Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 98
1.
Int J Mol Sci ; 23(24)2022 Dec 16.
Article En | MEDLINE | ID: mdl-36555720

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.


Collectins , Sepsis , Humans , Animals , Swine , Pulmonary Surfactant-Associated Protein D/chemistry , Mannans/metabolism , Ligands , Lectins/metabolism
2.
Curr Protein Pept Sci ; 23(12): 862-873, 2022.
Article En | MEDLINE | ID: mdl-36330647

BACKGROUND: Surfactant protein-S (SP-D) is a naturally occurring lung protein with the potential to treat pulmonary infections. A recombinant surfactant protein-D (SP-D) has been produced and was previously found to exist in multiple oligomeric states. INTRODUCTION: Separation and characterization of interconverting oligomeric states of a protein can be difficult using chromatographic methods, so an alternative separation technique was employed for SPD to characterize the different association states that exist. METHODS: Samples of SP-D were analyzed using asymmetrical flow field-flow fractionation (AF4) using UV and multi-angle laser light scattering (MALLS) detection. The AF4 method appears to be able to separate species as small as the monomer up to the dodecamer (the dominant species) to much larger species with a molar mass greater than 5 MDa. RESULTS: Consistent elution of four distinct peaks was observed after repeated injections. The largest species observed under the last peak (labeled as Peak 4) were termed "unstructured multimers" and were resolved fairly well from the other species. The AF4-MALLS data suggest that only a small fraction of Peak 4 truly corresponds to high molar mass unstructured multimers. All other peaks demonstrated significant molar mass homogeneity consistent with AFM results. CONCLUSION: AF4-MALLS technology appears to be a powerful analytical approach to characterize the complex and dynamic interplay among different protein oligomeric species of SP-D in an aqueous solution.


Protein Multimerization , Pulmonary Surfactant-Associated Protein D , Fractionation, Field Flow/methods , Protein Multimerization/physiology , Pulmonary Surfactant-Associated Protein D/chemistry , Recombinant Proteins/chemistry
3.
Int J Mol Sci ; 22(19)2021 Sep 28.
Article En | MEDLINE | ID: mdl-34638783

Nanoparticles are efficient drug delivery vehicles for targeting specific organs as well as systemic therapy for a range of diseases, including cancer. However, their interaction with the immune system offers an intriguing challenge. Due to the unique physico-chemical properties, carbon nanotubes (CNTs) are considered as nanocarriers of considerable interest in cancer diagnosis and therapy. CNTs, as a promising nanomaterial, are capable of both detecting as well as delivering drugs or small therapeutic molecules to tumour cells. In this study, we coupled a recombinant fragment of human surfactant protein D (rfhSP-D) with carboxymethyl-cellulose (CMC) CNTs (CMC-CNT, 10-20 nm diameter) for augmenting their apoptotic and immunotherapeutic properties using two leukemic cell lines. The cell viability of AML14.3D10 or K562 cancer cell lines was reduced when cultured with CMC-mwCNT-coupled-rfhSP-D (CNT + rfhSP-D) at 24 h. Increased levels of caspase 3, 7 and cleaved caspase 9 in CNT + rfhSP-D treated AML14.3D10 and K562 cells suggested an involvement of an intrinsic pathway of apoptosis. CNT + rfhSP-D treated leukemic cells also showed higher mRNA expression of p53 and cell cycle inhibitors (p21 and p27). This suggested a likely reduction in cdc2-cyclin B1, causing G2/M cell cycle arrest and p53-dependent apoptosis in AML14.3D10 cells, while p53-independent mechanisms appeared to be in operation in K562 cells. We suggest that CNT + rfhSP-D has therapeutic potential in targeting leukemic cells, irrespective of their p53 status, and thus, it is worth setting up pre-clinical trials in animal models.


Apoptosis/drug effects , Immunotherapy , Leukemia, Myeloid, Acute/therapy , Nanotubes, Carbon/chemistry , Pulmonary Surfactant-Associated Protein D , Humans , K562 Cells , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Pulmonary Surfactant-Associated Protein D/chemistry , Pulmonary Surfactant-Associated Protein D/pharmacology , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacology
4.
Am J Respir Cell Mol Biol ; 65(1): 41-53, 2021 07.
Article En | MEDLINE | ID: mdl-33784482

Coronavirus disease (COVID-19) is an acute infectious disease caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Human SP-D (surfactant protein D) is known to interact with the spike protein of SARS-CoV, but its immune surveillance against SARS-CoV-2 is not known. The current study aimed to examine the potential of a recombinant fragment of human SP-D (rfhSP-D) as an inhibitor of replication and infection of SARS-CoV-2. The interaction of rfhSP-D with the spike protein of SARS-CoV-2 and human ACE-2 (angiotensin-converting enzyme 2) receptor was predicted via docking analysis. The inhibition of interaction between the spike protein and ACE-2 by rfhSP-D was confirmed using direct and indirect ELISA. The effect of rfhSP-D on replication and infectivity of SARS-CoV-2 from clinical samples was assessed by measuring the expression of RdRp gene of the virus using quantitative PCR. In silico interaction studies indicated that three amino acid residues in the receptor-binding domain of spike protein of SARS-CoV-2 were commonly involved in interacting with rfhSP-D and ACE-2. Studies using clinical samples of SARS-CoV-2-positive cases (asymptomatic, n = 7; symptomatic, n = 8) and negative control samples (n = 15) demonstrated that treatment with 1.67 µM rfhSP-D inhibited viral replication by ∼5.5-fold and was more efficient than remdesivir (100 µM) in Vero cells. An approximately two-fold reduction in viral infectivity was also observed after treatment with 1.67 µM rfhSP-D. These results conclusively demonstrate that the rfhSP-D mediated calcium independent interaction between the receptor-binding domain of the S1 subunit of the SARS-CoV-2 spike protein and human ACE-2, its host cell receptor, and significantly reduced SARS-CoV-2 infection and replication in vitro.


COVID-19/metabolism , Pulmonary Surfactant-Associated Protein D , SARS-CoV-2/physiology , Spike Glycoprotein, Coronavirus , Virus Replication , Adult , Animals , Chlorocebus aethiops , Female , Humans , Male , Protein Binding , Pulmonary Surfactant-Associated Protein D/chemistry , Pulmonary Surfactant-Associated Protein D/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/metabolism , Vero Cells
5.
Am J Physiol Lung Cell Mol Physiol ; 319(1): L148-L158, 2020 07 01.
Article En | MEDLINE | ID: mdl-32432921

Surfactant protein D (SP-D) is a C-type lectin that participates in the innate immune defense of lungs. It binds pathogens through its carbohydrate recognition domain in a calcium-dependent manner. Human surfactant protein D (hSP-D) has been routinely obtained from bronchoalveolar lavage of patients suffering from pulmonary alveolar proteinosis (PAP) and from amniotic fluid (AF). As a consequence of the disease, hSP-D obtained from PAP is found in higher amounts and is mainly composed of higher order oligomeric forms. However, PAP-hSP-D has never been directly compared with nonpathological human protein in terms of structure and biological activity. Moreover, the quantitative distribution of the different hSP-D oligomeric forms in human protein obtained from a natural source has never been evaluated. In this work, we have determined the quantitative distribution of AF-hSP-D oligomers, characterized the sugars attached through the N-glycosylation site of the protein, and compared the activity of hSP-D from AF and PAP with respect to their ability to bind and agglutinate bacteria. We have found that fuzzy balls (40%) are the most abundant oligomeric form in AF-hSP-D, very closely followed by dodecamers (33%), with both together constituting 73% of the protein mass. The glycan attached to the N-glycosylation site was found to be composed of fucose, galactose, sialic acid, and N-acetylglucosamine. Finally, in the functional assays performed, hSP-D obtained from PAP showed higher potency, probably as a consequence of its higher proportion of large oligomers compared with hSP-D from AF.


Pulmonary Surfactant-Associated Protein D/chemistry , Pulmonary Surfactant-Associated Protein D/metabolism , Amniotic Fluid/metabolism , Asparagine/metabolism , Binding, Competitive , Chromatography, Affinity , Female , Glycosylation , Humans , Polysaccharides/metabolism , Pregnancy , Protein Binding , Protein Multimerization , Pulmonary Alveolar Proteinosis/metabolism , Pulmonary Surfactant-Associated Protein D/isolation & purification , Structure-Activity Relationship
6.
Biochim Biophys Acta Proteins Proteom ; 1868(8): 140436, 2020 08.
Article En | MEDLINE | ID: mdl-32325256

Surfactant Protein D (SP-D) is a collectin protein that participates in the innate immune defense of the lungs. SP-D mediates the clearance of invading microorganisms by opsonization, aggregation or direct killing, which are lately removed by macrophages. SP-D is found as a mixture of trimers, hexamers, dodecamers and higher order oligomers, "fuzzy balls". However, it is unknown whether there are differences between these oligomeric forms in functions, activity or potency. In the present work, we have obtained fractions enriched in trimers, hexamers and fuzzy balls of full-length recombinant human (rh) SP-D by size exclusion chromatography, in a sufficient amount to perform functional assays. We have evaluated the differences in protein lectin-dependent activity relative to aggregation and binding to E. coli, one of the ligands of SP-D in vivo. Fuzzy balls are the most active oligomeric form in terms of binding and aggregation of bacteria, achieving 2-fold binding higher than hexamers and 50% bacteria aggregation at very short times. Hexamers, recently described as a defined oligomeric form of the protein, have never been isolated or tested in terms of protein activity. rhSP-D hexamers efficiently bind and aggregate bacteria, achieving 50-60% aggregation at final time point and high protein concentrations. Nevertheless, trimers are not able to aggregate bacteria, although they bind to them. Therefore, SP-D potency, in functions that relay on the C-lectin activity of the protein, is proportional to the oligomeric state of the protein.


Pulmonary Surfactant-Associated Protein D/chemistry , Pulmonary Surfactant-Associated Protein D/isolation & purification , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Animals , CHO Cells , Chromatography, Gel/methods , Chromatography, Ion Exchange/methods , Cricetulus , Escherichia coli/chemistry , Gene Expression , Humans , Protein Aggregates , Protein Binding , Protein Multimerization , Pulmonary Surfactant-Associated Protein D/biosynthesis , Recombinant Proteins/biosynthesis
7.
Front Immunol ; 10: 2476, 2019.
Article En | MEDLINE | ID: mdl-31749796

Innate immunity is critical in the early containment of influenza A virus (IAV) infection and surfactant protein D (SP-D) plays a crucial role in innate defense against IAV in the lungs. Multivalent lectin-mediated interactions of SP-D with IAVs result in viral aggregation, reduced epithelial infection, and enhanced IAV clearance by phagocytic cells. Previous studies showed that porcine SP-D (pSP-D) exhibits distinct antiviral activity against IAV as compared to human SP-D (hSP-D), mainly due to key residues in the lectin domain of pSP-D that contribute to its profound neutralizing activity. These observations provided the basis for the design of a full-length recombinant mutant form of hSP-D, designated as "improved SP-D" (iSP-D). Inspired by pSP-D, the lectin domain of iSP-D has 5 amino acids replaced (Asp324Asn, Asp330Asn, Val251Glu, Lys287Gln, Glu289Lys) and 3 amino acids inserted (326Gly-Ser-Ser). Characterization of iSP-D revealed no major differences in protein assembly and saccharide binding selectivity as compared to hSP-D. However, hemagglutination inhibition measurements showed that iSP-D expressed strongly enhanced activity compared to hSP-D against 31 different IAV strains tested, including (pandemic) IAVs that were resistant for neutralization by hSP-D. Furthermore, iSP-D showed increased viral aggregation and enhanced protection of MDCK cells against infection by IAV. Importantly, prophylactic or therapeutic application of iSP-D decreased weight loss and reduced viral lung titers in a murine model of IAV infection using a clinical isolate of H1N1pdm09 virus. These studies demonstrate the potential of iSP-D as a novel human-based antiviral inhalation drug that may provide immediate protection against or recovery from respiratory (pandemic) IAV infections in humans.


Carbohydrates , Protein Engineering , Protein Interaction Domains and Motifs , Pulmonary Surfactant-Associated Protein D/chemistry , Pulmonary Surfactant-Associated Protein D/metabolism , Binding Sites , Carbohydrates/immunology , Disease Resistance/genetics , Disease Resistance/immunology , Glycosylation , Humans , Influenza A virus/immunology , Influenza, Human/genetics , Influenza, Human/immunology , Influenza, Human/virology , Models, Molecular , Molecular Conformation , Protein Binding , Protein Interaction Domains and Motifs/genetics , Pulmonary Surfactant-Associated Protein D/genetics , Pulmonary Surfactant-Associated Protein D/immunology , Recombinant Proteins , Structure-Activity Relationship
8.
Nanotoxicology ; 13(10): 1396-1408, 2019 12.
Article En | MEDLINE | ID: mdl-31512956

Titanium dioxide (TiO2) nanoparticles are typical and widely used nanomaterials, and there are many studies on the inflammatory responses induced by their inhalation. In this study, we conducted a 4-week inhalation exposure study of aerosolized TiO2> nanoparticles (P25) to male Wistar rats. The mean aerosol concentration measured at each day was 4.1 mg/m3 by dry powder dispersion of TiO2 nanoparticles. Control and exposure groups of rats were killed at 3 and 30 days after the termination of exposure, and bronchoalveolar lavage fluid (BALF) and serum were collected for analysis of total cell count, neutrophil count, and surfactant protein (SP-D) in BALF and SP-D in serum, as well as other serum biomarkers. SP-D is a component of lung surfactants produced in type II alveolar epithelial cells and Clara cells and secreted into the alveolar space and blood. The neutrophil count in the BALF was significantly elevated at 3 and 30 days. The levels of SP-D in the BALF were also elevated at 3 and 30 days, while the serum SP-D levels were elevated at 3 days only. We determined the amounts of TiO2 in the rat lungs in the exposure group at 3, 30, and 73 days to analyze the lung deposition fraction (10.2%) and the biological half-life time (72.4 days) of inhaled TiO2 nanoparticles. Histopathological analysis revealed mild pulmonary inflammation in lung tissue at 3 days. Serum SP-D was found to be a potential biomarker for exposure to TiO2 nanoparticles in this study.


Inhalation Exposure , Metal Nanoparticles/toxicity , Pulmonary Surfactant-Associated Protein D/metabolism , Pulmonary Surfactants/metabolism , Titanium/toxicity , Administration, Inhalation , Aerosols/metabolism , Animals , Bronchoalveolar Lavage Fluid , Leukocyte Count , Lung/metabolism , Male , Metal Nanoparticles/chemistry , Neutrophils , Pneumonia/chemically induced , Proteins/metabolism , Pulmonary Surfactant-Associated Protein D/blood , Pulmonary Surfactant-Associated Protein D/chemistry , Pulmonary Surfactants/blood , Pulmonary Surfactants/chemistry , Rats , Rats, Wistar , Surface-Active Agents/metabolism , Titanium/administration & dosage , Titanium/chemistry
9.
J Innate Immun ; 11(1): 13-28, 2019.
Article En | MEDLINE | ID: mdl-30293076

Innate recognition of viruses is an essential part of the immune response to viral pathogens. This is integral to the maintenance of healthy lungs, which are free from infection and efficient at gaseous exchange. An important component of innate immunity for identifying viruses is the family of C-type collagen-containing lectins, also known as collectins. These secreted, soluble proteins are pattern recognition receptors (PRRs) which recognise pathogen-associated molecular patterns (PAMPs), including viral glycoproteins. These innate immune proteins are composed of trimerized units which oligomerise into higher-order structures and facilitate the clearance of viral pathogens through multiple mechanisms. Similarly, many viral surface proteins form trimeric configurations, despite not showing primary protein sequence similarities across the virus classes and families to which they belong. In this review, we discuss the role of the lung collectins, i.e., surfactant proteins A and D (SP-A and SP-D) in viral recognition. We focus particularly on the structural similarity and complementarity of these trimeric collectins with the trimeric viral fusion proteins with which, we hypothesise, they have elegantly co-evolved. Recombinant versions of these innate immune proteins may have therapeutic potential in a range of infectious and inflammatory lung diseases including anti-viral therapeutics.


Collectins , Immunity, Innate , Pulmonary Surfactant-Associated Protein A , Pulmonary Surfactant-Associated Protein D , Receptors, Pattern Recognition , Viral Fusion Proteins/immunology , Animals , Collectins/chemistry , Collectins/immunology , Humans , Lung/immunology , Lung Diseases/immunology , Lung Diseases/therapy , Lung Diseases/virology , Protein Multimerization , Pulmonary Surfactant-Associated Protein A/chemistry , Pulmonary Surfactant-Associated Protein A/immunology , Pulmonary Surfactant-Associated Protein D/chemistry , Pulmonary Surfactant-Associated Protein D/immunology , Receptors, Pattern Recognition/chemistry , Receptors, Pattern Recognition/immunology , Viral Fusion Proteins/chemistry , Viruses/immunology
10.
Mol Immunol ; 105: 260-269, 2019 01.
Article En | MEDLINE | ID: mdl-30562646

At the lung lining innate defenses protect our lungs against inhaled fungal cells that could pose a threat to our health. These defenses are comprised of mucociliary clearance, soluble effector molecules and roaming phagocytic cells, such as macrophages and neutrophils. How important each of these defenses is during fungal clearance depends on the specific fungal pathogen in question and on the stage of infection. In this study the localization and antifungal activity of the lung surfactant protein D (SP-D) was studied in an environment mimicking the lung lining. To this end Calu-3 cells were grown on an air-liquid interface allowing them to polarize and to produce mucus at their apical surface. Additionally, neutrophils were added to study their role in fungal clearance. Two fungal pathogens were used for these experiments: Candida albicans and Aspergillus fumigatus, both of clinical relevance. During fungal infection SP-D localized strongly to both fungal surfaces and stayed bound through the different stages of infection. Furthermore, SP-D decreased fungal adhesion to the epithelium and increased fungal clearance by neutrophils from the epithelial surface. These findings suggest that SP-D plays an important role at the different stages of pulmonary defense against fungal intruders.


Antifungal Agents/immunology , Aspergillosis/immunology , Aspergillus fumigatus/immunology , Candida albicans/immunology , Candidiasis/immunology , Lung/immunology , Pulmonary Surfactant-Associated Protein D/immunology , Respiratory Mucosa/immunology , Antifungal Agents/chemistry , Aspergillosis/pathology , Candidiasis/pathology , Cell Line , Humans , Lung/microbiology , Pulmonary Surfactant-Associated Protein D/chemistry , Respiratory Mucosa/microbiology
11.
Viruses ; 11(1)2018 12 26.
Article En | MEDLINE | ID: mdl-30587835

Since the largest 2014⁻2016 Ebola virus disease outbreak in West Africa, understanding of Ebola virus infection has improved, notably the involvement of innate immune mediators. Amongst them, collectins are important players in the antiviral innate immune defense. A screening of Ebola glycoprotein (GP)-collectins interactions revealed the specific interaction of human surfactant protein D (hSP-D), a lectin expressed in lung and liver, two compartments where Ebola was found in vivo. Further analyses have demonstrated an involvement of hSP-D in the enhancement of virus infection in several in vitro models. Similar effects were observed for porcine SP-D (pSP-D). In addition, both hSP-D and pSP-D interacted with Reston virus (RESTV) GP and enhanced pseudoviral infection in pulmonary cells. Thus, our study reveals a novel partner of Ebola GP that may participate to enhance viral spread.


Ebolavirus/chemistry , Glycoproteins/chemistry , Hemorrhagic Fever, Ebola/immunology , Pulmonary Surfactant-Associated Protein D/chemistry , Animals , Chlorocebus aethiops , Collectins/chemistry , Ebolavirus/drug effects , HEK293 Cells , Host Microbial Interactions , Humans , Immunity, Innate , Protein Binding , Pulmonary Surfactant-Associated Protein D/genetics , Swine , Vero Cells , Viral Proteins/chemistry
12.
Front Immunol ; 9: 1126, 2018.
Article En | MEDLINE | ID: mdl-29915574

Human surfactant protein D (SP-D) is a potent innate immune molecule, which is emerging as a key molecule in the recognition and clearance of altered and non-self targets. Previous studies have shown that a recombinant fragment of human SP-D (rfhSP-D) induced apoptosis via p53-mediated apoptosis pathway in an eosinophilic leukemic cell line, AML14.3D10. Here, we report the ability of rfhSP-D to induce apoptosis via TNF-α/Fas-mediated pathway regardless of the p53 status in human pancreatic adenocarcinoma using Panc-1 (p53mt), MiaPaCa-2 (p53mt), and Capan-2 (p53wt) cell lines. Treatment of these cell lines with rfhSP-D for 24 h caused growth arrest in G1 cell cycle phase and triggered transcriptional upregulation of pro-apoptotic factors such as TNF-α and NF-κB. Translocation of NF-κB from the cytoplasm into the nucleus of pancreatic cancer cell lines was observed via immunofluorescence microscopy following treatment with rfhSP-D as compared to the untreated cells. The rfhSP-D treatment caused upregulation of pro-apoptotic marker Fas, as analyzed via qPCR and western blot, which then triggered caspase cascade, as evident from cleavage of caspase 8 and 3 analyzed via western blot at 48 h. The cell number following the rfhSP-D treatment was reduced in the order of Panc-1 (~67%) > MiaPaCa-2 (~60%) > Capan-2 (~35%). This study appears to suggest that rfhSP-D can potentially be used to therapeutically target pancreatic cancer cells irrespective of their p53 phenotype.


Apoptosis/drug effects , Pancreatic Neoplasms/metabolism , Peptide Fragments/pharmacology , Pulmonary Surfactant-Associated Protein D/pharmacology , Signal Transduction/drug effects , fas Receptor/metabolism , Apoptosis/genetics , Caspase 3/metabolism , Caspase 8/metabolism , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Humans , NF-kappa B/metabolism , Protein Transport , Pulmonary Surfactant-Associated Protein D/chemistry , Recombinant Proteins , TOR Serine-Threonine Kinases/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , fas Receptor/genetics , Pancreatic Neoplasms
13.
PLoS One ; 13(6): e0199175, 2018.
Article En | MEDLINE | ID: mdl-29912941

The crystal structures of a biologically and therapeutically active recombinant homotrimeric fragment of native human SP-D (hSP-D) complexed with the inner core oligosaccharide of the Salmonella enterica sv Minnesota rough strains R5 and R7 (rough mutant chemotypes Rc and Rd1) have been determined. The structures reveal that hSP-D specifically and preferentially targets the LPS inner core via the innermost conserved Hep-Kdo pair with the flexibility for alternative recognition when this preferred epitope is not available for binding. Hep-Kdo binding is achieved through calcium dependent recognition of the heptose dihydroxyethyl side chain coupled with specific interactions between the Kdo and the binding site flanking residues Arg343 and Asp325 with evidence for an extended binding site for LPS inner cores containing multiple Kdo residues. In one subunit of the R5-bound structure this preferred mode of binding is precluded by the crystal lattice and oligosaccharide is bound through the terminal inner core glucose. The structures presented here thus provide unique multiple insights into the recognition and binding of bacterial LPS by hSP-D. Not only is it demonstrated that hSP-D targets the highly conserved LPS proximal inner core Hep-Kdo motif, but also that hSP-D can recognise either terminal or non-terminal sugars and has the flexibility and versatility to adopt alternative strategies for bacterial recognition, utilising alternative LPS epitopes when the preferred inner core Hep-Kdo disaccharide is not available for binding.


Lipopolysaccharides/metabolism , Pulmonary Surfactant-Associated Protein D/metabolism , Salmonella enterica/metabolism , Binding Sites , Epitopes/chemistry , Lipopolysaccharides/chemistry , Molecular Structure , Oligosaccharides/chemistry , Oligosaccharides/metabolism , Pulmonary Surfactant-Associated Protein D/chemistry , Pulmonary Surfactant-Associated Protein D/isolation & purification , Recombinant Proteins
14.
J Biol Chem ; 293(27): 10646-10662, 2018 07 06.
Article En | MEDLINE | ID: mdl-29769321

Innate immunity is critical in the early containment of influenza A virus (IAV) infection, and surfactant protein D (SP-D) plays a crucial role in the pulmonary defense against IAV. In pigs, which are important intermediate hosts during the generation of pandemic IAVs, SP-D uses its unique carbohydrate recognition domain (CRD) to interact with IAV. An N-linked CRD glycosylation provides interactions with the sialic acid-binding site of IAV, and a tripeptide loop at the lectin-binding site facilitates enhanced interactions with IAV glycans. Here, to investigate both mechanisms of IAV neutralization in greater detail, we produced an N-glycosylated neck-CRD fragment of porcine SP-D (RpNCRD) in HEK293 cells. X-ray crystallography disclosed that the N-glycan did not alter the CRD backbone structure, including the lectin site conformation, but revealed a potential second nonlectin-binding site for glycans. IAV hemagglutination inhibition, IAV aggregation, and neutralization of IAV infection studies showed that RpNCRD, unlike the human analogue RhNCRD, exhibits potent neutralizing activity against pandemic A/Aichi/68 (H3N2), enabled by both porcine-specific structural features of its CRD. MS analysis revealed an N-glycan site-occupancy of >98% at Asn-303 of RpNCRD with complex-type, heterogeneously branched and predominantly α(2,3)-sialylated oligosaccharides. Glycan-binding array data characterized both RpNCRD and RhNCRD as mannose-type lectins. RpNCRD also bound LewisY structures, whereas RhNCRD bound polylactosamine-containing glycans. The presence of the N-glycan in the CRD increases the glycan-binding specificity of RpNCRD. These insights increase our understanding of porcine-specific innate defense against pandemic IAV and may inform the design of recombinant SP-D-based antiviral drugs.


Immunity, Innate/immunology , Influenza A virus/immunology , Lectins/metabolism , Orthomyxoviridae Infections/prevention & control , Polysaccharides/metabolism , Pulmonary Surfactant-Associated Protein D/metabolism , Sialic Acids/metabolism , Amino Acid Sequence , Animals , Binding Sites , Carbohydrate Conformation , Glycosylation , Hemagglutination Inhibition Tests , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Polysaccharides/chemistry , Pulmonary Surfactant-Associated Protein D/chemistry , Pulmonary Surfactant-Associated Protein D/genetics , Sequence Homology , Swine
15.
J Mol Biol ; 430(10): 1495-1509, 2018 05 11.
Article En | MEDLINE | ID: mdl-29626540

Pulmonary surfactant protein D (SP-D) is a glycoprotein from the collectin family that is a component of the lung surfactant system. It exhibits host defense and immune regulatory functions in addition to contributing to the homeostasis of the surfactant pool within the alveolar airspaces. It is known that the SP-D monomer forms trimers, which further associate into higher-order oligomers. However, the pathway and the interactions involved in the assembly of SP-D oligomers are not clearly understood. In the current study, a recombinant form of full-length human SP-D (rhSP-D) has been qualitatively and quantitatively studied by atomic force microscopy (AFM) and electrophoresis, with the aim to understand the conformational diversity and the determinants defining the oligomerization of the protein. The rhSP-D preparation studied is a mixture of trimers, hexamers, dodecamers and higher-order oligomeric species, with dodecamers accounting for more than 50% of the protein by mass. Similar structures were also found in hSP-D obtained from proteinosis patients, with the largest fuzzy-ball-like oligomers being more abundant in these samples. The proportion of dodecamer is increased under acidic conditions, accompanied by a conformational change into more compact configurations. Two hexamers appear to be the minimal necessary unit for dodecamer formation, with stabilization of the dodecamer occurring via non-covalent, ionic, and hydrophobic interactions between the individual N-terminal domains and the proximal area of the SP-D collagen stems.


Pulmonary Alveolar Proteinosis/metabolism , Pulmonary Surfactant-Associated Protein D/chemistry , Pulmonary Surfactant-Associated Protein D/metabolism , Humans , Hydrogen-Ion Concentration , Microscopy, Atomic Force , Models, Molecular , Protein Multimerization , Thermodynamics
16.
Biochemistry ; 56(31): 4095-4105, 2017 08 08.
Article En | MEDLINE | ID: mdl-28719181

Lung surfactant proteins (SPs) play critical roles in surfactant function and innate immunity. SP-A and SP-D, members of the collectin family of C-type lectins, exhibit distinct ligand specificities, effects on surfactant structure, and host defense functions despite extensive structural homology. SP-A binds to dipalmitoylphosphatidylcholine (DPPC), the major surfactant lipid component, but not phosphatidylinositol (PI), whereas SP-D shows the opposite preference. Additionally, SP-A and SP-D recognize widely divergent pathogen-associated molecular patterns. Previous studies suggested that a ligand-induced surface loop conformational change unique to SP-A contributes to lipid binding affinity. To test this hypothesis and define the structural features of SP-A and SP-D that determine their ligand binding specificities, a structure-guided approach was used to introduce key features of SP-D into SP-A. A quadruple mutant (E171D/P175E/R197N/K203D) that introduced an SP-D-like loop-stabilizing calcium binding site into the carbohydrate recognition domain was found to interconvert SP-A ligand binding preferences to an SP-D phenotype, exchanging DPPC for PI specificity, and resulting in the loss of lipid A binding and the acquisition of more avid mannan binding properties. Mutants with constituent single or triple mutations showed alterations in their lipid and sugar binding properties that were intermediate between those of SP-A and SP-D. Structures of mutant complexes with inositol or methyl-mannose revealed an attenuation of the ligand-induced conformational change relative to wild-type SP-A. These studies suggest that flexibility in a key surface loop supports the distinctive lipid binding functions of SP-A, thus contributing to its multiple functions in surfactant structure and regulation, and host defense.


Models, Molecular , Pulmonary Surfactant-Associated Protein A/metabolism , Pulmonary Surfactant-Associated Protein D/metabolism , 1,2-Dipalmitoylphosphatidylcholine/chemistry , 1,2-Dipalmitoylphosphatidylcholine/metabolism , Amino Acid Substitution , Animals , Binding Sites , Crystallography, X-Ray , Kinetics , Lectins, C-Type/chemistry , Lectins, C-Type/metabolism , Ligands , Lipid A/chemistry , Lipid A/metabolism , Liposomes , Mutagenesis, Site-Directed , Mutation , Phosphatidylinositols/chemistry , Phosphatidylinositols/metabolism , Protein Conformation , Protein Interaction Domains and Motifs , Protein Refolding , Protein Stability , Pulmonary Surfactant-Associated Protein A/chemistry , Pulmonary Surfactant-Associated Protein A/genetics , Pulmonary Surfactant-Associated Protein D/chemistry , Pulmonary Surfactant-Associated Protein D/genetics , Rats , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism
17.
Ann Anat ; 211: 184-201, 2017 May.
Article En | MEDLINE | ID: mdl-28351530

The collectins family encompasses several collagenous Ca2+-dependent defense lectins that are described as pathogen recognition molecules. They play an important role in both adaptive and innate immunity. Surfactant proteins A and D are two of these proteins which were initially discovered in association with surfactant in the pulmonary system. The structure, immune and inflammatory functions, and genetic variations have been well described in relation to their roles, function and pathophysiology in the pulmonary system. Subsequently, these proteins have been discovered in a wide range of other organs and organ systems. The role of these proteins outside the pulmonary system is currently an active area of research. This review intends to provide a current overview of the genetics, structure and extra-pulmonary functions of the surfactant collectin proteins.


Immunity, Innate/genetics , Immunity, Innate/immunology , Pulmonary Surfactant-Associated Protein A/genetics , Pulmonary Surfactant-Associated Protein A/immunology , Pulmonary Surfactant-Associated Protein D/genetics , Pulmonary Surfactant-Associated Protein D/immunology , Animals , Humans , Polymorphism, Single Nucleotide/genetics , Pulmonary Surfactant-Associated Protein A/chemistry , Pulmonary Surfactant-Associated Protein D/chemistry , Structure-Activity Relationship
18.
Nanoscale ; 9(3): 1097-1109, 2017 Jan 19.
Article En | MEDLINE | ID: mdl-27991644

Carbon nanotubes (CNTs) are increasingly being developed for use in biomedical applications, including drug delivery. One of the most promising applications under evaluation is in treating pulmonary diseases such as tuberculosis. Once inhaled or administered, the nanoparticles are likely to be recognised by innate immune molecules in the lungs such as hydrophilic pulmonary surfactant proteins. Here, we set out to examine the interaction between surfactant protein D (SP-D), a key lung pattern recognition molecule and CNTs, and possible downstream effects on the immune response via macrophages. We show here that a recombinant form of human SP-D (rhSP-D) bound to oxidised and carboxymethyl cellulose (CMC) coated CNTs via its C-type lectin domain and enhanced phagocytosis by U937 and THP-1 macrophages/monocytic cell lines, together with an increased pro-inflammatory response, suggesting that sequestration of SP-D by CNTs in the lungs can trigger an unwanted and damaging immune response. We also observed that functionalised CNTs, opsonised with rhSP-D, continued to activate complement via the classical pathway, suggesting that C1q, which is the recognition sub-component of the classical pathway, and SP-D have distinct pattern recognition sites on the CNTs. Consistent with our earlier reports, complement deposition on the rhSP-D opsonised CNTs led to dampening of the pro-inflammatory immune response by THP-1 macrophages, as evident from qPCR, cytokine array and NF-κB nuclear translocation analyses. This study highlights the importance of understanding the interplay between innate immune humoral factors including complement in devising nanoparticle based drug delivery strategies.


Macrophages/cytology , Nanotubes, Carbon/chemistry , Phagocytosis , Pulmonary Surfactant-Associated Protein D/chemistry , Complement C1q/chemistry , Complement Pathway, Classical , Cytokines/metabolism , Humans , Recombinant Proteins/chemistry , THP-1 Cells , Transcription Factors/metabolism , Transcriptome , U937 Cells
19.
Alcohol Clin Exp Res ; 40(12): 2563-2572, 2016 12.
Article En | MEDLINE | ID: mdl-27783409

BACKGROUND: Reactive aldehydes such as acetaldehyde and malondialdehyde generated as a result of alcohol metabolism and cigarette smoke exposure lead to the formation of malondialdehyde-acetaldehyde-adducted proteins (MAA adducts). These aldehydes can adduct to different proteins such as bovine serum albumin and surfactant protein A or surfactant protein D (SPD). Macrophages play an important role in innate immunity, but the effect of MAA adducts on macrophage function has not yet been examined. Because macrophage scavenger receptor A (SRA; CD204) mediates the uptake of modified proteins, we hypothesized that the effects of MAA-modified proteins on macrophage function are primarily mediated through SRA. METHODS: We tested this hypothesis by exposing SPD-MAA to macrophages and measuring functions. SPD-MAA treatment significantly stimulated pro-inflammatory cytokine tumor necrosis factor-alpha (TNF-α) release in the macrophage cell line, RAW 264.7. RESULTS: A significant reduction in phagocytosis of zymosan particles was also observed. SPD-MAA stimulated a significant dose-dependent increase in TNF-α and interleukin (IL)-6 release from peritoneal macrophages (PMs) of wild-type (WT) mice. But significantly less TNF-α and IL-6 were released from PMs of SRA-/- mice. We observed a significant reduction in phagocytosis of zymosan particles in PMs from WT mice treated with SPD-MAA. No further SPD-MAA-induced reduction was seen in PMs from SRA-/- mice. SPD-MAA treatment significantly increased SRA mRNA expression, but had no effect on surface receptor protein expression. Protein kinase C alpha inhibitor and NF-κB inhibitor significantly reduced pro-inflammatory cytokine release in response to SPD-MAA. CONCLUSIONS: In conclusion, our data demonstrate that SRA is important for MAA-adducted protein-mediated effect on macrophage functions.


Acetaldehyde/chemistry , Macrophages/drug effects , Macrophages/metabolism , Malondialdehyde/chemistry , Pulmonary Surfactant-Associated Protein A/pharmacology , Pulmonary Surfactant-Associated Protein D/pharmacology , Scavenger Receptors, Class A/genetics , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Humans , Interleukin-6/metabolism , Mice , Mice, Knockout , Phagocytosis/drug effects , Pulmonary Surfactant-Associated Protein A/chemistry , Pulmonary Surfactant-Associated Protein D/chemistry , Rats , Scavenger Receptors, Class A/biosynthesis , Tumor Necrosis Factor-alpha/metabolism
20.
J Struct Biol ; 195(3): 387-395, 2016 09.
Article En | MEDLINE | ID: mdl-27350640

As a major player of the innate immune system, surfactant protein D (SP-D) recognizes and promotes elimination of various pathogens such as Gram-negative bacteria. SP-D binds to l-glycero-d-manno-heptose (Hep), a constituent of the partially conserved lipopolysaccharide (LPS) inner core of many Gram-negative bacteria. Binding and affinity of trimeric human SP-D to Hep in distinct LPS inner core glycans differing in linkages and adjacent residues was elucidated using glycan array and surface plasmon resonance measurements that were compared to in silico interaction studies. The combination of in vitro assays using defined glycans and molecular docking and dynamic simulation approaches provides insights into the interaction of trimeric SP-D with those glycan ligands. Trimeric SP-D wildtype recognized larger LPS inner core oligosaccharides with slightly enhanced affinity than smaller compounds suggesting the involvement of stabilizing secondary interactions. A trimeric human SP-D mutant D324N+D325N+R343K resembling rat SP-D bound to various LPS inner core structures in a similar pattern as observed for the wildtype but with higher affinity. The selective mutation of SP-D promotes targeting of LPS inner core oligosaccharides on Gram-negative bacteria to develop novel therapeutic agents.


Lipopolysaccharides/chemistry , Pulmonary Surfactant-Associated Protein D/chemistry , Amino Acid Substitution , Crystallography, X-Ray , Humans , Kinetics , Molecular Docking Simulation , Protein Binding
...