Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 23
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
PLoS Biol ; 21(11): e3002353, 2023 Nov.
Article de Anglais | MEDLINE | ID: mdl-37943878

RÉSUMÉ

Wnt signaling pathways are transmitted via 10 homologous frizzled receptors (FZD1-10) in humans. Reagents broadly inhibiting Wnt signaling pathways reduce growth and metastasis of many tumors, but their therapeutic development has been hampered by the side effect. Inhibitors targeting specific Wnt-FZD pair(s) enriched in cancer cells may reduce side effect, but the therapeutic effect of narrow-spectrum Wnt-FZD inhibitors remains to be established in vivo. Here, we developed a fragment of C. difficile toxin B (TcdBFBD), which recognizes and inhibits a subclass of FZDs, FZD1/2/7, and examined whether targeting this FZD subgroup may offer therapeutic benefits for treating breast cancer models in mice. Utilizing 2 basal-like and 1 luminal-like breast cancer models, we found that TcdBFBD reduces tumor-initiating cells and attenuates growth of basal-like mammary tumor organoids and xenografted tumors, without damaging Wnt-sensitive tissues such as bones in vivo. Furthermore, FZD1/2/7-positive cells are enriched in chemotherapy-resistant cells in both basal-like and luminal mammary tumors treated with cisplatin, and TcdBFBD synergizes strongly with cisplatin in inhibiting both tumor types. These data demonstrate the therapeutic value of narrow-spectrum Wnt signaling inhibitor in treating breast cancers.


Sujet(s)
Toxines bactériennes , Tumeurs du sein , Clostridioides difficile , Tumeurs mammaires de l'animal , Humains , Animaux , Souris , Femelle , Voie de signalisation Wnt , Tumeurs du sein/métabolisme , Toxines bactériennes/métabolisme , Clostridioides difficile/métabolisme , Cisplatine
2.
Bioengineering (Basel) ; 10(8)2023 Jul 25.
Article de Anglais | MEDLINE | ID: mdl-37627769

RÉSUMÉ

Genetic screen technology has been applied to study the mechanism of action of bacterial toxins-a special class of virulence factors that contribute to the pathogenesis caused by bacterial infections. These screens aim to identify host factors that directly or indirectly facilitate toxin intoxication. Additionally, specific properties of certain toxins, such as membrane interaction, retrograde trafficking, and carbohydrate binding, provide robust probes to comprehensively investigate the lipid biosynthesis, membrane vesicle transport, and glycosylation pathways, respectively. This review specifically focuses on recent representative toxin-based genetic screens that have identified new players involved in and provided new insights into fundamental biological pathways, such as glycosphingolipid biosynthesis, protein glycosylation, and membrane vesicle trafficking pathways. Functionally characterizing these newly identified factors not only expands our current understanding of toxin biology but also enables a deeper comprehension of fundamental biological questions. Consequently, it stimulates the development of new therapeutic approaches targeting both bacterial infectious diseases and genetic disorders with defects in these factors and pathways.

3.
Nat Commun ; 13(1): 6786, 2022 11 09.
Article de Anglais | MEDLINE | ID: mdl-36351897

RÉSUMÉ

Toxin B (TcdB) is a major exotoxin responsible for diseases associated with Clostridioides difficile infection. Its sequence variations among clinical isolates may contribute to the difficulty in developing effective therapeutics. Here, we investigate receptor-binding specificity of major TcdB subtypes (TcdB1 to TcdB12). We find that representative members of subtypes 2, 4, 7, 10, 11, and 12 do not recognize the established host receptor, frizzled proteins (FZDs). Using a genome-wide CRISPR-Cas9-mediated screen, we identify tissue factor pathway inhibitor (TFPI) as a host receptor for TcdB4. TFPI is recognized by a region in TcdB4 that is homologous to the FZD-binding site in TcdB1. Analysis of 206 TcdB variant sequences reveals a set of six residues within this receptor-binding site that defines a TFPI binding-associated haplotype (designated B4/B7) that is present in all TcdB4 members, a subset of TcdB7, and one member of TcdB2. Intragenic micro-recombination (IR) events have occurred around this receptor-binding region in TcdB7 and TcdB2 members, resulting in either TFPI- or FZD-binding capabilities. Introduction of B4/B7-haplotype residues into TcdB1 enables dual recognition of TFPI and FZDs. Finally, TcdB10 also recognizes TFPI, although it does not belong to the B4/B7 haplotype, and shows species selectivity: it recognizes TFPI of chicken and to a lesser degree mouse, but not human, dog, or cattle versions. These findings identify TFPI as a TcdB receptor and reveal IR-driven changes on receptor-specificity among TcdB variants.


Sujet(s)
Toxines bactériennes , Clostridioides difficile , Animaux , Bovins , Chiens , Souris , Protéines bactériennes/métabolisme , Toxines bactériennes/composition chimique , Clostridioides difficile/génétique , Recombinaison génétique , Humains
5.
Nature ; 610(7931): 349-355, 2022 10.
Article de Anglais | MEDLINE | ID: mdl-36171290

RÉSUMÉ

Entomopathogenic nematodes are widely used as biopesticides1,2. Their insecticidal activity depends on symbiotic bacteria such as Photorhabdus luminescens, which produces toxin complex (Tc) toxins as major virulence factors3-6. No protein receptors are known for any Tc toxins, which limits our understanding of their specificity and pathogenesis. Here we use genome-wide CRISPR-Cas9-mediated knockout screening in Drosophila melanogaster S2R+ cells and identify Visgun (Vsg) as a receptor for an archetypal P. luminescens Tc toxin (pTc). The toxin recognizes the extracellular O-glycosylated mucin-like domain of Vsg that contains high-density repeats of proline, threonine and serine (HD-PTS). Vsg orthologues in mosquitoes and beetles contain HD-PTS and can function as pTc receptors, whereas orthologues without HD-PTS, such as moth and human versions, are not pTc receptors. Vsg is expressed in immune cells, including haemocytes and fat body cells. Haemocytes from Vsg knockout Drosophila are resistant to pTc and maintain phagocytosis in the presence of pTc, and their sensitivity to pTc is restored through the transgenic expression of mosquito Vsg. Last, Vsg knockout Drosophila show reduced bacterial loads and lethality from P. luminescens infection. Our findings identify a proteinaceous Tc toxin receptor, reveal how Tc toxins contribute to P. luminescens pathogenesis, and establish a genome-wide CRISPR screening approach for investigating insecticidal toxins and pathogens.


Sujet(s)
Toxines bactériennes , Systèmes CRISPR-Cas , Protéines de Drosophila , Drosophila melanogaster , Édition de gène , Facteurs de virulence , Animaux , Toxines bactériennes/métabolisme , Agents de lutte biologique , Culicidae , Protéines de Drosophila/génétique , Protéines de Drosophila/métabolisme , Drosophila melanogaster/cytologie , Drosophila melanogaster/génétique , Drosophila melanogaster/métabolisme , Drosophila melanogaster/microbiologie , Corps gras/cytologie , Techniques de knock-down de gènes , Hémocytes , Humains , Papillons de nuit , Mucines , Lutte biologique contre les nuisibles , Phagocytose , Photorhabdus/métabolisme , Séquences répétées d'acides aminés , Transgènes , Facteurs de virulence/métabolisme
6.
Cell Rep ; 38(10): 110476, 2022 03 08.
Article de Anglais | MEDLINE | ID: mdl-35263584

RÉSUMÉ

Targeted delivery of therapeutic proteins toward specific cells and across cell membranes remains major challenges. Here, we develop protein-based delivery systems utilizing detoxified single-chain bacterial toxins such as diphtheria toxin (DT) and botulinum neurotoxin (BoNT)-like toxin, BoNT/X, as carriers. The system can deliver large protein cargoes including Cas13a, CasRx, Cas9, and Cre recombinase into cells in a receptor-dependent manner, although delivery of ribonucleoproteins containing guide RNAs is not successful. Delivery of Cas13a and CasRx, together with guide RNA expression, reduces mRNAs encoding GFP, SARS-CoV-2 fragments, and endogenous proteins PPIB, KRAS, and CXCR4 in multiple cell lines. Delivery of Cre recombinase modifies the reporter loci in cells. Delivery of Cas9, together with guide RNA expression, generates mutations at the targeted genomic sites in cell lines and induced pluripotent stem cell (iPSC)-derived human neurons. These findings establish modular delivery systems based on single-chain bacterial toxins for delivery of membrane-impermeable therapeutics into targeted cells.


Sujet(s)
Toxines bactériennes , COVID-19 , Toxines bactériennes/génétique , Systèmes CRISPR-Cas , Édition de gène , Humains , /métabolisme , SARS-CoV-2
7.
Life Sci Alliance ; 5(6)2022 06.
Article de Anglais | MEDLINE | ID: mdl-35292538

RÉSUMÉ

Clostridioides difficile toxin A and B (TcdA and TcdB) are two major virulence factors responsible for diseases associated with C. difficile infection (CDI). Here, we report the 3.18-Å resolution crystal structure of a TcdA fragment (residues L843-T2481), which advances our understanding of the complete structure of TcdA holotoxin. Our structural analysis, together with complementary single molecule FRET and limited proteolysis studies, reveal that TcdA adopts a dynamic structure and its CROPs domain can sample a spectrum of open and closed conformations in a pH-dependent manner. Furthermore, a small globular subdomain (SGS) and the CROPs protect the pore-forming region of TcdA in the closed state at neutral pH, which could contribute to modulating the pH-dependent pore formation of TcdA. A rationally designed TcdA mutation that trapped the CROPs in the closed conformation showed drastically reduced cytotoxicity. Taken together, these studies shed new lights into the conformational dynamics of TcdA and its roles in TcdA intoxication.


Sujet(s)
Toxines bactériennes , Clostridioides difficile , Protéines bactériennes/génétique , Toxines bactériennes/génétique , Conformation moléculaire
8.
Cell ; 185(7): 1157-1171.e22, 2022 03 31.
Article de Anglais | MEDLINE | ID: mdl-35259335

RÉSUMÉ

Enterococci are a part of human microbiota and a leading cause of multidrug resistant infections. Here, we identify a family of Enterococcus pore-forming toxins (Epxs) in E. faecalis, E. faecium, and E. hirae strains isolated across the globe. Structural studies reveal that Epxs form a branch of ß-barrel pore-forming toxins with a ß-barrel protrusion (designated the top domain) sitting atop the cap domain. Through a genome-wide CRISPR-Cas9 screen, we identify human leukocyte antigen class I (HLA-I) complex as a receptor for two members (Epx2 and Epx3), which preferentially recognize human HLA-I and homologous MHC-I of equine, bovine, and porcine, but not murine, origin. Interferon exposure, which stimulates MHC-I expression, sensitizes human cells and intestinal organoids to Epx2 and Epx3 toxicity. Co-culture with Epx2-harboring E. faecium damages human peripheral blood mononuclear cells and intestinal organoids, and this toxicity is neutralized by an Epx2 antibody, demonstrating the toxin-mediated virulence of Epx-carrying Enterococcus.


Sujet(s)
Toxines bactériennes/métabolisme , Enterococcus , Agranulocytes , Facteurs de virulence/métabolisme , Animaux , Bovins , Enterococcus/métabolisme , Enterococcus/pathogénicité , Equus caballus , Souris , Tests de sensibilité microbienne , Suidae
9.
Sci Adv ; 7(43): eabi4582, 2021 Oct 22.
Article de Anglais | MEDLINE | ID: mdl-34678063

RÉSUMÉ

Toxin B (TcdB) is a primary cause of Clostridioides difficile infection (CDI). This toxin acts by glucosylating small GTPases in the Rho/Ras families, but the structural basis for TcdB recognition and selectivity of specific GTPase substrates remain unsolved. Here, we report the cocrystal structures of the glucosyltransferase domain (GTD) of two distinct TcdB variants in complex with human Cdc42 and R-Ras, respectively. These structures reveal a common structural mechanism by which TcdB recognizes Rho and R-Ras. Furthermore, we find selective clustering of adaptive residue changes in GTDs that determine their substrate preferences, which helps partition all known TcdB variants into two groups that display distinct specificities toward Rho or R-Ras. Mutations that selectively disrupt GTPases binding reduce the glucosyltransferase activity of the GTD and the toxicity of TcdB holotoxin. These findings establish the structural basis for TcdB recognition of small GTPases and reveal strategies for therapeutic interventions for CDI.

10.
FASEB J ; 35(7): e21647, 2021 07.
Article de Anglais | MEDLINE | ID: mdl-34165206

RÉSUMÉ

The Cytotoxic Necrotizing Factor Y (CNFY) is produced by the gram-negative, enteric pathogen Yersinia pseudotuberculosis. The bacterial toxin belongs to a family of deamidases, which constitutively activate Rho GTPases, thereby balancing inflammatory processes. We identified heparan sulfate proteoglycans as essential host cell factors for intoxication with CNFY. Using flow cytometry, microscopy, knockout cell lines, pulsed electron-electron double resonance, and bio-layer interferometry, we studied the role of glucosaminoglycans in the intoxication process of CNFY. Especially the C-terminal part of CNFY, which encompasses the catalytic activity, binds with high affinity to heparan sulfates. CNFY binding with the N-terminal domain to a hypothetical protein receptor may support the interaction between the C-terminal domain and heparan sulfates, which seems sterically hindered in the full toxin. A second conformational change occurs by acidification of the endosome, probably allowing insertion of the hydrophobic regions of the toxin into the endosomal membrane. Our findings suggest that heparan sulfates play a major role for intoxication within the endosome, rather than being relevant for an interaction at the cell surface.


Sujet(s)
Toxines bactériennes/métabolisme , Protéines Escherichia coli/métabolisme , Glycosaminoglycanes/métabolisme , Héparine/métabolisme , Lymphocytes/métabolisme , Protéines recombinantes/métabolisme , Yersinia pseudotuberculosis/composition chimique , Toxines bactériennes/composition chimique , Toxines bactériennes/génétique , Protéines Escherichia coli/composition chimique , Protéines Escherichia coli/génétique , Cellules HeLa , Humains , Conformation des protéines , Protéines recombinantes/génétique
11.
Nat Commun ; 12(1): 3748, 2021 06 18.
Article de Anglais | MEDLINE | ID: mdl-34145250

RÉSUMÉ

C. difficile is a major cause of antibiotic-associated gastrointestinal infections. Two C. difficile exotoxins (TcdA and TcdB) are major virulence factors associated with these infections, and chondroitin sulfate proteoglycan 4 (CSPG4) is a potential receptor for TcdB, but its pathophysiological relevance and the molecular details that govern recognition remain unknown. Here, we determine the cryo-EM structure of a TcdB-CSPG4 complex, revealing a unique binding site spatially composed of multiple discontinuous regions across TcdB. Mutations that selectively disrupt CSPG4 binding reduce TcdB toxicity in mice, while CSPG4-knockout mice show reduced damage to colonic tissues during C. difficile infections. We further show that bezlotoxumab, the only FDA approved anti-TcdB antibody, blocks CSPG4 binding via an allosteric mechanism, but it displays low neutralizing potency on many TcdB variants from epidemic hypervirulent strains due to sequence variations in its epitopes. In contrast, a CSPG4-mimicking decoy neutralizes major TcdB variants, suggesting a strategy to develop broad-spectrum therapeutics against TcdB.


Sujet(s)
Antigènes/métabolisme , Protéines bactériennes/métabolisme , Toxines bactériennes/métabolisme , Clostridioides difficile/pathogénicité , Entérocolite pseudomembraneuse/anatomopathologie , Protéoglycanes/métabolisme , Animaux , Anticorps monoclonaux/pharmacologie , Antigènes/génétique , Protéines bactériennes/génétique , Toxines bactériennes/génétique , Sites de fixation/physiologie , Anticorps neutralisants à large spectre/pharmacologie , Cryomicroscopie électronique , Entérocolite pseudomembraneuse/traitement médicamenteux , Souris , Souris de lignée C57BL , Souris knockout , Complexes multiprotéiques/métabolisme , Liaison aux protéines/effets des médicaments et des substances chimiques , Conformation des protéines , Protéoglycanes/génétique
12.
Toxins (Basel) ; 13(3)2021 03 18.
Article de Anglais | MEDLINE | ID: mdl-33803852

RÉSUMÉ

Shiga toxins (Stxs) are classic bacterial toxins and major virulence factors of toxigenic Shigella dysenteriae and enterohemorrhagic Escherichia coli (EHEC). These toxins recognize a glycosphingolipid globotriaosylceramide (Gb3/CD77) as their receptor and inhibit protein synthesis in cells by cleaving 28S ribosomal RNA. They are the major cause of life-threatening complications such as hemolytic uremic syndrome (HUS), associated with severe cases of EHEC infection, which is the leading cause of acute kidney injury in children. The threat of Stxs is exacerbated by the lack of toxin inhibitors and effective treatment for HUS. Here, we briefly summarize the Stx structure, subtypes, in vitro and in vivo models, Gb3 expression and HUS, and then introduce recent studies using CRISPR-Cas9-mediated genome-wide screens to identify the host cell factors required for Stx action. We also summarize the latest progress in utilizing and engineering Stx components for biomedical applications.


Sujet(s)
Infections à Escherichia coli/métabolisme , Syndrome hémolytique et urémique/métabolisme , Shiga-toxines/métabolisme , Escherichia coli producteur de Shiga-toxine/métabolisme , Trihexosylcéramide/métabolisme , Animaux , Systèmes CRISPR-Cas , Infections à Escherichia coli/génétique , Infections à Escherichia coli/microbiologie , Syndrome hémolytique et urémique/génétique , Syndrome hémolytique et urémique/microbiologie , Interactions hôte-pathogène , Humains , Immunotoxines/usage thérapeutique , Modèles moléculaires , Tumeurs/traitement médicamenteux , Tumeurs/immunologie , Tumeurs/métabolisme , Tumeurs/anatomopathologie , Conformation des protéines , Shiga-toxines/composition chimique , Shiga-toxines/génétique , Shiga-toxines/usage thérapeutique , Escherichia coli producteur de Shiga-toxine/génétique , Relation structure-activité
13.
Genet Med ; 23(6): 1158-1162, 2021 06.
Article de Anglais | MEDLINE | ID: mdl-33531666

RÉSUMÉ

PURPOSE: The endoplasmic reticulum membrane complex (EMC) is a highly conserved, multifunctional 10-protein complex related to membrane protein biology. In seven families, we identified 13 individuals with highly overlapping phenotypes who harbor a single identical homozygous frameshift variant in EMC10. METHODS: Using exome, genome, and Sanger sequencing, a recurrent frameshift EMC10 variant was identified in affected individuals in an international cohort of consanguineous families. Multiple families were independently identified and connected via Matchmaker Exchange and internal databases. We assessed the effect of the frameshift variant on EMC10 RNA and protein expression and evaluated EMC10 expression in normal human brain tissue using immunohistochemistry. RESULTS: A homozygous variant EMC10 c.287delG (Refseq NM_206538.3, p.Gly96Alafs*9) segregated with affected individuals in each family, who exhibited a phenotypic spectrum of intellectual disability (ID) and global developmental delay (GDD), variable seizures and variable dysmorphic features (elongated face, curly hair, cubitus valgus, and arachnodactyly). The variant arose on two founder haplotypes and results in significantly reduced EMC10 RNA expression and an unstable truncated EMC10 protein. CONCLUSION: We propose that a homozygous loss-of-function variant in EMC10 causes a novel syndromic neurodevelopmental phenotype. Remarkably, the recurrent variant is likely the result of a hypermutable site and arose on distinct founder haplotypes.


Sujet(s)
Incapacités de développement , Déficience intellectuelle , Enfant , Incapacités de développement/génétique , Mutation avec décalage du cadre de lecture , Homozygote , Humains , Déficience intellectuelle/génétique , Protéines membranaires/génétique , Pedigree , Phénotype , Crises épileptiques/génétique
14.
Cell Host Microbe ; 27(5): 782-792.e7, 2020 05 13.
Article de Anglais | MEDLINE | ID: mdl-32302524

RÉSUMÉ

The exotoxin TcsL is a major virulence factor in Paeniclostridium (Clostridium) sordellii and responsible for the high lethality rate associated with P. sordellii infection. Here, we present a genome-wide CRISPR-Cas9-mediated screen using a human lung carcinoma cell line and identify semaphorin (SEMA) 6A and 6B as receptors for TcsL. Disrupting SEMA6A/6B expression in several distinct human cell lines and primary human endothelial cells results in reduced TcsL sensitivity, while SEMA6A/6B over-expression increases their sensitivity. TcsL recognizes the extracellular domain (ECD) of SEMA6A/6B via a region homologous to the receptor-binding site in Clostridioides difficile toxin B (TcdB), which binds the human receptor Frizzled. Exchanging the receptor-binding interfaces between TcsL and TcdB switches their receptor-binding specificity. Finally, administration of SEMA6A-ECD proteins protects human cells from TcsL toxicity and reduces TcsL-induced damage to lung tissues and the lethality rate in mice. These findings establish SEMA6A and 6B as pathophysiologically relevant receptors for TcsL.


Sujet(s)
Toxines bactériennes/métabolisme , Clostridium sordellii/génétique , Clostridium sordellii/métabolisme , Sémaphorines/génétique , Sémaphorines/isolement et purification , Cellules A549 , Animaux , Protéines bactériennes , Sites de fixation , Systèmes CRISPR-Cas , Lignée cellulaire tumorale , Clustered regularly interspaced short palindromic repeats , Cellules endothéliales/métabolisme , Femelle , Techniques de knock-out de gènes , Cellules HeLa , Humains , Tumeurs du poumon , Mâle , Souris , Liaison aux protéines , Sémaphorines/métabolisme , Facteurs de virulence/métabolisme
15.
Life Sci Alliance ; 3(5)2020 05.
Article de Anglais | MEDLINE | ID: mdl-32234750

RÉSUMÉ

Osteosarcoma (OS) is a primary malignant bone neoplasm with high frequencies of tumor metastasis and recurrence. Although the Akt/PKB signaling pathway is known to play key roles in tumorigenesis, the roles of cyclin-dependent kinase-like 3 (CDKL3) in OS progression remain largely elusive. We have demonstrated the high expression levels of CDKL3 in OS human specimens and comprehensively investigated the role of CDKL3 in promoting OS progression both in vitro and in vivo. We found that CDKL3 regulates Akt activation and its downstream effects, including cell growth and autophagy. The up-regulation of CDKL3 in OS specimens appeared to be associated with Akt activation and shorter overall patient survival (P = 0.003). Our findings identify CDKL3 as a critical regulator that stimulates OS progression by enhancing Akt activation. CDKL3 represents both a biomarker for OS prognosis, and a potential therapeutic target in precision medicine by targeting CDKL3 to treat Akt hyper-activated OS.


Sujet(s)
Ostéosarcome/métabolisme , Protein-Serine-Threonine Kinases/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Animaux , Apoptose/génétique , Autophagie/génétique , Tumeurs osseuses/génétique , Carcinogenèse/génétique , Cycle cellulaire , Lignée cellulaire tumorale , Mouvement cellulaire/génétique , Prolifération cellulaire/génétique , Chine , Évolution de la maladie , Régulation de l'expression des gènes tumoraux/génétique , Humains , Mâle , Souris , Souris de lignée BALB C , Souris nude , Récidive tumorale locale/génétique , Ostéosarcome/génétique , Protein-Serine-Threonine Kinases/génétique , Protéines proto-oncogènes c-akt/génétique , Transduction du signal/génétique
16.
Metallomics ; 11(11): 1925-1936, 2019 11 01.
Article de Anglais | MEDLINE | ID: mdl-31631207

RÉSUMÉ

Auranofin (AuRF) has been reported to display anticancer activity and has entered several clinical trials; however, its mechanism of action remains largely unknown. In this work, the anticancer mechanism of auranofin was investigated using a proteomics strategy entailing subcellular fractionation prior to mass spectrometric analysis. Bioinformatics analysis of the nuclear sub-proteomes revealed that tumor suppressor p14ARF is a key regulator of transcription. Through independent analysis, we validated that up-regulation of p14ARF is associated with E2F-dependent transcription and increased p53 expression. Our analyses further reveal that 3-hydroxy-3-methylglutaryl-coenzyme A reductase (HMGCR), which is the rate-determining enzyme of the mevalonate pathway, is a novel target of auranofin with half maximal inhibitory concentration at micromolar levels. The auranofin-induced cancer cell death could be partially reverted by the addition of downstream products of the mevalonate pathway (mevalonolactone or geranyleranyl pyrophosphate (GGPP)), implying that auranofin may target the mevalonate pathway to exert its anticancer effect.


Sujet(s)
Antinéoplasiques/pharmacologie , Auranofine/pharmacologie , Hydroxymethylglutaryl-CoA reductases/métabolisme , Thérapie moléculaire ciblée , Lignée cellulaire tumorale , Facteurs de transcription E2F/métabolisme , Or/pharmacologie , Humains , Transduction du signal/effets des médicaments et des substances chimiques , Facteurs temps , Protéine p14(ARF) suppresseur de tumeur/métabolisme , Protéine p53 suppresseur de tumeur/métabolisme , Régulation positive/effets des médicaments et des substances chimiques
17.
Cell Rep ; 28(10): 2517-2526.e5, 2019 09 03.
Article de Anglais | MEDLINE | ID: mdl-31484065

RÉSUMÉ

The endoplasmic reticulum (ER) membrane protein complex (EMC) is a key contributor to biogenesis and membrane integration of transmembrane proteins, but our understanding of its mechanisms and the range of EMC-dependent proteins remains incomplete. Here, we carried out an unbiased mass spectrometry (MS)-based quantitative proteomic analysis comparing membrane proteins in EMC-deficient cells to wild-type (WT) cells and identified 36 EMC-dependent membrane proteins and 171 EMC-independent membrane proteins. Of these, six EMC-dependent and six EMC-independent proteins were further independently validated. We found that a common feature among EMC-dependent proteins is that they contain transmembrane domains (TMDs) with polar and/or charged residues. Mutagenesis studies demonstrate that EMC dependency can be converted in cells by removing or introducing polar and/or charged residues within TMDs. Our studies expand the list of validated EMC-dependent and EMC-independent proteins and suggest that the EMC is involved in handling TMDs with residues challenging for membrane integration.


Sujet(s)
Réticulum endoplasmique/métabolisme , Membranes intracellulaires/métabolisme , Protéines membranaires/métabolisme , Complexes multiprotéiques/métabolisme , Protéomique , Lignée cellulaire , Cellules HEK293 , Cellules HeLa , Humains , Protéines membranaires/composition chimique , Mutagenèse/génétique , Domaines protéiques , Reproductibilité des résultats , Réponse aux protéines mal repliées
18.
Nat Microbiol ; 4(10): 1760-1769, 2019 10.
Article de Anglais | MEDLINE | ID: mdl-31160825

RÉSUMÉ

Clostridium difficile toxin A (TcdA) is a major exotoxin contributing to disruption of the colonic epithelium during C. difficile infection. TcdA contains a carbohydrate-binding combined repetitive oligopeptides (CROPs) domain that mediates its attachment to cell surfaces, but recent data suggest the existence of CROPs-independent receptors. Here, we carried out genome-wide clustered regularly interspaced short palindromic repeats (CRISPR)-CRISPR-associated protein 9 (Cas9)-mediated screens using a truncated TcdA lacking the CROPs, and identified sulfated glycosaminoglycans (sGAGs) and low-density lipoprotein receptor (LDLR) as host factors contributing to binding and entry of TcdA. TcdA recognizes the sulfation group in sGAGs. Blocking sulfation and glycosaminoglycan synthesis reduces TcdA binding and entry into cells. Binding of TcdA to the colonic epithelium can be reduced by surfen, a small molecule that masks sGAGs, by GM-1111, a sulfated heparan sulfate analogue, and by sulfated cyclodextrin, a sulfated small molecule. Cells lacking LDLR also show reduced sensitivity to TcdA, although binding between LDLR and TcdA are not detected, suggesting that LDLR may facilitate endocytosis of TcdA. Finally, GM-1111 reduces TcdA-induced fluid accumulation and tissue damage in the colon in a mouse model in which TcdA is injected into the caecum. These data demonstrate in vivo and pathological relevance of TcdA-sGAGs interactions, and reveal a potential therapeutic approach of protecting colonic tissues by blocking these interactions.


Sujet(s)
Toxines bactériennes/métabolisme , Clostridioides difficile/composition chimique , Entérotoxines/métabolisme , Glycosaminoglycanes/métabolisme , Récepteurs aux lipoprotéines LDL/métabolisme , Animaux , Toxines bactériennes/composition chimique , Toxines bactériennes/génétique , Toxines bactériennes/toxicité , Membrane cellulaire/métabolisme , Côlon/effets des médicaments et des substances chimiques , Côlon/métabolisme , Endocytose , Entérotoxines/composition chimique , Entérotoxines/génétique , Entérotoxines/toxicité , Glycosaminoglycanes/déficit , Cellules HeLa , Héparitine sulfate/analogues et dérivés , Héparitine sulfate/pharmacologie , Humains , Muqueuse intestinale/métabolisme , Souris , Mutation , Oligopeptides/génétique , Liaison aux protéines , Récepteurs aux lipoprotéines LDL/déficit
19.
PLoS Biol ; 16(11): e2006951, 2018 11.
Article de Anglais | MEDLINE | ID: mdl-30481169

RÉSUMÉ

Glycosylation is a fundamental modification of proteins and membrane lipids. Toxins that utilize glycans as their receptors have served as powerful tools to identify key players in glycosylation processes. Here, we carried out Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)-Cas9-mediated genome-wide loss-of-function screens using two related bacterial toxins, Shiga-like toxins (Stxs) 1 and 2, which use a specific glycolipid, globotriaosylceramide (Gb3), as receptors, and the plant toxin ricin, which recognizes a broad range of glycans. The Stxs screens identified major glycosyltransferases (GTs) and transporters involved in Gb3 biosynthesis, while the ricin screen identified GTs and transporters involved in N-linked protein glycosylation and fucosylation. The screens also identified lysosomal-associated protein transmembrane 4 alpha (LAPTM4A), a poorly characterized four-pass membrane protein, as a factor specifically required for Stxs. Mass spectrometry analysis of glycolipids and their precursors demonstrates that LAPTM4A knockout (KO) cells lack Gb3 biosynthesis. This requirement of LAPTM4A for Gb3 synthesis is not shared by its homolog lysosomal-associated protein transmembrane 4 beta (LAPTM4B), and switching the domains between them determined that the second luminal domain of LAPTM4A is required, potentially acting as a specific "activator" for the GT that synthesizes Gb3. These screens also revealed two Golgi proteins, Transmembrane protein 165 (TMEM165) and Transmembrane 9 superfamily member 2 (TM9SF2), as shared factors required for both Stxs and ricin. TMEM165 KO and TM9SF2 KO cells both showed a reduction in not only Gb3 but also other glycosphingolipids, suggesting that they are required for maintaining proper levels of glycosylation in general in the Golgi. In addition, TM9SF2 KO cells also showed defective endosomal trafficking. These studies reveal key Golgi proteins critical for regulating glycosylation and glycolipid synthesis and provide novel therapeutic targets for blocking Stxs and ricin toxicity.


Sujet(s)
Ricine/génétique , Shiga-toxines/génétique , Toxines bactériennes/métabolisme , Systèmes CRISPR-Cas , Endosomes/métabolisme , Étude d'association pangénomique/méthodes , Glycolipides/métabolisme , Glycosphingolipides , Glycosylation , Appareil de Golgi/métabolisme , Appareil de Golgi/physiologie , Cellules HEK293 , Cellules HeLa , Humains , Mutation perte de fonction/génétique , Protéines membranaires/métabolisme , Protéines de transport membranaire/métabolisme , Protéines de transport membranaire/physiologie , Protéines oncogènes/métabolisme , Transport des protéines , Ricine/métabolisme , Shiga-toxines/métabolisme , Trihexosylcéramide/métabolisme , Trihexosylcéramide/physiologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...