Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.579
Filter
1.
PeerJ ; 12: e17350, 2024.
Article in English | MEDLINE | ID: mdl-38827297

ABSTRACT

Background: Pancreatic ductal adenocarcinoma (PDAC) is one of the leading causes of cancer-related deaths, with very limited therapeutic options available. This study aims to comprehensively depict the heterogeneity and identify prognostic targets for PDAC with single-cell RNA sequencing (scRNA-seq) analysis. Methods: ScRNA-seq analysis was performed on 16 primary PDAC and three adjacent lesions. A series of analytical methods were applied for analysis in cell clustering, gene profiling, lineage trajectory analysis and cell-to-cell interactions. In vitro experiments including colony formation, wound healing and sphere formation assay were performed to assess the role of makers. Results: A total of 32,480 cells were clustered into six major populations, among which the ductal cell cluster expressing high copy number variants (CNVs) was defined as malignant cells. Malignant cells were further subtyped into five subgroups which exhibited specific features in immunologic and metabolic activities. Pseudotime trajectory analysis indicated that components of various oncogenic pathways were differentially expressed along tumor progression. Furthermore, intensive substantial crosstalk between ductal cells and stromal cells was identified. Finally, genes (REG4 and SPINK1) screened out of differentially expressed genes (DEGs) were upregulated in PDAC cell lines. Silencing either of them significantly impaired proliferation, invasion, migration and stemness of PDAC cells. Conclusions: Our findings offer a valuable resource for deciphering the heterogeneity of malignant ductal cells in PDAC. REG4 and SPINK1 are expected to be promising targets for PDAC therapy.


Subject(s)
Carcinoma, Pancreatic Ductal , Lectins, C-Type , Pancreatic Neoplasms , Single-Cell Analysis , Transcriptome , Trypsin Inhibitor, Kazal Pancreatic , Humans , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/mortality , Trypsin Inhibitor, Kazal Pancreatic/genetics , Trypsin Inhibitor, Kazal Pancreatic/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/metabolism , Prognosis , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Female , Male , Pancreatitis-Associated Proteins
2.
Nat Commun ; 15(1): 4728, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38830864

ABSTRACT

Due to their exceptional solubility and stability, nanobodies have emerged as powerful building blocks for research tools and therapeutics. However, their generation in llamas is cumbersome and costly. Here, by inserting an engineered llama immunoglobulin heavy chain (IgH) locus into IgH-deficient mice, we generate a transgenic mouse line, which we refer to as 'LamaMouse'. We demonstrate that LamaMice solely express llama IgH molecules without association to Igκ or λ light chains. Immunization of LamaMice with AAV8, the receptor-binding domain of the SARS-CoV-2 spike protein, IgE, IgG2c, and CLEC9A enabled us to readily select respective target-specific nanobodies using classical hybridoma and phage display technologies, single B cell screening, and direct cloning of the nanobody-repertoire into a mammalian expression vector. Our work shows that the LamaMouse represents a flexible and broadly applicable platform for a facilitated selection of target-specific nanobodies.


Subject(s)
Camelids, New World , Immunoglobulin Heavy Chains , Mice, Transgenic , Single-Domain Antibodies , Spike Glycoprotein, Coronavirus , Animals , Single-Domain Antibodies/genetics , Single-Domain Antibodies/immunology , Camelids, New World/immunology , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/immunology , Mice , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/chemistry , Lectins, C-Type/metabolism , Lectins, C-Type/immunology , Lectins, C-Type/genetics , SARS-CoV-2/immunology , SARS-CoV-2/genetics , Immunoglobulin E/immunology , Humans , Dependovirus/genetics , Dependovirus/immunology , Immunoglobulin G/immunology , COVID-19/immunology , B-Lymphocytes/immunology
3.
Front Immunol ; 15: 1361351, 2024.
Article in English | MEDLINE | ID: mdl-38846954

ABSTRACT

Background: Gliomas constitute a category of malignant tumors originating from brain tissue, representing the majority of intracranial malignancies. Previous research has demonstrated the pivotal role of CLEC7A in the progression of various cancers, yet its specific implications within gliomas remain elusive. The primary objective of this study was to investigate the prognostic significance and immune therapeutic potential of CLEC7A in gliomas through the integration of bioinformatics and clinical pathological analyses. Methods: This investigation involved examining and validating the relationship between CLEC7A and glioma using samples from Hospital, along with data from TCGA, GEO, GTEx, and CGGA datasets. Subsequently, we explored its prognostic value, biological functions, expression location, and impact on immune cells within gliomas. Finally, we investigated its potential impact on the chemotaxis and polarization of macrophages. Results: The expression of CLEC7A is upregulated in gliomas, and its levels escalate with the malignancy of tumors, establishing it as an independent prognostic factor. Functional enrichment analysis revealed a significant correlation between CLEC7A and immune function. Subsequent examination of immune cell differential expression demonstrated a robust association between CLEC7A and M2 macrophages. This conclusion was further substantiated through single-cell analysis, immunofluorescence, and correlation studies. Finally, the knockout of CLEC7A in M2 macrophages resulted in a noteworthy reduction in macrophage chemotaxis and polarization factors. Conclusion: CLEC7A expression is intricately linked to the pathology and molecular characteristics of gliomas, establishing its role as an independent prognostic factor for gliomas and influencing macrophage function. It could be a promising target for immunotherapy in gliomas.


Subject(s)
Brain Neoplasms , Glioma , Lectins, C-Type , Macrophages , Tumor Microenvironment , Humans , Glioma/immunology , Glioma/genetics , Glioma/pathology , Tumor Microenvironment/immunology , Tumor Microenvironment/genetics , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Prognosis , Brain Neoplasms/immunology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Macrophages/immunology , Macrophages/metabolism , Biomarkers, Tumor/genetics , Gene Expression Regulation, Neoplastic , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism
4.
J Biomed Sci ; 31(1): 53, 2024 May 20.
Article in English | MEDLINE | ID: mdl-38764023

ABSTRACT

BACKGROUND: The C-type lectin family 18 (CLEC18) with lipid and glycan binding capabilities is important to metabolic regulation and innate immune responses against viral infection. However, human CLEC18 comprises three paralogous genes with highly similar sequences, making it challenging to distinguish genetic variations, expression patterns, and biological functions of individual CLEC18 paralogs. Additionally, the evolutionary relationship between human CLEC18 and its counterparts in other species remains unclear. METHODS: To identify the sequence variation and evolutionary divergence of human CLEC18 paralogs, we conducted a comprehensive analysis using various resources, including human and non-human primate reference genome assemblies, human pangenome assemblies, and long-read-based whole-genome and -transcriptome sequencing datasets. RESULTS: We uncovered paralogous sequence variants (PSVs) and polymorphic variants (PVs) of human CLEC18 proteins, and identified distinct signatures specific to each CLEC18 paralog. Furthermore, we unveiled a novel segmental duplication for human CLEC18A gene. By comparing CLEC18 across human and non-human primates, our research showed that the CLEC18 paralogy probably occurred in the common ancestor of human and closely related non-human primates, and the lipid-binding CAP/SCP/TAPS domain of CLEC18 is more diverse than its glycan-binding CTLD. Moreover, we found that certain amino acids alterations at variant positions are exclusive to human CLEC18 paralogs. CONCLUSIONS: Our findings offer a comprehensive profiling of the intricate variations and evolutionary characteristics of human CLEC18.


Subject(s)
Evolution, Molecular , Genetic Variation , Lectins, C-Type , Humans , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Animals , Primates/genetics
5.
Int J Mol Sci ; 25(9)2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38731839

ABSTRACT

CLEC4G, a glycan-binding receptor, has previously been demonstrated to inhibit Aß generation, yet its brain localization and functions in Alzheimer's disease (AD) are not clear. We explored the localization, function, and regulatory network of CLEC4G via experiments and analysis of RNA-seq databases. CLEC4G transcripts and proteins were identified in brain tissues, with the highest expression observed in neurons. Notably, AD was associated with reduced levels of CLEC4G transcripts. Bioinformatic analyses revealed interactions between CLEC4G and relevant genes such as BACE1, NPC1, PILRA, TYROBP, MGAT1, and MGAT3, all displaying a negative correlation trend. We further identified the upstream transcriptional regulators NR2F6 and XRCC4 for CLEC4G and confirmed a decrease in CLEC4G expression in APP/PS1 transgenic mice. This study highlights the role of CLEC4G in protecting against AD progression and the significance of CLEC4G for AD research and management.


Subject(s)
Alzheimer Disease , Lectins, C-Type , Mice, Transgenic , Neurons , Alzheimer Disease/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Animals , Neurons/metabolism , Mice , Humans , Lectins, C-Type/metabolism , Lectins, C-Type/genetics , Brain/metabolism , Brain/pathology , Gene Expression Regulation , Disease Models, Animal
6.
Int J Mol Sci ; 25(10)2024 May 17.
Article in English | MEDLINE | ID: mdl-38791534

ABSTRACT

C-type lectins play a crucial role as pathogen-recognition receptors for the dengue virus, which is responsible for causing both dengue fever (DF) and dengue hemorrhagic fever (DHF). DHF is a serious illness caused by the dengue virus, which exists in four different serotypes: DEN-1, DEN-2, DEN-3, and DEN-4. We conducted a genetic association study, during a significant DEN-2 outbreak in southern Taiwan, to explore how variations in the neck-region length of L-SIGN (also known as CD209L, CD299, or CLEC4M) impact the severity of dengue infection. PCR genotyping was utilized to identify polymorphisms in variable-number tandem repeats. We constructed L-SIGN variants containing either 7- or 9-tandem repeats and transfected these constructs into K562 and U937 cells, and cytokine and chemokine levels were evaluated using enzyme-linked immunosorbent assays (ELISAs) following DEN-2 virus infection. The L-SIGN allele 9 was observed to correlate with a heightened risk of developing DHF. Subsequent results revealed that the 9-tandem repeat was linked to elevated viral load alongside predominant T-helper 2 (Th2) cell responses (IL-4 and IL-10) in K562 and U937 cells. Transfecting K562 cells in vitro with L-SIGN variants containing 7- and 9-tandem repeats confirmed that the 9-tandem repeat transfectants facilitated a higher dengue viral load accompanied by increased cytokine production (MCP-1, IL-6, and IL-8). Considering the higher prevalence of DHF and an increased frequency of the L-SIGN neck's 9-tandem repeat in the Taiwanese population, individuals with the 9-tandem repeat may necessitate more stringent protection against mosquito bites during dengue outbreaks in Taiwan.


Subject(s)
Dengue Virus , Lectins, C-Type , Receptors, Cell Surface , Severe Dengue , Virus Replication , Humans , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Severe Dengue/immunology , Severe Dengue/virology , Severe Dengue/genetics , Dengue Virus/genetics , Dengue Virus/immunology , Virus Replication/genetics , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Male , K562 Cells , Female , U937 Cells , Taiwan/epidemiology , Minisatellite Repeats/genetics , Adult , Cytokines/metabolism , Cytokines/genetics , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Middle Aged , Viral Load
7.
PLoS One ; 19(5): e0303945, 2024.
Article in English | MEDLINE | ID: mdl-38776335

ABSTRACT

Killer cell lectin-like receptor G1 (KLRG1) has traditionally been regarded as an inhibitory receptor of T cell exhaustion in chronic infection and inflammation. However, its exact role in hepatitis B virus (HBV) infection remains elusive. CD8+ T cells from 190 patients with chronic hepatitis B were analyzed ex vivo for checkpoint and apoptosis markers, transcription factors, cytokines and subtypes in 190 patients with chronic hepatitis B. KLRG1+ and KLRG1- CD8+ T cells were sorted for transcriptome analysis. The impact of the KLRG1-E-cadherin pathway on the suppression of HBV replication mediated by virus-specific T cells was validated in vitro. As expected, HBV-specific CD8+ T cells expressed higher levels of KLRG1 and showed an exhausted molecular phenotype and function. However, despite being enriched for the inhibitory molecules, thymocyte selection-associated high mobility group box protein (TOX), eomesodermin (EOMES), and Helios, CD8+ T cells expressing KLRG1 produced significant levels of tumour necrosis factor (TNF)-α, interferon (IFN)-γ, perforin, and granzyme B, demonstrating not exhausted but active function. Consistent with the in vitro phenotypic assay results, RNA sequencing (RNA-seq) data showed that signature effector T cell and exhausted T cell genes were enriched in KLRG1+ CD8+ T cells. Furthermore, in vitro testing confirmed that KLRG1-E-cadherin binding inhibits the antiviral efficacy of HBV-specific CD8+ T cells. Based on these findings, we concluded that KLRG1+ CD8+ T cells are not only a terminally exhausted subgroup but also exhibit functional diversity, despite inhibitory signs in HBV infection.


Subject(s)
CD8-Positive T-Lymphocytes , Hepatitis B virus , Hepatitis B, Chronic , Lectins, C-Type , Receptors, Immunologic , Humans , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Lectins, C-Type/metabolism , Lectins, C-Type/genetics , Receptors, Immunologic/metabolism , Hepatitis B, Chronic/immunology , Hepatitis B, Chronic/virology , Female , Male , Hepatitis B virus/immunology , Adult , Middle Aged , Virus Replication , Cadherins/metabolism , Cadherins/genetics , Perforin/metabolism , Perforin/genetics
8.
Fish Shellfish Immunol ; 149: 109594, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38697376

ABSTRACT

Non-specific cytotoxic cells (NCCs) are vital immune cells involved in teleost's non-specific immunity. As a receptor molecule on the NCCs' surface, the non-specific cytotoxic cell receptor protein 1 (NCCRP-1) is known to play a crucial role in mediating their activity. Nevertheless, there have been limited studies on the signal molecule that transmits signals via NCCRP-1. In this study, a yeast two-hybrid (Y2H) library of tilapia liver and head kidney was constructed and subsequently screened with the bait vector NCCRP-1 of Oreochromis niloticus (On-NCCRP-1) to obtain a C-type lectin (On-CTL) with an interacting protein sequence. Consequently, the full-length sequence of On-CTL was cloned and analyzed. The expression analysis revealed that On-CTL is highly expressed in the liver and is widely distributed in other tissues. Furthermore, On-CTL expression was significantly up-regulated in the brain, intestine, and head kidney following a challenge with Streptococcus agalactiae. A point-to-point Y2H method was also used to confirm the binding between On-NCCRP-1 and On-CTL. The recombinant On-CTL (rOn-CTL) protein was purified. In vitro experiments demonstrated that rOn-CTL can up-regulate the expression of killer effector molecules in NCCs via its interaction with On-NCCRP-1. Moreover, activation of NCCs by rOn-CTL resulted in a remarkable enhancement in their ability to eliminate fathead minnow cells, indicating that rOn-CTL effectively modulates the killing activity of NCCs through the NCC receptor molecule On-NCCRP-1. These findings significantly contribute to our comprehension of the regulatory mechanisms governing NCC activity, paving the way for future research in this field.


Subject(s)
Cichlids , Fish Diseases , Fish Proteins , Lectins, C-Type , Streptococcus agalactiae , Animals , Cichlids/immunology , Cichlids/genetics , Lectins, C-Type/genetics , Lectins, C-Type/immunology , Lectins, C-Type/chemistry , Fish Proteins/genetics , Fish Proteins/immunology , Fish Diseases/immunology , Streptococcus agalactiae/physiology , Streptococcal Infections/immunology , Streptococcal Infections/veterinary , Gene Expression Regulation/immunology , Amino Acid Sequence , Immunity, Innate/genetics , Sequence Alignment/veterinary , Phylogeny , Gene Expression Profiling/veterinary
9.
Sci Rep ; 14(1): 10346, 2024 05 06.
Article in English | MEDLINE | ID: mdl-38710903

ABSTRACT

Mammals are generally resistant to Mycobacterium avium complex (MAC) infections. We report here on a primary immunodeficiency disorder causing increased susceptibility to MAC infections in a canine breed. Adult Miniature Schnauzers developing progressive systemic MAC infections were related to a common founder, and pedigree analysis was consistent with an autosomal recessive trait. A genome-wide association study and homozygosity mapping using 8 infected, 9 non-infected relatives, and 160 control Miniature Schnauzers detected an associated region on chromosome 9. Whole genome sequencing of 2 MAC-infected dogs identified a codon deletion in the CARD9 gene (c.493_495del; p.Lys165del). Genotyping of Miniature Schnauzers revealed the presence of this mutant CARD9 allele worldwide, and all tested MAC-infected dogs were homozygous mutants. Peripheral blood mononuclear cells from a dog homozygous for the CARD9 variant exhibited a dysfunctional CARD9 protein with impaired TNF-α production upon stimulation with the fungal polysaccharide ß-glucan that activates the CARD9-coupled C-type lectin receptor, Dectin-1. While CARD9-deficient knockout mice are susceptible to experimental challenges by fungi and mycobacteria, Miniature Schnauzer dogs with systemic MAC susceptibility represent the first spontaneous animal model of CARD9 deficiency, which will help to further elucidate host defense mechanisms against mycobacteria and fungi and assess potential therapies for animals and humans.


Subject(s)
CARD Signaling Adaptor Proteins , Dog Diseases , Genetic Predisposition to Disease , Genome-Wide Association Study , Mycobacterium avium Complex , Mycobacterium avium-intracellulare Infection , Animals , CARD Signaling Adaptor Proteins/genetics , Dogs , Mycobacterium avium-intracellulare Infection/veterinary , Mycobacterium avium-intracellulare Infection/genetics , Mycobacterium avium-intracellulare Infection/microbiology , Mycobacterium avium Complex/genetics , Dog Diseases/genetics , Dog Diseases/microbiology , Sequence Deletion , Pedigree , Female , Male , Whole Genome Sequencing , Homozygote , Lectins, C-Type/genetics
10.
Int J Mol Sci ; 25(9)2024 May 03.
Article in English | MEDLINE | ID: mdl-38732232

ABSTRACT

C-type lectins in organisms play an important role in the process of innate immunity. In this study, a C-type lectin belonging to the DC-SIGN class of Micropterus salmoides was identified. MsDC-SIGN is classified as a type II transmembrane protein. The extracellular segment of MsDC-SIGN possesses a coiled-coil region and a carbohydrate recognition domain (CRD). The key amino acid motifs of the extracellular CRD of MsDC-SIGN in Ca2+-binding site 2 were EPN (Glu-Pro-Asn) and WYD (Trp-Tyr-Asp). MsDC-SIGN-CRD can bind to four pathogen-associated molecular patterns (PAMPs), including lipopolysaccharide (LPS), glucan, peptidoglycan (PGN), and mannan. Moreover, it can also bind to Gram-positive, Gram-negative bacteria, and fungi. Its CRD can agglutinate microbes and displays D-mannose and D-galactose binding specificity. MsDC-SIGN was distributed in seven tissues of the largemouth bass, among which the highest expression was observed in the liver, followed by the spleen and intestine. Additionally, MsDC-SIGN was present on the membrane of M. salmoides leukocytes, thereby augmenting the phagocytic activity against bacteria. In a subsequent investigation, the expression patterns of the MsDC-SIGN gene and key genes associated with the TLR signaling pathway (TLR4, NF-κB, and IL10) exhibited an up-regulated expression response to the stimulation of Aeromonas hydrophila. Furthermore, through RNA interference of MsDC-SIGN, the expression level of the DC-SIGN signaling pathway-related gene (RAF1) and key genes associated with the TLR signaling pathway (TLR4, NF-κB, and IL10) was decreased. Therefore, MsDC-SIGN plays a pivotal role in the immune defense against A. hydrophila by modulating the TLR signaling pathway.


Subject(s)
Aeromonas hydrophila , Bass , Cell Adhesion Molecules , Fish Diseases , Signal Transduction , Animals , Aeromonas hydrophila/immunology , Bass/immunology , Bass/metabolism , Bass/microbiology , Bass/genetics , Cell Adhesion Molecules/metabolism , Cell Adhesion Molecules/genetics , Fish Diseases/immunology , Fish Diseases/microbiology , Fish Diseases/metabolism , Fish Proteins/metabolism , Fish Proteins/genetics , Fish Proteins/immunology , Gram-Negative Bacterial Infections/immunology , Gram-Negative Bacterial Infections/metabolism , Gram-Negative Bacterial Infections/microbiology , Immunity, Innate , Lectins, C-Type/metabolism , Lectins, C-Type/genetics , Lectins, C-Type/immunology , Pathogen-Associated Molecular Pattern Molecules/metabolism , Pathogen-Associated Molecular Pattern Molecules/immunology , Receptors, Cell Surface/metabolism , Receptors, Cell Surface/genetics , Toll-Like Receptors/metabolism , Toll-Like Receptors/genetics
11.
J Immunol ; 212(12): 1932-1944, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38709167

ABSTRACT

IFN regulatory factor 7 (IRF7) exerts anti-infective effects by promoting the production of IFNs in various bacterial and viral infections, but its role in highly morbid and fatal Candida albicans infections is unknown. We unexpectedly found that Irf7 gene expression levels were significantly upregulated in tissues or cells after C. albicans infection in humans and mice and that IRF7 actually exacerbates C. albicans infection in mice independent of its classical function in inducing IFNs production. Compared to controls, Irf7-/- mice showed stronger phagocytosis of fungus, upregulation of C-type lectin receptor CD209 expression, and enhanced P53-AMPK-mTOR-mediated autophagic signaling in macrophages after C. albicans infection. The administration of the CD209-neutralizing Ab significantly hindered the phagocytosis of Irf7-/- mouse macrophages, whereas the inhibition of p53 or autophagy impaired the killing function of these macrophages. Thus, IRF7 exacerbates C. albicans infection by compromising the phagocytosis and killing capacity of macrophages via regulating CD209 expression and p53-AMPK-mTOR-mediated autophagy, respectively. This finding reveals a novel function of IRF7 independent of its canonical IFNs production and its unexpected role in enhancing fungal infections, thus providing more specific and effective targets for antifungal therapy.


Subject(s)
Autophagy , Candida albicans , Candidiasis , Interferon Regulatory Factor-7 , Lectins, C-Type , Macrophages , Mice, Knockout , Phagocytosis , Receptors, Cell Surface , TOR Serine-Threonine Kinases , Animals , Mice , Phagocytosis/immunology , Autophagy/immunology , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Candidiasis/immunology , Candida albicans/immunology , Candida albicans/physiology , Interferon Regulatory Factor-7/genetics , Interferon Regulatory Factor-7/metabolism , Interferon Regulatory Factor-7/immunology , Macrophages/immunology , Humans , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , TOR Serine-Threonine Kinases/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , Mice, Inbred C57BL , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Signal Transduction/immunology
12.
Int Immunopharmacol ; 135: 112333, 2024 Jun 30.
Article in English | MEDLINE | ID: mdl-38805907

ABSTRACT

Macrophages are one of the important immune cells, which play important roles in innate and adaptive immune. However, the roles of macrophages in food allergy are not thoroughly understood. To investigate the roles of macrophages during food allergy, we focused on the relationship between macrophage polarization and allergic responses induced by tropomyosin (TM) in the present study. Arg 1 and CD206 expressions in the TM group were significantly higher than those of the PBS group, while iNOS and TNF-α expressions were no obvious difference, moreover, the morphology of macrophages stimulated by TM was similar to that of M2 macrophages. These results indicated macrophages were mainly polarized toward M2 phenotypes in vitro. The antibodies, mMCP-1, histamine and cytokines, revealed that macrophages could participate in food allergy, and macrophage polarization was associated with changes in allergic-related factors. The cytokine levels of M2 phenotypes were significantly higher than those of M1 phenotypes in peripheral blood. The mRNA expressions and protein levels of Arg1 and iNOS in the jejunum and peritoneal cells indicated that M2 phenotypes were the major macrophage in these tissues compared with M1 phenotypes. Hence, macrophage polarization plays an important role in food allergy.


Subject(s)
Arginase , Food Hypersensitivity , Macrophages , Mice, Inbred BALB C , Palaemonidae , Tropomyosin , Animals , Tropomyosin/immunology , Food Hypersensitivity/immunology , Mice , Macrophages/immunology , Arginase/metabolism , Palaemonidae/immunology , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide Synthase Type II/genetics , Cytokines/metabolism , Disease Models, Animal , Lectins, C-Type/metabolism , Lectins, C-Type/genetics , Receptors, Cell Surface/metabolism , Receptors, Cell Surface/genetics , Mannose-Binding Lectins/metabolism , Female , Mannose Receptor , Jejunum/immunology , Jejunum/pathology , Cells, Cultured , Histamine/metabolism , Macrophage Activation
13.
Toxicon ; 244: 107740, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38705487

ABSTRACT

Although non-front fanged snakes account for almost two-thirds of snake diversity, most studies on venom composition and evolution focus exclusively on front-fanged species, which comprise most of the clinically relevant accidents. Comprehensive reports on venom composition of non-front fanged snakes are still scarce for several groups. In this study, we address such shortage of knowledge by providing new insights about the venom composition among species of Phalotris, a poorly studied Neotropical dipsadid genus. Phalotris are known for their specialized venom delivery system and toxic venoms, which can cause life-threatening accidents in humans. We evaluate the venom-gland transcriptome of Phalotris, comparing the following three South American species: P. reticulatus for the Araucaria Pine forests, P. lemniscatus for the Pampa grasslands, and P. mertensi for the Brazilian Cerrado. Our results indicate similar venom profiles, in which they share a high expression level of Kunitz-type inhibitors (KUNZ). On the other hand, comparative analyses revealed substantial differences in the expression levels of C-type lectins (CTL) and snake venom metalloproteinases (SVMP). The diverse set of SVMP and CTL isoforms shows signals of positive selection, and we also identified truncated forms of type III SVMPs, which resemble type II and type I SVMPs of viperids. Additionally, we identified a CNP precursor hosting a proline-rich region containing a BPP motif resembling those commonly detected in viperid venoms with hypotensive activity. Altogether, our results suggest an evolutionary history favoring high expression levels of few KUNZ isoforms in Phalotris venoms, contrasting with a highly diverse set of SVMP and CTL isoforms. Such diversity can be comparable with the venom variability observed in some viperids. Our findings highlight the extreme phenotypic diversity of non-front fanged snakes and the importance to allocate greater effort to study neglected groups of Colubroidea.


Subject(s)
Transcriptome , Animals , Snake Venoms/genetics , Lectins, C-Type/genetics , Brazil , Metalloproteases/genetics
14.
Front Immunol ; 15: 1385696, 2024.
Article in English | MEDLINE | ID: mdl-38770013

ABSTRACT

Background: Recent studies have demonstrated a strong association between acute kidney injury (AKI) and chronic kidney disease (CKD), while the unresolved inflammation is believed to be a driving force for this chronic transition process. As a transmembrane pattern recognition receptor, Mincle (macrophage-inducible C-type lectin, Clec4e) was identified to participate in the early immune response after AKI. However, the impact of Mincle on the chronic transition of AKI remains largely unclear. Methods: We performed single-cell RNA sequencing (scRNA-seq) with the unilateral ischemia-reperfusion (UIR) murine model of AKI at days 1, 3, 14 and 28 after injury. Potential effects and mechanism of Mincle on renal inflammation and fibrosis were further validated in vivo utilizing Mincle knockout mice. Results: The dynamic expression of Mincle in macrophages and neutrophils throughout the transition from AKI to CKD was observed. For both cell types, Mincle expression was significantly up-regulated on day 1 following AKI, with a second rise observed on day 14. Notably, we identified distinct subclusters of Minclehigh neutrophils and Minclehigh macrophages that exhibited time-dependent influx with dual peaks characterized with remarkable pro-inflammatory and pro-fibrotic functions. Moreover, we identified that Minclehigh neutrophils represented an "aged" mature neutrophil subset derived from the "fresh" mature neutrophil cluster in kidney. Additionally, we observed a synergistic mechanism whereby Mincle-expressing macrophages and neutrophils sustained renal inflammation by tumor necrosis factor (TNF) production. Mincle-deficient mice exhibited reduced renal injury and fibrosis following AKI. Conclusion: The present findings have unveiled combined persistence of Minclehigh neutrophils and macrophages during AKI-to-CKD transition, contributing to unresolved inflammation followed by fibrosis via TNF-α as a central pro-inflammatory cytokine. Targeting Mincle may offer a novel therapeutic strategy for preventing the transition from AKI to CKD.


Subject(s)
Acute Kidney Injury , Disease Models, Animal , Lectins, C-Type , Macrophages , Membrane Proteins , Mice, Knockout , Neutrophils , Renal Insufficiency, Chronic , Animals , Lectins, C-Type/metabolism , Lectins, C-Type/genetics , Acute Kidney Injury/etiology , Acute Kidney Injury/immunology , Acute Kidney Injury/metabolism , Macrophages/immunology , Macrophages/metabolism , Neutrophils/immunology , Neutrophils/metabolism , Mice , Renal Insufficiency, Chronic/immunology , Renal Insufficiency, Chronic/etiology , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/pathology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Male , Inflammation/immunology , Mice, Inbred C57BL , Reperfusion Injury/immunology , Reperfusion Injury/metabolism , Fibrosis , Disease Progression
15.
In Vivo ; 38(3): 1042-1048, 2024.
Article in English | MEDLINE | ID: mdl-38688646

ABSTRACT

BACKGROUND/AIM: Oral epithelial cells serve as the primary defense against microbial exposure in the oral cavity, including the fungus Candida albicans. Dectin-1 is crucial for recognition of ß-glucan in fungi. However, expression and function of Dectin-1 in oral epithelial cells remain unclear. MATERIALS AND METHODS: We assessed Dectin-1 expression in Ca9-22 (gingiva), HSC-2 (mouth), HSC-3 (tongue), and HSC-4 (tongue) human oral epithelial cells using flow cytometry and real-time polymerase chain reaction. Cell treated with ß-glucan-rich zymosan were evaluated using real-time polymerase chain reaction. Phosphorylation of spleen-associated tyrosine kinase (SYK) was analyzed by western blotting. RESULTS: Dectin-1 was expressed in all four cell types, with high expression in Ca9-22 and HSC-2. In Ca9-22 cells, exposure to ß-glucan-rich zymosan did not alter the mRNA expression of chemokines nor of interleukin (IL)6, IL8, IL1ß, IL17A, and IL17F. Zymosan induced the expression of antimicrobial peptides ß-defensin-1 and LL-37, but not S100 calcium-binding protein A8 (S100A8) and S100A9. Furthermore, the expression of cylindromatosis (CYLD), a negative regulator of nuclear factor kappa B (NF-κB) signaling, was induced. In HSC-2 cells, zymosan induced the expression of IL17A. The expression of tumor necrosis factor alpha-induced protein 3 (TNFAIP3), a negative regulator of NF-κB signaling, was also induced. Expression of other cytokines and antimicrobial peptides remained unchanged. Zymosan induced phosphorylation of SYK in Ca9-22 cells, as well as NF-κB. CONCLUSION: Oral epithelial cells express Dectin-1 and recognize ß-glucan, which activates SYK and induces the expression of antimicrobial peptides and negative regulators of NF-κB, potentially maintaining oral homeostasis.


Subject(s)
Epithelial Cells , Lectins, C-Type , NF-kappa B , Signal Transduction , Syk Kinase , Humans , Lectins, C-Type/metabolism , Lectins, C-Type/genetics , NF-kappa B/metabolism , Syk Kinase/metabolism , Syk Kinase/genetics , Epithelial Cells/metabolism , Epithelial Cells/drug effects , Cell Line , Zymosan/pharmacology , Cytokines/metabolism , Cytokines/genetics , Phosphorylation , Mouth Mucosa/metabolism , Mouth Mucosa/immunology , Pore Forming Cytotoxic Proteins/metabolism , Pore Forming Cytotoxic Proteins/genetics , Antimicrobial Cationic Peptides/genetics , Antimicrobial Cationic Peptides/metabolism
16.
Int J Mol Sci ; 25(8)2024 Apr 13.
Article in English | MEDLINE | ID: mdl-38673902

ABSTRACT

Lectin-like transcript-1 (LLT1) expression is detected in different cancer types and is involved in immune evasion. The present study investigates the clinical relevance of tumoral and stromal LLT1 expression in oral squamous cell carcinoma (OSCC), and relationships with the immune infiltrate into the tumor immune microenvironment (TIME). Immunohistochemical analysis of LLT1 expression was performed in 124 OSCC specimens, together with PD-L1 expression and the infiltration of CD20+, CD4+, and CD8+ lymphocytes and CD68+ and CD163+-macrophages. Associations with clinicopathological variables, prognosis, and immune cell densities were further assessed. A total of 41 (33%) OSCC samples showed positive LLT1 staining in tumor cells and 55 (44%) positive LLT1 in tumor-infiltrating lymphocytes (TILs). Patients harboring tumor-intrinsic LLT1 expression exhibited poorer survival, suggesting an immunosuppressive role. Conversely, positive LLT1 expression in TILs was significantly associated with better disease-specific survival, and also an immune-active tumor microenvironment highly infiltrated by CD8+ T cells and M1/M2 macrophages. Furthermore, the combination of tumoral and stromal LLT1 was found to distinguish three prognostic categories (favorable, intermediate, and adverse; p = 0.029, Log-rank test). Together, these data demonstrate the prognostic relevance of tumoral and stromal LLT1 expression in OSCC, and its potential application to improve prognosis prediction and patient stratification.


Subject(s)
Lectins, C-Type , Receptors, Cell Surface , Squamous Cell Carcinoma of Head and Neck , Tumor Microenvironment , Adult , Female , Humans , Male , B7-H1 Antigen/metabolism , B7-H1 Antigen/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/genetics , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Macrophages/metabolism , Macrophages/immunology , Mouth Neoplasms/pathology , Mouth Neoplasms/immunology , Mouth Neoplasms/metabolism , Mouth Neoplasms/genetics , Mouth Neoplasms/mortality , Prognosis , Squamous Cell Carcinoma of Head and Neck/immunology , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/pathology , Squamous Cell Carcinoma of Head and Neck/mortality , Squamous Cell Carcinoma of Head and Neck/genetics , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Receptors, Cell Surface/genetics , Receptors, Cell Surface/immunology , Receptors, Cell Surface/metabolism , Lectins, C-Type/genetics , Lectins, C-Type/immunology
17.
Mol Med Rep ; 29(6)2024 06.
Article in English | MEDLINE | ID: mdl-38639174

ABSTRACT

Macrophage­inducible C­type lectin receptor (Mincle) is predominantly found on antigen­presenting cells. It can recognize specific ligands when stimulated by certain pathogens such as fungi and Mycobacterium tuberculosis. This recognition triggers the activation of the nuclear factor­κB pathway, leading to the production of inflammatory factors and contributing to the innate immune response of the host. Moreover, Mincle identifies lipid damage­related molecules discharged by injured cells, such as Sin3­associated protein 130, which triggers aseptic inflammation and ultimately hastens the advancement of renal damage, autoimmune disorders and malignancies by fostering tissue inflammation. Presently, research on the functioning of the Mincle receptor in different inflammatory and fibrosis­associated conditions has emerged as a popular topic. Nevertheless, there remains a lack of research on the impact of Mincle in promoting long­lasting inflammatory reactions and fibrosis. Additional investigation is required into the function of Mincle receptors in chronological inflammatory reactions and fibrosis of organ systems, including the progression from inflammation to fibrosis. Hence, the present study showed an overview of the primary roles and potential mechanism of Mincle in inflammation, fibrosis, as well as the progression of inflammation to fibrosis. The aim of the present study was to clarify the potential mechanism of Mincle in inflammation and fibrosis and to offer perspectives for the development of drugs that target Mincle.


Subject(s)
Inflammation , Mycobacterium tuberculosis , Animals , Mice , Inflammation/metabolism , Immunity, Innate , Mycobacterium tuberculosis/metabolism , NF-kappa B , Fibrosis , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Mice, Inbred C57BL
18.
Arterioscler Thromb Vasc Biol ; 44(6): 1318-1329, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38634281

ABSTRACT

BACKGROUND: Tissue resident memory T (TRM) cells are a T-cell subset that resides at the site of prior antigen recognition to protect the body against reoccurring encounters. Besides their protective function, TRM cells have also been implicated in inflammatory disorders. TRM cells are characterized by the expression of CD69 and transcription factors Hobit (homolog of Blimp-1 [B lymphocyte-induced maturation protein 1] in T cells) and Blimp-1. As the majority of T cells in the arterial intima expresses CD69, TRM cells may contribute to the pathogenesis of atherosclerosis as well. Here, we aimed to assess the presence and potential role of TRM cells in atherosclerosis. METHODS: To identify TRM cells in human atherosclerotic lesions, a single-cell RNA-sequencing data set was interrogated, and T-cell phenotypes were compared with that of integrated predefined TRM cells. The presence and phenotype of TRM in atherosclerotic lesions was corroborated using a mouse model that enabled tracking of Hobit-expressing TRM cells. To explore the function of TRM cells during atherogenesis, RAG1-/- (recombination activating gene 1 deficient) LDLr-/- (low-density lipoprotein receptor knockout) mice received a bone marrow transplant from HobitKO/CREBlimp-1flox/flox mice, which exhibit abrogated TRM cell formation, whereafter the mice were fed a Western-type diet for 10 weeks. RESULTS: Human atherosclerotic lesions contained T cells that exhibited a TRM cell-associated gene signature. Moreover, a fraction of these T cells clustered together with predefined TRM cells upon integration. The presence of Hobit-expressing TRM cells in the atherosclerotic lesion was confirmed in mice. These lesion-derived TRM cells were characterized by the expression of CD69 and CD49α. Moreover, we demonstrated that this small T-cell subset significantly affects lesion composition, by reducing the amount of intralesional macrophages and increasing collagen content. CONCLUSIONS: TRM cells, characterized by the expression of CD69 and CD49α, constitute a minor population in atherosclerotic lesions and are associated with increased lesion stability in a Hobit and Blimp-1 knockout mouse model.


Subject(s)
Atherosclerosis , Disease Models, Animal , Immunologic Memory , Macrophages , Memory T Cells , Mice, Inbred C57BL , Plaque, Atherosclerotic , Receptors, LDL , Animals , Atherosclerosis/pathology , Atherosclerosis/immunology , Atherosclerosis/metabolism , Atherosclerosis/genetics , Humans , Memory T Cells/immunology , Memory T Cells/metabolism , Macrophages/metabolism , Macrophages/immunology , Macrophages/pathology , Receptors, LDL/genetics , Receptors, LDL/deficiency , Mice , Male , Mice, Knockout , Antigens, Differentiation, T-Lymphocyte/metabolism , Antigens, Differentiation, T-Lymphocyte/genetics , Lectins, C-Type/metabolism , Lectins, C-Type/genetics , Phenotype , Female , Antigens, CD/metabolism , Antigens, CD/genetics , Aortic Diseases/pathology , Aortic Diseases/immunology , Aortic Diseases/genetics , Aortic Diseases/metabolism
19.
Int J Mol Sci ; 25(7)2024 Apr 04.
Article in English | MEDLINE | ID: mdl-38612837

ABSTRACT

Hashimoto's thyroiditis (HT) and Graves' disease (GD) are common autoimmune endocrine disorders in children. Studies indicate that apart from environmental factors, genetic background significantly contributes to the development of these diseases. This study aimed to assess the prevalence of selected single-nucleotide polymorphisms (SNPs) of Il7R, CD226, CAPSL, and CLEC16A genes in children with autoimmune thyroid diseases. We analyzed SNPs at the locus rs3194051, rs6897932 of IL7R, rs763361 of CD226, rs1010601 of CAPSL, and rs725613 of CLEC16A gene in 56 HT patients, 124 GD patients, and 156 healthy children. We observed significant differences in alleles IL7R (rs6897932) between HT males and the control group (C > T, p = 0.028) and between all GD patients and healthy children (C > T, p = 0.035) as well as GD females and controls (C > T, p = 0.018). Moreover, the C/T genotype was less frequent in GD patients at rs6897932 locus and in HT males at rs1010601 locus. The presence of the T allele in the IL7R (rs6897932) locus appears to have a protective effect against HT in males and GD in all children. Similarly, the presence of the T allele in the CAPSL locus (rs1010601) seems to reduce the risk of HT development in all patients.


Subject(s)
Autoimmune Diseases , Graves Disease , Hashimoto Disease , Child , Female , Male , Humans , Adolescent , Prevalence , Alleles , Hashimoto Disease/genetics , Polymorphism, Single Nucleotide , Graves Disease/genetics , Receptors, Interleukin-7/genetics , Monosaccharide Transport Proteins , Lectins, C-Type/genetics
20.
Pestic Biochem Physiol ; 201: 105852, 2024 May.
Article in English | MEDLINE | ID: mdl-38685211

ABSTRACT

C-type lectins (CTLs) play essential roles in humoral and cellular immune responses of invertebrates. Previous studies have demonstrated the involvement of CTLs in the humoral immunity of Tribolium castaneum, a worldwide pest in stored products. However, the function of CTLs in cellular immunity remains unclear. Here, we identified a CTL gene located on chromosome X and designated it as CTL2 (TcCTL2) from T. castaneum. It encodes a protein of 305 amino acids with a secretion signal peptide and a carbohydrate-recognition domain. TcCTL2 was mainly expressed in the early pupae and primarily distributed in the hemocytes in the late larvae. It was significantly upregulated after larvae were infected with Escherichia coli or Staphylococcus aureus, while knockdown of TcCTL2 exacerbates larval mortality and bacterial colonization after infection. The purified recombinant TcCTL2 (rTcCTL2) can bind to pathogen-associated molecular patterns and microbes and promote hemocyte-mediated encapsulation, melanization and phagocytosis in vitro. rTcCTL2 also induced bacterial agglutination in a Ca2+-dependent manner. Knockdown of TcCTL2 drastically suppressed encapsulation, melanization, and phagocytosis. Furthermore, silencing of TcCTL2 followed by bacterial infection significantly decreased the expression of transcription factors in Toll and IMD pathways, antimicrobial peptides, and prophenoloxidases and phenoloxidase activity. These results unveiled that TcCTL2 mediates both humoral and cellular immunity to promote bacterial clearance and protect T. castaneum from infectious microbes, which will deepen the understanding of the interaction between CTLs and innate immunity in T. castaneum and permit the optimization of pest control strategies by a combination of RNAi technology and bacterial infection.


Subject(s)
Immunity, Cellular , Immunity, Humoral , Insect Proteins , Lectins, C-Type , Staphylococcus aureus , Tribolium , Animals , Lectins, C-Type/metabolism , Lectins, C-Type/genetics , Staphylococcus aureus/immunology , Tribolium/immunology , Tribolium/genetics , Insect Proteins/metabolism , Insect Proteins/genetics , Hemocytes/immunology , Hemocytes/metabolism , Escherichia coli , Phagocytosis , Larva/immunology , Larva/microbiology
SELECTION OF CITATIONS
SEARCH DETAIL
...