Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 553
Filter
Add more filters

Publication year range
1.
Cell ; 179(5): 1160-1176.e24, 2019 11 14.
Article in English | MEDLINE | ID: mdl-31730855

ABSTRACT

Pediatric-onset colitis and inflammatory bowel disease (IBD) have significant effects on the growth of infants and children, but the etiopathogenesis underlying disease subtypes remains incompletely understood. Here, we report single-cell clustering, immune phenotyping, and risk gene analysis for children with undifferentiated colitis, Crohn's disease, and ulcerative colitis. We demonstrate disease-specific characteristics, as well as common pathogenesis marked by impaired cyclic AMP (cAMP)-response signaling. Specifically, infiltration of PDE4B- and TNF-expressing macrophages, decreased abundance of CD39-expressing intraepithelial T cells, and platelet aggregation and release of 5-hydroxytryptamine at the colonic mucosae were common in colitis and IBD patients. Targeting these pathways by using the phosphodiesterase inhibitor dipyridamole restored immune homeostasis and improved colitis symptoms in a pilot study. In summary, comprehensive analysis of the colonic mucosae has uncovered common pathogenesis and therapeutic targets for children with colitis and IBD.


Subject(s)
Inflammatory Bowel Diseases/pathology , Inflammatory Bowel Diseases/therapy , Intestinal Mucosa/pathology , Antigens, CD/metabolism , Apyrase/metabolism , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Cell Death/drug effects , Cellular Microenvironment/drug effects , Child , Cohort Studies , Colon/pathology , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Dipyridamole/pharmacology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression Regulation/drug effects , Genetic Predisposition to Disease , Homeostasis/drug effects , Humans , Immunoglobulin G/blood , Immunologic Memory , Inflammation/pathology , Inflammatory Bowel Diseases/blood , Inflammatory Bowel Diseases/genetics , Interferon Type I/metabolism , Macrophages/drug effects , Macrophages/metabolism , Methylprednisolone/pharmacology , Myeloid Cells/drug effects , Myeloid Cells/metabolism
2.
Cell ; 173(7): 1636-1649.e16, 2018 06 14.
Article in English | MEDLINE | ID: mdl-29754813

ABSTRACT

Hydrogen gas-evolving membrane-bound hydrogenase (MBH) and quinone-reducing complex I are homologous respiratory complexes with a common ancestor, but a structural basis for their evolutionary relationship is lacking. Here, we report the cryo-EM structure of a 14-subunit MBH from the hyperthermophile Pyrococcus furiosus. MBH contains a membrane-anchored hydrogenase module that is highly similar structurally to the quinone-binding Q-module of complex I while its membrane-embedded ion-translocation module can be divided into a H+- and a Na+-translocating unit. The H+-translocating unit is rotated 180° in-membrane with respect to its counterpart in complex I, leading to distinctive architectures for the two respiratory systems despite their largely conserved proton-pumping mechanisms. The Na+-translocating unit, absent in complex I, resembles that found in the Mrp H+/Na+ antiporter and enables hydrogen gas evolution by MBH to establish a Na+ gradient for ATP synthesis near 100°C. MBH also provides insights into Mrp structure and evolution of MBH-based respiratory enzymes.


Subject(s)
Archaeal Proteins/metabolism , Hydrogenase/metabolism , Pyrococcus furiosus/metabolism , Amino Acid Sequence , Archaeal Proteins/chemistry , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Binding Sites , Cell Membrane/chemistry , Cell Membrane/metabolism , Cryoelectron Microscopy , Electron Transport Complex I/chemistry , Electron Transport Complex I/metabolism , Evolution, Molecular , Hydrogen/metabolism , Hydrogenase/chemistry , Hydrogenase/genetics , Mutagenesis , Protein Structure, Quaternary , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Sequence Alignment , Sodium/chemistry , Sodium/metabolism , Sodium-Hydrogen Exchangers/chemistry , Sodium-Hydrogen Exchangers/metabolism
3.
Cell ; 158(4): 705-721, 2014 Aug 14.
Article in English | MEDLINE | ID: mdl-25126780

ABSTRACT

Acquisition of the intestinal microbiota begins at birth, and a stable microbial community develops from a succession of key organisms. Disruption of the microbiota during maturation by low-dose antibiotic exposure can alter host metabolism and adiposity. We now show that low-dose penicillin (LDP), delivered from birth, induces metabolic alterations and affects ileal expression of genes involved in immunity. LDP that is limited to early life transiently perturbs the microbiota, which is sufficient to induce sustained effects on body composition, indicating that microbiota interactions in infancy may be critical determinants of long-term host metabolic effects. In addition, LDP enhances the effect of high-fat diet induced obesity. The growth promotion phenotype is transferrable to germ-free hosts by LDP-selected microbiota, showing that the altered microbiota, not antibiotics per se, play a causal role. These studies characterize important variables in early-life microbe-host metabolic interaction and identify several taxa consistently linked with metabolic alterations. PAPERCLIP:


Subject(s)
Anti-Bacterial Agents/administration & dosage , Disease Models, Animal , Intestines/microbiology , Microbiota , Obesity/microbiology , Penicillins/administration & dosage , Animals , Bacteria/classification , Bacteria/metabolism , Female , Intestinal Mucosa/metabolism , Male , Mice , Mice, Inbred C57BL , Microbiota/drug effects , Obesity/metabolism
4.
EMBO J ; 43(8): 1634-1652, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38467832

ABSTRACT

During bacterial cell growth, hydrolases cleave peptide cross-links between strands of the peptidoglycan sacculus to allow new strand insertion. The Pseudomonas aeruginosa carboxyl-terminal processing protease (CTP) CtpA regulates some of these hydrolases by degrading them. CtpA assembles as an inactive hexamer composed of a trimer-of-dimers, but its lipoprotein binding partner LbcA activates CtpA by an unknown mechanism. Here, we report the cryo-EM structures of the CtpA-LbcA complex. LbcA has an N-terminal adaptor domain that binds to CtpA, and a C-terminal superhelical tetratricopeptide repeat domain. One LbcA molecule attaches to each of the three vertices of a CtpA hexamer. LbcA triggers relocation of the CtpA PDZ domain, remodeling of the substrate binding pocket, and realignment of the catalytic residues. Surprisingly, only one CtpA molecule in a CtpA dimer is activated upon LbcA binding. Also, a long loop from one CtpA dimer inserts into a neighboring dimer to facilitate the proteolytic activity. This work has revealed an activation mechanism for a bacterial CTP that is strikingly different from other CTPs that have been characterized structurally.


Subject(s)
Endopeptidases , Pseudomonas aeruginosa , Pseudomonas aeruginosa/metabolism , Endopeptidases/metabolism , Proteolysis
5.
Nature ; 604(7904): 127-133, 2022 04.
Article in English | MEDLINE | ID: mdl-35355010

ABSTRACT

Many aspects of plant photoperception are mediated by the phytochrome (Phy) family of bilin-containing photoreceptors that reversibly interconvert between inactive Pr and active Pfr conformers1,2. Despite extensive biochemical studies, full understanding of plant Phy signalling has remained unclear due to the absence of relevant 3D models. Here we report a cryo-electron microscopy structure of Arabidopsis PhyB in the Pr state that reveals a topologically complex dimeric organization that is substantially distinct from its prokaryotic relatives. Instead of an anticipated parallel architecture, the C-terminal histidine-kinase-related domains (HKRDs) associate head-to-head, whereas the N-terminal photosensory regions associate head-to-tail to form a parallelogram-shaped platform with near two-fold symmetry. The platform is internally linked by the second of two internal Per/Arnt/Sim domains that binds to the photosensory module of the opposing protomer and a preceding 'modulator' loop that assembles tightly with the photosensory module of its own protomer. Both connections accelerate the thermal reversion of Pfr back to Pr, consistent with an inverse relationship between dimer assembly and Pfr stability. Lopsided contacts between the HKRDs and the platform create profound asymmetry to PhyB that might imbue distinct signalling potentials to the protomers. We propose that this unique structural dynamism creates an extensive photostate-sensitive surface for conformation-dependent interactions between plant Phy photoreceptors and their signalling partners.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Phytochrome B , Arabidopsis/metabolism , Arabidopsis Proteins/chemistry , Arabidopsis Proteins/metabolism , Cryoelectron Microscopy , Light , Phytochrome B/chemistry , Phytochrome B/metabolism , Protein Domains , Protein Subunits/metabolism
6.
Nat Methods ; 2024 May 14.
Article in English | MEDLINE | ID: mdl-38744918

ABSTRACT

The combination of native electrospray ionization with top-down fragmentation in mass spectrometry (MS) allows simultaneous determination of the stoichiometry of noncovalent complexes and identification of their component proteoforms and cofactors. Although this approach is powerful, both native MS and top-down MS are not yet well standardized, and only a limited number of laboratories regularly carry out this type of research. To address this challenge, the Consortium for Top-Down Proteomics initiated a study to develop and test protocols for native MS combined with top-down fragmentation of proteins and protein complexes across 11 instruments in nine laboratories. Here we report the summary of the outcomes to provide robust benchmarks and a valuable entry point for the scientific community.

7.
Proc Natl Acad Sci U S A ; 121(18): e2319727121, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38669181

ABSTRACT

The DNA sliding clamp PCNA is a multipurpose platform for DNA polymerases and many other proteins involved in DNA metabolism. The topologically closed PCNA ring needs to be cracked open and loaded onto DNA by a clamp loader, e.g., the well-studied pentameric ATPase complex RFC (RFC1-5). The CTF18-RFC complex is an alternative clamp loader found recently to bind the leading strand DNA polymerase ε and load PCNA onto leading strand DNA, but its structure and the loading mechanism have been unknown. By cryo-EM analysis of in vitro assembled human CTF18-RFC-DNA-PCNA complex, we have captured seven loading intermediates, revealing a detailed PCNA loading mechanism onto a 3'-ss/dsDNA junction by CTF18-RFC. Interestingly, the alternative loader has evolved a highly mobile CTF18 AAA+ module likely to lower the loading activity, perhaps to avoid competition with the RFC and to limit its role to leading strand clamp loading. To compensate for the lost stability due to the mobile AAA+ module, CTF18 has evolved a unique ß-hairpin motif that reaches across RFC2 to interact with RFC5, thereby stabilizing the pentameric complex. Further, we found that CTF18 also contains a separation pin to locally melt DNA from the 3'-end of the primer; this ensures its ability to load PCNA to any 3'-ss/dsDNA junction, facilitated by the binding energy of the E-plug to the major groove. Our study reveals unique structural features of the human CTF18-RFC and contributes to a broader understanding of PCNA loading by the alternative clamp loaders.


Subject(s)
ATPases Associated with Diverse Cellular Activities , Cryoelectron Microscopy , Nuclear Proteins , Proliferating Cell Nuclear Antigen , Replication Protein C , Humans , Cryoelectron Microscopy/methods , DNA/metabolism , DNA/chemistry , DNA Replication , Models, Molecular , Proliferating Cell Nuclear Antigen/metabolism , Proliferating Cell Nuclear Antigen/chemistry , Protein Binding , Replication Protein C/metabolism , Replication Protein C/chemistry
8.
Nature ; 584(7821): 475-478, 2020 08.
Article in English | MEDLINE | ID: mdl-32494008

ABSTRACT

The endoplasmic reticulum (ER) membrane complex (EMC) cooperates with the Sec61 translocon to co-translationally insert a transmembrane helix (TMH) of many multi-pass integral membrane proteins into the ER membrane, and it is also responsible for inserting the TMH of some tail-anchored proteins1-3. How EMC accomplishes this feat has been unclear. Here we report the first, to our knowledge, cryo-electron microscopy structure of the eukaryotic EMC. We found that the Saccharomyces cerevisiae EMC contains eight subunits (Emc1-6, Emc7 and Emc10), has a large lumenal region and a smaller cytosolic region, and has a transmembrane region formed by Emc4, Emc5 and Emc6 plus the transmembrane domains of Emc1 and Emc3. We identified a five-TMH fold centred around Emc3 that resembles the prokaryotic YidC insertase and that delineates a largely hydrophilic client protein pocket. The transmembrane domain of Emc4 tilts away from the main transmembrane region of EMC and is partially mobile. Mutational studies demonstrated that the flexibility of Emc4 and the hydrophilicity of the client pocket are required for EMC function. The EMC structure reveals notable evolutionary conservation with the prokaryotic insertases4,5, suggests that eukaryotic TMH insertion involves a similar mechanism, and provides a framework for detailed understanding of membrane insertion for numerous eukaryotic integral membrane proteins and tail-anchored proteins.


Subject(s)
Cryoelectron Microscopy , Endoplasmic Reticulum/enzymology , Intracellular Membranes/enzymology , Multiprotein Complexes/chemistry , Multiprotein Complexes/ultrastructure , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/ultrastructure , Saccharomyces cerevisiae , Binding Sites , Endoplasmic Reticulum/chemistry , Endoplasmic Reticulum/ultrastructure , Evolution, Molecular , Hydrophobic and Hydrophilic Interactions , Intracellular Membranes/chemistry , Intracellular Membranes/ultrastructure , Models, Molecular , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Mutation , Protein Domains , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/ultrastructure , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Substrate Specificity
9.
J Biol Chem ; 300(6): 107387, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38763336

ABSTRACT

The cryo-EM resolution revolution has heralded a new era in our understanding of eukaryotic lipid flippases with a rapidly growing number of high-resolution structures. Flippases belong to the P4 family of ATPases (type IV P-type ATPases) that largely follow the reaction cycle proposed for the more extensively studied cation-transporting P-type ATPases. However, unlike the canonical P-type ATPases, no flippase cargos are transported in the phosphorylation half-reaction. Instead of being released into the intracellular or extracellular milieu, lipid cargos are transported to their destination at the inner leaflet of the membrane. Recent flippase structures have revealed multiple conformational states during the lipid transport cycle. Nonetheless, critical conformational states capturing the lipid cargo "in transit" are still missing. In this review, we highlight the amazing structural advances of these lipid transporters, discuss various perspectives on catalytic and regulatory mechanisms in the literature, and shed light on future directions in further deciphering the detailed molecular mechanisms of lipid flipping.


Subject(s)
Adenosine Triphosphatases , Humans , Animals , Adenosine Triphosphatases/metabolism , Adenosine Triphosphatases/chemistry , Cryoelectron Microscopy , Biological Transport , Phospholipid Transfer Proteins/metabolism , Phospholipid Transfer Proteins/chemistry , Phospholipid Transfer Proteins/genetics , Lipid Metabolism , Protein Conformation
10.
J Biol Chem ; : 107706, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39178950

ABSTRACT

Mutations that cause loss of function of GlcNAc-1-phosphotransferase (PTase) lead to the lysosomal storage disorder mucolipidosis II. PTase is the key enzyme of the mannose 6-phosphate (M6P) targeting system that is responsible for tagging lysosomal hydrolases with the M6P moiety for their delivery to the lysosome. We had previously generated a truncated hyperactive form of PTase termed S1S3 which was shown to notably increase the phosphorylation level of secreted lysosomal enzymes and enhance their uptake by cells. Here, we report the 3.4 Å cryo-EM structure of soluble S1S3 lacking both transmembrane domains and cytosolic tails. The structure reveals a high degree of conservation of the catalytic core to full-length PTase. In this dimeric structure, the EF hand of one protomer is observed interacting with the conserved region 4 of the other. In addition, we present a high-quality EM 3D map of the UDP-GlcNAc bound form of the full-length soluble protein showing the key molecular interactions between the nucleotide sugar donor and side chain amino acids of the protein. Finally, although the domain organization of S1S3 is very similar to that of the Drosophila melanogaster (fruit fly) PTase homolog, we establish that the latter does not act on lysosomal hydrolases.

11.
Mol Biol Evol ; 41(3)2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38421617

ABSTRACT

Polyploidy, a significant catalyst for speciation and evolutionary processes in both plant and animal kingdoms, has been recognized for a long time. However, the exact molecular mechanism that leads to polyploid formation, especially in vertebrates, is not fully understood. Our study aimed to elucidate this phenomenon using the zebrafish model. We successfully achieved an effective knockout of the cyclin N-terminal domain containing 1 (cntd1) using CRISPR/Cas9 technology. This resulted in impaired formation of meiotic crossovers, leading to cell-cycle arrest during meiotic metaphase and triggering apoptosis of spermatocytes in the testes. Despite these defects, the mutant (cntd1-/-) males were still able to produce a limited amount of sperm with normal ploidy and function. Interestingly, in the mutant females, it was the ploidy not the capacity of egg production that was altered. This resulted in the production of haploid, aneuploid, and unreduced gametes. This alteration enabled us to successfully obtain triploid and tetraploid zebrafish from cntd1-/- and cntd1-/-/- females, respectively. Furthermore, the tetraploid-heterozygous zebrafish produced reduced-diploid gametes and yielded all-triploid or all-tetraploid offspring when crossed with wild-type (WT) or tetraploid zebrafish, respectively. Collectively, our findings provide direct evidence supporting the crucial role of meiotic crossover defects in the process of polyploidization. This is particularly evident in the generation of unreduced eggs in fish and, potentially, other vertebrate species.


Subject(s)
Triploidy , Zebrafish , Male , Animals , Female , Tetraploidy , Seeds , Polyploidy , Ploidies
12.
Nat Chem Biol ; 19(5): 565-574, 2023 05.
Article in English | MEDLINE | ID: mdl-36593275

ABSTRACT

Heparan sulfate (HS) proteoglycans are extended (-GlcAß1,4GlcNAcα1,4-)n co-polymers containing decorations of sulfation and epimerization that are linked to cell surface and extracellular matrix proteins. In mammals, HS repeat units are extended by an obligate heterocomplex of two exostosin family members, EXT1 and EXT2, where each protein monomer contains distinct GT47 (GT-B fold) and GT64 (GT-A fold) glycosyltransferase domains. In this study, we generated human EXT1-EXT2 (EXT1-2) as a functional heterocomplex and determined its structure in the presence of bound donor and acceptor substrates. Structural data and enzyme activity of catalytic site mutants demonstrate that only two of the four glycosyltransferase domains are major contributors to co-polymer syntheses: the EXT1 GT-B fold ß1,4GlcA transferase domain and the EXT2 GT-A fold α1,4GlcNAc transferase domain. The two catalytic sites are over 90 Å apart, indicating that HS is synthesized by a dissociative process that involves a single catalytic site on each monomer.


Subject(s)
Heparitin Sulfate , Proteins , Animals , Humans , Heparitin Sulfate/chemistry , Glycosyltransferases/metabolism , N-Acetylglucosaminyltransferases/metabolism , Mammals
13.
FASEB J ; 38(15): e23865, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39096136

ABSTRACT

A thorough comprehension of age-related variances in orthodontic tooth movement (OTM) and bone remodeling response to mechanical force holds significant implications for enhancing orthodontic treatment. Mitophagy plays a crucial role in bone metabolism and various age-related diseases. However, the impact of mitophagy on the bone remodeling process during OTM remains elusive. Using adolescent (6 weeks old) and adult (12 months old) rats, we established OTM models and observed that orthodontic force increased the expression of the mitophagy proteins PTEN-induced putative kinase 1 (PINK1) and Parkin, as well as the number of tartrate-resistant acid phosphatase-positive osteoclasts and osteocalcin-positive osteoblasts. These biological changes were found to be age-related. In vitro, compression force loading promoted PINK1/Parkin-dependent mitophagy in periodontal ligament stem cells (PDLSCs) derived from adolescents (12-16 years old) and adults (25-35 years old). Furthermore, adult PDLSCs exhibited lower levels of mitophagy, impaired mitochondrial function, and a decreased ratio of RANKL/OPG compared to young PDLSCs after compression. Transfection of siRNA confirmed that inhibition of mitophagy in PDLSC resulted in decreased mitochondrial function and reduced RANKL/OPG ratio. Application of mitophagy inducer Urolithin A enhanced bone remodeling and accelerated OTM in rats, while the mitophagy inhibitor Mdivi-1 had the opposite effect. These findings indicate that force-stimulated PDLSC mitophagy contributes to alveolar bone remodeling during OTM, and age-related impairment of mitophagy negatively impacts the PDLSC response to mechanical stimulus. Our findings enhance the understanding of mitochondrial mechanotransduction and offer new targets to tackle current clinical challenges in orthodontic therapy.


Subject(s)
Mitochondria , Mitophagy , Osteoprotegerin , Periodontal Ligament , RANK Ligand , Tooth Movement Techniques , Animals , Mitophagy/physiology , Rats , RANK Ligand/metabolism , Periodontal Ligament/metabolism , Osteoprotegerin/metabolism , Mitochondria/metabolism , Male , Protein Kinases/metabolism , Rats, Sprague-Dawley , Adolescent , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Stem Cells/metabolism , Bone Remodeling/physiology , Cells, Cultured
14.
Cell ; 141(3): 446-57, 2010 Apr 30.
Article in English | MEDLINE | ID: mdl-20434985

ABSTRACT

The CED-4 homo-oligomer or apoptosome is required for initiation of programmed cell death in Caenorhabditis elegans by facilitating autocatalytic activation of the CED-3 caspase zymogen. How the CED-4 apoptosome assembles and activates CED-3 remains enigmatic. Here we report the crystal structure of the complete CED-4 apoptosome and show that it consists of eight CED-4 molecules, organized as a tetramer of an asymmetric dimer via a previously unreported interface among AAA(+) ATPases. These eight CED-4 molecules form a funnel-shaped structure. The mature CED-3 protease is monomeric in solution and forms an active holoenzyme with the CED-4 apoptosome, within which the protease activity of CED-3 is markedly stimulated. Unexpectedly, the octameric CED-4 apoptosome appears to bind only two, not eight, molecules of mature CED-3. The structure of the CED-4 apoptosome reveals shared principles for the NB-ARC family of AAA(+) ATPases and suggests a mechanism for the activation of CED-3.


Subject(s)
Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans/metabolism , Calcium-Binding Proteins/chemistry , Amino Acid Sequence , Animals , Apoptosomes/metabolism , Apoptotic Protease-Activating Factor 1/metabolism , Caenorhabditis elegans/chemistry , Caspases/chemistry , Crystallography, X-Ray , Models, Molecular , Sequence Alignment , X-Ray Diffraction
15.
Mol Cell ; 67(4): 646-658.e3, 2017 Aug 17.
Article in English | MEDLINE | ID: mdl-28781232

ABSTRACT

In miRNA-mediated gene silencing, the physical interaction between human Argonaute (hAgo) and GW182 (hGW182) is essential for facilitating the downstream silencing of the targeted mRNA. GW182 can interact with hAgo via three of the GW/WG repeats in its Argonaute-binding domain: motif-1, motif-2, and the hook motif. The structure of hAgo1 in complex with the hook motif of hGW182 reveals a "gate"-like interaction that is critical for GW182 docking into one of hAgo1's tryptophan-binding pockets. We show that hAgo1 and hAgo2 have a single GW182-binding site and that miRNA binding increases hAgo's affinity to GW182. With target binding occurring rapidly, this ensures that only mature RISC would be recruited for silencing. Finally, we show that hGW182 can recruit up to three copies of hAgo via its three GW motifs. This may explain the observed cooperativity in miRNA-mediated gene silencing.


Subject(s)
Argonaute Proteins/metabolism , Autoantigens/metabolism , Eukaryotic Initiation Factors/metabolism , Gene Silencing , MicroRNAs/metabolism , RNA, Guide, Kinetoplastida/metabolism , RNA-Binding Proteins/metabolism , Animals , Argonaute Proteins/chemistry , Argonaute Proteins/genetics , Autoantigens/chemistry , Autoantigens/genetics , Binding Sites , Binding, Competitive , Crystallography, X-Ray , Eukaryotic Initiation Factors/chemistry , Eukaryotic Initiation Factors/genetics , Humans , MicroRNAs/chemistry , MicroRNAs/genetics , Molecular Docking Simulation , Mutation , Nucleic Acid Conformation , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs , RNA, Guide, Kinetoplastida/chemistry , RNA, Guide, Kinetoplastida/genetics , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , Sf9 Cells , Structure-Activity Relationship , Transfection
16.
Am J Respir Crit Care Med ; 209(12): 1463-1476, 2024 06 15.
Article in English | MEDLINE | ID: mdl-38358857

ABSTRACT

Rationale: Acute cellular rejection (ACR) after lung transplant is a leading risk factor for chronic lung allograft dysfunction. Prior studies have demonstrated dynamic microbial changes occurring within the allograft and gut that influence local adaptive and innate immune responses. However, the lung microbiome's overall impact on ACR risk remains poorly understood. Objectives: To evaluate whether temporal changes in microbial signatures were associated with the development of ACR. Methods: We performed cross-sectional and longitudinal analyses (joint modeling of longitudinal and time-to-event data and trajectory comparisons) of 16S rRNA gene sequencing results derived from lung transplant recipient lower airway samples collected at multiple time points. Measurements and Main Results: Among 103 lung transplant recipients, 25 (24.3%) developed ACR. In comparing samples acquired 1 month after transplant, subjects who never developed ACR demonstrated lower airway enrichment with several oral commensals (e.g., Prevotella and Veillonella spp.) than those with current or future (beyond 1 mo) ACR. However, a subgroup analysis of those who developed ACR beyond 1 month revealed delayed enrichment with oral commensals occurring at the time of ACR diagnosis compared with baseline, when enrichment with more traditionally pathogenic taxa was present. In longitudinal models, dynamic changes in α-diversity (characterized by an initial decrease and a subsequent increase) and in the taxonomic trajectories of numerous oral commensals were more commonly observed in subjects with ACR. Conclusions: Dynamic changes in the lower airway microbiota are associated with the development of ACR, supporting its potential role as a useful biomarker or in ACR pathogenesis.


Subject(s)
Graft Rejection , Lung Transplantation , Humans , Lung Transplantation/adverse effects , Male , Graft Rejection/microbiology , Female , Middle Aged , Longitudinal Studies , Cross-Sectional Studies , Adult , Microbiota , RNA, Ribosomal, 16S/genetics , Lung/microbiology , Aged , Acute Disease
17.
Subcell Biochem ; 104: 383-408, 2024.
Article in English | MEDLINE | ID: mdl-38963493

ABSTRACT

Oxidoreductases facilitating electron transfer between molecules are pivotal in metabolic pathways. Flavin-based electron bifurcation (FBEB), a recently discovered energy coupling mechanism in oxidoreductases, enables the reversible division of electron pairs into two acceptors, bridging exergonic and otherwise unfeasible endergonic reactions. This chapter explores the four distinct FBEB complex families and highlights a decade of structural insights into FBEB complexes. In this chapter, we discuss the architecture, electron transfer routes, and conformational changes across all FBEB families, revealing the structural foundation that facilitate these remarkable functions.


Subject(s)
Flavins , Electron Transport , Flavins/metabolism , Flavins/chemistry , Oxidoreductases/metabolism , Oxidoreductases/chemistry , Protein Conformation , Models, Molecular , Oxidation-Reduction
18.
Proc Natl Acad Sci U S A ; 119(49): e2216240119, 2022 12 06.
Article in English | MEDLINE | ID: mdl-36442086

ABSTRACT

Duplication of DNA genomes requires unwinding of the double-strand (ds) DNA so that each single strand (ss) can be copied by a DNA polymerase. The genomes of eukaryotic cells are unwound by two ring-shaped hexameric helicases that initially encircle dsDNA but transition to ssDNA for function as replicative helicases. How the duplex is initially unwound, and the role of the two helicases in this process, is poorly understood. We recently described an initiation mechanism for eukaryotes in which the two helicases are directed inward toward one another and shear the duplex open by pulling on opposite strands of the duplex while encircling dsDNA [L. D. Langston, M. E. O'Donnell, eLife 8, e46515 (2019)]. Two head-to-head T-Antigen helicases are long known to be loaded at the SV40 origin. We show here that T-Antigen tracks head (N-tier) first on ssDNA, opposite the direction proposed for decades. We also find that SV40 T-Antigen tracks directionally while encircling dsDNA and mainly tracks on one strand of the duplex in the same orientation as during ssDNA translocation. Further, two inward directed T-Antigen helicases on dsDNA are able to melt a 150-bp duplex. These findings explain the "rabbit ear" DNA loops observed at the SV40 origin by electron microscopy and reconfigure how the DNA loops emerge from the double hexamer relative to earlier models. Thus, the mechanism of DNA shearing by two opposing helicases is conserved in a eukaryotic viral helicase and may be widely used to initiate origin unwinding of dsDNA genomes.


Subject(s)
Antigens, Viral, Tumor , DNA Helicases , Animals , Rabbits , Antigens, Viral, Tumor/genetics , DNA, Single-Stranded/genetics , DNA Replication , Eukaryota
19.
J Cell Mol Med ; 28(15): e18582, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39107876

ABSTRACT

Catheter ablation (CA) is an essential method for the interventional treatment of atrial fibrillation (AF), and it is very important to reduce long-term recurrence after CA. The mechanism of recurrence after CA is still unclear. We established a long-term model of beagle canines after circumferential pulmonary vein ablation (CPVA). The transcriptome and proteome were obtained using high-throughput sequencing and TMT-tagged LC-MS/LC analysis, respectively. Differentially expressed genes and proteins were screened and enriched, and the effect of fibrosis was found and verified in tissues. A downregulated protein, neuropeptide Y (NPY), was selected for validation and the results suggest that NPY may play a role in the long-term reinduction of AF after CPVA. Then, the molecular mechanism of NPY was further investigated. The results showed that the atrial effective refractory period (AERP) was shortened and fibrosis was increased after CPVA. Atrial myocyte apoptosis was alleviated by NPY intervention, and Akt activation was inhibited in cardiac fibroblasts. These results suggest that long-term suppression of NPY after CPVA may lead to induction of AF through promoting cardiomyocyte apoptosis and activating the Akt pathway in cardiac fibroblasts, which may make AF more likely to reinduce.


Subject(s)
Apoptosis , Atrial Fibrillation , Catheter Ablation , Myocardium , Neuropeptide Y , Pulmonary Veins , Animals , Dogs , Apoptosis/drug effects , Atrial Fibrillation/metabolism , Atrial Fibrillation/surgery , Atrial Fibrillation/pathology , Catheter Ablation/methods , Disease Models, Animal , Fibrosis , Heart Atria/metabolism , Heart Atria/pathology , Multiomics , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Neuropeptide Y/metabolism , Proteome/metabolism , Proteomics/methods , Proto-Oncogene Proteins c-akt/metabolism , Pulmonary Veins/metabolism , Pulmonary Veins/surgery , Transcriptome
20.
J Biol Chem ; 299(2): 102852, 2023 02.
Article in English | MEDLINE | ID: mdl-36592926

ABSTRACT

The Saccharomyces cerevisiae Yta7 is a chromatin remodeler harboring a histone-interacting bromodomain (BRD) and two AAA+ modules. It is not well understood how Yta7 recognizes the histone H3 tail to promote nucleosome disassembly for DNA replication or RNA transcription. By cryo-EM analysis, here we show that Yta7 assembles a three-tiered hexamer with a top BRD tier, a middle AAA1 tier, and a bottom AAA2 tier. Unexpectedly, the Yta7 BRD stabilizes a four-stranded ß-helix, termed BRD-interacting motif (BIM), of the largely disordered N-terminal region. The BIM motif is unique to the baker's yeast, and we show both BRD and BIM contribute to nucleosome recognition. We found that Yta7 binds both acetylated and nonacetylated H3 peptides but with a higher affinity for the unmodified peptide. This property is consistent with the absence of key residues of canonical BRDs involved in acetylated peptide recognition and the role of Yta7 in general nucleosome remodeling. Interestingly, the BRD tier exists in a spiral and a flat-ring form on top of the Yta7 AAA+ hexamer. The spiral is likely in a nucleosome-searching mode because the bottom BRD blocks the entry to the AAA+ chamber. The flat ring may be in a nucleosome disassembly state because the entry is unblocked and the H3 peptide has entered the AAA+ chamber and is stabilized by the AAA1 pore loops 1 and 2. Indeed, we show that the BRD tier is a flat ring when bound to the nucleosome. Overall, our study sheds light on the nucleosome disassembly by Yta7.


Subject(s)
Bromodomain Containing Proteins , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Adenosine Triphosphatases/metabolism , Bromodomain Containing Proteins/chemistry , Bromodomain Containing Proteins/genetics , Bromodomain Containing Proteins/metabolism , Chromatin Assembly and Disassembly , Chromosomal Proteins, Non-Histone/metabolism , Histones/metabolism , Nucleosomes/metabolism , Protein Conformation, beta-Strand , Protein Multimerization , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL