Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.849
Filter
Add more filters

Publication year range
1.
Cell ; 187(6): 1460-1475.e20, 2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38428423

ABSTRACT

Apelin is a key hormone in cardiovascular homeostasis that activates the apelin receptor (APLNR), which is regarded as a promising therapeutic target for cardiovascular disease. However, adverse effects through the ß-arrestin pathway limit its pharmacological use. Here, we report cryoelectron microscopy (cryo-EM) structures of APLNR-Gi1 complexes bound to three agonists with divergent signaling profiles. Combined with functional assays, we have identified "twin hotspots" in APLNR as key determinants for signaling bias, guiding the rational design of two exclusive G-protein-biased agonists WN353 and WN561. Cryo-EM structures of WN353- and WN561-stimulated APLNR-G protein complexes further confirm that the designed ligands adopt the desired poses. Pathophysiological experiments have provided evidence that WN561 demonstrates superior therapeutic effects against cardiac hypertrophy and reduced adverse effects compared with the established APLNR agonists. In summary, our designed APLNR modulator may facilitate the development of next-generation cardiovascular medications.


Subject(s)
Apelin Receptors , Cardiovascular Agents , Drug Design , Apelin Receptors/agonists , Apelin Receptors/chemistry , Apelin Receptors/ultrastructure , Cryoelectron Microscopy , GTP-Binding Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Humans , Cardiovascular Agents/chemistry
2.
Cell ; 186(26): 5784-5797.e17, 2023 12 21.
Article in English | MEDLINE | ID: mdl-38101408

ABSTRACT

Cannabis activates the cannabinoid receptor 1 (CB1), which elicits analgesic and emotion regulation benefits, along with adverse effects, via Gi and ß-arrestin signaling pathways. However, the lack of understanding of the mechanism of ß-arrestin-1 (ßarr1) coupling and signaling bias has hindered drug development targeting CB1. Here, we present the high-resolution cryo-electron microscopy structure of CB1-ßarr1 complex bound to the synthetic cannabinoid MDMB-Fubinaca (FUB), revealing notable differences in the transducer pocket and ligand-binding site compared with the Gi protein complex. ßarr1 occupies a wider transducer pocket promoting substantial outward movement of the TM6 and distinctive twin toggle switch rearrangements, whereas FUB adopts a different pose, inserting more deeply than the Gi-coupled state, suggesting the allosteric correlation between the orthosteric binding pocket and the partner protein site. Taken together, our findings unravel the molecular mechanism of signaling bias toward CB1, facilitating the development of CB1 agonists.


Subject(s)
Arrestin , Receptor, Cannabinoid, CB1 , Signal Transduction , Arrestin/metabolism , beta-Arrestin 1/metabolism , beta-Arrestins/metabolism , Cryoelectron Microscopy , Receptor, Cannabinoid, CB1/metabolism , Humans , Animals , Cell Line
3.
Cell ; 184(4): 931-942.e18, 2021 02 18.
Article in English | MEDLINE | ID: mdl-33571431

ABSTRACT

The D1- and D2-dopamine receptors (D1R and D2R), which signal through Gs and Gi, respectively, represent the principal stimulatory and inhibitory dopamine receptors in the central nervous system. D1R and D2R also represent the main therapeutic targets for Parkinson's disease, schizophrenia, and many other neuropsychiatric disorders, and insight into their signaling is essential for understanding both therapeutic and side effects of dopaminergic drugs. Here, we report four cryoelectron microscopy (cryo-EM) structures of D1R-Gs and D2R-Gi signaling complexes with selective and non-selective dopamine agonists, including two currently used anti-Parkinson's disease drugs, apomorphine and bromocriptine. These structures, together with mutagenesis studies, reveal the conserved binding mode of dopamine agonists, the unique pocket topology underlying ligand selectivity, the conformational changes in receptor activation, and potential structural determinants for G protein-coupling selectivity. These results provide both a molecular understanding of dopamine signaling and multiple structural templates for drug design targeting the dopaminergic system.


Subject(s)
Receptors, Dopamine D1/chemistry , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D2/chemistry , Receptors, Dopamine D2/metabolism , Signal Transduction , 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine/analogs & derivatives , 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine/pharmacology , Amino Acid Sequence , Conserved Sequence , Cryoelectron Microscopy , Cyclic AMP/metabolism , GTP-Binding Proteins/metabolism , HEK293 Cells , Humans , Ligands , Models, Molecular , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Receptors, Adrenergic, beta-2/metabolism , Receptors, Dopamine D1/ultrastructure , Receptors, Dopamine D2/ultrastructure , Structural Homology, Protein
4.
Mol Cell ; 84(3): 570-583.e7, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38215752

ABSTRACT

Adhesion G protein-coupled receptors (aGPCRs) are evolutionarily ancient receptors involved in a variety of physiological and pathophysiological processes. Modulators of aGPCR, particularly antagonists, hold therapeutic promise for diseases like cancer and immune and neurological disorders. Hindered by the inactive state structural information, our understanding of antagonist development and aGPCR activation faces challenges. Here, we report the cryo-electron microscopy structures of human CD97, a prototypical aGPCR that plays crucial roles in immune system, in its inactive apo and G13-bound fully active states. Compared with other family GPCRs, CD97 adopts a compact inactive conformation with a constrained ligand pocket. Activation induces significant conformational changes for both extracellular and intracellular sides, creating larger cavities for Stachel sequence binding and G13 engagement. Integrated with functional and metadynamics analyses, our study provides significant mechanistic insights into the activation and signaling of aGPCRs, paving the way for future drug discovery efforts.


Subject(s)
Antigens, CD , Receptors, G-Protein-Coupled , Signal Transduction , Humans , Cell Adhesion , Cryoelectron Microscopy , Platelet Glycoprotein GPIb-IX Complex , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/metabolism , Antigens, CD/chemistry , Antigens, CD/metabolism
5.
Development ; 151(6)2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38546043

ABSTRACT

The timely degradation of proteins that regulate the cell cycle is essential for oocyte maturation. Oocytes are equipped to degrade proteins via the ubiquitin-proteasome system. In meiosis, anaphase promoting complex/cyclosome (APC/C), an E3 ubiquitin-ligase, is responsible for the degradation of proteins. Ubiquitin-conjugating enzyme E2 S (UBE2S), an E2 ubiquitin-conjugating enzyme, delivers ubiquitin to APC/C. APC/C has been extensively studied, but the functions of UBE2S in oocyte maturation and mouse fertility are not clear. In this study, we used Ube2s knockout mice to explore the role of UBE2S in mouse oocytes. Ube2s-deleted oocytes were characterized by meiosis I arrest with normal spindle assembly and spindle assembly checkpoint dynamics. However, the absence of UBE2S affected the activity of APC/C. Cyclin B1 and securin are two substrates of APC/C, and their levels were consistently high, resulting in the failure of homologous chromosome separation. Unexpectedly, the oocytes arrested in meiosis I could be fertilized and the embryos could become implanted normally, but died before embryonic day 10.5. In conclusion, our findings reveal an indispensable regulatory role of UBE2S in mouse oocyte meiosis and female fertility.


Subject(s)
M Phase Cell Cycle Checkpoints , Meiosis , Animals , Female , Mice , Anaphase-Promoting Complex-Cyclosome/genetics , Anaphase-Promoting Complex-Cyclosome/metabolism , Oocytes/metabolism , Ubiquitins/metabolism
6.
Nature ; 592(7854): 469-473, 2021 04.
Article in English | MEDLINE | ID: mdl-33762731

ABSTRACT

Serotonin, or 5-hydroxytryptamine (5-HT), is an important neurotransmitter1,2 that activates the largest subtype family of G-protein-coupled receptors3. Drugs that target 5-HT1A, 5-HT1D, 5-HT1E and other 5-HT receptors are used to treat numerous disorders4. 5-HT receptors have high levels of basal activity and are subject to regulation by lipids, but the structural basis for the lipid regulation and basal activation of these receptors and the pan-agonism of 5-HT remains unclear. Here we report five structures of 5-HT receptor-G-protein complexes: 5-HT1A in the apo state, bound to 5-HT or bound to the antipsychotic drug aripiprazole; 5-HT1D bound to 5-HT; and 5-HT1E in complex with a 5-HT1E- and 5-HT1F-selective agonist, BRL-54443. Notably, the phospholipid phosphatidylinositol 4-phosphate is present at the G-protein-5-HT1A interface, and is able to increase 5-HT1A-mediated G-protein activity. The receptor transmembrane domain is surrounded by cholesterol molecules-particularly in the case of 5-HT1A, in which cholesterol molecules are directly involved in shaping the ligand-binding pocket that determines the specificity for aripiprazol. Within the ligand-binding pocket of apo-5-HT1A are structured water molecules that mimic 5-HT to activate the receptor. Together, our results address a long-standing question of how lipids and water molecules regulate G-protein-coupled receptors, reveal how 5-HT acts as a pan-agonist, and identify the determinants of drug recognition in 5-HT receptors.


Subject(s)
Cryoelectron Microscopy , Ligands , Lipids , Receptors, Serotonin, 5-HT1/metabolism , Receptors, Serotonin, 5-HT1/ultrastructure , Apoproteins/chemistry , Apoproteins/metabolism , Apoproteins/ultrastructure , Aripiprazole/metabolism , Aripiprazole/pharmacology , Binding Sites , Cholesterol/pharmacology , Heterotrimeric GTP-Binding Proteins/chemistry , Heterotrimeric GTP-Binding Proteins/metabolism , Heterotrimeric GTP-Binding Proteins/ultrastructure , Humans , Models, Molecular , Phosphatidylinositol Phosphates/chemistry , Phosphatidylinositol Phosphates/metabolism , Phosphatidylinositol Phosphates/pharmacology , Receptor, Serotonin, 5-HT1A/chemistry , Receptor, Serotonin, 5-HT1A/metabolism , Receptor, Serotonin, 5-HT1A/ultrastructure , Receptors, Serotonin, 5-HT1/chemistry , Serotonin 5-HT1 Receptor Agonists/chemistry , Serotonin 5-HT1 Receptor Agonists/metabolism , Serotonin 5-HT1 Receptor Agonists/pharmacology , Water/chemistry
7.
Proc Natl Acad Sci U S A ; 121(35): e2405845121, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39178231

ABSTRACT

Atherosclerosis is a chronic inflammatory disease of the arterial wall characterized by the accumulation of cholesterol-rich lipoproteins in macrophages. How macrophages commit to proinflammatory polarization under atherosclerosis conditions is not clear. Report here that the level of a circulating protein, leucine-rich alpha-2 glycoprotein 1 (LRG1), is elevated in the atherosclerotic tissue and serum samples from patients with coronary artery disease (CAD). LRG1 stimulated macrophages to proinflammatory M1-like polarization through the activation of extracellular signal-regulated kinase 1/2 (ERK1/2) and c-Jun N-terminal kinase (JNK) pathways. The LRG1 knockout mice showed significantly delayed atherogenesis progression and reduced levels of macrophage-related proinflammatory cytokines in a high-fat diet-induced Apoe-/- mouse atherosclerosis model. An anti-LRG1 neutralizing antibody also effectively blocked LRG1-induced macrophage M1-like polarization in vitro and conferred therapeutic benefits to animals with ApoE deficiency-induced atherosclerosis. LRG1 may therefore serve as an additional biomarker for CAD and targeting LRG1 could offer a potential therapeutic strategy for CAD patients by mitigating the proinflammatory response of macrophages.


Subject(s)
Atherosclerosis , Glycoproteins , Macrophages , Animals , Atherosclerosis/pathology , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/immunology , Macrophages/metabolism , Macrophages/immunology , Mice , Humans , Glycoproteins/metabolism , Glycoproteins/genetics , Mice, Knockout , Male , Apolipoproteins E/genetics , Apolipoproteins E/deficiency , Apolipoproteins E/metabolism , Disease Models, Animal , Cytokines/metabolism , Diet, High-Fat/adverse effects , Mice, Inbred C57BL , Coronary Artery Disease/pathology , Coronary Artery Disease/genetics , Coronary Artery Disease/metabolism , Coronary Artery Disease/immunology , Female , Mice, Knockout, ApoE , Macrophage Activation
8.
PLoS Pathog ; 20(8): e1012448, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39146384

ABSTRACT

The chemokine co-receptors CXCR4 and CCR5 mediate HIV entry and signal transduction necessary for viral infection. However, to date only the CCR5 antagonist maraviroc is approved for treating HIV-1 infection. Given that approximately 50% of late-stage HIV patients also develop CXCR4-tropic virus, clinical anti-HIV CXCR4 antagonists are needed. Here, we describe a novel allosteric CXCR4 antagonist TIQ-15 which inhibits CXCR4-tropic HIV-1 infection of primary and transformed CD4 T cells. TIQ-15 blocks HIV entry with an IC50 of 13 nM. TIQ-15 also inhibits SDF-1α/CXCR4-mediated cAMP production, cofilin activation, and chemotactic signaling. In addition, TIQ-15 induces CXCR4 receptor internalization without affecting the levels of the CD4 receptor, suggesting that TIQ-15 may act through a novel allosteric site on CXCR4 for blocking HIV entry. Furthermore, TIQ-15 did not inhibit VSV-G pseudotyped HIV-1 infection, demonstrating its specificity in blocking CXCR4-tropic virus entry, but not CXCR4-independent endocytosis or post-entry steps. When tested against a panel of clinical isolates, TIQ-15 showed potent inhibition against CXCR4-tropic and dual-tropic viruses, and moderate inhibition against CCR5-tropic isolates. This observation was followed by a co-dosing study with maraviroc, and TIQ-15 demonstrated synergistic activity. In summary, here we describe a novel HIV-1 entry inhibitor, TIQ-15, which potently inhibits CXCR4-tropic viruses while possessing low-level synergistic activities against CCR5-tropic viruses. TIQ-15 could potentially be co-dosed with the CCR5 inhibitor maraviroc to block viruses of mixed tropisms.

9.
Plant Cell ; 35(7): 2570-2591, 2023 06 26.
Article in English | MEDLINE | ID: mdl-37040621

ABSTRACT

SALT OVERLY SENSITIVE1 (SOS1) is a key component of plant salt tolerance. However, how SOS1 transcription is dynamically regulated in plant response to different salinity conditions remains elusive. Here, we report that C-type Cyclin1;1 (CycC1;1) negatively regulates salt tolerance by interfering with WRKY75-mediated transcriptional activation of SOS1 in Arabidopsis (Arabidopsis thaliana). Disruption of CycC1;1 promotes SOS1 expression and salt tolerance in Arabidopsis because CycC1;1 interferes with RNA polymerase II recruitment by occupying the SOS1 promoter. Enhanced salt tolerance of the cycc1;1 mutant was completely compromised by an SOS1 mutation. Moreover, CycC1;1 physically interacts with the transcription factor WRKY75, which can bind to the SOS1 promoter and activate SOS1 expression. In contrast to the cycc1;1 mutant, the wrky75 mutant has attenuated SOS1 expression and salt tolerance, whereas overexpression of SOS1 rescues the salt sensitivity of wrky75. Intriguingly, CycC1;1 inhibits WRKY75-mediated transcriptional activation of SOS1 via their interaction. Thus, increased SOS1 expression and salt tolerance in cycc1;1 were abolished by WRKY75 mutation. Our findings demonstrate that CycC1;1 forms a complex with WRKY75 to inactivate SOS1 transcription under low salinity conditions. By contrast, under high salinity conditions, SOS1 transcription and plant salt tolerance are activated at least partially by increased WRKY75 expression but decreased CycC1;1 expression.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Arabidopsis/metabolism , Salt Tolerance/genetics , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Gene Expression Regulation, Plant/genetics , Sodium-Hydrogen Exchangers/genetics , Sodium-Hydrogen Exchangers/metabolism
10.
Bioinformatics ; 40(8)2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39128019

ABSTRACT

MOTIVATION: High-throughput technologies yield a broad spectrum of multi-omics datasets, which offer unparalleled insights into complex biological systems. However, effectively analyzing this diverse array of data presents challenges, considering factors such as species diversity, data types, costs, and limitations of the available tools. RESULTS: Herein, we present ExpOmics, a comprehensive web platform featuring 7 applications and 4 toolkits, with 28 customizable analysis functions spanning various analyses of differential expression, co-expression, Weighted Gene Co-expression Network Analysis (WGCNA), feature selection, and functional enrichment. ExpOmics allows users to upload and explore multi-omics data without organism restrictions, supporting various expression data, including genes, mRNAs, lncRNAs, miRNAs, circRNAs, piRNAs, and proteins and is compatible with diverse gene nomenclatures and expression values. Moreover, ExpOmics enables users to analyze 22 427 transcriptomic datasets of 196 cancer subtypes sourced from 63 projects of The Cancer Genome Atlas Program (TCGA) to identify cancer biomarkers. The analysis results from ExpOmics are presented in high-quality graphical formats suitable for publication and are available for free download. A case study using ExpOmics identified two potential oncogenes, SERPINE1 and SLC43A1, that may regulate colorectal cancer through distinct biological processes. In summary, ExpOmics can serves as a robust platform for global researchers to explore multi-omics data, gain biological insights, and formulate testable hypotheses. AVAILABILITY AND IMPLEMENTATION: ExpOmics is available at http://www.biomedical-web.com/expomics.


Subject(s)
Software , Humans , Internet , Computational Biology/methods , Gene Regulatory Networks , Neoplasms/genetics , Neoplasms/metabolism , Databases, Genetic , Gene Expression Profiling/methods , Genomics/methods , Transcriptome/genetics , Multiomics
11.
Plant Physiol ; 195(2): 1624-1641, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38441329

ABSTRACT

Puccinia striiformis f. sp. tritici (Pst) secretes effector proteins that enter plant cells to manipulate host immune processes. In this report, we present an important Pst effector, Pst03724, whose mRNA expression level increases during Pst infection of wheat (Triticum aestivum). Silencing of Pst03724 reduced the growth and development of Pst. Pst03724 targeted the wheat calmodulin TaCaM3-2B, a positive regulator of wheat immunity. Subsequent investigations revealed that Pst03724 interferes with the TaCaM3-2B-NAD kinase (NADK) TaNADK2 association and thus inhibits the enzyme activity of TaNADK2 activated by TaCaM3-2B. Knocking down TaNADK2 expression by virus-mediated gene silencing significantly increased fungal growth and development, suggesting a decrease in resistance against Pst infection. In conclusion, our findings indicate that Pst effector Pst03724 inhibits the activity of NADK by interfering with the TaCaM3-2B-TaNADK2 association, thereby facilitating Pst infection.


Subject(s)
Calmodulin , Plant Diseases , Plant Immunity , Triticum , Calmodulin/metabolism , Calmodulin/genetics , Plant Diseases/microbiology , Plant Diseases/immunology , Triticum/microbiology , Triticum/genetics , Triticum/immunology , Triticum/metabolism , Plant Immunity/genetics , Puccinia/physiology , Plant Proteins/metabolism , Plant Proteins/genetics , Fungal Proteins/metabolism , Fungal Proteins/genetics , Gene Expression Regulation, Plant , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Phosphotransferases (Alcohol Group Acceptor)/genetics , Gene Silencing , Host-Pathogen Interactions , Enzyme Activation
12.
Plant Physiol ; 194(3): 1631-1645, 2024 Feb 29.
Article in English | MEDLINE | ID: mdl-38039102

ABSTRACT

PSI is a sophisticated photosynthesis protein complex that fuels the light reaction of photosynthesis in algae and vascular plants. While the structure and function of PSI have been studied extensively, the dynamic regulation on PSI oligomerization and high light response is less understood. In this work, we characterized a high light-responsive immunophilin gene FKB20-2 (FK506-binding protein 20-2) required for PSI oligomerization and high light tolerance in Chlamydomonas (Chlamydomonas reinhardtii). Biochemical assays and 77-K fluorescence measurement showed that loss of FKB20-2 led to the reduced accumulation of PSI core subunits and abnormal oligomerization of PSI complexes and, particularly, reduced PSI intermediate complexes in fkb20-2. It is noteworthy that the abnormal PSI oligomerization was observed in fkb20-2 even under dark and dim light growth conditions. Coimmunoprecipitation, MS, and yeast 2-hybrid assay revealed that FKB20-2 directly interacted with the low molecular weight PSI subunit PsaG, which might be involved in the dynamic regulation of PSI-light-harvesting complex I supercomplexes. Moreover, abnormal PSI oligomerization caused accelerated photodamage to PSII in fkb20-2 under high light stress. Together, we demonstrated that immunophilin FKB20-2 affects PSI oligomerization probably by interacting with PsaG and plays pivotal roles during Chlamydomonas tolerance to high light.


Subject(s)
Chlamydomonas reinhardtii , Chlamydomonas , Immunophilins , Photosystem I Protein Complex/genetics , Chlamydomonas/genetics , Peptidylprolyl Isomerase , Chlamydomonas reinhardtii/genetics
13.
Nat Chem Biol ; 19(7): 878-886, 2023 07.
Article in English | MEDLINE | ID: mdl-37142806

ABSTRACT

A diverse array of bacteria species naturally self-organize into durable macroscale patterns on solid surfaces via swarming motility-a highly coordinated and rapid movement of bacteria powered by flagella. Engineering swarming is an untapped opportunity to increase the scale and robustness of coordinated synthetic microbial systems. Here we engineer Proteus mirabilis, which natively forms centimeter-scale bullseye swarm patterns, to 'write' external inputs into visible spatial records. Specifically, we engineer tunable expression of swarming-related genes that modify pattern features, and we develop quantitative approaches to decoding. Next, we develop a dual-input system that modulates two swarming-related genes simultaneously, and we separately show that growing colonies can record dynamic environmental changes. We decode the resulting multicondition patterns with deep classification and segmentation models. Finally, we engineer a strain that records the presence of aqueous copper. This work creates an approach for building macroscale bacterial recorders, expanding the framework for engineering emergent microbial behaviors.


Subject(s)
Bacteria , Flagella
14.
J Med Genet ; 61(7): 652-660, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38508705

ABSTRACT

BACKGROUND: The ZFHX3 gene plays vital roles in embryonic development, cell proliferation, neuronal differentiation and neuronal death. This study aims to explore the relationship between ZFHX3 variants and epilepsy. METHODS: Whole-exome sequencing was performed in a cohort of 378 patients with partial (focal) epilepsy. A Drosophila Zfh2 knockdown model was used to validate the association between ZFHX3 and epilepsy. RESULTS: Compound heterozygous ZFHX3 variants were identified in eight unrelated cases. The burden of ZFHX3 variants was significantly higher in the case cohort, shown by multiple/specific statistical analyses. In Zfh2 knockdown flies, the incidence and duration of seizure-like behaviour were significantly greater than those in the controls. The Zfh2 knockdown flies exhibited more firing in excitatory neurons. All patients presented partial seizures. The five patients with variants in the C-terminus/N-terminus presented mild partial epilepsy. The other three patients included one who experienced frequent non-convulsive status epilepticus and two who had early spasms. These three patients had also neurodevelopmental abnormalities and were diagnosed as developmental epileptic encephalopathy (DEE), but achieved seizure-free after antiepileptic-drug treatment without adrenocorticotropic-hormone/steroids. The analyses of temporal expression (genetic dependent stages) indicated that ZFHX3 orthologous were highly expressed in the embryonic stage and decreased dramatically after birth. CONCLUSION: ZFHX3 is a novel causative gene of childhood partial epilepsy and DEE. The patients of infantile spasms achieved seizure-free after treatment without adrenocorticotropic-hormone/steroids implies a significance of genetic diagnosis in precise treatment. The genetic dependent stage provided an insight into the underlying mechanism of the evolutional course of illness.


Subject(s)
Epilepsies, Partial , Homeodomain Proteins , Spasms, Infantile , Animals , Child , Child, Preschool , Female , Humans , Infant , Male , Epilepsies, Partial/genetics , Epilepsies, Partial/drug therapy , Exome Sequencing , Genetic Predisposition to Disease , Homeodomain Proteins/genetics , Mutation , Spasms, Infantile/genetics , Drosophila
15.
Proc Natl Acad Sci U S A ; 119(14): e2121552119, 2022 04 05.
Article in English | MEDLINE | ID: mdl-35344427

ABSTRACT

SignificanceDiabetic neuropathy is a commonly occurring complication of diabetes that affects hundreds of millions of patients worldwide. Patients suffering from diabetic neuropathy experience abnormal sensations and have damage in their peripheral nerve axons as well as myelin, a tightly packed Schwann cell sheath that wraps around axons to provide insulation and increases electrical conductivity along the nerve fibers. The molecular events underlying myelin damage in diabetic neuropathy are largely unknown, and there is no efficacious treatment for the disease. The current study, using a diabetic mouse model and human patient nerve samples, uncovered a molecular mechanism underlying myelin sheath damage in diabetic neuropathy and provides a potential treatment strategy for the disease.


Subject(s)
Diabetes Mellitus , Diabetic Neuropathies , Animals , Axons , Diabetic Neuropathies/etiology , Diabetic Neuropathies/prevention & control , Humans , Mice , Myelin Sheath , Peripheral Nerves , Protein Kinases , Schwann Cells/physiology
16.
J Mol Cell Cardiol ; 186: 57-70, 2024 01.
Article in English | MEDLINE | ID: mdl-37984156

ABSTRACT

BACKGROUND: Macrophage-derived foam cells are a hallmark of atherosclerosis. Scavenger receptors, including lectin-like oxidized low-density lipoprotein (LDL) receptor-1 (OLR-1), are the principal receptors responsible for the uptake and modification of LDL, facilitating macrophage lipid load and the uptake of oxidized LDL by arterial wall cells. Krüppel-like factor 15 (KLF15) is a transcription factor that regulates the expression of genes by binding to the promoter during transcription. Therefore, this study aimed to investigate the precise role of macrophage KLF15 in atherogenesis. METHODS: We used two murine models of atherosclerosis: mice injected with an adeno-associated virus (AAV) encoding the Asp374-to-Tyr mutant version of human PCSK9, followed by 12 weeks on a high-fat diet (HFD), and ApoE-/-- mice on a HFD. We subsequently injected mice with AAV-KLF15 and AAV-LacZ to assess the role of KLF15 in the development of atherosclerosis in vivo. Oil Red O, H&E, and Masson's trichome staining were used to evaluate atherosclerotic lesions. Western blots and RT-qPCR were used to assess protein and mRNA levels, respectively. RESULTS: We determined that KLF15 expression was downregulated during atherosclerosis formation, and KLF15 overexpression prevented atherosclerosis progression. KLF15 expression levels did not affect body weight or serum lipid levels in mice. However, KLF15 overexpression in macrophages prevented foam cell formation by reducing OLR-1-meditated lipid uptake. KLF15 directly targeted and transcriptionally downregulated OLR-1 levels. Restoration of OLR-1 reversed the beneficial effects of KLF15 in atherosclerosis. CONCLUSION: Macrophage KLF15 transcriptionally downregulated OLR-1 expression to reduce lipid uptake, thereby preventing foam cell formation and atherosclerosis. Thus, our results suggest that KLF15 is a potential therapeutic target for atherosclerosis.


Subject(s)
Atherosclerosis , Foam Cells , Humans , Mice , Animals , Foam Cells/metabolism , Proprotein Convertase 9/metabolism , Macrophages/metabolism , Atherosclerosis/pathology , Lipoproteins, LDL/metabolism , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism
17.
Gene Ther ; 2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39025983

ABSTRACT

Persistence of human immunodeficiency virus (HIV) reservoirs prevents viral eradication, and consequently HIV-infected patients require lifetime treatment with antiretroviral therapy (ART) [1-5]. Currently, there are no effective therapeutics to prevent HIV rebound upon ART cessation. Here we describe an HIV/SIV Rev-dependent lentiviral particle that can be administered to inhibit viral rebound [6-9]. Using simian immunodeficiency virus (SIV)-infected rhesus macaques as a model, we demonstrate that the administration of pre-assembled SIV Rev-dependent lentiviral particles into SIVmac239-infected Indian rhesus macaques can lead to reduction of viral rebound upon ART termination. One of the injected animals, KC50, controlled plasma and CNS viremia to an undetectable level most of the time for over two years after ART termination. Surprisingly, detailed molecular and immunological characterization revealed that viremia control was concomitant with the induction of neutralizing antibodies (nAbs) following the administration of the Rev-dependent vectors. This study emphasizes the importance of neutralizing antibodies (nAbs) for viremia control [10-15], and also provides proof of concept that the Rev-dependent vector can be used to target viral reservoirs, including the CNS reservoirs, in vivo. However, future large-scale in vivo studies are needed to understand the potential mechanisms of viremia control induced by the Rev-dependent vector.

18.
J Am Chem Soc ; 146(19): 13201-13209, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38708998

ABSTRACT

The desirable superimposed stacking of two-dimensional covalent organic frameworks (2D COFs) benefits out-of-plane charge transfer, whereas the actual stacking deviation cannot leverage the potential of 2D COFs for optoelectrical applications. Herein, we report a chirality-induced strategy to control the parallel AA-stacking sequence for the ß-ketoenamine-linked COF film supported on a FTO substrate. The resulting chiral modules are periodically distributed at the framework node, ensuring identical mirrored configurations of layers for parallel stacking. Such unique architectonics exhibit the prolonged charge carrier lifetime, fast charge-transfer dynamics, and ultrahigh electron collection efficiency, thereby allowing for the excellent photocurrent response of 38 µA/cm2 at 0.25 V (vs RHE). The origin of superior performances lies in the intensified exciton gradient distribution and electron density for photoinduced electron-hole dissociation and charge transfer, in stark contrast to achiral analogues. This study highlights the stacking sequence regulated by chiral nanoarchitectonics and promises great potential of chiral COFs in photoelectrical catalysis.

19.
Anal Chem ; 96(18): 7281-7288, 2024 05 07.
Article in English | MEDLINE | ID: mdl-38663032

ABSTRACT

Single-cell spatial proteomic analysis holds great promise to advance our understanding of the composition, organization, interaction, and function of the various cell types in complex biological systems. However, the current multiplexed protein imaging technologies suffer from low detection sensitivity, limited multiplexing capacity, or are technically demanding. To tackle these issues, here, we report the development of a highly sensitive and multiplexed in situ protein profiling method using off-the-shelf antibodies. In this approach, the protein targets are stained with horseradish peroxidase (HRP) conjugated antibodies and cleavable fluorophores via click chemistry. Through repeated cycles of target staining, fluorescence imaging, and fluorophore cleavage, many proteins can be profiled in single cells in situ. Applying this approach, we successfully quantified 28 different proteins in human formalin-fixed paraffin-embedded (FFPE) tonsil tissue, which represents the highest multiplexing capacity among the tyramide signal amplification (TSA) methods. Based on their unique protein expression patterns and their microenvironment, ∼820,000 cells in the tissue are classified into distinct cell clusters. We also explored the cell-cell interactions between these varied cell clusters and observed that different subregions of the tissue are composed of cells from specific clusters.


Subject(s)
Click Chemistry , Fluorescent Dyes , Palatine Tonsil , Humans , Fluorescent Dyes/chemistry , Palatine Tonsil/cytology , Palatine Tonsil/chemistry , Palatine Tonsil/metabolism , Single-Cell Analysis , Proteins/analysis , Proteins/chemistry , Proteins/metabolism , Proteomics/methods , Horseradish Peroxidase/chemistry , Horseradish Peroxidase/metabolism , Optical Imaging , Paraffin Embedding
20.
Biochem Biophys Res Commun ; 725: 150260, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-38878760

ABSTRACT

This study introduces an innovative brain-targeted drug delivery system, RVG-Exo/CBD, utilizing rabies virus glycoprotein (RVG)-engineered exosomes for encapsulating cannabidiol (CBD). The novel delivery system was meticulously characterized, confirming the maintenance of exosomal integrity, size, and successful drug encapsulation with a high drug loading rate of 83.0 %. Evaluation of the RVG-Exo/CBD's brain-targeting capability demonstrated superior distribution and retention in brain tissue compared to unmodified exosomes, primarily validated through in vivo fluorescence imaging. The efficacy of this delivery system was assessed using a behavioral sensitization model in mice, where RVG-Exo/CBD notably suppressed methamphetamine-induced hyperactivity more effectively than CBD alone, indicating a reduction in effective dose and enhanced bioavailability. Overall, the RVG-Exo/CBD system emerges as a promising strategy for enhancing the therapeutic efficacy and safety of CBD, particularly for neurological applications, highlighting its potential for addressing the limitations associated with traditional CBD administration in clinical settings.


Subject(s)
Brain , Cannabidiol , Cannabidiol/administration & dosage , Cannabidiol/chemistry , Cannabidiol/pharmacology , Animals , Brain/metabolism , Brain/drug effects , Mice , Male , Glycoproteins/chemistry , Glycoproteins/metabolism , Glycoproteins/administration & dosage , Drug Delivery Systems/methods , Peptide Fragments , Viral Proteins
SELECTION OF CITATIONS
SEARCH DETAIL