Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Drug Deliv ; 28(1): 1419-1431, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34223777

ABSTRACT

Glucocorticoid (GC) hormone has been commonly used to treat systemic inflammation and immune disorders. However, the side effects associated with long-term use of high-dose GC hormone limit its clinical application seriously. GC hormone that can specifically target the lung might decrease the effective dosage and thus reduce GC-associated side effects. In this study, we successfully prepared human lung-targeting liposomal methylprednisolone crosslinked with nanobody (MPS-NSSLs-SPANb). Our findings indicate that MPS-NSSLs-SPANb may reduce the effective therapeutic dosage of MPS, achieve better efficacy, and reduce GC-associated side effects. In addition, MPS-NSSLs-SPANb showed higher efficacy and lower toxicity than conventional MPS.


Subject(s)
Idiopathic Pulmonary Fibrosis/drug therapy , Methylprednisolone/administration & dosage , Methylprednisolone/pharmacology , Pulmonary Surfactant-Associated Protein A/administration & dosage , Pulmonary Surfactant-Associated Protein A/pharmacology , Animals , Chemistry, Pharmaceutical , Drug Carriers/chemistry , Enzyme-Linked Immunosorbent Assay , Humans , Liposomes/chemistry , Lung/drug effects , Male , Mice , Mice, Nude , Random Allocation , Rats , Rats, Sprague-Dawley , Single-Domain Antibodies/administration & dosage , Single-Domain Antibodies/pharmacology
2.
Biol Reprod ; 99(3): 546-555, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29590302

ABSTRACT

Preterm birth accounts for the majority of neonatal morbidity and mortality in the developed world. A significant proportion of cases of spontaneous preterm labor are attributable to infections within gestational tissues. Surfactant protein A (SP-A), a collectin produced in the fetal lung and other tissues, has been shown previously in mice to suppress preterm delivery due to intrauterine (IU) instillation of sterile proinflammatory substances. Here we report a powerful antilabor effect for SP-A after IU infection with live Escherichia coli. SP-A abolished preterm birth (rate reduced from 100% to 0%) when it was administered into the uterus simultaneously with bacterial infection, reducing it by 75% when administered intravenously at the same time as IU bacterial inoculation, and by 48% when administered intravenously 4 h after IU bacterial infection. This effect on preterm delivery was accompanied by a parallel benefit on fetal survival in utero. SP-A had no effect on bacterial growth but reversed several major consequences of infection, including increased production of inflammatory mediators and a shift in macrophage polarization to the M1 phenotype. These findings suggest that exogenous SP-A has potential use to counteract infection-induced labor by reversing its proinflammatory consequences.


Subject(s)
Escherichia coli Infections/complications , Premature Birth/microbiology , Premature Birth/prevention & control , Pulmonary Surfactant-Associated Protein A/administration & dosage , Administration, Intravenous , Animals , Escherichia coli/drug effects , Escherichia coli/growth & development , Female , Inflammation/prevention & control , Macrophages/drug effects , Macrophages/physiology , Mice , Pregnancy , Uterus/drug effects
3.
PLoS One ; 10(5): e0126576, 2015.
Article in English | MEDLINE | ID: mdl-25965346

ABSTRACT

The surfactant protein (SP-A) receptor SP-R210 has been shown to increase phagocytosis of SP-A-bound pathogens and to modulate cytokine secretion by immune cells. SP-A plays an important role in pulmonary immunity by enhancing opsonization and clearance of pathogens and by modulating macrophage inflammatory responses. Alternative splicing of the Myo18A gene results in two isoforms: SP-R210S and SP-R210L, with the latter predominantly expressed in alveolar macrophages. In this study we show that SP-A is required for optimal expression of SP-R210L on alveolar macrophages. Interestingly, pre-treatment with SP-A prepared by different methods either enhances or suppresses responsiveness to LPS, possibly due to differential co-isolation of SP-B or other proteins. We also report that dominant negative disruption of SP-R210L augments expression of receptors including SR-A, CD14, and CD36, and enhances macrophages' inflammatory response to TLR stimulation. Finally, because SP-A is known to modulate CD14, we used a variety of techniques to investigate how SP-R210 mediates the effect of SP-A on CD14. These studies revealed a novel physical association between SP-R210S, CD14, and SR-A leading to an enhanced response to LPS, and found that SP-R210L and SP-R210S regulate internalization of CD14 via distinct macropinocytosis-like mechanisms. Together, our findings support a model in which SP-R210 isoforms differentially regulate trafficking, expression, and activation of innate immune receptors on macrophages.


Subject(s)
Inflammation/genetics , Lipopolysaccharide Receptors/genetics , Macrophages, Alveolar/immunology , Myosins/genetics , Pulmonary Surfactant-Associated Protein A/metabolism , Alternative Splicing/genetics , Humans , Immunity, Innate/genetics , Inflammation/chemically induced , Inflammation/immunology , Lipopolysaccharide Receptors/metabolism , Lipopolysaccharides/toxicity , Macrophages, Alveolar/metabolism , Myosins/metabolism , Phagocytosis , Protein Isoforms/genetics , Protein Isoforms/metabolism , Pulmonary Surfactant-Associated Protein A/administration & dosage
4.
J Pediatr Gastroenterol Nutr ; 60(5): 613-20, 2015 May.
Article in English | MEDLINE | ID: mdl-25539191

ABSTRACT

OBJECTIVES: Necrotizing enterocolitis (NEC) frequently results in significant morbidity and mortality in premature infants. Others reported that mice deficient in pulmonary surfactant protein-A (SP-A) born and raised in a nonhygienic environment succumb to significant gastrointestinal tract pathology, and enteral administration of purified SP-A significantly reduced mortality. We hypothesized that oral administration of purified SP-A can ameliorate pathology in an experimental model of neonatal NEC. METHODS: Experimental NEC was induced in newborn Sprague-Dawley rat pups by daily formula gavage and intermittent exposure to hypoxia. Purified human SP-A (5 µg/day) was administered by oral gavage. After 4 days, surviving pups were sacrificed, and intestinal pathology was assessed by histological examination of distal terminal ileal sections. Intestinal levels of inflammatory cytokines (IL-1ß, IFN-γ, and TNF-α) were assessed by enzyme-linked immunosorbent assay and levels of Toll-like receptor 4 (TLR4) by Western analysis. RESULTS: Sixty-one percent of the gavaged rat pups that survived to day 4 met the criteria for experimental NEC after hypoxia, whereas treatment with SP-A significantly reduced mortality and assessment of NEC. Intestinal levels of proinflammatory cytokines were significantly increased in pups exposed to hypoxia. Administration of SP-A to pups exposed to hypoxia significantly reduced IL-1ß and TNF-α levels, but had little effect on elevated levels of IFN-γ. SP-A treatment of hypoxia-exposed pups significantly reduced expression of intestinal TLR4, key in NEC pathogenesis. CONCLUSIONS: In a rat model of experimental neonatal NEC, oral administration of SP-A reduces intestinal levels of proinflammatory cytokines and TLR4 protein and ameliorates adverse outcomes associated with gastrointestinal pathologies.


Subject(s)
Cytokines/metabolism , Enterocolitis, Necrotizing/drug therapy , Pulmonary Surfactant-Associated Protein A/administration & dosage , Pulmonary Surfactants/administration & dosage , Administration, Oral , Animals , Disease Models, Animal , Enterocolitis, Necrotizing/metabolism , Enterocolitis, Necrotizing/pathology , Ileum/metabolism , Interferon-gamma/metabolism , Interleukin-1beta/metabolism , Rats , Rats, Sprague-Dawley , Toll-Like Receptor 4/metabolism , Tumor Necrosis Factor-alpha/metabolism
5.
Eur J Pharm Biopharm ; 77(3): 384-91, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21172435

ABSTRACT

Intratracheal administration of immunosuppressive agents to the lung is a novel treatment after lung transplantation. Nanoparticles of tacrolimus (FK506) might interact with human SP-A, which is the most abundant lipoprotein in the alveolar fluid. This study was undertaken to determine whether the formation of FK506/SP-A complexes interferes with FK506 immunosuppressive actions on stimulated human macrophage-like U937 cells. We found that SP-A was avidly bound to FK506 (K(d) = 35 ± 4nM), as determined by solid phase-binding assays and dynamic light scattering. Free FK506, at concentrations ≤ 1 µM, had no effect on the inflammatory response of LPS-stimulated U937 macrophages. However, coincubation of FK506 and SP-A, at concentrations where each component alone did not affect LPS-stimulated macrophage response, significantly inhibited LPS-induced NF-κB activation and TNF-alpha secretion. Free FK506, but not FK506/SP-A, functioned as substrate for the efflux transporter P-glycoprotein. FK506 bound to SP-A was delivered to macrophages by endocytosis, since several endocytosis inhibitors blocked FK506/SP-A anti-inflammatory effects. This process depended partly on SP-A binding to its receptor, SP-R210. These results indicate that FK506/SP-A complexes have a greater anti-inflammatory effect than either FK506 or SP-A alone and suggest that SP-A strengthened FK506 anti-inflammatory activity by facilitating FK506 entrance into the cell, overcoming P-glycoprotein.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Immunosuppressive Agents/pharmacology , Macrophages/drug effects , Pulmonary Surfactant-Associated Protein A/pharmacology , Tacrolimus/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Cell Survival/drug effects , Dose-Response Relationship, Drug , Endocytosis/drug effects , Humans , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/metabolism , Lipopolysaccharides/pharmacology , Macrophages/immunology , Macrophages/metabolism , Models, Molecular , Molecular Structure , Protein Binding , Pulmonary Surfactant-Associated Protein A/administration & dosage , Pulmonary Surfactant-Associated Protein A/metabolism , Receptors, Cell Surface/metabolism , Tacrolimus/administration & dosage , Tacrolimus/metabolism , U937 Cells
6.
FASEB J ; 24(10): 3662-73, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20484671

ABSTRACT

The purpose of this study was to investigate how surfactant membranes can be perturbed by C-reactive protein (CRP) and whether surfactant protein A (SP-A) might overcome CRP-induced surfactant membrane alterations. The effect of CRP on surfactant surface adsorption was evaluated in vivo after intratracheal instillation of CRP into rat lungs. Insertion of CRP into surfactant membranes was investigated through monolayer techniques. The effect of CRP on membrane structure was studied through differential scanning calorimetry and fluorescence spectroscopy and microscopy using large and giant unilamellar vesicles. Our results indicate that CRP inserts into surfactant membranes and drastically increases membrane fluidity, resulting in surfactant inactivation. At 10% CRP/phospholipid weight ratio, CRP causes disappearance of liquid-ordered/liquid-disordered phase coexistence distinctive of surfactant membranes. SP-A, the most abundant surfactant lipoprotein structurally similar to C1q, binds to CRP (K(d)=56±8 nM), as determined by solid-phase binding assays and dynamic light scattering. This novel SP-A/CRP interaction reduces CRP insertion and blocks CRP effects on surfactant membranes. In addition, intratracheal coinstillation of SP-A+CRP into rat lungs prevents surfactant inhibition induced by CRP, indicating that SP-A/CRP interactions might be an important factor in vivo in controlling harmful CRP effects in the alveolus.


Subject(s)
C-Reactive Protein/pharmacology , Membrane Fluidity/drug effects , Pulmonary Surfactant-Associated Protein A/pharmacology , Animals , Bronchoalveolar Lavage Fluid , C-Reactive Protein/administration & dosage , Calorimetry, Differential Scanning , Cell Membrane/metabolism , Male , Microscopy, Fluorescence , Pulmonary Surfactant-Associated Protein A/administration & dosage , Rats , Rats, Sprague-Dawley , Spectrometry, Fluorescence
7.
Recent Pat Antiinfect Drug Discov ; 5(2): 115-23, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20230362

ABSTRACT

Surfactant protein (SP)-A and SP-D belong to the "Soluble C-type Lectin" family of proteins and are collectively known as "Collectins". Based on their ability to recognize pathogens and to regulate the host defense, SP-A and SP-D have been recently categorized as "Secretory Pathogen Recognition Receptors". SP-A and SP-D were first identified in the lung; the expression of SP-A and SP-D has also been observed at other mucosal surfaces, such as lacrimal glands, gastrointestinal mucosa, genitourinary epithelium and periodontal surfaces. Since the role of these proteins is not fully elucidated at other mucosal surfaces, the focus of this article is on lung-SP-A and SP-D. It has become clear from research studies performed over a number of years that SP-A and SP-D are critical for the maintenance of lung homeostasis and the regulation of host defense and inflammation. However, none of the surfactant preparations available for clinical use have SP-A or SP-D. A review is presented here on SP-A- and SP-D-deficiencies in lung diseases, the importance of the administration of SP-A and SP-D, and recent patents and research directions that may lead to the design of novel SP-A- or SP-D-based therapeutics and surfactants.


Subject(s)
Anti-Infective Agents/administration & dosage , Immunologic Factors/therapeutic use , Inflammation/drug therapy , Lung Diseases/drug therapy , Pulmonary Surfactant-Associated Protein A/administration & dosage , Pulmonary Surfactant-Associated Protein D/administration & dosage , Animals , Anti-Infective Agents/pharmacology , Humans , Lung Diseases/metabolism , Patents as Topic , Pulmonary Surfactant-Associated Protein A/deficiency , Pulmonary Surfactant-Associated Protein A/pharmacology , Pulmonary Surfactant-Associated Protein D/deficiency , Pulmonary Surfactant-Associated Protein D/pharmacology , Pulmonary Surfactants/immunology , Pulmonary Surfactants/therapeutic use , Recombinant Proteins/therapeutic use , Surface-Active Agents/therapeutic use
8.
Pediatr Res ; 66(2): 162-7, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19390477

ABSTRACT

Ureaplasma respiratory tract colonization stimulates prolonged, dysregulated inflammation in the lungs of preterm infants, contributing to bronchopulmonary dysplasia (BPD) pathogenesis. Surfactant protein-A (SP-A), a lung collectin critical for bacterial clearance and regulating inflammation, is deficient in the preterm lung. To analyze the role of SP-A in modulating Ureaplasma-mediated lung inflammation, SP-A deficient (SP-A-/-) and WT mice were inoculated intratracheally with a mouse-adapted U. parvum isolate and indices of inflammation were sequentially assessed up to 28 d postinoculation. Compared with infected WT and noninfected controls, Ureaplasma-infected SP-A-/- mice exhibited an exaggerated inflammatory response evidenced by rapid influx of neutrophils and macrophages into the lung, and higher bronchoalveolar lavage TNF-alpha, mouse analogue of human growth-related protein alpha (KC), and monocyte chemotactic factor (MCP-1) concentrations. However, nitrite generation in response to Ureaplasma infection was blunted at 24 h and Ureaplasma clearance was delayed in SP-A-/- mice compared with WT mice. Coadministration of human SP-A with the Ureaplasma inoculum to SP-A-/- mice reduced the inflammatory response, but did not improve the bacterial clearance rate. SP-A deficiency may contribute to the prolonged inflammatory response in the Ureaplasma-infected preterm lung, but other factors may contribute to the impaired Ureaplasma clearance.


Subject(s)
Inflammation , Lung , Pneumonia/microbiology , Pneumonia/physiopathology , Pulmonary Surfactant-Associated Protein A/metabolism , Ureaplasma/metabolism , Animals , Bronchoalveolar Lavage Fluid/microbiology , Cytokines/metabolism , Humans , Infant, Newborn , Inflammation/microbiology , Inflammation/pathology , Lung/cytology , Lung/metabolism , Lung/microbiology , Mice , Mice, Knockout , Nitric Oxide/metabolism , Pneumonia/pathology , Pulmonary Surfactant-Associated Protein A/administration & dosage , Pulmonary Surfactant-Associated Protein A/genetics
9.
Infect Immun ; 76(1): 380-90, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17967856

ABSTRACT

The collectins surfactant-associated protein A (SP-A) and SP-D are components of innate immunity that are present before birth. Both proteins bind pathogens and assist in clearing infection. The significance of SP-A and SP-D as components of the neonatal immune system has not been investigated. To determine the role of SP-A and SP-D in neonatal immunity, wild-type, SP-A null, and SP-D null mice were bred in a bacterium-laden environment (corn dust bedding) or in a semisterile environment (cellulose fiber bedding). When reared in the corn dust bedding, SP-A null pups had significant mortality (P < 0.001) compared to both wild-type and SP-D null pups exposed to the same environment. The mortality of the SP-A null pups was associated with significant gastrointestinal tract pathology but little lung pathology. Moribund SP-A null newborn mice exhibited Bacillus sp. and Enterococcus sp. peritonitis. When the mother or newborn produced SP-A, newborn survival was significantly improved (P < 0.05) compared to the results when there was a complete absence of SP-A in both the mother and the pup. Significant sources of SP-A likely to protect a newborn include the neonatal lung and gastrointestinal tract but not the lactating mammary tissue of the mother. Furthermore, exogenous SP-A delivered by mouth to newborn SP-A null pups with SP-A null mothers improved newborn survival in the corn dust environment. Therefore, a lack of SP-D did not affect newborn survival, while SP-A produced by either the mother or the pup or oral exogenous SP-A significantly reduced newborn mortality associated with environmentally induced infection in SP-A null newborns.


Subject(s)
Animals, Newborn/immunology , Pulmonary Surfactant-Associated Protein A/immunology , Animals , Bedding and Linens , Dust , Female , Gene Deletion , Gene Expression , Humans , Immunity, Maternally-Acquired , Litter Size , Male , Mice , Pulmonary Surfactant-Associated Protein A/administration & dosage , Pulmonary Surfactant-Associated Protein A/genetics , Pulmonary Surfactant-Associated Protein D , Zea mays
10.
Respir Res ; 6: 97, 2005 Aug 24.
Article in English | MEDLINE | ID: mdl-16120217

ABSTRACT

BACKGROUND: The pulmonary surfactant protein (SP)-A has potent immunomodulatory activities but its role and regulation during allergic airway inflammation is unknown. METHODS: We studied changes in SP-A expression in the bronchoalveolar lavage (BAL) using a murine model of single Aspergillus fumigatus (Af) challenge of sensitized animals. RESULTS: SP-A protein levels in the BAL fluid showed a rapid, transient decline that reached the lowest values (25% of controls) 12 h after intranasal Af provocation of sensitized mice. Decrease of SP-A was associated with influx of inflammatory cells and increase of IL-4 and IL-5 mRNA and protein levels. Since levels of SP-A showed a significant negative correlation with these BAL cytokines (but not with IFN-gamma), we hypothesized that SP-A exerts an inhibitory effect on Th2-type immune responses. To study this hypothesis, we used an in vitro Af-rechallenge model. Af-induced lymphocyte proliferation of cells isolated from sensitized mice was inhibited in a dose-dependent manner by addition of purified human SP-A (0.1-10 microg/ml). Flow cytometric studies on Af-stimulated lymphocytes indicated that the numbers of CD4+ (but not CD8+) T cells were significantly increased in the parental population and decreased in the third and fourth generation in the presence of SP-A. Further, addition of SP-A to the tissue culture inhibited Af-induced IL-4 and IL-5 production suggesting that SP-A directly suppressed allergen-stimulated CD4+ T cell function. CONCLUSION: We speculate that a transient lack of this lung collectin following allergen exposure of the airways may significantly contribute to the development of a T-cell dependent allergic immune response.


Subject(s)
Aspergillosis/immunology , Aspergillosis/pathology , Aspergillus fumigatus/immunology , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Pulmonary Surfactant-Associated Protein A/immunology , T-Lymphocytes/immunology , Animals , Bronchial Provocation Tests , Bronchoalveolar Lavage Fluid/microbiology , Cells, Cultured , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Mice , Pulmonary Surfactant-Associated Protein A/administration & dosage
11.
J Immunol ; 174(11): 6943-54, 2005 Jun 01.
Article in English | MEDLINE | ID: mdl-15905537

ABSTRACT

Lung surfactant protein A (SP-A) and D (SP-D) are innate immune molecules which are known to interact with allergens and immune cells and modulate cytokine and chemokine profiles during host hypersensitivity response. We have previously shown therapeutic effects of SP-A and SP-D using a murine model of lung hypersensitivity to Aspergillus fumigatus (Afu) allergens. In this study, we have examined the susceptibility of SP-A (AKO) or SP-D gene-deficient (DKO) mice to the Afu allergen challenge, as compared with the wild-type mice. Both AKO and DKO mice exhibited intrinsic hypereosinophilia and several-fold increase in levels of IL-5 and IL-13, and lowering of IFN-gamma to IL-4 ratio in the lungs, suggesting a Th2 bias of immune response. This Th2 bias was reversible by treating AKO or DKO mice with SP-A or SP-D, respectively. The AKO and DKO mice showed distinct immune responses to Afu sensitization. DKO mice were found more susceptible than wild-type mice to pulmonary hypersensitivity induced by Afu allergens. AKO mice were found to be nearly resistant to Afu sensitization. Intranasal treatment with SP-D or rhSP-D (a recombinant fragment of human SP-D containing trimeric C-type lectin domains) was effective in rescuing the Afu-sensitized DKO mice, while SP-A-treated Afu-sensitized AKO mice showed several-fold elevated levels of IL-13 and IL-5, resulting in increased pulmonary eosinophilia and damaged lung tissue. These data reaffirm an important role for SP-A and SP-D in offering resistance to pulmonary allergenic challenge.


Subject(s)
Allergens/immunology , Antigens, Fungal/immunology , Aspergillosis, Allergic Bronchopulmonary/genetics , Aspergillosis, Allergic Bronchopulmonary/immunology , Aspergillus fumigatus/immunology , Genetic Predisposition to Disease/genetics , Pulmonary Surfactant-Associated Protein A/deficiency , Pulmonary Surfactant-Associated Protein D/deficiency , Allergens/administration & dosage , Animals , Antigens, Fungal/administration & dosage , Aspergillosis, Allergic Bronchopulmonary/pathology , Cells, Cultured , Erythropoietin/biosynthesis , Erythropoietin/metabolism , Female , Humans , Interleukin-13/antagonists & inhibitors , Interleukin-13/biosynthesis , Interleukin-15/antagonists & inhibitors , Interleukin-15/biosynthesis , Interleukin-5/biosynthesis , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pulmonary Eosinophilia/genetics , Pulmonary Eosinophilia/immunology , Pulmonary Eosinophilia/prevention & control , Pulmonary Surfactant-Associated Protein A/administration & dosage , Pulmonary Surfactant-Associated Protein A/genetics , Pulmonary Surfactant-Associated Protein D/administration & dosage , Pulmonary Surfactant-Associated Protein D/genetics , Pulmonary Surfactant-Associated Protein D/therapeutic use , Recombinant Proteins/administration & dosage , Recombinant Proteins/therapeutic use , Serum Albumin, Bovine/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...