Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 90
Filter
Add more filters

Publication year range
1.
Am J Pathol ; 193(6): 680-689, 2023 06.
Article in English | MEDLINE | ID: mdl-36868468

ABSTRACT

Respiratory viruses, including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), can trigger chronic lung disease that persists and even progresses after expected clearance of infectious virus. To gain an understanding of this process, the current study examined a series of consecutive fatal cases of coronavirus disease 2019 (COVID-19) that came to autopsy at 27 to 51 days after hospital admission. In each patient, a stereotyped bronchiolar-alveolar pattern of lung remodeling was identified with basal epithelial cell hyperplasia, immune activation, and mucinous differentiation. Remodeling regions featured macrophage infiltration and apoptosis and a marked depletion of alveolar type 1 and 2 epithelial cells. This pattern closely resembled findings from an experimental model of post-viral lung disease that requires basal-epithelial stem cell growth, immune activation, and differentiation. Together, these results provide evidence of basal epithelial cell reprogramming in long-term COVID-19 and thereby yield a pathway for explaining and correcting lung dysfunction in this type of disease.


Subject(s)
COVID-19 , Humans , Cellular Reprogramming , SARS-CoV-2 , Lung , Epithelial Cells
2.
Breast Cancer Res Treat ; 165(2): 355-364, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28612227

ABSTRACT

PURPOSE: The recent publication of the ACOSOG Z1031 trial results demonstrated that Ki-67 proliferation marker-based neoadjuvant endocrine therapy response monitoring could be used for tailoring the use of adjuvant chemotherapy in ER+HER2-negative breast cancer patients. In this paper, we describe the development of the Ki-67 clinical trial assay used for this study. METHODS: Ki-67 assay assessment focused on reproducing a 2.7% Ki-67 cut-point (CP) required for calculating the Preoperative Endocrine Prognostic Index and a 10% CP for poor endocrine therapy response identification within the first month of neoadjuvant endocrine treatment. Image analysis was assessed to increase the efficiency of the scoring process. Clinical outcome concordance for two independent Ki-67 scores was the primary performance metric. RESULTS: Discordant scores led to a triage approach where cases with complex histological features that software algorithms could not resolve were flagged for visual point counting (17%). The final Ki-67 scoring approach was run on T1/2 N0 cases from the P024 and POL trials (N = 58). The percent positive agreement for the 2.7% CP was 87.5% (95% CI 61.7-98.5%); percent negative agreement 88.9% (95% CI: 65.3-98.6%). Minor discordance did not affect the ability to predict similar relapse-free outcomes (Log-Rank P = 0.044 and P = 0.055). The data for the 10% early triage CP in the POL trial were similar (N = 66), the percentage positive agreement was 100%, and percent negative agreement 93.55% (95% CI: 78.58-99.21%). The independent survival predictions were concordant (Log-rank P = 0.0001 and P = 0.01). CONCLUSIONS: We have developed an efficient and reproducible Ki-67 scoring system that was approved by the Clinical Trials Evaluation Program for NCI-supported neoadjuvant endocrine therapy trials. Using the methodology described here, investigators are able to identify a subgroup of patients with ER+HER2-negative breast cancer that can be safely managed without the need of adjuvant chemotherapy.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Ki-67 Antigen/metabolism , Antineoplastic Agents, Hormonal/administration & dosage , Antineoplastic Agents, Hormonal/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers , Breast Neoplasms/diagnosis , Breast Neoplasms/mortality , Chemotherapy, Adjuvant , Clinical Decision-Making , Female , Humans , Kaplan-Meier Estimate , Neoadjuvant Therapy , Prognosis , ROC Curve , Reproducibility of Results , Treatment Outcome
3.
J Immunol ; 194(7): 3317-26, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25716998

ABSTRACT

Surfactant protein D (SP-D) is critical for maintenance of lung homeostasis and provides a first line of defense to pathogens at mucosal surfaces. Polymorphisms in the SP-D-encoding gene SFTPD have been associated with chronic obstructive pulmonary disease and ulcerative colitis. Identification of the immunoreceptors that bind SP-D is essential for understanding its contribution to lung homeostasis and mucosal defense. We located a putative binding motif for the osteoclast-associated receptor (OSCAR) within the SP-D collagenous domain. An OSCAR-Fc fusion protein specifically bound to the collagenous region of recombinant SP-D and captured native SP-D from human bronchoalveolar lavage. OSCAR localized in an intracellular compartment of alveolar macrophages together with SP-D. Moreover, we found OSCAR on the surface of interstitial lung and blood CCR2(+) inflammatory monocytes, which secreted TNF-α when exposed to SP-D in an OSCAR-dependent fashion. OSCAR and SP-D did not exclusively colocalize in lung, as they were also highly expressed in atherosclerotic plaques of human aorta, supporting a role for this interaction in atherosclerosis. Our results identify the OSCAR:SP-D interaction as a potential therapeutic target in chronic inflammatory diseases of the lung as well as other diseases involving tissue accumulation of SP-D, infiltration of inflammatory monocytes, and release of TNF-α.


Subject(s)
Inflammation/metabolism , Monocytes/metabolism , Pulmonary Surfactant-Associated Protein D/metabolism , Receptors, CCR2/metabolism , Receptors, Cell Surface/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Amino Acid Sequence , Carrier Proteins/metabolism , Cell Line , Cell Membrane/metabolism , Computational Biology , Foam Cells/immunology , Foam Cells/metabolism , Foam Cells/pathology , Gene Expression , Humans , Inflammation/immunology , Inflammation/pathology , Intracellular Space/metabolism , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Monocytes/immunology , Myeloid Cells/immunology , Myeloid Cells/metabolism , Plaque, Atherosclerotic/metabolism , Plaque, Atherosclerotic/pathology , Protein Binding , Protein Interaction Domains and Motifs , Protein Interaction Mapping , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
4.
J Struct Biol ; 195(3): 387-395, 2016 09.
Article in English | MEDLINE | ID: mdl-27350640

ABSTRACT

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.


Subject(s)
Lipopolysaccharides/chemistry , Pulmonary Surfactant-Associated Protein D/chemistry , Amino Acid Substitution , Crystallography, X-Ray , Humans , Kinetics , Molecular Docking Simulation , Protein Binding
5.
Am J Physiol Lung Cell Mol Physiol ; 306(11): L1036-44, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24705721

ABSTRACT

We recently reported that a trimeric neck and carbohydrate recognition domain (NCRD) fragment of human surfactant protein D (SP-D), a host defense lectin, with combinatorial substitutions at the 325 and 343 positions (D325A+R343V) exhibits markedly increased antiviral activity for seasonal strains of influenza A virus (IAV). The NCRD binds to glycan-rich viral envelope proteins including hemagglutinin (HA). We now show that replacement of D325 with serine to create D325S+R343V provided equal or increased neutralizing activity compared with D325A+R343V. The activity of the double mutants was significantly greater than that of either single mutant (D325A/S or R343V). D325A+R343V and D325S+R343V also strongly inhibited HA activity, and markedly aggregated, the 1968 pandemic H3N2 strain, Aichi68. D325S+R343V significantly reduced viral loads and mortality of mice infected with Aichi68, whereas wild-type SP-D NCRD did not. The pandemic H1N1 strains of 1918 and 2009 have only one N-linked glycan side on the head region of the HA and are fully resistant to inhibition by native SP-D. Importantly, we now show that D325A+R343V and D325S+R343V inhibited Cal09 H1N1 and related strains, and reduced uptake of Cal09 by epithelial cells. Inhibition of Cal09 was mediated by the lectin activity of the NCRDs. All known human pandemic strains have at least one glycan attachment on the top or side of the HA head, and our results indicate that they may be susceptible to inhibition by modified host defense lectins.


Subject(s)
Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/immunology , Influenza, Human/virology , Pulmonary Surfactant-Associated Protein D/genetics , Animals , Binding Sites , CHO Cells , Cricetinae , Cricetulus , Disease Resistance , Dogs , Female , Host-Pathogen Interactions , Humans , Influenza, Human/immunology , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred DBA , Mutation, Missense , Pandemics , Protein Binding , Pulmonary Surfactant-Associated Protein D/chemistry , Pulmonary Surfactant-Associated Protein D/metabolism , Viral Load
6.
J Immunol ; 188(6): 2759-68, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22345650

ABSTRACT

θ-Defensins are cyclic octadecapeptides found in nonhuman primates whose broad antiviral spectrum includes HIV-1, HSV-1, severe acute respiratory syndrome coronavirus, and influenza A virus (IAV). We previously reported that synthetic θ-defensins called retrocyclins can neutralize and aggregate various strains of IAV and increase IAV uptake by neutrophils. This study describes two families of peptides, hapivirins and diprovirins, whose design was inspired by retrocyclins. The goal was to develop smaller partially cyclic peptides that retain the antiviral activity of retrocyclins, while being easier to synthesize. The novel peptides also allowed for systemic substitution of key residues to evaluate the role of charge or hydrophobicity on antiviral activity. Seventy-two hapivirin or diprovirin peptides are described in this work, including several whose anti-IAV activity equals or exceeds that of normal α- or θ-defensins. Some of these also had strong antibacterial and antifungal activity. These new peptides were active against H3N2 and H1N1 strains of IAV. Structural features imparting strong antiviral activity were identified through iterative cycles of synthesis and testing. Our findings show the importance of hydrophobic residues for antiviral activity and show that pegylation, which often increases a peptide's serum t(1/2) in vivo, can increase the antiviral activity of DpVs. The new peptides acted at an early phase of viral infection, and, when combined with pulmonary surfactant protein D, their antiviral effects were additive. The peptides strongly increased neutrophil and macrophage uptake of IAV, while inhibiting monocyte cytokine generation. Development of modified θ-defensin analogs provides an approach for creating novel antiviral agents for IAV infections.


Subject(s)
Antiviral Agents/chemical synthesis , Antiviral Agents/pharmacology , Defensins/immunology , Defensins/pharmacology , Influenza A virus/immunology , Amino Acid Sequence , Animals , Antiviral Agents/immunology , Cell Line , Chemistry Techniques, Synthetic , Chromatography, High Pressure Liquid , Defensins/chemical synthesis , Dogs , Humans , Microscopy, Electron, Transmission , Molecular Sequence Data , Monocytes/virology , Neutrophils/virology , Peptides , Structure-Activity Relationship , Tumor Necrosis Factor-alpha/biosynthesis
7.
J Clin Invest ; 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39052353

ABSTRACT

Epithelial barriers are programmed for defense and repair but are also the site of long-term structural remodeling and disease. In general, this paradigm features epithelial stem cell (ESCs) that are called on to regenerate damaged tissues but can also be reprogrammed for detrimental remodeling. Here we identified a Wfdc21-dependent monocyte-derived dendritic cell (moDC) population that functioned as an early sentinel niche for basal-ESC reprogramming in mouse models of epithelial injury after respiratory viral infection. Niche function depended on moDC delivery of ligand GPNMB to basal-ESC receptor CD44 so that properly timed antibody blockade of ligand or receptor provided long-lasting correction of reprogramming and broad disease phenotypes. These same control points worked directly in mouse and human basal-ESC organoids. Together, the findings identify a mechanism to explain and modify what is otherwise a stereotyped but sometimes detrimental response to epithelial injury.

8.
bioRxiv ; 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38895360

ABSTRACT

All living organisms are charged with repair after injury particularly at epithelial barrier sites, but in some cases this response leads instead to structural remodeling and long-term disease. Identifying the molecular and cellular control of this divergence is key to disease modification. In that regard, stress kinase control of epithelial stem cells is a rational entry point for study. Here we examine the potential for mitogen-activated protein kinase 13 (MAPK13) regulation of epithelial stem cells using models of respiratory viral injury and post-viral lung disease. We show that Mapk13 gene-knockout mice handle acute infectious illness as expected but are protected against structural remodeling manifest as basal-epithelial stem cell (basal-ESC) hyperplasia-metaplasia, immune activation, and mucinous differentiation. In corresponding cell models, Mapk13-deficiency directly attenuates basal-ESC growth and organoid formation. Extension to human studies shows marked induction/activation of basal-cell MAPK13 in clinical samples of comparable remodeling found in asthma and COPD. Here again, MAPK13 gene-knockdown inhibits human basal-ESC growth in culture. Together, the data identify MAPK13 as a control for structural remodeling and disease after epithelial injury and as a suitable target for down-regulation as a disease-modifying strategy.

9.
JAMA Oncol ; 10(3): 362-371, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38236590

ABSTRACT

Importance: Adding fulvestrant to anastrozole (A+F) improved survival in postmenopausal women with advanced estrogen receptor (ER)-positive/ERBB2 (formerly HER2)-negative breast cancer. However, the combination has not been tested in early-stage disease. Objective: To determine whether neoadjuvant fulvestrant or A+F increases the rate of pathologic complete response or ypT1-2N0/N1mic/Ki67 2.7% or less residual disease (referred to as endocrine-sensitive disease) over anastrozole alone. Design, Setting, and Participants: A phase 3 randomized clinical trial assessing differences in clinical and correlative outcomes between each of the fulvestrant-containing arms and the anastrozole arm. Postmenopausal women with clinical stage II to III, ER-rich (Allred score 6-8 or >66%)/ERBB2-negative breast cancer were included. All analyses were based on data frozen on March 2, 2023. Interventions: Patients received anastrozole, fulvestrant, or a combination for 6 months preoperatively. Tumor Ki67 was assessed at week 4 and optionally at week 12, and if greater than 10% at either time point, the patient switched to neoadjuvant chemotherapy or immediate surgery. Main Outcomes and Measures: The primary outcome was the endocrine-sensitive disease rate (ESDR). A secondary outcome was the percentage change in Ki67 after 4 weeks of neoadjuvant endocrine therapy (NET) (week 4 Ki67 suppression). Results: Between February 2014 and November 2018, 1362 female patients (mean [SD] age, 65.0 [8.2] years) were enrolled. Among the 1298 evaluable patients, ESDRs were 18.7% (95% CI, 15.1%-22.7%), 22.8% (95% CI, 18.9%-27.1%), and 20.5% (95% CI, 16.8%-24.6%) with anastrozole, fulvestrant, and A+F, respectively. Compared to anastrozole, neither fulvestrant-containing regimen significantly improved ESDR or week 4 Ki67 suppression. The rate of week 4 or week 12 Ki67 greater than 10% was 25.1%, 24.2%, and 15.7% with anastrozole, fulvestrant, and A+F, respectively. Pathologic complete response/residual cancer burden class I occurred in 8 of 167 patients and 17 of 167 patients, respectively (15.0%; 95% CI, 9.9%-21.3%), after switching to neoadjuvant chemotherapy due to week 4 or week 12 Ki67 greater than 10%. PAM50 subtyping derived from RNA sequencing of baseline biopsies available for 753 patients (58%) identified 394 luminal A, 304 luminal B, and 55 nonluminal tumors. A+F led to a greater week 4 Ki67 suppression than anastrozole alone in luminal B tumors (median [IQR], -90.4% [-95.2 to -81.9%] vs -76.7% [-89.0 to -55.6%]; P < .001), but not luminal A tumors. Thirty-six nonluminal tumors (65.5%) had a week 4 or week 12 Ki67 greater than 10%. Conclusions and Relevance: In this randomized clinical trial, neither fulvestrant nor A+F significantly improved the 6-month ESDR over anastrozole in ER-rich/ERBB2-negative breast cancer. Aromatase inhibition remains the standard-of-care NET. Differential NET response by PAM50 subtype in exploratory analyses warrants further investigation. Trial Registration: ClinicalTrials.gov Identifier: NCT01953588.


Subject(s)
Breast Neoplasms , Triple Negative Breast Neoplasms , Aged , Female , Humans , Anastrozole/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Breast Neoplasms/pathology , Fulvestrant , Ki-67 Antigen , Neoadjuvant Therapy , Nitriles/adverse effects , Postmenopause , Receptor, ErbB-2 , Receptors, Estrogen , Triazoles/adverse effects , Triple Negative Breast Neoplasms/drug therapy , Middle Aged
10.
Biochemistry ; 52(47): 8527-38, 2013 Nov 26.
Article in English | MEDLINE | ID: mdl-24224757

ABSTRACT

Surfactant protein D (SP-D), a mammalian C-type lectin, is the primary innate inhibitor of influenza A virus (IAV) in the lung. Interactions of SP-D with highly branched viral N-linked glycans on hemagglutinin (HA), an abundant IAV envelope protein and critical virulence factor, promote viral aggregation and neutralization through as yet unknown molecular mechanisms. Two truncated human SP-D forms, wild-type (WT) and double mutant D325A+R343V, representing neck and carbohydrate recognition domains are compared in this study. Whereas both WT and D325A+R343V bind to isolated glycosylated HA, WT does not inhibit IAV in neutralization assays; in contrast, D325A+R343V neutralization compares well with that of full-length native SP-D. To elucidate the mechanism for these biochemical observations, we have determined crystal structures of D325A+R343V in the presence and absence of a viral nonamannoside (Man9). On the basis of the D325A+R343V-Man9 structure and other crystallographic data, models of complexes between HA and WT or D325A+R343V were produced and subjected to molecular dynamics. Simulations reveal that whereas WT and D325A+R343V both block the sialic acid receptor site of HA, the D325A+R343V complex is more stable, with stronger binding caused by additional hydrogen bonds and hydrophobic interactions with HA residues. Furthermore, the blocking mechanism of HA differs for WT and D325A+R343V because of alternate glycan binding modes. The combined results suggest a mechanism through which the mode of SP-D-HA interaction could significantly influence viral aggregation and neutralization. These studies provide the first atomic-level molecular view of an innate host defense lectin inhibiting its viral glycoprotein target.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Models, Molecular , Pulmonary Surfactant-Associated Protein D/chemistry , Adhesiveness , Amino Acid Substitution , Binding Sites , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Humans , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Influenza A Virus, H3N2 Subtype/chemistry , Influenza A Virus, H3N2 Subtype/growth & development , Influenza A Virus, H3N2 Subtype/metabolism , Microbial Viability , Molecular Dynamics Simulation , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Polysaccharides/chemistry , Polysaccharides/metabolism , Protein Conformation , Pulmonary Surfactant-Associated Protein D/genetics , Pulmonary Surfactant-Associated Protein D/metabolism , Receptors, Cell Surface/antagonists & inhibitors , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
11.
J Biol Chem ; 287(32): 26666-77, 2012 Aug 03.
Article in English | MEDLINE | ID: mdl-22685299

ABSTRACT

Pigs can act as intermediate hosts by which reassorted influenza A virus (IAV) strains can be transmitted to humans and cause pandemic influenza outbreaks. The innate host defense component surfactant protein D (SP-D) interacts with glycans on the hemagglutinin of IAV and contributes to protection against IAV infection in mammals. This study shows that a recombinant trimeric neck lectin fragment derived from porcine SP-D (pSP-D) exhibits profound inhibitory activity against IAV, in contrast to comparable fragments derived from human SP-D. Crystallographic analysis of the pSP-D fragment complexed with a viral sugar component shows that a unique tripeptide loop alters the lectin site conformation of pSP-D. Molecular dynamics simulations highlight the role of this flexible loop, which adopts a more stable conformation upon sugar binding and may facilitate binding to viral glycans through contact with distal portions of the branched mannoside. The combined data demonstrate that porcine-specific structural features of SP-D contribute significantly to its distinct anti-IAV activity. These findings could help explain why pigs serve as important reservoirs for newly emerging pathogenic IAV strains.


Subject(s)
Antiviral Agents/pharmacology , Carbohydrate Metabolism , Influenza A virus/drug effects , Pulmonary Surfactant-Associated Protein D/pharmacology , Animals , Antiviral Agents/chemistry , Base Sequence , Binding Sites , Cells, Cultured , Crystallization , DNA Primers , Dogs , Microbial Sensitivity Tests , Models, Molecular , Molecular Dynamics Simulation , Mutagenesis, Site-Directed , Pulmonary Surfactant-Associated Protein D/chemistry , Pulmonary Surfactant-Associated Protein D/genetics , Swine
12.
J Gen Virol ; 94(Pt 1): 40-49, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23052388

ABSTRACT

LL-37, the only human cathelicidin, is a cationic antimicrobial peptide with antibacterial and antifungal activity. LL-37 is released from neutrophil granules and produced by epithelial cells. It has been implicated in host defence against influenza A virus (IAV) in recent studies. We now demonstrate dose-related neutralizing activity of LL-37 against several seasonal and mouse-adapted IAV strains. The ability of LL-37 to inhibit these IAV strains resulted mainly from direct effects on the virus, since pre-incubation of virus with LL-37 was needed for optimal inhibition. LL-37 bound high-density lipoprotein (HDL), and pre-incubation of LL-37 with human serum or HDL reduced its antiviral activity. LL-37 did not inhibit viral association with epithelial cells as assessed by quantitative RT-PCR or confocal microscopy. This finding contrasted with results obtained with surfactant protein D (SP-D). Unlike collectins or human neutrophil defensins (HNPs), LL-37 did not induce viral aggregation under electron microscopy. In the electron microscopy studies, LL-37 appeared to cause disruption of viral membranes. LL-37 had additive antiviral activity when combined with other innate inhibitors like SP-D, surfactant protein A and HNPs. Unlike HNPs, LL-37 did not bind SP-D significantly. These findings indicate that LL-37 contributes to host defence against IAV through a mechanism distinct from that of SP-D and HNPs.


Subject(s)
Antimicrobial Cationic Peptides/pharmacology , Defensins/metabolism , Influenza A Virus, H3N2 Subtype/drug effects , Influenza A Virus, H3N2 Subtype/metabolism , Influenza A virus/metabolism , Pulmonary Surfactant-Associated Protein D/metabolism , Animals , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/metabolism , Antimicrobial Cationic Peptides/immunology , CHO Cells , Collectins/immunology , Collectins/metabolism , Cricetinae , Cricetulus , Defensins/immunology , Dogs , Epithelial Cells/immunology , Epithelial Cells/metabolism , Epithelial Cells/virology , Humans , Influenza A Virus, H3N2 Subtype/immunology , Influenza A virus/immunology , Lipoproteins, HDL/immunology , Lipoproteins, HDL/metabolism , Madin Darby Canine Kidney Cells , Mice , Neutrophils/immunology , Neutrophils/metabolism , Neutrophils/virology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/virology , Pulmonary Surfactant-Associated Protein A/immunology , Pulmonary Surfactant-Associated Protein A/metabolism , Pulmonary Surfactant-Associated Protein D/immunology , Cathelicidins
13.
Am J Respir Cell Mol Biol ; 47(6): 792-9, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23024061

ABSTRACT

Previously, we described the protective role of the neutrophil serine protease inhibitor serpinB1 in preventing early mortality of Pseudomonas aeruginosa lung infection by fostering bacterial clearance and limiting inflammatory cytokines and proteolytic damage. Surfactant protein D (SP-D), which maintains the antiinflammatory pulmonary environment and mediates bacterial removal, was degraded in infected serpinB1-deficient mice. Based on the hypothesis that increased SP-D would rescue or mitigate the pathological effects of serpinB1 deletion, we generated two serpinB1(-/-) lines overexpressing lung-specific rat SP-D and inoculated the mice with P. aeruginosa. Contrary to predictions, bacterial counts in the lungs of SP-D(low)serpinB1(-/-) and SP-D(high) serpinB1(-/-) mice were 4 logs higher than wild-type and not different from serpinB1(-/-) mice. SP-D overexpression also failed to mitigate inflammation (TNF-α), lung injury (free protein, albumin), or excess neutrophil death (free myeloperoxidase, elastase). These pathological markers were higher for infected SP-D(high)serpinB1(-/-) mice than for serpinB1(-/-) mice, although the differences were not significant after controlling for multiple comparisons. The failure of transgenic SP-D to rescue antibacterial defense of serpinB1-deficient mice occurred despite 5-fold or 20-fold increased expression levels, largely normal structure, and dose-dependent bacteria-aggregating activity. SP-D of infected wild-type mice was intact in 43-kD monomers by reducing SDS-PAGE. By contrast, proteolytic fragments of 35, 17, and 8 kD were found in infected SP-D(low)serpinB1(-/-), SP-D(high) serpinB1(-/-) mice, and serpinB1(-/-) mice. Thus, although therapies to increase lung concentration of SP-D may have beneficial applications, the findings suggest that therapy with SP-D may not be beneficial for lung inflammation or infection if the underlying clinical condition includes excess proteolysis.


Subject(s)
Pulmonary Surfactant-Associated Protein D/metabolism , Serpins/genetics , Animals , Bronchoalveolar Lavage Fluid , Cathepsin G/metabolism , Female , Lung Injury/immunology , Lung Injury/metabolism , Lung Injury/microbiology , Mice , Mice, 129 Strain , Mice, Knockout , Myeloblastin/metabolism , Neutrophils/enzymology , Pancreatic Elastase/metabolism , Pneumonia, Bacterial/immunology , Pneumonia, Bacterial/metabolism , Pneumonia, Bacterial/microbiology , Pseudomonas Infections/immunology , Pseudomonas Infections/metabolism , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/immunology , Pulmonary Surfactant-Associated Protein D/genetics , Serpins/deficiency
14.
J Biol Chem ; 286(47): 40681-92, 2011 Nov 25.
Article in English | MEDLINE | ID: mdl-21965658

ABSTRACT

The recognition of influenza A virus (IAV) by surfactant protein D (SP-D) is mediated by interactions between the SP-D carbohydrate recognition domains (CRD) and glycans displayed on envelope glycoproteins. Although native human SP-D shows potent antiviral and aggregating activity, trimeric recombinant neck+CRDs (NCRDs) show little or no capacity to influence IAV infection. A mutant trimeric NCRD, D325A/R343V, showed marked hemagglutination inhibition and viral neutralization, with viral aggregation and aggregation-dependent viral uptake by neutrophils. D325A/R343V exhibited glucose-sensitive binding to Phil82 hemagglutinin trimer (HA) by surface plasmon resonance. By contrast, there was very low binding to the HA trimer from another virus (PR8) that lacks glycans on the HA head. Mass spectrometry demonstrated the presence of high mannose glycans on the Phil82 HA at positions known to contribute to IAV binding. Molecular modeling predicted an enhanced capacity for bridging interactions between HA glycans and D325A/R343V. Finally, the trimeric D325A/R343V NCRD decreased morbidity and increased viral clearance in a murine model of IAV infection using a reassortant A/WSN/33 virus with a more heavily glycosylated HA. The combined data support a model in which altered binding by a truncated mutant SP-D to IAV HA glycans facilitates viral aggregation, leading to significant viral neutralization in vitro and in vivo. These studies demonstrate the potential utility of homology modeling and protein structure analysis for engineering effective collectin antivirals as in vivo therapeutics.


Subject(s)
Disease Resistance/genetics , Evolution, Molecular , Influenza A Virus, H1N1 Subtype/physiology , Pulmonary Surfactant-Associated Protein D/chemistry , Pulmonary Surfactant-Associated Protein D/genetics , Surface Plasmon Resonance/methods , Animals , Antiviral Agents/metabolism , Antiviral Agents/pharmacology , Crystallography, X-Ray , Female , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Humans , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H1N1 Subtype/metabolism , Mass Spectrometry , Mice , Models, Molecular , Mutagenesis, Site-Directed , Orthomyxoviridae Infections/virology , Protein Multimerization , Protein Structure, Quaternary , Protein Structure, Tertiary , Pulmonary Surfactant-Associated Protein D/metabolism , Species Specificity
15.
J Immunol ; 184(11): 6343-9, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20435925

ABSTRACT

Pulmonary surfactant protein D (SP-D), a member of the collectin family, is an innate immune molecule critical for defense that can also modulate adaptive immune responses. We previously showed that SP-D-deficient mice exhibit enhanced allergic responses and that SP-D induction requires lymphocytes. Thus, we postulated that SP-D may decrease adaptive allergic responses through interaction with T cells. In this study, we used two forms of SP-D, a dodecamer and a shorter fragment containing the trimeric neck and carbohydrate recognition domains (SP-D NCRD). Both forms decreased immune responses in vitro and in a murine model of pulmonary inflammation. SP-D NCRD increased transcription of CTLA4, a negative regulator of T cell activation, in T cells. SP-D NCRD no longer decreased lymphoproliferation and IL-2 cytokine production when CTLA4 signals were abrogated. Administration of SP-D NCRD in vivo no longer decreased allergen induced responses when CTLA4 was inhibited. Our results indicate that SP-D decreases allergen responses, an effect that may be mediated by increase of CTLA4 in T cells.


Subject(s)
Antigens, CD/immunology , Inflammation/immunology , Pulmonary Surfactant-Associated Protein D/immunology , Respiratory Hypersensitivity/immunology , T-Lymphocytes/immunology , Allergens/immunology , Animals , CTLA-4 Antigen , Enzyme-Linked Immunosorbent Assay , Immunoglobulin E/blood , Immunoglobulin E/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Ovalbumin/immunology , Reverse Transcriptase Polymerase Chain Reaction , Transfection
16.
J Immunol ; 185(7): 4284-91, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20817882

ABSTRACT

Acquired immune responses elicited to recent strains of seasonal H1N1 influenza viruses provide limited protection against emerging A(H1N1) pandemic viruses. Accordingly, pre-existing or rapidly induced innate immune defenses are of critical importance in limiting early infection. Respiratory secretions contain proteins of the innate immune system, including members of the collectin and pentraxin superfamilies. These mediate potent antiviral activity and act as an initial barrier to influenza infection. In this study, we have examined the sensitivity of H1N1 viruses, including pandemic virus strains, for their sensitivity to collectins (surfactant protein [SP]-D and mannose-binding lectin [MBL]) and to the pentraxin PTX3. Human SP-D and MBL inhibited virus-induced hemagglutinating activity, blocked the enzymatic activity of the viral neuraminidase, and neutralized the ability of H1N1 viruses to infect human respiratory epithelial cells in a manner that correlated with the degree of glycosylation in the globular head of the hemagglutinin. Recent seasonal H1N1 viruses expressed three to four N-glycosylation sequons on the head of hemagglutinin and were very sensitive to inhibition by SP-D or MBL, whereas A(H1N1) pandemic viruses expressed a single N-glycosylation sequon and were resistant to either collectin. Of interest, both seasonal and pandemic H1N1 viruses were resistant to PTX3. Thus, unlike recent seasonal H1N1 strains of influenza virus, A(H1N1) pandemic viruses are resistant to the antiviral activities of innate immune proteins of the collectin superfamily.


Subject(s)
C-Reactive Protein/immunology , Immune Evasion/immunology , Influenza A Virus, H1N1 Subtype/immunology , Mannose-Binding Lectin/immunology , Pulmonary Surfactant-Associated Protein D/immunology , Serum Amyloid P-Component/immunology , C-Reactive Protein/metabolism , Collectins/immunology , Disease Outbreaks , Enzyme-Linked Immunosorbent Assay , Glycosylation , Hemagglutination Tests , Hemagglutination, Viral , Hemagglutinins/chemistry , Hemagglutinins/genetics , Hemagglutinins/immunology , Humans , Immune Evasion/genetics , Immunity, Innate , Influenza A Virus, H1N1 Subtype/chemistry , Influenza A Virus, H1N1 Subtype/genetics , Mannose-Binding Lectin/metabolism , Neuraminidase/chemistry , Neuraminidase/genetics , Neuraminidase/immunology , Pulmonary Surfactant-Associated Protein D/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Serum Amyloid P-Component/metabolism
17.
medRxiv ; 2022 Sep 19.
Article in English | MEDLINE | ID: mdl-36172126

ABSTRACT

Respiratory viruses, including SARS-CoV-2, can trigger chronic lung disease that persists and even progresses after expected clearance of infectious virus. To gain an understanding of this process, we examined a series of consecutive fatal cases of Covid-19 that came to autopsy at 27-51 d after hospital admission. In each patient, we identify a stereotyped bronchiolar-alveolar pattern of lung remodeling with basal epithelial cell hyperplasia and mucinous differentiation. Remodeling regions also feature macrophage infiltration and apoptosis and a marked depletion of alveolar type 1 and 2 epithelial cells. This entire pattern closely resembles findings from an experimental model of post-viral lung disease that requires basal-epithelial stem cell growth, immune activation, and differentiation. The present results thereby provide evidence of possible basal epithelial cell reprogramming in long-term Covid-19 as well and thereby a pathway for explaining and correcting lung dysfunction in this type of disease.

18.
J Biol Chem ; 285(22): 16757-70, 2010 May 28.
Article in English | MEDLINE | ID: mdl-20228064

ABSTRACT

Surfactant protein D (SP-D) plays diverse and important roles in innate immunity and pulmonary homeostasis. Neutrophils and myeloperoxidase (MPO) colocalized with SP-D in a murine bacterial pneumonia model of acute inflammation, suggesting that MPO-derived reactive species might alter the function of SP-D. Exposure of SP-D to the complete MPO-H(2)O(2)-halide system caused loss of SP-D-dependent aggregating activity. Hypochlorous acid (HOCl), the major oxidant generated by MPO, caused a similar loss of aggregating activity, which was accompanied by the generation of abnormal disulfide-cross-linked oligomers. A full-length SP-D mutant lacking N-terminal cysteine residues and truncation mutants lacking the N-terminal domains were resistant to the oxidant-induced alterations in disulfide bonding. Mass spectroscopy of HOCl-treated human SP-D demonstrated several modifications, but none involved key ligand binding residues. There was detectable oxidation of cysteine 15, but no HOCl-induced cysteine modifications were observed in the C-terminal lectin domain. Together, the findings localize abnormal disulfide cross-links to the N-terminal domain. MPO-deficient mice showed decreased cross-linking of SP-D and increased SP-D-dependent aggregating activity in the pneumonia model. Thus, MPO-derived oxidants can lead to modifications of SP-D structure with associated alterations in its characteristic aggregating activity.


Subject(s)
Peroxidase/metabolism , Pulmonary Surfactant-Associated Protein D/chemistry , Animals , CHO Cells , Cricetinae , Cricetulus , Cysteine/chemistry , Disulfides/chemistry , Humans , In Vitro Techniques , Inflammation , Lectins/chemistry , Lung/metabolism , Mass Spectrometry/methods , Mice , Protein Structure, Tertiary , Rats
19.
PLoS Biol ; 6(11): e266, 2008 Nov 11.
Article in English | MEDLINE | ID: mdl-19007302

ABSTRACT

The pulmonary collectins, surfactant proteins A and D (SP-A and SP-D) have been implicated in the regulation of the innate immune system within the lung. In particular, SP-D appears to have both pro- and anti-inflammatory signaling functions. At present, the molecular mechanisms involved in switching between these functions remain unclear. SP-D differs in its quaternary structure from SP-A and the other members of the collectin family, such as C1q, in that it forms large multimers held together by the N-terminal domain, rather than aligning the triple helix domains in the traditional "bunch of flowers" arrangement. There are two cysteine residues within the hydrophobic N terminus of SP-D that are critical for multimer assembly and have been proposed to be involved in stabilizing disulfide bonds. Here we show that these cysteines exist within the reduced state in dodecameric SP-D and form a specific target for S-nitrosylation both in vitro and by endogenous, pulmonary derived nitric oxide (NO) within a rodent acute lung injury model. S-nitrosylation is becoming increasingly recognized as an important post-translational modification with signaling consequences. The formation of S-nitrosothiol (SNO)-SP-D both in vivo and in vitro results in a disruption of SP-D multimers such that trimers become evident. SNO-SP-D but not SP-D, either dodecameric or trimeric, is chemoattractive for macrophages and induces p38 MAPK phosphorylation. The signaling capacity of SNO-SP-D appears to be mediated by binding to calreticulin/CD91. We propose that NO controls the dichotomous nature of this pulmonary collectin and that posttranslational modification by S-nitrosylation causes quaternary structural alterations in SP-D, causing it to switch its inflammatory signaling role. This represents new insight into both the regulation of protein function by S-nitrosylation and NO's role in innate immunity.


Subject(s)
Acute Lung Injury/immunology , Inflammation/immunology , Nitric Oxide/metabolism , Pulmonary Surfactant-Associated Protein D/metabolism , S-Nitrosothiols/metabolism , Signal Transduction , Animals , Cysteine/chemistry , Cysteine/metabolism , Dimerization , Gene Expression Regulation , Humans , Immunity, Innate , Inflammation/metabolism , Lung/immunology , Lung/metabolism , Macrophages , Male , Mice , Pulmonary Surfactant-Associated Protein D/chemistry , Pulmonary Surfactant-Associated Protein D/genetics , Pulmonary Surfactant-Associated Protein D/immunology , Rats , Rats, Sprague-Dawley , Respiratory Mucosa/immunology
20.
J Immunol ; 182(12): 7878-87, 2009 Jun 15.
Article in English | MEDLINE | ID: mdl-19494312

ABSTRACT

We have reported that the alpha-defensins human neutrophil peptides (HNP)-1 and HNP-2 neutralize and aggregate influenza A virus (IAV) and promote uptake of IAV by neutrophils. These alpha-defensins were also shown to bind to surfactant protein (SP)-D and reduce its antiviral activity. In this study, we examined retrocyclin (RC)1 and RC2, humanized versions of the antiviral theta-defensins found in the leukocytes of certain nonhuman primates. RC1 was just as effective as HNP-1-3 in neutralizing IAV, and RC2 and RC101 (an analog of RC1) were more effective. In contrast, human beta-defensins (HBDs) showed less neutralizing activity. Human defensins 5 and 6 (mainly produced by intestinal Paneth cells) had viral neutralizing activity similar to HNP-1-3. Like HNP-1-3, RCs induced viral aggregation and promoted the uptake of IAV by neutrophils. We used surface plasmon resonance to evaluate binding of defensins to SP-D. HBDs, HD6, and HNP-4 bound minimally to SP-D. HNP-1-3 and RCs bound SP-D with high affinity; however, unlike HNP-1 and HNP-2, RCs did not inhibit SP-D antiviral activity. HBDs also did not inhibit antiviral activity of SP-D. Given their strong neutralizing activity and compatibility with SP-D, RCs may provide attractive prototypes for designing therapeutics that can prevent or treat respiratory infections caused by IAV.


Subject(s)
Defensins/immunology , Influenza A Virus, H1N1 Subtype/immunology , Pulmonary Surfactant-Associated Protein D/immunology , alpha-Defensins/immunology , beta-Defensins/immunology , Animals , Cell Line , Chickens , Cricetinae , Defensins/metabolism , Humans , Influenza A Virus, H1N1 Subtype/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice , Microscopy, Electron, Transmission , Neutrophils/immunology , Neutrophils/metabolism , Protein Binding , Pulmonary Surfactant-Associated Protein D/metabolism , alpha-Defensins/metabolism , beta-Defensins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL