Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 158
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 182(4): 812-827.e19, 2020 08 20.
Article in English | MEDLINE | ID: mdl-32697968

ABSTRACT

A SARS-CoV-2 variant carrying the Spike protein amino acid change D614G has become the most prevalent form in the global pandemic. Dynamic tracking of variant frequencies revealed a recurrent pattern of G614 increase at multiple geographic levels: national, regional, and municipal. The shift occurred even in local epidemics where the original D614 form was well established prior to introduction of the G614 variant. The consistency of this pattern was highly statistically significant, suggesting that the G614 variant may have a fitness advantage. We found that the G614 variant grows to a higher titer as pseudotyped virions. In infected individuals, G614 is associated with lower RT-PCR cycle thresholds, suggestive of higher upper respiratory tract viral loads, but not with increased disease severity. These findings illuminate changes important for a mechanistic understanding of the virus and support continuing surveillance of Spike mutations to aid with development of immunological interventions.


Subject(s)
Betacoronavirus/genetics , Betacoronavirus/pathogenicity , Coronavirus Infections/virology , Pneumonia, Viral/virology , Spike Glycoprotein, Coronavirus/genetics , COVID-19 , Coronavirus Infections/epidemiology , Coronavirus Infections/physiopathology , Epidemiological Monitoring , Genetic Fitness , Genetic Variation , Geographic Information Systems , Hospitalization , Humans , Pandemics , Phylogeny , Pneumonia, Viral/epidemiology , Pneumonia, Viral/physiopathology , Respiratory System/virology , SARS-CoV-2 , Severity of Illness Index , Viral Load
2.
Proc Natl Acad Sci U S A ; 120(28): e2301934120, 2023 07 11.
Article in English | MEDLINE | ID: mdl-37399373

ABSTRACT

E3 ubiquitin ligase Mdm2 facilitates ß-arrestin ubiquitination, leading to the internalization of G protein-coupled receptors (GPCRs). In this process, ß-arrestins bind to Mdm2 and recruit it to the receptor; however, the molecular architecture of the ß-arrestin-Mdm2 complex has not been elucidated yet. Here, we identified the ß-arrestin-binding region (ABR) on Mdm2 and solved the crystal structure of ß-arrestin1 in complex with Mdm2ABR peptide. The acidic residues of Mdm2ABR bind to the positively charged concave side of the ß-arrestin1 N-domain. The C-tail of ß-arrestin1 is still bound to the N-domain, indicating that Mdm2 binds to the inactive state of ß-arrestin1, whereas the phosphorylated C-terminal tail of GPCRs binds to activate ß-arrestins. The overlapped binding site of Mdm2 and GPCR C-tails on ß-arrestin1 suggests that the binding of GPCR C-tails might trigger the release of Mdm2. Moreover, hydrogen/deuterium exchange experiments further show that Mdm2ABR binding to ß-arrestin1 induces the interdomain interface to be more dynamic and uncouples the IP6-induced oligomer of ß-arrestin1. These results show how the E3 ligase, Mdm2, interacts with ß-arrestins to promote the internalization of GPCRs.


Subject(s)
Arrestins , Ubiquitin-Protein Ligases , beta-Arrestins/metabolism , Ubiquitin-Protein Ligases/metabolism , Arrestins/metabolism , beta-Arrestin 1/metabolism , Ubiquitination , Receptors, G-Protein-Coupled/metabolism , beta-Arrestin 2/metabolism , Phosphorylation
3.
Nucleic Acids Res ; 50(15): 8929-8946, 2022 08 26.
Article in English | MEDLINE | ID: mdl-35920317

ABSTRACT

Although both the p53 and forkhead box (FOX) family proteins are key transcription factors associated with cancer progression, their direct relationship is unknown. Here, we found that FOX family proteins bind to the non-canonical homotypic cluster of the p53 promoter region (TP53). Analysis of crystal structures of FOX proteins (FOXL2 and FOXA1) bound to the p53 homotypic cluster indicated that they interact with a 2:1 stoichiometry accommodated by FOX-induced DNA allostery. In particular, FOX proteins exhibited distinct dimerization patterns in recognition of the same p53-DNA; dimer formation of FOXA1 involved protein-protein interaction, but FOXL2 did not. Biochemical and biological functional analyses confirmed the cooperative binding of FOX proteins to the TP53 promoter for the transcriptional activation of TP53. In addition, up-regulation of TP53 was necessary for FOX proteins to exhibit anti-proliferative activity in cancer cells. These analyses reveal the presence of a discrete characteristic within FOX family proteins in which FOX proteins regulate the transcription activity of the p53 tumor suppressor via cooperative binding to the TP53 promoter in alternative dimer configurations.


Subject(s)
Forkhead Box Protein L2/metabolism , Forkhead Transcription Factors , Hepatocyte Nuclear Factor 3-alpha/metabolism , Tumor Suppressor Protein p53/genetics , Forkhead Transcription Factors/metabolism , Humans , Promoter Regions, Genetic , Tumor Suppressor Protein p53/metabolism
4.
Nucleic Acids Res ; 50(4): 2319-2333, 2022 02 28.
Article in English | MEDLINE | ID: mdl-35141752

ABSTRACT

Staphylococcus aureus is a notorious and globally distributed pathogenic bacterium. New strategies to develop novel antibiotics based on intrinsic bacterial toxin-antitoxin (TA) systems have been recently reported. Because TA systems are present only in bacteria and not in humans, these distinctive systems are attractive targets for developing antibiotics with new modes of action. S. aureus PemIK is a type II TA system, comprising the toxin protein PemK and the labile antitoxin protein PemI. Here, we determined the crystal structures of both PemK and the PemIK complex, in which PemK is neutralized by PemI. Our biochemical approaches, including fluorescence quenching and polarization assays, identified Glu20, Arg25, Thr48, Thr49, and Arg84 of PemK as being important for RNase function. Our study indicates that the active site and RNA-binding residues of PemK are covered by PemI, leading to unique conformational changes in PemK accompanied by repositioning of the loop between ß1 and ß2. These changes can interfere with RNA binding by PemK. Overall, PemK adopts particular open and closed forms for precise neutralization by PemI. This structural and functional information on PemIK will contribute to the discovery and development of novel antibiotics in the form of peptides or small molecules inhibiting direct binding between PemI and PemK.


Subject(s)
Antitoxins , Staphylococcus aureus , Anti-Bacterial Agents/metabolism , Antitoxins/genetics , Antitoxins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , RNA/metabolism , Staphylococcus aureus/genetics , Staphylococcus aureus/metabolism
5.
Int J Mol Sci ; 25(11)2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38892403

ABSTRACT

Bakanae disease (BD), caused by the fungal pathogen Fusarium fujikuroi, is a serious threat to rice production worldwide. Breeding elite rice varieties resistant to BD requires the identification of resistance genes. Previously, we discovered a resistant quantitative trait locus (QTL), qFfR1, in a Korean japonica rice variety, Nampyeong. In this study, we fine-mapped qFfR1 with a Junam*4/Nampyeong BC3F3 population and delimited its location to a 37.1 kb region on chromosome 1. Complementation experiments with seven candidate genes in this region revealed that OsI_02728 is the gene for qFfR1. This gene encodes a protein with a typical leucine-rich repeat (LRR) receptor-like protein structure. RNA-sequencing-based transcriptomic analysis revealed that FfR1 induces the transcription of defense genes, including lignin and terpenoid biosynthesis genes, pathogenesis-related genes, and thionin genes. These results may facilitate investigations into the molecular mechanisms underlying BD resistance, including molecular patterns of Fusarium fujikuroi interacting with FfR1 and players working in signal transduction pathways downstream of FfR1, and the breeding of new BD-resistant varieties by providing a BD resistance gene with its precise selection marker. This will contribute to efficient control of BD, which is becoming more prevalent according to temperature rises due to climate change.


Subject(s)
Chromosome Mapping , Disease Resistance , Fusarium , Oryza , Plant Diseases , Quantitative Trait Loci , Oryza/genetics , Oryza/microbiology , Disease Resistance/genetics , Plant Diseases/microbiology , Plant Diseases/genetics , Fusarium/pathogenicity , Cloning, Molecular , Plant Proteins/genetics , Plant Proteins/metabolism , Gene Expression Regulation, Plant , Chromosomes, Plant/genetics
6.
Emerg Infect Dis ; 29(5)2023 05.
Article in English | MEDLINE | ID: mdl-37054986

ABSTRACT

Since late 2020, SARS-CoV-2 variants have regularly emerged with competitive and phenotypic differences from previously circulating strains, sometimes with the potential to escape from immunity produced by prior exposure and infection. The Early Detection group is one of the constituent groups of the US National Institutes of Health National Institute of Allergy and Infectious Diseases SARS-CoV-2 Assessment of Viral Evolution program. The group uses bioinformatic methods to monitor the emergence, spread, and potential phenotypic properties of emerging and circulating strains to identify the most relevant variants for experimental groups within the program to phenotypically characterize. Since April 2021, the group has prioritized variants monthly. Prioritization successes include rapidly identifying most major variants of SARS-CoV-2 and providing experimental groups within the National Institutes of Health program easy access to regularly updated information on the recent evolution and epidemiology of SARS-CoV-2 that can be used to guide phenotypic investigations.


Subject(s)
COVID-19 , SARS-CoV-2 , United States/epidemiology , Humans , SARS-CoV-2/genetics , COVID-19/epidemiology , National Institutes of Health (U.S.)
7.
EMBO Rep ; 22(6): e51323, 2021 06 04.
Article in English | MEDLINE | ID: mdl-33938112

ABSTRACT

In eukaryotic cells, mitochondria are closely tethered to the endoplasmic reticulum (ER) at sites called mitochondria-associated ER membranes (MAMs). Ca2+ ion and phospholipid transfer occurs at MAMs to support diverse cellular functions. Unlike those in yeast, the protein complexes involved in phospholipid transfer at MAMs in humans have not been identified. Here, we determine the crystal structure of the tetratricopeptide repeat domain of PTPIP51 (PTPIP51_TPR), a mitochondrial protein that interacts with the ER-anchored VAPB protein at MAMs. The structure of PTPIP51_TPR shows an archetypal TPR fold, and an electron density map corresponding to an unidentified lipid-like molecule probably derived from the protein expression host is found in the structure. We reveal functions of PTPIP51 in phospholipid binding/transfer, particularly of phosphatidic acid, in vitro. Depletion of PTPIP51 in cells reduces the mitochondrial cardiolipin level. Additionally, we confirm that the PTPIP51-VAPB interaction is mediated by the FFAT-like motif of PTPIP51 and the MSP domain of VAPB. Our findings suggest that PTPIP51 is a phospholipid transfer protein with a MAM-tethering function.


Subject(s)
Calcium , Phospholipids , Calcium/metabolism , Endoplasmic Reticulum/metabolism , Humans , Mitochondria/genetics , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Phospholipids/metabolism , Protein Tyrosine Phosphatases
8.
Cell Mol Life Sci ; 78(1): 207-225, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32140747

ABSTRACT

NAD(P)-dependent steroid dehydrogenase-like (NSDHL), an essential enzyme in human cholesterol synthesis and a regulator of epidermal growth factor receptor (EGFR) trafficking pathways, has attracted interest as a therapeutic target due to its crucial relevance to cholesterol-related diseases and carcinomas. However, the development of pharmacological agents for targeting NSDHL has been hindered by the absence of the atomic details of NSDHL. In this study, we reported two X-ray crystal structures of human NSDHL, which revealed a detailed description of the coenzyme-binding site and the unique conformational change upon the binding of a coenzyme. A structure-based virtual screening and biochemical evaluation were performed and identified a novel inhibitor for NSDHL harboring suppressive activity towards EGFR. In EGFR-driven human cancer cells, treatment with the potent NSDHL inhibitor enhanced the antitumor effect of an EGFR kinase inhibitor. Overall, these findings could serve as good platforms for the development of therapeutic agents against NSDHL-related diseases.


Subject(s)
3-Hydroxysteroid Dehydrogenases/metabolism , Enzyme Inhibitors/metabolism , 3-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , 3-Hydroxysteroid Dehydrogenases/chemistry , 3-Hydroxysteroid Dehydrogenases/genetics , Binding Sites , Cell Line, Tumor , Cell Survival/drug effects , Cholesterol/chemistry , Crystallography, X-Ray , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Erlotinib Hydrochloride/chemistry , Erlotinib Hydrochloride/metabolism , Erlotinib Hydrochloride/pharmacology , Humans , Kinetics , Molecular Docking Simulation , Mutagenesis, Site-Directed , NAD/chemistry , NAD/metabolism , Protein Structure, Tertiary , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Signal Transduction
9.
J Neurochem ; 156(6): 967-978, 2021 03.
Article in English | MEDLINE | ID: mdl-32880929

ABSTRACT

The lysosomal membrane protein Niemann-Pick type C1 (NPC1) and Niemann-Pick type C2 (NPC2) are main players of cholesterol control in the lysosome and it is known that the mutation on these proteins leads to the cholesterol trafficking-related neurodegenerative disease, which is called the NPC disease. The mutation R518W or R518Q on the NPC1 is one of the type of disease-related mutation that causes cholesterol transports to be cut in half, which results in the accumulation of cholesterol and lipids in the late endosomal/lysosomal compartment of the cell. Even though there has been significant progress with understanding the cholesterol transport by NPC1 in combination with NPC2, especially after the structural determination of the full-length NPC1 in 2016, many details such as the interaction of the full-length NPC1 with the NPC2, the molecular motions responsible for the cholesterol transport during and after this interaction, and the structure and the function relations of many mutations are still not well understood. In this study, we report the extensive molecular dynamics simulations in order to gain insight into the structure and the dynamics of NPC1 lumenal domain for the cholesterol transport and the disease behind the mutation (R518W). It was found that the mutation induces a structural shift of the N-terminal domain, toward the loop region in the middle lumenal domain, which is believed to play a central role in the interaction with NPC2 protein, so the interaction with the NPC2 protein might be less favorable compared to the wild NPC1. Also, the simulation indicates the possible re-orientation of the N-terminal domain with both the wild and the R518W-mutated NPC1 after receiving the cholesterol from the NPC2 that align to form an internal tunnel, which is a possible pose for further action in cholesterol trafficking. We believe the current study can provide a better understanding of the cholesterol transport by NPC1 especially the role of NTD of NPC1 in combination with NPC2 interactions.


Subject(s)
Cholesterol/metabolism , Niemann-Pick Disease, Type C/genetics , Biological Transport , Endosomes/metabolism , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Lipid Metabolism/genetics , Lysosomes/metabolism , Models, Molecular , Molecular Dynamics Simulation , Mutation/genetics , Niemann-Pick C1 Protein , Vesicular Transport Proteins/chemistry , Vesicular Transport Proteins/genetics , X-Ray Diffraction
10.
FASEB J ; 34(2): 3051-3068, 2020 02.
Article in English | MEDLINE | ID: mdl-31908032

ABSTRACT

Bacterial toxin-antitoxin (TA) system has gained attention for its essential roles in cellular maintenance and survival under harsh environmental conditions such as nutrient deficiency and antibiotic treatment. There are at least 14 TA systems in Salmonella enterica serovar Typhimurium LT2, a pathogenic bacterium, and none of the structures of these TA systems have been determined. We determined the crystal structure of the VapBC TA complex from S. Typhimurium LT2 in proteolyzed and DNA-bound forms at 2.0 Å and 2.8 Å resolution, respectively. The VapC toxin possesses a pilT N-terminal domain (PIN-domain) that shows ribonuclease activity, and the VapB antitoxin has an AbrB-type DNA binding domain. In addition, the structure revealed details of interaction mode between VapBC and the cognate promoter DNA, including the inhibition of VapC by VapB and linear conformation of bound DNA in the VapBC complex. The complexation of VapBC with the linear DNA is not consistent with known structures of VapBC homologs in complex with bent DNA. We also identified VapC from S. Typhimurium LT2 as a putative Ca2+ -dependent ribonuclease, which differs from previous data showing that VapC homologs have Mg2+ or Mn2+ -dependent ribonuclease activities. The present studies could provide structural understanding of the physiology of VapBC systems and foundation for the development of new antibiotic drugs against Salmonella infection.


Subject(s)
Bacterial Proteins/chemistry , DNA, Bacterial/chemistry , Ribonucleases/chemistry , Salmonella typhimurium/enzymology , Crystallography, X-Ray , Protein Domains , Protein Structure, Quaternary
11.
Biochem Biophys Res Commun ; 522(1): 240-246, 2020 01 29.
Article in English | MEDLINE | ID: mdl-31759628

ABSTRACT

Fenestrations in choriocapillaris act as a window for molecular transports between the retina and choroid, and is crucial for maintaining visual function. Plasmalemma vesicle-associated protein (PLVAP) is essential for the development of endothelial fenestrations. There is little knowledge about how the choriocapillaris maintains the fenestrated endothelium. This study aimed to evaluate the role of vascular endothelial growth factor-A (VEGFA)-PLVAP axis in the maintenance of choroidal fenestrations using oxygen-induced retinopathy (OIR) model. In C57BL/6 J mice, the mice with OIR on postnatal day 12 (P12) presented thicker endothelium and less fenestration compared to the non-OIR mice. However, the OIR on 17 mice showed thinner endothelium with more fenestration compared to OIR on P12. In vivo angiography demonstrated the presence of hyperpermeable choroidal vessels on P17 in OIR mice. These dramatic changes in choriocapillaris were not observed in the BALB/cJ OIR model. The ultrastructural changes in the choriocapillaris were correlated with temporal variations in the expression of VEGFA and PLVAP. VEGFA stimulated expression of PLVAP in the choroidal endothelial cells. Loss of PLVAP disrupts the polarized structure of the choriocapillaris leading to retinal degeneration. These results indicate that the expression of retinal VEGFA is essential for maintaining the structure and function of choriocapillaris by preserving the endothelial PLVAP.


Subject(s)
Choroid/metabolism , Membrane Proteins/metabolism , Retina/metabolism , Retinal Diseases/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Cells, Cultured , Choroid/pathology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Humans , Mice, Inbred BALB C , Mice, Inbred C57BL , Oxygen/metabolism , Retina/pathology , Retinal Diseases/pathology
12.
Nucleic Acids Res ; 46(12): 6371-6386, 2018 07 06.
Article in English | MEDLINE | ID: mdl-29878152

ABSTRACT

Streptococcus pneumonia has attracted increasing attention due to its resistance to existing antibiotics. TA systems are essential for bacterial persistence under stressful conditions such as nutrient deprivation, antibiotic treatment, and immune system attacks. In particular, S. pneumoniae expresses the HicBA TA gene, which encodes the stable HicA toxin and the labile HicB antitoxin. These proteins interact to form a non-toxic TA complex under normal conditions, but the toxin is activated by release from the antitoxin in response to unfavorable growth conditions. Here, we present the first crystal structure showing the complete conformation of the HicBA complex from S. pneumonia. The structure reveals that the HicA toxin contains a double-stranded RNA-binding domain that is essential for RNA recognition and that the C-terminus of the HicB antitoxin folds into a ribbon-helix-helix DNA-binding motif. The active site of HicA is sterically blocked by the N-terminal region of HicB. RNase activity assays show that His36 is essential for the ribonuclease activity of HicA, and nuclear magnetic resonance (NMR) spectra show that several residues of HicB participate in binding to the promoter DNA of the HicBA operon. A toxin-mimicking peptide that inhibits TA complex formation and thereby increases toxin activity was designed, providing a novel approach to the development of new antibiotics.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Toxins/chemistry , Streptococcus pneumoniae , Toxin-Antitoxin Systems , Bacterial Proteins/metabolism , Bacterial Proteins/toxicity , Bacterial Toxins/metabolism , Bacterial Toxins/toxicity , Catalytic Domain , Models, Molecular , Promoter Regions, Genetic , Protein Domains , Ribonucleases/chemistry , Ribonucleases/metabolism
13.
J Cell Physiol ; 234(7): 11490-11499, 2019 07.
Article in English | MEDLINE | ID: mdl-30478996

ABSTRACT

G protein-coupled receptor 119 (GPR119) is known to be a promising therapeutic target for type 2 diabetes. Recently, it has been reported that the GPR119 agonist increases bone mineral density in an animal model of diabetes, suggesting that GPR119 may play a key role in bone metabolism. In this study, we investigated the functional role of GPR119 in receptor activator of nuclear factor-κB ligand (RANKL)-induced osteoclast formation. We found that the GPR119 expression was markedly increased in preosteoclasts and then downregulated in mature osteoclasts. Activation of GPR119 with AS1269574, a potent selective agonist for GPR119, inhibited the generation of multinuclear osteoclasts from bone marrow-derived macrophages. Confirming this observation, targeted silencing of GPR119 using short hairpin RNA abrogated the AS1269574-mediated suppressive effect on osteoclast formation. GPR119 activation attenuated the expression of c-Fos and nuclear factor of activated T cells cytoplasmic 1 (NFATc1) and blocked RANKL-stimulated phosphorylation of IκBα, c-Jun N-terminal protein kinase (JNK), and extracellular signal-regulated kinase (ERK) but not p38. In addition, GPR119 activation suppressed preosteoclast fusion by downregulating the expression of the dendritic cell-specific transmembrane (DC-STAMP), a molecule that is essential for cell-cell fusion in osteoclast formation. Furthermore, ectopic expression of DC-STAMP restored AS1269574-mediated inhibition of osteoclast fusion. Taken together, our findings demonstrate that GPR119 plays a negative role in osteoclast differentiation and fusion induced by RANKL, and therefore may represent a potential target for bone resorption-associated diseases.


Subject(s)
Gene Expression Regulation/drug effects , Osteoclasts/physiology , RANK Ligand/pharmacology , Receptors, G-Protein-Coupled/metabolism , Animals , Cell Differentiation , Cell Fusion , Cell Survival , Dimethyl Sulfoxide/pharmacology , Ethanolamines/pharmacology , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Silencing , JNK Mitogen-Activated Protein Kinases/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Macrophage Colony-Stimulating Factor/pharmacology , Mice , NF-KappaB Inhibitor alpha/genetics , NF-KappaB Inhibitor alpha/metabolism , Pyrimidines/pharmacology , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/genetics
14.
J Cell Physiol ; 234(2): 1659-1670, 2019 02.
Article in English | MEDLINE | ID: mdl-30076703

ABSTRACT

Estrogen-related receptor γ (ERRγ) is an orphan nuclear receptor that plays an important role in various metabolic processes under physiological and pathophysiological conditions. Here, we report that ERRγ functions as a negative regulator in receptor activator of nuclear factor κΒ ligand (RANKL)-induced osteoclast differentiation. We observed that ERRγ was strongly expressed in osteoclast precursors, bone marrow-derived macrophages (BMMs) while its expression was significantly reduced by RANKL during osteoclastogenesis. Overexpression of ERRγ in BMMs suppressed the formation of multinucleated osteoclasts and attenuated the induction of c-Fos and nuclear factor of activated T cells c1, which are critical modulators in osteoclastogenesis. Similarly, the treatment of ERRγ agonists, N-(4-(diethylaminobenzylidenyl)-N'-(4-hydroxybenzoyl)-hydrazine (DY131) or GSK4716, also inhibited osteoclast generation and the expression of these key modulators. On the other hand, shRNA-mediated knockdown of ERRγ accelerated the formation of bone-resorbing cells and the expression of osteoclastogenic markers. Forced expression of ERRγ blocked RANKL-stimulated phosphorylation of the nuclear factor κB (NF-κB) inhibitor IκBα and suppressed NF-κB transcriptional activity induced by RANKL or the NF-κB subunit p65. Furthermore, by employing a pharmacological approach, we showed that the ERRγ agonist DY131 protected against inflammatory bone loss induced by lipopolysaccharide in vivo. Together, our findings reveal that ERRγ is a pivotal regulator in RANKL-mediated osteoclastogenesis and suggest that ERRγ may have potential as a therapeutic target for pathological bone loss.


Subject(s)
Macrophages/metabolism , Osteoclasts/metabolism , Osteogenesis , Osteoporosis/metabolism , Receptors, Estrogen/metabolism , Animals , Cell Differentiation , Disease Models, Animal , Estrogens/pharmacology , Gene Expression Regulation , Macrophages/drug effects , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , NFATC Transcription Factors/metabolism , Osteoclasts/drug effects , Osteoclasts/pathology , Osteogenesis/drug effects , Osteoporosis/genetics , Osteoporosis/pathology , Osteoporosis/prevention & control , RANK Ligand/pharmacology , RAW 264.7 Cells , Receptors, Estrogen/drug effects , Receptors, Estrogen/genetics , Signal Transduction
15.
Biochem Biophys Res Commun ; 518(1): 114-119, 2019 10 08.
Article in English | MEDLINE | ID: mdl-31420168

ABSTRACT

C-di-GMP, bis-(3'-5')-cyclic dimeric guanosine monophosphate, is a key signaling molecule that regulates many important physiological processes in bacteria. C-di-GMP is synthesized by diguanylate cyclase (DGC) containing the homodimeric GGDEF domain. There are many uncharacterized hypothetical proteins annotated as a putative DGC in bacteria including hyperthermophiles; however, their structures still remain unexplored. Here, we solved the crystal structure of the GGDEF-like domain of Tm0107 protein from Thermotoga maritima at a resolution of 2.1 Å, which shares sequence similarities with DGC proteins in other bacteria. Tm0107 consists of an N-terminal coiled-coil and C-terminal GGDEF-like domain. We showed that the GGDEF-like domain of Tm0107 exists as monomer in solution and is structurally similar to other GGDEF domains. Two zinc ions are coordinated at the interface between two Tm0107 monomers. Based on our measurements of the Stokes radii of Tm0107 by analytical gel filtration, we propose a dimer model of Tm0107 containing both the N-terminal coiled coil and C-terminal GGDEF-like domains. Based on the model, Tm0107 forms a homodimer in a manner different compared to other structurally characterized DGC proteins. These results provide useful structural information about putative DGC proteins containing protein sequences similar to that of Tm0107, which is widely conserved in hyperthermophiles.


Subject(s)
Conserved Sequence , Escherichia coli Proteins/chemistry , Phosphorus-Oxygen Lyases/chemistry , Temperature , Thermotoga maritima/enzymology , Amino Acid Sequence , Escherichia coli Proteins/metabolism , Ions , Models, Molecular , Phosphorus-Oxygen Lyases/metabolism , Protein Domains , Protein Multimerization , Structural Homology, Protein , Zinc/metabolism
16.
Nucleic Acids Res ; 45(14): 8564-8580, 2017 Aug 21.
Article in English | MEDLINE | ID: mdl-28575388

ABSTRACT

Toxin-antitoxin (TA) systems are essential for bacterial persistence under stressful conditions. In particular, Mycobacterium tuberculosis express VapBC TA genes that encode the stable VapC toxin and the labile VapB antitoxin. Under normal conditions, these proteins interact to form a non-toxic TA complex, but the toxin is activated by release from the antitoxin in response to unfavorable conditions. Here, we present the crystal structure of the M. tuberculosis VapBC26 complex and show that the VapC26 toxin contains a pilus retraction protein (PilT) N-terminal (PIN) domain that is essential for ribonuclease activity and that, the VapB26 antitoxin folds into a ribbon-helix-helix DNA-binding motif at the N-terminus. The active site of VapC26 is sterically blocked by the flexible C-terminal region of VapB26. The C-terminal region of free VapB26 adopts an unfolded conformation but forms a helix upon binding to VapC26. The results of RNase activity assays show that Mg2+ and Mn2+ are essential for the ribonuclease activity of VapC26. As shown in the nuclear magnetic resonance spectra, several residues of VapB26 participate in the specific binding to the promoter region of the VapBC26 operon. In addition, toxin-mimicking peptides were designed that inhibit TA complex formation and thereby increase toxin activity, providing a novel approach to the development of new antibiotics.


Subject(s)
Antimicrobial Cationic Peptides/metabolism , Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , DNA-Binding Proteins/metabolism , Membrane Glycoproteins/metabolism , Mycobacterium tuberculosis/metabolism , Amino Acid Sequence , Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/pharmacology , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Toxins/chemistry , Bacterial Toxins/genetics , Calorimetry/methods , Crystallography, X-Ray , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Hydrophobic and Hydrophilic Interactions , Magnesium/chemistry , Magnesium/metabolism , Magnesium/pharmacology , Manganese/chemistry , Manganese/metabolism , Manganese/pharmacology , Mass Spectrometry/methods , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/genetics , Models, Molecular , Mutation , Mycobacterium tuberculosis/genetics , Protein Binding/drug effects , Protein Multimerization , Protein Structure, Secondary , Protein Structure, Tertiary , Ribonucleases/chemistry , Ribonucleases/genetics , Ribonucleases/metabolism , Sequence Homology, Amino Acid
17.
Proc Natl Acad Sci U S A ; 113(35): E5202-11, 2016 08 30.
Article in English | MEDLINE | ID: mdl-27531959

ABSTRACT

For bacteria, cysteine thiol groups in proteins are commonly used as thiol-based switches for redox sensing to activate specific detoxification pathways and restore the redox balance. Among the known thiol-based regulatory systems, the MarR/DUF24 family regulators have been reported to sense and respond to reactive electrophilic species, including diamide, quinones, and aldehydes, with high specificity. Here, we report that the prototypical regulator YodB of the MarR/DUF24 family from Bacillus subtilis uses two distinct pathways to regulate transcription in response to two reactive electrophilic species (diamide or methyl-p-benzoquinone), as revealed by X-ray crystallography, NMR spectroscopy, and biochemical experiments. Diamide induces structural changes in the YodB dimer by promoting the formation of disulfide bonds, whereas methyl-p-benzoquinone allows the YodB dimer to be dissociated from DNA, with little effect on the YodB dimer. The results indicate that B. subtilis may discriminate toxic quinones, such as methyl-p-benzoquinone, from diamide to efficiently manage multiple oxidative signals. These results also provide evidence that different thiol-reactive compounds induce dissimilar conformational changes in the regulator to trigger the separate regulation of target DNA. This specific control of YodB is dependent upon the type of thiol-reactive compound present, is linked to its direct transcriptional activity, and is important for the survival of B. subtilis This study of B. subtilis YodB also provides a structural basis for the relationship that exists between the ligand-induced conformational changes adopted by the protein and its functional switch.


Subject(s)
Bacillus subtilis/genetics , Bacterial Proteins/genetics , Gene Expression Regulation, Bacterial , Signal Transduction/genetics , Bacillus subtilis/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Benzoquinones/chemistry , Benzoquinones/pharmacology , Crystallography, X-Ray , Diamide/chemistry , Diamide/pharmacology , Oxidation-Reduction , Protein Conformation/drug effects , Protein Multimerization/drug effects
18.
Sensors (Basel) ; 19(22)2019 Nov 19.
Article in English | MEDLINE | ID: mdl-31752423

ABSTRACT

Slip-forming in concrete construction enables the continuous placement of concrete using a climbing form, the efficiency of which depends on appropriate slip-up timing. This implies the importance of knowing accurately the development of concrete strength over time, which has been assessed manually to date in construction fields. This paper presents a method for automating the slip-forming process by determining the optimal slip-up time using the in-situ strength of concrete. The strength of concrete is evaluated by a formula relating the strength to the surface wave velocity measured with ultrasonic sensors. Specifically, this study validates the applicability of the slip-form system with ultrasonic sensors for continuously monitoring the hardening of concrete through its application in several construction sites. To this end, a slip-form system with a pair of ultrasonic modules at the bottom of the panel was tested and the time variation of surface wave velocity in the concrete material was monitored during the slip-forming process. The results show that the proposed method can provide the optimal slip-up time of the form to automate the slip-forming process. This approach is expected to apply to other construction technologies that required the continuous monitoring of concrete strength for construction efficiency as well as quality maintenance.

19.
J Biol Chem ; 292(46): 18832-18847, 2017 11 17.
Article in English | MEDLINE | ID: mdl-28972145

ABSTRACT

The bacterial toxin-antitoxin MazEF system in the tuberculosis (TB)-causing bacterium Mycobacterium tuberculosis is activated under unfavorable conditions, including starvation, antibiotic exposure, and oxidative stress. This system contains the ribonucleolytic enzyme MazF and has emerged as a promising drug target for TB treatments targeting the latent stage of M. tuberculosis infection and reportedly mediates a cell death process via a peptide called extracellular death factor (EDF). Although it is well established that the increase in EDF-mediated toxicity of MazF drives a cell-killing phenomenon, the molecular details are poorly understood. Moreover, the divergence in sequences among reported EDFs suggests that each bacterial species has a unique EDF. To address these open questions, we report here the structures of MazF4 and MazEF4 complexes from M. tuberculosis, representing the first MazEF structures from this organism. We found that MazF4 possesses a negatively charged MazE4-binding pocket in contrast to the positively charged MazE-binding pockets in homologous MazEF complex structures from other bacteria. Moreover, using NMR spectroscopy and biochemical assays, we unraveled the molecular interactions of MazF4 with its RNA substrate and with a new EDF homolog originating from M. tuberculosis The EDF homolog discovered here possesses a positively charged residue at the C terminus, making this EDF distinct from previously reported EDFs. Overall, our results suggest that M. tuberculosis evolved a unique MazF and EDF and that the distinctive EDF sequence could serve as a starting point for designing new anti-tuberculosis drugs. We therefore conclude that this study might contribute to the development of a new line of anti-tuberculosis agents.


Subject(s)
Antitoxins/metabolism , Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Endoribonucleases/metabolism , Mycobacterium tuberculosis/metabolism , Peptides/metabolism , Amino Acid Sequence , Antitoxins/chemistry , Bacterial Proteins/chemistry , Bacterial Toxins/chemistry , Crystallography, X-Ray , Drug Discovery , Endoribonucleases/chemistry , Humans , Models, Molecular , Mycobacterium tuberculosis/chemistry , Mycobacterium tuberculosis/physiology , Peptides/chemistry , Protein Conformation , Protein Multimerization , Quorum Sensing , Sequence Alignment , Tuberculosis/microbiology
20.
J Cell Physiol ; 233(2): 1481-1489, 2018 Feb.
Article in English | MEDLINE | ID: mdl-28574596

ABSTRACT

GPR84, a member of the G protein-coupled receptor family, is found predominantly in immune cells, such as macrophages, and functions as a pivotal modulator of inflammatory responses. In this study, we investigated the role of GPR84 in receptor activator of nuclear factor-κB ligand (RANKL)-induced osteoclast differentiation. Our microarray data showed that GPR84 was significantly downregulated in osteoclasts compared to in their precursors, macrophages. The overexpression of GPR84 in bone marrow-derived macrophages suppressed the formation of multinucleated osteoclasts without affecting precursor proliferation. In addition, GPR84 overexpression attenuated the induction of c-Fos and nuclear factor of activated T cells, cytoplasmic 1 (NFATc1), which are transcription factors that are critical for osteoclastogenesis. Furthermore, knockdown of GPR84 using a small hairpin RNA promoted RANKL-mediated osteoclast differentiation and gene expression of osteoclastogenic markers. Mechanistically, GPR84 overexpression blocked RANKL-stimulated phosphorylation of IκBα and three MAPKs, JNK, ERK, and p38. GPR84 also suppressed NF-κB transcriptional activity mediated by RANKL. Conversely, GPR84 knockdown enhanced RANKL-induced activation of IκBα and the three MAPKs. Collectively, our results revealed that GPR84 functions as a negative regulator of osteoclastogenesis, suggesting that it may be a potential therapeutic target for osteoclast-mediated bone-destructive diseases.


Subject(s)
Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Osteoclasts/enzymology , Osteogenesis , Receptors, G-Protein-Coupled/metabolism , Animals , Cell Differentiation , Gene Expression Regulation , Mice , Mice, Inbred C57BL , NF-KappaB Inhibitor alpha/genetics , NF-KappaB Inhibitor alpha/metabolism , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Phosphorylation , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , RANK Ligand/genetics , RANK Ligand/metabolism , RAW 264.7 Cells , RNA Interference , Receptors, G-Protein-Coupled/genetics , Signal Transduction , Time Factors , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL