Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 812
Filter
1.
J Immunol ; 212(4): 645-662, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38180157

ABSTRACT

Collectin is a crucial component of the innate immune system and plays a vital role in the initial line of defense against pathogen infection. In mammals, collectin kidney 1 (CL-K1) is a soluble collectin that has recently been identified to have significant functions in host defense. However, the evolutionary origins of immune defense of CL-K1 and its mechanism in clearance of pathogenic microorganisms remain unclear, especially in early vertebrates. In this study, the Oreochromis niloticus CL-K1 (OnCL-K1) protein was purified and identified, which was capable of binding to two important pathogens of tilapia, Streptococcus agalactiae and Aeromonas hydrophila. Interestingly, OnCL-K1 exhibited direct bactericidal activity by binding to lipoteichoic acid or LPS on cell walls, disrupting the permeability and integrity of the bacterial membrane in vitro. Upon bacterial challenge, OnCL-K1 significantly inhibited the proliferation of pathogenic bacteria, reduced the inflammatory response, and improved the survival of tilapia. Further research revealed that OnCL-K1 could associate with OnMASPs to initiate and regulate the lectin complement pathway. Additionally, OnCD93 reduced the complement-mediated hemolysis by competing with OnMASPs for binding to OnCL-K1. More importantly, OnCL-K1 could facilitate phagocytosis by collaborating with cell surface CD93 in a lectin pathway-independent manner. Moreover, OnCL-K1 also promoted the formation of phagolysosomes, which degraded and killed ingested bacteria. Therefore, this study reveals the antibacterial response mechanism of CL-K1 in primitive vertebrates, including promoting complement activation, enhancing opsonophagocytosis, and killing of macrophages, as well as its internal links, all of which provide (to our knowledge) new insights into the understanding of the evolutionary origins and regulatory roles of the collectins in innate immunity.


Subject(s)
Macrophages , Opsonization , Animals , Macrophages/metabolism , Complement Activation , Kidney/metabolism , Vertebrates , Collectins/metabolism , Fish Proteins/metabolism , Mammals/metabolism
2.
FASEB J ; 38(5): e23543, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38466278

ABSTRACT

Collectin-11 (CL-11) is a pattern recognition molecule of the lectin pathway capable of interacting with collectin-10 (CL-10) and the MASPs to activate the complement cascade. Alternative splicing of the COLEC11 gene gives rise to two different isoforms found in serum (A and D). These isoforms vary in the length of their collagen-like region, which is involved in the stabilization of the trimeric subunit and the interaction with the MASPs. Here we aim at elucidating the biological differences of naturally occurring CL-11 isoforms A and D. We produced recombinant CL-11 as independent isoforms (CL-11A and CL-11D) and together with CL-10 (CL-10/11A, CL-10/11D). Both CL-11 isoforms associated with CL-10, but CL-11D did so to a lesser extent. CL-10/11 heterocomplexes were composed of trimeric subunits of CL-10 and CL-11, as opposed to CL-10 and CL-11 homotrimers. Heterocomplexes were more stable and migrated with higher apparent molecular weights. Immunoprecipitation of serum CL-11 and subsequent mass spectrometry analysis confirmed that native CL-11 circulates in the form of CL-10/11 heterocomplexes that associate with MASP-1, and MASP-3, but not necessarily MASP-2. Despite a shorter collagen region, CL-11D was capable to bind to the MASPs, suggesting that the missing exon 4 is not required for MASP association CL-11D had a reduced ligand binding compared to full-length CL-11A. Based on its reduced ability to oligomerize, form CL-10/11 heterocomplexes, and bind to ligands, we hypothesize that CL-11D may have a limited complement activation potential compared to full-length CL-11A.


Subject(s)
Alternative Splicing , Mannose-Binding Protein-Associated Serine Proteases , Protein Isoforms/genetics , Collagen , Collectins/genetics
3.
Kidney Int ; 105(3): 524-539, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38158182

ABSTRACT

The urinary tract is constantly exposed to microorganisms. Host defense mechanisms in protection from microbial colonization and development of urinary tract infections require better understanding to control kidney infection. Here we report that the lectin collectin 11 (CL-11), particularly kidney produced, has a pivotal role in host defense against uropathogen infection. CL-11 was found in mouse urine under normal and pathological conditions. Mice with global gene ablation of Colec11 had increased susceptibility to and severity of kidney and to an extent, bladder infection. Mice with kidney-specific Colec11 ablation exhibited a similar disease phenotype to that observed in global Colec11 deficient mice, indicating the importance of kidney produced CL-11 for protection against kidney and bladder infection. Conversely, intravesical or systemic administration of recombinant CL-11 reduced susceptibility to and severity of kidney and bladder infection. Mechanism analysis revealed that CL-11 can mediate several key innate defense mechanisms (agglutination, anti- adhesion, opsonophagocytosis), and limit local inflammatory responses to pathogens. Furthermore, CL-11-mediated innate defense mechanisms can act on clinically relevant microorganisms including multiple antibiotic resistant strains. CL-11 was detectable in eight of 24 urine samples from patients with urinary tract infections but not detectable in urine samples from ten healthy individuals. Thus, our findings demonstrate that CL-11 is a key factor of host defense mechanisms in kidney and bladder infection with therapeutic potential for human application.


Subject(s)
Cystitis , Escherichia coli Infections , Urinary Tract Infections , Humans , Mice , Animals , Urinary Bladder , Kidney , Collectins/genetics
4.
Pharmacol Rev ; 73(2): 792-827, 2021 04.
Article in English | MEDLINE | ID: mdl-33687995

ABSTRACT

The complement system was discovered at the end of the 19th century as a heat-labile plasma component that "complemented" the antibodies in killing microbes, hence the name "complement." Complement is also part of the innate immune system, protecting the host by recognition of pathogen-associated molecular patterns. However, complement is multifunctional far beyond infectious defense. It contributes to organ development, such as sculpting neuron synapses, promoting tissue regeneration and repair, and rapidly engaging and synergizing with a number of processes, including hemostasis leading to thromboinflammation. Complement is a double-edged sword. Although it usually protects the host, it may cause tissue damage when dysregulated or overactivated, such as in the systemic inflammatory reaction seen in trauma and sepsis and severe coronavirus disease 2019 (COVID-19). Damage-associated molecular patterns generated during ischemia-reperfusion injuries (myocardial infarction, stroke, and transplant dysfunction) and in chronic neurologic and rheumatic disease activate complement, thereby increasing damaging inflammation. Despite the long list of diseases with potential for ameliorating complement modulation, only a few rare diseases are approved for clinical treatment targeting complement. Those currently being efficiently treated include paroxysmal nocturnal hemoglobinuria, atypical hemolytic-uremic syndrome, myasthenia gravis, and neuromyelitis optica spectrum disorders. Rare diseases, unfortunately, preclude robust clinical trials. The increasing evidence for complement as a pathogenetic driver in many more common diseases suggests an opportunity for future complement therapy, which, however, requires robust clinical trials; one ongoing example is COVID-19 disease. The current review aims to discuss complement in disease pathogenesis and discuss future pharmacological strategies to treat these diseases with complement-targeted therapies. SIGNIFICANCE STATEMENT: The complement system is the host's defense friend by protecting it from invading pathogens, promoting tissue repair, and maintaining homeostasis. Complement is a double-edged sword, since when dysregulated or overactivated it becomes the host's enemy, leading to tissue damage, organ failure, and, in worst case, death. A number of acute and chronic diseases are candidates for pharmacological treatment to avoid complement-dependent damage, ranging from the well established treatment for rare diseases to possible future treatment of large patient groups like the pandemic coronavirus disease 2019.


Subject(s)
COVID-19/epidemiology , COVID-19/physiopathology , Complement System Proteins/physiology , Rare Diseases/physiopathology , Collectins/metabolism , Complement Activating Enzymes/metabolism , Complement C3/metabolism , Complement Inactivating Agents/pharmacology , Genetic Therapy/methods , Humans , Inflammation Mediators/metabolism , Lectins/metabolism , Mannose-Binding Protein-Associated Serine Proteases/metabolism , Pandemics , SARS-CoV-2 , Synapses/metabolism , Ficolins
5.
Int J Mol Sci ; 25(5)2024 Feb 21.
Article in English | MEDLINE | ID: mdl-38473757

ABSTRACT

Collectin-K1 (CL-K1) is a multifunctional C-type lectin that has been identified as playing a crucial role in innate immunity. It can bind to carbohydrates on pathogens, leading to direct neutralization, agglutination, and/or opsonization, thereby inhibiting pathogenic infection. In this study, we investigated a homolog of CL-K1 (OnCL-K1) in Nile tilapia (Oreochromis niloticus) and its role in promoting the clearance of the pathogen Streptococcus agalactiae (S. agalactiae) and enhancing the antibacterial ability of the fish. Our analysis of bacterial load displayed that OnCL-K1 substantially reduced the amount of S. agalactiae in tissues of the liver, spleen, anterior kidney, and brain in Nile tilapia. Furthermore, examination of tissue sections revealed that OnCL-K1 effectively alleviated tissue damage and inflammatory response in the liver, anterior kidney, spleen, and brain tissue of tilapia following S. agalactiae infection. Additionally, OnCL-K1 was found to decrease the expression of the pro-inflammatory factor IL-6 and migration inhibitor MIF, while increasing the expression of anti-inflammatory factor IL-10 and chemokine IL-8 in the spleen, anterior kidney, and brain tissues of tilapia. Moreover, statistical analysis of survival rates demonstrated that OnCL-K1 significantly improved the survival rate of tilapia after infection, with a survival rate of 90%. Collectively, our findings suggest that OnCL-K1 plays a vital role in the innate immune defense of resisting bacterial infection in Nile tilapia. It promotes the removal of bacterial pathogens from the host, inhibits pathogen proliferation in vivo, reduces damage to host tissues caused by pathogens, and improves the survival rate of the host.


Subject(s)
Cichlids , Streptococcal Infections , Tilapia , Animals , Cichlids/metabolism , Streptococcus agalactiae , Gene Expression Regulation , Amino Acid Sequence , Tilapia/metabolism , Collectins/genetics
6.
Lab Invest ; 103(7): 100130, 2023 07.
Article in English | MEDLINE | ID: mdl-36925047

ABSTRACT

Collectin subfamily member 10 (COLEC10), a C-type lectin mainly expressed in the liver, is involved in the development of hepatocellular carcinoma (HCC). However, its underlying molecular mechanism in HCC progression remains unknown. In this study, reduced COLEC10 expression in tumor tissues was validated using various HCC cohorts and was associated with poor patient prognosis. COLEC10 overexpression attenuated HCC cell growth and migration abilities in vitro and in vivo. We identified that COLEC10 was a novel interactor of 78-kDa glucose-regulated protein (GRP78), a master modulator of the unfolded protein response in the endoplasmic reticulum (ER). COLEC10 overexpression potentiated ER stress in HCC cells, as demonstrated by elevated expression levels of phosphorylated protein kinase RNA-like ER kinase, phosphorylated inositol-requiring protein 1α, activating transcription factor 4, DNA damage-inducible transcript 3, and X-box-binding protein 1s. The ER in COLEC10-overexpressing cells also showed a dilated and fragmented pattern. Mechanistically, COLEC10 overexpression increases GRP78 occupancy through direct binding by the C-terminal carbohydrate recognition domain in the ER, which released and activated the ER stress transducers protein kinase RNA-like ER kinase and phosphorylated inositol-requiring protein 1α, triggering the unfolded protein response activity. COLEC10-overexpressing HCC cells generated a relatively high reactive oxygen species level and switched to apoptotic cell death under sorafenib-treated conditions. Our study provides the first novel view that COLEC10 inhibits HCC progression by regulating GRP78-mediated ER stress signaling and may serve as a promising therapeutic and prognostic biomarker.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Carcinoma, Hepatocellular/metabolism , Endoplasmic Reticulum Chaperone BiP , Liver Neoplasms/metabolism , Endoplasmic Reticulum Stress , Apoptosis , RNA , Protein Kinases , Collectins
7.
Fish Shellfish Immunol ; 140: 108972, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37488038

ABSTRACT

With the tremendous success of the artificial breeding of Hexagrammos otakii, the yield has been substantially improved. However, intensive farming often results in bacterial diseases; hence it is imperative to find new antimicrobial molecules. In the present study, we identified a homologous cDNA fragment of collectin-10 from H. otakii, designated as HoCL-10. The cDNA length is 899 bp, which contains an open reading frame (ORF) of 816 bp encoding a secreted protein with 272 amino acid residues. The peptide of HoCL-10 contains an N-terminal collagen domain, a neck region, and a C-terminal carbohydrate recognition domain. The qRT-PCR results revealed that HoCL-10 mRNA was highest expressed in the liver and skin and was significantly induced post-LPS stimulation. The sugar and bacteria binding assay suggested that the recombinant HoCL-10 (rHoCL-10) could recognize various pathogen-associated molecular patterns (PAMPs) and bacteria. For effect on cells, rHoCL-10 enhanced the phagocytosis and migration ability of the macrophage indicated using pro-phagocytosis assay and trans-well assay. To determine the role of HoCL-10 in the complement system, the interaction between HoCL-10 and mannose-binding lectin associated serine protease 1, 2 (MASP-1, 2) were analyzed and demonstrated using ELISA and Far-western. And in vivo, the concentration of membrane-attack complex (MAC) in fish plasma was significantly down-regulated post-injection with HoCL-10 antibody. Finally, the bacteria challenge experiment was performed, implying that HoCL-10 may assist the host in defending against microbial invasion. The findings suggest that HoCL-10 may play crucial roles in host defense against microorganisms, possibly through opsonizing pathogens and activating the complement system.


Subject(s)
Bacterial Infections , Perciformes , Animals , DNA, Complementary , Bacteria/genetics , Complement Activation , Perciformes/genetics , Complement System Proteins , Carbohydrates , Collectins/genetics
8.
Fish Shellfish Immunol ; 133: 108543, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36669601

ABSTRACT

The collectin subfamily member 11 (Colec11), plays an important role in innate immunity as a pattern recognition molecule. In the present study, a colec11 homolog was identified and characterised from Qihe crucian carp, namely, Ca-colec11. The full-length cDNA of Ca-colec11 was composed of 1129 bp, with a 99 bp 5'-untranslated region (UTR), 816 bp open reading frame (ORF) encoding a 271-aa protein and 214 bp 3'-UTR with a polyadenylation signal sequence (aataaa) and a poly(A) tail. The deduced amino acid sequence of Ca-Colec11 contained a si gnal peptide, collagen domain, neck region and carbohydrate-recognition domain (CRD), which had four conserved cysteine residues (Cys170-Cys256 and Cys242-Cys264) and an EPN/WND motif required for carbohydrate-binding specificity. Tissue expression profile analysis by quantitative real-time polymerase chain reaction (RT-qPCR) showed that Ca-colec11 was ubiquitously distributed in the tested tissues and highly expressed in the liver. The gene expression levels of Ca-colec11 were evidently up-regulated in the liver, spleen, kidney and head kidney after infection with A. hydrophila and S. aureus. The recombinant Ca-Colec11 (rCa-Colec11) purified from Escherichia coli BL21 (DE3) could agglutinate A. hydrophila and S. aureus, and it possessed haemagglutination activity against rabbit erythrocytes, which was inhibited by various carbohydrates, including d-Mannose, N-Acetyl-d-mannosamine, l-Fucose, d-Glucose, N-Acetyl-d-glucosamine, d-Galactose, LPS and PGN. Furthermore, rCa-Colec11 could inhibit the growth of A. hydrophila and S. aureus. These findings collectively demonstrated that Ca-Colec11, as a PRR, could play a role in the immune defence of Qihe crucian carp.


Subject(s)
Carps , Goldfish , Animals , Rabbits , Carps/genetics , Carps/metabolism , Staphylococcus aureus/metabolism , Aeromonas hydrophila/genetics , Base Sequence , Fish Proteins/chemistry , Collectins/genetics , Carbohydrates , Immunity, Innate/genetics , Phylogeny
9.
Fish Shellfish Immunol ; 132: 108491, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36503059

ABSTRACT

It has been known that vitamin D3 (VD3) not only plays an important role in regulating calcium and phosphorus metabolism in animals, but also has extensive effects on immune functions. In this study, the mechanism how VD3 influences bactericidal ability in turbot was explored. The transcriptomic analysis identified that dietary VD3 significantly upregulated the gene expression of C-type lectin receptors (CLRs), including mannose receptors (mrc1, mrc2, pla2r1) and collectins (collectin 11 and collectin 12) in turbot intestine. Further results obtained from in vitro experiments confirmed that the gene expression of mannose receptors and collectins in head-kidney macrophages (HKMs) of turbot was induced after the cells were incubated with different concentrations of VD3 (0, 1, 10 nM) or 1,25(OH)2D3 (0, 10, 100 pM). Meanwhile, both phagocytosis and bactericidal functions of HKMs were significantly improved in VD3 or 1,25(OH)2D3-incubated HKMs. Furthermore, phagocytosis and bacterial killing of HKMs decreased after collectin 11 was knocked down. Moreover, VD3-enhanced antibacterial activities diminished in collectin 11-interfered cells. Interestingly, the evidence was provided in the present study that inactive VD3 could be metabolized into active 1,25(OH)2D3 via hydroxylases encoded by cyp27a1 and cyp27b1 in fish macrophages. In conclusion, VD3 could be metabolized to 1,25(OH)2D3 in HKMs, which promoted the expression of CLRs in macrophages, leading to enhanced bacterial clearance.


Subject(s)
Cholecalciferol , Flatfishes , Animals , Cholecalciferol/pharmacology , Cholecalciferol/metabolism , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Mannose Receptor , Flatfishes/genetics , Flatfishes/metabolism , Macrophages , Collectins , Kidney/metabolism
10.
Genet Med ; 24(8): 1653-1663, 2022 08.
Article in English | MEDLINE | ID: mdl-35511137

ABSTRACT

PURPOSE: Emerging evidence suggest that infection-dependent hyperactivation of complement system (CS) may worsen COVID-19 outcome. We investigated the role of predicted high impact rare variants - referred as qualifying variants (QVs) - of CS genes in predisposing asymptomatic COVID-19 in elderly individuals, known to be more susceptible to severe disease. METHODS: Exploiting exome sequencing data and 56 CS genes, we performed a gene-based collapsing test between 164 asymptomatic subjects (aged ≥60 years) and 56,885 European individuals from the Genome Aggregation Database. We replicated this test comparing the same asymptomatic individuals with 147 hospitalized patients with COVID-19. RESULTS: We found an enrichment of QVs in 3 genes (MASP1, COLEC11, and COLEC10), which belong to the lectin pathway, in the asymptomatic cohort. Analyses of complement activity in serum showed decreased activity of lectin pathway in asymptomatic individuals with QVs. Finally, we found allelic variants associated with asymptomatic COVID-19 phenotype and with a decreased expression of MASP1, COLEC11, and COLEC10 in lung tissue. CONCLUSION: This study suggests that genetic rare variants can protect from severe COVID-19 by mitigating the activity of lectin pathway and prothrombin. The genetic data obtained through ES of 786 asymptomatic and 147 hospitalized individuals are publicly available at http://espocovid.ceinge.unina.it/.


Subject(s)
COVID-19 , Aged , COVID-19/genetics , Collectins/genetics , Collectins/metabolism , Germ Cells , Humans , Lectins/genetics , SARS-CoV-2 , Exome Sequencing
11.
BMC Cancer ; 22(1): 380, 2022 Apr 09.
Article in English | MEDLINE | ID: mdl-35397600

ABSTRACT

BACKGROUND: Hepatocellular carcinoma (HCC) is the third-most deadly cancer worldwide. More breakthroughs are needed in the clinical practice for liver cancer are needed, and new treatment strategies are required. This study aims to determine the significant differences in genes associated with LIHC and further analyze its prognostic value further. METHODS: Here, we used the TCGA-LIHC database and the profiles of GSE25097 from GEO to explore the differentially co-expressed genes in HCC tissues compared with paratumor (or healthy) tissues. Then, we utilized WGCNA to screen differentially co-expressed genes. Finally, we explored the function of FYN in HCC cells and xenograft tumor models. RESULTS: We identified ten hub genes in the protein-protein interaction (PPI) network, but only three (COLEC10, TGFBR3, and FYN) appeared closely related to the prognosis. The expression of FYN was positively correlated with the prognosis of HCC patients. The xenograft model showed that overexpression of FYN could significantly inhibit malignant tumor behaviors and promote tumor cell apoptosis. CONCLUSION: Thus, FYN may be central to the development of LIHC and maybe a novel biomarker for clinical diagnosis and treatment.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Proto-Oncogene Proteins c-fyn , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/pathology , Collectins/genetics , Collectins/metabolism , Computational Biology , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Humans , Liver Neoplasms/pathology , Prognosis , Proto-Oncogene Proteins c-fyn/genetics , Proto-Oncogenes
12.
Am J Med Genet A ; 188(10): 3110-3117, 2022 10.
Article in English | MEDLINE | ID: mdl-35943032

ABSTRACT

Bi-allelic variants in COLEC11 and MASP1 have been associated with 3MC syndrome, a clinical entity made of up four rare autosomal recessive disorders: Carnevale, Mingarelli, Malpuech, and Michels syndromes, characterized by variable expression of facial dysmorphia, cleft lip/palate, postnatal growth deficiency, hearing loss, cognitive impairment, craniosynostosis, radioulnar synostosis, and genital and vesicorenal anomalies. More recently, bi-allelic variants in COLEC10 have been described to be associated with 3MC syndrome. Syndromic features seen in 3MC syndrome are thought to be due to disruption of the chemoattractant properties that influence neural crest cell migration. We identified nine individuals from five families of Ashkenazi Jewish descent with homozygosity of the c.311G > T (p.Gly104Val) variant in COLEC10 and phenotype consistent with 3MC syndrome. Carrier frequency was calculated among 52,278 individuals of Jewish descent. Testing revealed 400 carriers out of 39,750 individuals of Ashkenazi Jewish descent, giving a carrier frequency of 1 in 99 or 1.01%. Molecular protein modeling suggested that the p.Gly104Val substitution alters local conformation. The c.311G > T (p.Gly104Val) variant likely represents a founder variant, and homozygosity is associated with features of 3MC syndrome. 3MC syndrome should be in the differential diagnosis for individuals with short stature, radioulnar synostosis, cleft lip and cleft palate.


Subject(s)
Abnormalities, Multiple , Cleft Lip , Cleft Palate , Abnormalities, Multiple/diagnosis , Abnormalities, Multiple/genetics , Cleft Lip/diagnosis , Cleft Lip/genetics , Cleft Palate/diagnosis , Cleft Palate/genetics , Collectins/genetics , Humans , Jews/genetics , Mutation , Phenotype , Radius/abnormalities , Synostosis , Ulna/abnormalities
13.
Fish Shellfish Immunol ; 131: 527-536, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36265742

ABSTRACT

CL-11 (Collectin-11, also known as Collectin kidney-1 or CL-K1) is a member of collectin family that works as a pattern recognition molecule (PRM) and participating in lectin-complement pathway in host defense against pathogens. We identified the CL-11 homologue SsCL-11 in black rockfish (Sebastes schlegelii) and investigated the functional characteristics in this study. The SsCL-11 has conserved protein modules, i.e. an N-terminal hydrophobic region, a collagen-like region, an α-helical neck region and a carbohydrate recognition domain (CRD). SsCL-11 has varying degrees of expressions in difference tissues, among which the highest expression is observed in liver. It also shows induced expressions in immune-related tissues following Aeromonas salmonicida (A. salmonicida) infection. In addition, SsCL-11 exhibits binding abilities to different kinds of carbohydrates, pathogen-associated molecular patterns (PAMPs) and bacteria. It exhibits comparatively strong binding to l-fucose, d-mannose, and d-glucose, which is consistent with the functional EPN motif in its CRD. SsCL-11 also shows agglutinating effects on various bacteria in the presence of Ca2+. Furthermore, SsCL-11 is confirmed to be a secretory lectin and can form multimers. These findings collectively demonstrate that SsCL-11 can function as a recognition molecule in pathogen resistance in black rockfish, which will promote our understanding of immunological roles of fish collectins.


Subject(s)
Fish Diseases , Perciformes , Animals , Fish Proteins , Amino Acid Sequence , Collectins , Pathogen-Associated Molecular Pattern Molecules
14.
J Immunol ; 204(7): 1919-1928, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32094208

ABSTRACT

The complement system constitutes an important part of the innate immune system. The collectins and the ficolins are soluble pattern recognition molecules that contribute to complement activation via the lectin pathway. During previous experiments with ficolin-2 and ficolin-3, we have observed that the molecules may interact. We therefore hypothesized the existence of stable ficolin-2/-3 heterocomplexes. We could demonstrate ficolin-2/-3 heterocomplexes in normal human serum and plasma by ELISA using Abs specific for ficolin-2 and ficolin-3. The formation of heteromeric protein complexes were validated by coimmunoprecipitation and Western blot analysis. When recombinant ficolin-2 and recombinant ficolin-3 were mixed, no complexes were formed. However, when coexpressing ficolin-2 and ficolin-3 in Chinese hamster ovary cells, we could detect ficolin-2/-3 heterocomplexes in the supernatant. Furthermore, we measured concentration of the ficolin-2/-3 heterocomplexes in arbitrary units in 94 healthy individuals. We also established the relationship between the concentrations of ficolin-2, ficolin-3, and the ficolin-2/-3 heterocomplexes. We observed that the concentration of the ficolin-2/-3 heterocomplex correlated significantly with ficolin-2 (ρ: 0.24, p < 0.018) and ficolin-3 concentrations (ρ: 0.46, p < 0.0001). In conclusion, we describe a novel protein complex between ficolin-2 and ficolin-3 present in serum and plasma, which might be of additional biological relevance apart from the native ficolin-2 and ficolin-3 molecules.


Subject(s)
Lectins/blood , Animals , CHO Cells , Cell Line , Collectins/metabolism , Complement Activation/physiology , Complement Pathway, Mannose-Binding Lectin/physiology , Complement System Proteins/metabolism , Cricetulus , Humans , Mannose-Binding Protein-Associated Serine Proteases/metabolism , Mice , Ficolins
15.
J Immunol ; 204(6): 1598-1606, 2020 03 15.
Article in English | MEDLINE | ID: mdl-32041782

ABSTRACT

C1q/TNF-related protein (CTRP) 6 is a member of the CTRP protein family associated with the regulation of cellular and endocrine processes. CTRP6 contains collagen and globular structures, resembling the pattern recognition molecules (PRMs) of the classical and lectin complement pathways. We expressed human CTRP6 in Chinese hamster ovary cells and investigated the binding to different putative ligands (acetylated BSA [AcBSA], zymosan, mannan, and LPS from Escherichia coli and Salmonella as well as to the monosaccharides l-fucose, d-mannose, N-acetylglucosamine, N-acetylgalactosamine, and galactose). Furthermore, we investigated the binding of CTRP6 to various Gram-negative bacteria as well as PRMs and enzymes of the lectin complement pathway. We found that CTRP6 bound to AcBSA and to a lesser extent to zymosan. Using EDTA as chelating agent, we observed an increased binding to AcBSA, zymosan and the two strains of LPS. We detected no binding to mannan and BSA. We identified l-fucose as a ligand for CTRP6 and that it bound to certain enteroaggregative Escherichia coli and Pseudomonas aeruginosa isolates, whereas to other bacterial isolates, no binding was observed. CTRP6 did not appear to interact directly with the activating enzymes of the lectin pathway; however, we could show the specific recruitment of collectin-11 and subsequent initiation of the complement cascade through deposition of C4. In conclusion, our results demonstrate the binding of CTRP6 to a variety of microbial and endogenous ligands identifying CTRP6 as a novel human lectin and PRM of importance for complement recognition and innate immunity.


Subject(s)
Antigens, Bacterial/metabolism , Collagen/metabolism , Collectins/metabolism , Complement C4/metabolism , Complement Pathway, Mannose-Binding Lectin/immunology , Animals , Antigens, Bacterial/immunology , CHO Cells , Collagen/genetics , Collagen/isolation & purification , Complement Activation , Cricetulus , Escherichia coli/immunology , Escherichia coli/metabolism , Ligands , Mannose-Binding Protein-Associated Serine Proteases/metabolism , Pseudomonas aeruginosa/immunology , Pseudomonas aeruginosa/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism
16.
Int J Mol Sci ; 23(24)2022 Dec 16.
Article in English | MEDLINE | ID: mdl-36555720

ABSTRACT

Establishing the rapid and accurate diagnosis of sepsis is a key component to the improvement of clinical outcomes. The ability of analytical platforms to rapidly detect pathogen-associated molecular patterns (PAMP) in blood could provide a powerful host-independent biomarker of sepsis. A novel concept was investigated based on the idea that a pre-bound and fluorescent ligand could be released from lectins in contact with high-affinity ligands (such as PAMPs). To create fluorescent ligands with precise avidity, the kinetically followed TEMPO oxidation of yeast mannan and carbodiimide coupling were used. The chemical modifications led to decreases in avidity between mannan and human collectins, such as the mannan-binding lectin (MBL) and human surfactant protein D (SP-D), but not in porcine SP-D. Despite this effect, these fluorescent derivatives were captured by human lectins using highly concentrated solutions. The resulting fluorescent beads were exposed to different solutions, and the results showed that displacements occur in contact with higher affinity ligands, proving that two-stage competition processes can occur in collectin carbohydrate recognition mechanisms. Moreover, the fluorescence loss depends on the discrepancy between the respective avidities of the recognized ligand and the fluorescent mannan. Chemically modulated fluorescent ligands associated with a diversity of collectins may lead to the creation of diagnostic tools suitable for multiplex array assays and the identification of high-avidity ligands.


Subject(s)
Collectins , Sepsis , Humans , Animals , Swine , Pulmonary Surfactant-Associated Protein D/chemistry , Mannans/metabolism , Ligands , Lectins/metabolism
17.
J Biol Chem ; 295(27): 9157-9170, 2020 07 03.
Article in English | MEDLINE | ID: mdl-32424040

ABSTRACT

C-type lectins that contain collagen-like domains are known as collectins. These proteins are present both in the circulation and in extravascular compartments and are central players of the innate immune system, contributing to first-line defenses against viral, bacterial, and fungal pathogens. The collectins mannose-binding lectin (MBL) and surfactant protein D (SP-D) are regulated by tissue fibroblasts at extravascular sites via an endocytic mechanism governed by urokinase plasminogen activator receptor-associated protein (uPARAP or Endo180), which is also a collagen receptor. Here, we investigated the molecular mechanisms that drive the uPARAP-mediated cellular uptake of MBL and SP-D. We found that the uptake depends on residues within a protruding loop in the fibronectin type-II (FNII) domain of uPARAP that are also critical for collagen uptake. Importantly, however, we also identified FNII domain residues having an exclusive role in collectin uptake. We noted that these residues are absent in the related collagen receptor, the mannose receptor (MR or CD206), which consistently does not interact with collectins. We also show that the second C-type lectin-like domain (CTLD2) is critical for the uptake of SP-D, but not MBL, indicating an additional level of complexity in the interactions between collectins and uPARAP. Finally, we demonstrate that the same molecular mechanisms enable uPARAP to engage MBL immobilized on the surface of pathogens, thereby expanding the potential biological implications of this interaction. Our study reveals molecular details of the receptor-mediated cellular regulation of collectins and offers critical clues for future investigations into collectin biology and pathology.


Subject(s)
Collectins/metabolism , Endocytosis/physiology , Receptors, Mitogen/genetics , Animals , CHO Cells , Carrier Proteins/metabolism , Collagen/metabolism , Cricetulus , Fibroblasts/metabolism , HEK293 Cells , Humans , Lectins, C-Type , Mannose Receptor , Mannose-Binding Lectin/metabolism , Mannose-Binding Lectins , Membrane Glycoproteins/metabolism , Pulmonary Surfactant-Associated Protein D/metabolism , Receptors, Cell Surface , Receptors, Collagen/metabolism , Receptors, Mitogen/metabolism , Receptors, Urokinase Plasminogen Activator/genetics , Receptors, Urokinase Plasminogen Activator/metabolism
18.
Eur J Immunol ; 50(5): 624-642, 2020 05.
Article in English | MEDLINE | ID: mdl-32246830

ABSTRACT

Maintenance of homeostasis at body barriers that are constantly challenged by microbes, toxins and potentially bioactive (macro)molecules requires complex, highly orchestrated mechanisms of protection. Recent discoveries in respiratory research have shed light on the unprecedented role of airway epithelial cells (AEC), which, besides immune cells homing to the lung, also significantly contribute to host defence by expressing membrane-bound and soluble pattern recognition receptors (sPRR). Recent evidence suggests that distinct, evolutionary ancient, sPRR secreted by AEC might become activated by usually innocuous proteins, commonly referred to as allergens. We here provide a systematic overview on sPRR detectable in the mucus lining of AEC. Some of them become actively produced and secreted by AECs (like the pentraxins C-reactive protein and pentraxin 3; the collectins mannose binding protein and surfactant proteins A and D; H-ficolin; serum amyloid A; and the complement components C3 and C5). Others are elaborated by innate and adaptive immune cells such as monocytes/macrophages and T cells (like the pentraxins C-reactive protein and pentraxin 3; L-ficolin; serum amyloid A; and the complement components C3 and C5). Herein we discuss how sPRRs may contribute to homeostasis but sometimes also to overt disease (e.g. airway hyperreactivity and asthma) at the alveolar-air interface.


Subject(s)
Asthma/immunology , Bronchial Hyperreactivity/immunology , C-Reactive Protein/immunology , Homeostasis/immunology , Receptors, Pattern Recognition/immunology , Respiratory Mucosa/immunology , Allergens/administration & dosage , Animals , Asthma/genetics , Asthma/pathology , Bronchial Hyperreactivity/genetics , Bronchial Hyperreactivity/pathology , C-Reactive Protein/genetics , Collectins/genetics , Collectins/immunology , Complement C3/genetics , Complement C3/immunology , Complement C5/genetics , Complement C5/immunology , Epithelial Cells/immunology , Epithelial Cells/pathology , Gene Expression Regulation , Homeostasis/genetics , Humans , Lectins/genetics , Lectins/immunology , Receptors, Pattern Recognition/genetics , Respiratory Mucosa/pathology , Serum Amyloid A Protein/genetics , Serum Amyloid A Protein/immunology , Serum Amyloid P-Component/genetics , Serum Amyloid P-Component/immunology
19.
Am J Med Genet A ; 185(7): 2267-2270, 2021 07.
Article in English | MEDLINE | ID: mdl-33765348

ABSTRACT

3MC syndrome is a rare condition manifesting with typical facial appearance, postnatal growth deficiency, skeletal manifestations, and genitourinary tract anomalies. 3MC is caused by biallelic pathogenic variants in MASP1, COLEC11, or COLEC10. Here, we report an affected subject of Kurdish origin from Turkey presenting with facial dysmorphisms, such as, hypertelorism, blepharophimosis, blepharoptosis, highly arched eyebrows, umbilical hernia, and caudal appendage. These features were compatible with 3MC syndrome. Molecular analysis revealed a novel homozygous pathogenic variant, c.310C > T; p.Gln104Ter in the MASP1 gene, resulting in a premature stop codon. Few subjects with 3MC syndrome have been reported in the literature so far. Thus, detailed study of this subject contributes to the evolving clinical and genetic characterization of 3MC syndrome.


Subject(s)
Abnormalities, Multiple/genetics , Collectins/genetics , Craniofacial Abnormalities/genetics , Mannose-Binding Protein-Associated Serine Proteases/genetics , Muscular Atrophy/genetics , Abnormalities, Multiple/pathology , Blepharophimosis/genetics , Blepharophimosis/pathology , Blepharoptosis/genetics , Blepharoptosis/pathology , Cleft Lip/genetics , Cleft Lip/pathology , Cleft Palate/genetics , Cleft Palate/pathology , Craniofacial Abnormalities/pathology , Craniosynostoses/genetics , Craniosynostoses/pathology , Eye Abnormalities/genetics , Eye Abnormalities/pathology , Humans , Hypertelorism/genetics , Hypertelorism/pathology , Infant , Male , Muscular Atrophy/pathology , Turkey/epidemiology
20.
Pediatr Nephrol ; 36(5): 1065-1073, 2021 05.
Article in English | MEDLINE | ID: mdl-32472330

ABSTRACT

Ischaemia/reperfusion injury (IRI) is an inevitable and damaging consequence of the process of kidney transplantation, ultimately leading to delayed graft function and increased risk of graft loss. A key driver of this adverse reaction in kidneys is activation of the complement system, an important part of the innate immune system. This activation causes deposition of complement C3 on renal tubules as well as infiltration of immune cells and ultimately damage to the tubules resulting in reduced kidney function. Collectin-11 (CL-11) is a pattern recognition molecule of the lectin pathway of complement. CL-11 binds to a ligand that is exposed on the renal tubules by the stress caused by IRI, and through attached proteases, CL-11 activates complement and this contributes to the consequences outlined above. Recent work in our lab has shown that this damage-associated ligand contains a fucose residue that aids CL-11 binding and promotes complement activation. In this review, we will discuss the clinical context of renal transplantation, the relevance of the complement system in IRI, and outline the evidence for the role of CL-11 binding to a fucosylated ligand in IRI as well as its downstream effects. Finally, we will detail the simple but elegant theory that increasing the level of free fucose in the kidney acts as a decoy molecule, greatly reducing the clinical consequences of IRI mediated by CL-11.


Subject(s)
Collectins/metabolism , Fucose/metabolism , Kidney Transplantation , Reperfusion Injury , Humans , Kidney , Kidney Transplantation/adverse effects , Ligands , Reperfusion Injury/etiology
SELECTION OF CITATIONS
SEARCH DETAIL