Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 2.204
Filtrer
1.
Sci Rep ; 14(1): 22286, 2024 Sep 27.
Article de Anglais | MEDLINE | ID: mdl-39333229

RÉSUMÉ

Intelectin-1 (ITLN1; also Omentin-1, OMNT1) is secreted by adipose tissue (AT) and plays an important role in glucose metabolism regulation, with links to obesity-associated diseases. ITLN1 activity so far has rarely been investigated using RNA-sequencing and in larger cohorts. We evaluated ITLN1 expression among three clinical cohorts of the Leipzig Obesity BioBank-a cross-sectional cohort comprising of 1480 people, a cohort of people with metabolically healthy or unhealthy obesity (31 insulin-sensitive, 42 insulin-resistant individuals with obesity), and a longitudinal two-step bariatric surgery cohort (n = 65). We hypothesized that AT ITLN1 expression is associated with serum omentin-1, clinical parameters associated with obesity, and with weight loss after bariatric surgery. We also investigated the correlation of AT ITLN1 expression with genes related to inflammatory response, lipid metabolism, obesity, and regulation of energy balance. Likewise, we inspected gene group expression and metabolic pathways associated with ITLN1 expression using gene set enrichment and gene correlation analysis. We show that ITLN1 expression differs in VAT and SAT, and should therefore be analyzed separately. Furthermore, ITLN1 expression increases with VAT tissue mass, but is negatively affected by AT tissue dysfunction among individuals with unhealthy obesity, corroborated by interplay with genes related to tissue inflammation. Gene set enrichment and gene correlation analysis of ITLN1 expression suggest that AT ITLN1 expression is related to local inflammatory processes in AT, but also in processes such as regulation of appetite, energy balance, and maintenance of body weight.


Sujet(s)
Chirurgie bariatrique , Cytokines , Protéines liées au GPI , Insulinorésistance , Lectines , Obésité , Humains , Protéines liées au GPI/génétique , Protéines liées au GPI/métabolisme , Insulinorésistance/génétique , Lectines/génétique , Lectines/métabolisme , Lectines/sang , Obésité/métabolisme , Obésité/chirurgie , Obésité/génétique , Cytokines/métabolisme , Cytokines/sang , Mâle , Femelle , Adulte d'âge moyen , Adulte , Études transversales , Phénotype , Surpoids/métabolisme , Surpoids/génétique , Perte de poids/génétique , Tissu adipeux/métabolisme , Études de cohortes
2.
Front Immunol ; 15: 1411072, 2024.
Article de Anglais | MEDLINE | ID: mdl-39211050

RÉSUMÉ

Introduction: Gliomas are the most common primary intracranial tumors, known for their high invasiveness and destructiveness. Sialic acid-binding immunoglobulin-like lectin 7 (SIGLEC7) is present in various immune cells, especially macrophages, and significantly affects immune homeostasis and cancer cell response. However, research on the role and prognostic impact of SIGLEC7 in glioma patients is currently limited. Methods: We utilized transcriptomic data from 702 glioma patients in The Cancer Genome Atlas (TCGA) and 693 glioma patients in the Chinese Glioma Genome Atlas (CGGA), along with clinical samples we collected, to comprehensively investigate the impact of SIGLEC7 on glioma expression patterns, biological functions, and prognostic value. We focused on its role in glioma-related immune responses and immune cell infiltration and analyzed its expression at the single-cell level. Finally, we validated the role of SIGLEC7 in gliomas through tissue and cell experiments. Results: SIGLEC7 expression was significantly increased in glioma patients with malignant characteristics. Survival analysis indicated that glioma patients with high SIGLEC7 expression had significantly lower survival rates. Gene function analysis revealed that SIGLEC7 is primarily involved in immune and inflammatory responses and is strongly negatively correlated with tumor-associated immune regulation. Additionally, the expression of most immune checkpoints was positively correlated with SIGLEC7, and immune cell infiltration analysis clearly demonstrated a significant positive correlation between SIGLEC7 expression and M2 macrophage infiltration levels. Single-cell analysis, along with tissue and cell experiments, confirmed that SIGLEC7 enhances macrophage polarization towards the M2 phenotype, thereby promoting glioma invasiveness through the immunosuppressive effects of M2 macrophages. Cox regression analysis and the establishment of survival prediction models indicated that high SIGLEC7 expression is an unfavorable prognostic factor for glioma patients. Discussion: High SIGLEC7 expression predicts poor prognosis in glioma patients and is closely associated with M2 macrophages in the tumor environment. In the future, SIGLEC7 may become a promising target for glioma immunotherapy.


Sujet(s)
Tumeurs du cerveau , Gliome , Macrophages , Humains , Gliome/immunologie , Gliome/génétique , Gliome/mortalité , Gliome/anatomopathologie , Pronostic , Tumeurs du cerveau/immunologie , Tumeurs du cerveau/génétique , Tumeurs du cerveau/mortalité , Tumeurs du cerveau/anatomopathologie , Macrophages/immunologie , Macrophages/métabolisme , Mâle , Femelle , Lectines/génétique , Lectines/métabolisme , Régulation de l'expression des gènes tumoraux , Microenvironnement tumoral/immunologie , Activation des macrophages/génétique , Marqueurs biologiques tumoraux/génétique , Adulte d'âge moyen , Antigènes de différenciation des lymphocytes B/génétique , Antigènes de différenciation des myélomonocytes
3.
Int J Biol Macromol ; 278(Pt 3): 134851, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39168212

RÉSUMÉ

In mammals, siglec7, an integral component of the siglecs, is principally found on the surface of natural killer (NK) cells, macrophages, and monocytes, where it interacts with various pathogens to perform immunological regulatory activities. Nonetheless, the immune defense and mechanism of siglec7 in early vertebrates remain unknown. In this study, we identified siglec7 from Oreochromis niloticus (OnSiglec7) and revealed its immune functions. Specifically, OnSiglec7 was abundantly expressed in immune-related tissues of healthy tilapia and its transcription level was strongly activated after being challenged with A. hydrophila, S. agalactiae, and Poly: IC. Meanwhile, OnSiglec7 protein was purified and analyzed, which could recognize multiple pathogens through binding and agglutinating activity. Moreover, OnSiglec7-positive cells were mainly distributed in non-specific cytotoxic cells (NCC) of tilapia HKLs and showed cell membrane localization. Furthermore, OnSiglec7 blockage affected multiple innate immune responses (inflammation, apoptosis, and pyroptosis process) by regulating the activation of MAPK, NF-κB, TLR, and JAK-STAT pathways. Finally, OnSiglec7 blockage also greatly enhanced the cytotoxic effect of tilapia NCC. Summarily, this study uncovers immune functions and mechanisms of siglec7 in primitive vertebrates, thereby enhancing our understanding of the systemic evolution and ancient functions of other siglecs within the host's innate immune system (to our knowledge).


Sujet(s)
Immunité innée , Animaux , Cichlides/immunologie , Cichlides/métabolisme , Lectines/immunologie , Lectines/métabolisme , Lectines/génétique , Protéines de poisson/génétique , Protéines de poisson/immunologie , Protéines de poisson/métabolisme , Transduction du signal
4.
mSphere ; 9(9): e0046624, 2024 Sep 25.
Article de Anglais | MEDLINE | ID: mdl-39136454

RÉSUMÉ

The cyst wall of the eye pathogen Acanthamoeba castellanii contains cellulose and has ectocyst and endocyst layers connected by conical ostioles. Cyst walls contain families of lectins that localize to the ectocyst layer (Jonah) or the endocyst layer and ostioles (Luke and Leo). How lectins and an abundant laccase bind cellulose and why proteins go to locations in the wall are not known and are the focus of the studies here. Structural predictions identified ß-jelly-roll folds (BJRFs) of Luke and sets of four disulfide knots (4DKs) of Leo, each of which contains linear arrays of aromatic amino acids, also present in carbohydrate-binding modules of bacterial and plant endocellulases. Ala mutations showed that these aromatics are necessary for cellulose binding and proper localization of Luke and Leo in the Acanthamoeba cyst wall. BJRFs of Luke, 4DKs of Leo, a single ß-helical fold (BHF) of Jonah, and a copper oxidase domain of the laccase each bind to glycopolymers in both layers of deproteinated cyst walls. Promoter swaps showed that ectocyst localization does not just correlate with but is caused by early encystation-specific expression, while localization in the endocyst layer and ostioles is caused by later expression. Evolutionary studies showed distinct modes of assembly of duplicated domains in Luke, Leo, and Jonah lectins and suggested Jonah BHFs originated from bacteria, Luke BJRFs share common ancestry with slime molds, while 4DKs of Leo are unique to Acanthamoeba.IMPORTANCEAcanthamoebae is the only human parasite with cellulose in its cyst wall and conical ostioles that connect its inner and outer layers. Cyst walls are important virulence factors because they make Acanthamoebae resistant to surface disinfectants, hand sanitizers, contact lens sterilizers, and antibiotics applied to the eye. The goal here was to understand better how proteins are targeted to specific locations in the cyst wall. To this end, we identified three new proteins in the outer layer of the cyst wall, which may be targets for diagnostic antibodies in corneal scrapings. We used structural predictions and mutated proteins to show linear arrays of aromatic amino acids of two unrelated wall proteins are necessary for binding cellulose and proper wall localization. We showed early expression during encystation causes proteins to localize to the outer layer, while later expression causes proteins to localize to the inner layer and the ostioles.


Sujet(s)
Acanthamoeba castellanii , Cellulose , Protéines de protozoaire , Cellulose/métabolisme , Acanthamoeba castellanii/génétique , Acanthamoeba castellanii/métabolisme , Protéines de protozoaire/génétique , Protéines de protozoaire/métabolisme , Protéines de protozoaire/composition chimique , Paroi cellulaire/métabolisme , Paroi cellulaire/composition chimique , Paroi cellulaire/génétique , Liaison aux protéines , Lectines/génétique , Lectines/métabolisme , Acanthamoeba/génétique , Acanthamoeba/métabolisme , Transport des protéines , Laccase/génétique , Laccase/métabolisme , Laccase/composition chimique
5.
BMC Genomics ; 25(1): 805, 2024 Aug 27.
Article de Anglais | MEDLINE | ID: mdl-39187771

RÉSUMÉ

BACKGROUND: Hermansky-Pudlak Syndrome (HPS), a rare autosomal recessive disorder, is characterized by oculocutaneous albinism, bleeding diathesis, and sometimes severe lung problems and inflammatory bowel disease. Symptoms include skin and hair pigmentation variations, along with visual impairments. Variants in eleven genes encoding protein complexes essential for membrane trafficking and intracellular endosomal transport pathways underlie various recognized HPS subtypes. This study focuses on HPS-9, a subtype of Hermansky-Pudlak Syndrome caused by a variant in the BLOC1S6 gene, which is a subunit of the BLOC1 complex. In this study, a novel Copy Number Variation (CNV) in the aforementioned gene in an Iranian family is reported. The study aims to better understand the etiology of HPS-9 symptoms by identifying and confirming the variant and determining whether the gene is expressed despite the deletion. There have only been five reports of this syndrome in the literature thus far. Our novel CNV represents a significant contribution to understanding the genetic basis of HPS-9. RESULTS: This study investigates a male patient presenting with albinism. Whole Exome Sequencing (WES) identified a homozygous deletion of approximately 350 bp using CNV analysis. The deletion affects the intronic region of the BLOC1S6 gene, causing uncertainties in defining the exact boundaries due to WES limitations. Primer walking and GAP-PCR techniques were used to define the deletion boundaries. Subsequent assessments of this variant across other family members helped identify homozygous affected members and heterozygous carriers. The absence of BLOC1S6 expression in the affected individual was confirmed through Real-time PCR experiments. These findings underscore the importance of understanding the implications for the patient's healthcare and potential therapeutic approaches. CONCLUSION: This study introduces a case of Hermansky-Pudlak Syndrome Type 9 (HPS-9) caused by a homozygous deletion in the BLOC1S6 gene. We identified an approximately 7-kb deletion encompassing exon 1 and the intronic region of the gene. The absence of BLOC1S6 expression, confirmed via Real-time PCR, highlights the importance of studying the pathogenicity of the deletion and its impact on the patient's health. Our findings contribute to the sparse knowledge on HPS-9 and underscore the need for further exploration into the genetic causes of this rare disorder.


Sujet(s)
Protéines de transport , Variations de nombre de copies de segment d'ADN , Syndrome d'Hermanski-Pudlak , Lectines , Femelle , Humains , Mâle , , Syndrome d'Hermanski-Pudlak/génétique , Iran , Pedigree , Délétion de séquence , Protéines de transport/génétique , Lectines/génétique
6.
JCI Insight ; 9(16)2024 Jul 11.
Article de Anglais | MEDLINE | ID: mdl-38990656

RÉSUMÉ

Carcinomas are common in humans but rare among closely related "great apes." Plausible explanations, including human-specific genomic alterations affecting the biology of sialic acids, are proposed, but causality remains unproven. Here, an integrated evolutionary genetics-phenome-transcriptome approach studied the role of SIGLEC12 gene (encoding Siglec-XII) in epithelial transformation and cancer. Exogenous expression of the protein in cell lines and genetically engineered mice recapitulated approximately 30% of the human population in whom the protein is expressed in a form that cannot bind ligand because of a fixed, homozygous, human-universal missense mutation. Siglec-XII-null cells/mice recapitulated the remaining approximately 70% of the human population in whom an additional polymorphic frameshift mutation eliminates the entire protein. Siglec-XII expression drove several pro-oncogenic phenotypes in cell lines and increased tumor burden in mice challenged with chemical carcinogen and inflammation. Transcriptomic studies yielded a 29-gene signature of Siglec-XII-positive disease and when used as a computational tool for navigating human data sets, pinpointed with surprising precision that SIGLEC12 expression (model) recapitulates a very specific type of colorectal carcinomas (disease) that is associated with mismatch-repair defects and inflammation, disproportionately affects European Americans, and carries a favorable prognosis. They revealed a hitherto-unknown evolutionary genetic mechanism for an ethnic/environmental predisposition of carcinogenesis.


Sujet(s)
Tumeurs colorectales , Inflammation , Animaux , Humains , Souris , Tumeurs colorectales/génétique , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/métabolisme , Inflammation/génétique , Inflammation/métabolisme , Lectines/génétique , Lectines/métabolisme , Lectines liant l'acide sialique apparentées aux immunoglobulines/génétique , Lectines liant l'acide sialique apparentées aux immunoglobulines/métabolisme , Souris knockout
7.
Dev Comp Immunol ; 159: 105222, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-38964676

RÉSUMÉ

Invertebrate lectins exhibit structural diversity and play crucial roles in the innate immune responses by recognizing and eliminating pathogens. In the present study, a novel lectin containing a Gal_Lectin, a CUB and a transmembrane domain was identified from the Pacific oyster Crassostrea gigas (defined as CgGal-CUB). CgGal-CUB mRNA was detectable in all the examined tissues with the highest expression in adductor muscle (11.00-fold of that in haemocytes, p < 0.05). The expression level of CgGal-CUB mRNA in haemocytes was significantly up-regulated at 3, 24, 48 and 72 h (8.37-fold, 12.13-fold, 4.28-fold and 10.14-fold of that in the control group, respectively) after Vibrio splendidus stimulation. The recombinant CgGal-CUB (rCgGal-CUB) displayed binding capability to Mannan (MAN), peptidoglycan (PGN), D-(+)-Galactose and L-Rhamnose monohydrate, as well as Gram-negative bacteria (Escherichia coli, V. splendidus and Vibrio anguillarum), Gram-positive bacteria (Micrococcus luteus, Staphylococcus aureus, and Bacillus sybtilis) and fungus (Pichia pastoris). rCgGal-CUB was also able to agglutinate V. splendidus, and inhibit V. splendidus growth. Furthermore, rCgGal-CUB exhibited the activities of enhancing the haemocyte phagocytosis towards V. splendidus, and the phagocytosis rate of haemocytes was descended in blockage assay with CgGal-CUB antibody. These results suggested that CgGal-CUB served as a pattern recognition receptor to bind various PAMPs and bacteria, and enhanced the haemocyte phagocytosis towards V. splendidus.


Sujet(s)
Crassostrea , Hémocytes , Immunité innée , Lectines , Phagocytose , Vibrio , Animaux , Hémocytes/immunologie , Hémocytes/métabolisme , Crassostrea/immunologie , Vibrio/immunologie , Vibrio/physiologie , Lectines/métabolisme , Lectines/génétique , Lectines/immunologie , Mannanes/métabolisme , Mannanes/immunologie , Domaines protéiques/génétique , Peptidoglycane/immunologie , Peptidoglycane/métabolisme , Galactose/métabolisme , Galactose/immunologie , Infections à Vibrio/immunologie
8.
mBio ; 15(8): e0110724, 2024 Aug 14.
Article de Anglais | MEDLINE | ID: mdl-39041817

RÉSUMÉ

Neisserial adhesin A (NadA) is a meningococcal surface protein included as recombinant antigen in 4CMenB, a protein-based vaccine able to induce protective immune responses against Neisseria meningitidis serogroup B (MenB). Although NadA is involved in the adhesion/invasion of epithelial cells and human myeloid cells, its function in meningococcal physiology is still poorly understood. To clarify the role played by NadA in the host-pathogen interaction, we sought to identify its cellular receptors. We screened a protein microarray encompassing 2,846 human and 297 mouse surface/secreted recombinant proteins using recombinant NadA as probe. Efficient NadA binding was revealed on the paired sialic acid-binding immunoglobulin-type lectins receptors 5 and 14 (Siglec-5 and Siglec-14), but not on Siglec-9 therein used as control. The interaction was confirmed by biochemical tools with the determination of the KD value in the order of nanomolar and the identification of the NadA binding site by hydrogen-deuterium exchange coupled to mass spectrometry. The N-terminal domain of the Siglec-5 that recognizes the sialic acid was identified as the NadA binding domain. Intriguingly, exogenously added recombinant soluble Siglecs, including Siglec-9, were found to decorate N. meningitidis surface in a NadA-dependent manner. However, Siglec-5 and Siglec-14 transiently expressed in CHO-K1 cells endorsed NadA binding and increased N. meningitidis adhesion/invasion while Siglec-9 did not. Taken together, Siglec-5 and Siglec-14 satisfy all features of NadA receptors suggesting a possible role of NadA in the acute meningococcal infection.IMPORTANCEBacteria have developed several strategies for cell colonization and immune evasion. Knowledge of the host and pathogen factors involved in these mechanisms is crucial to build efficacious countermoves. Neisserial adhesin A (NadA) is a meningococcal surface protein included in the anti-meningococcus B vaccine 4CMenB, which mediates adhesion to and invasion of epithelial cells. Although NadA has been shown to bind to other cell types, like myeloid and endothelial cells, it still remains orphan of a defined host receptor. We have identified two strong NadA interactors, Siglec-5 and Siglec-14, which are mainly expressed on myeloid cells. This showcases that NadA is an additional and key player among the Neisseria meningitidis factors targeting immune cells. We thus provide novel insights on the strategies exploited by N. meningitidis during the infection process, which can progress to a severe illness and death.


Sujet(s)
Adhésines bactériennes , Antigènes CD , Antigènes de différenciation des myélomonocytes , Adhérence bactérienne , Interactions hôte-pathogène , Lectines , Humains , Adhésines bactériennes/métabolisme , Adhésines bactériennes/génétique , Antigènes CD/métabolisme , Antigènes CD/génétique , Lectines/métabolisme , Lectines/génétique , Lectines/immunologie , Animaux , Antigènes de différenciation des myélomonocytes/métabolisme , Antigènes de différenciation des myélomonocytes/génétique , Liaison aux protéines , Souris , Cellules CHO , Cricetulus , Neisseria meningitidis/génétique , Neisseria meningitidis/métabolisme , Neisseria meningitidis/immunologie , Protéines recombinantes/métabolisme , Protéines recombinantes/génétique , Lectines liant l'acide sialique apparentées aux immunoglobulines/métabolisme , Lectines liant l'acide sialique apparentées aux immunoglobulines/génétique , Cellules épithéliales/microbiologie , Cellules épithéliales/métabolisme , Cellules épithéliales/immunologie , Infections à méningocoques/microbiologie , Infections à méningocoques/immunologie , Récepteurs de surface cellulaire/métabolisme , Récepteurs de surface cellulaire/génétique , Neisseria meningitidis sérogroupe B/génétique , Neisseria meningitidis sérogroupe B/immunologie , Neisseria meningitidis sérogroupe B/métabolisme
9.
Open Biol ; 14(6): 230451, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38862023

RÉSUMÉ

Plasmodium species encode a unique set of six modular proteins named LCCL lectin domain adhesive-like proteins (LAPs) that operate as a complex and that are essential for malaria parasite transmission from mosquito to vertebrate. LAPs possess complex architectures obtained through unique assemblies of conserved domains associated with lipid, protein and carbohydrate interactions, including the name-defining LCCL domain. Here, we assessed the prevalence of Plasmodium LAP orthologues across eukaryotic life. Our findings show orthologous conservation in all apicomplexans, with lineage-specific repertoires acquired through differential lap gene loss and duplication. Besides Apicomplexa, LAPs are found in their closest relatives: the photosynthetic chromerids, which encode the broadest repertoire including a novel membrane-bound LCCL protein. LAPs are notably absent from other alveolate lineages (dinoflagellates, perkinsids and ciliates), but are encoded by predatory colponemids, a sister group to the alveolates. These results reveal that the LAPs are much older than previously thought and pre-date not only the Apicomplexa but the Alveolata altogether.


Sujet(s)
Évolution moléculaire , Phylogenèse , Plasmodium , Protéines de protozoaire , Protéines de protozoaire/génétique , Protéines de protozoaire/composition chimique , Protéines de protozoaire/métabolisme , Plasmodium/génétique , Plasmodium/métabolisme , Alveolata/génétique , Alveolata/métabolisme , Domaines protéiques , Apicomplexa/génétique , Apicomplexa/métabolisme , Lectines/génétique , Lectines/métabolisme , Lectines/composition chimique
10.
Int J Biol Macromol ; 275(Pt 1): 133311, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38909728

RÉSUMÉ

Lectins are proteins that bind specifically and reversibly to carbohydrates, and some of them have significant anti-tumor activities. Compared to those of lectins from land plants, there are far fewer studies on algal lectins, despite of the high biodiversity of algae. However, canonical strategies based on chromatographic feature-oriented screening cannot satisfy the requirement for algal lectin discovery. In this study, prospecting for novel OAAH family lectins throughout 358 genomes of red algae and cyanobacteria was conducted. Then 35 candidate lectins and 1843 of their simulated mutated forms were virtually screened based on predicted binding specificities to characteristic carbohydrates on cancer cells inferred by a deep learning model. A new lectin, named Siye, was discovered in Kappaphycus alvarezii genome and further verified on different cancer cells. Without causing agglutination of erythrocytes, Siye showed significant cytotoxicity to four human cancer cell lines (IC50 values ranging from 0.11 to 3.95 µg/mL), including breast adenocarcinoma HCC1937, lung carcinoma A549, liver cancer HepG2 and romyelocytic leukemia HL60. And the cytotoxicity was induced through promoting apoptosis by regulating the caspase and the p53 pathway within 24 h. This study testifies the feasibility and efficiency of the genome mining guided by evolutionary theory and artificial intelligence in the discovery of algal lectins.


Sujet(s)
Antinéoplasiques , Simulation numérique , Rhodophyta , Humains , Rhodophyta/composition chimique , Rhodophyta/génétique , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Lectines/pharmacologie , Lectines/composition chimique , Lectines/génétique , Lectines/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Génome ,
11.
BMC Genomics ; 25(1): 643, 2024 Jun 27.
Article de Anglais | MEDLINE | ID: mdl-38937673

RÉSUMÉ

BACKGROUND: The CBM13 family comprises carbohydrate-binding modules that occur mainly in enzymes and in several ricin-B lectins. The ricin-B lectin domain resembles the CBM13 module to a large extent. Historically, ricin-B lectins and CBM13 proteins were considered completely distinct, despite their structural and functional similarities. RESULTS: In this data mining study, we investigate structural and functional similarities of these intertwined protein groups. Because of the high structural and functional similarities, and differences in nomenclature usage in several databases, confusion can arise. First, we demonstrate how public protein databases use different nomenclature systems to describe CBM13 modules and putative ricin-B lectin domains. We suggest the introduction of a novel CBM13 domain identifier, as well as the extension of CAZy cross-references in UniProt to guard the distinction between CAZy and non-CAZy entries in public databases. Since similar problems may occur with other lectin families and CBM families, we suggest the introduction of novel CBM InterPro domain identifiers to all existing CBM families. Second, we investigated phylogenetic, nomenclatural and structural similarities between putative ricin-B lectin domains and CBM13 modules, making use of sequence similarity networks. We concluded that the ricin-B/CBM13 superfamily may be larger than initially thought and that several putative ricin-B lectin domains may display CAZyme functionalities, although biochemical proof remains to be delivered. CONCLUSIONS: Ricin-B lectin domains and CBM13 modules are associated groups of proteins whose database semantics are currently biased towards ricin-B lectins. Revision of the CAZy cross-reference in UniProt and introduction of a dedicated CBM13 domain identifier in InterPro may resolve this issue. In addition, our analyses show that several proteins with putative ricin-B lectin domains show very strong structural similarity to CBM13 modules. Therefore ricin-B lectin domains and CBM13 modules could be considered distant members of a larger ricin-B/CBM13 superfamily.


Sujet(s)
Lectines , Phylogenèse , Domaines protéiques , Ricine , Ricine/composition chimique , Ricine/génétique , Lectines/composition chimique , Lectines/génétique , Lectines/métabolisme , Bases de données de protéines , Séquence d'acides aminés , Similitude de séquences d'acides aminés
12.
Int J Mol Sci ; 25(9)2024 Apr 27.
Article de Anglais | MEDLINE | ID: mdl-38732017

RÉSUMÉ

Intelectins belong to a family of lectins with specific and transitory carbohydrate interaction capabilities. These interactions are related to the activity of agglutinating pathogens, as intelectins play a significant role in immunity. Despite the prominent immune defense function of intelectins, limited information about its structural characteristics and carbohydrate interaction properties is available. This study investigated an intelectin transcript identified in RNA-seq data obtained from the South American lungfish (Lepidosiren paradoxa), namely LpITLN2-B. The structural analyses predicted LpITLN2-B to be a homo-trimeric globular protein with the fibrinogen-like functional domain (FReD), exhibiting a molecular mass of 57 kDa. The quaternary structure is subdivided into three monomers, A, B, and C, and each domain comprises 11 ß-sheets: an anti-parallel ß-sheet, a ß-hairpin, and a disordered ß-sheet structure. Molecular docking demonstrates a significant interaction with disaccharides rather than monosaccharides. The preferential interaction with disaccharides highlights the potential interaction with pathogen molecules, such as LPS and Poly(I:C). The hemagglutination assay inhibited lectins activity, especially maltose and sucrose, highlighting lectin activity in L. paradoxa samples. Overall, our results show the potential relevance of LpITLN2-B in L. paradoxa immune defense against pathogens.


Sujet(s)
Protéines de poisson , Poissons , Immunité innée , Lectines , Animaux , Lectines/composition chimique , Lectines/métabolisme , Lectines/immunologie , Lectines/génétique , Poissons/immunologie , Poissons/génétique , Protéines de poisson/génétique , Protéines de poisson/composition chimique , Protéines de poisson/immunologie , Protéines de poisson/métabolisme , Simulation de docking moléculaire , Séquence d'acides aminés , Protéines liées au GPI/composition chimique , Protéines liées au GPI/métabolisme , Protéines liées au GPI/génétique , Protéines liées au GPI/immunologie
13.
Immunohorizons ; 8(5): 384-396, 2024 May 01.
Article de Anglais | MEDLINE | ID: mdl-38809232

RÉSUMÉ

The mammalian Siglec receptor sialoadhesin (Siglec1, CD169) confers innate immunity against the encapsulated pathogen group B Streptococcus (GBS). Newborn lung macrophages have lower expression levels of sialoadhesin at birth compared with the postnatal period, increasing their susceptibility to GBS infection. In this study, we investigate the mechanisms regulating sialoadhesin expression in the newborn mouse lung. In both neonatal and adult mice, GBS lung infection reduced Siglec1 expression, potentially delaying acquisition of immunity in neonates. Suppression of Siglec1 expression required interactions between sialic acid on the GBS capsule and the inhibitory host receptor Siglec-E. The Siglec1 gene contains multiple STAT binding motifs, which could regulate expression of sialoadhesin downstream of innate immune signals. Although GBS infection reduced STAT1 expression in the lungs of wild-type newborn mice, we observed increased numbers of STAT1+ cells in Siglece-/- lungs. To test if innate immune activation could increase sialoadhesin at birth, we first demonstrated that treatment of neonatal lung macrophages ex vivo with inflammatory activators increased sialoadhesin expression. However, overcoming the low sialoadhesin expression at birth using in vivo prenatal exposures or treatments with inflammatory stimuli were not successful. The suppression of sialoadhesin expression by GBS-Siglec-E engagement may therefore contribute to disease pathogenesis in newborns and represent a challenging but potentially appealing therapeutic opportunity to augment immunity at birth.


Sujet(s)
Animaux nouveau-nés , Souris knockout , Acide N-acétyl-neuraminique , Facteur de transcription STAT-1 , Lectine-1 de type Ig liant l'acide sialique , Infections à streptocoques , Streptococcus agalactiae , Animaux , Souris , Streptococcus agalactiae/immunologie , Acide N-acétyl-neuraminique/métabolisme , Lectine-1 de type Ig liant l'acide sialique/métabolisme , Infections à streptocoques/immunologie , Infections à streptocoques/microbiologie , Facteur de transcription STAT-1/métabolisme , Facteur de transcription STAT-1/génétique , Immunité innée , Souris de lignée C57BL , Poumon/immunologie , Poumon/microbiologie , Poumon/métabolisme , Macrophages alvéolaires/immunologie , Macrophages alvéolaires/métabolisme , Femelle , Macrophages/immunologie , Macrophages/métabolisme , Lectines/métabolisme , Lectines/génétique , Lectines liant l'acide sialique apparentées aux immunoglobulines/métabolisme , Lectines liant l'acide sialique apparentées aux immunoglobulines/génétique , Antigènes CD/métabolisme , Antigènes CD/génétique , Antigènes de différenciation des lymphocytes B
14.
Int J Biol Macromol ; 272(Pt 1): 132682, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38815947

RÉSUMÉ

The thermal stability of trimeric lectin BC2L-CN was investigated and found to be considerably altered when mutating residue 83, originally a threonine, located at the fucose-binding loop. Mutants were analyzed using differential scanning calorimetry and isothermal microcalorimetry. Although most mutations decreased the affinity of the protein for oligosaccharide H type 1, six mutations increased the melting temperature (Tm) by >5 °C; one mutation, T83P, increased the Tm value by 18.2 °C(T83P, Tm = 96.3 °C). In molecular dynamic simulations, the investigated thermostable mutants, T83P, T83A, and T83S, had decreased fluctuations in the loop containing residue 83. In the T83S mutation, the side-chain hydroxyl group of serine formed a hydrogen bond with a nearby residue, suggesting that the restricted movement of the side-chain resulted in fewer fluctuations and enhanced thermal stability. Residue 83 is located at the interface and near the upstream end of the equivalent loop in a different protomer; therefore, fluctuations by this residue likely propagate throughout the loop. Our study of the dramatic change in thermal stability by a single amino acid mutation provides useful insights into the rational design of protein structures, especially the structures of oligomeric proteins.


Sujet(s)
Simulation de dynamique moléculaire , Mutation , Stabilité protéique , Thréonine , Thréonine/composition chimique , Thréonine/génétique , Lectines/composition chimique , Lectines/génétique , Température , Liaison hydrogène
15.
Drug Discov Ther ; 18(3): 178-187, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38777764

RÉSUMÉ

In humans, Entamoeba histolytica is the main pathogen causing various amoebiases, while E. moshkovskii falls between being a pathogen and non-pathogen. The two species have similar behavior patterns but differ significantly in pathogenicity, with previous studies and clinical data indicating that E. moshkovskii has a low level of pathogenicity. Meaningfully, the biological characteristics of E. moshkovskii make it a potential model organism and a protein display platform for studying the functions of important Entamoeba proteins. Here, an Amoeba-pcDNA3.1 vector capable of overexpressing E. histolytica-sourced Igl-C protein was constructed and successfully transfected into E. moshkovskii. High levels of expression of the Igl-C, EGFP, and NeoR genes were identified in Igl-C-transfected trophozoites using qRT-PCR, and they were subsequently confirmed using immunoblotting. Transfection of Igl-C protein improved the adherence and phagocytosis of E. moshkovskii, demonstrating that E. histolytica Igl mediated amoebic adhesion. Moreover, as a manifestation of protein virulence, the ability of post-transfected trophozoites to induce inflammation in host macrophages was also enhanced. In conclusion, this study utilizing the characteristics of E. moshkovskii confirmed its potential to serve as a model organism. E. moshkovskii could replace E. histolytica as the target of gene editing, allowing more efficient study of amoebic pathogenicity.


Sujet(s)
Entamoeba histolytica , Entamoeba , Protéines de protozoaire , Trophozoïtes , Entamoeba/génétique , Entamoeba/pathogénicité , Protéines de protozoaire/génétique , Protéines de protozoaire/métabolisme , Entamoeba histolytica/génétique , Entamoeba histolytica/pathogénicité , Entamoeba histolytica/métabolisme , Trophozoïtes/métabolisme , Phagocytose , Lectines/métabolisme , Lectines/génétique , Humains , Animaux , Transfection , Virulence/génétique , Infection à Entamoeba/parasitologie , Souris
16.
Cell Commun Signal ; 22(1): 271, 2024 May 15.
Article de Anglais | MEDLINE | ID: mdl-38750493

RÉSUMÉ

BACKGROUND: Macrophages are key inflammatory immune cells that orchestrate the initiation and progression of autoimmune diseases. The characters of macrophage in diseases are determined by its phenotype in response to the local microenvironment. Ficolins have been confirmed as crucial contributors to autoimmune diseases, with Ficolin-2 being particularly elevated in patients with autoimmune diseases. However, whether Ficolin-A stimulates macrophage polarization is still poorly understood. METHODS: We investigated the transcriptomic expression profile of murine bone marrow-derived macrophages (BMDMs) stimulated with Ficolin-A using RNA-sequencing. To further confirm a distinct phenotype activated by Ficolin-A, quantitative RT-PCR and Luminex assay were performed in this study. Additionally, we assessed the activation of underlying cell signaling pathways triggered by Ficolin-A. Finally, the impact of Ficolin-A on macrophages were investigated in vivo through building Collagen-induced arthritis (CIA) and Dextran Sulfate Sodium Salt (DSS)-induced colitis mouse models with Fcna-/- mice. RESULTS: Ficolin-A activated macrophages into a pro-inflammatory phenotype distinct to LPS-, IFN-γ- and IFN-γ + LPS-induced phenotypes. The transcriptomic profile induced by Ficolin-A was primarily characterized by upregulation of interleukins, chemokines, iNOS, and Arginase 1, along with downregulation of CD86 and CD206, setting it apart from the M1 and M2 phenotypes. The activation effect of Ficolin-A on macrophages deteriorated the symptoms of CIA and DSS mouse models, and the deletion of Fcna significantly alleviated the severity of diseases in mice. CONCLUSION: Our work used transcriptomic analysis by RNA-Seq to investigate the impact of Ficolin-A on macrophage polarization. Our findings demonstrate that Ficolin-A induces a novel pro-inflammatory phenotype distinct to the phenotypes activated by LPS, IFN-γ and IFN-γ + LPS on macrophages.


Sujet(s)
, Inflammation , Lectines , Macrophages , Souris de lignée C57BL , Phénotype , Animaux , Macrophages/métabolisme , Macrophages/effets des médicaments et des substances chimiques , Lectines/génétique , Lectines/métabolisme , Souris , Inflammation/génétique , Inflammation/anatomopathologie , Activation des macrophages/effets des médicaments et des substances chimiques , Colite/induit chimiquement , Colite/anatomopathologie , Colite/génétique , Polarité de la cellule/effets des médicaments et des substances chimiques , Arthrite expérimentale/génétique , Arthrite expérimentale/anatomopathologie , Transduction du signal/effets des médicaments et des substances chimiques
17.
Nat Commun ; 15(1): 3900, 2024 May 09.
Article de Anglais | MEDLINE | ID: mdl-38724552

RÉSUMÉ

By incompletely understood mechanisms, type 2 (T2) inflammation present in the airways of severe asthmatics drives the formation of pathologic mucus which leads to airway mucus plugging. Here we investigate the molecular role and clinical significance of intelectin-1 (ITLN-1) in the development of pathologic airway mucus in asthma. Through analyses of human airway epithelial cells we find that ITLN1 gene expression is highly induced by interleukin-13 (IL-13) in a subset of metaplastic MUC5AC+ mucus secretory cells, and that ITLN-1 protein is a secreted component of IL-13-induced mucus. Additionally, we find ITLN-1 protein binds the C-terminus of the MUC5AC mucin and that its deletion in airway epithelial cells partially reverses IL-13-induced mucostasis. Through analysis of nasal airway epithelial brushings, we find that ITLN1 is highly expressed in T2-high asthmatics, when compared to T2-low children. Furthermore, we demonstrate that both ITLN-1 gene expression and protein levels are significantly reduced by a common genetic variant that is associated with protection from the formation of mucus plugs in T2-high asthma. This work identifies an important biomarker and targetable pathways for the treatment of mucus obstruction in asthma.


Sujet(s)
Asthme , Protéines liées au GPI , Interleukine-13 , Lectines , Mucine-5AC , Mucus , Enfant , Humains , Asthme/génétique , Asthme/métabolisme , Cytokines , Cellules épithéliales/métabolisme , Protéines liées au GPI/génétique , Protéines liées au GPI/métabolisme , Interleukine-13/génétique , Interleukine-13/métabolisme , Lectines/génétique , Lectines/métabolisme , Mucine-5AC/génétique , Mucine-5AC/métabolisme , Mucus/métabolisme , Muqueuse nasale/métabolisme , Polymorphisme génétique , Muqueuse respiratoire/métabolisme
18.
Cell Rep ; 43(4): 114012, 2024 Apr 23.
Article de Anglais | MEDLINE | ID: mdl-38573856

RÉSUMÉ

Plasmodium falciparum is a human-adapted apicomplexan parasite that causes the most dangerous form of malaria. P. falciparum cysteine-rich protective antigen (PfCyRPA) is an invasion complex protein essential for erythrocyte invasion. The precise role of PfCyRPA in this process has not been resolved. Here, we show that PfCyRPA is a lectin targeting glycans terminating with α2-6-linked N-acetylneuraminic acid (Neu5Ac). PfCyRPA has a >50-fold binding preference for human, α2-6-linked Neu5Ac over non-human, α2-6-linked N-glycolylneuraminic acid. PfCyRPA lectin sites were predicted by molecular modeling and validated by mutagenesis studies. Transgenic parasite lines expressing endogenous PfCyRPA with single amino acid exchange mutants indicated that the lectin activity of PfCyRPA has an important role in parasite invasion. Blocking PfCyRPA lectin activity with small molecules or with lectin-site-specific monoclonal antibodies can inhibit blood-stage parasite multiplication. Therefore, targeting PfCyRPA lectin activity with drugs, immunotherapy, or a vaccine-primed immune response is a promising strategy to prevent and treat malaria.


Sujet(s)
Érythrocytes , Plasmodium falciparum , Polyosides , Protéines de protozoaire , Humains , Antigènes de protozoaire/métabolisme , Antigènes de protozoaire/immunologie , Antigènes de protozoaire/génétique , Érythrocytes/parasitologie , Érythrocytes/métabolisme , Lectines/métabolisme , Lectines/génétique , Paludisme à Plasmodium falciparum/parasitologie , Plasmodium falciparum/métabolisme , Polyosides/métabolisme , Liaison aux protéines , Protéines de protozoaire/métabolisme , Protéines de protozoaire/génétique
19.
Nature ; 629(8010): 165-173, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38632398

RÉSUMÉ

Streptomyces are a genus of ubiquitous soil bacteria from which the majority of clinically utilized antibiotics derive1. The production of these antibacterial molecules reflects the relentless competition Streptomyces engage in with other bacteria, including other Streptomyces species1,2. Here we show that in addition to small-molecule antibiotics, Streptomyces produce and secrete antibacterial protein complexes that feature a large, degenerate repeat-containing polymorphic toxin protein. A cryo-electron microscopy structure of these particles reveals an extended stalk topped by a ringed crown comprising the toxin repeats scaffolding five lectin-tipped spokes, which led us to name them umbrella particles. Streptomyces coelicolor encodes three umbrella particles with distinct toxin and lectin composition. Notably, supernatant containing these toxins specifically and potently inhibits the growth of select Streptomyces species from among a diverse collection of bacteria screened. For one target, Streptomyces griseus, inhibition relies on a single toxin and that intoxication manifests as rapid cessation of vegetative hyphal growth. Our data show that Streptomyces umbrella particles mediate competition among vegetative mycelia of related species, a function distinct from small-molecule antibiotics, which are produced at the onset of reproductive growth and act broadly3,4. Sequence analyses suggest that this role of umbrella particles extends beyond Streptomyces, as we identified umbrella loci in nearly 1,000 species across Actinobacteria.


Sujet(s)
Antibiose , Protéines bactériennes , Toxines bactériennes , Streptomyces , Antibactériens/biosynthèse , Antibactériens/composition chimique , Antibactériens/métabolisme , Antibactériens/pharmacologie , Antibiose/effets des médicaments et des substances chimiques , Protéines bactériennes/composition chimique , Protéines bactériennes/génétique , Protéines bactériennes/métabolisme , Protéines bactériennes/pharmacologie , Protéines bactériennes/ultrastructure , Toxines bactériennes/composition chimique , Toxines bactériennes/génétique , Toxines bactériennes/métabolisme , Toxines bactériennes/pharmacologie , Cryomicroscopie électronique , Lectines/composition chimique , Lectines/génétique , Lectines/métabolisme , Lectines/ultrastructure , Tests de sensibilité microbienne , Modèles moléculaires , Streptomyces/composition chimique , Streptomyces/effets des médicaments et des substances chimiques , Streptomyces/génétique , Streptomyces/croissance et développement , Streptomyces coelicolor/composition chimique , Streptomyces coelicolor/génétique , Streptomyces coelicolor/métabolisme , Streptomyces griseus/effets des médicaments et des substances chimiques , Streptomyces griseus/génétique , Streptomyces griseus/croissance et développement , Streptomyces griseus/métabolisme
20.
Int Immunopharmacol ; 133: 112102, 2024 May 30.
Article de Anglais | MEDLINE | ID: mdl-38652971

RÉSUMÉ

Anaplastic thyroid carcinoma (ATC) is the most aggressive subtype of thyroid cancer with few effective therapies. Though immunotherapies such as targeting PD-1/PD-L1 axis have benefited patients with solid tumor, the druggable immune checkpoints are quite limited in ATC. In our study, we focused on the anti-tumor potential of sialic acid-binding Ig-like lectins (Siglecs) in ATC. Through screening by integrating microarray datasets including 216 thyroid-cancer tissues and single-cell RNA-sequencing, SIGLEC family members CD33, SIGLEC1, SIGLEC10 and SIGLEC15 were significantly overexpressed in ATC, among which SIGLEC15 increased highest and mainly expressed on cancer cells. SIGLEC15high ATC cells are characterized by high expression of serine protease PRSS23 and cancer stem cell marker CD44. Compared with SIGLEC15low cancer cells, SIGLEC15high ATC cells exhibited higher interaction frequency with tumor microenvironment cells. Further study showed that SIGLEC15high cancer cells mainly interacted with T cells by immunosuppressive signals such as MIF-TNFRSF14 and CXCL12-CXCR4. Notably, treatment of anti-SIGLEC15 antibody profoundly increased the cytotoxic ability of CD8+ T cells in a co-culture model and zebrafish-derived ATC xenografts. Consistently, administration of anti-SIGLEC15 antibody significantly inhibited tumor growth and prolonged mouse survival in an immunocompetent model of murine ATC, which was associated with increase of M1/M2, natural killer (NK) cells and CD8+ T cells, and decrease of myeloid-derived suppressor cells (MDSCs). SIGLEC15 inhibited T cell activation by reducing NFAT1, NFAT2, and NF-κB signals. Blocking SIGLEC15 increased the secretion of IFN-γ and IL-2 in vitro and in vivo. In conclusion, our finding demonstrates that SIGLEC15 is an emerging and promising target for immunotherapy in ATC.


Sujet(s)
Immunothérapie , Protéines membranaires , Carcinome anaplasique de la thyroïde , Tumeurs de la thyroïde , Animaux , Humains , Souris , Antinéoplasiques immunologiques/pharmacologie , Antinéoplasiques immunologiques/usage thérapeutique , Lymphocytes T CD8+/immunologie , Lignée cellulaire tumorale , Immunoglobulines , Immunothérapie/méthodes , Lectines/génétique , Lectines/métabolisme , Carcinome anaplasique de la thyroïde/thérapie , Carcinome anaplasique de la thyroïde/immunologie , Carcinome anaplasique de la thyroïde/génétique , Tumeurs de la thyroïde/thérapie , Tumeurs de la thyroïde/immunologie , Tumeurs de la thyroïde/génétique , Microenvironnement tumoral/immunologie , Tests d'activité antitumorale sur modèle de xénogreffe
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE