Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
Add more filters










Publication year range
1.
J Lipid Res ; 65(4): 100530, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38479648

ABSTRACT

Atherosclerosis results from the deposition and oxidation of LDL and immune cell infiltration in the sub-arterial space leading to arterial occlusion. Studies have shown that transcytosis transports circulating LDL across endothelial cells lining blood vessels. LDL transcytosis is initiated by binding to either scavenger receptor B1 (SR-B1) or activin A receptor-like kinase 1 on the apical side of endothelial cells leading to its transit and release on the basolateral side. HDL is thought to partly protect individuals from atherosclerosis due to its ability to remove excess cholesterol and act as an antioxidant. Apolipoprotein A1 (APOA1), an HDL constituent, can bind to SR-B1, raising the possibility that APOA1/HDL can compete with LDL for SR-B1 binding, thereby limiting LDL deposition in the sub-arterial space. To examine this possibility, we used in vitro approaches to quantify the internalization and transcytosis of fluorescent LDL in coronary endothelial cells. Using microscale thermophoresis and affinity capture, we find that SR-B1 and APOA1 interact and that binding is enhanced when using the cardioprotective variant of APOA1 termed Milano (APOA1-Milano). In male mice, transiently increasing the levels of HDL reduced the acute deposition of fluorescently labeled LDL in the atheroprone inner curvature of the aorta. Reduced LDL deposition was also observed when increasing circulating wild-type APOA1 or the APOA1-Milano variant, with a more robust inhibition from the APOA1-Milano. The results suggest that HDL may limit SR-B1-mediated LDL transcytosis and deposition, adding to the mechanisms by which it can act as an atheroprotective particle.


Subject(s)
Apolipoprotein A-I , Lipoproteins, HDL , Lipoproteins, LDL , Transcytosis , Animals , Humans , Male , Mice , Apolipoprotein A-I/metabolism , Atherosclerosis/metabolism , Endothelial Cells/metabolism , Lipoproteins, HDL/metabolism , Lipoproteins, LDL/metabolism , Protein Binding , Scavenger Receptors, Class B/metabolism
2.
Nat Commun ; 15(1): 2553, 2024 Mar 22.
Article in English | MEDLINE | ID: mdl-38519472

ABSTRACT

Lysosomal Storage Disorders (LSDs), which share common phenotypes, including enlarged lysosomes and defective lysosomal storage, are caused by mutations in lysosome-related genes. Although gene therapies and enzyme replacement therapies have been explored, there are currently no effective routine therapies against LSDs. During lysosome reformation, which occurs when the functional lysosome pool is reduced, lysosomal lipids and proteins are recycled to restore lysosome functions. Here we report that the sorting nexin protein SNX8 promotes lysosome tubulation, a process that is required for lysosome reformation, and that loss of SNX8 leads to phenotypes characteristic of LSDs in human cells. SNX8 overexpression rescued features of LSDs in cells, and AAV-based delivery of SNX8 to the brain rescued LSD phenotypes in mice. Importantly, by screening a natural compound library, we identified three small molecules that enhanced SNX8-lysosome binding and reversed LSD phenotypes in human cells and in mice. Altogether, our results provide a potential solution for the treatment of LSDs.


Subject(s)
Lysosomal Storage Diseases , Mice , Animals , Humans , Lysosomal Storage Diseases/genetics , Lysosomal Storage Diseases/therapy , Lysosomal Storage Diseases/metabolism , Proteins/metabolism , Brain/metabolism , Mutation , Lysosomes/metabolism , Sorting Nexins/genetics , Sorting Nexins/metabolism
3.
Nat Cell Biol ; 26(2): 219-234, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38253667

ABSTRACT

Lysosomal storage disorders (LSDs), which are characterized by genetic and metabolic lysosomal dysfunctions, constitute over 60 degenerative diseases with considerable health and economic burdens. However, the mechanisms driving the progressive death of functional cells due to lysosomal defects remain incompletely understood, and broad-spectrum therapeutics against LSDs are lacking. Here, we found that various gene abnormalities that cause LSDs, including Hexb, Gla, Npc1, Ctsd and Gba, all shared mutual properties to robustly autoactivate neuron-intrinsic cGAS-STING signalling, driving neuronal death and disease progression. This signalling was triggered by excessive cytoplasmic congregation of the dsDNA and DNA sensor cGAS in neurons. Genetic ablation of cGAS or STING, digestion of neuronal cytosolic dsDNA by DNase, and repair of neuronal lysosomal dysfunction alleviated symptoms of Sandhoff disease, Fabry disease and Niemann-Pick disease, with substantially reduced neuronal loss. We therefore identify a ubiquitous mechanism mediating the pathogenesis of a variety of LSDs, unveil an inherent connection between lysosomal defects and innate immunity, and suggest a uniform strategy for curing LSDs.


Subject(s)
Lysosomal Storage Diseases , Niemann-Pick Disease, Type C , Humans , Lysosomal Storage Diseases/genetics , Lysosomal Storage Diseases/metabolism , Lysosomal Storage Diseases/pathology , Niemann-Pick Disease, Type C/genetics , Niemann-Pick Disease, Type C/pathology , Lysosomes/metabolism , Immunity, Innate , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism
4.
Adv Sci (Weinh) ; 11(10): e2303341, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38145352

ABSTRACT

High-fat diet (HFD)-induced obesity is a crucial risk factor for metabolic syndrome, mainly due to adipose tissue dysfunctions associated with it. However, the underlying mechanism remains unclear. This study has used genetic screening to identify an obesity-associated human lncRNA LINK-A as a critical molecule bridging the metabolic microenvironment and energy expenditure in vivo by establishing the HFD-induced obesity knock-in (KI) mouse model. Mechanistically, HFD LINK-A KI mice induce the infiltration of inflammatory factors, including IL-1ß and CXCL16, through the LINK-A/HB-EGF/HIF1α feedback loop axis in a self-amplified manner, thereby promoting the adipose tissue microenvironment remodeling and adaptive thermogenesis disorder, ultimately leading to obesity and insulin resistance. Notably, LINK-A expression is positively correlated with inflammatory factor expression in individuals who are overweight. Of note, targeting LINK-A via nucleic acid drug antisense oligonucleotides (ASO) attenuate HFD-induced obesity and metabolic syndrome, pointing out LINK-A as a valuable and effective therapeutic target for treating HFD-induced obesity. Briefly, the results reveale the roles of lncRNAs (such as LINK-A) in remodeling tissue inflammatory microenvironments to promote HFD-induced obesity.


Subject(s)
Insulin Resistance , Metabolic Syndrome , RNA, Long Noncoding , Humans , Animals , Mice , RNA, Long Noncoding/metabolism , Metabolic Syndrome/complications , Metabolic Syndrome/metabolism , Obesity/metabolism , Adipose Tissue/metabolism , Diet, High-Fat
5.
Mol Cell ; 83(23): 4370-4385.e9, 2023 Dec 07.
Article in English | MEDLINE | ID: mdl-38016475

ABSTRACT

Targeting epigenetic regulators to potentiate anti-PD-1 immunotherapy converges on the activation of type I interferon (IFN-I) response, mimicking cellular response to viral infection, but how its strength and duration are regulated to impact combination therapy efficacy remains largely unknown. Here, we show that mitochondrial CPT1A downregulation following viral infection restrains, while its induction by epigenetic perturbations sustains, a double-stranded RNA-activated IFN-I response. Mechanistically, CPT1A recruits the endoplasmic reticulum-localized ZDHHC4 to catalyze MAVS Cys79-palmitoylation, which promotes MAVS stabilization and activation by inhibiting K48- but facilitating K63-linked ubiquitination. Further elevation of CPT1A incrementally increases MAVS palmitoylation and amplifies the IFN-I response, which enhances control of viral infection and epigenetic perturbation-induced antitumor immunity. Moreover, CPT1A chemical inducers augment the therapeutic effect of combined epigenetic treatment with PD-1 blockade in refractory tumors. Our study identifies CPT1A as a stabilizer of MAVS activation, and its link to epigenetic perturbation can be exploited for cancer immunotherapy.


Subject(s)
Interferon Type I , Virus Diseases , Humans , Signal Transduction , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Lipoylation , Epigenesis, Genetic , Immunity, Innate
6.
Mol Cell ; 83(19): 3485-3501.e11, 2023 Oct 05.
Article in English | MEDLINE | ID: mdl-37802024

ABSTRACT

p62 is a well-characterized autophagy receptor that recognizes and sequesters specific cargoes into autophagosomes for degradation. p62 promotes the assembly and removal of ubiquitinated proteins by forming p62-liquid droplets. However, it remains unclear how autophagosomes efficiently sequester p62 droplets. Herein, we report that p62 undergoes reversible S-acylation in multiple human-, rat-, and mouse-derived cell lines, catalyzed by zinc-finger Asp-His-His-Cys S-acyltransferase 19 (ZDHHC19) and deacylated by acyl protein thioesterase 1 (APT1). S-acylation of p62 enhances the affinity of p62 for microtubule-associated protein 1 light chain 3 (LC3)-positive membranes and promotes autophagic membrane localization of p62 droplets, thereby leading to the production of small LC3-positive p62 droplets and efficient autophagic degradation of p62-cargo complexes. Specifically, increasing p62 acylation by upregulating ZDHHC19 or by genetic knockout of APT1 accelerates p62 degradation and p62-mediated autophagic clearance of ubiquitinated proteins. Thus, the protein S-acylation-deacylation cycle regulates p62 droplet recruitment to the autophagic membrane and selective autophagic flux, thereby contributing to the control of selective autophagic clearance of ubiquitinated proteins.


Subject(s)
Autophagosomes , Ubiquitinated Proteins , Mice , Rats , Humans , Animals , Autophagosomes/metabolism , Ubiquitinated Proteins/metabolism , Sequestosome-1 Protein/genetics , Sequestosome-1 Protein/metabolism , Autophagy/genetics , Acylation , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Mammals/metabolism
7.
Protein Cell ; 14(9): 653-667, 2023 09 14.
Article in English | MEDLINE | ID: mdl-37707322

ABSTRACT

Lipophagy, the selective engulfment of lipid droplets (LDs) by autophagosomes for lysosomal degradation, is critical to lipid and energy homeostasis. Here we show that the lipid transfer protein ORP8 is located on LDs and mediates the encapsulation of LDs by autophagosomal membranes. This function of ORP8 is independent of its lipid transporter activity and is achieved through direct interaction with phagophore-anchored LC3/GABARAPs. Upon lipophagy induction, ORP8 has increased localization on LDs and is phosphorylated by AMPK, thereby enhancing its affinity for LC3/GABARAPs. Deletion of ORP8 or interruption of ORP8-LC3/GABARAP interaction results in accumulation of LDs and increased intracellular triglyceride. Overexpression of ORP8 alleviates LD and triglyceride deposition in the liver of ob/ob mice, and Osbpl8-/- mice exhibit liver lipid clearance defects. Our results suggest that ORP8 is a lipophagy receptor that plays a key role in cellular lipid metabolism.


Subject(s)
Autophagy , Lipid Droplets , Animals , Mice , Autophagosomes , Homeostasis , Triglycerides
8.
J Cell Biol ; 222(5)2023 05 01.
Article in English | MEDLINE | ID: mdl-37043189

ABSTRACT

Macroautophagy (autophagy) utilizes a serial of receptors to specifically recognize and degrade autophagy cargoes, including damaged organelles, to maintain cellular homeostasis. Upstream signals spatiotemporally regulate the biological functions of selective autophagy receptors through protein post-translational modifications (PTM) such as phosphorylation. However, it is unclear how acetylation directly controls autophagy receptors in selective autophagy. Here, we report that an ER-phagy receptor FAM134B is acetylated by CBP acetyltransferase, eliciting intense ER-phagy. Furthermore, FAM134B acetylation promoted CAMKII-mediated phosphorylation to sustain a mode of milder ER-phagy. Conversely, SIRT7 deacetylated FAM134B to temper its activities in ER-phagy to avoid excessive ER degradation. Together, this work provides further mechanistic insights into how ER-phagy receptor perceives environmental signals for fine-tuning of ER homeostasis and demonstrates how nucleus-derived factors are programmed to control ER stress by modulating ER-phagy.


Subject(s)
Autophagy , Endoplasmic Reticulum , Intracellular Signaling Peptides and Proteins , Membrane Proteins , Sirtuins , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress , Homeostasis , Hydrolases/metabolism , Macroautophagy , Membrane Proteins/genetics , Membrane Proteins/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Sirtuins/genetics , Sirtuins/metabolism
9.
J Hepatol ; 78(4): 704-716, 2023 04.
Article in English | MEDLINE | ID: mdl-36574921

ABSTRACT

BACKGROUND & AIMS: Appropriate treatment options are lacking for hepatitis E virus (HEV)-infected pregnant women and immunocompromised individuals. Thus, we aimed to identify efficient anti-HEV drugs through high-throughput screening, validate them in vitro and in vivo (in a preclinical animal study), and elucidate their underlying antiviral mechanism of action. METHODS: Using appropriate cellular and rodent HEV infection models, we studied a critical pathway for host-HEV interactions and performed a preclinical study of the corresponding antivirals, which target proteostasis of the HEV replicase. RESULTS: We found 17 inhibitors that target HEV-HSP90 interactions by unbiased compound library screening on human hepatocytes harboring an HEV replicon. Inhibitors of HSP90 (iHSP90) markedly suppressed HEV replication with efficacy exceeding that of conventional antivirals (IFNα and ribavirin) in vitro. Mechanistically, iHSP90 treatment released the viral replicase ORF1 protein from the ORF1-HSP90 complex and triggered rapid ubiquitin/proteasome-mediated degradation of ORF1, resulting in abrogated HEV replication. Furthermore, a preclinical trial in a Mongolian gerbil HEV infection model showed this novel anti-HEV strategy to be safe, efficient, and able to prevent HEV-induced liver damage. CONCLUSIONS: In this study, we uncover a proteostatic pathway that is critical for host-HEV interactions and we provide a foundation from which to translate this new understanding of the HEV life cycle into clinically promising antivirals. IMPACT AND IMPLICATIONS: Appropriate treatment options for hepatitis E virus (HEV)-infected pregnant women and immunocompromised patients are lacking; hence, there is an urgent need for safe and effective HEV-specific therapies. This study identified new antivirals (inhibitors of HSP90) that significantly limit HEV infection by targeting the viral replicase for degradation. Moreover, these anti-HEV drugs were validated in an HEV rodent model and were found to be safe and efficient for prevention of HEV-induced liver injury in preclinical experiments. Our findings substantially promote the understanding of HEV pathobiology and pave the way for antiviral development.


Subject(s)
Hepatitis E virus , Hepatitis E , Animals , Humans , Female , Pregnancy , Proteostasis , Viral Replicase Complex Proteins , Hepatitis E/drug therapy , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Viral Proteins , Virus Replication
10.
Front Immunol ; 13: 1011590, 2022.
Article in English | MEDLINE | ID: mdl-36311750

ABSTRACT

Background: Current stratification systems for tumor prognostic prediction and immunotherapeutic efficacy evaluation are less satisfying in colorectal cancer (CRC). As infiltrating immune cells in tumor microenvironment (TME) played a key role in tumor progression and responses to immune checkpoint blockade (ICB) therapy, we want to construct an immune-related scoring system with detailed immune profiles to stratify CRC patients. Methods: We developed a scoring system based on immune-related signatures and validated its ability to predict prognosis and immunotherapeutic outcomes in CRC. CD45+ cells from CRC patients were sorted to investigate detailed immune profiles of the stratification system using mass cytometry. A single-cell RNA sequencing dataset was used to analyze transcriptomic profiles. Results: We constructed an immune-related signature score (IRScore) based on 54 recurrence-free survival (RFS)-related immune signatures to stratify CRC patients. We revealed that IRScore was positively correlated with RFS and favorable outcomes in ICB treatment. Moreover, we depicted a detailed immune profile in TME using mass cytometry and identified that CD103+CD39+ T cells, characterized by an exhaustive, cytotoxic and proliferative phenotype, were enriched in CRC patients with high IRScore. As a beneficial immune signature, CD103+CD39+ T cells could predict prognosis and responses to ICB therapy in CRC. Conclusions: All the analyses above revealed that IRScore could be a valuable tool for predicting prognosis and facilitating the development of new therapeutic strategies in CRC, and CD103+CD39+ T cells were one of defined immune signatures in IRScore, which might be a key factor for antitumor immunity.


Subject(s)
Colorectal Neoplasms , T-Lymphocytes , Humans , T-Lymphocytes/pathology , Colorectal Neoplasms/genetics , Prognosis , Immunotherapy , Lymphocyte Count , Tumor Microenvironment
11.
Cell Mol Immunol ; 18(10): 2372-2382, 2021 10.
Article in English | MEDLINE | ID: mdl-34480147

ABSTRACT

The nucleotide-binding domain, leucine-rich-repeat containing family, pyrin domain-containing 3 (NLRP3) inflammasome is essential in inflammation and inflammatory disorders. Phosphorylation at various sites on NLRP3 differentially regulates inflammasome activation. The Ser725 phosphorylation site on NLRP3 is depicted in multiple inflammasome activation scenarios, but the importance and regulation of this site has not been clarified. The present study revealed that the phosphorylation of Ser725 was an essential step for the priming of the NLRP3 inflammasome in macrophages. We also showed that Ser725 was directly phosphorylated by misshapen (Msn)/NIK-related kinase 1 (MINK1), depending on the direct interaction between MINK1 and the NLRP3 LRR domain. MINK1 deficiency reduced NLRP3 activation and suppressed inflammatory responses in mouse models of acute sepsis and peritonitis. Reactive oxygen species (ROS) upregulated the kinase activity of MINK1 and subsequently promoted inflammasome priming via NLRP3 Ser725 phosphorylation. Eliminating ROS suppressed NLRP3 activation and reduced sepsis and peritonitis symptoms in a MINK1-dependent manner. Altogether, our study reveals a direct regulation of the NLRP3 inflammasome by Msn family kinase MINK1 and suggests that modulation of MINK1 activity is a potential intervention strategy for inflammasome-related diseases.


Subject(s)
Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Animals , Inflammation , Macrophages , Mice , Reactive Oxygen Species
12.
EMBO J ; 40(13): e108812, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34132411

ABSTRACT

The cytosolic NOD1 and NOD2 pattern recognition receptors are typically known as sensors of bacterial peptidoglycan fragments. A new study in this issue links NOD1/2 activation with ER homeostasis through the bioactive metabolite sphingosine-1-phosphate (S1P).


Subject(s)
Nod1 Signaling Adaptor Protein , Nod2 Signaling Adaptor Protein , Lysophospholipids , Nod1 Signaling Adaptor Protein/genetics , Nod1 Signaling Adaptor Protein/metabolism , Nod2 Signaling Adaptor Protein/genetics , Nod2 Signaling Adaptor Protein/metabolism , Receptors, Pattern Recognition/metabolism , Signal Transduction , Sphingosine/analogs & derivatives
13.
Nat Commun ; 12(1): 2829, 2021 05 14.
Article in English | MEDLINE | ID: mdl-33990559

ABSTRACT

Polycomb group (PcG) proteins maintain cell identity by repressing gene expression during development. Surprisingly, emerging studies have recently reported that a number of PcG proteins directly activate gene expression during cell fate determination process. However, the mechanisms by which they direct gene activation in pluripotency remain poorly understood. Here, we show that Phc1, a subunit of canonical polycomb repressive complex 1 (cPRC1), can exert its function in pluripotency maintenance via a PRC1-independent activation of Nanog. Ablation of Phc1 reduces the expression of Nanog and overexpression of Nanog partially rescues impaired pluripotency caused by Phc1 depletion. We find that Phc1 interacts with Nanog and activates Nanog transcription by stabilizing the genome-wide chromatin interactions of the Nanog locus. This adds to the already known canonical function of PRC1 in pluripotency maintenance via a PRC1-dependent repression of differentiation genes. Overall, our study reveals a function of Phc1 to activate Nanog transcription through regulating chromatin architecture and proposes a paradigm for PcG proteins to maintain pluripotency.


Subject(s)
Chromatin/genetics , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/physiology , Nanog Homeobox Protein/genetics , Polycomb Repressive Complex 1/genetics , Polycomb Repressive Complex 1/physiology , Animals , Cells, Cultured , Gene Knockdown Techniques , Gene Knockout Techniques , Genome, Human , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/physiology , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Models, Genetic , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/physiology , Polycomb Repressive Complex 1/antagonists & inhibitors , Polycomb Repressive Complex 1/deficiency
14.
Hum Genomics ; 14(1): 45, 2020 12 07.
Article in English | MEDLINE | ID: mdl-33287903

ABSTRACT

BACKGROUND: Germline variants of ten keratin genes (K1, K2, K5, K6A, K6B, K9, K10, K14, K16, and K17) have been reported for causing different types of genodermatoses with an autosomal dominant mode of inheritance. Among all the variants of these ten keratin genes, most of them are missense variants. Unlike pathogenic and likely pathogenic variants, understanding the clinical importance of novel missense variants or variants of uncertain significance (VUS) is the biggest challenge for clinicians or medical geneticists. Functional characterization is the only way to understand the clinical association of novel missense variants or VUS but it is time consuming, costly, and depends on the availability of patient's samples. Existing databases report the pathogenic variants of the keratin genes, but never emphasize the systematic effects of these variants on keratin protein structure and genotype-phenotype correlation. RESULTS: To address this need, we developed a comprehensive database KVarPredDB, which contains information of all ten keratin genes associated with genodermatoses. We integrated and curated 400 reported pathogenic missense variants as well as 4629 missense VUS. KVarPredDB predicts the pathogenicity of novel missense variants as well as to understand the severity of disease phenotype, based on four criteria; firstly, the difference in physico-chemical properties between the wild type and substituted amino acids; secondly, the loss of inter/intra-chain interactions; thirdly, evolutionary conservation of the wild type amino acids and lastly, the effect of the substituted amino acids in the heptad repeat. Molecular docking simulations based on resolved crystal structures were adopted to predict stability changes and get the binding energy to compare the wild type protein with the mutated one. We use this basic information to determine the structural and functional impact of novel missense variants on the keratin coiled-coil heterodimer. KVarPredDB was built under the integrative web application development framework SSM (SpringBoot, Spring MVC, MyBatis) and implemented in Java, Bootstrap, React-mutation-mapper, MySQL, Tomcat. The website can be accessed through http://bioinfo.zju.edu.cn/KVarPredDB . The genomic variants and analysis results are freely available under the Creative Commons license. CONCLUSIONS: KVarPredDB provides an intuitive and user-friendly interface with computational analytical investigation for each missense variant of the keratin genes associated with genodermatoses.


Subject(s)
Computational Biology/methods , Databases, Genetic , Genetic Predisposition to Disease/genetics , Keratins/genetics , Mutation, Missense , Skin Diseases, Genetic/genetics , Humans , Phenotype , Polymorphism, Single Nucleotide , Protein Isoforms/genetics , Reproducibility of Results
15.
Biochem J ; 477(17): 3387-3399, 2020 09 18.
Article in English | MEDLINE | ID: mdl-32830849

ABSTRACT

The pseudokinase (PK) RNase L is a functional ribonuclease and plays important roles in human innate immunity. The ribonuclease activity of RNase L can be regulated by the kinase inhibitor sunitinib. The combined use of oncolytic virus and sunitinib has been shown to exert synergistic effects in anticancer therapy. In this study, we aimed to uncover the mechanism of action through which sunitinib inhibits RNase L. We solved the crystal structures of RNase L in complex with sunitinib and its analogs toceranib and SU11652. Our results showed that sunitinib bound to the ATP-binding pocket of RNase L. Unexpectedly, the αA helix linking the ankyrin repeat-domain and the PK domain affected the binding mode of sunitinib and resulted in an unusual flipped orientation relative to other structures in PDB. Molecular dynamics simulations and dynamic light scattering results support that the binding of sunitinib in the PK domain destabilized the dimer conformation of RNase L and allosterically inhibited its ribonuclease activity. Our study suggested that dimer destabilization could be an effective strategy for the discovery of RNase L inhibitors and that targeting the ATP-binding pocket in the PK domain of RNase L was an efficient approach for modulating its ribonuclease activity.


Subject(s)
Endoribonucleases , Protein Multimerization , Sunitinib/chemistry , Crystallography, X-Ray , Endoribonucleases/antagonists & inhibitors , Endoribonucleases/chemistry , Humans , Protein Conformation, alpha-Helical , Protein Domains
16.
Nat Commun ; 11(1): 2429, 2020 05 15.
Article in English | MEDLINE | ID: mdl-32415067

ABSTRACT

Proton-linked monocarboxylate transporters (MCTs) must transport monocarboxylate efficiently to facilitate monocarboxylate efflux in glycolytically active cells, and transport monocarboxylate slowly or even shut down to maintain a physiological monocarboxylate concentration in glycolytically inactive cells. To discover how MCTs solve this fundamental aspect of intracellular monocarboxylate homeostasis in the context of multicellular organisms, we analyzed pyruvate transport activity of human monocarboxylate transporter 2 (MCT2). Here we show that MCT2 transport activity exhibits steep dependence on substrate concentration. This property allows MCTs to turn on almost like a switch, which is physiologically crucial to the operation of MCTs in the cellular context. We further determined the cryo-electron microscopy structure of the human MCT2, demonstrating that the concentration sensitivity of MCT2 arises from the strong inter-subunit cooperativity of the MCT2 dimer during transport. These data establish definitively a clear example of evolutionary optimization of protein function.


Subject(s)
Monocarboxylic Acid Transporters/metabolism , Amino Acid Motifs , Cryoelectron Microscopy , Green Fluorescent Proteins/chemistry , HEK293 Cells , Homeostasis , Humans , Microscopy, Fluorescence , Molecular Docking Simulation , Protein Binding , Protein Multimerization , Protein Transport
17.
Trends Cell Biol ; 30(6): 452-466, 2020 06.
Article in English | MEDLINE | ID: mdl-32413315

ABSTRACT

Lysosomes are of major importance for the regulation of cellular cholesterol homeostasis. Food-derived cholesterol and cholesterol esters contained within lipoproteins are delivered to lysosomes by endocytosis. From the lysosomal lumen, cholesterol is transported to the inner surface of the lysosomal membrane through the glycocalyx; this shuttling requires Niemann-Pick C (NPC) 1 and NPC2 proteins. The lysosomal membrane proteins lysosomal-associated membrane protein (LAMP)-2 and lysosomal integral membrane protein (LIMP)-2/SCARB2 also bind cholesterol. LAMP-2 may serve as a cholesterol reservoir, whereas LIMP-2, like NPC1, is able to transport cholesterol through a transglycocalyx tunnel. Contact sites and fusion events between lysosomes and other organelles mediate the distribution of cholesterol. Lysosomal cholesterol content is sensed thereby regulating mammalian target of rapamycin complex (mTORC)-dependent signaling. This review summarizes our understanding of the major steps in cholesterol handling from the moment it enters the lysosome until it leaves this compartment.


Subject(s)
Cholesterol/metabolism , Lysosomes/metabolism , Animals , Endocytosis , Humans , Lipid Metabolism , Models, Biological , Signal Transduction
18.
EMBO J ; 39(5): e102608, 2020 03 02.
Article in English | MEDLINE | ID: mdl-31930741

ABSTRACT

Degradation of endoplasmic reticulum (ER) by selective autophagy (ER-phagy) is crucial for ER homeostasis. However, it remains unclear how ER scission is regulated for subsequent autophagosomal sequestration and lysosomal degradation. Here, we show that oligomerization of ER-phagy receptor FAM134B (also referred to as reticulophagy regulator 1 or RETREG1) through its reticulon-homology domain is required for membrane fragmentation in vitro and ER-phagy in vivo. Under ER-stress conditions, activated CAMK2B phosphorylates the reticulon-homology domain of FAM134B, which enhances FAM134B oligomerization and activity in membrane fragmentation to accommodate high demand for ER-phagy. Unexpectedly, FAM134B G216R, a variant derived from a type II hereditary sensory and autonomic neuropathy (HSAN) patient, exhibits gain-of-function defects, such as hyperactive self-association and membrane scission, which results in excessive ER-phagy and sensory neuron death. Therefore, this study reveals a mechanism of ER membrane fragmentation in ER-phagy, along with a signaling pathway in regulating ER turnover, and suggests a potential implication of excessive selective autophagy in human diseases.


Subject(s)
Autophagy , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Endoplasmic Reticulum Stress , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Signal Transduction , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Cell Membrane/metabolism , Cytokinesis/physiology , Endoplasmic Reticulum/metabolism , Gain of Function Mutation , Humans , Intracellular Signaling Peptides and Proteins/genetics , Lysosomes/metabolism , Membrane Proteins/genetics , Polymerization
19.
Science ; 366(6464): 460-467, 2019 10 25.
Article in English | MEDLINE | ID: mdl-31649195

ABSTRACT

The nucleotide oligomerization domain (NOD)-like receptors 1 and 2 (NOD1/2) are intracellular pattern-recognition proteins that activate immune signaling pathways in response to peptidoglycans associated with microorganisms. Recruitment to bacteria-containing endosomes and other intracellular membranes is required for NOD1/2 signaling, and NOD1/2 mutations that disrupt membrane localization are associated with inflammatory bowel disease and other inflammatory conditions. However, little is known about this recruitment process. We found that NOD1/2 S-palmitoylation is required for membrane recruitment and immune signaling. ZDHHC5 was identified as the palmitoyltransferase responsible for this critical posttranslational modification, and several disease-associated mutations in NOD2 were found to be associated with defective S-palmitoylation. Thus, ZDHHC5-mediated S-palmitoylation of NOD1/2 is critical for their ability to respond to peptidoglycans and to mount an effective immune response.


Subject(s)
Acyltransferases/metabolism , Lipoylation , Nod1 Signaling Adaptor Protein/chemistry , Nod2 Signaling Adaptor Protein/chemistry , Signal Transduction , Animals , Cysteine/chemistry , HCT116 Cells , HEK293 Cells , Humans , Macrophages/immunology , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Peptidoglycan , Phagosomes/immunology , Phagosomes/microbiology , Protein Processing, Post-Translational , RAW 264.7 Cells , Salmonella typhimurium
20.
Nat Commun ; 10(1): 3521, 2019 08 06.
Article in English | MEDLINE | ID: mdl-31387993

ABSTRACT

The intracellular transport of cholesterol is subject to tight regulation. The structure of the lysosomal integral membrane protein type 2 (LIMP-2, also known as SCARB2) reveals a large cavity that traverses the molecule and resembles the cavity in SR-B1 that mediates lipid transfer. The detection of cholesterol within the LIMP-2 structure and the formation of cholesterol-like inclusions in LIMP-2 knockout mice suggested the possibility that LIMP2 transports cholesterol in lysosomes. We present results of molecular modeling, crosslinking studies, microscale thermophoresis and cell-based assays that support a role of LIMP-2 in cholesterol transport. We show that the cavity in the luminal domain of LIMP-2 can bind and deliver exogenous cholesterol to the lysosomal membrane and later to lipid droplets. Depletion of LIMP-2 alters SREBP-2-mediated cholesterol regulation, as well as LDL-receptor levels. Our data indicate that LIMP-2 operates in parallel with Niemann Pick (NPC)-proteins, mediating a slower mode of lysosomal cholesterol export.


Subject(s)
CD36 Antigens/metabolism , Cholesterol, LDL/metabolism , Lysosomal Membrane Proteins/metabolism , Lysosomes/metabolism , Receptors, Scavenger/metabolism , Animals , CD36 Antigens/genetics , CHO Cells , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cricetulus , Fibroblasts , Gene Knockout Techniques , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins , Lipid Droplets/metabolism , Lysosomal Membrane Proteins/genetics , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Niemann-Pick C1 Protein , Protein Domains , RNA, Small Interfering/metabolism , Receptors, Scavenger/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...