composed of two head-to-head dimers in a head-to-tail arrangement. The DmpR-phenol complex exhibits altered conformations within the C-termini of the sensory domains and shows an asymmetric orientation and angle in its coiled-coil linkers. The structural changes within the phenol binding sites and the downstream ATPase domains suggest that the effector binding signal is propagated through the coiled-coil helixes. The tetrameric DmpR-phenol complex interacts with the σ54 subunit of RNA polymerase in presence of an ATP analogue, indicating that DmpR-like bEBPs tetramers utilize a mechanistic mode distinct from that of hexameric AAA+ ATPases to activate σ54-dependent transcription.


Subject(s)
Adenosine Triphosphatases/chemistry , Bacterial Proteins/chemistry , DNA-Binding Proteins/chemistry , Protein Conformation , Protein Multimerization , Trans-Activators/chemistry , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , DNA-Directed RNA Polymerases/metabolism , Gene Expression Regulation, Bacterial , Phenol/metabolism , Protein Binding , Pseudomonas putida/enzymology , Pseudomonas putida/genetics , Sequence Homology, Amino Acid , Trans-Activators/genetics , Trans-Activators/metabolism
5.
J Biol Chem ; 295(9): 2839-2849, 2020 02 28.
Article in English | MEDLINE | ID: mdl-31980458

ABSTRACT

Understanding the mechanisms by which viruses evade host cell immune defenses is important for developing improved antiviral therapies. In an unusual twist, human cytomegalovirus co-opts the antiviral radical SAM enzyme viperin (virus-inhibitory protein, endoplasmic reticulum-associated, interferon-inducible) to enhance viral infectivity. This process involves translocation of viperin to the mitochondrion, where it binds the ß-subunit (HADHB) of the mitochondrial trifunctional enzyme complex that catalyzes thiolysis of ß-ketoacyl-CoA esters as part of fatty acid ß-oxidation. Here we investigated how the interaction between these two enzymes alters their activities and affects cellular ATP levels. Experiments with purified enzymes indicated that viperin inhibits the thiolase activity of HADHB, but, unexpectedly, HADHB activates viperin, leading to synthesis of the antiviral nucleotide 3'-deoxy-3',4'-didehydro-CTP. Measurements of enzyme activities in lysates prepared from transfected HEK293T cells expressing these enzymes mirrored the findings obtained with purified enzymes. Thus, localizing viperin to mitochondria decreased thiolase activity, and coexpression of HADHB significantly increased viperin activity. Furthermore, targeting viperin to mitochondria also increased the rate at which HADHB is retrotranslocated out of mitochondria and degraded, providing an additional mechanism by which viperin reduces HADHB activity. Targeting viperin to mitochondria decreased cellular ATP levels by more than 50%, consistent with the enzyme disrupting fatty acid catabolism. These results provide biochemical insight into the mechanism by which human cytomegalovirus subverts viperin; they also provide a biochemical rationale for viperin's recently discovered role in regulating thermogenesis in adipose tissues.


Subject(s)
Acetyl-CoA C-Acetyltransferase/antagonists & inhibitors , Mitochondria/metabolism , Proteins/metabolism , Adenosine Triphosphate/metabolism , Cytomegalovirus/physiology , HEK293 Cells , Humans , Immune Evasion , Mitochondrial Trifunctional Protein, beta Subunit/antagonists & inhibitors , Mitochondrial Trifunctional Protein, beta Subunit/metabolism , Mitochondrial Trifunctional Protein, beta Subunit/physiology , Oxidoreductases Acting on CH-CH Group Donors
6.
Biochemistry ; 59(6): 780-789, 2020 02 18.
Article in English | MEDLINE | ID: mdl-31977203

ABSTRACT

The radical SAM enzyme, viperin, exerts a wide range of antiviral effects through both the synthesis of the antiviral nucleotide 3'-deoxy-3',4'-didehydro-CTP (ddhCTP) and through its interactions with various cellular and viral proteins. Here we investigate the interaction of viperin with hepatitis C virus nonstructural protein 5A (NS5A) and the host sterol regulatory protein, vesicle-associated membrane protein A (VAP-33). NS5A and VAP-33 form part of the viral replication complex that is essential for replicating the RNA genome of the hepatitis C virus. Using transfected enzymes in HEK293T cells, we show that viperin binds independently to both NS5A and the C-terminal domain of VAP-33 (VAP-33C) and that this interaction is dependent on the proteins being colocalized to the ER membrane. Coexpression of VAP-33C and NS5A resulted in changes to the catalytic activity of viperin that depended upon viperin being colocalized to the ER membrane. The viperin-NS5A-VAP-33C complex exhibited the lowest specific activity, indicating that NS5A may inhibit viperin's ability to synthesize ddhCTP. Coexpression of viperin with NS5A was also found to significantly reduce cellular NS5A levels, most likely by increasing the rate of proteasomal degradation. An inactive mutant of viperin, unable to bind the iron-sulfur cluster, was similarly effective at reducing cellular NS5A levels.


Subject(s)
Proteins/metabolism , Proteolysis , Vesicular Transport Proteins/metabolism , Viral Nonstructural Proteins/metabolism , Amino Acid Sequence , Enzyme Activation/physiology , HEK293 Cells , Hepacivirus/metabolism , Humans , Oxidoreductases Acting on CH-CH Group Donors , Protein Binding/physiology , Protein Structure, Secondary , Proteins/chemistry , Vesicular Transport Proteins/chemistry , Viral Core Proteins/chemistry , Viral Core Proteins/metabolism , Viral Nonstructural Proteins/chemistry , Virus Replication/physiology
7.
J Biol Chem ; 294(17): 6888-6898, 2019 04 26.
Article in English | MEDLINE | ID: mdl-30872404

ABSTRACT

Virus-inhibitory protein, endoplasmic reticulum-associated, interferon-inducible (viperin) is a radical SAM enzyme that plays a multifaceted role in the cellular antiviral response. Viperin has recently been shown to catalyze the SAM-dependent formation of 3'-deoxy-3',4'-didehydro-CTP (ddhCTP), which inhibits some viral RNA polymerases. Viperin is also implicated in regulating Lys-63-linked polyubiquitination of interleukin-1 receptor-associated kinase-1 (IRAK1) by the E3 ubiquitin ligase tumor necrosis factor receptor-associated factor 6 (TRAF6) as part of the Toll-like receptor-7 and -9 (TLR7/9) innate immune signaling pathways. In these pathways, the poly-ubiquitination of IRAK1 by TRAF6 is necessary to activate IRAK1, which then phosphorylates downstream targets and ultimately leads to the production of type I interferons. That viperin is a component of these pathways suggested that its enzymatic activity might be regulated by interactions with partner proteins. To test this idea, we have reconstituted the interactions between viperin, IRAK1, and TRAF6 by transiently expressing these enzymes in HEK 293T cells. We show that IRAK1 and TRAF6 increase viperin activity ∼10-fold to efficiently catalyze the radical-mediated dehydration of CTP to ddhCTP. Furthermore, we found that TRAF6-mediated ubiquitination of IRAK1 requires the association of viperin with both IRAK1 and TRAF6. Ubiquitination appears to depend on structural changes in viperin induced by SAM binding, but, significantly, does not require catalytically active viperin. We conclude that the synergistic activation of viperin and IRAK1 provides a mechanism that couples innate immune signaling with the production of the antiviral nucleotide ddhCTP.


Subject(s)
Antiviral Agents/metabolism , Cytidine Triphosphate/biosynthesis , Immunity, Innate , Interleukin-1 Receptor-Associated Kinases/metabolism , Proteins/metabolism , Signal Transduction , TNF Receptor-Associated Factor 6/metabolism , Adenosine/administration & dosage , Adenosine/analogs & derivatives , HEK293 Cells , Half-Life , Humans , Intracellular Signaling Peptides and Proteins , Oxidoreductases Acting on CH-CH Group Donors , Phosphorylation , Protein Binding , S-Adenosylmethionine/metabolism , Ubiquitination
8.
J Microbiol Biotechnol ; 22(12): 1724-30, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23221536

ABSTRACT

An alpha-L-arabinofuranosidase (TmAFase) from Thermotoga maritima MSB8 is a highly thermostable exo-acting hemicellulase that exhibits a relatively higher activity towards arabinan and arabinoxylan, compared with other glycoside hydrolase 51 family enzymes. In the present study, we carried out the enzymatic characterization and structural analysis of TmAFase. Tight domain associations found in TmAFase, such as an inter-domain disulfide bond (Cys306 and Cys476) in each monomer, a novel extended arm (amino acids 374-385) at the dimer interface, and total 12 salt bridges in the hexamer, may account for the thermostability of the enzyme. One of the xylan binding determinants (Trp96) was identified in the active site, and a region of amino acids (374-385) protrudes out forming an obvious wall at the substrate-binding groove to generate a cavity. The altered cavity shape with a strong negative electrostatic distribution is likely related to the unique substrate preference of TmAFase towards branched polymeric substrates.


Subject(s)
Bacterial Proteins/chemistry , Glycoside Hydrolases/chemistry , Thermotoga maritima/enzymology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Catalytic Domain , Crystallography, X-Ray , Enzyme Stability , Escherichia coli/genetics , Glycoside Hydrolases/genetics , Glycoside Hydrolases/metabolism , Models, Molecular , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Thermotoga maritima/genetics
9.
Analyst ; 135(11): 2879-86, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20877819

ABSTRACT

Phytoestrogens are plant compounds that mimic the actions of endogenous estrogens. The abundance of these chemicals in nature and their potential effects on health require the development of a convenient method to detect phytoestrogens. We have developed a nanoparticle (NP)-conjugated FRET probe based on the human estrogen receptor α (ER) ligand-binding domain (LBD) to detect phytoestrogens. The NP-conjugated FRET probe showed fluorescence signals for genistein, resveratrol and daidzein compounds with Δ ratios of 1.65, 2.60 and 1.37 respectively, which are approximately six times greater compared to individual FRET probes. A significantly higher signal for resveratrol versus genistein and daidzein indicates that the probe can differentiate between antagonistic phytoalexin substances and agonistic isoflavone compounds. NP-conjugated probes demonstrated a wide dynamic range, ranging from 10(-18) to 10(-1) M with EC(50) values of 9.6 × 10(-10), 9.0 × 10(-10) and 9.2 × 10(-10) M for genistein, daidzein and resveratrol respectively, whereas individual probes detected concentrations of 10(-13) to 10(-4) M for phytoestrogens compounds. The time profile revealed that the NP-conjugated probe is stable over 30 h and there is not a significant deviation in the FRET signal at room temperature. These data demonstrate that conjugation of a FRET probe to nanoparticles is able to serve as an effective FRET sensor for monitoring bioactive compounds with significantly increased sensitivity, dynamic range and stability.


Subject(s)
Fluorescence Resonance Energy Transfer/methods , Fluorescent Dyes/chemistry , Nanoparticles/chemistry , Phytoestrogens/analysis , Binding Sites , Estrogen Receptor alpha/chemistry , Genistein/analysis , Humans , Isoflavones/analysis , Ligands , Resveratrol , Sensitivity and Specificity , Stilbenes/analysis
10.
Proteins ; 78(2): 348-56, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-19768689

ABSTRACT

The debranching enzyme Nostoc punctiforme debranching enzyme (NPDE) from the cyanobacterium Nostoc punctiforme (PCC73102) hydrolyzes the alpha-1,6 glycosidic linkages of malto-oligosaccharides. Despite its high homology to cyclodextrin/pullulan (CD/PUL)-hydrolyzing enzymes from glycosyl hydrolase 13 family (GH-13), NPDE exhibits a unique catalytic preference for longer malto-oligosaccharides (>G8), performing hydrolysis without the transgylcosylation or CD-hydrolyzing activities of other GH-13 enzymes. To investigate the molecular basis for the property of NPDE, we determined the structure of NPDE at 2.37-A resolution. NPDE lacks the typical N-terminal domain of other CD/PUL-hydrolyzing enzymes and forms an elongated dimer in a head-to-head configuration. The unique orientation of residues 25-55 in NPDE yields an extended substrate binding groove from the catalytic center to the dimeric interface. The substrate binding groove with a lengthy cavity beyond the -1 subsite exhibits a suitable architecture for binding longer malto-oligosaccharides (>G8). These structural results may provide a molecular basis for the substrate specificity and catalytic function of this cyanobacterial enzyme, distinguishing it from the classical neopullulanases and CD/PUL-hydrolyzing enzymes.


Subject(s)
Bacterial Proteins/chemistry , Nostoc/enzymology , Amino Acid Sequence , Catalytic Domain , Crystallography, X-Ray , Models, Molecular , Molecular Sequence Data , Protein Conformation , Protein Multimerization , Sequence Alignment , Substrate Specificity
11.
Appl Environ Microbiol ; 74(1): 333-5, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17981933

ABSTRACT

Efficient lactic acid production from cane sugar molasses by Lactobacillus delbrueckii mutant Uc-3 in batch fermentation process is demonstrated. Lactic acid fermentation using molasses was not significantly affected by yeast extract concentrations. The final lactic acid concentration increased with increases of molasses sugar concentrations up to 190 g/liter. The maximum lactic acid concentration of 166 g/liter was obtained at a molasses sugar concentration of 190 g/liter with a productivity of 4.15 g/liter/h. Such a high concentration of lactic acid with high productivity from molasses has not been reported previously, and hence mutant Uc-3 could be a potential candidate for economical production of lactic acid from molasses at a commercial scale.


Subject(s)
Carbohydrate Metabolism , Lactic Acid/metabolism , Lactobacillus delbrueckii/metabolism , Molasses , Culture Media/chemistry , Fermentation
SELECTION OF CITATIONS
SEARCH DETAIL
...