Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 251
Filter
Add more filters

Publication year range
1.
Annu Rev Immunol ; 33: 393-416, 2015.
Article in English | MEDLINE | ID: mdl-25622194

ABSTRACT

Innate immune responses depend on timely recognition of pathogenic or danger signals by multiple cell surface or cytoplasmic receptors and transmission of signals for proper counteractions through adaptor and effector molecules. At the forefront of innate immunity are four major signaling pathways, including those elicited by Toll-like receptors, RIG-I-like receptors, inflammasomes, or cGAS, each with its own cellular localization, ligand specificity, and signal relay mechanism. They collectively engage a number of overlapping signaling outcomes, such as NF-κB activation, interferon response, cytokine maturation, and cell death. Several proteins often assemble into a supramolecular complex to enable signal transduction and amplification. In this article, we review the recent progress in mechanistic delineation of proteins in these pathways, their structural features, modes of ligand recognition, conformational changes, and homo- and hetero-oligomeric interactions within the supramolecular complexes. Regardless of seemingly distinct interactions and mechanisms, the recurring themes appear to consist of autoinhibited resting-state receptors, ligand-induced conformational changes, and higher-order assemblies of activated receptors, adaptors, and signaling enzymes through conserved protein-protein interactions.


Subject(s)
Immunity, Innate/physiology , Animals , Humans , Inflammasomes/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Protein Binding , Receptors, Pattern Recognition/chemistry , Receptors, Pattern Recognition/metabolism , Signal Transduction , Structure-Activity Relationship
2.
Cell ; 184(20): 5107-5121.e14, 2021 09 30.
Article in English | MEDLINE | ID: mdl-34551316

ABSTRACT

Neural circuit assembly features simultaneous targeting of numerous neuronal processes from constituent neuron types, yet the dynamics is poorly understood. Here, we use the Drosophila olfactory circuit to investigate dynamic cellular processes by which olfactory receptor neurons (ORNs) target axons precisely to specific glomeruli in the ipsi- and contralateral antennal lobes. Time-lapse imaging of individual axons from 30 ORN types revealed a rich diversity in extension speed, innervation timing, and ipsilateral branch locations and identified that ipsilateral targeting occurs via stabilization of transient interstitial branches. Fast imaging using adaptive optics-corrected lattice light-sheet microscopy showed that upon approaching target, many ORN types exhibiting "exploring branches" consisted of parallel microtubule-based terminal branches emanating from an F-actin-rich hub. Antennal nerve ablations uncovered essential roles for bilateral axons in contralateral target selection and for ORN axons to facilitate dendritic refinement of postsynaptic partner neurons. Altogether, these observations provide cellular bases for wiring specificity establishment.


Subject(s)
Olfactory Pathways/cytology , Olfactory Pathways/diagnostic imaging , Time-Lapse Imaging , Animals , Axons/physiology , Cells, Cultured , Dendrites/physiology , Drosophila melanogaster/cytology , Drosophila melanogaster/physiology , Microtubules/metabolism , Olfactory Receptor Neurons/physiology , Time Factors
3.
Cell ; 184(23): 5759-5774.e20, 2021 11 11.
Article in English | MEDLINE | ID: mdl-34678144

ABSTRACT

NLRP6 is important in host defense by inducing functional outcomes including inflammasome activation and interferon production. Here, we show that NLRP6 undergoes liquid-liquid phase separation (LLPS) upon interaction with double-stranded RNA (dsRNA) in vitro and in cells, and an intrinsically disordered poly-lysine sequence (K350-354) of NLRP6 is important for multivalent interactions, phase separation, and inflammasome activation. Nlrp6-deficient or Nlrp6K350-354A mutant mice show reduced inflammasome activation upon mouse hepatitis virus or rotavirus infection, and in steady state stimulated by intestinal microbiota, implicating NLRP6 LLPS in anti-microbial immunity. Recruitment of ASC via helical assembly solidifies NLRP6 condensates, and ASC further recruits and activates caspase-1. Lipoteichoic acid, a known NLRP6 ligand, also promotes NLRP6 LLPS, and DHX15, a helicase in NLRP6-induced interferon signaling, co-forms condensates with NLRP6 and dsRNA. Thus, LLPS of NLRP6 is a common response to ligand stimulation, which serves to direct NLRP6 to distinct functional outcomes depending on the cellular context.


Subject(s)
Inflammasomes/metabolism , RNA Viruses/physiology , Receptors, Cell Surface/metabolism , Amino Acid Sequence , Animals , CARD Signaling Adaptor Proteins/metabolism , Hepatocytes/virology , Intestines/virology , Intrinsically Disordered Proteins/chemistry , Lipopolysaccharides/metabolism , Liver/virology , Mice , Polylysine/metabolism , Protein Binding , RNA, Double-Stranded/metabolism , Receptors, Cell Surface/chemistry , Signal Transduction , Teichoic Acids/metabolism
4.
Cell ; 176(6): 1477-1489.e14, 2019 03 07.
Article in English | MEDLINE | ID: mdl-30827683

ABSTRACT

Receptor clustering on the cell membrane is critical in the signaling of many immunoreceptors, and this mechanism has previously been attributed to the extracellular and/or the intracellular interactions. Here, we report an unexpected finding that for death receptor 5 (DR5), a receptor in the tumor necrosis factor receptor superfamily, the transmembrane helix (TMH) alone in the receptor directly assembles a higher-order structure to drive signaling and that this structure is inhibited by the unliganded ectodomain. Nuclear magnetic resonance structure of the TMH in bicelles shows distinct trimerization and dimerization faces, allowing formation of dimer-trimer interaction networks. Single-TMH mutations that disrupt either trimerization or dimerization abolish ligand-induced receptor activation. Surprisingly, proteolytic removal of the DR5 ectodomain can fully activate downstream signaling in the absence of ligand. Our data suggest a receptor activation mechanism in which binding of ligand or antibodies to overcome the pre-ligand autoinhibition allows TMH clustering and thus signaling.


Subject(s)
Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Apoptosis , Cell Line, Tumor , Cell Membrane/metabolism , HEK293 Cells , Humans , Ligands , Models, Molecular , Mutagenesis, Site-Directed/methods , Protein Binding , Proteolysis , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/chemistry , Receptors, TNF-Related Apoptosis-Inducing Ligand/ultrastructure , Signal Transduction
5.
Mol Cell ; 83(24): 4586-4599.e5, 2023 Dec 21.
Article in English | MEDLINE | ID: mdl-38096827

ABSTRACT

SIR2-HerA, a bacterial two-protein anti-phage defense system, induces bacterial death by depleting NAD+ upon phage infection. Biochemical reconstitution of SIR2, HerA, and the SIR2-HerA complex reveals a dynamic assembly process. Unlike other ATPases, HerA can form various oligomers, ranging from dimers to nonamers. When assembled with SIR2, HerA forms a hexamer and converts SIR2 from a nuclease to an NAD+ hydrolase, representing an unexpected regulatory mechanism mediated by protein assembly. Furthermore, high concentrations of ATP can inhibit NAD+ hydrolysis by the SIR2-HerA complex. Cryo-EM structures of the SIR2-HerA complex reveal a giant supramolecular assembly up to 1 MDa, with SIR2 as a dodecamer and HerA as a hexamer, crucial for anti-phage defense. Unexpectedly, the HerA hexamer resembles a spiral staircase and exhibits helicase activities toward dual-forked DNA. Together, we reveal the supramolecular assembly of SIR2-HerA as a unique mechanism for switching enzymatic activities and bolstering anti-phage defense strategies.


Subject(s)
Escherichia coli Proteins , Escherichia coli , Sirtuins , T-Phages , Adenosine Triphosphatases/genetics , Bacterial Proteins/genetics , NAD , Sirtuins/metabolism , Escherichia coli/enzymology , Escherichia coli/virology , Escherichia coli Proteins/metabolism
6.
Cell ; 163(5): 1138-1152, 2015 Nov 19.
Article in English | MEDLINE | ID: mdl-26548953

ABSTRACT

Diverse repertoires of antigen-receptor genes that result from combinatorial splicing of coding segments by V(D)J recombination are hallmarks of vertebrate immunity. The (RAG1-RAG2)2 recombinase (RAG) recognizes recombination signal sequences (RSSs) containing a heptamer, a spacer of 12 or 23 base pairs, and a nonamer (12-RSS or 23-RSS) and introduces precise breaks at RSS-coding segment junctions. RAG forms synaptic complexes only with one 12-RSS and one 23-RSS, a dogma known as the 12/23 rule that governs the recombination fidelity. We report cryo-electron microscopy structures of synaptic RAG complexes at up to 3.4 Å resolution, which reveal a closed conformation with base flipping and base-specific recognition of RSSs. Distortion at RSS-coding segment junctions and base flipping in coding segments uncover the two-metal-ion catalytic mechanism. Induced asymmetry involving tilting of the nonamer-binding domain dimer of RAG1 upon binding of HMGB1-bent 12-RSS or 23-RSS underlies the molecular mechanism for the 12/23 rule.


Subject(s)
DNA-Binding Proteins/chemistry , Homeodomain Proteins/chemistry , V(D)J Recombination , Amino Acid Sequence , Animals , Cryoelectron Microscopy , DNA-Binding Proteins/genetics , DNA-Binding Proteins/ultrastructure , Homeodomain Proteins/genetics , Homeodomain Proteins/ultrastructure , Humans , Mice , Molecular Sequence Data , Multiprotein Complexes/chemistry , Multiprotein Complexes/ultrastructure , Mutation , Sequence Alignment , Zebrafish
7.
Nature ; 621(7977): 154-161, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37494956

ABSTRACT

Although eukaryotic and long prokaryotic Argonaute proteins (pAgos) cleave nucleic acids, some short pAgos lack nuclease activity and hydrolyse NAD(P)+ to induce bacterial cell death1. Here we present a hierarchical activation pathway for SPARTA, a short pAgo consisting of an Argonaute (Ago) protein and TIR-APAZ, an associated protein2. SPARTA progresses through distinct oligomeric forms, including a monomeric apo state, a monomeric RNA-DNA-bound state, two dimeric RNA-DNA-bound states and a tetrameric RNA-DNA-bound active state. These snapshots together identify oligomerization as a mechanistic principle of SPARTA activation. The RNA-DNA-binding channel of apo inactive SPARTA is occupied by an auto-inhibitory motif in TIR-APAZ. After the binding of RNA-DNA, SPARTA transitions from a monomer to a symmetric dimer and then an asymmetric dimer, in which two TIR domains interact through charge and shape complementarity. Next, two dimers assemble into a tetramer with a central TIR cluster responsible for hydrolysing NAD(P)+. In addition, we observe unique features of interactions between SPARTA and RNA-DNA, including competition between the DNA 3' end and the auto-inhibitory motif, interactions between the RNA G2 nucleotide and Ago, and splaying of the RNA-DNA duplex by two loops exclusive to short pAgos. Together, our findings provide a mechanistic basis for the activation of short pAgos, a large section of the Ago superfamily.


Subject(s)
Argonaute Proteins , Prokaryotic Cells , Apoproteins/chemistry , Apoproteins/metabolism , Argonaute Proteins/chemistry , Argonaute Proteins/classification , Argonaute Proteins/metabolism , DNA/metabolism , Enzyme Activation , NAD/metabolism , Prokaryotic Cells/metabolism , RNA/metabolism
8.
Mol Cell ; 80(3): 501-511.e3, 2020 11 05.
Article in English | MEDLINE | ID: mdl-33065002

ABSTRACT

Vesicular- or vacuolar-type adenosine triphosphatases (V-ATPases) are ATP-driven proton pumps comprised of a cytoplasmic V1 complex for ATP hydrolysis and a membrane-embedded Vo complex for proton transfer. They play important roles in acidification of intracellular vesicles, organelles, and the extracellular milieu in eukaryotes. Here, we report cryoelectron microscopy structures of human V-ATPase in three rotational states at up to 2.9-Å resolution. Aided by mass spectrometry, we build all known protein subunits with associated N-linked glycans and identify glycolipids and phospholipids in the Vo complex. We define ATP6AP1 as a structural hub for Vo complex assembly because it connects to multiple Vo subunits and phospholipids in the c-ring. The glycolipids and the glycosylated Vo subunits form a luminal glycan coat critical for V-ATPase folding, localization, and stability. This study identifies mechanisms of V-ATPase assembly and biogenesis that rely on the integrated roles of ATP6AP1, glycans, and lipids.


Subject(s)
Vacuolar Proton-Translocating ATPases/metabolism , Vacuolar Proton-Translocating ATPases/physiology , Vacuolar Proton-Translocating ATPases/ultrastructure , Cryoelectron Microscopy/methods , Cytoplasm/metabolism , Cytosol/metabolism , HEK293 Cells , Humans , Protein Subunits/metabolism , Structure-Activity Relationship
9.
Nature ; 593(7860): 607-611, 2021 05.
Article in English | MEDLINE | ID: mdl-33883744

ABSTRACT

As organelles of the innate immune system, inflammasomes activate caspase-1 and other inflammatory caspases that cleave gasdermin D (GSDMD). Caspase-1 also cleaves inactive precursors of the interleukin (IL)-1 family to generate mature cytokines such as IL-1ß and IL-18. Cleaved GSDMD forms transmembrane pores to enable the release of IL-1 and to drive cell lysis through pyroptosis1-9. Here we report cryo-electron microscopy structures of the pore and the prepore of GSDMD. These structures reveal the different conformations of the two states, as well as extensive membrane-binding elements including a hydrophobic anchor and three positively charged patches. The GSDMD pore conduit is predominantly negatively charged. By contrast, IL-1 precursors have an acidic domain that is proteolytically removed by caspase-110. When permeabilized by GSDMD pores, unlysed liposomes release positively charged and neutral cargoes faster than negatively charged cargoes of similar sizes, and the pores favour the passage of IL-1ß and IL-18 over that of their precursors. Consistent with these findings, living-but not pyroptotic-macrophages preferentially release mature IL-1ß upon perforation by GSDMD. Mutation of the acidic residues of GSDMD compromises this preference, hindering intracellular retention of the precursor and secretion of the mature cytokine. The GSDMD pore therefore mediates IL-1 release by electrostatic filtering, which suggests the importance of charge in addition to size in the transport of cargoes across this large channel.


Subject(s)
Inflammasomes/chemistry , Interleukin-1beta/metabolism , Intracellular Signaling Peptides and Proteins/chemistry , Macrophages/metabolism , Phosphate-Binding Proteins/chemistry , Animals , Caspase 1/metabolism , Cryoelectron Microscopy , Humans , Interleukin-1/metabolism , Mice, Inbred C57BL , Protein Precursors/metabolism , Protein Structure, Quaternary , Static Electricity
10.
Mol Cell ; 69(4): 535-536, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29452633

ABSTRACT

While innate immunity is crucial for host defense, dysregulated signaling activation leads to pathological inflammation. In this issue of Molecular Cell, Goncharov et al. (2018) present a strategy to combat inflammatory diseases by disrupting RIP2-XIAP interaction in NOD2-mediated signaling.


Subject(s)
Nod2 Signaling Adaptor Protein , Receptor-Interacting Protein Serine-Threonine Kinase 2 , Humans , Immunity, Innate , Inflammation , Signal Transduction , X-Linked Inhibitor of Apoptosis Protein
11.
PLoS Pathog ; 19(12): e1011818, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38048362

ABSTRACT

During asexual growth and replication cycles inside red blood cells, the malaria parasite Plasmodium falciparum primarily relies on glycolysis for energy supply, as its single mitochondrion performs little or no oxidative phosphorylation. Post merozoite invasion of a host red blood cell, the ring stage lasts approximately 20 hours and was traditionally thought to be metabolically quiescent. However, recent studies have shown that the ring stage is active in several energy-costly processes, including gene transcription, protein translation, protein export, and movement inside the host cell. It has remained unclear whether a low glycolytic flux alone can meet the energy demand of the ring stage over a long period post invasion. Here, we demonstrate that the metabolic by-product pyrophosphate (PPi) is a critical energy source for the development of the ring stage and its transition to the trophozoite stage. During early phases of the asexual development, the parasite utilizes Plasmodium falciparum vacuolar pyrophosphatase 1 (PfVP1), an ancient pyrophosphate-driven proton pump, to export protons across the parasite plasma membrane. Conditional deletion of PfVP1 leads to a delayed ring stage that lasts nearly 48 hours and a complete blockage of the ring-to-trophozoite transition before the onset of parasite death. This developmental arrest can be partially rescued by an orthologous vacuolar pyrophosphatase from Arabidopsis thaliana, but not by the soluble pyrophosphatase from Saccharomyces cerevisiae, which lacks proton pumping activities. Since proton-pumping pyrophosphatases have been evolutionarily lost in human hosts, the essentiality of PfVP1 suggests its potential as an antimalarial drug target. A drug target of the ring stage is highly desired, as current antimalarials have limited efficacy against this stage.


Subject(s)
Antimalarials , Malaria, Falciparum , Animals , Humans , Plasmodium falciparum/metabolism , Proton Pumps/metabolism , Trophozoites/metabolism , Diphosphates/metabolism , Protons , Erythrocytes/parasitology , Pyrophosphatases/metabolism , Malaria, Falciparum/parasitology , Antimalarials/metabolism
12.
FASEB J ; 38(5): e23515, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38470367

ABSTRACT

Endometriosis is a benign gynecological disease that shares some common features of malignancy. Autophagy plays vital roles in endometriosis and influences endometrial cell metastasis, and hypoxia was identified as the initiator of this pathological process through hypoxia inducible factor 1 alpha (HIF-1α). A newly discovered circular RNA FOXO3 (circFOXO3) is critical in cell autophagy, migration, and invasion of various diseases and is reported to be related to hypoxia, although its role in endometriosis remains to be elucidated up to now. In this study, a lower circFOXO3 expression in ectopic endometrium was investigated. Furthermore, we verified that circFOXO3 could regulate autophagy by downregulating the level of p53 protein to mediate the migration and invasion of human endometrial stromal cells (T HESCs). Additionally, the effects of HIF-1α on circFOXO3 and autophagy were examined in T HESCs. Notably, overexpression of HIF-1α could induce autophagy and inhibit circFOXO3 expression, whereas overexpressing of circFOXO3 under hypoxia significantly inhibited hypoxia-induced autophagy. Mechanistically, the direct combination between HIF-1α and HIF-1α-binding site on adenosine deaminase 1 acting on RNA (ADAR1) promoter increased the level of ADAR1 protein, which bind directly with circFOXO3 pre-mRNA to block the cyclization of circFOXO3. All these results support that hypoxia-mediated ADAR1 elevation inhibited the expression of circFOXO3, and then autophagy was induced upon loss of circFOXO3 via inhibition of p53 degradation, participating in the development of endometriosis.


Subject(s)
Endometriosis , Female , Humans , Endometriosis/genetics , Tumor Suppressor Protein p53 , RNA , RNA, Circular/genetics , Autophagy , Hypoxia
13.
Proc Natl Acad Sci U S A ; 119(35): e2203742119, 2022 08 30.
Article in English | MEDLINE | ID: mdl-35994636

ABSTRACT

Vacuolar-type adenosine triphosphatases (V-ATPases) not only function as rotary proton pumps in cellular organelles but also serve as signaling hubs. To identify the endogenous binding partners of V-ATPase, we collected a large dataset of human V-ATPases and did extensive classification and focused refinement of human V-ATPases. Unexpectedly, about 17% of particles in state 2 of human V-ATPases display additional density with an overall resolution of 3.3 Å. Structural analysis combined with artificial intelligence modeling enables us to identify this additional density as mEAK-7, a protein involved in mechanistic target of rapamycin (mTOR) signaling in mammals. Our structure shows that mEAK-7 interacts with subunits A, B, D, and E of V-ATPases in state 2. Thus, we propose that mEAK-7 may regulate V-ATPase function through binding to V-ATPases in state 2 as well as mediate mTOR signaling.


Subject(s)
Cryoelectron Microscopy , Data Mining , Vacuolar Proton-Translocating ATPases , Animals , Artificial Intelligence , Cryoelectron Microscopy/methods , Data Mining/methods , Humans , Mammals/genetics , Mammals/metabolism , Protein Binding/genetics , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Vacuolar Proton-Translocating ATPases/genetics , Vacuolar Proton-Translocating ATPases/metabolism
14.
Proc Natl Acad Sci U S A ; 119(9)2022 03 01.
Article in English | MEDLINE | ID: mdl-35149555

ABSTRACT

SARS-CoV-2 is a highly pathogenic virus that evades antiviral immunity by interfering with host protein synthesis, mRNA stability, and protein trafficking. The SARS-CoV-2 nonstructural protein 1 (Nsp1) uses its C-terminal domain to block the messenger RNA (mRNA) entry channel of the 40S ribosome to inhibit host protein synthesis. However, how SARS-CoV-2 circumvents Nsp1-mediated suppression for viral protein synthesis and if the mechanism can be targeted therapeutically remain unclear. Here, we show that N- and C-terminal domains of Nsp1 coordinate to drive a tuned ratio of viral to host translation, likely to maintain a certain level of host fitness while maximizing replication. We reveal that the stem-loop 1 (SL1) region of the SARS-CoV-2 5' untranslated region (5' UTR) is necessary and sufficient to evade Nsp1-mediated translational suppression. Targeting SL1 with locked nucleic acid antisense oligonucleotides inhibits viral translation and makes SARS-CoV-2 5' UTR vulnerable to Nsp1 suppression, hindering viral replication in vitro at a nanomolar concentration, as well as providing protection against SARS-CoV-2-induced lethality in transgenic mice expressing human ACE2. Thus, SL1 allows Nsp1 to switch infected cells from host to SARS-CoV-2 translation, presenting a therapeutic target against COVID-19 that is conserved among immune-evasive variants. This unique strategy of unleashing a virus' own virulence mechanism against itself could force a critical trade-off between drug resistance and pathogenicity.


Subject(s)
5' Untranslated Regions/genetics , Immune Evasion/genetics , Protein Biosynthesis , SARS-CoV-2/genetics , Viral Nonstructural Proteins/genetics , Animals , Base Sequence , Chlorocebus aethiops , HEK293 Cells , Host-Pathogen Interactions/drug effects , Host-Pathogen Interactions/genetics , Humans , Immune Evasion/drug effects , Mice, Transgenic , Models, Biological , Oligonucleotides, Antisense/pharmacology , Protein Biosynthesis/drug effects , SARS-CoV-2/drug effects , Vero Cells , Virus Replication/drug effects
15.
J Nutr ; 154(6): 1861-1868, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38677479

ABSTRACT

BACKGROUND: Cumulative preclinical evidence reported quercetin, a major flavonoid, can attenuate the disease activity of inflammatory bowel diseases (IBD). However, there is limited evidence that supports the benefits of quercetin for patients with IBD. OBJECTIVES: To investigate whether dietary quercetin intake is associated with adverse outcomes among individuals with IBD in a prospective cohort study. METHODS: We included 2293 participants with IBD (764 Crohn's disease [CD] and 1529 ulcerative colitis [UC]) from the UK Biobank. Dietary information was collected using validated 24-h dietary assessments, and quercetin intake was estimated based on national nutrient databases. Two outcomes, enterotomy and all-cause mortality, were obtained based on the national data. Cox proportional hazard models were used to estimate the hazard ratios (HRs) and 95% confidence intervals (CIs). RESULTS: After a mean (standard deviation) follow-up of 9.6 (1.8) y, we documented 193 enterotomy events and 176 deaths. Compared with participants with the lowest quartile intake of quercetin, those in the highest quartiles were associated with lower risk of enterotomy (HR: 0.46; 95% CI: 0.28, 0.76) and all-cause mortality (HR: 0.53; 95% CI: 0.33, 0.83) in IBD. The inverse associations between quercetin and enterotomy were consistent in CD (HR: 0.30; 95% CI: 0.12, 0.78) but not UC (HR: 0.58; 95% CI: 0.32, 1.07), while the inverse associations between quercetin and mortality were consistent both in CD (HR: 0.37; 95% CI: 0.15, 0.92) and UC (HR: 0.55; 95% CI: 0.31, 0.95). CONCLUSIONS: Higher dietary intake of quercetin was associated with lower risk of enterotomy and all-cause mortality in IBD. Our study provides novel evidence that further suggests the benefits of quercetin for patients with IBD, while also calling for further validation in other cohorts and clinical trials.


Subject(s)
Diet , Inflammatory Bowel Diseases , Quercetin , Humans , Quercetin/administration & dosage , Quercetin/pharmacology , Prospective Studies , Female , Male , Middle Aged , Adult , Cohort Studies , Proportional Hazards Models , Crohn Disease , Risk Factors
16.
BMC Cancer ; 24(1): 727, 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38877445

ABSTRACT

BACKGROUND: The Naples Prognostic Score (NPS), integrating inflammatory and nutritional biomarkers, has been reported to be associated with the prognosis of various malignancies, but there is no report on intrahepatic cholangiocarcinoma (ICC). This study aimed to explore the prognostic value of NPS in patients with ICC. METHODS: Patients with ICC after hepatectomy were collected, and divided into three groups. The prognosis factors were determined by Cox regression analysis. Predictive efficacy was evaluated by the time-dependent receiver operating characteristic (ROC) curves. RESULTS: A total of 174 patients were included (Group 1: 33 (19.0%) patients; Group 2: 83 (47.7%) patients; and Group 3: 58 (33.3%) patients). The baseline characteristics showed the higher the NPS, the higher the proportion of patients with cirrhosis and Child-Pugh B, and more advanced tumors. The Kaplan-Meier curves reflect higher NPS were associated with poor survival. Multivariable analysis showed NPS was an independent risk factor of overall survival (NPS group 2 vs. 1: HR = 1.671, 95% CI: 1.022-3.027, p = 0.009; NPS group 3 vs. 1: HR = 2.208, 95% CI: 1.259-4.780, p = 0.007) and recurrence-free survival (NPS group 2 vs. 1: HR = 1.506, 95% CI: 1.184-3.498, p = 0.010; NPS group 3 vs. 1: HR = 2.141, 95% CI: 2.519-4.087, P = 0.001). The time ROC indicated NPS was superior to other models in predicting prognosis. CONCLUSIONS: NPS is a simple and effective tool for predicting the long-term survival of patients with ICC after hepatectomy. Patients with high NPS require close follow-up, and improving NPS may prolong the survival time.


Subject(s)
Bile Duct Neoplasms , Cholangiocarcinoma , Hepatectomy , Humans , Cholangiocarcinoma/surgery , Cholangiocarcinoma/mortality , Cholangiocarcinoma/pathology , Male , Female , Middle Aged , Prognosis , Bile Duct Neoplasms/surgery , Bile Duct Neoplasms/mortality , Bile Duct Neoplasms/pathology , Aged , ROC Curve , Retrospective Studies , Kaplan-Meier Estimate , Adult , Risk Factors
17.
Mol Cell ; 61(4): 602-613, 2016 Feb 18.
Article in English | MEDLINE | ID: mdl-26853147

ABSTRACT

Fas (CD95, Apo-1, or TNFRSF6) is a prototypical apoptosis-inducing death receptor in the tumor necrosis factor receptor (TNFR) superfamily. While the extracellular domains of TNFRs form trimeric complexes with their ligands and the intracellular domains engage in higher-order oligomerization, the role of the transmembrane (TM) domains is unknown. We determined the NMR structures of mouse and human Fas TM domains in bicelles that mimic lipid bilayers. Surprisingly, these domains use proline motifs to create optimal packing in homotrimer assembly distinct from classical trimeric coiled-coils in solution. Cancer-associated and structure-based mutations in Fas TM disrupt trimerization in vitro and reduce apoptosis induction in vivo, indicating the essential role of intramembrane trimerization in receptor activity. Our data suggest that the structures represent the signaling-active conformation of Fas TM, which appears to be different from the pre-ligand conformation. Analysis of other TNFR sequences suggests proline-containing sequences as common motifs for receptor TM trimerization.


Subject(s)
Lipid Bilayers/metabolism , Proline/metabolism , fas Receptor/chemistry , fas Receptor/metabolism , Animals , Apoptosis , HEK293 Cells , HeLa Cells , Humans , Magnetic Resonance Imaging , Mice , Models, Molecular , Mutation , Protein Multimerization , Protein Structure, Tertiary , Signal Transduction , fas Receptor/genetics
18.
Mol Cell ; 64(2): 236-250, 2016 10 20.
Article in English | MEDLINE | ID: mdl-27746017

ABSTRACT

Caspase-8 activation can be triggered by death receptor-mediated formation of the death-inducing signaling complex (DISC) and by the inflammasome adaptor ASC. Caspase-8 assembles with FADD at the DISC and with ASC at the inflammasome through its tandem death effector domain (tDED), which is regulated by the tDED-containing cellular inhibitor cFLIP and the viral inhibitor MC159. Here we present the caspase-8 tDED filament structure determined by cryoelectron microscopy. Extensive assembly interfaces not predicted by the previously proposed linear DED chain model were uncovered, and were further confirmed by structure-based mutagenesis in filament formation in vitro and Fas-induced apoptosis and ASC-mediated caspase-8 recruitment in cells. Structurally, the two DEDs in caspase-8 use quasi-equivalent contacts to enable assembly. Using the tDED filament structure as a template, structural analyses reveal the interaction surfaces between FADD and caspase-8 and the distinct mechanisms of regulation by cFLIP and MC159 through comingling and capping, respectively.


Subject(s)
CASP8 and FADD-Like Apoptosis Regulating Protein/chemistry , Caspase 8/chemistry , Death Domain Receptor Signaling Adaptor Proteins/chemistry , Fas-Associated Death Domain Protein/chemistry , Viral Proteins/chemistry , Amino Acid Sequence , Apoptosis/drug effects , Binding Sites , CARD Signaling Adaptor Proteins , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Caspase 8/genetics , Caspase 8/metabolism , Cryoelectron Microscopy , Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Death Domain Receptor Signaling Adaptor Proteins/genetics , Death Domain Receptor Signaling Adaptor Proteins/metabolism , Death Effector Domain , Fas-Associated Death Domain Protein/genetics , Fas-Associated Death Domain Protein/metabolism , Gene Expression , Humans , Jurkat Cells , Plasmids/chemistry , Plasmids/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Transfection , Viral Proteins/genetics , Viral Proteins/metabolism , fas Receptor/pharmacology
19.
BMC Surg ; 24(1): 148, 2024 May 11.
Article in English | MEDLINE | ID: mdl-38734630

ABSTRACT

BACKGROUND & AIMS: Complications after laparoscopic liver resection (LLR) are important factors affecting the prognosis of patients, especially for complex hepatobiliary diseases. The present study aimed to evaluate the value of a three-dimensional (3D) printed dry-laboratory model in the precise planning of LLR for complex hepatobiliary diseases. METHODS: Patients with complex hepatobiliary diseases who underwent LLR were preoperatively enrolled, and divided into two groups according to whether using a 3D-printed dry-laboratory model (3D vs. control group). Clinical variables were assessed and complications were graded by the Clavien-Dindo classification. The Comprehensive Complication Index (CCI) scores were calculated and compared for each patient. Multivariable analysis was performed to determine the risk factors of postoperative complications. RESULTS: Sixty-two patients with complex hepatobiliary diseases underwent the precise planning of LLR. Among them, thirty-one patients acquired the guidance of a 3D-printed dry-laboratory model, and others were only guided by traditional enhanced CT or MRI. The results showed no significant differences between the two groups in baseline characters. However, compared to the control group, the 3D group had a lower incidence of intraoperative blood loss, as well as postoperative 30-day and major complications, especially bile leakage (all P < 0.05). The median score on the CCI was 20.9 (range 8.7-51.8) in the control group and 8.7 (range 8.7-43.4) in the 3D group (mean difference, -12.2, P = 0.004). Multivariable analysis showed the 3D model was an independent protective factor in decreasing postoperative complications. Subgroup analysis also showed that a 3D model could decrease postoperative complications, especially for bile leakage in patients with intrahepatic cholelithiasis. CONCLUSION: The 3D-printed models can help reduce postoperative complications. The 3D-printed models should be recommended for patients with complex hepatobiliary diseases undergoing precise planning LLR.


Subject(s)
Laparoscopy , Liver Diseases , Postoperative Complications , Printing, Three-Dimensional , Humans , Female , Male , Middle Aged , Laparoscopy/methods , Laparoscopy/adverse effects , Postoperative Complications/prevention & control , Postoperative Complications/etiology , Liver Diseases/surgery , Aged , Biliary Tract Diseases/prevention & control , Biliary Tract Diseases/surgery , Biliary Tract Diseases/etiology , Hepatectomy/methods , Hepatectomy/adverse effects , Adult , Retrospective Studies , Cohort Studies
20.
J Sci Food Agric ; 2024 Mar 24.
Article in English | MEDLINE | ID: mdl-38523359

ABSTRACT

BACKGROUND: Banana flour can provide a solution to people with gluten intolerance, as it is gluten-free. Native banana flour may have limited functionality in certain applications. In this study, banana flour was modified by ultrasonic (US) and annealing (ANN) treatments at four incubation time spans, namely 12, 24, 36 and 72 h, separately or combined sequentially (US-ANN) to enhance the physicochemical and digestive properties. RESULTS: US led to exposed granular surfaces and damaged non-starch components. Both treatments, at extended incubation time, increased crystallinity, resulting in a narrower starch gelatinization temperature range. The swelling power was significantly lower for ANN and US-ANN compared to US alone, providing a delay of gelatinization temperature. However, none of the treatments affected the gelatinization enthalpy. Furthermore, US increased peak viscosity, breakdown, final viscosity and setback whereas the opposite results were obtained for ANN and US-ANN. Additionally, US prior to ANN significantly increased the resistant starch (RS) content for annealing times over 24 h, especially for the US-ANN treatment for 72 h, which provided the highest RS content (49.3%) compared to ANN treatment for 72 h (44.0%) and native flour (36.3%). CONCLUSIONS: US prior to ANN treatment offers an alternative method to improve the functional and digestive properties of banana flour, extending the range of applications. © 2024 Society of Chemical Industry.

SELECTION OF CITATIONS
SEARCH DETAIL