Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
Nat Chem Biol ; 20(7): 847-856, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38167918

ABSTRACT

Pharmacological activation of voltage-gated ion channels by ligands serves as the basis for therapy and mainly involves a classic gating mechanism that augments the native voltage-dependent open probability. Through structure-based virtual screening, we identified a new scaffold compound, Ebio1, serving as a potent and subtype-selective activator for the voltage-gated potassium channel KCNQ2 and featuring a new activation mechanism. Single-channel patch-clamp, cryogenic-electron microscopy and molecular dynamic simulations, along with chemical derivatives, reveal that Ebio1 engages the KCNQ2 activation by generating an extended channel gate with a larger conductance at the saturating voltage (+50 mV). This mechanism is different from the previously observed activation mechanism of ligands on voltage-gated ion channels. Ebio1 caused S6 helices from residues S303 and F305 to perform a twist-to-open movement, which was sufficient to open the KCNQ2 gate. Overall, our findings provide mechanistic insights into the activation of KCNQ2 channel by Ebio1 and lend support for KCNQ-related drug development.


Subject(s)
Ion Channel Gating , KCNQ2 Potassium Channel , Molecular Dynamics Simulation , KCNQ2 Potassium Channel/metabolism , KCNQ2 Potassium Channel/chemistry , Humans , Ion Channel Gating/drug effects , Small Molecule Libraries/pharmacology , Small Molecule Libraries/chemistry , Animals , Patch-Clamp Techniques , Cryoelectron Microscopy , HEK293 Cells , Structure-Activity Relationship
2.
EMBO Rep ; 24(10): e56948, 2023 Oct 09.
Article in English | MEDLINE | ID: mdl-37672005

ABSTRACT

The maintenance of lysosome homeostasis is crucial for cell growth. Lysosome-dependent degradation and metabolism sustain tumor cell survival. Here, we demonstrate that CCDC50 serves as a lysophagy receptor, promoting tumor progression and invasion by controlling lysosomal integrity and renewal. CCDC50 monitors lysosomal damage, recognizes galectin-3 and K63-linked polyubiquitination on damaged lysosomes, and specifically targets them for autophagy-dependent degradation. CCDC50 deficiency causes the accumulation of ruptured lysosomes, impaired autophagic flux, and superfluous reactive oxygen species, consequently leading to cell death and tumor suppression. CCDC50 expression is associated with malignancy, progression to metastasis, and poor overall survival in human melanoma. Targeting CCDC50 suppresses tumor growth and lung metastasis, and enhances the effect of BRAFV600E inhibition. Thus, we demonstrate critical roles of CCDC50-mediated clearance of damaged lysosomes in supporting tumor growth, hereby identifying a potential therapeutic target of melanoma.

3.
Proc Natl Acad Sci U S A ; 119(45): e2207067119, 2022 11 08.
Article in English | MEDLINE | ID: mdl-36763058

ABSTRACT

The cardiac KCNQ1 potassium channel carries the important IKs current and controls the heart rhythm. Hundreds of mutations in KCNQ1 can cause life-threatening cardiac arrhythmia. Although KCNQ1 structures have been recently resolved, the structural basis for the dynamic electro-mechanical coupling, also known as the voltage sensor domain-pore domain (VSD-PD) coupling, remains largely unknown. In this study, utilizing two VSD-PD coupling enhancers, namely, the membrane lipid phosphatidylinositol 4,5-bisphosphate (PIP2) and a small-molecule ML277, we determined 2.5-3.5 Å resolution cryo-electron microscopy structures of full-length human KCNQ1-calmodulin (CaM) complex in the apo closed, ML277-bound open, and ML277-PIP2-bound open states. ML277 binds at the "elbow" pocket above the S4-S5 linker and directly induces an upward movement of the S4-S5 linker and the opening of the activation gate without affecting the C-terminal domain (CTD) of KCNQ1. PIP2 binds at the cleft between the VSD and the PD and brings a large structural rearrangement of the CTD together with the CaM to activate the PD. These findings not only elucidate the structural basis for the dynamic VSD-PD coupling process during KCNQ1 gating but also pave the way to develop new therapeutics for anti-arrhythmia.


Subject(s)
Heart , KCNQ1 Potassium Channel , Humans , KCNQ1 Potassium Channel/metabolism , Cryoelectron Microscopy , Piperidines
4.
J Med Virol ; 96(2): e29411, 2024 02.
Article in English | MEDLINE | ID: mdl-38285434

ABSTRACT

Cap RNA methylations play important roles in the replication, evasion of host RNA sensor recognition, and pathogenesis. Coronaviruses possess both guanine N7- and 2'-O-ribose methyltransferases (N7-MTase and 2'-O-MTase) encoded by nonstructural protein (nsp) 14 and nsp16/10 complex, respectively. In this study, we reconstituted the two-step RNA methylations of N7-MTase and 2'-O-MTase of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in vitro and demonstrated its common and different features in comparison with that of SARS-CoV. We revealed that the nsp16/10 2'-O-MTase of SARS-CoV-2 has a broader substrate selectivity than the counterpart of SARS-CoV and can accommodate both unmethylated and uncapped RNA substrates in a sequence-independent manner. Most intriguingly, the substrate selectivity of nsp16/10 complex is not determined by the apoenzyme of nsp16 MTase but by its cofactor nsp10. These results provide insight into the unique features of SARS-CoV-2 MTases and may help develop strategies to precisely intervene in the methylation pathway and pathogenesis of SARS-CoV-2.


Subject(s)
COVID-19 , Methyltransferases , Humans , Methyltransferases/genetics , SARS-CoV-2/genetics , RNA Methylation , RNA Caps
5.
EMBO Rep ; 23(5): e54453, 2022 05 04.
Article in English | MEDLINE | ID: mdl-35343634

ABSTRACT

The NLRP3-directed inflammasome complex is crucial for the host to resist microbial infection and monitor cellular damage. However, the hyperactivation of NLRP3 inflammasome is implicated in pathogenesis of inflammatory diseases, including inflammatory bowel disease (IBD). Autophagy and autophagy-related genes are closely linked to NLRP3-mediated inflammation in these inflammatory disorders. Here, we report that CCDC50, a novel autophagy cargo receptor, negatively regulates NLRP3 inflammasome assembly and suppresses the cleavage of pro-caspase-1 and interleukin 1ß (IL-1ß) release by delivering NLRP3 for autophagic degradation. Transcriptome analysis showed that knockdown of CCDC50 results in upregulation of signaling pathways associated with autoinflammatory diseases. CCDC50 deficiency leads to enhanced proinflammatory cytokine response triggered by a wide range of endogenous and exogenous NLRP3 stimuli. Ccdc50-deficient mice are more susceptible to dextran sulfate (DSS)-induced colitis and exhibit more severe gut inflammation with elevated NLRP3 inflammasome activity. These results illustrate the physiological significance of CCDC50 in the pathogenicity of inflammatory diseases, suggesting protective roles of CCDC50 in keeping gut inflammation under control.


Subject(s)
Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Animals , Autophagy , Dextran Sulfate/toxicity , Inflammasomes/genetics , Inflammation , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/genetics
6.
Proc Natl Acad Sci U S A ; 118(3)2021 01 19.
Article in English | MEDLINE | ID: mdl-33431678

ABSTRACT

Nuclear factor κB (NF-κB)-mediated signaling pathway plays a crucial role in the regulation of inflammatory process, innate and adaptive immune responses. The hyperactivation of inflammatory response causes host cell death, tissue damage, and autoinflammatory disorders, such as sepsis and inflammatory bowel disease. However, how these processes are precisely controlled is still poorly understood. In this study, we demonstrated that ankyrin repeat and suppressor of cytokine signaling box containing 1 (ASB1) is involved in the positive regulation of inflammatory responses by enhancing the stability of TAB2 and its downstream signaling pathways, including NF-κB and mitogen-activated protein kinase pathways. Mechanistically, unlike other members of the ASB family that induce ubiquitination-mediated degradation of their target proteins, ASB1 associates with TAB2 to inhibit K48-linked polyubiquitination and thereby promote the stability of TAB2 upon stimulation of cytokines and lipopolysaccharide (LPS), which indicates that ASB1 plays a noncanonical role to further stabilize the target protein rather than induce its degradation. The deficiency of Asb1 protects mice from Salmonella typhimurium- or LPS-induced septic shock and increases the survival of mice. Moreover, Asb1-deficient mice exhibited less severe colitis and intestinal inflammation induced by dextran sodium sulfate. Given the crucial role of ASB proteins in inflammatory signaling pathways, our study offers insights into the immune regulation in pathogen infection and inflammatory disorders with therapeutic implications.


Subject(s)
Colitis/immunology , NF-kappa B/immunology , Protein Processing, Post-Translational , Salmonella Infections/immunology , Shock, Septic/immunology , Suppressor of Cytokine Signaling Proteins/immunology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Animals , Colitis/chemically induced , Colitis/genetics , Colitis/mortality , Dextran Sulfate , Genes, Reporter , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Lipopolysaccharides , Luciferases/genetics , Luciferases/immunology , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/immunology , Mice , Mice, Knockout , NF-kappa B/genetics , Protein Binding , Salmonella Infections/genetics , Salmonella Infections/microbiology , Salmonella Infections/mortality , Salmonella typhimurium/immunology , Salmonella typhimurium/pathogenicity , Shock, Septic/chemically induced , Shock, Septic/genetics , Shock, Septic/mortality , Signal Transduction , Suppressor of Cytokine Signaling Proteins/genetics , Survival Analysis , Ubiquitination
7.
Proc Natl Acad Sci U S A ; 118(20)2021 05 18.
Article in English | MEDLINE | ID: mdl-33990467

ABSTRACT

Cardiac arrhythmias are the most common cause of sudden cardiac death worldwide. Lengthening the ventricular action potential duration (APD), either congenitally or via pathologic or pharmacologic means, predisposes to a life-threatening ventricular arrhythmia, Torsade de Pointes. IKs (KCNQ1+KCNE1), a slowly activating K+ current, plays a role in action potential repolarization. In this study, we screened a chemical library in silico by docking compounds to the voltage-sensing domain (VSD) of the IKs channel. Here, we show that C28 specifically shifted IKs VSD activation in ventricle to more negative voltages and reversed the drug-induced lengthening of APD. At the same dosage, C28 did not cause significant changes of the normal APD in either ventricle or atrium. This study provides evidence in support of a computational prediction of IKs VSD activation as a potential therapeutic approach for all forms of APD prolongation. This outcome could expand the therapeutic efficacy of a myriad of currently approved drugs that may trigger arrhythmias.


Subject(s)
Action Potentials/drug effects , KCNQ1 Potassium Channel/genetics , Myocytes, Cardiac/metabolism , Small Molecule Libraries/pharmacology , Action Potentials/physiology , Amino Acid Substitution , Animals , Arrhythmias, Cardiac/drug therapy , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/pathology , Calcium/metabolism , Dogs , Furans/pharmacology , Gene Expression , Guinea Pigs , Heart Atria/cytology , Heart Atria/metabolism , Heart Ventricles/cytology , Heart Ventricles/metabolism , Humans , KCNQ1 Potassium Channel/chemistry , KCNQ1 Potassium Channel/metabolism , Moxifloxacin/pharmacology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Oocytes/cytology , Oocytes/drug effects , Oocytes/metabolism , Patch-Clamp Techniques , Phenethylamines/pharmacology , Potassium/metabolism , Primary Cell Culture , Pyridines/pharmacology , Pyrimidines/pharmacology , Sodium/metabolism , Sulfonamides/pharmacology , Transgenes , Xenopus laevis
8.
Pharmacol Res ; 192: 106765, 2023 06.
Article in English | MEDLINE | ID: mdl-37075871

ABSTRACT

Cardiovascular disease (CVD) remains the leading cause of morbidity and mortality, imposing an increasing global health burden. Cardiac ion channels (voltage-gated NaV, CaV, KVs, and others) synergistically shape the cardiac action potential (AP) and control the heartbeat. Dysfunction of these channels, due to genetic mutations, transcriptional or post-translational modifications, may disturb the AP and lead to arrhythmia, a major risk for CVD patients. Although there are five classes of anti-arrhythmic drugs available, they can have varying levels of efficacies and side effects on patients, possibly due to the complex pathogenesis of arrhythmias. As an alternative treatment option, Chinese herbal remedies have shown promise in regulating cardiac ion channels and providing anti-arrhythmic effects. In this review, we first discuss the role of cardiac ion channels in maintaining normal heart function and the pathogenesis of CVD, then summarize the classification of Chinese herbal compounds, and elaborate detailed mechanisms of their efficacy in regulating cardiac ion channels and in alleviating arrhythmia and CVD. We also address current limitations and opportunities for developing new anti-CVD drugs based on Chinese herbal medicines.


Subject(s)
Cardiovascular Diseases , Drugs, Chinese Herbal , Humans , Anti-Arrhythmia Agents/therapeutic use , Cardiovascular Diseases/drug therapy , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , Ion Channels/physiology , Arrhythmias, Cardiac/drug therapy
9.
Circ Res ; 124(4): 539-552, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30566038

ABSTRACT

RATIONALE: Mutations in the SCN5A gene, encoding the α subunit of the Nav1.5 channel, cause a life-threatening form of cardiac arrhythmia, long QT syndrome type 3 (LQT3). Mexiletine, which is structurally related to the Na+ channel-blocking anesthetic lidocaine, is used to treat LQT3 patients. However, the patient response is variable, depending on the genetic mutation in SCN5A. OBJECTIVE: The goal of this study is to understand the molecular basis of patients' variable responses and build a predictive statistical model that can be used to personalize mexiletine treatment based on patient's genetic variant. METHODS AND RESULTS: We monitored the cardiac Na+ channel voltage-sensing domain (VSD) conformational dynamics simultaneously with other gating properties for the LQT3 variants. To systematically identify the relationship between mexiletine block and channel biophysical properties, we used a system-based statistical modeling approach to connect the multivariate properties to patient phenotype. We found that mexiletine altered the conformation of the Domain III VSD, which is the same VSD that many tested LQT3 mutations affect. Analysis of 15 LQT3 variants showed a strong correlation between the activation of the Domain III-VSD and the strength of the inhibition of the channel by mexiletine. Based on this improved molecular-level understanding, we generated a systems-based model based on a dataset of 32 LQT3 patients, which then successfully predicted the response of 7 out of 8 patients to mexiletine in a blinded, retrospective trial. CONCLUSIONS: Our results imply that the modulated receptor theory of local anesthetic action, which confines local anesthetic binding effects to the channel pore, should be revised to include drug interaction with the Domain III-VSD. Using an algorithm that incorporates this mode of action, we can predict patient-specific responses to mexiletine, improving therapeutic decision making.


Subject(s)
Anti-Arrhythmia Agents/therapeutic use , Long QT Syndrome/genetics , Mexiletine/therapeutic use , NAV1.5 Voltage-Gated Sodium Channel/genetics , Pharmacogenomic Variants , Sodium Channel Blockers/therapeutic use , Adolescent , Adult , Animals , Anti-Arrhythmia Agents/pharmacology , Female , HEK293 Cells , Humans , Ion Channel Gating , Long QT Syndrome/drug therapy , Male , Mexiletine/pharmacology , Mutation, Missense , NAV1.5 Voltage-Gated Sodium Channel/chemistry , NAV1.5 Voltage-Gated Sodium Channel/metabolism , Sodium Channel Blockers/pharmacology , Xenopus
10.
J Environ Sci (China) ; 81: 4-16, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30975328

ABSTRACT

Core-shell magnetic seeds with certain adsorption capacity that were prepared by sulfated roasting, served as the core of a magnetic separation technology for purification of starch wastewater. XRD and SEM results indicate that magnetite's surface transformed to be porous α-Fe2O3 structure. Compared with magnetite particles, the specific surface area was significantly improved to be 8.361 from 2.591 m2/g, with little decrease in specific susceptibility. Zeta potential, FT-IR and XPS experiments indicate that both phosphate and starch adsorbed on the surface of the core-shell magnetic seeds by chemical adsorption, which fits well with the Langmuir adsorption model. The porous surface structure of magnetic seeds significantly contributes to the adsorption of phosphate and starch species, which can be efficiently removed to be 1.51 mg/L (phosphate) and 9.51 mg/L (starch) using magnetic separation.


Subject(s)
Phosphorus/analysis , Starch/analysis , Waste Disposal, Fluid/methods , Wastewater/chemistry , Water Pollutants, Chemical/analysis , Ferrosoferric Oxide/chemistry , Magnetics , Sulfates/chemistry
11.
J Cell Biochem ; 118(11): 4020-4032, 2017 11.
Article in English | MEDLINE | ID: mdl-28422315

ABSTRACT

Positive transcription elongation factor-b (P-TEFb) is required for the release of RNA polymerase II (RNAPII) from its pause near the gene promoters and thus for efficient proceeding to the transcription elongation. It consists of two core subunits-CDK9 and one of T-typed or K-typed cyclin, of which, cyclin T1/CDK9 is the major and most studied combination. We have previously identified a novel splice variant of cyclin T1, cyclin T1b, which negatively regulates the transcription elongation of HIV-1 genes as well as several host genes. In this study, we revealed the serine-arginine-rich protein, ASF/SF2, as a regulatory factor of the alternative splicing of cyclin T1 gene. ASF/SF2 promotes the production of cyclin T1b versus cyclin T1a and regulates the expression of cyclin T1-depedent genes at the transcription level. We further found that a cis-element on exon 8 is responsible for the skipping of exon 7 mediated by ASF/SF2. Collectively, ASF/SF2 is identified as a splicing regulator of cyclin T1, which contributes to the control of the subsequent transcription events. J. Cell. Biochem. 118: 4020-4032, 2017. © 2017 Wiley Periodicals, Inc.


Subject(s)
Alternative Splicing/physiology , Cyclin T/biosynthesis , Serine-Arginine Splicing Factors/metabolism , Cell Line , Cyclin T/genetics , Humans , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , Serine-Arginine Splicing Factors/genetics
12.
Retrovirology ; 14(1): 51, 2017 Nov 15.
Article in English | MEDLINE | ID: mdl-29141633

ABSTRACT

BACKGROUND: The CRISPR/Cas9 system has been widely used for genome editing in mammalian cells. CXCR4 is a co-receptor for human immunodeficiency virus type 1 (HIV-1) entry, and loss of CXCR4 function can protect cells from CXCR4 (X4)-tropic HIV-1 infection, making CXCR4 an important target for HIV-1 gene therapy. However, the large size of the CRISPR/SpCas9 system presents an obstacle to its efficient delivery into primary CD4+ T cells. Recently, a small Staphylococcus aureus Cas9 (SaCas9) has been developed as a genome editing tool can address this question. Therefore, it provides a promising strategy for HIV-1 gene therapy if it is used to target CXCR4. RESULTS: Here, we employed a short version of Cas9 from Staphylococcus aureus (SaCas9) for targeting CXCR4. We demonstrated that transduction of lenti-virus expressing SaCas9 and selected single-guided RNAs of CXCR4 in human CD4+ T cell lines efficiently induced the editing of the CXCR4 gene, making these cell lines resistant to X4-tropic HIV-1 infection. Moreover, we efficiently transduced primary human CD4+ T cells using adeno-associated virus-delivered CRISPR/SaCas9 and disrupted CXCR4 expression. We also showed that CXCR4-edited primary CD4+ T cells proliferated normally and were resistant to HIV-1 infection. CONCLUSIONS: Our study provides a basis for possible application of CXCR4-targeted genome editing by CRISPR/SaCas9 in HIV-1 gene therapy.


Subject(s)
CD4-Positive T-Lymphocytes/virology , CRISPR-Cas Systems/genetics , Disease Resistance/genetics , Gene Editing/methods , HIV Infections/genetics , Receptors, CXCR4/genetics , Staphylococcus aureus/enzymology , CD4-Positive T-Lymphocytes/metabolism , Cells, Cultured , Endonucleases/metabolism , Gene Expression Regulation , Gene Knockout Techniques , HEK293 Cells , HIV Infections/metabolism , HIV Infections/virology , HIV-1 , Host-Pathogen Interactions/genetics , Humans , Jurkat Cells , Receptors, CXCR4/metabolism
13.
J Biol Chem ; 289(24): 16914-23, 2014 Jun 13.
Article in English | MEDLINE | ID: mdl-24764303

ABSTRACT

Large conductance Ca(2+)- and voltage-activated potassium (BK) channels, composed of pore-forming α subunits and auxiliary ß subunits, play important roles in diverse physiological activities. The ß1 is predominately expressed in smooth muscle cells, where it greatly enhances the Ca(2+) sensitivity of BK channels for proper regulation of smooth muscle tone. However, the structural basis underlying dynamic interaction between BK mSlo1 α and ß1 remains elusive. Using macroscopic ionic current recordings in various Ca(2+) and Mg(2+) concentrations, we identified two binding sites on the cytosolic N terminus of ß1, namely the electrostatic enhancing site (mSlo1(K392,R393)-ß1(E13,T14)), increasing the calcium sensitivity of BK channels, and the hydrophobic site (mSlo1(L906,L908)-ß1(L5,V6,M7)), passing the physical force from the Ca(2+) bowl onto the enhancing site and S6 C-linker. Dynamic binding of these sites affects the interaction between the cytosolic domain and voltage-sensing domain, leading to the reduction of Mg(2+) sensitivity. A comprehensive structural model of the BK(mSlo1 α-ß1) complex was reconstructed based on these functional studies, which provides structural and mechanistic insights for understanding BK gating.


Subject(s)
Calcium/metabolism , Ion Channel Gating , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Magnesium/metabolism , Action Potentials , Amino Acid Sequence , Binding Sites , HEK293 Cells , Humans , Ice , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/chemistry , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/genetics , Molecular Dynamics Simulation , Molecular Sequence Data , Protein Binding , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism
14.
Biochem Biophys Res Commun ; 459(2): 270-276, 2015 Apr 03.
Article in English | MEDLINE | ID: mdl-25732088

ABSTRACT

Eukaryotic cellular and most viral RNAs carry a 5'-terminal cap structure, a 5'-5' triphosphate linkage between the 5' end of the RNA and a guanosine nucleotide (cap-0). SARS coronavirus (SARS-CoV) nonstructural protein nsp16 functions as a methyltransferase, to methylate mRNA cap-0 structure at the ribose 2'-O position of the first nucleotide to form cap-1 structures. However, whether there is interplay between nsp16 and host proteins was not yet clear. In this report, we identified several potential cellular nsp16-interacting proteins from a human thymus cDNA library by yeast two-hybrid screening. VHL, one of these proteins, was proven to interact with nsp16 both in vitro and in vivo. Further studies showed that VHL can inhibit SARS-CoV replication by regulating nsp16 ubiquitination and promoting its degradation. Our results have revealed the role of cellular VHL in the regulation of SARS-CoV replication.


Subject(s)
Methyltransferases/metabolism , Severe acute respiratory syndrome-related coronavirus/physiology , Viral Nonstructural Proteins/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , Animals , Chlorocebus aethiops , Exoribonucleases/genetics , Exoribonucleases/metabolism , Host-Pathogen Interactions/physiology , Humans , Methyltransferases/chemistry , Methyltransferases/genetics , Protein Stability , Proteolysis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Severe acute respiratory syndrome-related coronavirus/genetics , Two-Hybrid System Techniques , Ubiquitination , Vero Cells , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/genetics , Virus Replication/physiology , Von Hippel-Lindau Tumor Suppressor Protein/genetics
15.
bioRxiv ; 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-39005479

ABSTRACT

The cardiac KCNQ1+KCNE1 (I Ks ) channel regulates heart rhythm in both normal and stress conditions. Under stress, the ß-adrenergic stimulation elevates the intracellular cAMP level, leading to KCNQ1 phosphorylation by protein kinase A and increased I Ks , which shortens action potentials to adapt to accelerated heart rate. An impaired response to the ß-adrenergic stimulation due to KCNQ1 mutations is associated with the occurrence of a lethal congenital long QT syndrome (type 1, also known as LQT1). However, the underlying mechanism of ß-adrenergic stimulation of I Ks remains unclear, impeding the development of new therapeutics. Here we find that the unique properties of KCNQ1 channel gating with two distinct open states are key to this mechanism. KCNQ1's fully activated open (AO) state is more sensitive to cAMP than its' intermediate open (IO) state. By enhancing the AO state occupancy, the small molecules ML277 and C28 are found to effectively enhance the cAMP sensitivity of the KCNQ1 channel, independent of KCNE1 association. This finding of enhancing AO state occupancy leads to a potential novel strategy to rescue the response of I Ks to ß-adrenergic stimulation in LQT1 mutants. The success of this approach is demonstrated in cardiac myocytes and also in a high-risk LQT1 mutation. In conclusion the present study not only uncovers the key role of the AO state in I Ks channel phosphorylation, but also provides a new target for anti-arrhythmic strategy. Significance statement: The increase of I Ks potassium currents with adrenalin stimulation is important for "fight-or-flight" responses. Mutations of the IKs channel reducing adrenalin responses are associated with more lethal form of the type-1 long-QT syndrome (LQT). The alpha subunit of the IKs channel, KCNQ1 opens in two distinct open states, the intermediate-open (IO) and activated-open (AO) states, following a two-step voltage sensing domain (VSD) activation process. We found that the AO state, but not the IO state, is responsible for the adrenalin response. Modulators that specifically enhance the AO state occupancy can enhance adrenalin responses of the WT and LQT-associated mutant channels. These results reveal a mechanism of state dependent modulation of ion channels and provide an anti-arrhythmic strategy.

16.
Autophagy ; 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38869076

ABSTRACT

Protein aggregation caused by the disruption of proteostasis will lead to cellular cytotoxicity and even cell death, which is implicated in multiple neurodegenerative diseases. The elimination of aggregated proteins is mediated by selective macroautophagy receptors, which is termed aggrephagy. However, the identity and redundancy of aggrephagy receptors in recognizing substrates remain largely unexplored. Here, we find that CCDC50, a highly expressed autophagy receptor in brain, is recruited to proteotoxic stresses-induced polyubiquitinated protein aggregates and ectopically expressed aggregation-prone proteins. CCDC50 recognizes and further clears these cytotoxic aggregates through autophagy. The ectopic expression of CCDC50 increases the tolerance to stress-induced proteotoxicity and hence improved cell survival in neuron cells, whereas CCDC50 deficiency caused accumulation of lipid deposits and polyubiquitinated protein conjugates in the brain of one-year-old mice. Our study illustrates how aggrephagy receptor CCDC50 combats proteotoxic stress for the benefit of neuronal cell survival, thus suggesting a protective role in neurotoxic proteinopathy.

17.
Autophagy ; 19(1): 365-366, 2023 01.
Article in English | MEDLINE | ID: mdl-35620989

ABSTRACT

The assembly of the NLRP3 inflammasome can be initiated by a wide range of stimuli including exogenous infection as well as endogenous damage. Therefore, the tight regulation of the NLRP3 inflammasome is crucial for the host to resist microbial invasion and maintain homeostasis. Our recent work has identified a negative regulator of NLRP3-mediated inflammation, namely CCDC50 (coiled-coil domain containing protein 50). CCDC50 can be induced by NLRP3 agonists and then functions as a macroautophagy/autophagy cargo receptor to recognize K63-polyubiquitinated NLRP3 and deliver it to MAP1LC3/LC3-conjugated phagophores for degradation. CCDC50 inhibits the polymerization of NLRP3 and the recruitment of PYCARD/ASC, consequently suppressing the assembly of inflammasomes. ccdc50-knockout mice are more susceptible to dextran-sulfate (DSS)-induced colitis and exhibit more severe gut inflammation with elevated NLRP3 inflammasome activity, suggesting a protective role of CCDC50 in the pathology and progression of inflammatory bowel disease (IBD). Our finding reveals a function of autophagy-related proteins in the regulation of NLRP3-mediated inflammation, thus demonstrating the intricate crosstalk between autophagy and inflammation.


Subject(s)
Colitis , Inflammasomes , Intracellular Signaling Peptides and Proteins , NLR Family, Pyrin Domain-Containing 3 Protein , Animals , Mice , Autophagy , Colitis/chemically induced , Colitis/metabolism , Inflammasomes/metabolism , Inflammation/metabolism , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Intracellular Signaling Peptides and Proteins/metabolism
18.
Nat Commun ; 14(1): 6632, 2023 10 19.
Article in English | MEDLINE | ID: mdl-37857637

ABSTRACT

The human voltage-gated potassium channel KCNQ2/KCNQ3 carries the neuronal M-current, which helps to stabilize the membrane potential. KCNQ2 can be activated by analgesics and antiepileptic drugs but their activation mechanisms remain unclear. Here we report cryo-electron microscopy (cryo-EM) structures of human KCNQ2-CaM in complex with three activators, namely the antiepileptic drug cannabidiol (CBD), the lipid phosphatidylinositol 4,5-bisphosphate (PIP2), and HN37 (pynegabine), an antiepileptic drug in the clinical trial, in an either closed or open conformation. The activator-bound structures, along with electrophysiology analyses, reveal the binding modes of two CBD, one PIP2, and two HN37 molecules in each KCNQ2 subunit, and elucidate their activation mechanisms on the KCNQ2 channel. These structures may guide the development of antiepileptic drugs and analgesics that target KCNQ2.


Subject(s)
Analgesics , Anticonvulsants , Humans , Anticonvulsants/pharmacology , Cryoelectron Microscopy , Ligands , Membrane Potentials , KCNQ2 Potassium Channel/chemistry , KCNQ2 Potassium Channel/metabolism , KCNQ3 Potassium Channel/metabolism
19.
Cancer Res ; 82(18): 3359-3374, 2022 Sep 16.
Article in English | MEDLINE | ID: mdl-35792620

ABSTRACT

Glioblastoma (GBM) is among the most aggressive human cancers. Although oncolytic virus (OV) therapy has been proposed as a potential approach to treat GBM, it frequently fails because GBM cells are usually nonpermissive to OV. Here, we describe a dual-step drug screen for identifying chemical enhancers of OV in GBM. From a high-throughput screen of 1416 FDA-approved drugs, an inhibitor of CDK4/6 was identified as the top enhancer, selectively increasing potency of two OV strains, VSVΔ51 and Zika virus. Mechanistically, CDK4/6 inhibition promoted autophagic degradation of MAVS, resulting in impaired antiviral responses and enhanced tumor-selective replication of VSVΔ51 in vitro and in vivo. CDK4/6 inhibition cooperated with VSVΔ51 to induce severe DNA damage stress and amplify oncolysis. In GBM xenograft models, combined treatment with CDK4/6 inhibitor and VSVΔ51 significantly inhibited tumor growth and prolonged the survival of tumor-bearing mice. Further investigation revealed that CDK4/6 inhibitor and VSVΔ51 synergistically induced immunogenic cell death and boosted antitumor immunity. Together, this study features a promising approach of treating aggressive GBM through the combination of CDK4/6 inhibitor with OV. SIGNIFICANCE: This study proposes inhibition of cyclin-dependent kinases as a clinically translatable combinatorial strategy to enhance the efficacy of oncolytic virotherapy in GBM.


Subject(s)
Brain Neoplasms , Glioblastoma , Oncolytic Virotherapy , Oncolytic Viruses , Zika Virus Infection , Zika Virus , Animals , Antiviral Agents , Brain Neoplasms/metabolism , Cell Death , Cell Line, Tumor , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinases , Glioblastoma/pathology , Humans , Mice , Oncolytic Virotherapy/methods , T-Lymphocytes/metabolism , Xenograft Model Antitumor Assays , Zika Virus Infection/drug therapy
20.
Commun Biol ; 5(1): 301, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35365746

ABSTRACT

Loss-of-function mutations in Kv7.1 often lead to long QT syndrome (LQTS), a cardiac repolarization disorder associated with arrhythmia and subsequent sudden cardiac death. The discovery of agonistic IKs modulators may offer a new potential strategy in pharmacological treatment of this disorder. The benzodiazepine derivative (R)-L3 potently activates Kv7.1 channels and shortens action potential duration, thus may represent a starting point for drug development. However, the molecular mechanisms underlying modulation by (R)-L3 are still unknown. By combining alanine scanning mutagenesis, non-canonical amino acid incorporation, voltage-clamp electrophysiology and fluorometry, and in silico protein modelling, we show that (R)-L3 not only stimulates currents by allosteric modulation of the pore domain but also alters the kinetics independently from the pore domain effects. We identify novel (R)-L3-interacting key residues in the lower S4-segment of Kv7.1 and observed an uncoupling of the outer S4 segment with the inner S5, S6 and selectivity filter segments.


Subject(s)
Benzodiazepines , Ion Channel Gating , Benzodiazepines/pharmacology , Mutation
SELECTION OF CITATIONS
SEARCH DETAIL