Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 30.709
Filtrer
2.
Physiol Rep ; 12(12): e16012, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38959068

RÉSUMÉ

Pulmonary fibrosis is an interstitial scarring disease of the lung characterized by poor prognosis and limited treatment options. Tissue transglutaminase 2 (TG2) is believed to promote lung fibrosis by crosslinking extracellular matrix components and activating latent TGFß. This study assessed physiologic pulmonary function and metabolic alterations in the mouse bleomycin model with TG2 genetic deletion. TG2-deficient mice demonstrated attenuated the fibrosis and preservation of lung function, with significant reduction in elastance and increases in compliance and inspiratory capacity compared to control mice treated with bleomycin. Bleomycin induced metabolic changes in the mouse lung that were consistent with increased aerobic glycolysis, including increased expression of lactate dehydrogenase A and increased production of lactate, as well as increased glutamine, glutamate, and aspartate. TG2-deficient mice treated with bleomycin exhibited similar metabolic changes but with reduced magnitude. Our results demonstrate that TG2 is required for a typical fibrosis response to injury. In the absence of TG2, the fibrotic response is biochemically similar to wild-type, but lesions are smaller and lung function is preserved. We also show for the first time that profibrotic pathways of tissue stiffening and metabolic reprogramming are interconnected, and that metabolic disruptions in fibrosis go beyond glycolysis.


Sujet(s)
Bléomycine , Poumon , Souris knockout , Protein glutamine gamma glutamyltransferase-2 , Fibrose pulmonaire , Transglutaminases , Animaux , Bléomycine/toxicité , Protein glutamine gamma glutamyltransferase-2/métabolisme , Transglutaminases/métabolisme , Transglutaminases/génétique , Fibrose pulmonaire/induit chimiquement , Fibrose pulmonaire/métabolisme , Fibrose pulmonaire/génétique , Fibrose pulmonaire/anatomopathologie , Souris , Poumon/anatomopathologie , Poumon/métabolisme , Poumon/effets des médicaments et des substances chimiques , Protéines G/métabolisme , Protéines G/génétique , Souris de lignée C57BL , Glycolyse , Mâle
3.
Nat Commun ; 15(1): 5664, 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38969660

RÉSUMÉ

Mitochondrial gene expression relies on mitoribosomes to translate mitochondrial mRNAs. The biogenesis of mitoribosomes is an intricate process involving multiple assembly factors. Among these factors, GTP-binding proteins (GTPBPs) play important roles. In bacterial systems, numerous GTPBPs are required for ribosome subunit maturation, with EngB being a GTPBP involved in the ribosomal large subunit assembly. In this study, we focus on exploring the function of GTPBP8, the human homolog of EngB. We find that ablation of GTPBP8 leads to the inhibition of mitochondrial translation, resulting in significant impairment of oxidative phosphorylation. Structural analysis of mitoribosomes from GTPBP8 knock-out cells shows the accumulation of mitoribosomal large subunit assembly intermediates that are incapable of forming functional monosomes. Furthermore, fPAR-CLIP analysis reveals that GTPBP8 is an RNA-binding protein that interacts specifically with the mitochondrial ribosome large subunit 16 S rRNA. Our study highlights the role of GTPBP8 as a component of the mitochondrial gene expression machinery involved in mitochondrial large subunit maturation.


Sujet(s)
Protéines G , Mitochondries , Ribosomes mitochondriaux , Phosphorylation oxydative , Humains , Ribosomes mitochondriaux/métabolisme , Mitochondries/métabolisme , Protéines G/métabolisme , Protéines G/génétique , ARN ribosomique 16S/génétique , ARN ribosomique 16S/métabolisme , Protéines de liaison à l'ARN/métabolisme , Protéines de liaison à l'ARN/génétique , Protéines mitochondriales/métabolisme , Protéines mitochondriales/génétique , Cellules HEK293 , Biosynthèse des protéines , ARN messager/métabolisme , ARN messager/génétique , Cellules HeLa
4.
Cancer Med ; 13(13): e7431, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38978333

RÉSUMÉ

BACKGROUND: Cancer utilizes immunosuppressive mechanisms to create a tumor microenvironment favorable for its progression. The purpose of this study is to histologically characterize the immunological properties of the tumor microenvironment of oral squamous cell carcinoma (OSCC) and identify key molecules involved in the immunological microenvironment and patient prognosis. METHODS: First, overlapping differentially expressed genes (DEGs) were screened from OSCC transcriptome data in public databases. Correlation analysis of DEGs with known immune-related genes identified genes involved in the immune microenvironment of OSCC. Next, stromal patterns of tumor were classified and immunohistochemical staining was performed for immune cell markers (CD3, CD4, Foxp3, CD8, CD20, CD68, and CD163), programmed death-ligand 1 (PD-L1), and guanylate binding protein 5 (GBP5) in resected specimens obtained from 110 patients with OSCC who underwent resection. Correlations between each factor and their prognostic impact were analyzed. RESULTS: Among the novel OSCC-specific immune-related genes screened (including ADAMDEC1, CXCL9, CXCL13, DPT, GBP5, IDO1, and PLA2G7), GBP5 was selected as the target gene. Histopathologic analysis showed that multiple T-cell subsets and CD20-positive cells were less common in the advanced stages, whereas CD163-positive cells were more common in advanced stages. The immature type in the stromal pattern category was associated with less immune cell infiltration, lower expression of PD-L1 in immune cells, lower expression of GBP5 in the stroma, and shorter overall survival and recurrence-free survival. Expression of GBP5 in the tumor and stroma correlated with immune cell infiltration of tumors and PD-L1 expression in tumor and immune cells. Patients with low tumor GBP5 expression and high stromal expression had significantly longer overall survival and recurrence-free survival. CONCLUSIONS: The stromal pattern category may reflect both invasive and immunomodulatory potentials of cancer-associated fibroblasts in OSCC. GBP5 has been suggested as a potential biomarker to predict the prognosis and therapeutic efficacy of immune checkpoint inhibitors.


Sujet(s)
Marqueurs biologiques tumoraux , Biologie informatique , Tumeurs de la bouche , Microenvironnement tumoral , Humains , Tumeurs de la bouche/immunologie , Tumeurs de la bouche/anatomopathologie , Tumeurs de la bouche/génétique , Tumeurs de la bouche/mortalité , Tumeurs de la bouche/métabolisme , Tumeurs de la bouche/chirurgie , Mâle , Femelle , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/métabolisme , Pronostic , Biologie informatique/méthodes , Microenvironnement tumoral/immunologie , Adulte d'âge moyen , Études rétrospectives , Sujet âgé , Protéines G/génétique , Protéines G/immunologie , Protéines G/métabolisme , Adulte , Régulation de l'expression des gènes tumoraux , Carcinome épidermoïde/génétique , Carcinome épidermoïde/immunologie , Carcinome épidermoïde/anatomopathologie , Carcinome épidermoïde/mortalité , Carcinome épidermoïde/métabolisme , Antigène CD274/métabolisme , Antigène CD274/génétique , Lymphocytes TIL/immunologie , Lymphocytes TIL/métabolisme
5.
Proc Natl Acad Sci U S A ; 121(28): e2407066121, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38959038

RÉSUMÉ

Mammalian transglutaminases, a family of Ca2+-dependent proteins, are implicated in a variety of diseases. For example, celiac disease (CeD) is an autoimmune disorder whose pathogenesis requires transglutaminase 2 (TG2) to deamidate select glutamine residues in diet-derived gluten peptides. Deamidation involves the formation of transient γ-glutamyl thioester intermediates. Recent studies have revealed that in addition to the deamidated gluten peptides themselves, their corresponding thioester intermediates are also pathogenically relevant. A mechanistic understanding of this relevance is hindered by the absence of any structure of Ca2+-bound TG2. We report the X-ray crystallographic structure of human TG2 bound to an inhibitory gluten peptidomimetic and two Ca2+ ions in sites previously designated as S1 and S3. Together with additional structure-guided experiments, this structure provides a mechanistic explanation for how S1 regulates formation of an inhibitory disulfide bond in TG2, while also establishing that S3 is essential for γ-glutamyl thioester formation. Furthermore, our crystallographic findings and associated analyses have revealed that i) two interacting residues, H305 and E363, play a critical role in resolving the thioester intermediate into an isopeptide bond (transamidation) but not in thioester hydrolysis (deamidation); and ii) residues N333 and K176 stabilize preferred TG2 substrates and inhibitors via hydrogen bonding to nonreactive backbone atoms. Overall, the intermediate-state conformer of TG2 reported here represents a superior model to previously characterized conformers for both transition states of the TG2-catalyzed reaction.


Sujet(s)
Calcium , Protéines G , Protein glutamine gamma glutamyltransferase-2 , Transglutaminases , Transglutaminases/métabolisme , Transglutaminases/composition chimique , Protein glutamine gamma glutamyltransferase-2/métabolisme , Humains , Calcium/métabolisme , Protéines G/métabolisme , Protéines G/composition chimique , Cristallographie aux rayons X , Glutens/métabolisme , Glutens/composition chimique , Modèles moléculaires , Conformation des protéines , Maladie coeliaque/métabolisme , Liaison aux protéines
6.
Allergol Immunopathol (Madr) ; 52(4): 46-52, 2024.
Article de Anglais | MEDLINE | ID: mdl-38970264

RÉSUMÉ

BACKGROUND: Lung adenocarcinoma (LUAD) is a leading cause of tumor-associated mortality, and it is needed to find new target to combat this disease. Guanine nucleotide-binding -protein-like 3 (GNL3) mediates cell proliferation and apoptosis in several cancers, but its role in LUAD remains unclear. OBJECTIVE: To explore the expression and function of Guanine nucleotide-binding protein-like 3 (GNL3) in lung adenocarcinoma (LUAD) and its potential mechanism in inhibiting the growth of LUAD cells. METHODS: We evaluated the expression of GNL3 in LUAD tissues and its association with patient prognosis using databases and immunohistochemistry. Cell proliferation was assessed by CCK-8 assay as well as colony formation, while apoptosis was evaluated by FCM. The effect of GNL3 knockdown on the Wnt/ß-catenin axis was investigated by Immunoblot analysis. RESULTS: GNL3 is overexpressed in LUAD tissues and is correlated with poor prognosis. Knockdown of GNL3 significantly inhibited the growth as well as induced apoptosis in A549 as well as H1299 cells. Furthermore, we found that the inhibitory effect of GNL3 knockdown on LUAD cell growth is associated with the downregulation of the Wnt/ß-catenin axis. CONCLUSION: GNL3 is key in the progression of LUAD by metiating Wnt/ß-catenin axis. Targeting GNL3 may represent a novel therapeutic method for LUAD treatment.


Sujet(s)
Adénocarcinome pulmonaire , Apoptose , Prolifération cellulaire , Techniques de knock-down de gènes , Tumeurs du poumon , Voie de signalisation Wnt , Humains , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/métabolisme , Tumeurs du poumon/génétique , Adénocarcinome pulmonaire/génétique , Adénocarcinome pulmonaire/anatomopathologie , Adénocarcinome pulmonaire/métabolisme , Lignée cellulaire tumorale , Pronostic , Protéines G/métabolisme , Protéines G/génétique , bêta-Caténine/métabolisme , Régulation de l'expression des gènes tumoraux , Cellules A549 , Protéines nucléaires
7.
Cells ; 13(13)2024 Jun 27.
Article de Anglais | MEDLINE | ID: mdl-38994966

RÉSUMÉ

Fluorescence resonance energy transfer (FRET) biosensors have proven to be an indispensable tool in cell biology and, more specifically, in the study of G-protein signalling. The best method of measuring the activation status or FRET state of a biosensor is often fluorescence lifetime imaging microscopy (FLIM), as it does away with many disadvantages inherent to fluorescence intensity-based methods and is easily quantitated. Despite the significant potential, there is a lack of reliable FLIM-FRET biosensors, and the data processing and analysis workflows reported previously face reproducibility challenges. Here, we established a system in live primary mouse pancreatic ductal adenocarcinoma cells, where we can detect the activation of an mNeonGreen-Gαi3-mCherry-Gγ2 biosensor through the lysophosphatidic acid receptor (LPAR) with 2-photon time-correlated single-photon counting (TCSPC) FLIM. This combination gave a superior signal to the commonly used mTurquoise2-mVenus G-protein biosensor. This system has potential as a platform for drug screening, or to answer basic cell biology questions in the field of G-protein signalling.


Sujet(s)
Techniques de biocapteur , Transfert d'énergie par résonance de fluorescence , Animaux , Transfert d'énergie par résonance de fluorescence/méthodes , Souris , Techniques de biocapteur/méthodes , Protéines G/métabolisme , Humains , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas/anatomopathologie , Lignée cellulaire tumorale , Récepteurs à l'acide phosphatidique/métabolisme , Carcinome du canal pancréatique/métabolisme , Carcinome du canal pancréatique/anatomopathologie
8.
Sci Rep ; 14(1): 15981, 2024 Jul 10.
Article de Anglais | MEDLINE | ID: mdl-38987418

RÉSUMÉ

Human tissue transglutaminase (tTG) is an intriguing multifunctional enzyme involved in various diseases, including celiac disease and neurological disorders. Although a number of tTG inhibitors have been developed, the molecular determinants governing ligand binding remain incomplete due to the lack of high-resolution structural data in the vicinity of its active site. In this study, we obtained the complete high-resolution model of tTG by in silico methods based on available PDB structures. We discovered significant differences in the active site architecture between our and known tTG models, revealing an additional loop which affects the ligand binding affinity. We assembled a library of new potential tTG inhibitors based on the obtained complete model of the enzyme. Our library substantially expands the spectrum of possible drug candidates targeting tTG and encompasses twelve molecular scaffolds, eleven of which are novel and exhibit higher binding affinity then already known ones, according to our in silico studies. The results of this study open new directions for structure-based drug design of tTG inhibitors, offering the complete protein model and suggesting a wide range of new compounds for further experimental validation.


Sujet(s)
Domaine catalytique , Protéines G , Protein glutamine gamma glutamyltransferase-2 , Transglutaminases , Transglutaminases/métabolisme , Transglutaminases/composition chimique , Transglutaminases/antagonistes et inhibiteurs , Humains , Protéines G/métabolisme , Protéines G/composition chimique , Protéines G/antagonistes et inhibiteurs , Simulation numérique , Liaison aux protéines , Modèles moléculaires , Antienzymes/composition chimique , Antienzymes/pharmacologie , Simulation de docking moléculaire , Ligands , Conformation des protéines
10.
NPJ Syst Biol Appl ; 10(1): 75, 2024 Jul 16.
Article de Anglais | MEDLINE | ID: mdl-39013872

RÉSUMÉ

Mathematical models of biochemical reaction networks are an important and emerging tool for the study of cell signaling networks involved in disease processes. One promising potential application of such mathematical models is the study of how disease-causing mutations promote the signaling phenotype that contributes to the disease. It is commonly assumed that one must have a thorough characterization of the network readily available for mathematical modeling to be useful, but we hypothesized that mathematical modeling could be useful when there is incomplete knowledge and that it could be a tool for discovery that opens new areas for further exploration. In the present study, we first develop a mechanistic mathematical model of a G-protein coupled receptor signaling network that is mutated in almost all cases of uveal melanoma and use model-driven explorations to uncover and explore multiple new areas for investigating this disease. Modeling the two major, mutually-exclusive, oncogenic mutations (Gαq/11 and CysLT2R) revealed the potential for previously unknown qualitative differences between seemingly interchangeable disease-promoting mutations, and our experiments confirmed oncogenic CysLT2R was impaired at activating the FAK/YAP/TAZ pathway relative to Gαq/11. This led us to hypothesize that CYSLTR2 mutations in UM must co-occur with other mutations to activate FAK/YAP/TAZ signaling, and our bioinformatic analysis uncovers a role for co-occurring mutations involving the plexin/semaphorin pathway, which has been shown capable of activating this pathway. Overall, this work highlights the power of mechanism-based computational systems biology as a discovery tool that can leverage available information to open new research areas.


Sujet(s)
Mutation , Récepteurs couplés aux protéines G , Transduction du signal , Humains , Transduction du signal/génétique , Transduction du signal/physiologie , Récepteurs couplés aux protéines G/génétique , Récepteurs couplés aux protéines G/métabolisme , Mutation/génétique , Tumeurs de l'uvée/génétique , Tumeurs de l'uvée/métabolisme , Biologie des systèmes/méthodes , Modèles biologiques , Mélanome/génétique , Mélanome/métabolisme , Protéines G/génétique , Protéines G/métabolisme
12.
Cell Rep Methods ; 4(7): 100816, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38981474

RÉSUMÉ

We developed a method that utilizes fluorescent labeling of nuclear envelopes alongside cytometry sorting for the selective isolation of Purkinje cell (PC) nuclei. Beginning with SUN1 reporter mice, we GFP-tagged envelopes to confirm that PC nuclei could be accurately separated from other cell types. We then developed an antibody-based protocol to make PC nuclear isolation more robust and adaptable to cerebellar tissues of any genotypic background. Immunofluorescent labeling of the nuclear membrane protein RanBP2 enabled the isolation of PC nuclei from C57BL/6 cerebellum. By analyzing the expression of PC markers, nuclear size, and nucleoli number, we confirmed that our method delivers a pure fraction of PC nuclei. To demonstrate its applicability, we isolated PC nuclei from spinocerebellar ataxia type 7 (SCA7) mice and identified transcriptional changes in known and new disease-associated genes. Access to pure PC nuclei offers insights into PC biology and pathology, including the nature of selective neuronal vulnerability.


Sujet(s)
Souris de lignée C57BL , Cellules de Purkinje , Animaux , Cellules de Purkinje/métabolisme , Souris , Noyau de la cellule/métabolisme , Cervelet/métabolisme , Cervelet/cytologie , Anticorps , Protéines G , Serine-type D-Ala-D-Ala carboxypeptidase
13.
Nutrients ; 16(11)2024 May 30.
Article de Anglais | MEDLINE | ID: mdl-38892641

RÉSUMÉ

Potential celiac disease (PCD) is a clinical condition characterised by the presence of a positive CD-specific serology and a normal intestinal architecture. Asymptomatic PCD patients are generally advised to continue on a gluten-containing diet (GCD), but long-term risks of this approach have never been explored. In the present study, we aimed to investigate nutritional and autoimmune complications possibly developing overtime in a cohort of asymptomatic PCD children on a GCD. We compared children's parameters of growth, nutritional status, and autoimmunity between the time of diagnosis and on the occasion of their last medical check, after a long-term gluten-containing diet. Altogether, we collected data from 171 PCD children with a mean follow-up time of 3 years (range 0.35-15.3 years). During follow-up, although patients did not reduce their amount of daily gluten intake, their anti-tissue transglutaminase (anti-TG2) antibodies spontaneously and significantly decreased. Most parameters analysed had not changed during follow-up (height centile, ferritin, albumin, cholesterol, calcium, alkaline phosphatase, parathormone, and vitamin D) or even improved significantly (weight and BMI centile, haemoglobin, blood iron, HDL, glycaemia, and HbA1C, p < 0.05), always remaining within the limit of normality. Equally, autoantibodies for other concomitant autoimmune disorders did not increase overtime. Similar results were obtained excluding from analysis patients who had stopped producing anti-TG2 and those with a follow-up time < 3 years. Our pilot study has provided reassuring results regarding the maintenance of a gluten-containing diet in asymptomatic PCD children, even when long-term follow-up was considered.


Sujet(s)
Autoanticorps , Maladie coeliaque , Régime sans gluten , État nutritionnel , Humains , Maladie coeliaque/diétothérapie , Maladie coeliaque/immunologie , Enfant , Mâle , Femelle , Enfant d'âge préscolaire , Adolescent , Autoanticorps/sang , Protein glutamine gamma glutamyltransferase-2 , Protéines G/immunologie , Transglutaminases/immunologie , Glutens/effets indésirables , Glutens/immunologie , État de santé , Nourrisson , Études de suivi , Auto-immunité
14.
BMC Oral Health ; 24(1): 659, 2024 Jun 05.
Article de Anglais | MEDLINE | ID: mdl-38840172

RÉSUMÉ

BACKGROUND: Peri-implantitis (PI) is a frequent inflammatory disorder characterised by progressive loss of the supporting bone. Not all patients with recognised risk factors develop PI. The aim of this study is to evaluate the presence of single nucleotide polymorphisms (SNP) of inflammatory and bone metabolism related proteins in a population treated with dental implants from the Basque Country (Spain). METHODS: We included 80 patients with diagnosis of PI and 81 patients without PI, 91 women and 70 men, with a mean age of 60.90 years. SNPs of BMP-4, BRINP3, CD14, FGF-3, FGF-10, GBP-1, IL-1α, IL-1ß, IL-10, LTF, OPG and RANKL proteins were selected. We performed a univariate and bivariate analysis using IBM SPSS® v.28 statistical software. RESULTS: Presence of SNPs GBP1 rs7911 (p = 0.041) and BRINP3 rs1935881 (p = 0.012) was significantly more common in patients with PI. Patients with PI who smoked (> 10 cig/day) showed a higher presence of OPG rs2073617 SNP (p = 0.034). Also, BMP-4 rs17563 (p = 0.018) and FGF-3 rs1893047 (p = 0.014) SNPs were more frequent in patients with PI and Type II diabetes mellitus. CONCLUSIONS: Our findings suggest that PI could be favoured by an alteration in the osseointegration of dental implants, based on an abnormal immunological response to peri-implant infection in patients from the Basque Country (Spain).


Sujet(s)
Implants dentaires , Péri-implantite , Polymorphisme de nucléotide simple , Humains , Mâle , Femelle , Études cas-témoins , Adulte d'âge moyen , Espagne , Péri-implantite/génétique , Ostéoprotégérine/génétique , Sujet âgé , Protéine morphogénétique osseuse de type 4/génétique , Protéines G/génétique , Ligand de RANK/génétique , Interleukine-1 alpha/génétique , Phosphodiesterases , Pyrophosphatases
15.
Exp Dermatol ; 33(6): e15112, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38840385

RÉSUMÉ

Cutaneous squamous cell carcinoma (cSCC) ranks as the second most prevalent skin tumour (excluding melanoma). However, the molecular mechanisms driving cSCC progression remain elusive. This study aimed to investigate GBP1 expression in cSCC and elucidate its potential molecular mechanisms underlying cSCC development. GBP1 expression was assessed across public databases, cell lines and tissue samples. Various assays, including clone formation, CCK8 and EdU were employed to evaluate cell proliferation, while wound healing and transwell assays determined cell migration and invasion. Subcutaneous tumour assays were conducted to assess in vivo tumour proliferation, and molecular mechanisms were explored through western blotting, immunofluorescence and immunoprecipitation. Results identified GBP1 as an oncogene in cSCC, with elevated expression in both tumour tissues and cells, strongly correlating with tumour stage and grade. In vitro and in vivo investigations revealed that increased GBP1 expression significantly enhanced cSCC cell proliferation, migration and invasion. Mechanistically, GBP1 interaction with SP1 promoted STAT3 activation, contributing to malignant behaviours. In conclusion, the study highlights the crucial role of the GBP1/SP1/STAT3 signalling axis in regulating tumour progression in cSCC. These findings provide valuable insights into the molecular mechanisms of cSCC development and offer potential therapeutic targets for interventions against cSCC.


Sujet(s)
Carcinome épidermoïde , Mouvement cellulaire , Prolifération cellulaire , Protéines G , Invasion tumorale , Facteur de transcription STAT-3 , Tumeurs cutanées , Facteur de transcription Sp1 , Facteur de transcription STAT-3/métabolisme , Humains , Tumeurs cutanées/métabolisme , Tumeurs cutanées/anatomopathologie , Tumeurs cutanées/génétique , Carcinome épidermoïde/métabolisme , Carcinome épidermoïde/anatomopathologie , Carcinome épidermoïde/génétique , Facteur de transcription Sp1/métabolisme , Protéines G/métabolisme , Protéines G/génétique , Lignée cellulaire tumorale , Animaux , Souris , Transduction du signal , Femelle , Souris nude
16.
Proc Natl Acad Sci U S A ; 121(24): e2321532121, 2024 Jun 11.
Article de Anglais | MEDLINE | ID: mdl-38830102

RÉSUMÉ

Cannabis sativa is known for its therapeutic benefit in various diseases including pain relief by targeting cannabinoid receptors. The primary component of cannabis, Δ9-tetrahydrocannabinol (THC), and other agonists engage the orthosteric site of CB1, activating both Gi and ß-arrestin signaling pathways. The activation of diverse pathways could result in on-target side effects and cannabis addiction, which may hinder therapeutic potential. A significant challenge in pharmacology is the design of a ligand that can modulate specific signaling of CB1. By leveraging insights from the structure-function selectivity relationship (SFSR), we have identified Gi signaling-biased agonist-allosteric modulators (ago-BAMs). Further, two cryoelectron microscopy (cryo-EM) structures reveal the binding mode of ago-BAM at the extrahelical allosteric site of CB1. Combining mutagenesis and pharmacological studies, we elucidated the detailed mechanism of ago-BAM-mediated biased signaling. Notably, ago-BAM CB-05 demonstrated analgesic efficacy with fewer side effects, minimal drug toxicity and no cannabis addiction in mouse pain models. In summary, our finding not only suggests that ago-BAMs of CB1 provide a potential nonopioid strategy for pain management but also sheds light on BAM identification for GPCRs.


Sujet(s)
Cryomicroscopie électronique , Récepteur cannabinoïde de type CB1 , Transduction du signal , Récepteur cannabinoïde de type CB1/métabolisme , Récepteur cannabinoïde de type CB1/génétique , Récepteur cannabinoïde de type CB1/composition chimique , Animaux , Régulation allostérique/effets des médicaments et des substances chimiques , Souris , Humains , Transduction du signal/effets des médicaments et des substances chimiques , Protéines G/métabolisme , Protéines G/génétique , Cellules HEK293 , Relation structure-activité , Dronabinol/pharmacologie , Dronabinol/composition chimique , Dronabinol/analogues et dérivés , Cannabis/composition chimique , Cannabis/métabolisme
17.
J Proteome Res ; 23(7): 2651-2660, 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38838187

RÉSUMÉ

Histone dopaminylation is a newly identified epigenetic mark that plays a role in the regulation of gene transcription, where an isopeptide bond is formed between the fifth amino acid of H3 (i.e., glutamine) and dopamine. Recently, we developed a chemical probe to specifically label and enrich histone dopaminylation via bioorthogonal chemistry. Given this powerful tool, we found that histone H3 glutamine 5 dopaminylation (H3Q5dop) was highly enriched in colorectal tumors, which could be attributed to the high expression level of its regulator, transglutaminase 2 (TGM2), in colon cancer cells. Due to the enzyme promiscuity of TGM2, nonhistone proteins have also been identified as dopaminylation targets; however, the dopaminylated proteome in cancer cells still remains elusive. Here, we utilized our chemical probe to enrich dopaminylated proteins from colorectal cancer cells in a bioorthogonal manner and performed the chemical proteomics analysis. Therefore, 425 dopaminylated proteins were identified, many of which are involved in nucleic acid metabolism and transcription pathways. More importantly, a number of dopaminylation sites were identified and attributed to the successful application of our chemical probe. Overall, these findings shed light on the significant association between cellular protein dopaminylation and cancer development, further suggesting that targeting these pathways may become a promising anticancer strategy.


Sujet(s)
Tumeurs colorectales , Histone , Protein glutamine gamma glutamyltransferase-2 , Protéomique , Humains , Tumeurs colorectales/métabolisme , Tumeurs colorectales/génétique , Protéomique/méthodes , Protein glutamine gamma glutamyltransferase-2/métabolisme , Histone/métabolisme , Transglutaminases/métabolisme , Transglutaminases/génétique , Protéines G/métabolisme , Protéines G/génétique , Lignée cellulaire tumorale , Protéome/analyse , Protéome/métabolisme , Maturation post-traductionnelle des protéines , Glutamine/métabolisme , Glutamine/composition chimique , Épigenèse génétique
18.
J Med Virol ; 96(6): e29730, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38860570

RÉSUMÉ

Hantaan virus (HTNV) infection can cause hemorrhagic fever with renal syndrome (HFRS) in humans, and currently, there are no long-standing protective vaccines or specific antivirals available. Guanylate-binding protein 1 (GBP1) is an interferon-stimulated gene that defends against various pathogen infections. However, the function of GBP1 in HTNV infection remains unknown. Here, we describe how GBP1 prevents HTNV infection by obstructing virus entry. We found that HTNV infection induced GBP1 expression and that overexpression of GBP1 inhibited HTNV infection, while knockout of GBP1 had the opposite effect. Interestingly, GBP1 did not affect interferon (IFN) signaling during HTNV infection. Instead, GBP1 prevented HTNV from entering cells through clathrin-mediated endocytosis (CME). We also discovered that GBP1 specifically interacted with actin but not dynamin 2 (DNM2) and made it difficult for DNM2 to be recruited by actin, which may account for the suppression of CME during HTNV infection. These findings establish an antiviral role for GBP1 in inhibiting HTNV infection and help us better understand how GBP1 regulates HTNV entry and could potentially aid in developing treatments for this virus.


Sujet(s)
Endocytose , Protéines G , Virus Hantaan , Pénétration virale , Humains , Actines/métabolisme , Lignée cellulaire , Dynamine-II/métabolisme , Dynamine-II/génétique , Protéines G/génétique , Protéines G/métabolisme , Virus Hantaan/physiologie , Cellules HEK293 , Fièvre hémorragique avec syndrome rénal/virologie , Interactions hôte-pathogène
19.
Pol J Microbiol ; 73(2): 217-235, 2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38905278

RÉSUMÉ

Interferon-alpha (IFN-α) is a first-line drug for treating chronic hepatitis B (CHB). Guanylate-binding protein 1 (GBP1) is one of the interferon-stimulating factors, which participates in the innate immunity of the host and plays an antiviral and antibacterial role. In this study, we explored how GBP1 is involved in IFN-α antiviral activity against HBV. Before being gathered, HepG2-NTCP and HepG2 2.15 cells were transfected with the wild-type hGBP1 plasmid or si-GBP1, respectively, and followed by stimulation with Peg-IFNα-2b. We systematically explored the role of GBP1 in regulating HBV infection in cell models. Additionally, we also examined GBP1 levels in CHB patients. GBP1 activity increased, and its half-life was prolonged after HBV infection. Overexpression of GBP1 inhibited the production of HBsAg and HBeAg, as well as HBs protein and HBV total RNA levels, whereas silencing of GBP1 inhibited its ability to block viral infections. Interestingly, overexpressing GBP1 co-treatment with Peg-IFNα-2b further increased the antiviral effect of IFN-α, while GBP1 silencing co-treatment with Peg-IFNα-2b partly restored its inhibitory effect on HBV. Mechanistically, GBP1 mediates the anti-HBV response of Peg-IFNα-2b by targeting HBs. Analysis of clinical samples revealed that GBP1 was elevated in CHB patients and increased with Peg-IFNα-2b treatment, while GBP1 showed good stability in the interferon response group. Our study demonstrates that GBP1 inhibits HBV replication and promotes HBsAg clearance. It is possible to achieve antiviral effects through the regulation of IFN-α induced immune responses in response to HBV.


Sujet(s)
Antiviraux , Protéines G , Virus de l'hépatite B , Hépatite B chronique , Interféron alpha , Humains , Interféron alpha/pharmacologie , Interféron alpha/immunologie , Virus de l'hépatite B/effets des médicaments et des substances chimiques , Virus de l'hépatite B/physiologie , Antiviraux/pharmacologie , Protéines G/génétique , Protéines G/métabolisme , Protéines G/immunologie , Cellules HepG2 , Hépatite B chronique/traitement médicamenteux , Hépatite B chronique/virologie , Hépatite B chronique/immunologie , Mâle , Antigènes de surface du virus de l'hépatite B/immunologie , Antigènes de surface du virus de l'hépatite B/métabolisme , Femelle , Adulte , Réplication virale/effets des médicaments et des substances chimiques , Hépatite B/virologie , Hépatite B/immunologie , Hépatite B/traitement médicamenteux
20.
Nat Immunol ; 25(7): 1218-1230, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38914866

RÉSUMÉ

Transglutaminase 2 (TG2) plays a pivotal role in the pathogenesis of celiac disease (CeD) by deamidating dietary gluten peptides, which facilitates antigenic presentation and a strong anti-gluten T cell response. Here, we elucidate the molecular mechanisms underlying the efficacy of the TG2 inhibitor ZED1227 by performing transcriptional analysis of duodenal biopsies from individuals with CeD on a long-term gluten-free diet before and after a 6-week gluten challenge combined with 100 mg per day ZED1227 or placebo. At the transcriptome level, orally administered ZED1227 effectively prevented gluten-induced intestinal damage and inflammation, providing molecular-level evidence that TG2 inhibition is an effective strategy for treating CeD. ZED1227 treatment preserved transcriptome signatures associated with mucosal morphology, inflammation, cell differentiation and nutrient absorption to the level of the gluten-free diet group. Nearly half of the gluten-induced gene expression changes in CeD were associated with the epithelial interferon-γ response. Moreover, data suggest that deamidated gluten-induced adaptive immunity is a sufficient step to set the stage for CeD pathogenesis. Our results, with the limited sample size, also suggest that individuals with CeD might benefit from an HLA-DQ2/HLA-DQ8 stratification based on gene doses to maximally eliminate the interferon-γ-induced mucosal damage triggered by gluten.


Sujet(s)
Maladie coeliaque , Régime sans gluten , Protéines G , Analyse de profil d'expression de gènes , Glutens , Muqueuse intestinale , Protein glutamine gamma glutamyltransferase-2 , Transglutaminases , Maladie coeliaque/immunologie , Humains , Glutens/immunologie , Transglutaminases/métabolisme , Transglutaminases/antagonistes et inhibiteurs , Protéines G/métabolisme , Protéines G/antagonistes et inhibiteurs , Protéines G/génétique , Muqueuse intestinale/anatomopathologie , Muqueuse intestinale/métabolisme , Muqueuse intestinale/immunologie , Muqueuse intestinale/effets des médicaments et des substances chimiques , Femelle , Mâle , Adulte , Transcriptome , Duodénum/anatomopathologie , Duodénum/immunologie , Duodénum/métabolisme , Interféron gamma/métabolisme , Adulte d'âge moyen , Antigènes HLA-DQ/génétique , Antigènes HLA-DQ/immunologie , Jeune adulte , Immunité acquise/effets des médicaments et des substances chimiques
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...