Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Sci Transl Med ; 16(754): eadl3848, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38959329

ABSTRACT

Autoantibodies to nuclear antigens are hallmarks of systemic lupus erythematosus (SLE) where they contribute to pathogenesis. However, there remains a gap in our knowledge regarding how different isotypes of autoantibodies contribute to this autoimmune disease, including the production of the critical type I interferon (IFN) cytokines by plasmacytoid dendritic cells (pDCs) in response to immune complexes (ICs). We focused on IgA, which is the second-most prevalent isotype in serum and, along with IgG, is deposited in glomeruli in individuals with lupus nephritis. We show that individuals with SLE have serum IgA autoantibodies against most nuclear antigens, correlating with IgG against the same antigen. We investigated whether IgA autoantibodies against a major SLE autoantigen, Smith ribonucleoprotein (Sm/RNP), played a role in IC activation of pDCs. We found that pDCs expressed the IgA-specific Fc receptor, FcαR, and IgA1 autoantibodies synergized with IgG in RNA-containing ICs to generate robust primary blood pDC IFN-α responses in vitro. pDC responses to these ICs required both FcαR and FcγRIIa, showing synergy between these Fc receptors. Sm/RNP IC binding to and internalization by pDCs were greater when ICs contained both IgA1 and IgG. Circulating pDCs from individuals with SLE had higher binding of IgA1-containing ICs and higher expression of FcαR than pDCs from healthy control individuals. Although pDC FcαR expression correlated with the blood IFN-stimulated gene signature in SLE, Toll-like receptor 7 agonists, but not IFN-α, up-regulated pDC FcαR expression in vitro. Together, we show a mechanism by which IgA1 autoantibodies contribute to SLE pathogenesis.


Subject(s)
Antigen-Antibody Complex , Autoantibodies , Dendritic Cells , Immunoglobulin A , Immunoglobulin G , Lupus Erythematosus, Systemic , Humans , Dendritic Cells/immunology , Dendritic Cells/metabolism , Immunoglobulin A/immunology , Immunoglobulin A/metabolism , Immunoglobulin A/blood , Autoantibodies/immunology , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Antigen-Antibody Complex/immunology , Antigen-Antibody Complex/metabolism , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/blood , RNA/metabolism , Female , Interferon-alpha/metabolism , Adult , Receptors, Fc/metabolism , Receptors, Fc/immunology , Toll-Like Receptor 7/metabolism , Male , Receptors, IgG/metabolism
2.
bioRxiv ; 2024 Apr 04.
Article in English | MEDLINE | ID: mdl-37745328

ABSTRACT

Autoantibodies to nuclear antigens are hallmarks of the autoimmune disease systemic lupus erythematosus (SLE) where they contribute to pathogenesis. However, there remains a gap in our knowledge regarding how different isotypes of autoantibodies contribute to disease, including the production of the critical type I interferon (IFN) cytokines by plasmacytoid dendritic cells (pDCs) in response to immune complexes (ICs). We focused on IgA, which is the second most prevalent isotype in serum, and along with IgG is deposited in glomeruli in lupus nephritis. Here, we show that individuals with SLE have IgA autoantibodies against most nuclear antigens, correlating with IgG against the same antigen. We investigated whether IgA autoantibodies against a major SLE autoantigen, Smith ribonucleoproteins (Sm/RNPs), play a role in IC activation of pDCs. We found that pDCs express the IgA-specific Fc receptor, FcαR, and there was a striking ability of IgA1 autoantibodies to synergize with IgG in RNA-containing ICs to generate robust pDC IFNα responses. pDC responses to these ICs required both FcαR and FcγRIIa, showing a potent synergy between these Fc receptors. Sm/RNP IC binding to and internalization by pDCs were greater when ICs contained both IgA1 and IgG. pDCs from individuals with SLE had higher binding of IgA1-containing ICs and higher expression of FcαR than pDCs from healthy control individuals. Whereas pDC FcαR expression correlated with blood ISG signature in SLE, TLR7 agonists, but not IFNα, upregulated pDC FcαR expression in vitro. Together, we show a new mechanism by which IgA1 autoantibodies contribute to SLE pathogenesis.

3.
Sci Signal ; 12(581)2019 05 14.
Article in English | MEDLINE | ID: mdl-31088976

ABSTRACT

B cell adaptor for phosphoinositide 3-kinase (PI3K) (BCAP) is a signaling adaptor that activates the PI3K pathway downstream of B cell receptor signaling in B cells and Toll-like receptor (TLR) signaling in macrophages. BCAP binds to the regulatory p85 subunit of class I PI3K and is a large, multidomain protein. We used proteomic analysis to identify other BCAP-interacting proteins in macrophages and found that BCAP specifically associated with the caspase-1 pseudosubstrate inhibitor Flightless-1 and its binding partner leucine-rich repeat flightless-interacting protein 2. Because these proteins inhibit the NLRP3 inflammasome, we investigated the role of BCAP in inflammasome function. Independent of its effects on TLR priming, BCAP inhibited NLRP3- and NLRC4-induced caspase-1 activation, cell death, and IL-1ß release from macrophages. Accordingly, caspase-1-dependent clearance of a Yersinia pseudotuberculosis mutant was enhanced in BCAP-deficient mice. Mechanistically, BCAP delayed the recruitment and activation of pro-caspase-1 within the NLRP3/ASC preinflammasome through its association with Flightless-1. Thus, BCAP is a multifunctional signaling adaptor that inhibits key pathogen-sensing pathways in macrophages.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism , Calcium-Binding Proteins/metabolism , Inflammasomes/metabolism , Macrophages/metabolism , Microfilament Proteins/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Trans-Activators/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Caspase 1/genetics , Caspase 1/metabolism , Cells, Cultured , HEK293 Cells , Humans , Macrophages/microbiology , Mice, Inbred C57BL , Mice, Knockout , Mutation , Protein Binding , Yersinia pseudotuberculosis/genetics , Yersinia pseudotuberculosis/physiology
4.
J Immunol ; 202(9): 2529-2534, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30936294

ABSTRACT

Systemic lupus erythematosus severity correlates with elevated serum levels of type I IFNs, cytokines produced in large quantities by plasmacytoid dendritic cells (pDC) in response to engagement of TLR7 and TLR9 with endocytosed nucleic acids. B cell adaptor for PI3K (BCAP) promoted many aspects of TLR7-driven lupus-like disease, including Isg15 and Ifit1 expression in blood and an immature pDC phenotype associated with higher IFN production. BCAP-/- mice produced significantly less serum IFN-α than wild-type mice after injection of TLR9 agonist, and BCAP promoted TLR7 and TLR9-induced IFN-α production specifically in pDC. TLR-induced IFN-α production in pDC requires DOCK2-mediated activation of Rac1 leading to activation of IKKα, a mechanism we show was dependent on BCAP. BCAP-/- pDC had decreased actin polymerization and Rac1 activation and reduced IKKα phosphorylation upon TLR9 stimulation. We show a novel role for BCAP in promoting TLR-induced IFN-α production in pDC and in systemic lupus erythematosus pathogenesis.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Dendritic Cells/immunology , Interferon-alpha/immunology , Lupus Erythematosus, Systemic/immunology , Membrane Glycoproteins/immunology , Plasma Cells/immunology , Toll-Like Receptor 7/immunology , Toll-Like Receptor 9/immunology , Adaptor Proteins, Signal Transducing/genetics , Animals , Cytokines/genetics , Cytokines/immunology , Dendritic Cells/pathology , Female , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/immunology , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/immunology , Interferon-alpha/genetics , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/pathology , Male , Membrane Glycoproteins/genetics , Mice , Mice, Knockout , Neuropeptides/genetics , Neuropeptides/immunology , Plasma Cells/pathology , RNA-Binding Proteins/genetics , RNA-Binding Proteins/immunology , Toll-Like Receptor 7/genetics , Toll-Like Receptor 9/genetics , Ubiquitins/genetics , Ubiquitins/immunology , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/immunology
5.
J Exp Med ; 215(9): 2429-2443, 2018 09 03.
Article in English | MEDLINE | ID: mdl-30093532

ABSTRACT

CD8+ T cells respond to signals via the T cell receptor (TCR), costimulatory molecules, and immunoregulatory cytokines by developing into diverse populations of effector and memory cells. The relative strength of phosphoinositide 3-kinase (PI3K) signaling early in the T cell response can dramatically influence downstream effector and memory T cell differentiation. We show that initial PI3K signaling during T cell activation results in up-regulation of the signaling scaffold B cell adaptor for PI3K (BCAP), which further potentiates PI3K signaling and promotes the accumulation of CD8+ T cells with a terminally differentiated effector phenotype. Accordingly, BCAP-deficient CD8+ T cells have attenuated clonal expansion and altered effector and memory T cell development following infection with Listeria monocytogenes Thus, induction of BCAP serves as a positive feedback circuit to enhance PI3K signaling in activated CD8+ T cells, thereby acting as a molecular checkpoint regulating effector and memory T cell development.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , CD8-Positive T-Lymphocytes/immunology , Carrier Proteins/immunology , Cell Differentiation/immunology , Immunologic Memory , Signal Transduction/immunology , Adaptor Proteins, Signal Transducing/genetics , Animals , CD8-Positive T-Lymphocytes/pathology , Carrier Proteins/genetics , Cell Differentiation/genetics , Humans , Listeria monocytogenes/immunology , Listeriosis/genetics , Listeriosis/immunology , Listeriosis/pathology , Lymphocyte Activation , Mice , Mice, Transgenic , Signal Transduction/genetics
6.
Immunol Rev ; 269(1): 212-27, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26683155

ABSTRACT

Toll-like receptors (TLR) are transmembrane pattern recognition receptors that recognize microbial ligands and signal for production of inflammatory cytokines and type I interferon in macrophages and dendritic cells (DC). Whereas TLR-induced inflammatory mediators are required for pathogen clearance, many are toxic to the host and can cause pathological inflammation when over-produced. This is demonstrated by the role of TLR-induced cytokines in autoimmune diseases, such as rheumatoid arthritis, inflammatory bowel disease, and systemic lupus erythematosus. Because of the potent effects of TLR-induced cytokines, we have diverse mechanisms to dampen TLR signaling. Here, we highlight three pathways that participate in inhibition of TLR responses in macrophages and DC, and their implications in autoimmunity; A20, encoded by the TNFAIP3 gene, Lyp encoded by the PTPN22 gene, and the BCAP/PI3K pathway. We present new findings that Lyp promotes TLR responses in primary human monocytes and that the autoimmunity risk Lyp620W variant is more effective at promoting TLR-induced interleukin-6 than the non-risk Lyp620R protein. This suggests that Lyp serves to downregulate a TLR inhibitory pathway in monocytes, and we propose that Lyp inhibits the TREM2/DAP12 inhibitory pathway. Overall, these pathways demonstrate distinct mechanisms of negative regulation of TLR responses, and all impact autoimmune disease pathogenesis and treatment.


Subject(s)
Autoimmune Diseases/immunology , Dendritic Cells/immunology , Macrophages/immunology , Myeloid Cells/immunology , Toll-Like Receptors/metabolism , Animals , Carrier Proteins/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Humans , Immunomodulation , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 22/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 22/metabolism , Signal Transduction , Tumor Necrosis Factor alpha-Induced Protein 3
7.
Proc Natl Acad Sci U S A ; 109(1): 267-72, 2012 Jan 03.
Article in English | MEDLINE | ID: mdl-22187458

ABSTRACT

Toll-like receptors (TLRs) recognize pathogens and their components, thereby initiating immune responses to infectious organisms. TLR ligation leads to the activation of NF-κB and MAPKs through well-defined pathways, but it has remained unclear how TLR signaling activates PI3K, which provides an inhibitory pathway limiting TLR responses. Here, we show that the signaling adapter B-cell adaptor for PI3K (BCAP) links TLR signaling to PI3K activation. BCAP-deficient macrophages and mice are hyperresponsive to TLR agonists and have reduced PI3K activation. The ability of BCAP to inhibit TLR responses requires its capacity to bind PI3K. BCAP is constitutively phosphorylated and associated with the p85 subunit of PI3K in macrophages. This tyrosine-phosphorylated BCAP is transiently enriched in the membrane fraction in response to LPS treatment, suggesting a model whereby TLR signaling causes the phosphorylation of the small amount of BCAP that is associated with membranes in the resting state or the translocation of phosphorylated BCAP from the cytoplasm to the membrane. This accumulation of tyrosine-phosphorylated BCAP at the membrane with its associated PI3K would then allow for the catalysis of Ptd Ins P2 to Ptd Ins P3 and downstream PI3K-dependent signals. Therefore, BCAP is an essential activator of the PI3K pathway downstream of TLR signaling, providing a brake to limit potentially pathogenic excessive TLR responses.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , B-Lymphocytes/enzymology , Phosphatidylinositol 3-Kinases/metabolism , Toll-Like Receptors/metabolism , Adaptor Proteins, Signal Transducing/deficiency , Animals , B-Lymphocytes/drug effects , Cytokines/biosynthesis , Enzyme Activation/drug effects , I-kappa B Proteins/metabolism , Inflammation/pathology , Intracellular Signaling Peptides and Proteins/metabolism , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/enzymology , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/metabolism , NF-KappaB Inhibitor alpha , Phosphorylation/drug effects , Phosphotyrosine/metabolism , Protein Binding/drug effects , Protein Subunits/metabolism , Protein-Tyrosine Kinases/metabolism , Proteolysis/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism , Syk Kinase
8.
Immunol Rev ; 232(1): 42-58, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19909355

ABSTRACT

The adapter proteins DAP12 and FcRgamma associate with a wide spectrum of receptors in a variety of innate immune cells to mediate intracellular signaling pathways when their cognate receptor is engaged. These adapter proteins are coupled to their receptors through charged residues within the transmembrane regions of the adapter and receptor. DAP12 and FcRgamma contain specific protein domains (referred to as immunoreceptor tyrosine-based activation motifs) that serve as the substrates and docking sites for kinases, allowing amplification of intracellular signaling reactions. Recent research has broadened the repertoire of receptors that utilize these adapters for signaling to include not only novel immunoglobulin superfamily members but also cytokine receptors, integrins, and other adhesion molecules. There is abundant evidence that these multifunctional signaling adapters also mediate inhibitory activity, downmodulating signaling from Toll-like receptors and other heterologous receptors. In this review, we discuss the newly described receptors that utilize DAP12 and/or FcRgamma adapters to modulate innate immune responses.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Membrane Proteins/metabolism , Myeloid Cells/metabolism , Receptors, IgG/metabolism , Adaptor Proteins, Signal Transducing/immunology , Animals , Humans , Immunity, Innate , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/immunology , Myeloid Cells/cytology , Myeloid Cells/immunology , Protein Interaction Domains and Motifs , Receptors, IgG/immunology , Signal Transduction/immunology , Tyrosine/metabolism
9.
Curr Eye Res ; 34(2): 92-8, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19219679

ABSTRACT

PURPOSE: To use protein arrays to delineate the spectrum of angiogenic bioactive protein modulators that might be secreted and up-regulated by the corneal epithelium in response to killed bacterial products. METHODS: Immortalized human corneal epithelial cells were grown in culture, serum starved, and exposed to heat-killed Pseudomonas aeruginosa in a dose-dependent manner. The resultant culture medium was screened by antibody arrays for 43 proteins that can modulate angiogenesis and immune and inflammatory processes. Parallel analysis was carried out on tears recovered in the open and closed eye phases (OTF and CTF) of the diurnal cycle. RESULTS: Array analysis reveals that the immortalized cells constitutively secrete several proteins and up-regulate the secretion of IL-6, IL-8, and GRO in response to killed bacteria. Also evident was the emergence of a strong signal for GM-CSF and moderate/weak signals for MCP-1, MMP-9, Leptin, and INF gamma in a dose-dependent manner. Several of these proteins, including IL-6, IL-8, GRO, MMP-9, TIMP-1, and MCP-1, accumulate in the CTF. Other proteins are unique to tear fluid. CONCLUSIONS: Nine proteins were identified that are secreted by epithelium in response to killed bacteria that contribute to the innate and adaptive defense system through potentiating PMN and macrophage recruitment, activation, and opsonization in a cooperative manner. The vast majority of these proteins are angiogenic modulators, perhaps contributing to the imbalance between angiogenic and angiostatic processes and risk of corneal vascularization.


Subject(s)
Epithelium, Corneal/metabolism , Eye Proteins/metabolism , Protein Array Analysis/methods , Pseudomonas aeruginosa/physiology , Angiogenesis Modulating Agents/metabolism , Cell Line , Cell Membrane , Cytokines/metabolism , Epithelium, Corneal/microbiology , Epithelium, Corneal/pathology , Humans , Immunity, Innate , Tears/metabolism , Up-Regulation
10.
FEMS Immunol Med Microbiol ; 54(2): 177-84, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18657106

ABSTRACT

We reported previously that surfactant protein D (SP-D) was present in human tears and corneal epithelial cells, and that it contributed to tear fluid protection of those cells against Pseudomonas aeruginosa invasion. This suggested a role in ocular innate immunity. Here, we explored the effects of bacterial challenge on SP-D expression by human corneal epithelial cells. Results showed that these cells produced and secreted SP-D constitutively in culture, and that production (mRNA, protein) and secretion of SP-D were upregulated after exposure to heat-killed P. aeruginosa or to purified flagellin or lipopolysaccharide. To begin exploring the mechanism for flagellin-mediated SP-D induction, cells were exposed to purified flagellin or flagellin mutated in the TLR-5-binding domain (L94A, L88A) which reduces IL-8 secretion by A549 respiratory cells. Mutated flagellin did not upregulate IL-8 expression in corneal epithelial cells, but did induce SP-D responses. Mitogen-activated protein kinase inhibitors, especially the JNK inhibitor SP600125, reduced secretion of SP-D, but not production, in the presence of P. aeruginosa. These data show that while SP-D and IL-8 corneal responses are each induced by P. aeruginosa or its antigens, they can involve different regions of the same ligand. The data suggest that separate mechanisms may regulate SP-D secretion and production by human corneal epithelia.


Subject(s)
Epithelium, Corneal/immunology , Epithelium, Corneal/metabolism , Pseudomonas aeruginosa/immunology , Pulmonary Surfactant-Associated Protein D/metabolism , Analysis of Variance , Antigens, Bacterial/genetics , Antigens, Bacterial/metabolism , Cell Line, Transformed , Enzyme Inhibitors/pharmacology , Epithelium, Corneal/microbiology , Flagellin/genetics , Flagellin/immunology , Gene Expression Regulation , Humans , Interleukin-8/metabolism , Ligands , Lipopolysaccharides/immunology , Point Mutation , Pseudomonas aeruginosa/genetics , Pulmonary Surfactant-Associated Protein D/genetics , Up-Regulation
11.
Infect Immun ; 75(5): 2325-32, 2007 May.
Article in English | MEDLINE | ID: mdl-17325054

ABSTRACT

Pseudomonas aeruginosa keratitis is an acute sight-threatening infection. We previously reported that human tear fluid could protect individual human corneal epithelial cells in vitro against invasion by and cytotoxicity due to clinical and laboratory isolates of P. aeruginosa and that the protective mechanism was independent of bacteriostatic activity. In the present study, we examined the effects of human tear fluid in vivo. Tears were collected from healthy human volunteers and were studied in vivo in mice. The effects on the virulence of both invasive and cytotoxic clinical isolates of P. aeruginosa were examined. Tear fluid was found to reduce the severity of disease when corneas were challenged with cytotoxic bacteria immediately after scratch injury, and it completely protected against susceptibility to infection by a cytotoxic strain in a model in which corneas were infected during the healing process 6 h after scratching. Visible protection correlated with the inhibition of bacterial colonization 1, 4, and 48 h postinoculation. Tear fluid also significantly reduced the severity of infections caused by invasive P. aeruginosa in the 6-h-healing model. This result also coincided with significantly reduced bacterial colonization at 48 h. In vitro, human tear fluid significantly reduced the ability of invasive and cytotoxic bacteria to translocate across corneal epithelia and increased transepithelial resistance with or without bacterial inoculation. These data show that human tear fluid can protect against P. aeruginosa corneal infection in vivo and that the mechanism likely involves enhanced epithelial barrier function in addition to protection of individual epithelial cells against bacterial internalization and cytotoxicity.


Subject(s)
Keratitis/immunology , Pseudomonas Infections/immunology , Pseudomonas aeruginosa/pathogenicity , Tears/immunology , Animals , Cells, Cultured , Colony Count, Microbial , Cornea/cytology , Cornea/microbiology , Disease Models, Animal , Epithelial Cells/microbiology , Female , Humans , Keratitis/microbiology , Mice , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/growth & development , Pseudomonas aeruginosa/isolation & purification , Rabbits
12.
Infect Immun ; 73(4): 2147-56, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15784557

ABSTRACT

We have previously shown that human tear fluid protects corneal epithelial cells against Pseudomonas aeruginosa in vitro and in vivo and that protection does not depend upon tear bacteriostatic activity. We sought to identify the responsible tear component(s). The hypothesis tested was that collectins (collagenous calcium-dependent lectins) were involved. Reflex tear fluid was collected from healthy human subjects and examined for collectin content by enzyme-linked immunosorbent assay (ELISA) and Western blot with antibody against surfactant protein D (SP-D), SP-A, or mannose-binding lectin (MBL). SP-D, but not SP-A or MBL, was detected by ELISA of human reflex tear fluid. Western blot analysis of whole tears and of high-performance liquid chromatography tear fractions confirmed the presence of SP-D, most of which eluted in the same fraction as immunoglobulin A. SP-D tear concentrations were calculated at approximately 2 to 5 microg/ml. Depletion of SP-D with mannan-conjugated Sepharose or anti-SP-D antibody reduced the protective effect of tears against P. aeruginosa invasion. Recombinant human or mouse SP-D used alone reduced P. aeruginosa invasion of epithelial cells without detectable bacteriostatic activity or bacterial aggregation. Immunofluorescence microscopy revealed SP-D antibody labeling throughout the corneal epithelium of normal, but not gene-targeted SP-D knockout mice. SP-D was also detected in vitro in cultured human and mouse corneal epithelial cells. In conclusion, SP-D is present in human tear fluid and in human and mouse corneal epithelia. SP-D is involved in human tear fluid protection against P. aeruginosa invasion. Whether SP-D plays other roles in the regulation of other innate or adaptive immune responses at the ocular surface, as it does in the airways, remains to be explored.


Subject(s)
Cornea/chemistry , Pseudomonas aeruginosa/pathogenicity , Pulmonary Surfactant-Associated Protein D/physiology , Tears/chemistry , Animals , Cells, Cultured , Cytoprotection , Epithelial Cells/microbiology , Humans , Lipopolysaccharides/metabolism , Mice , Pulmonary Surfactant-Associated Protein D/analysis , Rabbits , Recombinant Proteins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...